Next Article in Journal
Adaptations of the Genus Bradyrhizobium to Selected Elements, Heavy Metals and Pesticides Present in the Soil Environment
Next Article in Special Issue
Immunostimulating and Anticancer Activities of the Pectic Polysaccharide from Panax ginseng Leaves Treated with High Pressure/Enzyme Process
Previous Article in Journal
Genetic Variation and Sex-Based Differences: Current Considerations for Anesthetic Management
Previous Article in Special Issue
Meroterpenoids from Terrestrial and Marine Fungi: Promising Agents for Neurodegenerative Disorders—An Updated Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Pinosylvin: A Multifunctional Stilbenoid with Antimicrobial, Antioxidant, and Anti-Inflammatory Potential

by
Argyrios Periferakis
1,2,3,*,†,
Aristodemos-Theodoros Periferakis
1,3,†,
Lamprini Troumpata
1,
Konstantinos Periferakis
2,4,
Spyrangelos Georgatos-Garcia
5,6,
Georgia Touriki
7,
Christiana Diana Maria Dragosloveanu
8,9,*,
Ana Caruntu
10,11,
Ilinca Savulescu-Fiedler
12,13,
Serban Dragosloveanu
14,15,
Andreea-Elena Scheau
16,
Ioana Anca Badarau
1,
Constantin Caruntu
1,17 and
Cristian Scheau
1,16
1
Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
2
Akadimia of Ancient Greek and Traditional Chinese Medicine, 16675 Athens, Greece
3
Elkyda, Research & Education Centre of Charismatheia, 17675 Athens, Greece
4
Pan-Hellenic Organization of Educational Programs (P.O.E.P.), 17236 Athens, Greece
5
Tilburg Institute for Law, Technology, and Society (TILT), Tilburg University, 5037 DE Tilburg, The Netherlands
6
Corvers Greece IKE, 15124 Athens, Greece
7
Faculty of Law, Democritus University of Thrace, 69100 Komotini, Greece
8
Department of Ophthalmology, Faculty of Dentistry, The “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
9
Department of Ophthalmology, Clinical Hospital for Ophthalmological Emergencies, 010464 Bucharest, Romania
10
Department of Oral and Maxillofacial Surgery, “Carol Davila” Central Military Emergency Hospital, 010825 Bucharest, Romania
11
Department of Oral and Maxillofacial Surgery, Faculty of Dental Medicine, Titu Maiorescu University, 031593 Bucharest, Romania
12
Department of Internal Medicine, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
13
Department of Internal Medicine and Cardiology, Coltea Clinical Hospital, 030167 Bucharest, Romania
14
Department of Orthopaedics and Traumatology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
15
Department of Orthopaedics, “Foisor” Clinical Hospital of Orthopaedics, Traumatology and Osteoarticular TB, 021382 Bucharest, Romania
16
Department of Radiology and Medical Imaging, “Foisor” Clinical Hospital of Orthopaedics, Traumatology and Osteoarticular TB, 021382 Bucharest, Romania
17
Department of Dermatology, “Prof. N.C. Paulescu” National Institute of Diabetes, Nutrition and Metabolic Diseases, 011233 Bucharest, Romania
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Curr. Issues Mol. Biol. 2025, 47(3), 204; https://doi.org/10.3390/cimb47030204
Submission received: 14 February 2025 / Revised: 13 March 2025 / Accepted: 15 March 2025 / Published: 18 March 2025
(This article belongs to the Special Issue Molecular Research in Bioactivity of Natural Products, 2nd Edition)

Abstract

:
Stilbenoids are a category of plant compounds exhibiting notable health-related benefits. After resveratrol, perhaps the most well-known stilbenoid is pinosylvin, a major phytochemical constituent of most plants characterised by the pine spines among others. Pinosylvin and its derivatives have been found to exert potent antibacterial and antifungal effects, while their antiparasitic and antiviral properties are still a subject of ongoing research. The antioxidant properties of pinosylvin are mostly based on its scavenging of free radicals, inhibition of iNOS and protein kinase C, and promotion of HO-1 expression. Its anti-inflammatory properties are based on a variety of mechanisms, such as COX-2 inhibition, NF-κB and TRPA1 activation inhibition, and reduction in IL-6 levels. Its anticancer properties are partly associated with its antioxidant and anti-inflammatory potential, although a number of other mechanisms are described, such as apoptosis induction and matrix metalloproteinase inhibition. A couple of experiments have also suggested a neuroprotective potential. A multitude of ethnomedical and ethnobotanical effects of pinosylvin-containing plants are reported, like antimicrobial, antioxidant, anti-inflammatory, hepatoprotective, and prokinetic actions; many of these are corroborated by recent research. The advent of novel methods of artificial pinosylvin synthesis may facilitate its mass production and adoption as a medical compound. Finally, pinosylvin may be a tool in promoting environmentally friendly pesticide and insecticide policies and be used in land remediation schemes.

1. Introduction

In current times, there is a wide availability of pharmaceutical agents for treating the causes and symptoms of a broad spectrum of pathologies [1,2,3]. However, many of these drugs, albeit effective, are associated with side-effects, which may be comparatively mild or even severe [4,5,6,7,8,9,10]. In the specific case of antimicrobial agents, microbial resistance has become another pronounced problem [11,12,13,14].
In an effort to find alternative therapeutical agents and based also on a rich medical–ethnobotanical tradition of different cultures, phytomedicine has been steadily gaining ground in the last decades [15,16,17,18,19,20,21,22,23,24,25,26,27,28]. Numerous compounds have been explored for their potential medicinal effects such as capsaicin, quercetin, curcumin, and kaempferol, among others [29,30,31,32,33,34,35,36,37,38,39].
Pinosylvin, a stilbenoid polyphenol mostly found in plants of the Pinaceae family, is the focus of this paper. It is believed that pinosylvin and its derivatives form part of a defence mechanism of the plants producing them, against microorganisms and insects [40]; it was isolated and identified for the first time in 1939 [41]. Currently, there exists a wealth of data on the antibacterial, antifungal, antiparasitic, antiviral, antioxidative, anti-inflammatory, anti-carcinogenic, and neuroprotective properties and potential.
In this review, we will provide a comprehensive overview of the current knowledge available on pinosylvin and its derivatives, presenting the research pertaining to its aforementioned activities and potential, in a clear and detailed manner. The associated mechanisms and potential links between the antioxidant and anti-inflammatory applications will also be explored, as well as the available evidence for its anticancer properties and main traditional uses of pinosylvin-containing plants. It is our intention, via this mode of presentation, and based on our detailed bibliographical research, using Google Scholar, PubMed, and Scopus databases, to incorporate the current state of knowledge on this topic and to provide directions for future research.

2. Biosynthesis and Bioavailability of Pinosylvin

In general, all stilbenoids are phenolic derivatives of the phenylpropanoid pathway [42]. Their common structural characteristic is the stilbene core of phenyl rings connected by a methylene bridge [43]; based on the precise arrangement of these phenyl rings and a variety of substitution patterns, stilbenoids are classified in various subclasses [44]. The stilbenoids are hydroxylated stilbene derivatives, having a biosynthesis pathway which shares many steps with that of chalcones [45,46] (Figure 1).

2.1. Synthesis of Pinosylvin in Plants

The first isolation of pinosylvin (3,5-dihydroxy-trans-stilbene) was performed in 1939, by V. H. Erdtman [47], in extracts of Pinus silvestris, hence its name; since then it has been identified in a number of plant species (Table 1). The common precursor molecule of all stilbenoids is phenylalanine or tyrosine, which are obtained by glucose metabolism, through the shikimate or arogenate pathways [48]. Pinosylvin, resveratrol, and other stilbenoids are classified as phytoalexins, and are synthesised by some plants in response to environmental stressors [49,50]. Both flavonoid and stilbenoid phytoalexins are derived through the universal phenylpropanoic–polymalonic acid pathway; endogenous and exogenous stimuli regulate the expression of these pathways [50]. Pinosylvin synthase, the key enzyme in the biosynthetic sequence, was isolated in 1984, using column chromatography [51]. A detailed description of stilbene biosynthesis is provided by Mendonça et al. [52].
A schematic of the biosynthetic and chemical pathways of pinosylvin synthesis is available in Figure 2.

2.2. Pharmacokinetics of Pinosylvin

Details on the metabolism of stilbenes and pinosylvin in particular are relatively unknown [82]; the particularities of the metabolism of different stilbenes hinder the development of a generalised understanding of their metabolism as a class [82,83].
A part of them is metabolised in the liver, via glucuronidation and sulfation; the other part is metabolised in the intestine [82,83,84,85,86,87]. Stilbene sulfation seems to be associated with their cardioprotective and anticancer activities [88]. Regarding intestinal metabolism, perhaps some elements can be inferred from related experiments on resveratrol, whose intestinal biotransformation peaks at about one hour post-consumption; however, hepatic biotransformation is much more important [89,90]. For resveratrol, the oral bioavailability has been found to be about 30% in rats, but in humans it is about 0.5% [91].

2.3. Methods of In Vitro Pinosylvin Synthesis

Pinosylvin does not naturally occur in appreciable amounts and so its isolation and purification by conventional methods requires a complex multi-step process, which is technically challenging and economically unsustainable [41]. Direct chemical synthesis also has similar drawbacks, coupled with the formation of dangerous by-products [92]. In the last few years, artificial biosynthesis of hydroxystilbenes and associated compounds has gained traction [93,94,95]. The currently available methods for pinosylvin synthesis are presented in Table 2 in chronological order.
The first method for artificial pinosylvin procurement was proposed by Jorgensen [96,97]; the method demonstrated that by causing mechanical damage to the stem and cambium of red pine, fungal penetration ensues and this leads to the synthesis of pinosylvin and its monomethyl ether. This method made possible a more detailed study of the biosynthetic pathway of pinosylvin, in plant cells in vitro [98]. So, in this case, pinosylvin is produced by the plant cells in response to desiccation. Improvements to this original scheme were soon proposed by Jorgensen and Balsillie [96].
Another approach is the production of pinosylvin from in vitro cell cultures of Pinus strobus L. cells in a specially modified culture medium [99]. It is also possible to induce pinosylvin accumulation in methanolic extracts, from Pinus sylvestris L. cell cultures by using an elicitor preparation of the spine needle pathogen [100].
Apart from these methods, which are centred on using plant cells, in one way or another, the production of pinosylvin using bacterial cells has been tried successfully [101]. Pinosylvin, as well as other stilbenes, has been produced by the genetic modification of E. coli as well by similar modifications on Pseudomonas taiwanensis and Corynebacterium glutamicium [102,103,104,105,106,107,108,109,110].
Table 2. Methods of artificial synthesis of pinosylvin and other associated stilbenes, by chrono-logical order.
Table 2. Methods of artificial synthesis of pinosylvin and other associated stilbenes, by chrono-logical order.
Pinosylvin ProducerMethodResultYearReferences
Cells of P. resinosa Stimulation of plant cells in response to desiccationProduction of pinosylvin and pinosylvin monomethyl ether1961[97]
Cells of P. resinosaStimulation of plant cells in response to desiccationProduction of pinosylvin and pinosylvin monomethyl ether1969[96]
Pinus sylvestris L. cellsTreatment of cells with an elicitor preparation from the pine needle pathogen L. seditiosumProduction of pinosylvin and pinosylvin-3-O-methyl ether1994[100]
Metabolically engineered E. coliConstruction of a pathway for stilbene biosynthesis inside E. coli cellsProduction of stilbene polyketides2006[102]
Metabolically engineered E. coliConstruction and subsequent modification of a pathway for stilbene biosynthesis inside E. coli cellsProduction of stilbene methyl ethers2007[103]
Metabolically engineered E. coliConstruction of a pathway for stilbene biosynthesis inside E. coli cellsProduction of pinosylvin2015[104]
Metabolically engineered E. coliEstablishment of a variety of biosynthetic pathways in E. coli cells using enzymes from different sourcesProduction of various phenylpropanoid derivatives2015[105]
Metabolically engineered E. coliDevelopment of three different bioengineering strategiesProduction of pinosylvin2016[106]
Metabolically engineered C. glutamiciumConstruction of a pathway for stilbene biosynthesis inside C. glutamicium cellsProduction of pinosylvin and other associated compounds2016[110]
Metabolically engineered E. coliConstruction and subsequent modification of a pathway for stilbene biosynthesis inside E. coli cellsProduction of pinosylvin2018[107]
Metabolically engineered E. coliReduction of specific gene expression in order to increase pinosylvin production in already-modified E. coliProduction of pinosylvin (increased compared to the originally modified strain)2018[108]
Callus cells of P. strobus L.Aging of callus cells in a specially modified culture mediumProduction of pinosylvin stilbenes2022[99]
Metabolically engineered P. taiwanensisConstruction of a pathway for polyketide biosynthesis inside P. taiwanensis cellsProduction of polyketides2023[109]

3. Pinosylvin as an Antibacterial Agent

In the last decades, plant metabolites have been steadily gaining ground as viable antibacterial agents which may help mitigate the problem of microbial resistance to antibiotics, that is caused both by the versatility and adaptability of bacteria—indeed, they survive in a host of adverse environments such as hot springs—and the overuse of antibiotics [29,111,112,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128,129,130]. Apart from potential resistance, most common antibiotics may be associated with allergies, hepatoxicity, changes to normal microbiota, and other side-effects [131,132,133,134,135,136,137,138].
The spectrum of antibacterial properties of pinosylvin and some of its derivatives is quite broad, ranging from relatively unknown pathogens to potentially dangerous and resistant strains (Table 3).

3.1. Antibacterial Properties Against Achromobacter xylosoxidans

This pathogen is associated with nosocomial infections and notable resistance to antibiotics [148,149]. Medical water, particularly sterile distilled water bottles, is one of its main reservoirs [148]. It can cause a wide range of pathologies, such as ecthyma gangrenosum, bacteraemia, and pneumonia [149,150].
According to Lindberg et al. [60], the strain A50182 was affected by the presence of pinosylvin though not as much as the Gram-positive bacteria included in the study. At any rate, the extracts P. sylvestris, P. resinosa, and P. contorta were the most impactful, possibly due to the presence of stilbenes.

3.2. Antibacterial Properties Against Arcobacter butzleri

This bacterium is closely related to Campylobacter spp., shows increased resistance, and was recognized as an emerging pathogen in 2002 [139,151,152,153]. It can be found in many different types of food such as meat, dairy products, vegetables, and seafood and usually affects the digestive tract [151,153]. The increased resistance means that research should be directed at finding additional treatment options.
Pinosylvin was found to be effective against two strains of A. butzleri, namely DQ46M1 and CR50-2, at a concentration of 128 μg/mL [139]. Another beneficial aspect is the synergistic action it displayed alongside several antibiotics, as was made evident by the reduction in MIC [139]. Interestingly, the results of Sousa et al. [139] also suggest that pinosylvin could potentially even revert antibiotic resistance. The efficacy of pinosylvin seems to be related to its ability to interact with the activity of efflux pumps, inhibiting them [139].

3.3. Antibacterial Properties Against Bacillus spp.

3.3.1. Bacillus cereus

This is a species of facultative anaerobic bacteria, which are most notable for their ability to form spores, thus becoming more resistant to a variety of factors [154,155]. B. cereus can induce significant mortality, especially when it affects the CNS [156,157]. It is commonly associated with ocular, respiratory, and wound infections, although it is most frequently encountered in contaminated food, such as in the case of the ‘‘fried rice syndrome’’ [157,158].
Pinosylvin was found to be potent in its action against B. cereus though the mechanism behind its significant effectiveness has not been elucidated [41,146]. In fact, Välimaa et al. [140] found it to be the most susceptible to pinosylvin alongside L. monocytogenes.

3.3.2. Bacillus coagulans

This probiotic bacterium has been very useful in its application for medicinal purposes, most notably its beneficial role in several intestinal pathologies, like constipation, diarrhoea, colitis, and, most notably, irritable bowel syndrome [159,160].
There is evidence that showcases the effectiveness of pinosylvin against this bacterium, more specifically the strain E50L1, especially when compared with its effectiveness against Gram-negative bacteria [60]. This is likely a result of the stilbenes contained in the extracts; the extracts of P. sylvestris, P. resinosa, and P. contorta were the most potent in that regard.

3.3.3. Bacillus subtilis

This bacterium not only is non-pathogenic but has also been studied extensively due to a multitude of factors, contributing greatly to our understanding of several biological aspects [161]. It has even been used as a probiotic, in order to regulate intestinal flora, and recently it is being looked into as a potential vaccine expression vector [162].
Pinosylvin is active against B. subtilis at a concentration of 64 g/mL [141]. This activity was corroborated by Bouarab-Chibane et al. [142]. It is presumed that the susceptibility of B. subtilis, as well as that of other Gram-positive bacteria, is somehow related to their membrane structure [163].

3.4. Antibacterial Properties Against Burkholderia multivorans

Burkholderia multivorans belongs to the Burkholderia cepacian complex, which is notable for its resistance to antibiotics [164,165]. This bacterium usually infects the lungs of patients suffering from cystic fibrosis, though it is also becoming an important cause of neonatal sepsis [164,165,166]; rarely, it may also cause bacterial meningitis [167].
The research of Lindberg et al. [60] found this bacterium to be susceptible to the action of pinosylvin, most probably because of the extracts’ stilbenes, though less so than the Gram-positive bacteria on which it was tested. From the extracts included in this research, P. sylvestris, P. resinosa, and P. contorta were the most effective. The specific strains which were found to be susceptible were F45L5 and F453DL1 [60].

3.5. Antibacterial Properties Against Campylobacter spp.

Both C. coli and C. jejuni can be isolated from poultry and poultry-related products [168]. Campylobacter spp., and C. jejuni in particular, are the leading cause of food-borne pathologies all over the globe [168,169,170]. The commonly associated condition is a usually self-limiting gastroenteritis [171]. However, it can also have serious consequences like miscarriage or even Guillain–Barré syndrome due to molecular mimicry [172,173]. Campylobacter coli can also cause severe conditions, albeit rarely, like bacteraemia, and recently, the first recorded case of infantile myocarditis [174,175]. Their resistance to antibiotics is an ever-growing concern that needs to be addressed [170].
Pinosylvin seems to be a promising agent against Campylobacter spp. [41]; the compound described by Silva et al. [144] exhibited colony count inhibition higher than 99% at a concentration as low as 0.08 mg/cm2. This research also demonstrated the effectiveness of coated pad-pinosylvin ICs in preventing Campylobacter contamination of fresh chicken. The effectiveness of pinosylvin in the case of Campylobacter spp. is attributed to its ability to damage the bacterial membrane, therefore acting in a bactericidal way [143].

3.6. Antibacterial Properties Against Escherichia coli

This bacterium makes up an important part of the physiological gut microbiota, but it may cause opportunistic infections, especially in the case of immunocompromised patients [176,177]. This becomes a significant problem in the wake of the appearance of multi-drug resistant (MDR) E. coli strains [178].
Pinosylvin was found to be effective in this regard, namely at a concentration of 250 µg/mL [145]; promising results were also discovered by Bouarab-Chibane et al. [142]. This effectiveness was corroborated by the findings of Hou et al. [141], though at a different concentration, namely 68 g/mL. However, related to the susceptibility of other bacteria, Välimaa et al. [140] concluded that pinosylvin is on the lower side of efficacy against E. coli.

3.7. Antibacterial Properties Against Listeria monocytogenes

These ubiquitous, intracellular bacteria are transmitted via the faecal–oral route and are the cause of foodborne pathologies, owing in large part to their ability to survive in refrigerator temperatures [179,180,181,182]. Infection with this pathogen can lead to severe complications like meningoencephalitis in immunocompromised patients and pregnant women and their foetuses, while healthy adults may experience the disease as a self-limiting gastroenteritis [179,180,181,182].
Pinosylvin derivatives have been found to be effective against L. monocytogenes [41,142,183]. It is at the storage temperature of 8 °C that pinosylvin exerts the highest effectiveness against inoculated L. monocytogenes, completely inhibiting its multiplication at a concentration of 140 μg/g [146]. As the temperature rose to 20 °C, the antibacterial effect was reported to last for 72 h, with bacterial growth becoming logarithmic and profuse after this temporal threshold [146]. These findings are corroborated by the research of Välimaa et al. [140] who found L. monocytogenes to be the most susceptible to pinosylvin alongside B. cereus.

3.8. Antibacterial Activity Against Proteus vulgaris

P. vulgaris, alongside P. mirabilis, are the most notable species of their genus. The disease most commonly associated with this bacterium are urinary tract infections, such as cystitis and pyelonephritis [184,185]. Alarmingly, P. vulgaris has been associated with resistant nosocomial infections [186]. Pinosylvin is active against P. vulgaris at a concentration of > 128 g/mL [141].

3.9. Antibacterial Activity Against Pseudomonas spp.

3.9.1. Pseudomonas aeruginosa

P. aeruginosa is commonly implicated in nosocomial infections [187,188,189]. These infections range from urinary tract infections to pneumonia and can have significant morbidity and mortality, especially when the patient also suffers from COPD or cystic fibrosis [187,188,189,190,191,192]. An exacerbating factor is the polyresistance commonly encountered in the strains of this bacterium [187,188,189]. Pinosylvin is active against P. aeruginosa at a concentration of >128 g/mL [141]. This activity is corroborated by the research of Bouarab-Chibane et al. [142].

3.9.2. Pseudomonas fluorescens

These ubiquitous bacteria can be found as part of the physiological microbiota on many parts of the human body and can occasionally cause opportunistic infections [193,194]. These infections range from bacteraemia to pneumonia or can even be ocular [18,194,195]. It is worth mentioning that drug-resistant strains are emerging [193], which is a cause for alarm. Pinosylvin has exhibited some potential as an agent against the growth of this bacterium, but not as notable as in the case of other bacteria [41,140].

3.10. Antibacterial Activity Against Salmonella spp.

Salmonella infantis, a serovar of S. enterica, has a zoonotic transmission and it is implicated in a significant number of foodborne infections worldwide, being mainly found in broilers and their meat [196,197]. Alarmingly, it presents with resistance to a wide range of antibiotics, including colistin which is among the last lines of defence, while also being highly virulent, this being attributed in large part to its biofilm-forming capacity [196,197]. These multidrug-resistant (MDR) strains have the potential to cause major health problems, especially in low- and middle-income countries [197].
Pinosylvin has exhibited some potential as an agent against the growth of this bacterium [41], though not nearly as prominently as in the case of other bacteria [140]. The research of Bouarab-Chibane et al. also highlighted the effectiveness of pinosylvin against S. enteritidis, another common serovar of salmonella associated with foodborne disease outbreaks [142,198].

3.11. Antibacterial Activity Against Staphylococcus spp.

3.11.1. Staphylococcus aureus

This bacterium often colonizes the human body, though it is also the causative agent of several infectious conditions such as food poisoning, scalded skin syndrome, pneumonia, and bacterial endocarditis [199,200,201,202]. Staphylococcus is particularly notable in the clinical setting due to its implications in implant infections during orthopaedic surgery, as well as to the existence of particularly resistant strains, namely the methicillin-resistant Staphylococcus aureus strains, which have a very negative impact on patient morbidity and mortality [203,204,205,206]. Due to the emergence of strains with reduced susceptibility to vancomycin, one of the last lines of defence against this pathogen [206], extensive research in order to formulate effective treatment schemes is necessary [199].
Pinosylvin has been found to be effective in inhibiting S. aureus at concentrations of 75 μg/mL to 250 µg/mL, while prenylated pinosylvin was able to inhibit the growth of even MRSA strains at a concentration of 12.5 µg/mL [145,146,147]. The findings of Bouarab-Chibane et al. were similar, as were those of Hou et al., the latter settling on a concentration of 64 g/mL, and regarding MRSA, pinosylvin acid derivatives exhibited effectiveness at a concentration of 16 g/mL [141,142]. In any case, the data provided by Välimaa et al. [140] suggest that the action of pinosylvin against S. aureus is considered moderate when compared to its effects on other bacteria.

3.11.2. Staphylococcus epidermidis

This bacterium can be found as a commensal on the human skin and occasionally in the nasopharynx [207]. Even though it has a beneficial role in maintaining the integrity of the skin as a barrier against infection, it can occasionally become pathogenic, especially when it comes to contaminating implants, and in a select few cases, it can even cause toxic shock syndrome [208,209,210,211]. Pinosylvin is active against S. epidermidis at a concentration of 128 g/mL [141].

4. Pinosylvin as an Antifungal Agent

In general, there are vastly fewer fungi which are pathogenic to humans compared to bacteria [212]. Even then, most fungal infections are not considered life-threatening, unless there exist particular risk factors. Be that as it may, fungal infections still represent an appreciable threat to human health, especially in some particular regions [213,214,215]. Just like in the case of bacteria, resistance is a potentiality [216,217,218], while some side-effects of antifungal drugs have also been noted.
The spectrum of antifungal properties of pinosylvin and some of its derivatives has not been tested as extensively as its antibacterial potential; nevertheless, the results are encouraging and justify further research on the subject (Table 4).

4.1. Antifungal Activity Against Aspergillus fumigatus

This is a ubiquitous saprophytic fungus [219]. Aspergillosis can be dangerous, particularly when it encounters a depressed immune system [219]. A notable case is that of allergic bronchopulmonary aspergillosis which affects patients with asthma or cystic fibrosis and can lead to bronchiectasis [220]. The emergence of triazole-resistant Aspergillus fumigatus strains is therefore a cause for concern [221].
There are studies that highlight the potency of pinosylvin against this microbe by Bakrim et al. [41] and Välimaa et al. [140]. The extracts tested in the latter study [140] were able to inhibit the germination and growth of the fungus.

4.2. Antifungal Activity Against Candida albicans

Candida spp. are often harmless but on occasion, particularly in the case of Candida albicans, they are capable of causing pathologies, most notably related to the vagina and the oral cavity, while also possibly playing a negative role in the exacerbation of chronic inflammatory bowel diseases [222,223,224,225]. Infections caused by this microorganism may be severe, leading to significant nosocomial complications [224,226,227,228].
Pinosylvin has potent antifungal effects against Candida albicans according to Barkim et al., Välimaa et al., and Lee et al., in the latter case, at a concentration of 62.5 μg/mL [41,140,145]. As noted in the work of Välimaa et al. [140], all Pinus extracts were effective in this regard, but the most prominent action was that exhibited by the extracts of P. resinosa, P. strobus, and P. sibirica. Pinocembrin, another flavonoid, is similarly effective though the mechanism behind this has not yet been elucidated [73].

4.3. Antifungal Activity Against Cladosporium herbarum

This microorganism has been known to occasionally cause hypersensitivity pneumonitis, a delayed allergic reaction [229,230]. Thankfully, barring the case of patients with compromised immunity status, they do not seem to be otherwise pathogenic [231].
Kostecki et al. [78] found that compounds with an unsubstituted aromatic ring A, like pinosylvin, exhibit notable antifungal activity, its EC50 being 10mg/mL. The findings of Pacher et al. [79] are similar and extend to four microfungi, namely Alternaria citri, Fusarium avenaceum, Pyricularia grisea, and Botrytis cinerea. Among them, P. grisea, a necrotrophic plant pathogen with widespread distribution that causes grey mould disease, was the most susceptible to the action of the stilbenoids [79,232].

4.4. Antifungal Activity Against Plasmopara viticola

This obligate biotrophic oomycete is the causative agent of the grapevine downy mildew [233], a major threat for grape cultures and wine production worldwide [233,234,235]. Its effectiveness as a pathogen is attributed to a great variety of pathogenicity effectors, and it has long since developed resistance to several fungicides [233,234,235].
The effectiveness of pinosylvin against this oomycete has been highlighted by Bakrim et al. and Gabaston et al. [41,73]. In particular, Gabaston et al. [73] assumed that the Pinus pinaster knot extract owes this effectiveness to its high concentration of pinosylvin, pinosylvin monomethyl ether, and pinocembrin. This claim is supported by the results of the bioassays of these molecules in the presence of Plasmopara viticola [73]. Based on this information, Gabaston et al. [73] suggest that the by-products of Pinus pinaster knot could be used in the field of viticulture as protective agents.

4.5. Antifungal Activity Against Penicillium brevirocompactum

This species of penicillium, alongside several others such as P. expansum and P. digitatum, is responsible for fruit spoilage, particularly when it comes to pears [236]. This has considerable implications as far as market profits are concerned [236]. As far as the field of medicine is concerned, in vitro models have exhibited the capacity of its metabolites to bring about inflammatory reactions and cytotoxic effects in mice [237]. In a similar vein, it has been proven to be a potent sensitizer for asthmatic patients [238].
Based on the writings of Bakrim et al. and Välimaa et al. [41,140], pinosylvin is a potent agent against P. brevicompactum. The extracts included in the tests of Välimaa et al. [140] were effective in inhibiting both the germination and the growth of this microorganism.

4.6. Antifungal Activity Against Saccharomyces cerevisiae

This microorganism has a host of applications, playing an important role in the production of fermentative foods and drinks, but also of biofuels [239,240]. Its genome has been domesticated and sequenced, allowing for many laboratory applications, such as the synthesis of an eukaryotic genome similar to its own [239,240]. From a purely medicinal point of view, it has been labelled as ‘‘generally regarded as safe’’ [240].
Pinosylvin has been found to be effective against this fungus according to both Bakrim et al., Välimaa et al., and Lee et al. [41,140,145]. In particular, Lee et al. [145] stated that the MIC is 125 μg/mL.

5. Pinosylvin as an Antiparasitic and Antiviral Agent

The data on the antiparasitic and antiviral properties of pinosylvin are scarce compared to the corresponding data on its antibacterial and antifungal properties.
Regarding its antiparasitic activity, pinosylvin monomethylether (PSM), alongside (−)-nortrachelogenin, were identified as nematicidal substances; the resistance of some pines, such as P. strobus and P. palustris, to the pinewood nematode Bursaphelenchus xylophilus is most likely attributed to them [71]. This is important because the parasite can cause pine wilt disease and has subsequently been quarantined in a number of countries following the application of strict protocols [241] due to extensive crop losses [242]. It should be noted that pinosylvin itself was also active against the parasite’s propagative larvae, though not to the same extent as PSM (LD50 of 4 ppm); this finding is one of several that suggest that a trans-type double bond conjugated with two aromatic rings and a hydroxyl group is necessary for nematicidal activity [71].
Regarding its antiviral activity, relevant research efforts were undertaken in the wake of the recent SARS-CoV-2 pandemic; this pandemic highlighted the highly contagious nature of the virus, and also had important psychological repercussions and effects on many people and age groups [243,244,245,246,247,248,249,250,251,252,253,254,255,256]. Though its main target is the lungs, where it brings about acute respiratory distress syndrome, the virus can find its way to several other parts of the body and has even been implicated in coagulopathies [250,257,258,259,260]. Due to the large size of its genome and its notable recombination capacity, it poses a challenge, as our understanding of the available effective therapeutic options is still developing [243,258,260].
The research of Naseem et al. [261] described the interaction of pinosylvin monomethyl ether as a ligand binding to the SARS-CoV-2 protein, Mpro. In this in silico model, the molecular docking study found out that this pinosylvin derivative was capable of binding to the protein, though not as effectively as other tested stilbene compounds [261]. Interestingly, an in vitro model testing synthesized stilbene derivatives found them to be similarly capable of inhibiting SARS coronavirus replication [262].

6. Antioxidant Properties of Pinosylvin

Pinosylvin, as well as its derivatives, possess a remarkable number of antioxidant properties exerted by different mechanisms (Table 5). There is evidence to suggest that the antioxidant properties of pinosylvin are associated with its anticancer and anti-inflammatory properties [263,264,265,266,267,268,269,270].
A number of researchers have identified the ability of such compounds to scavenge free radicals either in pure form or when present in extracts evaluated by specific in vitro assays [63,69,271,272].
The research team of Park et al. [266] investigated the effect of pinosylvin on the production of nitric oxide (NO), using lipopolysaccharide (LPS)-stimulated murine macrophage RAW 264.7 cells. It is known that NO is physiologically produced via a reaction involving nitric oxide synthase (NOS), which exists in three isoforms [273,274]. A deficiency of some NOS isoforms leads to disease [275,276], as well as excessive NOS expression [277]. An isoform of NOS, termed by the researchers as inducible NOS (iNOS), is stimulated, among others, by LPS, hence the treatment of the RAW 264.7 cells. It was found that the antioxidant effect of pinosylvin in this case was associated most probably with TRIF-mediated signalling, inhibition of iNOS directly, and via mRNA expression inhibition [266]. Toll/IL-1R domain-containing adaptor-inducing IFN-b (TRIF) and its associated signalling pathway are necessary for interferon and other pro-inflammatory mediator production [278].
The research of Jančinová et al. [267], who incubated human neutrophils with pinosylvin in the presence of different pro-oxidant stimuli, determined that pinosylvin could suppress oxidant generation, probably via the inhibition of protein kinase C; a very similar mechanism has been demonstrated for resveratrol-induced protein kinase C inhibition [279]. This enzyme has long been thought of as instrumental in redox modification, with an important role in health and disease [280,281].
Jeong et al. [268] used bovine aortic endothelial cells, harvested from descending thoracic aortas, and using various concentrations of pinosylvin and different incubation times, found that it exerted an anti-apoptotic, proliferative, remodelling, wound healing-inducing, and anti-atherogenic effect. All these effects stem from the antioxidant effect of pinosylvin, which is mediated through the NO generation pathway [268].
Moreover, it was found that antioxidative protection against oxidative stress can be conferred, via inducing heme oxygenase 1 (HO-1) in human retinal ARPE-19 cells, conferring protection against oxidative stress [282]. In general, HO enzymes are involved in heme degradation, utilising NADPH and oxygen [283,284,285]. Concurrent effects on modulating growth factors associated with macular degeneration and other retinopathies or neuropathies may be beneficial for addressing a variety of diseases involving imbalances between ROS generation and the antioxidant defence system [286,287,288,289,290].
The more recent experiment of Wang et al., conducted on mice, aimed to explore the effects of pinosylvin on male infertility caused by oligoasthenospermia [291,292]. It was found that pinosylvin increased epidydimal sperm concentration and motility and induced an increase in testosterone levels, by decreasing oxidative stress via activation of the nuclear factor E2-related factor 2-antioxidant response element (Nrf2-ARE) pathway, an intrinsic defence mechanism against oxidative stress, also implicated in the progress of neuroinflammation and neurodegenerative diseases [291,293].
In 2010, Macickova et al. [265] induced adjuvant arthritis in rats, a condition known to cause an increase in pro-oxidant compounds in blood and tissues [294,295,296]. Indeed, they determined that hind paw volume and the chemiluminescence of the affected joint were reduced, indicating reduced inflammation and pro-oxidant compound levels [265]. The induction of adjuvant arthritis in rats was also employed by Jančinová et al. [267], and the oral administration of pinosylvin had reduced both neutrophil number and oxidant formation, thereby reducing inflammation. A potent effect against adjuvant arthritis was also noted by Bauerova et al. [270].
Also, in rats with adjuvant arthritis, the administration of pinosylvin, either as a monotherapy or in combination with carnosine resulted in a decrease in most inflammatory markers; this effect is believed to be associated with the antioxidant properties of these compounds [269].
Table 5. Experiments on the antioxidant properties of pinosylvin and its derivatives.
Table 5. Experiments on the antioxidant properties of pinosylvin and its derivatives.
CompoundSourceTest TypeMechanismConcentrationAdministrationYearReference
Pinosylvinn/a (pure compound)In vitro—pulse radiolysis experimentsFree radical scavenging in pH values between 2 and 120.1 mM aqueous solutionn/a2002[271]
Pinosylvinn/a (laboratory synthesis)In vivo—rat modelInhibition of neutrophil infiltrationn/aoral daily dose of 30 mg/kg b.w. for 28 d2010[265]
Pinosylvinn/a (pure compound)In vitro—LPS-stimulated RAW 264.7 cells(probable) TRIF-mediated signalling, iNOS and mRNA expression inhibition39.9 μΜ (IC50) Pretreatment of cells with pinosylvin before LPS stimulation2011[266]
Pinosylvinn/a (laboratory synthesis)In vitro—human neutrophils(probable) inhibition of protein kinase C14.16 ± 1.46 μΜ/L (EC50)Incubation of cells with pinosylvin2012[267]
In vivo—rat modelReduction in neutrophilia and oxidants productionn/aOral daily dose of 30 mg/kg for 21 d
Pinosylvinn/a (laboratory synthesis)In vitro—bovine aortic endothelial cellsMediation of NO productionVarious (depending on different experimental protocols)Incubation of cells with pinosylvin2012[268]
Pinosylvinn/a (pure compound)In vivo—rat modelReduction in pro-oxidative processesn/aOral daily dose of 30 mg/kg b.w. per os for 28 d2012[269]
Pinosylvinn/a (pure compound)In vitro—human retinal pigment epithelial (ARPE-19) cellsPromotion of HO-1 expressionVarious (depending on different experimental protocols)Incubation of cells with pinosylvin2014[282]
Pinosylvinn/a (laboratory synthesis)In vivo—rat modelPromotion of hepatic and pulmonary NF-κB activation, increase in lung lipo-oxygenase and promotion of plasma antioxidant statusn/aOral daily dose of 50 mg/kg b.w. twice a week for 28 d2015[270]
Pinosylvin monomethyl ether, pinosylvin, pinosylvin dimethyl etherP. merkusiiIn vitro—free radical scavenging experimentsUptake of reactive oxygen species 11.4–25.8 mg/L (EC50 for extract)n/a2015[69]
Pinosylvinn/a (pure compound)In vitro—ORAC-FL, ABTS and FRAP assaysFree radical scavengingVarious (depending on the assay)n/a2017[272]
Pinosylvin n/a (laboratory synthesis)In vitro—mouse modelActivation of the Nrf2-ARE pathwayn/aIntragastric daily administration of 100 mg/kg b.w for 2 w2020[291]
Pinosylvin, pinosylvin monomethyl etherP. caribaeaIn vitro—antioxidant assays using DPPH and ABTS methodsFree radical scavenging (electron donation/cation scavenging)17.25 ± 0.78 μg/mL (IC50 for extract)n/a2023[63]

7. Anti-Inflammatory and Anti-Allergic Properties of Pinosylvin

In general, natural stilbenoids, as well as their synthetic analogues, are considered effective anti-inflammatory agents [297,298]; both pinosylvin and its derivatives are potent inhibitors of inflammation, as proven by the extensive testing performed in vitro and in laboratory animals (Table 6).
Perhaps the most important mechanism regulating the inflammatory response in humans is the cyclo-oxygenase 2 (COX) pathway [299,300]. This is a rate-limiting step in the synthesis of prostaglandins (PGEs) and thromboxane A2 (TXA2) [301,302]. Inhibition of COX-2 pathway will inevitably lead to reduction in inflammation [303,304].
The earliest experiments with pinosylvin and its derivatives regarding their anti-inflammatory role were performed by Park et al. [297]; it was demonstrated that pinosylvin and some of its derivatives were effective in inhibiting COX-2-associated prostaglandin (PGE) production [297,305].
Pinosylvin, apart from its direct COX-2 inhibition, also inhibits COX-2 indirectly, via the transcriptional inhibition of NF-κB and its associated pathway [306]; it is known that NF-κB-associated signalling is associated with COX-2 production [307,308]. Still relating to the production of inflammatory mediators, the research of Adams et al. [81] demonstrated the potential of pinosylvin and other stilbenoids to inhibit leukotriene biosynthesis. In recent years, leukotriene metabolism has been the focus of research, concerning therapeutical anti-inflammatory interventions [309].
The anti-inflammatory properties of pinosylvin, connected to its antioxidant properties, were also demonstrated by Park et al. [266], in an in vitro study, as presented in the previous section. It will be recalled that similar results linking the antioxidant activity of pinosylvin with its anti-inflammatory one have already been demonstrated [267,294].
Another link between the antioxidant and anti-inflammatory properties of pinosylvin and its derivatives was revealed by Laavola et al. [76]; reduced NO production and iNOS expression, NF-κB transcription, along with interleukin 6 (IL-6) and monocyte chemoattractant protein-1 (MCP-1) production. IL-6 is important in inflammation and host immunity, but its dysregulation is connected to chronic inflammation and autoimmune pathologies [310]; MCP-1 functions by attracting monocytes, thereby contributing towards immunological surveillance at a tissue level [311]. In the in vivo arm of the same research, the administration of pinosylvin in rats with λ-carrageenan-induced inflammation led to a reduction in paw oedema [76]. Similar results regarding the effect of pinosylvin on IL-6 and MCP-1 were produced by Eräsalo et al. [312].
Likewise, the release of TNFα and IL-6 were inhibited by pinosylvin, due to its ability to inhibit the JAK/STAT pathway; a molecular docking study found that pinosylvin is able to bind to the active site of the JAK2 protein [264]. This pathway has been linked to inflammatory, auto-immune, and stress-related pathologies [313,314].
The in vivo and in vitro research of Moilanen et al. demonstrated a dose-dependent and rapid inhibition of TRPA1, which, among other functions, is involved in pain and odour perception [315,316,317,318]. Pinosylvin monomethylether was found to be an excellent peroxisome proliferator-activated receptor gamma (PPARγ) activator and it also reduced IL-6 activation [53]; PPARγ expression was also enhanced in murine macrophages, promoting a resolution of inflammation [319]; a similar result on inflammation resolution was noted by Kwon et al. [320]. On the other hand, in the experiment of Modi et al., pinosylvin was found to downregulate PPARγ and C/EBPa. The former has various roles in inflammation and even auto-immune diseases [321,322,323]; the role of the latter is still a subject of research [324,325,326].
Another in vivo study, performed by Aalto et al. [57] using Drosophila melanogaster, found that pinosylvin and pinosylvin monomethylether administration can inhibit induced immune responses in the intestine of the fruit fly by a transient receptor ankyrin 1 (TrpA1)-dependent antagonism. This is a Ca2+-permeable, non-selective cation channel, found in certain cell types [327]; it is currently under research as a promising target in the treatment of inflammatory diseases, pruritus, asthma, and even Alzheimer’s disease [328].
Regarding the anti-allergic properties of pinosylvin, only a few researchers have concentrated on this. Labib et al. [56] isolated (Z)-pinosylvin mono methyl ether, (Z)-pinosylvin-3-O-b-D-glucoside, along with other compounds, from the leaves of Agonis flexuosa, and found that it had an affinity for the human histamine receptor, and thus a histamine release inhibition, although at the moment there are more promising phytochemicals in relation to this aspect [56]. Pinosylvin, extracted from H. dulcis Thunb., inhibited the release of IL-4, TNF-α, and PGE2 and the expression of IL-4, TNF-α, COX-2, NFKB1, and NFKB2 in a dose-dependent manner in RBL-2H3 cells treated with IgE [54]. All of these hormones and mediators are important in the process of allergic responses [292,329,330].
Table 6. Experiments on the anti-inflammatory and anti-allergic properties of pinosylvin and its derivatives.
Table 6. Experiments on the anti-inflammatory and anti-allergic properties of pinosylvin and its derivatives.
CompoundPlantTested onMechanismConcentrationAdministrationYearReference
Pinosylvin (and other derivatives)n/a (laboratory synthesis)In vitro—LPS-stimulated murine RAW 264.7 cellsInhibition of COX-2-induced PGE production10.6 μΜ (IC50)Pretreatment of cells with pinosylvin before LPS stimulation2004[297]
Pinosylvin, dihydropinosylvinn/a (laboratory synthesis) and S. tuberosa (dihydrop.)In vitro—activated human neutrophilsInhibition of leukotriene biosynthesis ~ 50 μΜ (IC50)Incubation of cells with test compounds2005[81]
Pinosylvinn/a (pure compound)In vitro—human THP-1 monocytesInhibition of LPS-induced NF-κB activationVariousIncubation of cells with pinosylvin2006[306]
Pinosylvinn/a (pure compound)In vitro—LPS-stimulated murine RAW 264.7 cells(probable) TRIF-mediated signalling, iNOS and mRNA expression inhibition39.9 μΜ (IC50)Pretreatment of cells with pinosylvin before LPS stimulation2011[266]
Pinosylvin, monomethylpinosylvinP. sylvestrisIn vitro—murine J774 macrophagesDecreased iNOS expression and NO production, decreased NF-κB transcription13–15 μΜ, 8–12 μΜ (ΕC50)Addition in fresh culture medium post-cell growth (for 72 h)2015[76]
In vivo—male C57BL/6 miceReduction of paw oedema100 mg/kgAdministered via intraperitoneal injection once
PinosylvinH. dulcis ThunbIn vitro—RBL-2H3 basophilic leukaemia cell lineInhibition of released and/or expressions of inflammatory mediators5–20 μg/mLTreatment of cells with pinosylvin for 1 h2015[54]
Pinosylvinn/a (pure compound)In vitro—HEK293 (human embryonic kidney) cellsInhibition of TRPA1 activation0.1–100 μΜ (IC50 = 16.7–26.5 μΜ)Pre-incubation of cells with pinosylvin2016[315]
In vivo—male C57BL/6N miceReduction of IL-6 in inflamed tissue10 mg/kgIntraperitoneal injection (pinosylvin dissolved in 250 μL of phosphate buffered saline solution)
Pinosylvin monomethyletherC. cajanIn vitro—LPS-stimulated murine RAW 264.7 cellsActivation of PPARγ and inhibition of IL-6 activationVarious IC50 valuesIncubation of cells with solution containing the target compound2016[53]
Pinosylvinn/a (pure compound)In vitro—mouse 3T3-L1 preadipocyte fibroblastsDownregulation of PPARγ and C/EBPa116.8 ± 7.5 μΜ (ΕC50)Incubation of cells with pinosylvin2017[331]
Pinosylvin, monomethylpinosylvinn/a (laboratory synthesis)In vitro—murine J774 macrophagesInhibition of PI3K/Akt activation and of IL-6, NO, and MCP-1 expressionVarious 1Incubation of cells with pinosylvin2018[312]
In vivo—male C57BL/6 miceReduction in carrageenan-induced paw oedema via inhibition of IL-6 and MCP-130 mg/kg Intraperitoneal injection 1 h prior to inflammation induction
Pinosylvinn/a (pure compound)In vitro—human THP-1 monocytes and human U937 cellsPromotion of leucocyte apoptosis via upregulation of ALOX15 expression Various 1Treatment of cells with pinosylvin2018[320]
(Z)-pinosylvin mono methyl ether, (Z)-pinosylvin-3-O-b-D-glucosideA. flexuosaIn vitro—U937 human monocytesInhibition of histamine releaseVarious (less than the IC50 of ciprofloxacin) 1Incubation of cells with target compounds2020[56]
Pinosylvin, monomethyl pinosylvinn/a (pure compound)In vitro—murine J774 macrophagesDownregulation of classical M1 macrophage activation and upregulation of alternative M2 activation10, 30, 60 μΜAddition of target compounds in fresh culture medium after differentiation of monocytes to macrophages2021[319]
Pinosylvin, pinosylvin monomethyletherP. abies, P. sylvestrisIn vivo—Drosophila melanogaster1 (TrpA1)-dependent antagonism of NF-kB-mediated intestinal immune
responses
100 μΜ or 500 μΜ24 h feeding of larvae of indicated concentrations mixed with fly food2023[57]
PinosylvinP. nigra laricio var. calabricaIn vitro—LPS-stimulated murine RAW 264.7 cellsInhibition of TNFα and IL-6 expression, via inhibition of the JAK/STAT pathway40 μΜ (IC50 = 10.6 μΜ)Pretreatment of cells with target compounds2023[264]
1 depending on different derivatives, experimental protocols, or tested in increasing concentrations for different actions.

8. Anti-Cancer Properties of Pinosylvin

Stilbenes, stilbenoids, and their derivatives have a host of anticancer properties [332,333,334,335,336,337,338]. Studies have demonstrated the antiproliferative properties of pinosylvin in particular, via inhibition of protein uptake, suppression of Src/ERK and GSK-3/β-catenin signalling, gene, metabolic and signalling modifications [339,340,341]. The Src/ERK pathway is associated with glucose metabolism and the metabolic plasticity of cancer cells [342], while GSK-3/β-catenin signalling is involved in a number of diverse pathways [343]; their suppression leads to stoppage of cells in the G0/G1 phase, as shown by Park et al. on human colorectal HCT 116 cancer cells [340].
Mellanen et al. determined that there was an increase in oestrogen expression and proliferation in certain breast cancer cells, when exposed to pinosylvin; in general, the expression of oestrogens and their receptors is an important component in breast cancers [344,345]. Several pinosylvin derivatives have shown effectiveness against MCF-7 breast cancer cells with pinostilbene being among the highest effective compounds [346].
A number of experiments have demonstrated the antimetastatic potential of pinosylvin and its derivatives, which mostly occurs via matrix metalloproteinase inhibition and/or downregulation of their expression, both in vitro and in vivo [263,347,348]. The downregulation of the ERK1/2 signalling pathway observed by Chen et al. on SAS and SCC-9 oral cancer cells is also noteworthy, as this is an important pathway in cancer as well as in other pathologies [263,349]; a downregulation of Akt phosphorylation was noted in the same research, with crucial consequences on tumorigenesis [350]. In addition, vimentin is highly expressed in metastatic cancers and correlates with poor patient prognosis, cadherins and zonula occludens (ZO) are important in cellular adhesion [351,352,353]; as demonstrated by Chuang et al. [348], pinosylvin decreases the expression of all these proteins. Finally, Du et al. determined that migration, in squamous cell carcinoma cells, is inhibited by regulating of the STX6/ITGA3/VASP pathway, which has a demonstrated association with tumour development and migration [354,355].
Pinosylvin is also characterised by a notable cytotoxic potential, which however applies both for healthy and cancer cells, albeit healthy cells have a higher tolerance [356]; the cytotoxic potential of pinosylvin is exerted by different mechanisms, as presented by Song et al. on THP-1 and U937 leukaemia cell lines, notably a downregulation of AMP-activated protein kinase α1 (AMPKα1), a central regulator of energy homeostasis [357,358]. Finally, effects of a chemopreventive nature, relating to COX-2 inhibition and antioxidant properties have been reported by Park et al. [359]. An overview of experiments on the anti-tumoral properties is provided in Table 7.

9. Neuroprotective Properties of Pinosylvin

The neuroprotective properties of stilbenes are mostly based on their anti-inflammatory and antioxidant properties [52]; relevant research already exists for resveratrol [360,361,362]. Current evidence is in favour of stilbenes in cases of neurodegenerative diseases [363]. Xu et al. specifically targeted the neuroprotective properties of pinosylvin [364] (Table 8).
In stroke and cerebral ischaemia, the subsequent reperfusion, if successful, must be accompanied by some form of neuroprotection, to avert neural cell death [365,366,367]. Pinosylvin was demonstrated as capable of reducing LDH levels, decreasing TUNEL-positive cells (i.e., where DNA fragmentation occurred [368]), and downregulating cleaved caspase 3 levels in PC 12 cells found to enhance Bcl-2 expression and decrease Bax expression. The alteration of mitochondrial function is associated with the pathological effects of cerebral ischaemia and contributes to neuron death [369,370]. Finally, pinosylvin activated the Nrf2 pathway, thus reducing oxidative stress and, thereby, mitochondrial dysfunction. In vitro, pinosylvin administration in rats improved the brain deficit and reduced the infarct volume [364].

10. Traditional Medical Applications of Pinosylvin-Producing Plants

A wide variety of plant species from the Dipterocarpaceae, Cyperaceae, Gnetaceae, Pinaceae, Leguminosae, Vitaceae families, and others produce stilbenes [371]. Such plants and their products have been included in the diets of numerous populations since ancient times and used as sources of drugs or as medicines themselves prominently at least until the 16th century in most cultures [372].
For pine trees in particular, pine nuts have been discovered in various cases in Nerja (Málaga) and Lattes in southern France, where the sites are dated to the Mesolithic period, indicating the importance of pine nuts in the diets of early humans [373]. The taste of pine nuts was mentioned by both the Ancient Greeks and Romans, and Roman legions carried pine nuts among their provisions [374]. We must note here that apart from plant parts and extracts, drinks and foods containing stilbenoids have long been used in folk medicine. For example, in Greece of the early 20th century, Malaka wine (from Malaga, Spain) was used as the basis of a medicinal elixir [375]; stilbenes, stilbenoids, and other phenols are detected in most wines [376,377]. Grapes have also been consumed since antiquity and are noted as having numerous health benefits [378]. Peanuts, another source of stilbenoids, and other bioactive compounds are considered an excellent functional food [147,379].
Stilbenes are included in many preparations of traditional medicine systems, such as Ayurveda and Japanese traditional medicine [380]; pinosylvin-producing plants have a prominent role in the pharmacopoeias and traditional medical systems of many cultures. In this section, we will provide a representative selection of the traditional uses of pinosylvin-producing plants, organised by region, and cross-reference the recorded uses against experimentally proven effects (Table 9).

10.1. Traditional Uses in Europe

In general, Greece, as a region, is characterised by an astonishing variety of plant diversity, and particularly of endemic plant species [413,414]. Accordingly, the most important doctors of Ancient Greek medical tradition, Hippocrates, Theophrastus, and Dioscorides, all focused on plants as the basis of their therapies [415,416,417,418,419,420].
Hippocrates refers to pine as an ingredient in an ointment meant to help with the healing of burns and possibly with lessening the post-burn scar [421]. The bark of pine trees was also mentioned by Dioscorides as being a treatment for burn wounds [421]—the antiseptic alkaloids found in pine bark are useful in that regard as confirmed by modern research [422,423]; pine nuts are also mentioned in the context of relieving chest infections [424]. Earlier references, from an Ancient Egyptian medical text, also mention pine nuts as a drug [425]. Galen, the most important physician of the Imperial Roman era [426]—his medical training was based on Ancient Greek teachings—also mentions the use of pine as an expectorant [424]. A similar action is described much later by Abu Ali al-Husayn ibn Sina, a Persian polymath of the 10th-11th centuries A.D., in his book Canon of Medicine [424]. Remarkably, it was also described by a rather more obscure Ancient Greek physician, Metrodora, in her book, On Women’s Diseases.
Moreover, parts of the pine tree had other uses, linked to religion and magic; for instance, Maenads, the female followers of the god Dionysus, would use a decoction containing pine and ivy fruits, along with other ingredients, to reach an altered mental state [427,428,429]. Such is the importance of the pine tree in Ancient Greek culture that pine cones have been found depicted in Ancient Greek coinage [429,430,431], and a pine wreath was awarded to the winner of the Isthmian games [432], an important ancient agonistic festival [433]. Pine nuts are also used in some local Greek culinary traditions and for wine storage [429].
In Italy, Pinus halepensis Mill. is found around the Mediterranean [434]; it is an evergreen tree which is found in arid and semi-arid climates at low altitudes [435]. It must be noted that the production of bioactive compounds of this plant, and hence its medicinal properties, depend upon the particular regional climate conditions [68]. The leaves of the plant are used in the treatment of various respiratory pathologies [436,437,438]; the resin of the buds has been used to aid in wound healing [439]. Based on the current evidence, the plant possesses a notable antioxidant activity [440,441,442,443,444,445,446,447,448], which may be associated with its wound-healing properties. The anti-inflammatory properties of the plant have also been investigated [446,449,450,451,452].
In another region of the country, namely Valfurva, the extract from Pinus mugo Turra is said to be used as an expectorant, although this property has not been verified using modern methods [405]. This specific Pinus species, which also grows in some other European countries like Romania, has been evaluated for its antifungal and antioxidant activities, which are promising according to the research of [453], although to the knowledge of the authors, no study has yet evaluated its pinosylvin content. Given the high pinosylvin content of other Pinus species, it seems reasonable that such content must be commensurate to that of the others.
Along with the Romans, the Dacians are part of the national identity of modern Romania [454]; Dacian medicinal knowledge was well-known and respected in the ancient world, and, indeed, many of the phytochemical traditions of these ancient people have carried over to traditional Romanian practices [455,456,457,458,459]. It seems that based on current knowledge, pinosylvin-producing plants were not prominent in their ethnopharmacological tradition.
However, in the regions of Romania formerly occupied by the Habsburg Empire, a number of pharmaceutical products derived from pine trees are mentioned [403]. Especially in Transylvania, plant-based pharmaceutical products are quite common in the medical tradition of the region [460,461,462]. Indeed, there is ample ethnobotanical research on the traditions and effectiveness of such remedies [403,463,464,465]. Both Pinus nigra and Pinus sylvestris are identified by a variety of terms by those inhabiting the regions [466,467,468,469]. Pinus nigra is used in the treatment of colds, cough and other respiratory complaints, furuncles, and warts [464,465,470,471,472,473,474]—the soot, needles or cones of the plant may be used depending on the occasion. The resin is also used to treat teeth decay, or, alternatively, for teeth cleaning [472,473,475]. Some reports also mention the use of parts of the plant for digestive complaints, [476], while a ritual use to ward off evil is mentioned in some Serbian regions [477].
The soot of P. sylvestris is used to treat asthma, coughs, and respiratory diseases [478], along with rheumatism [471,479]. The bark of the plant has also been mentioned as effective in the case of varicose veins [480]. Pine species were also used in ethnoveterinary practices [471].
Finally, in Spain, the inflorescence of P. halepensis Mill is used to treat asthma while leaf decoctions and infusions are used to treat colds; other parts of the plant are used to treat pain and baldness [481]. In other parts of Spain, various parts of the plant are used for a number of remedies including warts, the characteristic lesions caused by the human papilloma virus (HPV) [482,483]. Other uses of Aleppo pine’s parts and gum include expectorant and anti-abscess applications [484]. In the Balearic islands, the buds of the plant are used to treat bronchitis [485].

10.2. Traditional Uses in Africa

In Algeria, parts of P. halepensis Mill are used as a remedy for gastrointestinal system pathologies and as a disinfectant and antifungal agent [486,487]. Recent research has confirmed the antibacterial and antifungal [445,448,488,489,490,491,492,493,494,495,496,497,498,499] activities of the plant. In association with the treatment of the aforementioned gastrointestinal system pathologies, the cytoprotective properties of P. halepensis Mill extracts have been confirmed for hepatic and renal cells [500,501]. Compounds found in the plant have also been shown to be able to protect cellular DNA from damage and associated mutations [68,447].
A fruit infusion of the plant is used to treat haemorrhoids, ulcers, pulmonary pathologies, and tuberculosis [502]. In other regions of Algeria, parts of the plant are used for similar purposes, and also for renal inflammations [503,504,505]. A notable use is against delusional parasitosis, a mental condition, and as an adrenal gland stimulant [506,507,508].
In Morocco, the bark of P. halepensis Mill is widely used as a poultice to treat injuries, scars, and infections, and as an astringent [509,510,511,512]. The resin of the plant, along with its bark, is used in SW Morocco to treat pathologies of the digestive, integumentary, circulatory, and urogenital system [513]. Regarding circulatory system pathologies, extracts of the plant have a proven anti-coagulant and anti-haemolytic activity [442,446,447,514,515]. Although no anticancer use is specifically mentioned in medical traditions, it may be assumed that some of pathologies of the aforementioned systems may be associated with neoplasias; plant extracts have been found to possess a cytotoxic activity against human glioblastoma cell lines [516], human myeloma, and adenocarcinoma cells [517].
Notably, a bark decoction is used in NW Morocco to treat tuberculosis [518]. Infusions from the leaves of this plant are used in different parts of Morocco to treat infections and eczema [519]. A leaf decoction is also used against toothache [520,521]. Finally, in Nigeria, extracts of A. hypogaea are used as antidiabetic and anti-cholesterol medications, in cardiovascular pathologies, to promote weight loss, and even in cases of cancer [522,523] by the locals.

10.3. Traditional Uses in Asia

Traditional Chinese medicine (TCM) began developing in Ancient China, although at the moment, the exact timing of the formulation of its first theories is not entirely clear [524,525]. Regardless, even since the beginnings of these practices, medicinal plants had a prominent role [372,526].
Arachis hypogaea (peanut) is an important part of TCM; although initially believed to have been domesticated in Bolivia and Argentina and cultivated by the Incas, it was subsequently transported to Spain and Europe and then to Asia [527,528]. It is now considered to be amongst the most-cultivated crops worldwide. Peanut seed oil contains numerous phospholipids of medical significance and other bioactive compounds [529]. Stilbenes have been identified in different organs of the plant [381]. Stem and leaf extracts of the plant are used to treat sleep disorders, and it has been proven that such extracts can improve sleep behaviour in phenobarbital-treated rats [382,530,531,532]. The skin of the peanut is also used to treat haemorrhage and bronchitis [533]; the extract of the coat of the seed has been shown to have an antibacterial potential [383].
Parts of H. dulcis are used in China, as well as in Japan and Korea (discussed later on), as nutraceuticals and supplements [389]. A wealth of different research evidence, from animal experiments, suggests that the extracts of the plant have an anti-inflammatory, analgesic, and anti-allergic activity, use as a laxative, and a potential anti-osteoporotic effect [534,535,536,537,538,539,540,541,542]; it also seems to promote lipid metabolism [543,544,545]. The leaves of another plant, Cajanus cajan, are used as an analgesic to stem the flow of blood and to treat parasitic infections [387].
Korea has a rich medical tradition, which may be traced back to the medical systems of China and Japan [546]; for at least seven centuries, Korean traditional medicine (KTM) has existed as a distinct medical tradition, with its own therapeutical approaches and methods [547]. KTM is well-known for the use of herbs and plants as therapeutical agents, alone or in combinations and different modes of preparation [548,549,550].
Extracts of Hovenia dulcis have long been used for alcohol detoxification, and based on research evidence, it is indeed a property of extracts of the plant to lower blood alcohol concentrations [551,552,553,554,555,556,557]. It is believed that the extracts of the plant indirectly upregulate alcohol dehydrogenase and acetaldehyde dehydrogenase, the two enzymes involved in the alcohol metabolism pathway. On a related note, it has been proven that extracts of the plant have a hepatoprotective action, in chemically induced liver damage [555,558,559,560]. Given that free radicals are known to be a main cause of hepatic injury [561,562], the antioxidant capacity of the plant and of pinosylvin and its derivatives, along with other phytochemicals, in particular, appears to be in play here; in two experiments, glutathione-S-transferase activity was also upregulated [552,554]. Although some extracts of this plant have a demonstrated antidiabetic activity, this does not seem to be related to their pinosylvin content [388]. Pinus densiflora Sieb. et Zucc. has been widely used in the treatment of hypertension, atherosclerosis, stroke, diabetes, cancer, and balding [400].
In Pakistan, P. roxburgii Sargent is found in areas where the monsoons do not penetrate. Its resin is used as a stimulant, and it is also applied externally in cases of suppuration and swollen lymph nodes. Both the wood and the oleoresin of the plant are used to treat snake and scorpion bites; the oil of the plant is also used to cure gastrointestinal complaints and is considered a prokinetic [407,563].
On the Asiatic side of Turkey, the Turkish red pine (P. brutia Ten.) is found mostly in coastal areas, is mostly exploited for pine resin production [564], and, in many cases, for turpentine production [565,566]. In the region of the Kazdağ mountain, in Western Turkey, the resin of the plant, alone, or in combination with honey, is used to cure gastrointestinal complaints, as an anti-cough medication, and also to treat diabetes [395]. In Turkish folk medicine, other uses of the plant have been described, namely as a treatment of haemorrhoids and as a tonic [395]. A combination of resin and hot water is also applied externally for wound treatment [395].
The bark extract of P. brutia has been tested in combination with Pycnogenol®, and when applied on cotton fabrics, has a proven antibacterial activity against Aspergillus brasiliensis, a member of the black aspergilli section [567], and also promotes wound closure. This result indicates a promising research avenue in the development of eco-friendly natural antifungal finishes for medical textiles [397]. The antibacterial action of the extract in vitro, against Staphylococcus aureus, Bacillus cereus, Listeria monocytogenes, Salmonella Typhimurium, and Escherichia coli, was verified by the research of Erol et al. [399], who also noticed an interesting antiproliferative potential against certain cancer cell lines. Interestingly, the tar of the roots of the plant also possesses an antimicrobial potential [568].
Moreover, the cone extract of the plant has demonstrated a potential for preventing acute lung injury in certain concentrations [398]; the same combination has exhibited a potent anti-inflammatory potential in rat models [394]. The antioxidant potential of the extract of the plant has already been noted by Cretu et al. [396]. Finally, the extract of P. brutia along with that of other plants from the same family is of interest to the cosmetics industry [569].
In the Himalayas, P. roxhburghii Sargent, a native plant, contains pinosylvin and pinosylvin monomethyl ether in its bark, stem, and needle extracts [570,571]; it has various uses in the local medical systems, as well as in Ayurveda [75]. Extracts from various parts of the plant have demonstrated antibacterial activity against Agrobacterium tumefaciens, E. coli, Salmonella arizonae, S. typhi, B. subtilis, and other pathogens [572,573,574]; bark and needle extracts were also found to have notable antioxidant activity, analgesic activity, anticonvulsant activity, and anti-asthmatic activity [75,409,575,576,577]. Some marketed preparations of the plant are also available [578,579,580]. Wood oil from the plant also has a hepatoprotective activity, although pinosylvin has not been detected in it [581]. Finally, the plant also has some cultural uses [409]. Another widely used plant is C. cajan, especially for pathologies of the oral cavity, as a laxative [582], to treat intoxications, and also to induce lactation [583]. Other uses of the plant have been recorded in regions of Bangladesh and Trinidad and Tobacco [387].

10.4. Traditional Uses in North America

The regions of North America, in what is now Canada and USA, have had a long history of habitation before the arrival of European settlers [584]. In accordance with the norms observed in most other ancient civilisations, Native Americans relied mostly on plants and their extracts for treating the majority of ailments and pathologies [412].
In the British Columbia region of Canada, Picea glauca Voss is used for a variety of reasons. It is burned so that the smoke can clear the air of infectious agents; it is mixed with bear grease and applied on wounds as a healing ointment; in decoction form, it is used for sore mouths or it is mixed with fat and swallowed in cases of haemoptysis; finally, young buds of it are chewed in cases of strep throat and it is applied as a dab in the nostrils in cases of sinusitis [390]. The Carrier people are Native Americans, which have a long medicinal tradition, having identified and included in their medical practices a great variety of plants, of which they frequently use all parts [585,586,587,588].
Apart from the Carrier people, there are other indigenous people in the Canadian boreal forest, who use P. glauca extracts and ointments to treat chronic pain of various causes [393,589]. The pitch preparations of P. glauca have a proven antibacterial activity against E. coli, S. aureus, P. aeruginosa, C. albicans, and A. fumigatus [391]. Moreover, it seems that the extracts of different organs of the plant can be used to protect organs from glucose toxicity or deprivation [590].
Another tribe of Native Americans, the Cherokee, have an important ethnomedical and ethnobotanical tradition [591,592,593,594]. P. virginiana Miller grows from Pennsylvania to Alabama [595,596] and was widely used by the Cherokee Native Americans to produce a type of wash for sores and skin ulcers, with the sap of the plant being employed in wounds refusing to heal. The oil of the plant was used to bathe painful joints and a tea from the needles was used in cases of colds and fevers [412]. Based on the current evidence, although notable bioactive compounds are found in parts and extracts of the plant, there is no appreciable antimicrobial activity [412].

11. Discussion

All pine trees have been found to contain a very high amount of polyphenols and are, in general, high in minerals and nutritional value [597,598,599,600,601,602,603]. Of course, pinosylvin and its derivatives are present in other plant species, in lower quantities, as evidenced by the successful small-scale extractions of various researchers. Novel solutions for artificial pinosylvin synthesis are available [99,106,108,110] which may prove more feasible and cost-effective on a larger scale.

11.1. Health-Related Properties of Pinosylvin and Future Research Perspectives

Based on the current research evidence, the antibacterial, antifungal, antiparasitic, and antiviral properties of pinosylvin and its derivatives are notable in in vitro settings [41,71,140,261]. Given the potential for complications of certain bacterial infections like tuberculosis [604,605], antimicrobial applications represent an important aspect of phytochemical research. This is important both in the context of emerging bacterial and fungal resistance to antibiotics, the prevalence of parasitic infections, and the increasing risk of viral pandemics [606,607,608,609,610,611,612]; the septic potential and the significance of sepsis in clinical settings must also not be ignored [613,614,615,616]. Moreover, considering the possibility of adverse effects, in antibacterial, antifungal, and antiviral, and especially antiparasitic drugs, pinosylvin formulations should be developed as adjuvant antimicrobial treatments to lower the dose or length of drug administration, if and when needed [134,617,618,619,620,621,622,623,624,625].
The antioxidant properties of pinosylvin in scavenging free radicals [69,267,271] are well demonstrated. Apart from that, the antioxidant effects can be exerted via other mechanisms and pathways [268,282]. The antioxidant properties of pinosylvin and its derivatives are also related to its anti-inflammatory and anticancer properties [263,269,270]. Considering the importance of oxidative stress in health and longevity, and its relevance in the pathophysiology of different diseases [626,627,628,629,630,631], relevant uses of pinosylvin should be explored; additionally, the use of pinosylvin as a supportive treatment in heavy metal poisoning, given that one of the mechanisms of metal toxicity is oxidative stress induction, could demonstrate its utility [632,633].
Regarding its anti-inflammatory potential, pinosylvin is both a direct and an indirect inhibitor of COX-2 [297,306]; it also inhibits leukotriene biosynthesis directly [81]. Other anti-inflammatory mechanisms of pinosylvin include IL-6 and MCP-1 inhibition [312], inhibition of the JAK/STAT pathway [264], and PPARγ expression upregulation [319] (TrpA1)-dependent antagonism [57]. A couple of research studies [56,535] have commented specifically on the anti-allergic potential of pinosylvin and its derivatives and have demonstrated an anti-inflammatory effect in the intestine of D. melanogaster. While such an effect has yet to be translated into applications in humans, it could be a promising therapeutical application for patients with chronic intestinal inflammation, such as inflammatory bowel syndrome patients [634,635,636].
Regarding the anticancer properties of pinosylvin and its derivatives, there is a noted and quite important antimetastatic potential [347]. The related anticancer effects of pinosylvin are mediated by a variety of mechanisms [348,354]. The cytotoxicity of pinosylvin is another potential anticancer action although the toxic effect is relatively important for healthy cells too [356]. Chemopreventive effects were also noted by Park et al. [359]. Direct antiproliferative properties are exerted by a number of mechanisms [339,341]. With the exception of the in vivo arm of the experiment of Park et al. [347], all other experiments mentioned in the text were performed in vitro. Given that some traditional medical uses of pinosylvin are mentioned for ocular pathologies, perhaps pinosylvin may offer a viable therapeutical option in cases of ocular cancers, such as uveal melanoma [637,638,639]. Therefore, while considering the promising anticancer potential of pinosylvin and its derivatives, the distribution, bioavailability, and effective concentrations at target cites remain to be solved. Considering the multifactorial nature of cancer pathogenesis and development [640,641,642,643,644,645,646,647,648,649,650], the interplay between pinosylvin and different molecular mechanisms should be further explored.
On the subject of distribution and bioavailability, we should mention that current research efforts on novel delivery systems may go a long way towards simplifying administration-related complications. In recent years, liposomes, micelles, micro-emulsions and nano-emulsions, colloidal capsules, and solid nanoparticles have been developed, and these may be used as carriers for pinosylvin and its derivatives [651,652,653,654]. The introduction of pinosylvin in 3D-printed biomaterials is another option, considering the rapid developments in the field of orthopaedic surgery, and the associated risk of infections [203,655,656,657,658,659,660,661,662,663,664,665,666]. Such research is all the more important considering also the advances in imaging techniques [667,668,669,670,671,672,673] which allow for earlier diagnoses and more complex surgeries. As of yet, there do not exist detailed studies examining the toxicology of pinosylvin, in a manner similar to other phytochemicals, such as capsaicin [674]. Few researchers have examined the toxic doses of pinosylvin at a cellular level, with the toxicity limit being above the effective limit; the relevant research results are summarised by Bakrim et al. [41].
Apart from the heretofore presented properties of pinosylvin, other health-promoting effects have been noted. For example, it was found that pinosylvin can inhibit adipogenesis in murine adipocytes [321]; considering the deleterious health effects of obesity (e.g., [675,676,677,678]), further research on the anti-adipogenic effects of pinosylvin should be undertaken. A number of studies point towards the antidiabetic effects of stilbenes [52]; pinosylvin has been proven capable of reducing insulin resistance in rat skeletal muscles [331], and this is a promising research avenue for future in vitro research in human cells. It is not clear yet if pinosylvin can have a meaningful antidiabetic effect, but considering the diabetes-associated complications [679,680,681,682], and the existing evidence on the antidiabetic effect of certain stilbenes [683,684,685], future research in that direction seems to hold some potential.
Currently, no specific cardioprotective effects of pinosylvin have been identified. This is in contrast to other stilbenes and their analogues [686,687,688]. For example, resveratrol is known to exert cardioprotective effects by modulating cholesterol levels, impacting cardiovascular risk [689,690,691,692,693]; pinosylvin, which has similar properties, is very likely to have commensurate effects. Additionally, it has been demonstrated that it prevents necrosis in bovine aortic endothelial cells at sufficiently high concentrations [694].
As mentioned, parts of P. roxhburghii Sargent have hepatoprotective properties [581]; this might be partly attributed to the presence of pinosylvin. Already a number of phytochemical compounds with hepatoprotective action, including stilbenes, have been identified [695,696]. Pinosylvin, its analogues, and derivatives, if proven useful in that regard, could be administered in cases where liver damage is probable or existing, and as an adjunct therapy during therapy for hepatocellular carcinoma, a prominent form of hepatic cancer [697,698,699,700,701,702,703,704].
Another interesting research avenue would be the comparative effectives of the different pinosylvin stereoisomers. For instance, trans-resveratrol has been found to have better anticancer properties compared to cis-resveratrol [705] and trans-stilbenes, in general, exhibit a better inhibitory action against COX [706].
A number of stilbene-based drugs, such as raloxifene and tamoxifen, are currently available [707]; in fact, the stilbene core is a very versatile structure with very diverse and promising potential uses [708]. Further development along this axis may prove successful and provide alternatives to existing solutions or new avenues for pharmacotherapy.
Regarding the phytochemical uses, there is a wealth of ethnomedical and ethnobotanical evidence on the uses of pinosylvin-producing plants; many of these uses are verified by recent research efforts. Apart from the medical traditions directly mentioned heretofore, there are various other local medical systems from ancient people, such as that of the Incas of Peru [709,710,711], the Aztecs of Central America [712,713], and the aborigines of Oceania [714,715]. In all these medical traditions, along with those already mentioned, from Asia, Europe, North America, and Africa, there are a host of foodstuffs used as medicines [716,717,718,719,720]; we propose that concerted research on the constituents of these foodstuffs be performed so that their stilbenoid content in particular be explored and relevant applications are experimentally tested.
An overview of the most relevant and studied medical applications of pinosylvin and its derivatives is depicted in Figure 3.

11.2. Non-Medical Uses of Pinosylvin and Its Derivatives

It has also been reported that the bark of P. roxburghii has been tested for use in the removal of toxic waste from the water. Research results were encouraging at least in the case of Cr(VI) [579]. Pine bark has also been successfully tested in a similar capacity, with the addition of microalgae cultivated on it [721]; the use of pine bark instead of activated carbon is another possibility for pesticide removal [722]. Indeed, parts of plants have been explored as solutions in wastewater treatment and management systems in the last decades [723,724,725]. Heavy metal pollution is attributable both to mining in different sites and locations and industrial processes [650,726,727,728,729,730,731,732,733,734,735,736,737,738]; on the other hand, pesticide-associated pollution is an important issue both from historical and current uses [739,740,741,742,743]. Pesticides are a potential source of severe intoxication [744,745,746]. Currently, there are a number of initiatives and research regarding both the remediation of mining cites and the mitigation of pesticide pollution [747,748,749,750,751,752,753,754,755,756,757,758]. Perhaps, for such purposes, pinosylvin can be combined with or added to novel recovery mediums such as the ones proposed by a number of researchers [759,760,761].
It is important to mention at this point that parts of P. halepensis Mill possess a notable herbicidal activity [495,496,762,763,764]. Evidence suggests that the essential oils of the needles of this plant possess a larvicidal activity against the larvae of the Aedes albopictus mosquito [765]. On the other hand, up to a certain concentration, there were no discernible effects against the larvae of Drosophila melanogaster [493]. Moreover, Stemona collinsiae has also been used as an insecticide by the local people in Thailand—the ethanolic extract of the plant has been proven capable of eliminating the larval and adult stages of P. ruficornis [766] and the larvae of Musca domestica and Chrysomya megacephala [767].
A summary of the above-mentioned applications is available in Table 10.
These properties of P. halepensis Mill and Stemona collinsiae may prove important in the context of relevant policies of the European Union, which has developed and implemented a comprehensive policy and legal framework to regulate and promote the development and use of phytomedicines, plant-based substances, and natural products. First and foremost, it is widely known that the EU supports the use of plant-based substances as alternatives to synthetic chemical pesticides and herbicides, in an effort to reduce dependence on the latter [768,769]. Directive 2009/128/EC sets the ground rules for sustainable pesticide use, aiming to reduce health and environmental risks while promoting integrated pest management and alternative non-chemical methods [770]; the regulation (EC) No 1107/2009 lays down the main instruments for placing effective plant protection products (using pesticide substances) on the market that are safe for humans, animals, and the environment, while at the same time ensuring effective functioning of the internal market and improved agricultural production [771]. Meanwhile, the Horizon Europe framework [772] funds research into sustainable agriculture, including innovations in plant-based pesticides and herbicides. Therefore, it also aligns with the EU’s Farm to Fork strategy [773,774] under the European Green Deal [775] which aims to reduce pesticide and fertiliser use by 2030.
Moreover, there is phytoremediation which refers to the utilization of a plant-based substance for the removal or neutralization of contaminants like heavy metals in soil or wastewater [776,777,778]. The EU in the context of its broader sustainability goals, either by aligning with the United Nations (UN) 2030 Agenda which includes the 17 Sustainable Development Goals (SDGs) or by having sustainable development as its core principle (Art.3.3.) of the Treaty on European Union (TEU) [779,780] as well as implementing through strategies such as the European Green Deal, delineates and fosters phytoremediation directly and indirectly under the Waste Framework Directive (2008/98/EC) and the Water Framework Directive (2000/60/EC) [780,781,782]. Additionally, among the other EU policies and frameworks, there is the Urban Waste Water Treatment Directive (91/271/EEC) and its consolidated 2014 version [783,784,785] which promote the treatment of urban wastewater and encourage the exploration of innovative technologies, including plant-based and natural filtration methods.
Finally, the EU’s regulatory framework research and development programs have a significant focus on fostering innovation in plant-based substances offering at the same time funding opportunities through Horizon Europe, the largest research and innovation programme supporting projects in sustainable agriculture, biodiversity, and green technologies, as well as the LIFE Programme [772,786,787], the only EU funding initiative entirely dedicated to environmental, climate, and energy objectives and projects. Those initiatives and tools can easily support any aspiring phytoremediation and natural plant-based technologies/projects including but not limited to those with a pollution control orientation.
In the USA, biopesticides are regulated by the Environmental Protection Agency (EPA) under the Federal Insecticide, Fungicide, and Rodenticide Act (FIFRA) [788], which regulates specifically the distribution, sale, and use of pesticides while streamlining registration for biopesticides over chemical ones. Supplementarily, the Food Quality Protection Act (FQPA) is applicable and requires that all pesticides meet specific health standards, with an emphasis on sustainable use, while phytoremediation is guided by the National Environmental Policy Act | US EPA (NEPA), a US environmental law designed to promote the enhancement of the environment, and the Toxic Substances Control Act (TSCA) under EPA oversight [789,790,791].
In Japan, the Agricultural Chemicals Control Act [792] regulates all pesticides, including biopesticides, with a primary focus on safety. In the meantime, the Act on Promotion of Organic Agriculture [793] promotes sustainable use through reduced chemical inputs and supports integrated pest management (IPM) practices, in line with its environmental goals. Finally, the Environmental Conservation Law [794] enables phytoremediation projects, with a focus on urban and industrial applications under the oversight of the Japanese Ministry of the Environment.
In South Korea, the Framework Act on Environmental Policy promotes phytoremediation for soil health, aligning with its respective national environmental priorities which include but are not limited to sustainable development, low carbon, green growth, and environmental protection [795,796]. On another note, under the Agrochemicals Control Act [797], biopesticides must meet stringent safety standards. In addition, there is also the Sustainable Agriculture Promotion Act [798] that encourages reduced pesticide reliance, and supports organic farming initiatives, promoting IPM as part of sustainable agriculture practices.
Conclusively, each country’s regulatory framework is adjusted to unique national plans and policies, supporting overall reduced pesticide use and sustainable agricultural practices such as the introduction of safe biopesticides under certain conditions and fulfilling specific minimum requirements. Each country also seems to support phytoremediation either directly through funding or indirectly through its regulatory regime, yet we always need to consider the varying cultural and market priorities when comparing the state of play in multiple and diverse jurisdictions.

12. Conclusions

Pinosylvin and its derivatives represent a potent class of bioactive compounds which can be employed as antimicrobial and potentially as antiviral agents. At the same time, the antioxidant, anti-inflammatory, and anticancer actions of said compounds can be used to improve the actions of existing drugs and perhaps improve the prognoses in certain cases or reduce drug doses and associated side-effects. Questions of bioavailability can be addressed via the use of novel delivery systems. The multifaceted uses and applications of pinosylvin even comprise land remediation. Thus, it can be a potential contributor towards more environmentally friendly policies for insecticide and pesticide use, in accordance with global trends and European Union directives.

Author Contributions

Conceptualization, A.P., S.G.-G., S.D., I.A.B., C.C., and C.S.; methodology, A.P., A.-T.P., L.T., C.D.M.D., and S.D.; validation, A.P., A.-T.P., L.T., K.P., S.G.-G., G.T., I.S.-F., S.D., and A.-E.S.; investigation, A.P., A.-T.P., L.T., K.P., S.G.-G., G.T., A.C., and A.-E.S.; resources, A.P., A.-T.P., L.T., K.P., C.D.M.D., and A.-E.S.; data curation, A.P., A.-T.P., K.P., G.T., C.D.M.D., A.C., S.D., I.A.B., and C.S.; formal analysis, A.P., A.-T.P., S.G.-G., G.T., C.D.M.D., A.C., I.S.-F., S.D., A.-E.S., C.C., and C.S.; software, A.P. and A.-T.P.; writing—original draft preparation, A.P., A.-T.P., L.T., K.P., S.G.-G., G.T., C.D.M.D., A.C., I.S.-F., S.D., A.-E.S., I.A.B., C.C., and C.S.; writing—review and editing, A.P., A.C., I.A.B., C.C., and C.S.; visualization, A.P. and A.-T.P.; supervision, A.C., I.A.B., C.C., and C.S.; project administration, C.C. and C.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Kaitin, K.I.; DiMasi, J.A. Pharmaceutical innovation in the 21st century: New drug approvals in the first decade, 2000–2009. Clin. Pharmacol. Ther. 2011, 89, 183–188. [Google Scholar] [CrossRef]
  2. De la Torre, B.G.; Albericio, F. The pharmaceutical industry in 2023: An analysis of FDA drug approvals from the perspective of molecules. Molecules 2024, 29, 585. [Google Scholar] [CrossRef] [PubMed]
  3. Kumar, S. Innovative Drug Discovery Research by Pharmaceutical Companies in India and China; ACS Publications: Washington, DC, USA, 2024. [Google Scholar]
  4. Butt, J.; Barthel, J.; Hosokawa, M.; Moore, R. NSAIDs: A clinical approach to the problems of gastrointestinal side-effects. Aliment. Pharmacol. Ther. 1988, 2, 121–129. [Google Scholar] [CrossRef]
  5. Cunha, B.A. Antibiotic side effects. Med. Clin. N. Am. 2001, 85, 149–185. [Google Scholar] [CrossRef]
  6. Mackin, P. Cardiac side effects of psychiatric drugs. Hum. Psychopharmacol. Clin. Exp. 2008, 23, S3–S14. [Google Scholar] [CrossRef]
  7. Sostres, C.; Gargallo, C.J.; Arroyo, M.T.; Lanas, A. Adverse effects of non-steroidal anti-inflammatory drugs (NSAIDs, aspirin and coxibs) on upper gastrointestinal tract. Best Pract. Res. Clin. Gastroenterol. 2010, 24, 121–132. [Google Scholar] [CrossRef]
  8. Richardson, W.L.; Hammert, W.C. Adverse effects of common oral antibiotics. J. Hand Surg. 2014, 39, 989–991. [Google Scholar] [CrossRef]
  9. Rousan, T.A.; Mathew, S.T.; Thadani, U. The risk of cardiovascular side effects with anti-anginal drugs. Expert Opin. Drug Saf. 2016, 15, 1609–1623. [Google Scholar] [CrossRef]
  10. Bețiu, A.M.; Noveanu, L.; Hâncu, I.M.; Lascu, A.; Petrescu, L.; Maack, C.; Elmér, E.; Muntean, D.M. Mitochondrial effects of common cardiovascular medications: The good, the bad and the mixed. Int. J. Mol. Sci. 2022, 23, 13653. [Google Scholar] [CrossRef]
  11. McManus, M.C. Mechanisms of bacterial resistance to antimicrobial agents. Am. J. Health-Syst. Pharm. 1997, 54, 1420–1433. [Google Scholar] [CrossRef]
  12. Tenover, F.C. Development and spread of bacterial resistance to antimicrobial agents: An overview. Clin. Infect. Dis. 2001, 33, S108–S115. [Google Scholar] [CrossRef]
  13. Van Duijkeren, E.; Schink, A.K.; Roberts, M.C.; Wang, Y.; Schwarz, S. Mechanisms of bacterial resistance to antimicrobial agents. In Antimicrobial Resistance in Bacteria from Livestock and Companion Animals; John Wiley & Sons, Inc.: Hoboken, NJ, USA, 2018; pp. 51–82. [Google Scholar]
  14. Varela, M.F.; Stephen, J.; Lekshmi, M.; Ojha, M.; Wenzel, N.; Sanford, L.M.; Hernandez, A.J.; Parvathi, A.; Kumar, S.H. Bacterial resistance to antimicrobial agents. Antibiotics 2021, 10, 593. [Google Scholar] [CrossRef]
  15. Verma, A.R.; Vijayakumar, M.; Mathela, C.S.; Rao, C.V. In vitro and in vivo antioxidant properties of different fractions of Moringa oleifera leaves. Food Chem. Toxicol. 2009, 47, 2196–2201. [Google Scholar] [CrossRef]
  16. Gilca, M.; Gaman, L.; Panait, E.; Stoian, I.; Atanasiu, V. Chelidonium majus—An integrative review: Traditional knowledge versus modern findings. Forsch. Komplementmed. 2010, 17, 241–248. [Google Scholar] [CrossRef]
  17. Singh, N.; Bhalla, M.; de Jager, P.; Gilca, M. An overview on ashwagandha: A Rasayana (rejuvenator) of Ayurveda. Afr. J. Tradit. Complement. Altern. Med. 2011, 8. [Google Scholar] [CrossRef]
  18. Singh, N.; Pandey, B.; Verma, P.; Bhalla, M.; Gilca, M. Phyto-pharmacotherapeutics of Cyperus rotundus Linn. (Motha): An overview. IJNPR 2012, 3, 467–476. [Google Scholar]
  19. Petran, M.; Dragos, D.; Gilca, M. Historical ethnobotanical review of medicinal plants used to treat children diseases in Romania (1860s–1970s). J. Ethnobiol. Ethnomed. 2020, 16, 15. [Google Scholar] [CrossRef]
  20. Veeresham, C. Natural products derived from plants as a source of drugs. J. Adv. Pharm. Technol. Res. 2012, 3, 200–201. [Google Scholar] [CrossRef]
  21. Katiyar, C.; Gupta, A.; Kanjilal, S.; Katiyar, S. Drug discovery from plant sources: An integrated approach. AYU (Int. Q. J. Res. Ayurveda) 2012, 33, 10–19. [Google Scholar] [CrossRef]
  22. Mandal, S.C.; Mandal, V.; Konishi, T. Natural Products and Drug Discovery: An Integrated Approach; Elsevier: Amsterdam, The Netherlands, 2018. [Google Scholar]
  23. Seidel, V. Plant-Derived Chemicals: A Source of Inspiration for New Drugs. Plants 2020, 9, 1562. [Google Scholar] [CrossRef]
  24. Tesso, H.; König, W.A.; Kubeczka, K.H.; Bartnik, M.; Glowniak, K. Secondary metabolites of Peucedanum tauricum fruits. Phytochemistry 2005, 66, 707–713. [Google Scholar] [CrossRef]
  25. Turski, M.P.; Turska, M.; Zgrajka, W.; Bartnik, M.; Kocki, T.; Turski, W.A. Distribution, synthesis, and absorption of kynurenic acid in plants. Planta Med. 2011, 77, 858–864. [Google Scholar] [CrossRef]
  26. Koch, W.; Kukula-Koch, W.; Głowniak, K. Catechin Composition and Antioxidant Activity of Black Teas in Relation to Brewing Time. J. AOAC Int. 2017, 100, 1694–1699. [Google Scholar] [CrossRef]
  27. Kukula-Koch, W.; Grabarska, A.; Łuszczki, J.; Czernicka, L.; Nowosadzka, E.; Gumbarewicz, E.; Jarząb, A.; Audo, G.; Upadhyay, S.; Głowniak, K.; et al. Superior anticancer activity is demonstrated by total extract of Curcuma longa L. as opposed to individual curcuminoids separated by centrifugal partition chromatography. Phytother. Res. 2018, 32, 933–942. [Google Scholar] [CrossRef]
  28. Salehi, B.; Quispe, C.; Sharifi-Rad, J.; Cruz-Martins, N.; Nigam, M.; Mishra, A.P.; Konovalov, D.A.; Orobinskaya, V.; Abu-Reidah, I.M.; Zam, W.; et al. Phytosterols: From Preclinical Evidence to Potential Clinical Applications. Front. Pharmacol. 2020, 11, 599959. [Google Scholar] [CrossRef]
  29. Füchtbauer, S.; Mousavi, S.; Bereswill, S.; Heimesaat, M.M. Antibacterial properties of capsaicin and its derivatives and their potential to fight antibiotic resistance—A literature survey. Eur. J. Microbiol. Immunol. 2021, 11, 10–17. [Google Scholar] [CrossRef]
  30. Periferakis, A.-T.; Periferakis, A.; Periferakis, K.; Caruntu, A.; Badarau, I.A.; Savulescu-Fiedler, I.; Scheau, C.; Caruntu, C. Antimicrobial Properties of Capsaicin: Available Data and Future Research Perspectives. Nutrients 2023, 15, 4097. [Google Scholar] [CrossRef]
  31. Boots, A.W.; Haenen, G.R.; Bast, A. Health effects of quercetin: From antioxidant to nutraceutical. Eur. J. Pharmacol. 2008, 585, 325–337. [Google Scholar] [CrossRef]
  32. Dumitrache, M.D.; Jieanu, A.S.; Scheau, C.; Badarau, I.A.; Popescu, G.D.A.; Caruntu, A.; Costache, D.O.; Costache, R.S.; Constantin, C.; Neagu, M.; et al. Comparative effects of capsaicin in chronic obstructive pulmonary disease and asthma (Review). Exp. Ther. Med. 2021, 22, 917. [Google Scholar] [CrossRef]
  33. Kumar, R.; Vijayalakshmi, S.; Nadanasabapathi, S. Health benefits of quercetin. Def. Life Sci. J. 2017, 2, 142–151. [Google Scholar] [CrossRef]
  34. Aghababaei, F.; Hadidi, M. Recent advances in potential health benefits of quercetin. Pharmaceuticals 2023, 16, 1020. [Google Scholar] [CrossRef]
  35. Noorafshan, A.; Ashkani-Esfahani, S. A review of therapeutic effects of curcumin. Curr. Pharm. Des. 2013, 19, 2032–2046. [Google Scholar]
  36. Hewlings, S.J.; Kalman, D.S. Curcumin: A review of its effects on human health. Foods 2017, 6, 92. [Google Scholar] [CrossRef]
  37. Duvoix, A.; Blasius, R.; Delhalle, S.; Schnekenburger, M.; Morceau, F.; Henry, E.; Dicato, M.; Diederich, M. Chemopreventive and therapeutic effects of curcumin. Cancer Lett. 2005, 223, 181–190. [Google Scholar] [CrossRef]
  38. Periferakis, A.; Periferakis, K.; Badarau, I.A.; Petran, E.M.; Popa, D.C.; Caruntu, A.; Costache, R.S.; Scheau, C.; Caruntu, C.; Costache, D.O. Kaempferol: Antimicrobial Properties, Sources, Clinical, and Traditional Applications. Int. J. Mol. Sci. 2022, 23, 15054. [Google Scholar] [CrossRef]
  39. Periferakis, A.; Periferakis, A.-T.; Troumpata, L.; Periferakis, K.; Scheau, A.-E.; Savulescu-Fiedler, I.; Caruntu, A.; Badarau, I.A.; Caruntu, C.; Scheau, C. Kaempferol: A Review of Current Evidence of Its Antiviral Potential. Int. J. Mol. Sci. 2023, 24, 16299. [Google Scholar] [CrossRef]
  40. Castelli, G.; Bruno, F.; Vitale, F.; Roberti, M.; Colomba, C.; Giacomini, E.; Guidotti, L.; Cascio, A.; Tolomeo, M. In vitro antileishmanial activity of trans-stilbene and terphenyl compounds. Exp Parasitol 2016, 166, 1–9. [Google Scholar] [CrossRef]
  41. Bakrim, S.; Machate, H.; Benali, T.; Sahib, N.; Jaouadi, I.; Omari, N.E.; Aboulaghras, S.; Bangar, S.P.; Lorenzo, J.M.; Zengin, G.; et al. Natural Sources and Pharmacological Properties of Pinosylvin. Plants 2022, 11, 1541. [Google Scholar] [CrossRef]
  42. Rivière, C.; Pawlus, A.D.; Mérillon, J.M. Natural stilbenoids: Distribution in the plant kingdom and chemotaxonomic interest in Vitaceae. Nat. Prod. Rep. 2012, 29, 1317–1333. [Google Scholar] [CrossRef]
  43. Chu, L.L.; Tran, C.T.B.; Pham, D.T.K.; Nguyen, H.T.A.; Nguyen, M.H.; Pham, N.M.; Nguyen, A.T.V.; Phan, D.T.; Do, H.M.; Nguyen, Q.H. Metabolic Engineering of Corynebacterium glutamicum for the Production of Flavonoids and Stilbenoids. Molecules 2024, 29, 2252. [Google Scholar] [CrossRef]
  44. Akinwumi, B.C.; Bordun, K.M.; Anderson, H.D. Biological Activities of Stilbenoids. Int. J. Mol. Sci. 2018, 19, 792. [Google Scholar] [CrossRef] [PubMed]
  45. Sobolev, V.S.; Horn, B.W.; Potter, T.L.; Deyrup, S.T.; Gloer, J.B. Production of Stilbenoids and Phenolic Acids by the Peanut Plant at Early Stages of Growth. J. Agric. Food Chem. 2006, 54, 3505–3511. [Google Scholar] [CrossRef]
  46. Valletta, A.; Iozia, L.M.; Leonelli, F. Impact of Environmental Factors on Stilbene Biosynthesis. Plants 2021, 10, 90. [Google Scholar] [CrossRef]
  47. Erdtman, V.H. Tallvedkärnans extraktivämnen och deras inverkan på uppslutningen enligt sulfitmetoden. Sven. Papperstidning 1939, 42, 344–349. [Google Scholar]
  48. Shrestha, A.; Pandey, R.P.; Sohng, J.K. Biosynthesis of resveratrol and piceatannol in engineered microbial strains: Achievements and perspectives. Appl. Microbiol. Biotechnol. 2019, 103, 2959–2972. [Google Scholar] [CrossRef]
  49. Langcake, P.; Pryce, R.J. The production of resveratrol by Vitis vinifera and other members of the Vitaceae as a response to infection or injury. Physiol. Plant Pathol. 1976, 9, 77–86. [Google Scholar] [CrossRef]
  50. Jeandet, P.; Hébrard, C.; Deville, M.A.; Cordelier, S.; Dorey, S.; Aziz, A.; Crouzet, J. Deciphering the role of phytoalexins in plant-microorganism interactions and human health. Molecules 2014, 19, 18033–18056. [Google Scholar] [CrossRef]
  51. Schöppner, A.; Kindl, H. Purification and properties of a stilbene synthase from induced cell suspension cultures of peanut. J. Biol. Chem. 1984, 259, 6806–6811. [Google Scholar] [CrossRef]
  52. Mendonça, E.L.S.S.; Xavier, J.A.; Fragoso, M.B.T.; Silva, M.O.; Escodro, P.B.; Oliveira, A.C.M.; Tucci, P.; Saso, L.; Goulart, M.O.F. E-Stilbenes: General Chemical and Biological Aspects, Potential Pharmacological Activity Based on the Nrf2 Pathway. Pharmaceuticals 2024, 17, 232. [Google Scholar] [CrossRef]
  53. Schuster, R.; Holzer, W.; Doerfler, H.; Weckwerth, W.; Viernstein, H.; Okonogi, S.; Mueller, M. Cajanus cajan-a source of PPARγ activators leading to anti-inflammatory and cytotoxic effects. Food Funct. 2016, 7, 3798–3806. [Google Scholar] [CrossRef]
  54. Lim, S.J.; Kim, M.; Randy, A.; Nho, C.W. Inhibitory effect of the branches of Hovenia dulcis Thunb. and its constituent pinosylvin on the activities of IgE-mediated mast cells and passive cutaneous anaphylaxis in mice. Food Funct. 2015, 6, 1361–1370. [Google Scholar] [CrossRef]
  55. Fu, Y.; Sun, X.; Wang, L.; Chen, S. Pharmacokinetics and Tissue Distribution Study of Pinosylvin in Rats by Ultra-High-Performance Liquid Chromatography Coupled with Linear Trap Quadrupole Orbitrap Mass Spectrometry. Evid. Based Complement. Altern. Med. 2018, 2018, 4181084. [Google Scholar] [CrossRef]
  56. Labib, R.M.; Malak, L.G.; Youssef, F.S.; Ross, S.A. A new stilbene from Agonis flexuosa leaves and verification of its histamine release inhibitory activity using in silico and in vitro studies. S. Afr. J. Bot. 2020, 135, 384–390. [Google Scholar] [CrossRef]
  57. Aalto, A.L.; Saadabadi, A.; Lindholm, F.; Kietz, C.; Himmelroos, E.; Marimuthu, P.; Salo-Ahen, O.M.H.; Eklund, P.; Meinander, A. Stilbenoid compounds inhibit NF-κB-mediated inflammatory responses in the Drosophila intestine. Front. Immunol. 2023, 14, 1253805. [Google Scholar] [CrossRef]
  58. Celimene, C.C.; Micales, J.A.; Ferge, L.; Young, R.A. Efficacy of pinosylvins against white-rot and brown-rot fungi. Holzforschung 1999. [Google Scholar] [CrossRef]
  59. Rowe, J.W.; Bower, C.L.; Wagner, E.R. Extractives of jack pine bark: Occurrence of cis- and trans-pinosylvin dimethyl ether and ferulic acid esters. Phytochemistry 1969, 8, 235–241. [Google Scholar] [CrossRef]
  60. Lindberg, L.E.; Willför, S.M.; Holmbom, B.R. Antibacterial effects of knotwood extractives on paper mill bacteria. J. Ind. Microbiol. Biotechnol. 2004, 31, 137–147. [Google Scholar] [CrossRef]
  61. Pietarinen, S.P.; Willför, S.M.; Ahotupa, M.O.; Hemming, J.E.; Holmbom, B.R. Knotwood and bark extracts: Strong antioxidants from waste materials. J. Wood Sci. 2006, 52, 436–444. [Google Scholar] [CrossRef]
  62. Yildirim, H.; Holmbom, B. Investigations on the wood extractives of pine species from Turkey. Part III. Nonvolatile, nonpolar components in Pinus brutia (Henry). Acta Acad. Abo B 1977, 37, 1–9. [Google Scholar]
  63. Sinyeue, C.; Maerker, L.; Guentas, L.; Medevielle, V.; Bregier, F.; Chaleix, V.; Sol, V.; Lebouvier, N. Polyphenol Content, Antioxidant, and Antibiotic Activities of Pinus Caribaea Morelet Forestry Coproducts. Nat. Prod. Commun. 2023, 18, 1934578X231211958. [Google Scholar] [CrossRef]
  64. Willför, S.M.; Ahotupa, M.O.; Hemming, J.E.; Reunanen, M.H.; Eklund, P.C.; Sjöholm, R.E.; Eckerman, C.S.; Pohjamo, S.P.; Holmbom, B.R. Antioxidant activity of knotwood extractives and phenolic compounds of selected tree species. J. Agric. Food Chem. 2003, 51, 7600–7606. [Google Scholar] [CrossRef] [PubMed]
  65. Dumas, M.T.; Hubbes, M.; Strunz, G.M. Identification of some compounds associated with resistance of Pinus densiflora to Fomes annosus. Eur. J. For. Pathol. 1983, 13, 151–160. [Google Scholar] [CrossRef]
  66. Kodan, A.; Kuroda, H.; Sakai, F. A stilbene synthase from Japanese red pine (Pinus densiflora): Implications for phytoalexin accumulation and down-regulation of flavonoid biosynthesis. Proc. Natl. Acad. Sci. USA 2002, 99, 3335–3339. [Google Scholar] [CrossRef]
  67. Benouadah, N.; Pranovich, A.; Aliouche, D.; Hemming, J.; Smeds, A.; Willför, S. Analysis of extractives from Pinus halepensis and Eucalyptus camaldulensis as predominant trees in Algeria. Holzforschung 2018, 72, 97–104. [Google Scholar] [CrossRef]
  68. El Omari, N.; Ezzahrae Guaouguaou, F.; El Menyiy, N.; Benali, T.; Aanniz, T.; Chamkhi, I.; Balahbib, A.; Taha, D.; Shariati, M.A.; Zengin, G.; et al. Phytochemical and biological activities of Pinus halepensis mill., and their ethnomedicinal use. J. Ethnopharmacol. 2021, 268, 113661. [Google Scholar] [CrossRef]
  69. Wijayanto, A.; Dumarçay, S.; Gérardin-Charbonnier, C.; Sari, R.K.; Syafii, W.; Gérardin, P. Phenolic and lipophilic extractives in Pinus merkusii Jungh. et de Vries knots and stemwood. Ind. Crops Prod. 2015, 69, 466–471. [Google Scholar] [CrossRef]
  70. Ioannidis, K.; Melliou, E.; Alizoti, P.; Magiatis, P. Identification of black pine (Pinus nigra Arn.) heartwood as a rich source of bioactive stilbenes by qNMR. J. Sci. Food Agric. 2017, 97, 1708–1716. [Google Scholar] [CrossRef]
  71. Suga, T.; Ohta, S.; Munesada, K.; Ide, N.; Kurokawa, M.; Shimizu, M.; Ohta, E. Endogenous pine wood nematicidal substances in pines, Pinus massoniana, P. strobus and P. palustris. Phytochemistry 1993, 33, 1395–1401. [Google Scholar] [CrossRef]
  72. Conde, E.; Fang, W.; Hemming, J.; Willför, S.; Domínguez, H.; Parajó, J.C. Recovery of bioactive compounds from Pinus pinaster wood by consecutive extraction stages. Wood Sci. Technol. 2014, 48, 311–323. [Google Scholar] [CrossRef]
  73. Gabaston, J.; Richard, T.; Cluzet, S.; Palos Pinto, A.; Dufour, M.C.; Corio-Costet, M.F.; Mérillon, J.M. Pinus pinaster Knot: A Source of Polyphenols against Plasmopara viticola. J. Agric. Food Chem. 2017, 65, 8884–8891. [Google Scholar] [CrossRef]
  74. Gabaston, J.; Leborgne, C.; Waffo-Téguo, P.; Pedrot, E.; Richard, T.; Mérillon, J.-M.; Valls Fonayet, J. Separation and isolation of major polyphenols from maritime pine (Pinus pinaster) knots by two-step centrifugal partition chromatography monitored by LC-MS and NMR spectroscopy. J. Sep. Sci. 2020, 43, 1080–1088. [Google Scholar] [CrossRef]
  75. Kaushik, P.; Kaushik, D.; Khokra, S.L. Ethnobotany and phytopharmacology of Pinus roxburghii Sargent: A plant review. J. Integr. Med. 2013, 11, 371–376. [Google Scholar] [CrossRef] [PubMed]
  76. Laavola, M.; Nieminen, R.; Leppänen, T.; Eckerman, C.; Holmbom, B.; Moilanen, E. Pinosylvin and monomethylpinosylvin, constituents of an extract from the knot of Pinus sylvestris, reduce inflammatory gene expression and inflammatory responses in vivo. J. Agric. Food Chem. 2015, 63, 3445–3453. [Google Scholar] [CrossRef] [PubMed]
  77. Hemingway, R.W.; McGraw, G.W.; Barras, S.J. Polyphenols in Ceratocystis minor infected Pinus taeda: Fungal metabolites, phloem and xylem phenols. J. Agric. Food Chem. 1977, 25, 717–722. [Google Scholar] [CrossRef]
  78. Kostecki, K.; Engelmeier, D.; Pacher, T.; Hofer, O.; Vajrodaya, S.; Greger, H. Dihydrophenanthrenes and other antifungal stilbenoids from Stemona cf. pierrei. Phytochemistry 2004, 65, 99–106. [Google Scholar] [CrossRef] [PubMed]
  79. Pacher, T.; Seger, C.; Engelmeier, D.; Vajrodaya, S.; Hofer, O.; Greger, H. Antifungal Stilbenoids from Stemona collinsae. J. Nat. Prod. 2002, 65, 820–827. [Google Scholar] [CrossRef]
  80. Brem, B.; Seger, C.; Pacher, T.; Hofer, O.; Vajrodaya, S.; Greger, H. Feeding deterrence and contact toxicity of Stemona alkaloids-a source of potent natural insecticides. J. Agric. Food Chem. 2002, 50, 6383–6388. [Google Scholar] [CrossRef]
  81. Adams, M.; Pacher, T.; Greger, H.; Bauer, R. Inhibition of leukotriene biosynthesis by stilbenoids from Stemona species. J. Nat. Prod. 2005, 68, 83–85. [Google Scholar] [CrossRef]
  82. Wang, P.; Sang, S. Metabolism and pharmacokinetics of resveratrol and pterostilbene. Biofactors 2018, 44, 16–25. [Google Scholar] [CrossRef]
  83. Setoguchi, Y.; Oritani, Y.; Ito, R.; Inagaki, H.; Maruki-Uchida, H.; Ichiyanagi, T.; Ito, T. Absorption and Metabolism of Piceatannol in Rats. J. Agric. Food Chem. 2014, 62, 2541–2548. [Google Scholar] [CrossRef]
  84. Roupe, K.A.; Remsberg, C.M.; Yáñez, J.A.; Davies, N.M. Pharmacometrics of stilbenes: Seguing towards the clinic. Curr. Clin. Pharmacol. 2006, 1, 81–101. [Google Scholar] [CrossRef] [PubMed]
  85. Nunes, S.; Danesi, F.; Del Rio, D.; Silva, P. Resveratrol and inflammatory bowel disease: The evidence so far. Nutr. Res. Rev. 2018, 31, 85–97. [Google Scholar] [CrossRef]
  86. Danesi, F.; Ferguson, L.R. Could Pomegranate Juice Help in the Control of Inflammatory Diseases? Nutrients 2017, 9, 958. [Google Scholar] [CrossRef] [PubMed]
  87. Fu, J.; Wu, S.; Wang, M.; Tian, Y.; Zhang, Z.; Song, R. Intestinal metabolism of Polygonum cuspidatum in vitro and in vivo. Biomed. Chromatogr. 2018, 32, e4190. [Google Scholar] [CrossRef]
  88. Miksits, M.; Maier-Salamon, A.; Aust, S.; Thalhammer, T.; Reznicek, G.; Kunert, O.; Haslinger, E.; Szekeres, T.; Jaeger, W. Sulfation of resveratrol in human liver: Evidence of a major role for the sulfotransferases SULT1A1 and SULT1E1. Xenobiotica 2005, 35, 1101–1119. [Google Scholar] [CrossRef]
  89. Boocock, D.J.; Faust, G.E.; Patel, K.R.; Schinas, A.M.; Brown, V.A.; Ducharme, M.P.; Booth, T.D.; Crowell, J.A.; Perloff, M.; Gescher, A.J.; et al. Phase I dose escalation pharmacokinetic study in healthy volunteers of resveratrol, a potential cancer chemopreventive agent. Cancer Epidemiol. Biomark. Prev. 2007, 16, 1246–1252. [Google Scholar] [CrossRef]
  90. Springer, M.; Moco, S. Resveratrol and Its Human Metabolites-Effects on Metabolic Health and Obesity. Nutrients 2019, 11, 143. [Google Scholar] [CrossRef]
  91. Walle, T.; Hsieh, F.; DeLegge, M.H.; Oatis, J.E.; Walle, U.K. High absorption but very low bioavailability of oral resveratrol in humans. Drug Metab. Dispos. 2004, 32, 1377–1382. [Google Scholar] [CrossRef]
  92. Poljanšek, I.; Oven, P.; Vek, V.; Raitanen, J.-E.; Hemming, J.; Willför, S. Isolation of pure pinosylvins from industrial knotwood residue with non-chlorinated solvents. Holzforschung 2019, 73, 475–484. [Google Scholar] [CrossRef]
  93. Kim, H.; Rencoret, J.; Elder, T.J.; Del Río, J.C.; Ralph, J. Biomimetic oxidative copolymerization of hydroxystilbenes and monolignols. Sci. Adv. 2023, 9, eade5519. [Google Scholar] [CrossRef]
  94. Zhang, S.; Lei, Y.; Xu, X.; Shi, W.; Chen, S. [Synthesis of artificial diethylstilbestrol antigen for preparation of polyclonal antibodies]. Zhejiang Da Xue Xue Bao Yi Xue Ban 2013, 42, 25–31. [Google Scholar] [CrossRef]
  95. Wang, X.F.; Zhang, Y.; Lin, M.B.; Hou, Q.; Yao, C.S.; Shi, J.G. Biomimetic synthesis of active isorhapontigenin dimers. J. Asian Nat. Prod. Res. 2014, 16, 511–521. [Google Scholar] [CrossRef]
  96. Jorgensen, E.; Balsillie, D. Formation of heartwood phenols in callus tissue cultures of red pine (Pinus resinosa). Can. J. Bot. 1969, 47, 1015–1016. [Google Scholar] [CrossRef]
  97. Jorgensen, E. The formation of pinosylvin and its monomethyl ether in the sapwood of Pinus resinosa Ait. Can. J. Bot. 1961, 39, 1765–1772. [Google Scholar] [CrossRef]
  98. von Rudloff, E.; Jorgensen, E. The biosynthesis of pinosylvin in the sapwood of Pinus resinosa AIT. Phytochemistry 1963, 2, 297–304. [Google Scholar] [CrossRef]
  99. Koo, H.B.; Hwang, H.S.; Han, J.Y.; Cheong, E.J.; Kwon, Y.S.; Choi, Y.E. Enhanced production of pinosylvin stilbene with aging of Pinus strobus callus and nematicidal activity of callus extracts against pinewood nematodes. Sci. Rep. 2022, 12, 770. [Google Scholar] [CrossRef]
  100. Lange, B.M.; Trost, M.; Heller, W.; Langebartels, C.; Sandermann, H. Elicitor-induced formation of free and cell-wall-bound stilbenes in cell-suspension cultures of Scots pine (Pinus sylvestris L.). Planta 1994, 194, 143–148. [Google Scholar] [CrossRef]
  101. Mahdizade Ari, M.; Dadgar, L.; Elahi, Z.; Ghanavati, R.; Taheri, B. Genetically Engineered Microorganisms and Their Impact on Human Health. Int. J. Clin. Pract. 2024, 2024, 6638269. [Google Scholar] [CrossRef]
  102. Watts, K.T.; Lee, P.C.; Schmidt-Dannert, C. Biosynthesis of plant-specific stilbene polyketides in metabolically engineered Escherichia coli. BMC Biotechnol. 2006, 6, 22. [Google Scholar] [CrossRef]
  103. Katsuyama, Y.; Funa, N.; Horinouchi, S. Precursor-directed biosynthesis of stilbene methyl ethers in Escherichia coli. Biotechnol. J. 2007, 2, 1286–1293. [Google Scholar] [CrossRef]
  104. van Summeren-Wesenhagen, P.V.; Marienhagen, J. Metabolic engineering of Escherichia coli for the synthesis of the plant polyphenol pinosylvin. Appl. Environ. Microbiol. 2015, 81, 840–849. [Google Scholar] [CrossRef] [PubMed]
  105. Wang, S.; Zhang, S.; Xiao, A.; Rasmussen, M.; Skidmore, C.; Zhan, J. Metabolic engineering of Escherichia coli for the biosynthesis of various phenylpropanoid derivatives. Metab. Eng. 2015, 29, 153–159. [Google Scholar] [CrossRef]
  106. Liang, J.L.; Guo, L.Q.; Lin, J.F.; He, Z.Q.; Cai, F.J.; Chen, J.F. A novel process for obtaining pinosylvin using combinatorial bioengineering in Escherichia coli. World J. Microbiol. Biotechnol. 2016, 32, 102. [Google Scholar] [CrossRef]
  107. Xu, J.Y.; Xu, Y.; Chu, X.; Tan, M.; Ye, B.C. Protein Acylation Affects the Artificial Biosynthetic Pathway for Pinosylvin Production in Engineered E. coli. ACS Chem. Biol. 2018, 13, 1200–1208. [Google Scholar] [CrossRef] [PubMed]
  108. Salas-Navarrete, C.; Hernández-Chávez, G.; Flores, N.; Martínez, L.M.; Martinez, A.; Bolívar, F.; Barona-Gomez, F.; Gosset, G. Increasing pinosylvin production in Escherichia coli by reducing the expression level of the gene fabI-encoded enoyl-acyl carrier protein reductase. Electron. J. Biotechnol. 2018, 33, 11–16. [Google Scholar] [CrossRef]
  109. Schwanemann, T.; Otto, M.; Wynands, B.; Marienhagen, J.; Wierckx, N. A Pseudomonas taiwanensis malonyl-CoA platform strain for polyketide synthesis. Metab. Eng. 2023, 77, 219–230. [Google Scholar] [CrossRef] [PubMed]
  110. Kallscheuer, N.; Vogt, M.; Kappelmann, J.; Krumbach, K.; Noack, S.; Bott, M.; Marienhagen, J. Identification of the phd gene cluster responsible for phenylpropanoid utilization in Corynebacterium glutamicum. Appl. Microbiol. Biotechnol. 2016, 100, 1871–1881. [Google Scholar] [CrossRef]
  111. Wallace, R.J. Antimicrobial properties of plant secondary metabolites. Proc. Nutr. Soc. 2004, 63, 621–629. [Google Scholar] [CrossRef]
  112. Salehi, B.; Zakaria, Z.A.; Gyawali, R.; Ibrahim, S.A.; Rajkovic, J.; Shinwari, Z.K.; Khan, T.; Sharifi-Rad, J.; Ozleyen, A.; Turkdonmez, E. Piper species: A comprehensive review on their phytochemistry, biological activities and applications. Molecules 2019, 24, 1364. [Google Scholar] [CrossRef]
  113. Makuwa, S.C.; Serepa-Dlamini, M.H. The antibacterial activity of crude extracts of secondary metabolites from bacterial endophytes associated with Dicoma anomala. Int. J. Microbiol. 2021, 2021, 8812043. [Google Scholar] [CrossRef]
  114. Aminah, N.S.; Laili, E.; Rafi, M.; Rochman, A.; Insanu, M.; Tun, K. Secondary metabolite compounds from Sida genus and their bioactivity. Heliyon 2021, 7, e06682. [Google Scholar] [CrossRef]
  115. Jubair, N.; Rajagopal, M.; Chinnappan, S.; Abdullah, N.B.; Fatima, A. Review on the antibacterial mechanism of plant-derived compounds against multidrug-resistant bacteria (MDR). Evid.-Based Complement. Altern. Med. 2021, 2021, 3663315. [Google Scholar] [CrossRef] [PubMed]
  116. Bhatia, P.; Sharma, A.; George, A.J.; Anvitha, D.; Kumar, P.; Dwivedi, V.P.; Chandra, N.S. Antibacterial activity of medicinal plants against ESKAPE: An update. Heliyon 2021, 7, e06310. [Google Scholar] [CrossRef] [PubMed]
  117. Dorobăţ, O.-M.; Moisoiu, A.; Tălăpan, D. Bacteria isolated from pleural fluid and their resistance to antimicrobials. Pneumologia 2006, 55, 47–51. [Google Scholar]
  118. Dorobăţ, O.M.; Moisoiu, A.; Tălăpan, D. Incidence and resistance patterns of pathogens from lower respiratory tract infections (LRTI). Pneumologia 2007, 56, 7–15. [Google Scholar]
  119. Dorobăţ, O.M.; Bădicuţ, I.; Tălăpan, D.; Tenea, C.; Rafila, A. Antibiotic resistance of Gram-positive cocci isolated in 2008. Bacteriol. Virusol. Parazitol. Epidemiol. (Buchar. Rom. 1990) 2010, 55, 83–92. [Google Scholar]
  120. Popescu, G.A.; Șerban, R.; Iosif, I.; Codiță, I.; Dorobăț, O.; Tălăpan, D.; Buzea, M.; Szekely, E.; Dorneanu, O.; Bota, K. Antimicrobial resistance of germs isolated from invasive infections—Romania 2012. BMC Infect. Dis. 2013, 13, O16. [Google Scholar] [CrossRef]
  121. Rafila, A.; Talapan, D.; Dorobăţ, O.M.; Popescu, G.A.; Piţigoi, D.; Florea, D.; Buicu, F.C. Emergence of Carbapenemase-producing Enterobacteriaceae, a Public Health Threat: A Romanian Infectious Disease Hospital Based Study/Emergenţa Enterobacteriaceaelor producătoare de carbapenemaze, o ameninţare pentru sănătatea publică: Un studiu realizat într-un spital romanesc de boli infectioase. Rev. Romana Med. Lab. 2015, 23, 295–301. [Google Scholar]
  122. Kohler, P.P.; Melano, R.G.; Patel, S.N.; Shafinaz, S.; Faheem, A.; Coleman, B.L.; Green, K.; Armstrong, I.; Almohri, H.; Borgia, S. Emergence of carbapenemase-producing Enterobacteriaceae, south-central Ontario, Canada. Emerg. Infect. Dis. 2018, 24, 1674. [Google Scholar] [CrossRef]
  123. Tălăpan, D.; Rafila, A. Five-Year Survey of Asymptomatic Colonization with Multidrug-Resistant Organisms in a Romanian Tertiary Care Hospital. Infect. Drug Resist. 2022, 15, 2959–2967. [Google Scholar] [CrossRef]
  124. Cireșă, A.; Tălăpan, D.; Vasile, C.-C.; Popescu, C.; Popescu, G.-A. Evolution of Antimicrobial Resistance in Klebsiella pneumoniae over 3 Years (2019–2021) in a Tertiary Hospital in Bucharest, Romania. Antibiotics 2024, 13, 431. [Google Scholar] [CrossRef]
  125. Kanellopoulos, C.; Lamprinou, V.; Politi, A.; Voudouris, P.; Iliopoulos, I.; Kokkaliari, M.; Moforis, L.; Economou-Amilli, A. Microbial Mat Stratification in Travertine Depositions of Greek Hot Springs and Biomineralization Processes. Minerals 2022, 12, 1408. [Google Scholar] [CrossRef]
  126. Kanellopoulos, C.; Lamprinou, V.; Politi, A.; Voudouris, P.; Iliopoulos, I.; Kokkaliari, M.; Moforis, L.; Economou-Amilli, A. Speleothems and Biomineralization Processes in Hot Spring Environment: The Case of Aedipsos (Edipsos), Euboea (Evia) Island, Greece. J. Mar. Sci. Eng. 2022, 10, 1909. [Google Scholar] [CrossRef]
  127. Kanellopoulos, C.; Lamprinou, V.; Politi, A.; Voudouris, P.; Economou-Amilli, A. Pioneer species of Cyanobacteria in hot springs and their role to travertine formation: The case of Aedipsos hot springs, Euboea (Evia), Greece. Depos. Rec. 2022, 8, 1079–1092. [Google Scholar] [CrossRef]
  128. Ackerman, S.; Gonzales, R. The context of antibiotic overuse. Ann. Intern. Med. 2012, 157, 211–212. [Google Scholar] [CrossRef] [PubMed]
  129. Shallcross, L.J.; Davies, D.S.C. Antibiotic overuse: A key driver of antimicrobial resistance. Br. J. Gen. Pract. 2014, 64, 604–605. [Google Scholar] [CrossRef]
  130. Denyer Willis, L.; Chandler, C. Quick fix for care, productivity, hygiene and inequality: Reframing the entrenched problem of antibiotic overuse. BMJ Glob. Health 2019, 4, e001590. [Google Scholar] [CrossRef]
  131. Kitsos, N.; Cassimos, D.; Xinias, I.; Agakidis, C.; Mavroudi, A. Management of antibiotic allergy in children: A practical approach. Allergol. Immunopathol. 2022, 50, 30–38. [Google Scholar] [CrossRef]
  132. Green, E.A.; Fogarty, K.; Ishmael, F.T. Penicillin Allergy: Mechanisms, Diagnosis, and Management. Prim. Care 2023, 50, 221–235. [Google Scholar] [CrossRef]
  133. Wrynn, A.F. An overview of penicillin allergies for nurses. Nursing 2023, 53, 27–31. [Google Scholar] [CrossRef]
  134. Westphal, J.F.; Vetter, D.; Brogard, J.M. Hepatic side-effects of antibiotics. J. Antimicrob. Chemother. 1994, 33, 387–401. [Google Scholar] [CrossRef]
  135. Thiim, M.; Friedman, L.S. Hepatotoxicity of antibiotics and antifungals. Clin. Liver Dis. 2003, 7, 381–399. [Google Scholar] [CrossRef]
  136. Polson, J.E. Hepatotoxicity due to antibiotics. Clin. Liver Dis. 2007, 11, 549–561. [Google Scholar] [CrossRef]
  137. Andrade, R.J.; Tulkens, P.M. Hepatic safety of antibiotics used in primary care. J. Antimicrob. Chemother. 2011, 66, 1431–1446. [Google Scholar] [CrossRef] [PubMed]
  138. Rehman, K.; Kamran, S.H.; Hamid Akash, M.S. Chapter 16—Toxicity of antibiotics. In Antibiotics and Antimicrobial ResistanceGenes in the Environment; Hashmi, M.Z., Ed.; Elsevier: Amsterdam, The Netherlands, 2020; Volume 1, pp. 234–252. [Google Scholar]
  139. Sousa, V.; Luís, Â.; Oleastro, M.; Domingues, F.; Ferreira, S. Polyphenols as resistance modulators in Arcobacter butzleri. Folia Microbiol. 2019, 64, 547–554. [Google Scholar] [CrossRef]
  140. Välimaa, A.L.; Honkalampi-Hämäläinen, U.; Pietarinen, S.; Willför, S.; Holmbom, B.; von Wright, A. Antimicrobial and cytotoxic knotwood extracts and related pure compounds and their effects on food-associated microorganisms. Int. J. Food Microbiol. 2007, 115, 235–243. [Google Scholar] [CrossRef]
  141. Hou, W.; Sun, P.-H.; Geng, Z.-Z.; Xu, H.-G.; Lin, J.; Chen, W.-M. Design, Synthesis and Antibacterial Assay of Pinosylvin Acid Derivatives. J. Pharm. Biomed. Sci. 2016, 6, 369–373. [Google Scholar]
  142. Bouarab-Chibane, L.; Forquet, V.; Lantéri, P.; Clément, Y.; Léonard-Akkari, L.; Oulahal, N.; Degraeve, P.; Bordes, C. Antibacterial Properties of Polyphenols: Characterization and QSAR (Quantitative Structure–Activity Relationship) Models. Front. Microbiol. 2019, 10, 829. [Google Scholar] [CrossRef]
  143. Silva, F.; Nerín, C.; Domingues, F.C. Stilbene phytoallexins inclusion complexes: A natural-based strategy to control foodborne pathogen Campylobacter. Food Control 2015, 54, 66–73. [Google Scholar] [CrossRef]
  144. Silva, F.; Domingues, F.C.; Nerín, C. Control microbial growth on fresh chicken meat using pinosylvin inclusion complexes based packaging absorbent pads. LWT 2018, 89, 148–154. [Google Scholar] [CrossRef]
  145. Lee, S.K.; Lee, H.J.; Min, H.Y.; Park, E.J.; Lee, K.M.; Ahn, Y.H.; Cho, Y.J.; Pyee, J.H. Antibacterial and antifungal activity of pinosylvin, a constituent of pine. Fitoterapia 2005, 76, 258–260. [Google Scholar] [CrossRef] [PubMed]
  146. Plumed-Ferrer, C.; Väkeväinen, K.; Komulainen, H.; Rautiainen, M.; Smeds, A.; Raitanen, J.E.; Eklund, P.; Willför, S.; Alakomi, H.L.; Saarela, M.; et al. The antimicrobial effects of wood-associated polyphenols on food pathogens and spoilage organisms. Int. J. Food Microbiol. 2013, 164, 99–107. [Google Scholar] [CrossRef] [PubMed]
  147. de Bruijn, W.J.C.; Araya-Cloutier, C.; Bijlsma, J.; de Swart, A.; Sanders, M.G.; de Waard, P.; Gruppen, H.; Vincken, J.-P. Antibacterial prenylated stilbenoids from peanut (Arachis hypogaea). Phytochem. Lett. 2018, 28, 13–18. [Google Scholar] [CrossRef]
  148. Marion-Sanchez, K.; Olive, C.; Platon, M.G.; Cesarine, M.; Derancourt, C.; Pailla, K. Achromobacter xylosoxidans in hospital environments: Still waters run deep! Trans. R. Soc. Trop. Med. Hyg. 2020, 114, 470–472. [Google Scholar] [CrossRef]
  149. Bonis, B.M.; Hunter, R.C. JMM Profile: Achromobacter xylosoxidans: The cloak-and-dagger opportunist. J. Med. Microbiol. 2022, 71, 001505. [Google Scholar] [CrossRef]
  150. Alkan, S. Ecthyma gangrenosum caused by Achromobacter xylosoxidans bacteremia. Rev. Soc. Bras. Med. Trop. 2023, 56, e00712023. [Google Scholar] [CrossRef]
  151. Jiménez-Guerra, G.; Casanovas MorenoTorres, I.; Moldovan, T.D.; Navarro-Marí, J.M.; Gutiérrez-Fernández, J. Arcobacter butzleri and intestinal colonization. Rev. Esp. Quim. 2020, 33, 73–75. [Google Scholar] [CrossRef]
  152. Ruiz de Alegría Puig, C.; Fernández Martínez, M.; Pablo Marcos, D.; Agüero Balbín, J.; Calvo Montes, J. Outbreak of Arcobacter butzleri? An emerging enteropathogen. Enferm. Infecc. Microbiol. Clin. 2023, 41, 169–172. [Google Scholar] [CrossRef]
  153. Chieffi, D.; Fanelli, F.; Fusco, V. Arcobacter butzleri: Up-to-date taxonomy, ecology, and pathogenicity of an emerging pathogen. Compr. Rev. Food Sci. Food Saf. 2020, 19, 2071–2109. [Google Scholar] [CrossRef]
  154. Bottone, E.J. Bacillus cereus, a volatile human pathogen. Clin. Microbiol. Rev. 2010, 23, 382–398. [Google Scholar] [CrossRef]
  155. Brown, K.L. Control of bacterial spores. Br. Med. Bull. 2000, 56, 158–171. [Google Scholar] [CrossRef]
  156. Peláez Bejarano, A.; García de Lomas, J.M.; Franco-Huertas, M.; Martínez-Marcos, F.J.; Jiménez-Hidalgo, A. Successful treatment of postsurgical meningitis caused by Bacillus cereus: A case report and literature review. J. Chemother. 2023, 35, 570–575. [Google Scholar] [CrossRef] [PubMed]
  157. Drobniewski, F.A. Bacillus cereus and related species. Clin. Microbiol. Rev. 1993, 6, 324–338. [Google Scholar] [CrossRef] [PubMed]
  158. Leong, S.S.; Korel, F.; King, J.H. Bacillus cereus: A review of “fried rice syndrome” causative agents. Microb. Pathog. 2023, 185, 106418. [Google Scholar] [CrossRef]
  159. Mu, Y.; Cong, Y. Bacillus coagulans and its applications in medicine. Benef. Microbes 2019, 10, 679–688. [Google Scholar] [CrossRef]
  160. Gupta, A.K.; Maity, C. Efficacy and safety of Bacillus coagulans LBSC in irritable bowel syndrome: A prospective, interventional, randomized, double-blind, placebo-controlled clinical study [CONSORT Compliant]. Medicine 2021, 100, e23641. [Google Scholar] [CrossRef]
  161. Kovács, Á.T. Bacillus subtilis. Trends Microbiol. 2019, 27, 724–725. [Google Scholar] [CrossRef]
  162. Lv, P.; Song, Y.; Liu, C.; Yu, L.; Shang, Y.; Tang, H.; Sun, S.; Wang, F. Application of Bacillus subtilis as a live vaccine vector: A review. J. Vet. Med. Sci. 2020, 82, 1693–1699. [Google Scholar] [CrossRef]
  163. Inouye, S.; Yamaguchi, H.; Takizawa, T. Screening of the antibacterial effects of a variety of essential oils on respiratory tract pathogens, using a modified dilution assay method. J. Infect. Chemother. 2001, 7, 251–254. [Google Scholar] [CrossRef]
  164. Flanagan, J.N.; Kavanaugh, L.; Steck, T.R. Burkholderia multivorans Exhibits Antibiotic Collateral Sensitivity. Microb. Drug Resist. 2020, 26, 1–8. [Google Scholar] [CrossRef]
  165. Dogra, S.; Angrup, A.; Kanaujia, R.; Vig, S.; Kaur, R.; Paul, R.A.; Biswal, M.; Samujh, R.; Ray, P. Burkholderia multivorans Sepsis Outbreak in a Neonatal Surgical Unit of a Tertiary Care Hospital. Indian. J. Pediatr. 2021, 88, 725. [Google Scholar] [CrossRef]
  166. Courtney, J.M.; Dunbar, K.E.; McDowell, A.; Moore, J.E.; Warke, T.J.; Stevenson, M.; Elborn, J.S. Clinical outcome of Burkholderia cepacia complex infection in cystic fibrosis adults. J. Cyst. Fibros. 2004, 3, 93–98. [Google Scholar] [CrossRef]
  167. Peralta, D.P.; Chang, A.Y.; Ariza-Hutchinson, A.; Ho, C.A. Burkholderia multivorans: A rare yet emerging cause of bacterial meningitis. IDCases 2018, 11, 61–63. [Google Scholar] [CrossRef] [PubMed]
  168. Tegtmeyer, N.; Sharafutdinov, I.; Harrer, A.; Soltan Esmaeili, D.; Linz, B.; Backert, S. Campylobacter Virulence Factors and Molecular Host-Pathogen Interactions. Curr. Top. Microbiol. Immunol. 2021, 431, 169–202. [Google Scholar] [CrossRef] [PubMed]
  169. Elmi, A.; Nasher, F.; Dorrell, N.; Wren, B.; Gundogdu, O. Revisiting Campylobacter jejuni Virulence and Fitness Factors: Role in Sensing, Adapting, and Competing. Front. Cell Infect. Microbiol. 2020, 10, 607704. [Google Scholar] [CrossRef]
  170. Gao, F.; Tu, L.; Chen, M.; Chen, H.; Zhang, X.; Zhuang, Y.; Luo, J.; Chen, M. Erythromycin resistance of clinical Campylobacter jejuni and Campylobacter coli in Shanghai, China. Front. Microbiol. 2023, 14, 1145581. [Google Scholar] [CrossRef]
  171. Barker, C.R.; Painset, A.; Swift, C.; Jenkins, C.; Godbole, G.; Maiden, M.C.J.; Dallman, T.J. Microevolution of Campylobacter jejuni during long-term infection in an immunocompromised host. Sci. Rep. 2020, 10, 10109. [Google Scholar] [CrossRef]
  172. Mariette, F.; Amrane, S.; Couteau, C.; Lagier, J.C.; Eldin, C. Campylobacter jejuni infection associated with miscarriage, a case report and literature review. J. Reprod. Immunol. 2020, 141, 103153. [Google Scholar] [CrossRef]
  173. Shang, P.; Zhu, M.; Wang, Y.; Zheng, X.; Wu, X.; Zhu, J.; Feng, J.; Zhang, H.L. Axonal variants of Guillain-Barré syndrome: An update. J. Neurol. 2021, 268, 2402–2419. [Google Scholar] [CrossRef]
  174. Adrianza, A.; Pourfarrokh, N.; Choi, H.; Hwang, M.; Lukey, J.; Jinadatha, C.; Navarathna, D.H. Campylobacter coli bacteremia associated with diarrhea. IDCases 2023, 31, e01734. [Google Scholar] [CrossRef]
  175. Gomes, S.A.; Trigo, C.; Pinto, F.F. Campylobacter coli Myocarditis: A case report. Cardiol. Young 2022, 32, 1172–1174. [Google Scholar] [CrossRef]
  176. Martinson, J.N.V.; Walk, S.T. Escherichia coli Residency in the Gut of Healthy Human Adults. EcoSal Plus 2020, 9, 10-1128. [Google Scholar] [CrossRef] [PubMed]
  177. Kaper, J.B.; Nataro, J.P.; Mobley, H.L.T. Pathogenic Escherichia coli. Nat. Rev. Microbiol. 2004, 2, 123–140. [Google Scholar] [CrossRef] [PubMed]
  178. Cerceo, E.; Deitelzweig, S.B.; Sherman, B.M.; Amin, A.N. Multidrug-Resistant Gram-Negative Bacterial Infections in the Hospital Setting: Overview, Implications for Clinical Practice, and Emerging Treatment Options. Microb. Drug Resist. 2016, 22, 412–431. [Google Scholar] [CrossRef]
  179. Farber, J.M.; Peterkin, P.I. Listeria monocytogenes, a food-borne pathogen. Microbiol. Rev. 1991, 55, 476–511. [Google Scholar] [CrossRef]
  180. Disson, O.; Moura, A.; Lecuit, M. Making Sense of the Biodiversity and Virulence of Listeria monocytogenes. Trends Microbiol. 2021, 29, 811–822. [Google Scholar] [CrossRef]
  181. Finn, L.; Onyeaka, H.; O’Neill, S. Listeria monocytogenes Biofilms in Food-Associated Environments: A Persistent Enigma. Foods 2023, 12, 3339. [Google Scholar] [CrossRef]
  182. Rogalla, D.; Bomar, P.A. Listeria Monocytogenes. In StatPearls; StatPearls Publishing LLC.: Treasure Island, FL, USA, 2024. [Google Scholar]
  183. Bouarab Chibane, L.; Degraeve, P.; Ferhout, H.; Bouajila, J.; Oulahal, N. Plant antimicrobial polyphenols as potential natural food preservatives. J. Sci. Food Agric. 2019, 99, 1457–1474. [Google Scholar] [CrossRef]
  184. Matthews, S.J.; Lancaster, J.W. Urinary Tract Infections in the Elderly Population. Am. J. Geriatr. Pharmacother. 2011, 9, 286–309. [Google Scholar] [CrossRef]
  185. Rodriguez-Mañas, L. Urinary tract infections in the elderly: A review of disease characteristics and current treatment options. Drugs Context 2020. [Google Scholar] [CrossRef]
  186. Charkhian, H.; Bodaqlouie, A.; Soleimannezhadbari, E.; Lotfollahi, L.; Shaykh-Baygloo, N.; Hosseinzadeh, R.; Yousefi, N.; Khodayar, M. Comparing the Bacteriostatic Effects of Different Metal Nanoparticles Against Proteus vulgaris. Curr. Microbiol. 2020, 77, 2674–2684. [Google Scholar] [CrossRef] [PubMed]
  187. Buhl, M.; Peter, S.; Willmann, M. Prevalence and risk factors associated with colonization and infection of extensively drug-resistant Pseudomonas aeruginosa: A systematic review. Expert. Rev. Anti Infect. Ther. 2015, 13, 1159–1170. [Google Scholar] [CrossRef]
  188. Paz-Zarza, V.M.; Mangwani-Mordani, S.; Martínez-Maldonado, A.; Álvarez-Hernández, D.; Solano-Gálvez, S.G.; Vázquez-López, R. [Pseudomonas aeruginosa: Pathogenicity and antimicrobial resistance in urinary tract infection]. Rev. Chil. Infectol. 2019, 36, 180–189. [Google Scholar] [CrossRef] [PubMed]
  189. Reynolds, D.; Kollef, M. The Epidemiology and Pathogenesis and Treatment of Pseudomonas aeruginosa Infections: An Update. Drugs 2021, 81, 2117–2131. [Google Scholar] [CrossRef] [PubMed]
  190. Klockgether, J.; Tümmler, B. Recent advances in understanding Pseudomonas aeruginosa as a pathogen. F1000Research 2017, 6, 1261. [Google Scholar] [CrossRef]
  191. Murphy, T.F. Pseudomonas aeruginosa in adults with chronic obstructive pulmonary disease. Curr. Opin. Pulm. Med. 2009, 15, 138–142. [Google Scholar] [CrossRef]
  192. Gonçalves-de-Albuquerque, C.F.; Silva, A.R.; Burth, P.; Rocco, P.R.; Castro-Faria, M.V.; Castro-Faria-Neto, H.C. Possible mechanisms of Pseudomonas aeruginosa-associated lung disease. Int. J. Med. Microbiol. 2016, 306, 20–28. [Google Scholar] [CrossRef]
  193. Silverio, M.P.; Kraychete, G.B.; Rosado, A.S.; Bonelli, R.R. Pseudomonas fluorescens Complex and Its Intrinsic, Adaptive, and Acquired Antimicrobial Resistance Mechanisms in Pristine and Human-Impacted Sites. Antibiotics 2022, 11, 985. [Google Scholar] [CrossRef]
  194. Ishii, H.; Kushima, H.; Koide, Y.; Kinoshita, Y. Pseudomonas fluorescens pneumonia. Int. J. Infect. Dis. 2024, 140, 92–94. [Google Scholar] [CrossRef]
  195. Mitra, S.; Rath, S.; Das, S.; Basu, S. Ocular infection by a psychrophile: Pseudomonas fluorescens. Indian. J. Med. Microbiol. 2019, 37, 289–291. [Google Scholar] [CrossRef]
  196. Montoro-Dasi, L.; Lorenzo-Rebenaque, L.; Marco-Fuertes, A.; Vega, S.; Marin, C. Holistic Strategies to Control Salmonella Infantis: An Emerging Challenge in the European Broiler Sector. Microorganisms 2023, 11, 1765. [Google Scholar] [CrossRef] [PubMed]
  197. Alvarez, D.M.; Barrón-Montenegro, R.; Conejeros, J.; Rivera, D.; Undurraga, E.A.; Moreno-Switt, A.I. A review of the global emergence of multidrug-resistant Salmonella enterica subsp. enterica Serovar Infantis. Int. J. Food Microbiol. 2023, 403, 110297. [Google Scholar] [CrossRef]
  198. Yuan, H.; Liu, F.; Long, J.; Duan, G.; Yang, H. A review on circular RNAs and bacterial infections. Int. J. Biol. Macromol. 2023, 244, 125391. [Google Scholar] [CrossRef] [PubMed]
  199. Hsieh, R.C.; Liu, R.; Burgin, D.J.; Otto, M. Understanding mechanisms of virulence in MRSA: Implications for antivirulence treatment strategies. Expert. Rev. Anti Infect. Ther. 2023, 21, 911–928. [Google Scholar] [CrossRef] [PubMed]
  200. Leung, A.K.C.; Barankin, B.; Leong, K.F. Staphylococcal-scalded skin syndrome: Evaluation, diagnosis, and management. World J. Pediatr. 2018, 14, 116–120. [Google Scholar] [CrossRef]
  201. Lowy, F.D. Staphylococcus aureus infections. N. Engl. J. Med. 1998, 339, 520–532. [Google Scholar] [CrossRef]
  202. Tong, S.Y.; Davis, J.S.; Eichenberger, E.; Holland, T.L.; Fowler, V.G., Jr. Staphylococcus aureus infections: Epidemiology, pathophysiology, clinical manifestations, and management. Clin. Microbiol. Rev. 2015, 28, 603–661. [Google Scholar] [CrossRef]
  203. Periferakis, A.; Periferakis, A.T.; Troumpata, L.; Dragosloveanu, S.; Timofticiuc, I.A.; Georgatos-Garcia, S.; Scheau, A.E.; Periferakis, K.; Caruntu, A.; Badarau, I.A.; et al. Use of Biomaterials in 3D Printing as a Solution to Microbial Infections in Arthroplasty and Osseous Reconstruction. Biomimetics 2024, 9, 154. [Google Scholar] [CrossRef]
  204. Malachowa, N.; DeLeo, F.R. Mobile genetic elements of Staphylococcus aureus. Cell Mol. Life Sci. 2010, 67, 3057–3071. [Google Scholar] [CrossRef]
  205. Cristea, S.; Predescu, V.; Dragosloveanu, S.; Cuculici, S.; Marandici, N. Surgical Approaches for Total Knee Arthroplasty. In Arthroplasty—A Comprehensive Review; Bagaria, V., Ed.; IntechOpen: London, UK, 2016; pp. 25–47. [Google Scholar]
  206. Howden, B.P.; Davies, J.K.; Johnson, P.D.; Stinear, T.P.; Grayson, M.L. Reduced vancomycin susceptibility in Staphylococcus aureus, including vancomycin-intermediate and heterogeneous vancomycin-intermediate strains: Resistance mechanisms, laboratory detection, and clinical implications. Clin. Microbiol. Rev. 2010, 23, 99–139. [Google Scholar] [CrossRef]
  207. Burke, T.L.; Rupp, M.E.; Fey, P.D. Staphylococcus epidermidis. Trends Microbiol. 2023, 31, 763–764. [Google Scholar] [CrossRef] [PubMed]
  208. Armeftis, C.; Ioannou, A.; Lazarou, T.; Giannopoulos, A.; Dimitriadou, E.; Makrides, K.; Pana, Z.D. Staphylococcus epidermidis induced toxic shock syndrome (TSS) secondary to influenza infection. BMC Infect. Dis. 2023, 23, 583. [Google Scholar] [CrossRef] [PubMed]
  209. Brown, M.M.; Horswill, A.R. Staphylococcus epidermidis-Skin friend or foe? PLoS Pathog. 2020, 16, e1009026. [Google Scholar] [CrossRef]
  210. Zheng, Y.; Hunt, R.L.; Villaruz, A.E.; Fisher, E.L.; Liu, R.; Liu, Q.; Cheung, G.Y.C.; Li, M.; Otto, M. Commensal Staphylococcus epidermidis contributes to skin barrier homeostasis by generating protective ceramides. Cell Host Microbe 2022, 30, 301–313.e309. [Google Scholar] [CrossRef]
  211. Severn, M.M.; Horswill, A.R. Staphylococcus epidermidis and its dual lifestyle in skin health and infection. Nat. Rev. Microbiol. 2023, 21, 97–111. [Google Scholar] [CrossRef]
  212. Brown, G.D.; Denning, D.W.; Levitz, S.M. Tackling human fungal infections. Science 2012, 336, 647. [Google Scholar] [CrossRef]
  213. Rodriguez-Tudela, J.L.; Alastruey-Izquierdo, A.; Gago, S.; Cuenca-Estrella, M.; León, C.; Miro, J.M.; Nuñez Boluda, A.; Ruiz Camps, I.; Sole, A.; Denning, D.W. Burden of serious fungal infections in Spain. Clin. Microbiol. Infect. 2015, 21, 183–189. [Google Scholar] [CrossRef]
  214. Alvarez-Moreno, C.A.; Cortes, J.A.; Denning, D.W. Burden of Fungal Infections in Colombia. J. Fungi 2018, 4, 41. [Google Scholar] [CrossRef]
  215. Tufa, T.B.; Denning, D.W. The Burden of Fungal Infections in Ethiopia. J. Fungi 2019, 5, 109. [Google Scholar] [CrossRef]
  216. Fisher, M.C.; Hawkins, N.J.; Sanglard, D.; Gurr, S.J. Worldwide emergence of resistance to antifungal drugs challenges human health and food security. Science 2018, 360, 739–742. [Google Scholar] [CrossRef]
  217. Revie, N.M.; Iyer, K.R.; Robbins, N.; Cowen, L.E. Antifungal drug resistance: Evolution, mechanisms and impact. Curr. Opin. Microbiol. 2018, 45, 70–76. [Google Scholar] [CrossRef] [PubMed]
  218. Ben-Ami, R.; Kontoyiannis, D.P. Resistance to Antifungal Drugs. Infect. Dis. Clin. N. Am. 2021, 35, 279–311. [Google Scholar] [CrossRef]
  219. Matthaiou, E.I.; Sass, G.; Stevens, D.A.; Hsu, J.L. Iron: An essential nutrient for Aspergillus fumigatus and a fulcrum for pathogenesis. Curr. Opin. Infect. Dis. 2018, 31, 506–511. [Google Scholar] [CrossRef] [PubMed]
  220. Greenberger, P.A. Allergic bronchopulmonary aspergillosis. J. Allergy Clin. Immunol. 2002, 110, 685–692. [Google Scholar] [CrossRef]
  221. Amona, F.M.; Oladele, R.O.; Resendiz-Sharpe, A.; Denning, D.W.; Kosmidis, C.; Lagrou, K.; Zhong, H.; Han, L. Triazole resistance in Aspergillus fumigatus isolates in Africa: A systematic review. Med. Mycol. 2022, 60, myac059. [Google Scholar] [CrossRef]
  222. Hurley, R.; De Louvois, J. Candida vaginitis. Postgrad. Med. J. 1979, 55, 645–647. [Google Scholar] [CrossRef]
  223. Sobel, J.D. Vulvovaginal candidosis. Lancet 2007, 369, 1961–1971. [Google Scholar] [CrossRef]
  224. Poulain, D. Candida albicans, plasticity and pathogenesis. Crit. Rev. Microbiol. 2015, 41, 208–217. [Google Scholar] [CrossRef]
  225. Millsop, J.W.; Fazel, N. Oral candidiasis. Clin. Dermatol. 2016, 34, 487–494. [Google Scholar] [CrossRef]
  226. Mayer, F.L.; Wilson, D.; Hube, B. Candida albicans pathogenicity mechanisms. Virulence 2013, 4, 119–128. [Google Scholar] [CrossRef]
  227. Pfaller, M.A.; Diekema, D.J. Epidemiology of invasive candidiasis: A persistent public health problem. Clin. Microbiol. Rev. 2007, 20, 133–163. [Google Scholar] [CrossRef]
  228. Pfaller, M.A.; Diekema, D.J. Epidemiology of invasive mycoses in North America. Crit. Rev. Microbiol. 2010, 36, 1–53. [Google Scholar] [CrossRef]
  229. Pedro Abreu, J.; Esteves, J.; Boncoraglio, M.T.; Pereira, F.M.; Costa, C.; Oliveira, C. Cladosporium herbarum Hot-Tub Lung Hypersensitivity Pneumonitis in a Greenhouse Worker. Eur. J. Case Rep. Intern. Med. 2020, 7, 001565. [Google Scholar] [CrossRef]
  230. Planté-Bordeneuve, T.; Gilbert, O.; Latinne, D.; Bruffaerts, N.; Ghaye, B.; Froidure, A. Familial hypersensitivity pneumonitis triggered by Cladosporium herbarum exposure during carpooling. ERJ Open Res. 2020, 6, 00233-2020. [Google Scholar] [CrossRef] [PubMed]
  231. Breitenbach, M.; Simon-Nobbe, B. The allergens of Cladosporium herbarum and Alternaria alternata. Chem. Immunol. 2002, 81, 48–72. [Google Scholar] [CrossRef] [PubMed]
  232. Bi, K.; Liang, Y.; Mengiste, T.; Sharon, A. Killing softly: A roadmap of Botrytis cinerea pathogenicity. Trends Plant Sci. 2023, 28, 211–222. [Google Scholar] [CrossRef] [PubMed]
  233. Koledenkova, K.; Esmaeel, Q.; Jacquard, C.; Nowak, J.; Clément, C.; Ait Barka, E. Plasmopara viticola the Causal Agent of Downy Mildew of Grapevine: From Its Taxonomy to Disease Management. Front. Microbiol. 2022, 13, 889472. [Google Scholar] [CrossRef]
  234. Fu, Q.; Wang, Y.; Yang, J.; Jiao, Y.; Li, W.; Yang, F.; Yin, X.; Shang, B.; Liu, R.; Zhang, Y.; et al. Plasmopara viticola RxLR effector PvAvh77 triggers cell death and governs immunity responses in grapevine. J. Exp. Bot. 2023, 74, 2047–2066. [Google Scholar] [CrossRef]
  235. Gouveia, C.; Santos, R.B.; Paiva-Silva, C.; Buchholz, G.; Malhó, R.; Figueiredo, A. The pathogenicity of Plasmopara viticola: A review of evolutionary dynamics, infection strategies and effector molecules. BMC Plant Biol. 2024, 24, 327. [Google Scholar] [CrossRef]
  236. Louw, J.P.; Korsten, L. Pathogenic Penicillium spp. on Apple and Pear. Plant Dis. 2014, 98, 590–598. [Google Scholar] [CrossRef]
  237. Rand, T.G.; Giles, S.; Flemming, J.; Miller, J.D.; Puniani, E. Inflammatory and cytotoxic responses in mouse lungs exposed to purified toxins from building isolated Penicillium brevicompactum Dierckx and P. chrysogenum Thom. Toxicol. Sci. 2005, 87, 213–222. [Google Scholar] [CrossRef]
  238. Vincent, M.; Percier, P.; De Prins, S.; Huygen, K.; Potemberg, G.; Muraille, E.; Romano, M.; Michel, O.; Denis, O. Investigation of inflammatory and allergic responses to common mold species: Results from in vitro experiments, from a mouse model of asthma, and from a group of asthmatic patients. Indoor Air 2017, 27, 933–945. [Google Scholar] [CrossRef]
  239. Eldarov, M.A.; Kishkovskaia, S.A.; Tanaschuk, T.N.; Mardanov, A.V. Genomics and Biochemistry of Saccharomyces cerevisiae Wine Yeast Strains. Biochemistry 2016, 81, 1650–1668. [Google Scholar] [CrossRef]
  240. Belda, I.; Ruiz, J.; Santos, A.; Van Wyk, N.; Pretorius, I.S. Saccharomyces cerevisiae. Trends Genet. 2019, 35, 956–957. [Google Scholar] [CrossRef]
  241. Pires, D.; Vicente, C.S.L.; Inácio, M.L.; Mota, M. The Potential of Esteya spp. for the Biocontrol of the Pinewood Nematode, Bursaphelenchus xylophilus. Microorganisms 2022, 10, 168. [Google Scholar] [CrossRef]
  242. Koenning, S.R.; Overstreet, C.; Noling, J.W.; Donald, P.A.; Becker, J.O.; Fortnum, B.A. Survey of crop losses in response to phytoparasitic nematodes in the United States for 1994. J Nematol 1999, 31, 587–618. [Google Scholar]
  243. Yang, H.; Rao, Z. Structural biology of SARS-CoV-2 and implications for therapeutic development. Nat. Rev. Microbiol. 2021, 19, 685–700. [Google Scholar] [CrossRef]
  244. Daia, C.; Scheau, C.; Neagu, G.; Andone, I.; Spanu, A.; Popescu, C.; Stoica, S.I.; Verenca, M.C.; Onose, G. Nerve conduction study and electromyography findings in patients recovering from COVID-19-Case report. Int. J. Infect. Dis. 2021, 103, 420–422. [Google Scholar] [CrossRef]
  245. Fertig, T.E.; Chitoiu, L.; Terinte-Balcan, G.; Peteu, V.-E.; Marta, D.; Gherghiceanu, M. The atomic portrait of SARS-CoV-2 as captured by cryo-electron microscopy. J. Cell. Mol. Med. 2022, 26, 25–34. [Google Scholar] [CrossRef]
  246. Lupașcu, R.E.; Ilie, M.I.; Velescu, B.Ș.; Udeanu, D.I.; Sultana, C.; Ruță, S.; Arsene, A.L. COVID-19-Current Therapeutical Approaches and Future Perspectives. Processes 2022, 10, 1053. [Google Scholar] [CrossRef]
  247. Zamfir, M.A.; Moraru, L.; Dobrea, C.; Scheau, A.E.; Iacob, S.; Moldovan, C.; Scheau, C.; Caruntu, C.; Caruntu, A. Hematologic Malignancies Diagnosed in the Context of the mRNA COVID-19 Vaccination Campaign: A Report of Two Cases. Medicina 2022, 58, 874. [Google Scholar] [CrossRef] [PubMed]
  248. Kirtipal, N.; Bharadwaj, S.; Kang, S.G. From SARS to SARS-CoV-2, insights on structure, pathogenicity and immunity aspects of pandemic human coronaviruses. Infect. Genet. Evol. 2020, 85, 104502. [Google Scholar] [CrossRef]
  249. Santos-López, G.; Cortés-Hernández, P.; Vallejo-Ruiz, V.; Reyes-Leyva, J. SARS-CoV-2: Basic concepts, origin and treatment advances. Gac. Med. Mex. 2021, 157, 84–89. [Google Scholar] [CrossRef] [PubMed]
  250. Hu, B.; Guo, H.; Zhou, P.; Shi, Z.L. Characteristics of SARS-CoV-2 and COVID-19. Nat. Rev. Microbiol. 2021, 19, 141–154. [Google Scholar] [CrossRef]
  251. Talevi, D.; Socci, V.; Carai, M.; Carnaghi, G.; Faleri, S.; Trebbi, E.; Di Bernardo, A.; Capelli, F.; Pacitti, F. Mental health outcomes of the COVID-19 pandemic. Riv. Di Psichiatr. 2020, 55, 137–144. [Google Scholar]
  252. Pedrosa, A.L.; Bitencourt, L.; Fróes, A.C.F.; Cazumbá, M.L.B.; Campos, R.G.B.; de Brito, S.B.C.S.; Simões e Silva, A.C. Emotional, behavioral, and psychological impact of the COVID-19 pandemic. Front. Psychol. 2020, 11, 566212. [Google Scholar] [CrossRef]
  253. Cullen, W.; Gulati, G.; Kelly, B.D. Mental health in the COVID-19 pandemic. QJM Int. J. Med. 2020, 113, 311–312. [Google Scholar] [CrossRef]
  254. Mihailescu, A.; Graur, A.; Ioniță, I.; Styliadis, V.; Serban, T.; Bubulac, L.; Diaconescu, L.; Popa-Velea, O. Associations between personality characteristics and perceived quality of life in medical students during the COVID-19 pandemic. J. Psychosom. Res. 2022, 157, 110883. [Google Scholar] [CrossRef]
  255. Papandreou, A.; Mavrogalou, A.; Periferakis, A.-T.; Periferakis, A.; Badarau, I.A.; Popa-Velea, O.; Scheau, C. The Effects of COVID-19 on the Emotional and Social Stability, Motivation and Attitudes of Gifted and Non-Gifted Children in Greece. Children 2023, 10, 706. [Google Scholar] [CrossRef]
  256. Mihăilescu, A.I.; Popa-Velea, O.; Ciobanu, A.M.; Diaconescu, L.V.; Graur, A.; Ioniţă, I.; Carsote, M. Psychological Factors Associated with General Quality of Life in the Context of COVID-19 Pandemic: A Cross-Sectional Study on a Multicultural Sample of Romanian Medical Students. Healthcare 2024, 12, 1243. [Google Scholar] [CrossRef]
  257. Bourgonje, A.R.; Abdulle, A.E.; Timens, W.; Hillebrands, J.L.; Navis, G.J.; Gordijn, S.J.; Bolling, M.C.; Dijkstra, G.; Voors, A.A.; Osterhaus, A.D.; et al. Angiotensin-converting enzyme 2 (ACE2), SARS-CoV-2 and the pathophysiology of coronavirus disease 2019 (COVID-19). J. Pathol. 2020, 251, 228–248. [Google Scholar] [CrossRef]
  258. Stein, S.R.; Ramelli, S.C.; Grazioli, A.; Chung, J.Y.; Singh, M.; Yinda, C.K.; Winkler, C.W.; Sun, J.; Dickey, J.M.; Ylaya, K.; et al. SARS-CoV-2 infection and persistence in the human body and brain at autopsy. Nature 2022, 612, 758–763. [Google Scholar] [CrossRef]
  259. Jackson, C.B.; Farzan, M.; Chen, B.; Choe, H. Mechanisms of SARS-CoV-2 entry into cells. Nat. Rev. Mol. Cell Biol. 2022, 23, 3–20. [Google Scholar] [CrossRef] [PubMed]
  260. Andrews, H.S.; Herman, J.D.; Gandhi, R.T. Treatments for COVID-19. Annu. Rev. Med. 2024, 75, 145–157. [Google Scholar] [CrossRef]
  261. Naseem, A.; Rasool, F.; Ahmed, A.; Carter, W.G. The Potential of Stilbene Compounds to Inhibit M(pro) Protease as a Natural Treatment Strategy for Coronavirus Disease-2019. Curr. Issues Mol. Biol. 2022, 45, 12–32. [Google Scholar] [CrossRef]
  262. Li, Y.Q.; Li, Z.L.; Zhao, W.J.; Wen, R.X.; Meng, Q.W.; Zeng, Y. Synthesis of stilbene derivatives with inhibition of SARS coronavirus replication. Eur. J. Med. Chem. 2006, 41, 1084–1089. [Google Scholar] [CrossRef]
  263. Chen, M.-K.; Liu, Y.-T.; Lin, J.-T.; Lin, C.-C.; Chuang, Y.-C.; Lo, Y.-S.; Hsi, Y.-T.; Hsieh, M.-J. Pinosylvin reduced migration and invasion of oral cancer carcinoma by regulating matrix metalloproteinase-2 expression and extracellular signal-regulated kinase pathway. Biomed. Pharmacother. 2019, 117, 109160. [Google Scholar] [CrossRef]
  264. Perri, M.R.; Pellegrino, M.; Marrelli, M.; Aquaro, S.; Cavaliere, F.; Grande, F.; Occhiuzzi, M.A.; Lupia, C.; Toma, C.C.; Conforti, F.; et al. Identification of Pinosylvin in Pinus nigra subsp. laricio: A Naturally Occurring Stilbenoid Suppressing LPS-Induced Expression of Pro-Inflammatory Cytokines and Mediators and Inhibiting the JAK/STAT Signaling Pathway. Pharmaceuticals 2023, 16, 718. [Google Scholar] [CrossRef]
  265. Macickova, T.; Drabikova, K.; Nosal, R.; Bauerova, K.; Mihalova, D.; Harmatha, J.; Pecivova, J. In vivo effect of pinosylvin and pterostilbene in the animal model of adjuvant arthritis. Neuro Endocrinol. Lett. 2010, 31 (Suppl. 2), 91–95. [Google Scholar]
  266. Park, E.J.; Min, H.Y.; Chung, H.J.; Ahn, Y.H.; Pyee, J.H.; Lee, S.K. Pinosylvin suppresses LPS-stimulated inducible nitric oxide synthase expression via the MyD88-independent, but TRIF-dependent downregulation of IRF-3 signaling pathway in mouse macrophage cells. Cell Physiol. Biochem. 2011, 27, 353–362. [Google Scholar] [CrossRef]
  267. Jančinová, V.; Perečko, T.; Nosáľ, R.; Harmatha, J.; Smidrkal, J.; Drábiková, K. The natural stilbenoid pinosylvin and activated neutrophils: Effects on oxidative burst, protein kinase C, apoptosis and efficiency in adjuvant arthritis. Acta Pharmacol. Sin. 2012, 33, 1285–1292. [Google Scholar] [CrossRef] [PubMed]
  268. Jeong, E.; Lee, H.R.; Pyee, J.; Park, H. Pinosylvin induces cell survival, migration and anti-adhesiveness of endothelial cells via nitric oxide production. Phytother. Res. 2012, 27, 610–617. [Google Scholar] [CrossRef]
  269. Drafi, F.; Bauerova, K.; Kuncirova, V.; Ponist, S.; Mihalova, D.; Fedorova, T.; Harmatha, J.; Nosal, R. Pharmacological influence on processes of adjuvant arthritis: Effect of the combination of an antioxidant active substance with methotrexate. Interdiscip. Toxicol. 2012, 5, 84–91. [Google Scholar] [CrossRef] [PubMed]
  270. Bauerova, K.; Acquaviva, A.; Ponist, S.; Gardi, C.; Vecchio, D.; Drafi, F.; Arezzini, B.; Bezakova, L.; Kuncirova, V.; Mihalova, D.; et al. Markers of inflammation and oxidative stress studied in adjuvant-induced arthritis in the rat on systemic and local level affected by pinosylvin and methotrexate and their combination. Autoimmunity 2015, 48, 46–56. [Google Scholar] [CrossRef]
  271. Stojanović, S.; Brede, O. Elementary reactions of the antioxidant action of trans-stilbene derivatives: Resveratrol, pinosylvin and 4-hydroxystilbene. Phys. Chem. Chem. Phys. 2002, 4, 757–764. [Google Scholar] [CrossRef]
  272. Rodríguez-Bonilla, P.; Gandía-Herrero, F.; Matencio, A.; García-Carmona, F.; López-Nicolás, J.M. Comparative Study of the Antioxidant Capacity of Four Stilbenes Using ORAC, ABTS+, and FRAP Techniques. Food Anal. Methods 2017, 10, 2994–3000. [Google Scholar] [CrossRef]
  273. Król, M.; Kepinska, M. Human Nitric Oxide Synthase-Its Functions, Polymorphisms, and Inhibitors in the Context of Inflammation, Diabetes and Cardiovascular Diseases. Int. J. Mol. Sci. 2020, 22, 56. [Google Scholar] [CrossRef]
  274. Förstermann, U.; Sessa, W.C. Nitric oxide synthases: Regulation and function. Eur. Heart J. 2012, 33, 829–837. [Google Scholar] [CrossRef]
  275. Feng, Q.; Hedner, T. Endothelium-derived relaxing factor (EDRF) and nitric oxide (NO). II. Physiology, pharmacology and pathophysiological implications. Clin. Physiol. 1990, 10, 503–526. [Google Scholar] [CrossRef]
  276. Naseem, K.M. The role of nitric oxide in cardiovascular diseases. Mol. Asp. Med. 2005, 26, 33–65. [Google Scholar] [CrossRef]
  277. Patel, R.P.; McAndrew, J.; Sellak, H.; White, C.R.; Jo, H.; Freeman, B.A.; Darley-Usmar, V.M. Biological aspects of reactive nitrogen species. Biochim. Biophys. Acta (BBA) Bioenerg. 1999, 1411, 385–400. [Google Scholar] [CrossRef]
  278. Ullah, M.O.; Sweet, M.J.; Mansell, A.; Kellie, S.; Kobe, B. TRIF-dependent TLR signaling, its functions in host defense and inflammation, and its potential as a therapeutic target. J. Leukoc. Biol. 2016, 100, 27–45. [Google Scholar] [CrossRef]
  279. Slater, S.J.; Seiz, J.L.; Cook, A.C.; Stagliano, B.A.; Buzas, C.J. Inhibition of protein kinase C by resveratrol. Biochim. Biophys. Acta 2003, 1637, 59–69. [Google Scholar] [CrossRef] [PubMed]
  280. Gopalakrishna, R.; Jaken, S. Protein kinase C signaling and oxidative stress. Free Radic. Biol. Med. 2000, 28, 1349–1361. [Google Scholar] [CrossRef] [PubMed]
  281. Giorgi, C.; Agnoletto, C.; Baldini, C.; Bononi, A.; Bonora, M.; Marchi, S.; Missiroli, S.; Patergnani, S.; Poletti, F.; Rimessi, A. Redox control of protein kinase C: Cell-and disease-specific aspects. Antioxid. Redox Signal. 2010, 13, 1051–1085. [Google Scholar] [CrossRef]
  282. Koskela, A.; Reinisalo, M.; Hyttinen, J.M.; Kaarniranta, K.; Karjalainen, R.O. Pinosylvin-mediated protection against oxidative stress in human retinal pigment epithelial cells. Mol. Vis. 2014, 20, 760–769. [Google Scholar]
  283. Wegiel, B.; Nemeth, Z.; Correa-Costa, M.; Bulmer, A.C.; Otterbein, L.E. Heme oxygenase-1: A metabolic nike. Antioxid. Redox Signal 2014, 20, 1709–1722. [Google Scholar] [CrossRef]
  284. Flück, C.E.; Rojas Velazquez, M.N.; Pandey, A.V. Chapter 12—P450 oxidoreductase deficiency. In Genetic Steroid Disorders, 2nd ed.; New, M.I., Ed.; Academic Press: San Diego, CA, USA, 2023; pp. 239–264. [Google Scholar] [CrossRef]
  285. Swan, R.; Kim, S.J.; Campbell, J.P.; Chan, R.V.P.; Sonmez, K.; Taylor, K.D.; Li, X.; Chen, Y.-D.I.; Rotter, J.I.; Simmons, C.; et al. The Genetics of Retinopathy of Prematurity: A Model for Neovascular Retinal Disease. Ophthalmol. Retin. 2018, 2, 949–962. [Google Scholar] [CrossRef]
  286. Reinisalo, M.; Kårlund, A.; Koskela, A.; Kaarniranta, K.; Karjalainen, R.O. Polyphenol Stilbenes: Molecular Mechanisms of Defence against Oxidative Stress and Aging-Related Diseases. Oxidative Med. Cell. Longev. 2015, 2015, 340520. [Google Scholar] [CrossRef]
  287. Dragosloveanu, C.D.M.; Potop, V.; Coviltir, V.; Dinu, V.; Păsărică, M.; Ducan, I.L.; Maier, C.; Dragosloveanu, Ş. Prematurity-Risk Factor or Coincidence in Congenital Glaucoma? Medicina 2022, 58, 334. [Google Scholar] [CrossRef]
  288. Srejovic, J.V.; Muric, M.D.; Jakovljevic, V.L.; Srejovic, I.M.; Sreckovic, S.B.; Petrovic, N.T.; Todorovic, D.Z.; Bolevich, S.B.; Sarenac Vulovic, T.S. Molecular and Cellular Mechanisms Involved in the Pathophysiology of Retinal Vascular Disease-Interplay Between Inflammation and Oxidative Stress. Int. J. Mol. Sci. 2024, 25, 11850. [Google Scholar] [CrossRef]
  289. Dragosloveanu, C.D.M.; Celea, C.G.; Dragosloveanu, Ş. Comparison of 360° circumferential trabeculotomy and conventional trabeculotomy in primary pediatric glaucoma surgery: Complications, reinterventions and preoperative predictive risk factors. Int. Ophthalmol. 2020, 40, 3547–3554. [Google Scholar] [CrossRef]
  290. Su, E.; Kesavamoorthy, N.; Junge, J.A.; Zheng, M.; Craft, C.M.; Ameri, H. Comparison of Retinal Metabolic Activity and Structural Development between rd10 Mice and Normal Mice Using Multiphoton Fluorescence Lifetime Imaging Microscopy. Curr. Issues Mol. Biol. 2024, 46, 612–620. [Google Scholar] [CrossRef]
  291. Wang, C.N.; Sang, M.M.; Gong, S.N.; Yang, J.F.; Cheng, C.Y.; Sun, F. Two resveratrol analogs, pinosylvin and 4,4′-dihydroxystilbene, improve oligoasthenospermia in a mouse model by attenuating oxidative stress via the Nrf2-ARE pathway. Bioorg. Chem. 2020, 104, 104295. [Google Scholar] [CrossRef]
  292. Wang, Z.; Zhou, Z.; Zhang, L.; Li, X.; Li, M.; Pan, Y.; Jiao, T.; Shi, X.; Liu, Q.; Wang, C.; et al. Efficacy and safety of nonpharmacological strategies for the treatment of oligoasthenospermia: A systematic review and Bayesian network meta-analysis. Eur. J. Med. Res. 2023, 28, 6. [Google Scholar] [CrossRef]
  293. Buendia, I.; Michalska, P.; Navarro, E.; Gameiro, I.; Egea, J.; León, R. Nrf2-ARE pathway: An emerging target against oxidative stress and neuroinflammation in neurodegenerative diseases. Pharmacol. Ther. 2016, 157, 84–104. [Google Scholar] [CrossRef] [PubMed]
  294. Bauerová, K.; Ponist, S.; Ondrejicková, O.; Komendová, D.; Mihalová, D. Association between tissue gamma-glutamyl-transferase and clinical markers of adjuvant arthritis in Lewis rats. Neuro Endocrinol. Lett. 2006, 27 (Suppl. 2), 172–175. [Google Scholar]
  295. Drábiková, K.; Nosal, R.; Bauerova, K.; Mihalová, D.; Jancinová, V.; Petríková, M.; Košťálová, D. Chemiluminescence as a measure of oxidative stress in paws and spleen of rats with adjuvant arthritis. Chem. Listy 2007, 101, s180–s182. [Google Scholar]
  296. Jančinová, V.; Petríková, M.; Perečko, T.; Drábiková, K.; Nosáľ, R.; Bauerová, K.; Poništ, S.; Košťálová, D. Inhibition of neutrophil oxidative burst with arbutin. Effects in vitro and in adjuvant arthritis. Chem. Listy 2007, 101, 189–191. [Google Scholar]
  297. Park, E.J.; Min, H.Y.; Ahn, Y.H.; Bae, C.M.; Pyee, J.H.; Lee, S.K. Synthesis and inhibitory effects of pinosylvin derivatives on prostaglandin E2 production in lipopolysaccharide-induced mouse macrophage cells. Bioorg. Med. Chem. Lett. 2004, 14, 5895–5898. [Google Scholar] [CrossRef]
  298. Lee, S.K.; Min, H.Y.; Huh, S.K.; Kim, E.Y.; Lee, E.; Song, S.; Kim, S. Styrylheterocycles: A novel class of inhibitors on lipopolysaccharide-Induced nitric oxide production. Bioorg. Med. Chem. Lett. 2003, 13, 3689–3692. [Google Scholar] [CrossRef] [PubMed]
  299. Vane, J.R.; Bakhle, Y.S.; Botting, R.M. Cyclooxygenases 1 and 2. Annu. Rev. Pharmacol. Toxicol. 1998, 38, 97–120. [Google Scholar] [CrossRef]
  300. Hla, T.; Bishop-Bailey, D.; Liu, C.H.; Schaefers, H.J.; Trifan, O.C. Cyclooxygenase-1 and -2 isoenzymes. Int. J. Biochem. Cell Biol. 1999, 31, 551–557. [Google Scholar] [CrossRef]
  301. Adelizzi, R.A. COX-1 and COX-2 in health and disease. J. Am. Osteopath. Assoc. 1999, 99, S7–S12. [Google Scholar] [CrossRef] [PubMed]
  302. Chandrasekharan, N.V.; Dai, H.; Roos, K.L.; Evanson, N.K.; Tomsik, J.; Elton, T.S.; Simmons, D.L. COX-3, a cyclooxygenase-1 variant inhibited by acetaminophen and other analgesic/antipyretic drugs: Cloning, structure, and expression. Proc. Natl. Acad. Sci. USA 2002, 99, 13926–13931. [Google Scholar] [CrossRef]
  303. Ju, Z.; Li, M.; Xu, J.; Howell, D.C.; Li, Z.; Chen, F.E. Recent development on COX-2 inhibitors as promising anti-inflammatory agents: The past 10 years. Acta Pharm. Sin. B 2022, 12, 2790–2807. [Google Scholar] [CrossRef]
  304. Zarghi, A.; Arfaei, S. Selective COX-2 Inhibitors: A Review of Their Structure-Activity Relationships. Iran. J. Pharm. Res. 2011, 10, 655–683. [Google Scholar]
  305. Ricciotti, E.; FitzGerald, G.A. Prostaglandins and inflammation. Arter. Thromb. Vasc. Biol. 2011, 31, 986–1000. [Google Scholar] [CrossRef]
  306. Lee, J.; Jung, E.; Lim, J.; Lee, J.; Hur, S.; Kim, S.S.; Lim, S.; Hyun, C.G.; Kim, Y.S.; Park, D. Involvement of nuclear factor-kappaB in the inhibition of pro-inflammatory mediators by pinosylvin. Planta Med. 2006, 72, 801–806. [Google Scholar] [CrossRef]
  307. Yamamoto, K.; Arakawa, T.; Ueda, N.; Yamamoto, S. Transcriptional roles of nuclear factor kappa B and nuclear factor-interleukin-6 in the tumor necrosis factor alpha-dependent induction of cyclooxygenase-2 in MC3T3-E1 cells. J. Biol. Chem. 1995, 270, 31315–31320. [Google Scholar] [CrossRef]
  308. Deng, J.; Miller, S.A.; Wang, H.Y.; Xia, W.; Wen, Y.; Zhou, B.P.; Li, Y.; Lin, S.Y.; Hung, M.C. beta-catenin interacts with and inhibits NF-kappa B in human colon and breast cancer. Cancer Cell 2002, 2, 323–334. [Google Scholar] [CrossRef]
  309. Haeggström, J.Z. Leukotriene biosynthetic enzymes as therapeutic targets. J. Clin. Investig. 2018, 128, 2680–2690. [Google Scholar] [CrossRef]
  310. Tanaka, T.; Narazaki, M.; Kishimoto, T. IL-6 in inflammation, immunity, and disease. Cold Spring Harb. Perspect. Biol. 2014, 6, a016295. [Google Scholar] [CrossRef] [PubMed]
  311. Deshmane, S.L.; Kremlev, S.; Amini, S.; Sawaya, B.E. Monocyte chemoattractant protein-1 (MCP-1): An overview. J. Interferon Cytokine Res. 2009, 29, 313–326. [Google Scholar] [CrossRef]
  312. Eräsalo, H.; Hämäläinen, M.; Leppänen, T.; Mäki-Opas, I.; Laavola, M.; Haavikko, R.; Yli-Kauhaluoma, J.; Moilanen, E. Natural Stilbenoids Have Anti-Inflammatory Properties in Vivo and Down-Regulate the Production of Inflammatory Mediators NO, IL6, and MCP1 Possibly in a PI3K/Akt-Dependent Manner. J. Nat. Prod. 2018, 81, 1131–1142. [Google Scholar] [CrossRef]
  313. Banerjee, S.; Biehl, A.; Gadina, M.; Hasni, S.; Schwartz, D.M. JAK-STAT Signaling as a Target for Inflammatory and Autoimmune Diseases: Current and Future Prospects. Drugs 2017, 77, 521–546. [Google Scholar] [CrossRef] [PubMed]
  314. Sarapultsev, A.; Gusev, E.; Komelkova, M.; Utepova, I.; Luo, S.; Hu, D. JAK-STAT signaling in inflammation and stress-related diseases: Implications for therapeutic interventions. Mol. Biomed. 2023, 4, 40. [Google Scholar] [CrossRef]
  315. Moilanen, L.J.; Hämäläinen, M.; Lehtimäki, L.; Nieminen, R.M.; Muraki, K.; Moilanen, E. Pinosylvin Inhibits TRPA1-Induced Calcium Influx In Vitro and TRPA1-Mediated Acute Paw Inflammation In Vivo. Basic. Clin. Pharmacol. Toxicol. 2016, 118, 238–242. [Google Scholar] [CrossRef]
  316. Namer, B.; Seifert, F.; Handwerker, H.O.; Maihöfner, C. TRPA1 and TRPM8 activation in humans: Effects of cinnamaldehyde and menthol. Neuroreport 2005, 16, 955–959. [Google Scholar] [CrossRef]
  317. Schütz, M.; Oertel, B.G.; Heimann, D.; Doehring, A.; Walter, C.; Dimova, V.; Geisslinger, G.; Lötsch, J. Consequences of a human TRPA1 genetic variant on the perception of nociceptive and olfactory stimuli. PLoS ONE 2014, 9, e95592. [Google Scholar] [CrossRef]
  318. Kremeyer, B.; Lopera, F.; Cox, J.J.; Momin, A.; Rugiero, F.; Marsh, S.; Woods, C.G.; Jones, N.G.; Paterson, K.J.; Fricker, F.R.; et al. A gain-of-function mutation in TRPA1 causes familial episodic pain syndrome. Neuron 2010, 66, 671–680. [Google Scholar] [CrossRef] [PubMed]
  319. Kivimäki, K.; Leppänen, T.; Hämäläinen, M.; Vuolteenaho, K.; Moilanen, E. Pinosylvin Shifts Macrophage Polarization to Support Resolution of Inflammation. Molecules 2021, 26, 2772. [Google Scholar] [CrossRef]
  320. Kwon, O.; Seo, Y.; Park, H. Pinosylvin exacerbates LPS-induced apoptosis via ALOX 15 upregulation in leukocytes. BMB Rep. 2018, 51, 302–307. [Google Scholar] [CrossRef]
  321. Modi, S.; Yaluri, N.; Kokkola, T. Strigolactone GR24 and pinosylvin attenuate adipogenesis and inflammation of white adipocytes. Biochem. Biophys. Res. Commun. 2018, 499, 164–169. [Google Scholar] [CrossRef] [PubMed]
  322. Villapol, S. Roles of Peroxisome Proliferator-Activated Receptor Gamma on Brain and Peripheral Inflammation. Cell Mol. Neurobiol. 2018, 38, 121–132. [Google Scholar] [CrossRef]
  323. Liu, Y.; Wang, J.; Luo, S.; Zhan, Y.; Lu, Q. The roles of PPARγ and its agonists in autoimmune diseases: A comprehensive review. J. Autoimmun. 2020, 113, 102510. [Google Scholar] [CrossRef]
  324. Moseti, D.; Regassa, A.; Kim, W.K. Molecular Regulation of Adipogenesis and Potential Anti-Adipogenic Bioactive Molecules. Int. J. Mol. Sci. 2016, 17, 124. [Google Scholar] [CrossRef]
  325. Tao, L.L.; Zhai, Y.Z.; Ding, D.; Yin, W.H.; Liu, X.P.; Yu, G.Y. The role of C/EBP-α expression in human liver and liver fibrosis and its relationship with autophagy. Int. J. Clin. Exp. Pathol. 2015, 8, 13102–13107. [Google Scholar]
  326. Koleva, R.I.; Ficarro, S.B.; Radomska, H.S.; Carrasco-Alfonso, M.J.; Alberta, J.A.; Webber, J.T.; Luckey, C.J.; Marcucci, G.; Tenen, D.G.; Marto, J.A. C/EBPα and DEK coordinately regulate myeloid differentiation. Blood 2012, 119, 4878–4888. [Google Scholar] [CrossRef]
  327. Alvarado, M.G.; Thakore, P.; Earley, S. Transient Receptor Potential Channel Ankyrin 1: A Unique Regulator of Vascular Function. Cells 2021, 10, 1167. [Google Scholar] [CrossRef]
  328. Hu, Z.; Zhang, Y.; Yu, W.; Li, J.; Yao, J.; Zhang, J.; Wang, J.; Wang, C. Transient receptor potential ankyrin 1 (TRPA1) modulators: Recent update and future perspective. Eur. J. Med. Chem. 2023, 257, 115392. [Google Scholar] [CrossRef]
  329. White, M.V.; Kaliner, M.A. Mediators of allergic rhinitis. J. Allergy Clin. Immunol. 1992, 90, 699–704. [Google Scholar] [CrossRef] [PubMed]
  330. Kubo, T.; Morita, H.; Sugita, K.; Akdis, C.A. Chapter 1—Introduction to Mechanisms of Allergic Diseases. In Middleton’s Allergy Essentials; O’Hehir, R.E., Holgate, S.T., Sheikh, A., Eds.; Elsevier: Amsterdam, The Netherlands, 2017; pp. 1–27. [Google Scholar] [CrossRef]
  331. Modi, S.; Yaluri, N.; Kokkola, T.; Laakso, M. Plant-derived compounds strigolactone GR24 and pinosylvin activate SIRT1 and enhance glucose uptake in rat skeletal muscle cells. Sci. Rep. 2017, 7, 17606. [Google Scholar] [CrossRef] [PubMed]
  332. Sirerol, J.A.; Rodríguez, M.L.; Mena, S.; Asensi, M.A.; Estrela, J.M.; Ortega, A.L. Role of Natural Stilbenes in the Prevention of Cancer. Oxid. Med. Cell Longev. 2016, 2016, 3128951. [Google Scholar] [CrossRef]
  333. De Filippis, B.; Ammazzalorso, A.; Fantacuzzi, M.; Giampietro, L.; Maccallini, C.; Amoroso, R. Anticancer Activity of Stilbene-Based Derivatives. ChemMedChem 2017, 12, 558–570. [Google Scholar] [CrossRef]
  334. Yousuf, M.; Jinka, S.; Adhikari, S.S.; Banerjee, R. Methoxy-enriched cationic stilbenes as anticancer therapeutics. Bioorg. Chem. 2020, 98, 103719. [Google Scholar] [CrossRef]
  335. Borys, F.; Tobiasz, P.; Poterała, M.; Fabczak, H.; Krawczyk, H.; Joachimiak, E. Systematic Studies on Anti-Cancer Evaluation of Stilbene and Dibenzo[b,f]oxepine Derivatives. Molecules 2023, 28, 3558. [Google Scholar] [CrossRef]
  336. Scheau, C.; Mihai, L.G.; Badarau, I.A.; Caruntu, C. Emerging applications of some important natural compounds in the field of oncology. Farmacia 2020, 68, 984–991. [Google Scholar] [CrossRef]
  337. Duta-Bratu, C.-G.; Nitulescu, G.M.; Mihai, D.P.; Olaru, O.T. Resveratrol and Other Natural Oligomeric Stilbenoid Compounds and Their Therapeutic Applications. Plants 2023, 12, 2935. [Google Scholar] [CrossRef]
  338. Hassan, A.H.E.; Wang, C.Y.; Oh, T.; Ham, G.; Lee, S.K.; Lee, Y.S. Discovery of a stilbenoid-flavanone hybrid as an antitumor Wnt/β-catenin signaling pathway inhibitor. Bioorganic Chem. 2024, 145, 107178. [Google Scholar] [CrossRef]
  339. Skinnider, L.; Stoessl, A. The effect of the phytoalexins, lubimin, (−)-maackiain, pinosylvin, and the related compounds dehydroloroglossol and hordatine M on human lymphoblastoid cell lines. Experientia 1986, 42, 568–570. [Google Scholar] [CrossRef] [PubMed]
  340. Park, E.J.; Chung, H.J.; Park, H.J.; Kim, G.D.; Ahn, Y.H.; Lee, S.K. Suppression of Src/ERK and GSK-3/β-catenin signaling by pinosylvin inhibits the growth of human colorectal cancer cells. Food Chem. Toxicol. 2013, 55, 424–433. [Google Scholar] [CrossRef] [PubMed]
  341. Ketola, K.; Viitala, M.; Kohonen, P.; Fey, V.; Culig, Z.; Kallioniemi, O.; Iljin, K. High-throughput cell-based compound screen identifies pinosylvin methyl ether and tanshinone IIA as inhibitors of castration-resistant prostate cancer. J. Mol. Biochem. 2016, 5, 12–22. [Google Scholar]
  342. Pelaz, S.G.; Tabernero, A. Src: Coordinating metabolism in cancer. Oncogene 2022, 41, 4917–4928. [Google Scholar] [CrossRef]
  343. McCubrey, J.A.; Steelman, L.S.; Bertrand, F.E.; Davis, N.M.; Abrams, S.L.; Montalto, G.; D’Assoro, A.B.; Libra, M.; Nicoletti, F.; Maestro, R.; et al. Multifaceted roles of GSK-3 and Wnt/β-catenin in hematopoiesis and leukemogenesis: Opportunities for therapeutic intervention. Leukemia 2014, 28, 15–33. [Google Scholar] [CrossRef]
  344. Mellanen, P.; Petänen, T.; Lehtimäki, J.; Mäkelä, S.; Bylund, G.; Holmbom, B.; Mannila, E.; Oikari, A.; Santti, R. Wood-derived estrogens: Studies in vitro with breast cancer cell lines and in vivo in trout. Toxicol. Appl. Pharmacol. 1996, 136, 381–388. [Google Scholar] [CrossRef]
  345. Hua, H.; Zhang, H.; Kong, Q.; Jiang, Y. Mechanisms for estrogen receptor expression in human cancer. Exp. Hematol. Oncol. 2018, 7, 24. [Google Scholar] [CrossRef]
  346. Tanwar, A.K.; Dhiman, N.; Kumar, A.; Jaitak, V. Engagement of phytoestrogens in breast cancer suppression: Structural classification and mechanistic approach. Eur. J. Med. Chem. 2021, 213, 113037. [Google Scholar] [CrossRef]
  347. Park, E.J.; Park, H.J.; Chung, H.J.; Shin, Y.; Min, H.Y.; Hong, J.Y.; Kang, Y.J.; Ahn, Y.H.; Pyee, J.H.; Lee, S.K. Antimetastatic activity of pinosylvin, a natural stilbenoid, is associated with the suppression of matrix metalloproteinases. J. Nutr. Biochem. 2012, 23, 946–952. [Google Scholar] [CrossRef]
  348. Chuang, Y.C.; Hsieh, M.C.; Lin, C.C.; Lo, Y.S.; Ho, H.Y.; Hsieh, M.J.; Lin, J.T. Pinosylvin inhibits migration and invasion of nasopharyngeal carcinoma cancer cells via regulation of epithelial-mesenchymal transition and inhibition of MMP-2. Oncol. Rep. 2021, 46, 143. [Google Scholar] [CrossRef]
  349. Sun, J.; Nan, G. The extracellular signal-regulated kinase 1/2 pathway in neurological diseases: A potential therapeutic target (Review). Int. J. Mol. Med. 2017, 39, 1338–1346. [Google Scholar] [CrossRef] [PubMed]
  350. Gao, Y.; Moten, A.; Lin, H.-K. Akt: A new activation mechanism. Cell Res. 2014, 24, 785–786. [Google Scholar] [CrossRef]
  351. Berr, A.L.; Wiese, K.; dos Santos, G.; Koch, C.M.; Anekalla, K.R.; Kidd, M.; Davis, J.M.; Cheng, Y.; Hu, Y.-S.; Ridge, K.M. Vimentin is required for tumor progression and metastasis in a mouse model of non–small cell lung cancer. Oncogene 2023, 42, 2074–2087. [Google Scholar] [CrossRef]
  352. Kaszak, I.; Witkowska-Piłaszewicz, O.; Niewiadomska, Z.; Dworecka-Kaszak, B.; Ngosa Toka, F.; Jurka, P. Role of Cadherins in Cancer-A Review. Int. J. Mol. Sci. 2020, 21, 7624. [Google Scholar] [CrossRef]
  353. Ram, A.K.; Vairappan, B. Role of zonula occludens in gastrointestinal and liver cancers. World J. Clin. Cases 2022, 10, 3647–3661. [Google Scholar] [CrossRef]
  354. Du, J.; Zhao, Y.; Hu, D.; Li, H.; Gao, L.; Xing, X.; Shi, K. Pinosylvin Inhibits Esophageal Squamous Cell Carcinoma Migration and Invasion by Regulating STX6/ITGA3/VASP Pathway. Rev. Bras. De Farmacogn. 2023, 33, 173–181. [Google Scholar] [CrossRef]
  355. Du, J.; Liu, X.; Wu, Y.; Zhu, J.; Tang, Y. Essential role of STX6 in esophageal squamous cell carcinoma growth and migration. Biochem. Biophys. Res. Commun. 2016, 472, 60–67. [Google Scholar] [CrossRef]
  356. Simard, F.; Legault, J.; Lavoie, S.; Mshvildadze, V.; Pichette, A. Isolation and identification of cytotoxic compounds from the wood of Pinus resinosa. Phytother. Res. 2008, 22, 919–922. [Google Scholar] [CrossRef]
  357. Song, J.; Seo, Y.; Park, H. Pinosylvin enhances leukemia cell death via down-regulation of AMPKα expression. Phytother. Res. 2018, 32, 2097–2104. [Google Scholar] [CrossRef]
  358. Steinberg, G.R.; Carling, D. AMP-activated protein kinase: The current landscape for drug development. Nat. Rev. Drug Discov. 2019, 18, 527–551. [Google Scholar] [CrossRef]
  359. Park, E.-J.; Min, H.-Y.; Kim, M.-S.; Pyee, J.-H.; Ahn, Y.-H.; Lee, S.-K. Suppressive effects of pinosylvin on prostaglandin E2 and nitric oxide production in lipopolysaccharide-stimulated RAW 264.7 cells. In Proceedings of the Annual Meeting of KSAP: International Symposium on Pharmaceutical and Biomedical Sciences on Obesity, Seoul, Republic of Korea, 1 November 2003; p. 102. [Google Scholar]
  360. Marambaud, P.; Zhao, H.; Davies, P. Resveratrol Promotes Clearance of Alzheimer’s Disease Amyloid-β Peptides*. J. Biol. Chem. 2005, 280, 37377–37382. [Google Scholar] [CrossRef]
  361. Lu, K.T.; Ko, M.C.; Chen, B.Y.; Huang, J.C.; Hsieh, C.W.; Lee, M.C.; Chiou, R.Y.; Wung, B.S.; Peng, C.H.; Yang, Y.L. Neuroprotective effects of resveratrol on MPTP-induced neuron loss mediated by free radical scavenging. J. Agric. Food Chem. 2008, 56, 6910–6913. [Google Scholar] [CrossRef] [PubMed]
  362. Zhang, F.; Shi, J.S.; Zhou, H.; Wilson, B.; Hong, J.S.; Gao, H.M. Resveratrol protects dopamine neurons against lipopolysaccharide-induced neurotoxicity through its anti-inflammatory actions. Mol. Pharmacol. 2010, 78, 466–477. [Google Scholar] [CrossRef]
  363. Temsamani, H.; Krisa, S.; Mérillon, J.-M.; Richard, T. Promising neuroprotective effects of oligostilbenes. Nutr. Aging 2015, 3, 49–54. [Google Scholar] [CrossRef]
  364. Xu, H.; Deng, R.; Li, E.T.S.; Shen, J.; Wang, M. Pinosylvin provides neuroprotection against cerebral ischemia and reperfusion injury through enhancing PINK1/Parkin mediated mitophagy and Nrf2 pathway. J. Funct. Foods 2020, 71, 104019. [Google Scholar] [CrossRef]
  365. Lee, R.H.C.; Lee, M.H.H.; Wu, C.Y.C.; Couto, E.S.A.; Possoit, H.E.; Hsieh, T.H.; Minagar, A.; Lin, H.W. Cerebral ischemia and neuroregeneration. Neural Regen. Res. 2018, 13, 373–385. [Google Scholar] [CrossRef]
  366. Kunz, A.; Dirnagl, U.; Mergenthaler, P. Acute pathophysiological processes after ischaemic and traumatic brain injury. Best Pract. Res. Clin. Anaesthesiol. 2010, 24, 495–509. [Google Scholar] [CrossRef]
  367. Xing, C.; Arai, K.; Lo, E.H.; Hommel, M. Pathophysiologic cascades in ischemic stroke. Int. J. Stroke 2012, 7, 378–385. [Google Scholar] [CrossRef] [PubMed]
  368. Gorczyca, W.; Traganos, F.; Jesionowska, H.; Darzynkiewicz, Z. Presence of DNA Strand Breaks and Increased Sensitivity of DNA in Situ to Denaturation in Abnormal Human Sperm Cells: Analogy to Apoptosis of Somatic Cells. Exp. Cell Res. 1993, 207, 202–205. [Google Scholar] [CrossRef]
  369. Moro, M.A.; Almeida, A.; Bolaños, J.P.; Lizasoain, I. Mitochondrial respiratory chain and free radical generation in stroke. Free Radic. Biol. Med. 2005, 39, 1291–1304. [Google Scholar] [CrossRef]
  370. Chan, P.H. Mitochondria and neuronal death/survival signaling pathways in cerebral ischemia. Neurochem. Res. 2004, 29, 1943–1949. [Google Scholar] [CrossRef] [PubMed]
  371. Cassidy, A.; Hanley, B.; Lamuela-Raventos, R.M. Isoflavones, lignans and stilbenes–Origins, metabolism and potential importance to human health. J. Sci. Food Agric. 2000, 80, 1044–1062. [Google Scholar] [CrossRef]
  372. Petrovska, B.B. Historical review of medicinal plants’ usage. Pharmacogn. Rev. 2012, 6, 1–5. [Google Scholar] [CrossRef]
  373. Gil, L. Las Transformaciones Históricas del Paisaje: La Permanencia y la Extinción Local del Pino Piñonero. Los Montes y su Historia: Una Perspectiva Política, Económica y Social; Universidad de Huelva: Huelva, Spain, 1999; pp. 151–186. [Google Scholar]
  374. Whited, T.L.; Engels, J.I.; Hoffmann, R.C.; Ibsen, H.; Verstegen, W. Northern Europe: An Environmental History; Bloomsbury Publishing USA: New York, NY, USA, 2005. [Google Scholar]
  375. Tsoucalas, G.; Vladimiros, L. The medical use of Málaka wine and cola nuts in late 19th/early 20th century Greece. DELTOS 2024, 33, 85–92. [Google Scholar] [CrossRef]
  376. Rodríguez-Delgado, M.A.; González, G.; Pérez-Trujillo, J.P.; García-Montelongo, F.J. Trans-resveratrol in wines from the Canary Islands (Spain). Analysis by high performance liquid chromatography. Food Chem. 2002, 76, 371–375. [Google Scholar] [CrossRef]
  377. Suprun, A.R.; Dubrovina, A.S.; Tyunin, A.P.; Kiselev, K.V. Profile of Stilbenes and Other Phenolics in Fanagoria White and Red Russian Wines. Metabolites 2021, 11, 231. [Google Scholar] [CrossRef]
  378. Zhou, D.D.; Li, J.; Xiong, R.G.; Saimaiti, A.; Huang, S.Y.; Wu, S.X.; Yang, Z.J.; Shang, A.; Zhao, C.N.; Gan, R.Y.; et al. Bioactive Compounds, Health Benefits and Food Applications of Grape. Foods 2022, 11, 2755. [Google Scholar] [CrossRef] [PubMed]
  379. Arya, S.S.; Salve, A.R.; Chauhan, S. Peanuts as functional food: A review. J. Food Sci. Technol. 2016, 53, 31–41. [Google Scholar] [CrossRef] [PubMed]
  380. Vijayan, N.; Haridas, M.; Abdulhameed, S. Stilbenes and Their Derivatives in Traditional Medicine. In Bioresources and Bioprocess in Biotechnology: Volume 2: Exploring Potential Biomolecules; Sugathan, S., Pradeep, N.S., Abdulhameed, S., Eds.; Springer: Singapore, 2017; pp. 407–418. [Google Scholar] [CrossRef]
  381. Lopes, R.M.; Agostini-Costa, T.d.S.; Gimenes, M.A.; Silveira, D. Chemical Composition and Biological Activities of Arachis Species. J. Agric. Food Chem. 2011, 59, 4321–4330. [Google Scholar] [CrossRef]
  382. Zu, X.Y.; Xiong, G.Q.; Geng, S.R.; Liao, T.; Li, X.; Zhang, Z.Y. Arachis hypogaea L. stem and leaf extract improves the sleep behavior of pentobarbital-treated rats. Biomed. Rep. 2014, 2, 388–391. [Google Scholar] [CrossRef]
  383. Al-Azawi, A.H.; Hassan, Z.H. Antibacterial Activity of Arachis hypogaea L. Seed Coat Extract Cultivated in Iraq. Pak. J. Biotechnol. Vol. 2017, 14, 601–605. [Google Scholar]
  384. Luo, Q.F.; Sun, L.; Si, J.Y.; Chen, D.H. Hypocholesterolemic effect of stilbenes containing extract-fraction from Cajanus cajan L. on diet-induced hypercholesterolemia in mice. Phytomedicine 2008, 15, 932–939. [Google Scholar] [CrossRef] [PubMed]
  385. Kong, Y.; Fu, Y.-J.; Zu, Y.-G.; Chang, F.-R.; Chen, Y.-H.; Liu, X.-L.; Stelten, J.; Schiebel, H.-M. Cajanuslactone, a new coumarin with anti-bacterial activity from pigeon pea [Cajanus cajan (L.) Millsp.] leaves. Food Chem. 2010, 121, 1150–1155. [Google Scholar] [CrossRef]
  386. Nicholson, R.A.; David, L.S.; Pan, R.L.; Liu, X.M. Pinostrobin from Cajanus cajan (L.) Millsp. inhibits sodium channel-activated depolarization of mouse brain synaptoneurosomes. Fitoterapia 2010, 81, 826–829. [Google Scholar] [CrossRef] [PubMed]
  387. Pal, D.; Mishra, P.; Sachan, N.; Ghosh, A.K. Biological activities and medicinal properties of Cajanus cajan (L) Millsp. J. Adv. Pharm. Technol. Res. 2011, 2, 207–214. [Google Scholar] [CrossRef]
  388. Hyun, T.K.; Eom, S.H.; Yu, C.Y.; Roitsch, T. Hovenia dulcis–an Asian traditional herb. Planta Medica 2010, 76, 943–949. [Google Scholar] [CrossRef]
  389. Sferrazza, G.; Brusotti, G.; Zonfrillo, M.; Temporini, C.; Tengattini, S.; Bononi, M.; Tateo, F.; Calleri, E.; Pierimarchi, P. Hovenia dulcis Thumberg: Phytochemistry, Pharmacology, Toxicology and Regulatory Framework for Its Use in the European Union. Molecules 2021, 26, 903. [Google Scholar] [CrossRef]
  390. Ritch-Krc, E.M.; Thomas, S.; Turner, N.J.; Towers, G.H.N. Carrier herbal medicine: Traditional and contemporary plant use. J. Ethnopharmacol. 1996, 52, 85–94. [Google Scholar] [CrossRef]
  391. Ritch-Krc, E.M.; Turner, N.J.; Towers, G.H.N. Carrier herbal medicine: An evaluation of the antimicrobial and anticancer activity in some frequently used remedies. J. Ethnopharmacol. 1996, 52, 151–156. [Google Scholar] [CrossRef]
  392. Legault, J.; Girard-Lalancette, K.; Dufour, D.; Pichette, A. Antioxidant Potential of Bark Extracts from Boreal Forest Conifers. Antioxidants 2013, 2, 77–89. [Google Scholar] [CrossRef]
  393. Uprety, Y.; Lacasse, A.; Asselin, H. Traditional Uses of Medicinal Plants from the Canadian Boreal Forest for the Management of Chronic Pain Syndromes. Pain Pract. 2016, 16, 459–466. [Google Scholar] [CrossRef] [PubMed]
  394. Ince, I.; Yesil-Celiktas, O.; Karabay-Yavasoglu, N.U.; Elgin, G. Effects of Pinus brutia bark extract and Pycnogenol® in a rat model of carrageenan induced inflammation. Phytomedicine 2009, 16, 1101–1104. [Google Scholar] [CrossRef]
  395. Satil, F.; Selvi, S.; Polat, R. Ethnic uses of pine resin production from Pinus brutia by native people on the Kazdag Mountain (Mt. Ida) in Western Turkey. J. Food Agric. Environ. 2011, 9, 1059–1063. [Google Scholar]
  396. Cretu, E.; Karonen, M.; Salminen, J.-P.; Mircea, C.; Trifan, A.; Charalambous, C.; Constantinou, A.I.; Miron, A. In Vitro Study on the Antioxidant Activity of a Polyphenol-Rich Extract from Pinus brutia Bark and Its Fractions. J. Med. Food 2013, 16, 984–991. [Google Scholar] [CrossRef]
  397. Secim-Karakaya, P.; Saglam-Metiner, P.; Yesil-Celiktas, O. Antimicrobial and wound healing properties of cotton fabrics functionalized with oil-in-water emulsions containing Pinus brutia bark extract and Pycnogenol® for biomedical applications. Cytotechnology 2021, 73, 423–431. [Google Scholar] [CrossRef]
  398. Süntar, I.; Demirel, M.A.; Taban, K.; Çeribaşı, A.O.; Gök, H.N.; Metkin, G. Assessment of the preventive activity of Pinus brutia Ten. (Pinaceae) against in vivo acute lung injury model. Phytochem. Lett. 2023, 58, 8–18. [Google Scholar] [CrossRef]
  399. Erol, K.F.; Kutlu, G.; Tornuk, F.; Guzel, M.; Donmez, I. Determination of antioxidant, anticancer, antidiabetic and antimicrobial activities of Turkish red pine (Pinus brutia Ten.) bark ultrasound-assisted extract as a functional food additive. Acta Aliment. 2023, 52, 102–112. [Google Scholar] [CrossRef]
  400. Kim, J.H.; Seo, J.E.; Choi, S.W. Quantitative Changes of Phenolic Compounds in Pine Twigs by Variety, Harvest Season, and Growing Region. Prev. Nutr. Food Sci. 2022, 27, 299–308. [Google Scholar] [CrossRef] [PubMed]
  401. Šarac, Z.; Matejić, J.S.; Stojanović-Radić, Z.Z.; Veselinović, J.B.; Džamić, A.M.; Bojović, S.; Marin, P.D. Biological activity of Pinus nigra terpenes—Evaluation of FtsZ inhibition by selected compounds as contribution to their antimicrobial activity. Comput. Biol. Med. 2014, 54, 72–78. [Google Scholar] [CrossRef]
  402. Milić, N.; Milanović, M.; Četojević-Simin, D.; Malenčić, Đ.; Prvulović, D.; Pavkov, N.; Radulović, Z.; Milošević, N.; Rašković, A.; Mandić, A. Phytochemical characterization and effects on cell proliferation of Pinus nigra Arn. bark. Arch. Pharm. 2021, 354, e2000416. [Google Scholar] [CrossRef]
  403. Papp, N.; Purger, D.; Czigle, S.; Czégényi, D.; Stranczinger, S.; Tóth, M.; Dénes, T.; Kocsis, M.; Takácsi-Nagy, A.; Filep, R. The Importance of Pine Species in the Ethnomedicine of Transylvania (Romania). Plants 2022, 11, 2331. [Google Scholar] [CrossRef] [PubMed]
  404. Demirtas, İ.; Ozen, T.; Marah, S.; Mutlu, D.; Arslan, Ş.; Gül, F. Functional food components and activities of Pinus nigra and Pinus sylvestris barks as food supplements. Int. J. Chem. Technol. 2023, 7, 229–238. [Google Scholar] [CrossRef]
  405. Dei Cas, L.; Pugni, F.; Fico, G. Tradition of use on medicinal species in Valfurva (Sondrio, Italy). J. Ethnopharmacol. 2015, 163, 113–134. [Google Scholar] [CrossRef]
  406. Manzione, M.G.; Vitalini, S.; Sharopov, F.; Capó, X.; Iriti, M.; Martorell, M.; Pezzani, R. Pinus mugo Turra and its Therapeutic Potential: A Narrative Review. Nat. Prod. Commun. 2024, 19, 1934578X241265934. [Google Scholar] [CrossRef]
  407. Hussain, M.; Shah, G.M.; Khan, M.A. Traditional medicinal and economic uses of Gymnosperms of Kaghan valley, Pakistan. Ethnobot. Leafl. 2006, 2006, 7. [Google Scholar]
  408. Khare, C.P. Indian Medicinal Plants: An Illustrated Dictionary; Springer Science & Business Media: Berlin/Heidelberg, Germany, 2008. [Google Scholar]
  409. Siddiqui, M.; Mesaik, A.; Wahab, M.; Khan, N.; Khan, U.; Nazim, K.; Hussain, S. Phytosociology of Pinus roxburghii Sargent. (Chir pine) in Lesser Himalayan and Hindu Kush range of Pakistan. Pak. J. Bot. 2009, 41, 2357–2369. [Google Scholar]
  410. Abbasi, A.M.; Khan, M.A.; Ahmad, M.; Zafar, M.; Jahan, S.; Sultana, S. Ethnopharmacological application of medicinal plants to cure skin diseases and in folk cosmetics among the tribal communities of North-West Frontier Province, Pakistan. J. Ethnopharmacol. 2010, 128, 322–335. [Google Scholar] [CrossRef] [PubMed]
  411. Piechowiak, T.; Matłok, N.; Balawejder, M. Extraction of phytochemicals from young shoots of Pinus sylvestris L. and analysis of their selected biological properties. LWT 2023, 188, 115404. [Google Scholar] [CrossRef]
  412. Stewart, C.D.; Jones, C.D.; Setzer, W.N. Essential oil compositions of Juniperus virginiana and Pinus virginiana, two important trees in Cherokee traditional medicine. Am. J. Essent. Oil Nat. Prod. 2014, 2, 17–24. [Google Scholar]
  413. Georghiou, K.; Delipetrou, P. Patterns and traits of the endemic plants of Greece. Bot. J. Linn. Soc. 2010, 162, 130–153. [Google Scholar] [CrossRef]
  414. Axiotis, E.; Halabalaki, M.; Skaltsounis, L.A. An Ethnobotanical Study of Medicinal Plants in the Greek Islands of North Aegean Region. Front. Pharmacol. 2018, 9, 409. [Google Scholar] [CrossRef]
  415. Sigerist, H.E. On Hippocrates. Bull. Inst. Hist. Med. 1934, 2, 190–214. [Google Scholar]
  416. Goldberg, H. Hippocrates: Father of Medicine; iUniverse: Lincoln, NE, USA, 2006. [Google Scholar]
  417. Scarborough, J. Theophrastus on herbals and herbal remedies. J. Hist. Biol. 1978, 11, 353–385. [Google Scholar] [CrossRef]
  418. Sharples, R.W.; Huby, P.M.; Fortenbaugh, W.W. Theophrastus of Eresus: Sources on Biology; Brill: Leiden, The Netherlands, 1995; Volume 64. [Google Scholar]
  419. Riddle, J.M. Dioscorides on Pharmacy and Medicine; University of Texas Press: Austin, TX, USA, 1985. [Google Scholar]
  420. Staub, P.O.; Casu, L.; Leonti, M. Back to the roots: A quantitative survey of herbal drugs in Dioscorides’ De Materia Medica (ex Matthioli, 1568). Phytomedicine 2016, 23, 1043–1052. [Google Scholar] [CrossRef]
  421. Wallner, C.; Moormann, E.; Lulof, P.; Drysch, M.; Lehnhardt, M.; Behr, B. Burn Care in the Greek and Roman Antiquity. Medicina 2020, 56, 657. [Google Scholar] [CrossRef]
  422. De Armas, E.; Sarracent, Y.; Marrero, E.; Fernández, O.; Branford-White, C. Efficacy of Rhizophora mangle aqueous bark extract (RMABE) in the treatment of aphthous ulcers: A pilot study. Curr. Med. Res. Opin. 2005, 21, 1711–1715. [Google Scholar] [CrossRef]
  423. Gonzales, M.V.M.; Tolentino, A.G. Extraction and isolation of the alkaloids from the Samanea saman (Acacia) bark: Its antiseptic potential. IInternational J. Sci. Technol. Res. 2014, 3, 119–124. [Google Scholar]
  424. Casas-Agustench, P.; Salas-Huetos, A.; Salas-Salvadó, J. Mediterranean nuts: Origins, ancient medicinal benefits and symbolism. Public. Health Nutr. 2011, 14, 2296–2301. [Google Scholar] [CrossRef]
  425. Ghalioungui, P. La Médecine des Pharaons: Magie et Science Médicale Dans l’Egypte Ancienne; Robert Laffont: Paris, France, 1983. [Google Scholar]
  426. Mattern, S.P. Galen and the Rhetoric of Healing; JHU Press: Baltimore, MD, USA, 2008. [Google Scholar]
  427. McNally, S. The maenad in early Greek art. Arethusa 1978, 11, 101–135. [Google Scholar]
  428. Hedreen, G. Silens, nymphs, and maenads. J. Hell. Stud. 1994, 114, 47–69. [Google Scholar]
  429. Brussell, D.E. Medicinal plants of Mt. Pelion, Greece. Econ. Bot. 2004, 58, S174–S202. [Google Scholar] [CrossRef]
  430. Seltman, C. Greek Sculpture and some festival coins. Hesperia J. Am. Sch. Class. Stud. Athens 1948, 17, 71–85. [Google Scholar] [CrossRef]
  431. Ghosh, S. Representation of Greek gods/goddesses in Graeco-Bactrian and Indo-Greek visual culture. In The Graeco-Bactrian and Indo-Greek World; Routledge: London, UK, 2020; pp. 570–579. [Google Scholar]
  432. Broneer, O. The Isthmian victory crown. Am. J. Archaeol. 1962, 66, 259–263. [Google Scholar] [CrossRef]
  433. Tillyard, E. Theseus, Sinis, and the Isthmian Games. J. Hell. Stud. 1913, 33, 296–312. [Google Scholar] [CrossRef]
  434. Ruiz Daniels, R.; Taylor, R.S.; González-Martínez, S.C.; Vendramin, G.G.; Fady, B.; Oddou-Muratorio, S.; Piotti, A.; Simioni, G.; Grivet, D.; Beaumont, M.A. Looking for local adaptation: Convergent microevolution in Aleppo pine (Pinus halepensis). Genes 2019, 10, 673. [Google Scholar] [CrossRef]
  435. Mauri, A.; Di Leo, M.; de Rigo, D.; Caudullo, G. Pinus halepensis and Pinus brutia in Europe: Distribution, habitat, usage and threats. In European Atlas of Forest Tree Species; San-Miguel-Ayanz, J., de Rigo, D., Caudullo, G., Houston Durrant, T., Mauri, A., Eds.; Publication Office of the European Union: Luxembourg, 2016; pp. 122–123. [Google Scholar]
  436. Scherrer, A.M.; Motti, R.; Weckerle, C.S. Traditional plant use in the areas of Monte Vesole and Ascea, Cilento National Park (Campania, Southern Italy). J. Ethnopharmacol. 2005, 97, 129–143. [Google Scholar] [CrossRef] [PubMed]
  437. Passalacqua, N.G.; Guarrera, P.M.; De Fine, G. Contribution to the knowledge of the folk plant medicine in Calabria region (Southern Italy). Fitoterapia 2007, 78, 52–68. [Google Scholar] [CrossRef]
  438. Motti, R.; Motti, P. An Ethnobotanical Survey of Useful Plants in the Agro Nocerino Sarnese (Campania, Southern Italy). Hum. Ecol. 2017, 45, 865–878. [Google Scholar] [CrossRef]
  439. Menale, B.; Muoio, R. Use of medicinal plants in the South-Eastern area of the Partenio Regional Park (Campania, Southern Italy). J. Ethnopharmacol. 2014, 153, 297–307. [Google Scholar] [CrossRef]
  440. Guri, A.; Kefalas, P.; Roussis, V. Antioxidant potential of six pine species. Phytother. Res. 2006, 20, 263–266. [Google Scholar] [CrossRef]
  441. Dhibi, M.; Mechri, B.; Brahmi, F.; Skhiri, F.; Alsaif, M.A.; Hammami, M. Fatty acid profiles, antioxidant compounds and antiradical properties of Pinus halepensis Mill. cones and seeds. J. Sci. Food Agric. 2012, 92, 1702–1708. [Google Scholar] [CrossRef]
  442. Dhibi, M.; Issaoui, M.; Brahmi, F.; Mechri, B.; Mnari, A.; Cheraif, I.; Skhiri, F.; Gazzah, N.; Hammami, M. Nutritional quality of fresh and heated Aleppo pine (Pinus halepensis Mill.) seed oil: Trans-fatty acid isomers profiles and antioxidant properties. J. Food Sci. Technol. 2014, 51, 1442–1452. [Google Scholar] [CrossRef] [PubMed]
  443. Djerrad, Z.; Kadik, L.; Djouahri, A. Chemical variability and antioxidant activities among Pinus halepensis Mill. essential oils provenances, depending on geographic variation and environmental conditions. Ind. Crops Prod. 2015, 74, 440–449. [Google Scholar] [CrossRef]
  444. Djerrad, Z.; Djouahri, A.; Kadik, L. Variability of Pinus halepensis Mill. Essential Oils and Their Antioxidant Activities Depending on the Stage of Growth During Vegetative Cycle. Chem. Biodivers. 2017, 14, e1600340. [Google Scholar] [CrossRef] [PubMed]
  445. Salim, H.; Rimawi, W.H.; Shaheen, S.; Mjahed, A. Phytochemical Analysis and Antibacterial Activity of Extracts from Palestinian Aleppo Pine Seeds, Bark and Cones. Asian J. Chem. 2018, 31, 143–147. [Google Scholar] [CrossRef]
  446. Abbou, A.; Kadri, N.; Debbache, N.; Dairi, S.; Remini, H.; Dahmoune, F.; Berkani, F.; Adel, K.; Belbahi, A.; Madani, K. Effect of precipitation solvent on some biological activities of polysaccharides from Pinus halepensis Mill. seeds. Int. J. Biol. Macromol. 2019, 141, 663–670. [Google Scholar] [CrossRef]
  447. Meziti, H.; Bouriche, H.; Kada, S.; Demirtas, I.; Kizil, M.; Senator, A.; Garrido, G. Phytochemical analysis, and antioxidant, anti-hemolytic and genoprotective effects of Quercus ilex L. and Pinus halepensis Mill. methanolic extracts. J. Pharm. Pharmacogn. Res. 2019, 7, 260–272. [Google Scholar] [CrossRef]
  448. Bouyahya, A.; Belmehdi, O.; Abrini, J.; Dakka, N.; Bakri, Y. Chemical composition of Mentha suaveolens and Pinus halepensis essential oils and their antibacterial and antioxidant activities. Asian Pac. J. Trop. Med. 2019, 12, 117–122. [Google Scholar] [CrossRef]
  449. Duganath, N.; Kumar, S.R.; Kumanan, R.; Jayaveera, K.N. Evaluation of anti-denaturation property and anti-oxidant activity of traditionally used medicinal plants. Int. J. Pharma Biosci. 2010, 1, PS110. [Google Scholar]
  450. Leelaprakash, G.; Dass, S.M. Invitro anti-inflammatory activity of methanol extract of Enicostemma axillare. Int. J. Drug Dev. Res. 2011, 3, 189–196. [Google Scholar]
  451. Chandra, S.; Chatterjee, P.; Dey, P.; Bhattacharya, S. Evaluation of in vitro anti-inflammatory activity of coffee against the denaturation of protein. Asian Pac. J. Trop. Biomed. 2012, 2, S178–S180. [Google Scholar] [CrossRef]
  452. Süntar, I.; Tumen, I.; Ustün, O.; Keleş, H.; Küpeli Akkol, E. Appraisal on the wound healing and anti-inflammatory activities of the essential oils obtained from the cones and needles of Pinus species by in vivo and in vitro experimental models. J. Ethnopharmacol. 2012, 139, 533–540. [Google Scholar] [CrossRef]
  453. Popescu, D.I.; Lengyel, E.; Apostolescu, F.G.; Soare, L.C.; Botoran, O.R.; Șuțan, N.A. Volatile Compounds and Antioxidant and Antifungal Activity of Bud and Needle Extracts from Three Populations of Pinus mugo Turra Growing in Romania. Horticulturae 2022, 8, 952. [Google Scholar] [CrossRef]
  454. Popa, C.; Hanscam, E. Of Romans, Dacians and Romanians. Archaica 2020, 7–8, 244–251. [Google Scholar]
  455. Segneanu, A.-E.; Cepan, C.; Grozescu, I.; Cziple, F.; Olariu, S.; Ratiu, S.; Lazar, V.; Murariu, S.M.; Velciov, S.M.; Marti, T.D. Therapeutic Use of Some Romanian Medicinal Plants. In Pharmacognosy; Shagufta, P., Areej, A.-T., Eds.; IntechOpen: Rijeka, Croatia, 2019; Chapter 7. [Google Scholar] [CrossRef]
  456. Žuškin, E.; Lipozenčić, J.; Pucarin-Cvetković, J.; Mustajbegović, J.; Schachter, N.; Mučić-Pučić, B.; Neralić-Meniga, I. Ancient medicine-a review. Acta Dermatovenerol. Croat. 2008, 16, 149–157. [Google Scholar]
  457. Carciumaru, M. Plants used by traco-geto-dacians (Attempt of synthesis)(V). Thraco-Dacica 1987, 8, 171–176. [Google Scholar]
  458. Fierascu, R.C.; Fierascu, I.; Ortan, A.; Avramescu, S.M.; Dinu-Pirvu, C.E.; Ionescu, D. Romanian aromatic and medicinal plants: From tradition to science. In Aromatic and Medicinal Plants-Back to Nature; IntechOpen: London, UK, 2017; pp. 149–173. [Google Scholar]
  459. Pogăcias, A. The Dacian Society–Fierce Warriors and Their Women Sources and Representations. Hiperboreea 2017, 4, 5–22. [Google Scholar]
  460. Melius, J.P. Herbarium; Heltai Gaspárné, Colosvár: Kolozsvár, Romania, 1578. [Google Scholar]
  461. Papp, N.; Birkás-Frendl, K.; Farkas, Á.; Pieroni, A. An ethnobotanical study on home gardens in a Transylvanian Hungarian Csángó village (Romania). Genet. Resour. Crop Evol. 2013, 60, 1423–1432. [Google Scholar] [CrossRef]
  462. Bartha, S.G.; Quave, C.L.; Balogh, L.; Papp, N. Ethnoveterinary practices of covasna county, transylvania, Romania. J. Ethnobiol. Ethnomedicine 2015, 11, 1–22. [Google Scholar] [CrossRef]
  463. Kóczián, G.; Pintér, I.; Szabó, L.G. Adatok a gyimesi csángók népi gyógyászatához. Gyógyszerészet 1975, 19, 226–230. [Google Scholar]
  464. Halászné, Z. Data to the medicinal plants usage of the Hungarians in Moldova. Gyógyszerészet 1981, 25, 361–367. [Google Scholar]
  465. Péntek, J.; Szabó, A. Ember és Növényvilág: Kalotaszeg Növényzete és Népi Növényismerete; Kriterion könyvkiadó: Cluj-Napoca, Romania, 1985. [Google Scholar]
  466. Martindale, W.; Westcott, W.W. The Extra Pharmacopoeia of Martindale and Westcott; HK Lewis: London, UK, 1925; Volume 2. [Google Scholar]
  467. Dörfler, H.; Roselt, G. Heilpfanzen Gestern und Heute; Urania: Leipzig, Germany; Jena, Germany; Berlin, Germany, 1988. [Google Scholar]
  468. Richardson, D.M. Ecology and Biogeography of Pinus; Cambridge University Press: Cambridge, UK, 2000. [Google Scholar]
  469. Žuna Pfeiffer, T.; Krstin, L.; Špoljarić Maronić, D.; Hmura, M.; Eržić, I.; Bek, N.; Stević, F. An ethnobotanical survey of useful wild plants in the north-eastern part of Croatia (Pannonian region). Plant Biosyst.-Int. J. Deal. All Asp. Plant Biol. 2020, 154, 463–473. [Google Scholar] [CrossRef]
  470. Koczian, G.; Szabo, I.; Szabo, L. Etnobotanikai adatok Kalotaszebrol. Bot. Kozlemenyek 1977, 64, 23–29. [Google Scholar]
  471. Butură, V. Enciclopedie de Etnobotanică Românească; Editura Științifică și Enciclopedică: Bucharest, Romania, 1979. [Google Scholar]
  472. Gub, J. Adatok a Nagy-Homoród és a Nagy-Küküllő közötti terület népi növényismeretéhez. Néprajzi Látóhatár 1993, 1, 95–110. [Google Scholar]
  473. Halász, P. Növények a Moldvai Magyarok Hagyományában és Mindennapjaiban; General Press Konyvkiado: Budapest, Hungary, 2010. [Google Scholar]
  474. Janaćković, P.; Gavrilović, M.; Savić, J.; Marin, P.D.; Stevanović, Z.D. Traditional knowledge on plant use from Negotin Krajina (Eastern Serbia): An ethnobotanical study. Indian J. Tradit. Knowl. 2019, 18, 25–33. [Google Scholar]
  475. Gunda, B. Egy kárpátaljai magyar falu ethnobotanikája. Nyíregyházi Jósa András Múzeum Évkönyve 1978, 1978, 25–41. [Google Scholar]
  476. Kóczián, G.; Szabó, L.G. Nagyatád Egy Kertészgazdaságának Leírása, Agrobotanikai Értékei És Géntartaléka (Description of a Garden in Nagyatád, Its Agrobotanical Values and Gene Resources). Agrártudományi Szle 1978, 1–2, 181–197. [Google Scholar]
  477. Matejić, J.S.; Stefanović, N.; Ivković, M.; Živanović, N.; Marin, P.D.; Džamić, A.M. Traditional uses of autochthonous medicinal and ritual plants and other remedies for health in Eastern and South-Eastern Serbia. J. Ethnopharmacol. 2020, 261, 113186. [Google Scholar] [CrossRef]
  478. Tucakov, J. Lečenje Biljem (Healing with Herbs); Rad: Belgrade, Serbia, 1986. [Google Scholar]
  479. Kóczian, G. Hagyományos Parasztgazdálkodás, Termesztett, a Gyujtögeto Gazdálkodás, Vad Növényfajainak Etnobotanikai Értékelése. (Ethnobotanical Evaluation of Traditional Farming, Cultivation, and Collecting of Wild Plant Species); Nagyatádi Kulturális és Sport Központ: Nagyatád, Hungary, 2014. [Google Scholar]
  480. Keszeg, V. A Mezőségi Detrehemtelep Népi Gyógyászata (Folk Medicine of the Detrehemtelep in the Field). In Népismereti Dolgozatok; Kriterion Könyvkiadó: Bucharest, Romania, 1981; pp. 97–117. [Google Scholar]
  481. Benítez, G.; González-Tejero, M.R.; Molero-Mesa, J. Pharmaceutical ethnobotany in the western part of Granada province (southern Spain): Ethnopharmacological synthesis. J. Ethnopharmacol. 2010, 129, 87–105. [Google Scholar] [CrossRef]
  482. Belda, A.; Peiró, V.; Seva, E. The Relationship between Plants Used to Sustain Finches (Fringillidae) and Uses for Human Medicine in Southeast Spain. Evid.-Based Complement. Altern. Med. 2012, 2012, 360913. [Google Scholar] [CrossRef]
  483. Zhu, P.; Qi, R.-Q.; Yang, Y.; Huo, W.; Zhang, Y.; He, L.; Wang, G.; Xu, J.; Zhang, F.; Yang, R.; et al. Clinical guideline for the diagnosis and treatment of cutaneous warts (2022). J. Evid.-Based Med. 2022, 15, 284–301. [Google Scholar] [CrossRef]
  484. Gras, A.; Serrasolses, G.; Vallès, J.; Garnatje, T. Traditional knowledge in semi-rural close to industrial areas: Ethnobotanical studies in western Gironès (Catalonia, Iberian Peninsula). J. Ethnobiol. Ethnomed. 2019, 15, 19. [Google Scholar] [CrossRef] [PubMed]
  485. Carrió, E.; Vallès, J. Ethnobotany of medicinal plants used in Eastern Mallorca (Balearic Islands, Mediterranean Sea). J. Ethnopharmacol. 2012, 141, 1021–1040. [Google Scholar] [CrossRef]
  486. Benarba, B.; Belabid, L.; Righi, K.; Bekkar, A.a.; Elouissi, M.; Khaldi, A.; Hamimed, A. Ethnobotanical study of medicinal plants used by traditional healers in Mascara (North West of Algeria). J. Ethnopharmacol. 2015, 175, 626–637. [Google Scholar] [CrossRef]
  487. Chermat, S.; Gharzouli, R. Ethnobotanical study of medicinal flora in the North East of Algeria-An empirical knowledge in Djebel Zdimm (Setif). J. Mater. Sci. Eng. 2015, 5, 50–59. [Google Scholar]
  488. Hmamouchi, M.; Hamamouchi, J.; Zouhdi, M.; Bessiere, J.M. Chemical and Antimicrobial Properties of Essential Oils of Five Moroccan Pinaceae. J. Essent. Oil Res. 2001, 13, 298–302. [Google Scholar] [CrossRef]
  489. Ghanmi, M.; Satrani, B.; Chaouch, A.; Aafi, A.; Abid, A.E.; Ismaili, M.R.; Farah, A. Composition chimique et activité antimicrobienne de l’essence de térébenthine du pin maritime (Pinus pinaster) et du pin d’Alep (Pinus hale- pensis) du Maroc. Acta Bot. Gall. 2007, 154, 293–300. [Google Scholar] [CrossRef]
  490. Bachir, R. Antibacterial potential of essential oils of the needles of Pinus halepensis against Staphylococcus aureus and Escherichia coli. J. Coast. Life Med. 2014, 2, 651–655. [Google Scholar] [CrossRef]
  491. Fekih, N.; Allali, H.; Merghache, S.; Chaïb, F.; Merghache, D.; El Amine, M.; Djabou, N.; Muselli, A.; Tabti, B.; Costa, J. Chemical composition and antibacterial activity of Pinus halepensis Miller growing in West Northern of Algeria. Asian Pac. J. Trop. Dis. 2014, 4, 97–103. [Google Scholar] [CrossRef]
  492. Sadou, N.; Seridi, R.; Djahoudi, A.; Hadef, Y. Composition chimique et activité antibactérienne des Huiles Essentielles des aiguilles de Pinus halepensis Mill. du Nord est Algérien. Synthèse Rev. Sci. Technol. 2015, 30, 33–39. [Google Scholar]
  493. Mitić, Z.S.; Jovanović, B.; Jovanović, S.Č.; Stojanović-Radić, Z.Z.; Mihajilov-Krstev, T.; Jovanović, N.M.; Nikolić, B.M.; Marin, P.D.; Zlatković, B.K.; Stojanović, G.S. Essential oils of Pinus halepensis and P. heldreichii: Chemical composition, antimicrobial and insect larvicidal activity. Ind. Crops Prod. 2019, 140, 111702. [Google Scholar] [CrossRef]
  494. Ashmawy, N.A.; Al Farraj, D.A.; Salem, M.Z.M.; Elshikh, M.S.; Al-Kufaidy, R.; Alshammari, M.K.; Salem, A.Z.M. Potential impacts of Pinus halepensis Miller trees as a source of phytochemical compounds: Antibacterial activity of the cones essential oil and n-butanol extract. Agrofor. Syst. 2020, 94, 1403–1413. [Google Scholar] [CrossRef]
  495. Amri, I.; Hamrouni, L.; Hanana, M.; Gargouri, S.; Fezzani, T.; Jamoussi, B. Chemical composition, physico-chemical properties, antifungal and herbicidal activities of Pinus halepensis Miller essential oils. Biol. Agric. Hortic. 2013, 29, 91–106. [Google Scholar] [CrossRef]
  496. Hamrouni, L.; Hanana, M.; Amri, I.; Romane, A.E.; Gargouri, S.; Jamoussi, B. Allelopathic effects of essential oils of Pinus halepensis Miller: Chemical composition and study of their antifungal and herbicidal activities. Arch. Phytopathol. Plant Prot. 2015, 48, 145–158. [Google Scholar] [CrossRef]
  497. Eryilmaz, M.; Tosun, A.; Tumen, I. Antimicrobial Activity of Some Species from Pinaceae and Cupressaceae. Turk. J. Pharm. Sci. 2015, 13, 35–40. [Google Scholar] [CrossRef]
  498. Al-Bazaz, H.K.; Al-jubori, I.S.; Eldalawy, R.; Nasser, N.M. Antimicrobial and Antioxidant Activity Pinus halepensis Miller. Cone Extract which grown in Iraq. Res. J. Pharm. Technol. 2018, 11, 4977–4980. [Google Scholar] [CrossRef]
  499. Mohamed, A.A.; Behiry, S.I.; Ali, H.M.; EL-Hefny, M.; Salem, M.Z.M.; Ashmawy, N.A. Phytochemical Compounds of Branches from P. halepensis Oily Liquid Extract and S. terebinthifolius Essential Oil and Their Potential Antifungal Activity. Processes 2020, 8, 330. [Google Scholar] [CrossRef]
  500. Bouzenna, H.; Samout, N.; Amani, E.; Mbarki, S.; Tlili, Z.; Rjeibi, I.; Elfeki, A.; Talarmin, H.; Hfaiedh, N. Protective Effects of Pinus halepensis L. Essential Oil on Aspirin-induced Acute Liver and Kidney Damage in Female Wistar Albino Rats. J. Oleo Sci. 2016, 65, 701–712. [Google Scholar] [CrossRef]
  501. Bouzenna, H.; Hfaiedh, N.; Bouaziz, M.; Giroux-Metges, M.-A.; Elfeki, A.; Talarmin, H. Cytoprotective effects of essential oil of Pinus halepensis L. against aspirin-induced toxicity in IEC-6 cells. Arch. Physiol. Biochem. 2017, 123, 364–370. [Google Scholar] [CrossRef]
  502. Sarri, M.; Boudjelal, A.; Hendel, N.; Sarri, D.; Benkhaled, A. Flora and ethnobotany of medicinal plants in the southeast of the capital of Hodna (Algeria). Arab. J. Med. Aromat. Plants 2015, 1, 24–30. [Google Scholar]
  503. Senouci, F.; Ababou, A.; Chouieb, M. Ethnobotanical Survey of the Medicinal Plants used in the Southern Mediterranean. Case Study: The Region of Bissa (Northeastern Dahra Mountains, Algeria). Pharmacogn. J. 2019, 11, 647–659. [Google Scholar] [CrossRef]
  504. Chohra, D.; Ferchichi, L. Ethnobotanical study of Belezma National Park (BNP) plants in Batna: East of Algeria. Acta Sci. Nat. 2019, 6, 40–54. [Google Scholar] [CrossRef]
  505. Nazim, B.; Houarı, T.; Ismaıl, B. Ethnobotanical Survey of Some Plants Used in Tessala Region, Algeria. Curr. Perspect. Med. Aromat. Plants 2020, 3, 25–30. [Google Scholar] [CrossRef]
  506. Miara, M.D.; Bendif, H.; Rebbas, K.; Rabah, B.; Hammou, M.A.; Maggi, F. Medicinal plants and their traditional uses in the highland region of Bordj Bou Arreridj (Northeast Algeria). J. Herb. Med. 2019, 16, 100262. [Google Scholar] [CrossRef]
  507. Mendelsohn, A.; Sato, T.; Subedi, A.; Wurcel, A.G. State-of-the-Art Review: Evaluation and Management of Delusional Infestation. Clin. Infect. Dis. 2024, 79, e1–e10. [Google Scholar] [CrossRef]
  508. Laid, B.; Rebbas, K.; Mouloud, G.; Rabah, B.; Brini, F.; Bouzerzour, H. Ethnobotanical study of medicinal plants in Jebel Messaad region (M’sila, Algeria). Glob. J. Res. Med. Plants Indig. Med. 2014, 3, 445–459. [Google Scholar]
  509. Moussi, M.; Filali, H.; Tazi, A.; Hakkou, F. Ethnobotanical survey of healing medicinal plants traditionally used in the main Moroccan cities. J. Pharmacogn. Phytother. 2015, 7, 164–182. [Google Scholar]
  510. Salhi, N.; Bouyahya, A.; Fettach, S.; Zellou, A.; Cherrah, Y. Ethnopharmacological study of medicinal plants used in the treatment of skin burns in occidental Morocco (area of Rabat). S. Afr. J. Bot. 2019, 121, 128–142. [Google Scholar] [CrossRef]
  511. Mouhajir, F.; Hudson, J.B.; Rejdali, M.; Towers, G.H.N. Multiple Antiviral Activities of Endemic Medicinal Plants Used by Berber Peoples of Morocco. Pharm. Biol. 2001, 39, 364–374. [Google Scholar] [CrossRef]
  512. El Abbouyi, A.; Filali-Ansari, N.; Khyari, P.; Loukili, H. Inventory of medicinal plants prescribed by traditional healers in El Jadida city and suburbs (Morocco). Int. J. Green Pharm. 2014, 8, 242–251. [Google Scholar] [CrossRef]
  513. Ouhaddou, H.; Boubaker, H.; Msanda, F.; El Mousadik, A. An ethnobotanical study of medicinal plants of the Agadir Ida Ou Tanane province (southwest Morocco). J. Appl. Biosci. 2014, 84, 7707–7722. [Google Scholar] [CrossRef]
  514. Shinde, U.A.; Phadke, A.S.; Nair, A.M.; Mungantiwar, A.A.; Dikshit, V.J.; Saraf, M.N. Membrane stabilizing activity—A possible mechanism of action for the anti-inflammatory activity of Cedrus deodara wood oil. Fitoterapia 1999, 70, 251–257. [Google Scholar] [CrossRef]
  515. Umapathy, E.; Ndebia, E.; Meeme, A.; Adam, B.; Menziwa, P.; Nkeh-Chungag, B.; Iputo, J. An experimental evaluation of Albuca setosa aqueous extract on membrane stabilization, protein denaturation and white blood cell migration during acute inflammation. J. Med. Plants Res. 2010, 4, 789–795. [Google Scholar]
  516. Ghayth, R.; Jebali, J.; Ghazghazi, H.; Riguene, H.; Khouja, M.; Salem, R. Chemical composition and biological activities of Pinus halepensis Mill. Oil. Rev. Roum. De Chim. 2019, 64, 999–1006. [Google Scholar] [CrossRef]
  517. Kadri, N.; Khettal, B.; Adjebli, A.; Cresteil, T.; Yahiaoui-Zaidi, R.; Barragan-Montero, V.; Montero, J.-L. Antiangiogenic activity of neutral lipids, glycolipids, and phospholipids fractions of Pinus halepensis Mill. seeds. Ind. Crops Prod. 2014, 54, 6–12. [Google Scholar] [CrossRef]
  518. Merzouki, A.; Ed-derfoufi, F.; Molero Mesa, J. Contribution to the knowledge of Rifian traditional medicine. II: Folk medicine in Ksar Lakbir district (NW Morocco). Fitoterapia 2000, 71, 278–307. [Google Scholar] [CrossRef]
  519. Youbi, A.E.; Ouahidi, I.; Mansouri, L.E.; Daoudi, A.; Bousta, D. Ethnopharmacological survey of plants used for immunological diseases in four regions of Morocco. Eur. J. Med. Plants 2016, 13, 1. [Google Scholar] [CrossRef]
  520. Benlamdini, N.; Elhafian, M.; Rochdi, A.; Zidane, L. Étude floristique et ethnobotanique de la flore médicinale du Haut Atlas oriental (Haute Moulouya). J. Appl. Biosci. 2014, 78, 6771–6787. [Google Scholar] [CrossRef]
  521. Bouyahya, A.; Abrini, J.; Et-Touys, A.; Bakri, Y.; Dakka, N. Indigenous knowledge of the use of medicinal plants in the North-West of Morocco and their biological activities. Eur. J. Integr. Med. 2017, 13, 9–25. [Google Scholar] [CrossRef]
  522. Bilbis, L.S.; Shehu, R.A.; Abubakar, M.G. Hypoglycemic and hypolipidemic effects of aqueous extract of Arachis hypogaea in normal and Alloxan-induced diabetic rats. Phytomedicine 2002, 9, 553–555. [Google Scholar] [CrossRef]
  523. King, J.C.; Blumberg, J.; Ingwersen, L.; Jenab, M.; Tucker, K.L. Tree nuts and peanuts as components of a healthy diet. J. Nutr. 2008, 138, 1736s–1740s. [Google Scholar] [CrossRef] [PubMed]
  524. Periferakis, A.; Periferakis, K. On the Dissemination of Acupuncture to Europe. JournalNX 2020, 6, 201–209. [Google Scholar]
  525. Periferakis, A. H Εμφάνιση του Βελονισμού στην Ευρώπη, 1683–1850-Ascensus Acupuncturae in Europam, 1683–1850; Akadimia of Ancient Greek and Traditional Chinese Medicine: Athens, Greece, 2021. [Google Scholar]
  526. Wiart, C. Ethnopharmacology of medicinal plants: Asia and the Pacific; Springer Science & Business Media: Berlin/Heidelberg, Germany, 2007. [Google Scholar]
  527. Bertioli, D.; Seijo, G.; Freitas, F.; Valls, J.; Leal-Bertioli, S.; Moretzsohn, M. An overview of peanut and its wild relatives. Plant Genet. Resour. 2011, 9, 134–149. [Google Scholar] [CrossRef]
  528. Hammons, R.O.; Herman, D.; Stalker, H.T. Chapter 1—Origin and Early History of the Peanut. In Peanuts; Stalker, H.T., F. Wilson, R., Eds.; AOCS Press: Champaign, IL, USA, 2016; pp. 1–26. [Google Scholar] [CrossRef]
  529. Akram, N.A.; Shafiq, F.; Ashraf, M. Peanut (Arachis hypogaea L.): A Prospective Legume Crop to Offer Multiple Health Benefits Under Changing Climate. Compr. Rev. Food Sci. Food Saf. 2018, 17, 1325–1338. [Google Scholar] [CrossRef]
  530. Wang, Q.; Xu, J.; Shi, M. Clinical efficacy of Groundnut leaves on insomnia treatments. Shanghai J. Trad. Chin. Med. 2001, 5, 8–10. [Google Scholar]
  531. Hu, P.; Fan, R.; Li, Y.; Pang, C. Studies on pharmacological action of Luohuashengzhiye extract. Chin. Trad. Pat. Med. 2001, 23, 919–920. [Google Scholar]
  532. Wang, Y.; Li, H.; Xu, Y.; Zhang, Y.; Xu, D.; Xiao, L.; Li, X. Clinical confirmation of preparation from the branch and leaf of peanut in treating insomnia. Shanghai J. Tradit. Chin. Med. 2001, 5, 8–10. [Google Scholar]
  533. Lou, H.; Yamazaki, Y.; Sasaki, T.; Uchida, M.; Tanaka, H.; Oka, S. A-type proanthocyanidins from peanut skins. Phytochemistry 1999, 51, 297–308. [Google Scholar] [CrossRef]
  534. Lee, J.-S.; Lee, M.-K.; Kim, Y.-K.; Kim, K.-E.; Hyun, K.-Y. Attenuant effects of Hovenia dulcis extract on inflammatory orifacial pain in rats. J. Korea Acad.-Ind. Coop. Soc. 2014, 15, 5088–5094. [Google Scholar]
  535. Lim, S.J.; Kim, M.; Randy, A.; Nam, E.J.; Nho, C.W. Effects of Hovenia dulcis Thunb. extract and methyl vanillate on atopic dermatitis-like skin lesions and TNF-α/IFN-γ-induced chemokines production in HaCaT cells. J. Pharm. Pharmacol. 2016, 68, 1465–1479. [Google Scholar] [CrossRef]
  536. Park, J.-Y.; Moon, J.-Y.; Park, S.-D.; Park, W.-H.; Kim, H.; Kim, J.-E. Fruits extracts of Hovenia dulcis Thunb. suppresses lipopolysaccharide-stimulated inflammatory responses through nuclear factor-kappaB pathway in Raw 264.7 cells. Asian Pac. J. Trop. Med. 2016, 9, 357–365. [Google Scholar] [CrossRef] [PubMed]
  537. Jeong, Y.H.; Oh, Y.-C.; Cho, W.-K.; Yim, N.-H.; Ma, J.Y. Hoveniae Semen Seu Fructus Ethanol Extract Exhibits Anti-Inflammatory Activity via MAPK, AP-1, and STAT Signaling Pathways in LPS-Stimulated RAW 264.7 and Mouse Peritoneal Macrophages. Mediat. Inflamm. 2019, 2019, 9184769. [Google Scholar] [CrossRef]
  538. Xu, B.-J.; Deng, Y.-Q.; Sung, C.-K. Advances in studies on bioactivity of Hovenia dulcis. Agric. Chem. Biotechnol. 2004, 47, 1–5. [Google Scholar]
  539. Yoshikawa, M.; Ueda, T.; Muraoka, O.; Aoyama, H.; Matsuda, H.; Shimoda, H.; Yamahara, J.; Murakami, N. Absolute stereostructures of hovenidulciosides A1 and A2, bioactive novel triterpene glycosides from hoveniae semen seu fructus, the seeds and fruit of Hovenia dulcis Thunb. Chem. Pharm. Bull. 1995, 43, 532–534. [Google Scholar] [CrossRef] [PubMed]
  540. Yoshikawa, M.; Murakami, T.; Ueda, T.; Matsuda, H.; Yamahara, J.; Murakami, N. Bioactive Saponins and Glycosides. IV. Four Methyl-Migrated 16, 17-Seco-Dammarane Triterpene Glycosides from Chinese Natural Medicine, Hoveniae Semen Seu Fructus, the Seeds and Fruit of Hovenia dulcis Thunb: Absolute Stereostructures and Inhibitory Activity on Histamine Release of Hovenidulcioseds A1, A2, B1, and B2. Chem. Pharm. Bull. 1996, 44, 1736–1743. [Google Scholar]
  541. Choi, C.-Y.; Cho, S.-S.; Yoon, I.-S. Hot-water extract of the branches of Hovenia dulcis Thunb.(Rhamnaceae) ameliorates low-fiber diet-induced constipation in rats. Drug Des. Dev. Ther. 2018, 12, 695–703. [Google Scholar] [CrossRef] [PubMed]
  542. Cha, P.-H.; Shin, W.; Zahoor, M.; Kim, H.-Y.; Min, D.S.; Choi, K.-Y. Hovenia dulcis Thunb extract and its ingredient methyl vanillate activate wnt/β-catenin pathway and increase bone mass in growing or ovariectomized mice. PLoS ONE 2014, 9, e85546. [Google Scholar] [CrossRef]
  543. Kim, H.-L.; Sim, J.-E.; Choi, H.-M.; Choi, I.-Y.; Jeong, M.-Y.; Park, J.; Jung, Y.; Youn, D.-H.; Cho, J.-H.; Kim, J.-H. The AMPK pathway mediates an anti-adipogenic effect of fruits of Hovenia dulcis Thunb. Food Funct. 2014, 5, 2961–2968. [Google Scholar] [CrossRef]
  544. Pinto, J.T.; Oliveira, T.T.d.; Alvarenga, L.F.; Barbosa, A.S.; Pizziolo, V.R.; Costa, M.R.d. Pharmacological activity of the hydroalcoholic extract from Hovenia dulcis thunberg fruit and the flavonoid dihydromyricetin during hypercholesterolemia induced in rats. Braz. J. Pharm. Sci. 2014, 50, 727–735. [Google Scholar] [CrossRef]
  545. Yang, B.; Wu, Q.; Song, X.; Yang, Q.; Kan, J. Physicochemical properties and bioactive function of Japanese grape (Hovenia dulcis) pomace insoluble dietary fibre modified by ball milling and complex enzyme treatment. Int. J. Food Sci. Technol. 2019, 54, 2363–2373. [Google Scholar] [CrossRef]
  546. Cha, W.S.; Oh, J.H.; Park, H.J.; Ahn, S.W.; Hong, S.Y.; Kim, N.I. Historical difference between traditional Korean medicine and traditional Chinese medicine. Neurol. Res. 2007, 29 (Suppl. 1), S5–S9. [Google Scholar] [CrossRef] [PubMed]
  547. Patwardhan, B.; Mutalik, G.; Tillu, G. Chapter 2—Evolution of Medicine. In Integrative Approaches for Health; Patwardhan, B., Mutalik, G., Tillu, G., Eds.; Academic Press: Boston, MA, USA, 2015; pp. 27–52. [Google Scholar] [CrossRef]
  548. Kumar, H.; Song, S.Y.; More, S.V.; Kang, S.M.; Kim, B.W.; Kim, I.S.; Choi, D.K. Traditional Korean East Asian medicines and herbal formulations for cognitive impairment. Molecules 2013, 18, 14670–14693. [Google Scholar] [CrossRef]
  549. Kuete, V.; Seo, E.-J.; Krusche, B.; Oswald, M.; Wiench, B.; Schröder, S.; Greten, H.J.; Lee, I.-S.; Efferth, T. Cytotoxicity and pharmacogenomics of medicinal plants from traditional Korean medicine. Evid.-Based Complement. Altern. Med. 2013, 2013, 341724. [Google Scholar] [CrossRef] [PubMed]
  550. Ryu, J.Y.; Park, J.Y.; Sung, A.D.; Sung, S.H. The utilization of traditional herbal medicine for treatment in traditional Korean medicine clinics. In Medicinal Plants-Use in Prevention and Treatment of Diseases; Hassan, B.A.R., Ed.; IntechOpen: London, UK, 2019; pp. 111–126. [Google Scholar]
  551. Sakai, K.; Yamane, T.; Saitoh, Y.; Ikawa, C.; Nishihata, T. Effect of water extracts of crude drugs in decreasing blood ethanol concentrations in rats. Chem. Pharm. Bull. 1987, 35, 4597–4604. [Google Scholar] [CrossRef]
  552. An, S.-W.; Kim, Y.-G.; Kim, M.-H.; Lee, B.-I.; Lee, S.-H.; Kwon, H.-I.; Hwang, B.; Lee, H.-Y. Comparison of Hepatic Detoxification activity and reducing Serum Alcohol concentration of Hovenia dulsis Thunb and Alnus japonica Steud. Korean J. Med. Crop Sci. 1999, 7, 263–268. [Google Scholar]
  553. Kim, M.-H.; Chung, Y.-T.; Lee, J.-H.; Park, Y.-S.; Shin, M.-K.; Kim, H.-S.; Kim, D.-H.; Lee, H.-Y. Hepatic detoxification activity and reduction of serum alcohol concentration of Hovenia dulcis Thunb from Korea and China. Korean J. Med. Crop Sci. 2000, 8, 225–233. [Google Scholar]
  554. Cha, B.-C.; Lee, E.-H.; Lee, E.; Park, H.-H. Activity of glutathione S-transferase and effect of alcohol decomposition on the fruit of Hovenia dulcis Thunb. Yakhak Hoeji 2004, 48, 213–217. [Google Scholar]
  555. Sung-Hee, K.; Jin-Yeul, M. A study on the extraction and efficacy of bioactive compound from Hovenia dulcis. KSBB J. 2006, 21, 11–15. [Google Scholar]
  556. Hänsel, R.; Sticher, O. Pharmakognosie-Phytopharmazie; Springer: Berlin/Heidelberg, Germany, 2006. [Google Scholar]
  557. Chen, S.; Zhong, G.; Li, A.; Li, S.; Wu, L. Influence of Hovenia dulcis on alcohol concentration in blood and activity of alcohol dehydrogenase (ADH) of animals after drinking. Zhongguo Zhong Yao Za Zhi = Zhongguo Zhongyao Zazhi = China J. Chin. Mater. Medica 2006, 31, 1094–1096. [Google Scholar]
  558. Hase, K.; Ohsugi, M.; Xiong, Q.; Basnet, P.; Kadota, S.; Namba, T. Hepatoprotective effect of Hovenia dulcis Thunb. on experimental liver injuries induced by carbon tetrachloride or D-galactosamine: Lipopolysaccharide. Biol. Pharm. Bull. 1997, 20, 381–385. [Google Scholar] [CrossRef]
  559. Yoshikawa, M.; Murakami, T.; Ueda, T.; Yoshizumi, S.; Ninomiya, K.; Murakami, N.; Matsuda, H.; Saito, M.; Fujii, W.; Tanaka, T. Bioactive constituents of Chinese natural medicines. III. Absolute stereostructures of new dihydroflavonols, hovenitins I, II, and III, isolated from hoveniae semen seu fructus, the seed and fruit of Hovenia dulcis THUNB.(Rhamnaceae): Inhibitory effect on alcohol-induced muscular relaxation and hepatoprotective activity. Yakugaku Zasshi J. Pharm. Soc. Jpn. 1997, 117, 108–118. [Google Scholar]
  560. Ji, Y.; Lu, H. Protective effect of the water extract from Hovenia dulcis Thunb. on CCl4-induced experimental hepatic injury. Lishizhen Med. Mater. Med. Res. 2002, 13, 327–328. [Google Scholar]
  561. Cederbaum, A.I. Introduction: Role of lipid peroxidation and oxidative stress in alcohol toxicity. Free Radic. Biol. Med. 1989, 7, 537–539. [Google Scholar] [CrossRef] [PubMed]
  562. Sultana, R.; Raju, B.S.S.; Sharma, V.; Reddanna, P.; Babu, P.P. Formation of acetaldehyde adducts of glutathione S-transferase A3 in the liver of rats administered alcohol chronically. Alcohol 2005, 35, 57–66. [Google Scholar] [CrossRef] [PubMed]
  563. Jan, G.; Khan, M.A.; Jan, F. Traditional medicinal and economic uses of gymnosperms of Dir Kohistan Valleys, NWFP, Pakistan. Ethnobot. Leafl. 2009, 2009, 9. [Google Scholar]
  564. Acar, İ.; Gül, G.; Örtel, E. Türkiye’de kızılçam ormanlarından akma reçine üretiminde asit pasta tahrik tekniğinin uygulanması esasları üzerine araştırmalar. Ege Or. Ar. En. Müd. Teknik. Bülten 1996, 5, 1–52. [Google Scholar]
  565. Rezzi, S.; Bighelli, A.; Castola, V.; Casanova, J. Composition and chemical variability of the oleoresin of Pinus nigra ssp. laricio from Corsica. Ind. Crops Prod. 2005, 21, 71–79. [Google Scholar] [CrossRef]
  566. Tumen, I.; Reunanen, M. A Comparative Study on Turpentine Oils of Oleoresins of Pinus sylvestris L. from Three Districts of Denizli. Rec. Nat. Prod. 2010, 4, 224–229. [Google Scholar]
  567. Varga, J.; Kocsubé, S.; Tóth, B.; Frisvad, J.C.; Perrone, G.; Susca, A.; Meijer, M.; Samson, R.A. Aspergillus brasiliensis sp. nov., a biseriate black Aspergillus species with world-wide distribution. Int. J. Syst. Evol. Microbiol. 2007, 57, 1925–1932. [Google Scholar] [CrossRef]
  568. Kizil, M.; Kizil, G.; Yavuz, M.; Aytekin, Ç. Antimicrobial Activity of the Tar Obtained from the Roots and Stems of Pinus brutia. Pharm. Biol. 2002, 40, 135–138. [Google Scholar] [CrossRef]
  569. Chammam, A.; Fillaudeau, L.; Romdhane, M.; Bouajila, J. Chemical Composition and In Vitro Bioactivities of Extracts from Cones of P. halepensis, P. brutia, and P. pinea: Insights into Pharmaceutical and Cosmetic Potential. Plants 2024, 13, 1802. [Google Scholar] [CrossRef] [PubMed]
  570. Coppen, J.J.W.; Robinson, J.M.; Kaushal, A.N. Composition of xylem resin from Pinus wallichiana and P. roxburghii. Phytochemistry 1988, 27, 2873–2875. [Google Scholar] [CrossRef]
  571. Willför, S.; Ali, M.; Karonen, M.; Reunanen, M.; Arfan, M.; Harlamow, R. Extractives in bark of different conifer species growing in Pakistan. Holzforschung 2009, 63, 551–558. [Google Scholar] [CrossRef]
  572. Parihar, P.; Parihar, L.; Bohra, A. Antibacterial activity of extracts of Pinus roxburghii Sarg. Bangladesh J. Bot. 2006, 35, 85–86. [Google Scholar]
  573. Zafar, I.; Fatima, A.; Khan, S.; Rehman, Z.; Mehmud, S. GC-MS studies of needles essential oil of Pinus roxburghaii and their antimicrobial activity from. Electron. J. Environ. Agric. Food Chem. 2010, 9, 468–473. [Google Scholar]
  574. Chaudhary, A.K.; Ahmad, S.; Mazumder, A. Study of antibacterial and antifungal activity of traditional Cedrus deodara and Pinus roxburghii Sarg. CellMed 2012, 2, 37.31–37.34. [Google Scholar]
  575. Maimoona, A.; Naeem, I.; Shujaat, S.; Saddiqe, Z.; Mughal, T.; Mehmood, T. Comparison of Radical Scavenging Capacity of Different Extracts of Barks and Needles of Pinus roxburghii and Pinus wallichiana. Asian J. Chem. 2011, 23, 819–822. [Google Scholar]
  576. Kaushik, D.; Kumar, A.; Kaushik, P.; Rana, A.C. Analgesic and Anti-Inflammatory Activity of Pinus roxburghii Sarg. Adv. Pharmacol. Sci. 2012, 2012, 245431. [Google Scholar] [CrossRef]
  577. Kaushik, D.; Kumar, A.; Kaushik, P.; Rana, A.C. Anticonvulsant activity of alcoholic extract of bark of Pinus roxburghii Sarg. Zhong Xi Yi Jie He Xue Bao 2012, 10, 1056–1060. [Google Scholar] [CrossRef]
  578. Rastogi, S.; Shukla, A.; Kolhapure, S. Evaluation of the clinical efficacy and safety of RG-01 (Rumalaya gel) in the management of chronic sub-acute inflammatory joint disorder. Med. Update 2004, 12, 31–37. [Google Scholar]
  579. Ahmad, R.; Rao, R.A.K.; Masood, M.M. Removal and recovery of Cr (VI) from synthetic and industrial wastewater using bark of Pinus roxburghii as an adsorbent. Water Qual. Res. J. 2005, 40, 462–468. [Google Scholar] [CrossRef]
  580. Azmat, A.; Ahmed, K.Z.; Ahmed, M.; Tariq, B. Antinociceptive effects of poly herbal oil extract (PHOE). Pak. J. Pharmacol. 2006, 23, 1–7. [Google Scholar]
  581. Khan, I.; Singh, V.; Chaudhary, A.K. Hepatoprotective activity of Pinus roxburghii Sarg. wood oil against carbon tetrachloride-and ethanol-induced hepatotoxicity. Bangladesh J. Pharmacol. 2012, 7, 94–99. [Google Scholar] [CrossRef]
  582. Ahsan, R.; Islam, M. In vitro antibacterial screening and toxicological study of some useful plants (Cajanus cajan). Eur. J. Sci. Res. 2009, 41, 227–232. [Google Scholar]
  583. Zu, Y.G.; Liu, X.L.; Fu, Y.J.; Wu, N.; Kong, Y.; Wink, M. Chemical composition of the SFE-CO extracts from Cajanus cajan (L.) Huth and their antimicrobial activity in vitro and in vivo. Phytomedicine 2010, 17, 1095–1101. [Google Scholar] [CrossRef] [PubMed]
  584. Arnaiz-Villena, A.; Parga-Lozano, C.; Moreno, E.; Areces, C.; Rey, D.; Gomez-Prieto, P. The Origin of Amerindians and the Peopling of the Americas According to HLA Genes: Admixture with Asian and Pacific People. Curr. Genom. 2010, 11, 103–114. [Google Scholar] [CrossRef]
  585. Chandler, R.F.; Freeman, L.; Hooper, S.N. Herbal remedies of the maritime Indians. J. Ethnopharmacol. 1979, 1, 49–68. [Google Scholar] [CrossRef]
  586. Arnason, T.; Hebda, R.J.; Johns, T. Use of plants for food and medicine by Native Peoples of eastern Canada. Can. J. Bot. 1981, 59, 2189–2325. [Google Scholar] [CrossRef]
  587. Turner, N.J.; Hebda, R.J. Contemporary use of bark for medicine by two salishan native elders of Southeast Vancouver Island, Canada. J. Ethnopharmacol. 1990, 29, 59–72. [Google Scholar] [CrossRef]
  588. Gottesfeld, L.M.J. Wet’Suwet’en ethnobotany: Traditional plant uses. J. Ethnobiol. 1994, 14, 185–210. [Google Scholar]
  589. Uprety, Y.; Asselin, H.; Dhakal, A.; Julien, N. Traditional use of medicinal plants in the boreal forest of Canada: Review and perspectives. J. Ethnobiol. Ethnomed. 2012, 8, 7. [Google Scholar] [CrossRef] [PubMed]
  590. Harris, C.S.; Lambert, J.; Saleem, A.; Coonishish, J.; Martineau, L.C.; Cuerrier, A.; Haddad, P.S.; Arnason, J.T.; Bennett, S.A.L. Antidiabetic Activity of Extracts from Needle, Bark, and Cone of Picea glauca.: Organ-Specific Protection from Glucose Toxicity and Glucose Deprivation. Pharm. Biol. 2008, 46, 126–134. [Google Scholar] [CrossRef]
  591. Mooney, J. The sacred formulas of the Cherokees. In Seventh Annual Report of the Bureau of Ethnology; Powell, J.W., Ed.; Government Printing Office: Washington, DC, USA, 1891; Volume 7, pp. 301–397. [Google Scholar]
  592. Hamel, P.B.; Chiltoskey, M.U. Cherokee Plants and Their Uses: A 400 Year History; Herald Publishing Company: Sylva, NC, USA, 1975. [Google Scholar]
  593. Garrett, J.T. The Cherokee Herbal: Native Plant Medicine from the Four Directions; Simon and Schuster: Rochester, NY, USA, 2003. [Google Scholar]
  594. Setzer, W.N. The Phytochemistry of Cherokee Aromatic Medicinal Plants. Medicines 2018, 5, 121. [Google Scholar] [CrossRef] [PubMed]
  595. Brown, C.L.; Kirkman, L.K. Trees of Georgia and Adjacent States; Timber Press: Portland, OR, USA, 2000. [Google Scholar]
  596. Radford, A.E.; Ahles, H.E.; Bell, C.R. Manual of the Vascular Flora of the Carolinas; Univ of North Carolina Press: Chapel Hill, NC, USA, 2010. [Google Scholar]
  597. Boag, D.; Kiceniuk, J. Protein and caloric content of lodge pole pine needles. For. Chron. 1968, 44, 28–31. [Google Scholar] [CrossRef]
  598. Anäs, E.; Ekman, R.; Holmbom, B. Composition of nonpolar extractives in bark of Norway spruce and Scots pine. J. Wood Chem. Technol. 1983, 3, 119–130. [Google Scholar] [CrossRef]
  599. Fischer, C.; Höll, W. Food reserves of Scots pine (Pinus sylvestris L.) I. Seasonal changes in the carbohydrate and fat reserves of pine needles. Trees 1991, 5, 187–195. [Google Scholar] [CrossRef]
  600. Nergiz, C.; Dönmez, I. Chemical composition and nutritive value of Pinus pinea L. seeds. Food Chem. 2004, 86, 365–368. [Google Scholar] [CrossRef]
  601. Miranda, I.; Gominho, J.; Mirra, I.; Pereira, H. Chemical characterization of barks from Picea abies and Pinus sylvestris after fractioning into different particle sizes. Ind. Crops Prod. 2012, 36, 395–400. [Google Scholar] [CrossRef]
  602. Angel, H.Z.; Priest, J.S.; Stovall, J.P.; Oswald, B.P.; Weng, Y.; Williams, H.M. Individual tree and stand-level carbon and nutrient contents across one rotation of loblolly pine plantations on a reclaimed surface mine. New For. 2019, 50, 733–753. [Google Scholar] [CrossRef]
  603. Dziedzinski, M.; Kobus-Cisowska, J.; Szymanowska, D.; Stuper-Szablewska, K.; Baranowska, M. Identification of polyphenols from coniferous shoots as natural antioxidants and antimicrobial compounds. Molecules 2020, 25, 3527. [Google Scholar] [CrossRef]
  604. Nedelea, D.-G.; Vulpe, D.E.; Viscopoleanu, G.; Radulescu, A.C.; Mihailescu, A.A.; Gradinaru, S.; Orghidan, M.; Scheau, C.; Cergan, R.; Dragosloveanu, S. Progressive Thoracolumbar Tuberculosis in a Young Male: Diagnostic, Therapeutic, and Surgical Insights. Infect. Dis. Rep. 2024, 16, 1005–1016. [Google Scholar] [CrossRef] [PubMed]
  605. Shah, M.; Reed, C. Complications of tuberculosis. Curr. Opin. Infect. Dis. 2014, 27, 403–410. [Google Scholar] [CrossRef]
  606. Nadgir, C.A.; Biswas, D.A. Antibiotic Resistance and Its Impact on Disease Management. Cureus 2023, 15, e38251. [Google Scholar] [CrossRef] [PubMed]
  607. Vitiello, A.; Ferrara, F.; Boccellino, M.; Ponzo, A.; Cimmino, C.; Comberiati, E.; Zovi, A.; Clemente, S.; Sabbatucci, M. Antifungal Drug Resistance: An Emergent Health Threat. Biomedicines 2023, 11, 1063. [Google Scholar] [CrossRef] [PubMed]
  608. Herrick, J.A.; Nordstrom, M.; Maloney, P.; Rodriguez, M.; Naceanceno, K.; Gallo, G.; Mejia, R.; Hershow, R. Parasitic infections represent a significant health threat among recent immigrants in Chicago. Parasitol. Res. 2020, 119, 1139–1148. [Google Scholar] [CrossRef] [PubMed]
  609. Ahmed, M. Intestinal Parasitic Infections in 2023. Gastroenterol. Res 2023, 16, 127–140. [Google Scholar] [CrossRef] [PubMed]
  610. Mathison, B.A.; Pritt, B.S. The Landscape of Parasitic Infections in the United States. Mod. Pathol. 2023, 36, 100217. [Google Scholar] [CrossRef]
  611. Mishra, B.; Rath, S.; Mohanty, M.; Mohapatra, P.R. The Threat of Impending Pandemics: A Proactive Approach. Cureus 2023, 15, e36723. [Google Scholar] [CrossRef]
  612. Casadevall, A. Pandemics past, present, and future: Progress and persistent risks. J. Clin. Investig. 2024, 134, e179519. [Google Scholar] [CrossRef]
  613. Scott, H.F.; Greenwald, E.E.; Bajaj, L.; Davies, S.J.D.; Brou, L.; Kempe, A. The sensitivity of clinician diagnosis of sepsis in tertiary and community-based emergency settings. J. Pediatr. 2018, 195, 220–227.e221. [Google Scholar] [CrossRef] [PubMed]
  614. Wentowski, C.; Mewada, N.; Nielsen, N.D. Sepsis in 2018: A review. Anaesth. Intensive Care Med. 2019, 20, 6–13. [Google Scholar] [CrossRef]
  615. Cuesta-Montero, P.; Navarro-Martínez, J.; Yedro, M.; Galiana-Ivars, M. Sepsis and Clinical Simulation: What Is New?(and Old). J. Pers. Med. 2023, 13, 1475. [Google Scholar] [CrossRef] [PubMed]
  616. Gorecki, G.P.; Tomescu, D.R.; Pleș, L.; Panaitescu, A.M.; Dragosloveanu, Ș.; Scheau, C.; Sima, R.M.; Coman, I.S.; Grigorean, V.T.; Cochior, D. Implications of using artificial intelligence in the diagnosis of sepsis/sepsis shock. Germs 2024, 14, 77–84. [Google Scholar] [CrossRef]
  617. Couce, A.; Blázquez, J. Side effects of antibiotics on genetic variability. FEMS Microbiol. Rev. 2009, 33, 531–538. [Google Scholar] [CrossRef] [PubMed]
  618. McManus, D.S.; Shah, S. Antifungal drugs. Side Eff. Drugs Annu. 2019, 41, 285–292. [Google Scholar]
  619. Houšť, J.; Spížek, J.; Havlíček, V. Antifungal drugs. Metabolites 2020, 10, 106. [Google Scholar] [CrossRef] [PubMed]
  620. De Clercq, E. Antiviral drugs: Current state of the art. J. Clin. Virol. 2001, 22, 73–89. [Google Scholar] [CrossRef]
  621. Abers, M.S.; Shandera, W.X.; Kass, J.S. Neurological and psychiatric adverse effects of antiretroviral drugs. CNS Drugs 2014, 28, 131–145. [Google Scholar] [CrossRef]
  622. Rosenblatt, J.E. Antiparasitic agents. In Mayo Clinic Proceedings; Elsevier: Amsterdam, The Netherlands, 2019; pp. 1161–1174. [Google Scholar]
  623. Baggish, A.L.; Hill, D.R. Antiparasitic agent atovaquone. Antimicrob. Agents Chemother. 2002, 46, 1163–1173. [Google Scholar] [CrossRef]
  624. Kappagoda, S.; Singh, U.; Blackburn, B.G. Antiparasitic therapy. In Mayo Clinic Proceedings; Elsevier: Amsterdam, The Netherlands, 2019; pp. 561–583. [Google Scholar]
  625. Periferakis, A.; Caruntu, A.; Periferakis, A.-T.; Scheau, A.-E.; Badarau, I.A.; Caruntu, C.; Scheau, C. Availability, Toxicology and Medical Significance of Antimony. Int. J. Environ. Res. Public. Health 2022, 19, 4669. [Google Scholar] [CrossRef]
  626. Tan, B.L.; Norhaizan, M.E.; Liew, W.-P.-P.; Sulaiman Rahman, H. Antioxidant and oxidative stress: A mutual interplay in age-related diseases. Front. Pharmacol. 2018, 9, 1162. [Google Scholar] [CrossRef] [PubMed]
  627. Belenguer-Varea, Á.; Tarazona-Santabalbina, F.J.; Avellana-Zaragoza, J.A.; Martínez-Reig, M.; Mas-Bargues, C.; Inglés, M. Oxidative stress and exceptional human longevity: Systematic review. Free Radic. Biol. Med. 2020, 149, 51–63. [Google Scholar] [CrossRef]
  628. Spector, A. Review: Oxidative Stress and Disease. J. Ocul. Pharmacol. Ther. 2000, 16, 193–201. [Google Scholar] [CrossRef] [PubMed]
  629. Shoham, A.; Hadziahmetovic, M.; Dunaief, J.L.; Mydlarski, M.B.; Schipper, H.M. Oxidative stress in diseases of the human cornea. Free Radic. Biol. Med. 2008, 45, 1047–1055. [Google Scholar] [CrossRef]
  630. Căruntu, C.; Boda, D.; Musat, S.; Căruntu, A.; Mandache, E. Stress-induced mast cell activation in glabrous and hairy skin. Mediat. Inflamm. 2014, 2014, 105950. [Google Scholar] [CrossRef]
  631. Liguori, I.; Russo, G.; Curcio, F.; Bulli, G.; Aran, L.; Della-Morte, D.; Gargiulo, G.; Testa, G.; Cacciatore, F.; Bonaduce, D.; et al. Oxidative stress, aging, and diseases. Clin. Interv. Aging 2018, 13, 757–772. [Google Scholar] [CrossRef]
  632. Mateo, R.; Beyer, W.N.; Spann, J.; Hoffman, D.; Ramis, A. Relationship Between Oxidative Stress, Pathology, and Behavioral Signs of Lead Poisoning in Mallards. J. Toxicol. Environ. Health Part A 2003, 66, 1371–1389. [Google Scholar] [CrossRef]
  633. Farina, M.; Avila, D.S.; da Rocha, J.B.T.; Aschner, M. Metals, oxidative stress and neurodegeneration: A focus on iron, manganese and mercury. Neurochem. Int. 2013, 62, 575–594. [Google Scholar] [CrossRef] [PubMed]
  634. Baumgart, D.C.; Carding, S.R. Inflammatory bowel disease: Cause and immunobiology. Lancet 2007, 369, 1627–1640. [Google Scholar] [CrossRef]
  635. Sands, B.E. Biomarkers of inflammation in inflammatory bowel disease. Gastroenterology 2015, 149, 1275–1285.e1272. [Google Scholar] [CrossRef]
  636. Ramos, G.P.; Papadakis, K.A. Mechanisms of disease: Inflammatory bowel diseases. In Mayo Clinic Proceedings; Elsevier: Amsterdam, The Netherlands, 2019; pp. 155–165. [Google Scholar]
  637. Păsărică, M.A.; Curcă, P.F.; Dragosloveanu, C.D.M.; Tătaru, C.I.; Manole, I.R.; Murgoi, G.E.; Grigorescu, A.C. Underlying Ciliary Body Uveal Melanoma in a Patient with Chronic Lymphocytic Leukemia Presenting for Hyphema. Diagnostics 2022, 12, 1312. [Google Scholar] [CrossRef] [PubMed]
  638. Păsărică, M.A.; Curcă, P.F.; Burcea, M.; Schmitzer, S.; Dragosloveanu, C.D.M.; Grigorescu, A.C. The Effects of Oncological Treatment on Redox Balance in Patients with Uveal Melanoma. Diagnostics 2023, 13, 1907. [Google Scholar] [CrossRef]
  639. Păsărică, M.A.; Curcă, P.F.; Dragosloveanu, C.D.M.; Grigorescu, A.C.; Nisipașu, C.I. Pathological and Molecular Diagnosis of Uveal Melanoma. Diagnostics 2024, 14, 958. [Google Scholar] [CrossRef] [PubMed]
  640. Caruntu, C.; Boda, D.; Constantin, C.; Caruntu, A.; Neagu, M. Catecholamines Increase in Vitro Proliferation of Murine B16F10 Melanoma Cells. Acta Endocrinol. 2014, 10, 545–558. [Google Scholar] [CrossRef]
  641. Zabaleta, J. Multifactorial etiology of gastric cancer. Cancer Epigenetics: Methods Protocols; Springer: Berlin/Heidelberg, Germany, 2012; pp. 411–435. [Google Scholar]
  642. Ion, A.; Popa, I.M.; Papagheorghe, L.M.; Lisievici, C.; Lupu, M.; Voiculescu, V.; Caruntu, C.; Boda, D. Proteomic Approaches to Biomarker Discovery in Cutaneous T-Cell Lymphoma. Dis. Markers 2016, 2016, 9602472. [Google Scholar] [CrossRef]
  643. Hausman, D.M. What is cancer? Perspect. Biol. Med. 2019, 62, 778–784. [Google Scholar] [CrossRef] [PubMed]
  644. Tampa, M.; Georgescu, S.R.; Mitran, C.I.; Mitran, M.I.; Matei, C.; Scheau, C.; Constantin, C.; Neagu, M. Recent Advances in Signaling Pathways Comprehension as Carcinogenesis Triggers in Basal Cell Carcinoma. J. Clin. Med. 2020, 9, 3010. [Google Scholar] [CrossRef]
  645. Wan, M.-l.; Wang, Y.; Zeng, Z.; Deng, B.; Zhu, B.-s.; Cao, T.; Li, Y.-k.; Xiao, J.; Han, Q.; Wu, Q. Colorectal cancer (CRC) as a multifactorial disease and its causal correlations with multiple signaling pathways. Biosci. Rep. 2020, 40, BSR20200265. [Google Scholar] [CrossRef]
  646. Tampa, M.; Mitran, C.I.; Mitran, M.I.; Nicolae, I.; Dumitru, A.; Matei, C.; Manolescu, L.; Popa, G.L.; Caruntu, C.; Georgescu, S.R. The Role of Beta HPV Types and HPV-Associated Inflammatory Processes in Cutaneous Squamous Cell Carcinoma. J. Immunol. Res. 2020, 2020, 5701639. [Google Scholar] [CrossRef]
  647. Scheau, C.; Draghici, C.; Ilie, M.A.; Lupu, M.; Solomon, I.; Tampa, M.; Georgescu, S.R.; Caruntu, A.; Constantin, C.; Neagu, M.; et al. Neuroendocrine Factors in Melanoma Pathogenesis. Cancers 2021, 13, 2277. [Google Scholar] [CrossRef]
  648. Caruntu, A.; Moraru, L.; Surcel, M.; Munteanu, A.; Costache, D.O.; Tanase, C.; Constantin, C.; Scheau, C.; Neagu, M.; Caruntu, C. Persistent Changes of Peripheral Blood Lymphocyte Subsets in Patients with Oral Squamous Cell Carcinoma. Healthcare 2022, 10, 342. [Google Scholar] [CrossRef] [PubMed]
  649. Cretu, B.; Zamfir, A.; Bucurica, S.; Scheau, A.E.; Savulescu Fiedler, I.; Caruntu, C.; Caruntu, A.; Scheau, C. Role of Cannabinoids in Oral Cancer. Int. J. Mol. Sci. 2024, 25, 969. [Google Scholar] [CrossRef] [PubMed]
  650. Periferakis, A.; Paresoglou, I.; Paresoglou, N. The significance of the Lavrion mines in Greek and European Geoheritage. Eur. Geol. 2019, 48, 24–27. [Google Scholar]
  651. Matei, A.-M.; Caruntu, C.; Tampa, M.; Georgescu, S.R.; Matei, C.; Constantin, M.M.; Constantin, T.V.; Calina, D.; Ciubotaru, D.A.; Badarau, I.A. Applications of nanosized-lipid-based drug delivery systems in wound care. Appl. Sci. 2021, 11, 4915. [Google Scholar] [CrossRef]
  652. Zhen, X.; Xie, C.; Jiang, Y.; Ai, X.; Xing, B.; Pu, K. Semiconducting Photothermal Nanoagonist for Remote-Controlled Specific Cancer Therapy. Nano Lett. 2018, 18, 1498–1505. [Google Scholar] [CrossRef]
  653. Bhagwat, D.A.; Swami, P.A.; Nadaf, S.J.; Choudhari, P.B.; Kumbar, V.M.; More, H.N.; Killedar, S.G.; Kawtikwar, P.S. Capsaicin Loaded Solid SNEDDS for Enhanced Bioavailability and Anticancer Activity: In-Vitro, In-Silico, and In-Vivo Characterization. J. Pharm. Sci. 2021, 110, 280–291. [Google Scholar] [CrossRef]
  654. Xu, M.; Zhang, J.; Mu, Y.; Foda, M.F.; Han, H. Activation of TRPV1 by capsaicin-loaded CaCO(3) nanoparticle for tumor-specific therapy. Biomaterials 2022, 284, 121520. [Google Scholar] [CrossRef]
  655. Timofticiuc, I.A.; Călinescu, O.; Iftime, A.; Dragosloveanu, S.; Caruntu, A.; Scheau, A.E.; Badarau, I.A.; Didilescu, A.C.; Caruntu, C.; Scheau, C. Biomaterials Adapted to Vat Photopolymerization in 3D Printing: Characteristics and Medical Applications. J. Funct. Biomater. 2023, 15, 7. [Google Scholar] [CrossRef]
  656. Chae, S.; Cho, D.W. Biomaterial-based 3D bioprinting strategy for orthopedic tissue engineering. Acta Biomater. 2023, 156, 4–20. [Google Scholar] [CrossRef]
  657. Timofticiuc, I.-A.; Dragosloveanu, S.; Caruntu, A.; Scheau, A.-E.; Badarau, I.A.; Garofil, N.D.; Didilescu, A.C.; Caruntu, C.; Scheau, C. 3D Bioprinting in Limb Salvage Surgery. J. Funct. Biomater. 2024, 15, 383. [Google Scholar] [CrossRef]
  658. He, Y.; Chen, J.; Chen, Y.; Qian, H. Effect of Operating Room Nursing Management on Nosocomial Infection in Orthopedic Surgery: A Meta-Analysis. J. Healthc. Eng. 2022, 2022, 4193932. [Google Scholar] [CrossRef]
  659. Dragosloveanu, S.; Petre, M.A.; Capitanu, B.S.; Dragosloveanu, C.D.M.; Cergan, R.; Scheau, C. Initial Learning Curve for Robot-Assisted Total Knee Arthroplasty in a Dedicated Orthopedics Center. J. Clin. Med. 2023, 12, 6950. [Google Scholar] [CrossRef] [PubMed]
  660. Gaudias, J. Antibiotic prophylaxis in orthopedics-traumatology. Orthop. Traumatol. Surg. Res. 2021, 107, 102751. [Google Scholar] [CrossRef] [PubMed]
  661. Dragosloveanu, S.; Petre, M.-A.; Gherghe, M.E.; Nedelea, D.-G.; Scheau, C.; Cergan, R. Overall Accuracy of Radiological Digital Planning for Total Hip Arthroplasty in a Specialized Orthopaedics Hospital. J. Clin. Med. 2023, 12, 4503. [Google Scholar] [CrossRef] [PubMed]
  662. Barros, J.; Monteiro, F.J.; Ferraz, M.P. Bioengineering Approaches to Fight against Orthopedic Biomaterials Related-Infections. Int. J. Mol. Sci. 2022, 23, 11658. [Google Scholar] [CrossRef]
  663. Dragosloveanu, S.; Petre, M.-A.; Gherghe, M.E.; Baz, R.O.; Cergan, R.; Scheau, C. Elbow Reconstruction with Megaprosthesis: An Effective Strategy for Salvage Surgery in Trauma Patients. Diagnostics 2024, 14, 724. [Google Scholar] [CrossRef]
  664. Yuan, S.; Zhang, T.; Zhang, D.; He, Q.; Du, M.; Zeng, F. Impact of negative pressure wound treatment on incidence of surgical site infection in varied orthopedic surgeries: A systematic review and meta-analysis. Int. Wound J. 2023, 20, 2334–2345. [Google Scholar] [CrossRef]
  665. Bahadır, B.; Atik, O.; Kanatlı, U.; Sarıkaya, B. A brief introduction to medical image processing, designing and 3D printing for orthopedic surgeons. Jt. Dis. Relat. Surg. 2023, 34, 451–454. [Google Scholar] [CrossRef]
  666. Dragosloveanu, Ș.; Dragosloveanu, C.D.; Cotor, D.C.; Stoica, C.I. Short vs. long intramedullary nail systems in trochanteric fractures: A randomized prospective single center study. Exp. Ther. Med. 2022, 23, 1–5. [Google Scholar] [CrossRef]
  667. Huang, H.K.; Aberle, D.R.; Lufkin, R.; Grant, E.G.; Hanafee, W.N.; Kangarloo, H. Advances in medical imaging. Ann. Intern. Med. 1990, 112, 203–220. [Google Scholar] [CrossRef]
  668. Căruntu, C.; Boda, D.; Căruntu, A.; Rotaru, M.; Baderca, F.; Zurac, S. In vivo imaging techniques for psoriatic lesions. Rom. J. Morphol. Embryol. 2014, 55, 1191–1196. [Google Scholar] [PubMed]
  669. Periferakis, A.; Bolocan, A.; Ion, D. A Review of Innovation in Medicine. Technol. Innov. Life Sci. 2022, 1, 42–48. [Google Scholar] [CrossRef]
  670. Enyedi, M.; Scheau, C.; Baz, R.O.; Didilescu, A.C. Circle of Willis: Anatomical variations of configuration. A magnetic resonance angiography study. Folia Morphol. 2023, 82, 24–29. [Google Scholar] [CrossRef] [PubMed]
  671. Dragosloveanu, S.; Capitanu, B.-S.; Josanu, R.; Vulpe, D.; Cergan, R.; Scheau, C. Radiological Assessment of Coronal Plane Alignment of the Knee Phenotypes in the Romanian Population. J. Clin. Med. 2024, 13, 4223. [Google Scholar] [CrossRef]
  672. Nedelea, D.-G.; Vulpe, D.; Enyedi, M.; Cergan, R.; Scheau, C.; Baz, R.O.; Dragosloveanu, S. Comprehensive Approach of the Diagnosis, Treatment, and Medical Rehabilitation of Patients with Spondylolisthesis. Balneo PRM Res. J. 2024, 15, 2. [Google Scholar] [CrossRef]
  673. Rong, J.; Liu, Y. Advances in medical imaging techniques. BMC Methods 2024, 1, 10. [Google Scholar] [CrossRef]
  674. Petran, E.M.; Periferakis, A.; Troumpata, L.; Periferakis, A.-T.; Scheau, A.-E.; Badarau, I.A.; Periferakis, K.; Caruntu, A.; Savulescu-Fiedler, I.; Sima, R.-M.; et al. Capsaicin: Emerging Pharmacological and Therapeutic Insights. Curr. Issues Mol. Biol. 2024, 46, 7895–7943. [Google Scholar] [CrossRef] [PubMed]
  675. Dixon, J.B. The effect of obesity on health outcomes. Mol. Cell. Endocrinol. 2010, 316, 104–108. [Google Scholar] [CrossRef] [PubMed]
  676. Norman, J.E. The adverse effects of obesity on reproduction. Reproduction 2010, 140, 343. [Google Scholar] [CrossRef]
  677. Lam, B.C.C.; Lim, A.Y.L.; Chan, S.L.; Yum, M.P.S.; Koh, N.S.Y.; Finkelstein, E.A. The impact of obesity: A narrative review. Singap. Med. J. 2023, 64, 163–171. [Google Scholar] [CrossRef]
  678. Savulescu-Fiedler, I.; Mihalcea, R.; Dragosloveanu, S.; Scheau, C.; Baz, R.O.; Caruntu, A.; Scheau, A.-E.; Caruntu, C.; Benea, S.N. The Interplay between Obesity and Inflammation. Life 2024, 14, 856. [Google Scholar] [CrossRef] [PubMed]
  679. Neagu, M.; Constantin, C.; Surcel, M.; Munteanu, A.; Scheau, C.; Savulescu-Fiedler, I.; Caruntu, C. Diabetic neuropathy: A NRF2 disease? J. Diabetes 2024, 16, e13524. [Google Scholar] [CrossRef]
  680. Pouwer, F.; Nefs, G.; Nouwen, A. Adverse effects of depression on glycemic control and health outcomes in people with diabetes: A review. Endocrinol. Metab. Clin. 2013, 42, 529–544. [Google Scholar] [CrossRef] [PubMed]
  681. Labib, A.-M.; Martins, A.P.; Raposo, J.F.; Torre, C. The association between polypharmacy and adverse health consequences in elderly type 2 diabetes mellitus patients; a systematic review and meta-analysis. Diabetes Res. Clin. Pract. 2019, 155, 107804. [Google Scholar]
  682. King, K.; Rosenthal, A. The adverse effects of diabetes on osteoarthritis: Update on clinical evidence and molecular mechanisms. Osteoarthr. Cartil. 2015, 23, 841–850. [Google Scholar] [CrossRef]
  683. Kubyshkin, A.; Shevandova, A.; Petrenko, V.; Fomochkina, I.; Sorokina, L.; Kucherenko, A.; Gordienko, A.; Khimich, N.; Zyablitskaya, E.; Makalish, T. Anti-inflammatory and antidiabetic effects of grape-derived stilbene concentrate in the experimental metabolic syndrome. J. Diabetes Metab. Disord. 2020, 19, 1205–1214. [Google Scholar] [CrossRef]
  684. Choi, S.Z.; Lee, S.O.; Jang, K.U.; Chung, S.H.; Park, S.H.; Kang, H.C.; Yang, E.Y.; Cho, H.J.; Lee, K.R. Antidiabetic stilbene and anthraquinone derivatives from Rheum undulatum. Arch. Pharmacal Res. 2005, 28, 1027–1030. [Google Scholar] [CrossRef]
  685. Peng, J.; Lu, C.; Luo, Y.; Su, X.; Li, S.; Ho, C.-T. Hypoglycemic effects and associated mechanisms of resveratrol and related stilbenes in diet. Food Funct. 2024, 15, 2381–2405. [Google Scholar] [CrossRef]
  686. Abbas, A.M. Cardioprotective effect of resveratrol analogue isorhapontigenin versus omega-3 fatty acids in isoproterenol-induced myocardial infarction in rats. J. Physiol. Biochem. 2016, 72, 469–484. [Google Scholar] [CrossRef]
  687. Riba, A.; Deres, L.; Sumegi, B.; Toth, K.; Szabados, E.; Halmosi, R. Cardioprotective Effect of Resveratrol in a Postinfarction Heart Failure Model. Oxid. Med. Cell. Longev. 2017, 2017, 6819281. [Google Scholar] [CrossRef]
  688. Teka, T.; Zhang, L.; Ge, X.; Li, Y.; Han, L.; Yan, X. Stilbenes: Source plants, chemistry, biosynthesis, pharmacology, application and problems related to their clinical Application-A comprehensive review. Phytochemistry 2022, 197, 113128. [Google Scholar] [CrossRef] [PubMed]
  689. Liu, Y.; Ma, W.; Zhang, P.; He, S.; Huang, D. Effect of resveratrol on blood pressure: A meta-analysis of randomized controlled trials. Clin. Nutr. 2015, 34, 27–34. [Google Scholar] [CrossRef] [PubMed]
  690. Savulescu-Fiedler, I.; Dorobantu-Lungu, L.R.; Dragosloveanu, S.; Benea, S.N.; Dragosloveanu, C.D.M.; Caruntu, A.; Scheau, A.E.; Caruntu, C.; Scheau, C. The Cross-Talk Between the Peripheral and Brain Cholesterol Metabolisms. Curr. Issues Mol. Biol. 2025, 47, 115. [Google Scholar] [CrossRef]
  691. Li, X.; Dai, Y.; Yan, S.; Shi, Y.; Li, J.; Liu, J.; Cha, L.; Mu, J. Resveratrol lowers blood pressure in spontaneously hypertensive rats via calcium-dependent endothelial NO production. Clin. Exp. Hypertens. 2016, 38, 287–293. [Google Scholar] [CrossRef] [PubMed]
  692. Colica, C.; Milanović, M.; Milić, N.; Aiello, V.; De Lorenzo, A.; Abenavoli, L. A Systematic Review on Natural Antioxidant Properties of Resveratrol. Nat. Prod. Commun. 2018, 13, 1934578X1801300923. [Google Scholar] [CrossRef]
  693. Choi, K. A Pilot Study of the Total Cholesterol/High-Density Lipoprotein Ratio as a Prognostic Indicator of Hyperlipidemia-Related Diseases in Dogs and Cats. Curr. Issues Mol. Biol. 2024, 46, 12174–12182. [Google Scholar] [CrossRef]
  694. Park, J.; Pyee, J.; Park, H. Pinosylvin at a high concentration induces AMPK-mediated autophagy for preventing necrosis in bovine aortic endothelial cells. Can. J. Physiol. Pharmacol. 2014, 92, 993–999. [Google Scholar] [CrossRef]
  695. Kungwani, N.; Singh, P.; Kumar, V. Medicinal plants: Adjunct treatment to tuberculosis chemotherapy to prevent hepatic damage. J. Ayurveda Integr. Med. 2019, 11, 522–528. [Google Scholar] [CrossRef]
  696. Madrigal-Santillán, E.; Madrigal-Bujaidar, E.; Álvarez-González, I.; Sumaya-Martínez, M.T.; Gutiérrez-Salinas, J.; Bautista, M.; Morales-González, Á.; García-Luna y González-Rubio, M.; Aguilar-Faisal, J.L.; Morales-González, J.A. Review of natural products with hepatoprotective effects. World J. Gastroenterol. 2014, 20, 14787–14804. [Google Scholar] [CrossRef]
  697. Zhang, C.; Cui, S.; Mao, G.; Li, G. Clinical characteristics and the risk factors of hepatic injury in 221 children with infectious mononucleosis. Front. Pediatr. 2022, 9, 809005. [Google Scholar] [CrossRef]
  698. Shen, R.; Zhou, Y.; Zhang, L.; Yang, S. The value of bile acid spectrum in the evaluation of hepatic injury in children with infectious mononucleosis caused by Epstein Barr virus infection. Front. Pediatr. 2023, 11, 1109762. [Google Scholar] [CrossRef] [PubMed]
  699. Ramadori, G.; Moriconi, F.; Malik, I.; Dudas, J. Physiology and pathophysiology of liver inflammation, damage and repair. J. Physiol. Pharmacol. 2008, 59, 107–117. [Google Scholar]
  700. Dong, V.; Nanchal, R.; Karvellas, C.J. Pathophysiology of acute liver failure. Nutr. Clin. Pract. 2020, 35, 24–29. [Google Scholar] [CrossRef]
  701. Pinzani, M. Pathophysiology of liver fibrosis. Dig. Dis. 2015, 33, 492–497. [Google Scholar] [CrossRef] [PubMed]
  702. Periferakis, A.; Tsigas, G.; Periferakis, A.-T.; Badarau, I.A.; Scheau, A.-E.; Tampa, M.; Georgescu, S.R.; Didilescu, A.C.; Scheau, C.; Caruntu, C. Antitumoral and Anti-inflammatory Roles of Somatostatin and Its Analogs in Hepatocellular Carcinoma. Anal. Cell. Pathol. 2021, 2021, 1840069. [Google Scholar] [CrossRef]
  703. Periferakis, A.; Tsigas, G.; Periferakis, A.-T.; Tone, C.M.; Hemes, D.A.; Periferakis, K.; Troumpata, L.; Badarau, I.A.; Scheau, C.; Caruntu, A.; et al. Agonists, Antagonists and Receptors of Somatostatin: Pathophysiological and Therapeutical Implications in Neoplasias. Curr. Issues Mol. Biol. 2024, 46, 9721–9759. [Google Scholar] [CrossRef] [PubMed]
  704. Scheau, A.-E.; Jurca, S.O.; Scheau, C.; Lupescu, I.G. Non-Surgical Treatment for Hepatocellular Carcinoma: What to Expect at Follow-Up Magnetic Resonance Imaging—A Pictorial Review. Appl. Sci. 2024, 14, 9159. [Google Scholar] [CrossRef]
  705. Roberti, M.; Pizzirani, D.; Simoni, D.; Rondanin, R.; Baruchello, R.; Bonora, C.; Buscemi, F.; Grimaudo, S.; Tolomeo, M. Synthesis and biological evaluation of resveratrol and analogues as apoptosis-inducing agents. J. Med. Chem. 2003, 46, 3546–3554. [Google Scholar] [CrossRef]
  706. Waffo-Téguo, P.; Hawthorne, M.E.; Cuendet, M.; Mérillon, J.M.; Kinghorn, A.D.; Pezzuto, J.M.; Mehta, R.G. Potential cancer-chemopreventive activities of wine stilbenoids and flavans extracted from grape (Vitis vinifera) cell cultures. Nutr. Cancer 2001, 40, 173–179. [Google Scholar] [CrossRef]
  707. Shagufta, A.I. Tamoxifen a pioneering drug: An update on the therapeutic potential of tamoxifen derivatives. Eur. J. Med. Chem. 2018, 143, 515–531. [Google Scholar] [CrossRef]
  708. Pecyna, P.; Wargula, J.; Murias, M.; Kucinska, M. More Than Resveratrol: New Insights into Stilbene-Based Compounds. Biomolecules 2020, 10, 1111. [Google Scholar] [CrossRef]
  709. Mendoza, R.G. Lords of the medicine bag: Medical science and traditional practice in ancient Peru and South America. In Medicine across Cultures: History and Practice of Medicine in Non-Western Cultures; Springer: Berlin/Heidelberg, Germany, 2003; pp. 225–257. [Google Scholar]
  710. Elferink, J.G. The Inca healer: Empirical medical knowledge and magic in pre-Columbian Peru. Rev. De Indias 2015, 75, 323–350. [Google Scholar] [CrossRef]
  711. Periferakis, A. A review of obsidian source exploitation in pre-columbian south America. Bull. Geol. Soc. Greece 2019, 55, 65–108. [Google Scholar] [CrossRef]
  712. Guerra, F. Aztec medicine. Med. Hist. 1966, 10, 315–338. [Google Scholar] [CrossRef] [PubMed]
  713. Musso, C.G. Finding the present in the past: Ancient Mexican medicine. Hum. Med. 2004, 2, 1–3. [Google Scholar]
  714. Maher, P. A review of ‘traditional’Aboriginal health beliefs. Aust. J. Rural Health 1999, 7, 229–236. [Google Scholar] [CrossRef]
  715. Oliver, S.J. The role of traditional medicine practice in primary health care within Aboriginal Australia: A review of the literature. J. Ethnobiol. Ethnomed. 2013, 9, 1–8. [Google Scholar] [CrossRef]
  716. Pieroni, A. Medicinal plants and food medicines in the folk traditions of the upper Lucca Province, Italy. J. Ethnopharmacol. 2000, 70, 235–273. [Google Scholar] [CrossRef]
  717. Ramalingum, N.; Mahomoodally, M.F. The therapeutic potential of medicinal foods. Adv. Pharmacol. Pharm. Sci. 2014, 2014, 354264. [Google Scholar] [CrossRef]
  718. Brain, A.; John, M. Phenolic content and antioxidant activity of selected Ugandan traditional medicinal foods. Afr. J. Food Sci. 2014, 8, 427–434. [Google Scholar]
  719. Tavakkoli-Kakhki, M.; Motavasselian, M.; Mosaddegh, M.; Esfahani, M.M.; Kamalinejad, M.; Nematy, M.; Eslami, S. Omega-3 and omega-6 content of medicinal foods for depressed patients: Implications from the Iranian Traditional Medicine. Avicenna J. Phytomed. 2014, 4, 225. [Google Scholar]
  720. Hou, Y.; Wang, X.; Zhang, Y.; Wang, S.; Meng, X. Highland mate: Edible and functional foods in traditional medicine for the prevention and treatment of hypoxia-related symptoms. Curr. Opin. Pharmacol. 2021, 60, 306–314. [Google Scholar] [CrossRef] [PubMed]
  721. Garbowski, T.; Charazińska, S.; Pulikowski, K.; Wiercik, P. Application of microalgae cultivated on pine bark for the treatment of municipal wastewater in cylindrical photobioreactors. Water Environ. J. 2020, 34, 949–959. [Google Scholar] [CrossRef]
  722. Sousa, S.; Jiménez-Guerrero, P.; Ruiz, A.; Ratola, N.; Alves, A. Organochlorine pesticides removal from wastewater by pine bark adsorption after activated sludge treatment. Environ. Technol. 2011, 32, 673–683. [Google Scholar] [CrossRef] [PubMed]
  723. Peterson, S.B.; Teal, J.M. The role of plants in ecologically engineered wastewater treatment systems. Ecol. Eng. 1996, 6, 137–148. [Google Scholar] [CrossRef]
  724. Eisenberg, D.; Soller, J.; Sakaji, R.; Olivieri, A. A methodology to evaluate water and wastewater treatment plant reliability. Water Sci. Technol. 2001, 43, 91–99. [Google Scholar] [CrossRef]
  725. Alasino, N.; Mussati, M.C.; Scenna, N. Wastewater treatment plant synthesis and design. Ind. Eng. Chem. Res. 2007, 46, 7497–7512. [Google Scholar] [CrossRef]
  726. Periferakis, A. The Yukon Gold Rush: Early Examples of the Socioeconomic and Environmental Impact of Mining. In Proceedings of the 15th International Congress of the Geological Society of Greece, Athens, Greece, 22–24 May 2019; pp. 710–711. [Google Scholar]
  727. Periferakis, A. The Keramos Antimonite Mines in Chios Island, Greece: Mining History and Current Situation. News Miner. 2020, 35, 5–21. [Google Scholar]
  728. Lombardi, G.; Lanzirotti, A.; Qualls, C.; Socola, F.; Ali, A.M.; Appenzeller, O. Five hundred years of mercury exposure and adaptation. J. Biomed. Biotechnol. 2012, 2012, 472858. [Google Scholar] [CrossRef]
  729. Clackett, S.P.; Porter, T.J.; Lehnherr, I. The tree-ring mercury record of Klondike gold mining at Bear Creek, central Yukon. Environ. Pollut. 2021, 268, 115777. [Google Scholar] [CrossRef]
  730. Strosnider, W.H.J.; Llanos López, F.S.; LaBar, J.A.; Palmer, K.J.; Nairn, R.W. Unabated acid mine drainage from Cerro Rico de Potosí, Bolivia: Uncommon constituents of concern impact the Rio Pilcomayo headwaters. Environ. Earth Sci. 2014, 71, 3223–3234. [Google Scholar] [CrossRef]
  731. Haghighizadeh, A.; Rajabi, O.; Nezarat, A.; Hajyani, Z.; Haghmohammadi, M.; Hedayatikhah, S.; Asl, S.D.; Aghababai Beni, A. Comprehensive analysis of heavy metal soil contamination in mining Environments: Impacts, monitoring Techniques, and remediation strategies. Arab. J. Chem. 2024, 17, 105777. [Google Scholar] [CrossRef]
  732. Stamatis, G.; Voudouris, K.; Karefilakis, F. Groundwater Pollution by Heavy Metals in Historical Mining Area of Lavrio, Attica, Greece. Water Air Soil Pollut. 2001, 128, 61–83. [Google Scholar] [CrossRef]
  733. Voudouris, P.; Melfos, V.; Mavrogonatos, C.; Photiades, A.; Moraiti, E.; Rieck, B.; Kolitsch, U.; Tarantola, A.; Scheffer, C.; Morin, D.; et al. The Lavrion Mines: A Unique Site of Geological and Mineralogical Heritage. Minerals 2021, 11, 76. [Google Scholar] [CrossRef]
  734. Antoniadis, V.; Thalassinos, G.; Levizou, E.; Wang, J.; Wang, S.-L.; Shaheen, S.M.; Rinklebe, J. Hazardous enrichment of toxic elements in soils and olives in the urban zone of Lavrio, Greece, a legacy, millennia-old silver/lead mining area and related health risk assessment. J. Hazard. Mater. 2022, 434, 128906. [Google Scholar] [CrossRef]
  735. Fashola, M.O.; Ngole-Jeme, V.M.; Babalola, O.O. Heavy Metal Pollution from Gold Mines: Environmental Effects and Bacterial Strategies for Resistance. Int. J. Environ. Res. Public Health 2016, 13, 1047. [Google Scholar] [CrossRef]
  736. Bozatzi, A.; Periferakis, A. Geodidactics and CLIL: Synthesizing a syllabus for the ELT classroom through games, stories and technology. In Proceedings of the 16th International Congress of the Geological Society of Greece, Patras, Greece, 17–19 October 2022. [Google Scholar]
  737. Tchounwou, P.B.; Yedjou, C.G.; Patlolla, A.K.; Sutton, D.J. Heavy metal toxicity and the environment. Exp. Suppl. 2012, 101, 133–164. [Google Scholar] [CrossRef]
  738. Briffa, J.; Sinagra, E.; Blundell, R. Heavy metal pollution in the environment and their toxicological effects on humans. Heliyon 2020, 6, e04691. [Google Scholar] [CrossRef]
  739. Van Der Werf, H.M. Assessing the impact of pesticides on the environment. Agric. Ecosyst. Environ. 1996, 60, 81–96. [Google Scholar] [CrossRef]
  740. Gill, H.K.; Garg, H. Pesticide: Environmental impacts and management strategies. Pestic.-Toxic. Asp. 2014, 8, 10–5772. [Google Scholar]
  741. Stefanidis, K.; Christopoulou, A.; Poulos, S.; Dassenakis, E.; Dimitriou, E. Nitrogen and Phosphorus Loads in Greek Rivers: Implications for Management in Compliance with the Water Framework Directive. Water 2020, 12, 1531. [Google Scholar] [CrossRef]
  742. Della Rossa, P.; Jannoyer, M.; Mottes, C.; Plet, J.; Bazizi, A.; Arnaud, L.; Jestin, A.; Woignier, T.; Gaude, J.-M.; Cattan, P. Linking current river pollution to historical pesticide use: Insights for territorial management? Sci. Total Environ. 2017, 574, 1232–1242. [Google Scholar] [CrossRef] [PubMed]
  743. Kirchhelle, C. Toxic Tales—Recent Histories of Pollution, Poisoning, and Pesticides (ca. 1800–2010). NTM Z. Gesch. Wiss. Tech. Med. 2018, 26, 213–229. [Google Scholar] [CrossRef] [PubMed]
  744. Cobilinschi, C.; Tincu, R.; Totan, A.; Ghiorghiu, Z.; Neagu, T.P.; Macovei, R. Thyroid dysfunction caused by organophosphate poisoning. Toxicol. Lett. 2017, 280, S169. [Google Scholar] [CrossRef]
  745. Cobilinschi, C.; Țincu, R.; Ungureanu, R.; Dumitru, I.; Băetu, A.; Isac, S.; Cobilinschi, C.O.; Grințescu, I.M.; Mirea, L. Toxic-Induced Nonthyroidal Illness Syndrome Induced by Acute Low-Dose Pesticides Exposure—Preliminary In Vivo Study. Toxics 2022, 10, 511. [Google Scholar] [CrossRef]
  746. Cobilinschi, C.; Olteanu, A.; Cobilinschi, C.; Grintescu, I.; Macovei, R.; Tudosie, M.; Serban, D.; Tincu, R. From the discovery of combat gases to insecticides and bioterrorism—A brief history. Rom. J. Mil. Med. 2022, 125, 247–252. [Google Scholar] [CrossRef]
  747. Franklin, M.R.; Fernandes, H.M. Identifying and overcoming the constraints that prevent the full implementation of decommissioning and remediation programs in uranium mining sites. J. Environ. Radioact. 2013, 119, 48–54. [Google Scholar] [CrossRef]
  748. Lima, A.T.; Mitchell, K.; O’Connell, D.W.; Verhoeven, J.; Van Cappellen, P. The legacy of surface mining: Remediation, restoration, reclamation and rehabilitation. Environ. Sci. Policy 2016, 66, 227–233. [Google Scholar] [CrossRef]
  749. Guzman, E.R. Canadian Financial Assurance Frameworks for the Remediation of Mining Sites: An Assessment of Ontario’s, British Columbia’s and Quebec’s Schemes and Three Potential Reform Initiatives. J. Environ. Law Pract. 2017, 31, 1–35. [Google Scholar]
  750. Mulligan, D.; Grigg, A.; Madsen, T.; Pearce, A.; Roe, P. Research initiatives for the remediation of land following open-cut coal mining in central Queensland. In Remediation and Management of Degraded Lands; Routledge: London, UK, 2018; pp. 25–36. [Google Scholar]
  751. O’Brien, R.M.; Phelan, T.J.; Smith, N.M.; Smits, K.M. Remediation in developing countries: A review of previously implemented projects and analysis of stakeholder participation efforts. Crit. Rev. Environ. Sci. Technol. 2021, 51, 1259–1280. [Google Scholar] [CrossRef]
  752. Miu, B.A.; Pop, C.-E.; Crăciun, N.; Deák, G. Bringing life back into former mining sites: A mini-review on soil remediation using organic amendments. Sustainability 2022, 14, 12469. [Google Scholar] [CrossRef]
  753. Reichenberger, S.; Bach, M.; Skitschak, A.; Frede, H.-G. Mitigation strategies to reduce pesticide inputs into ground-and surface water and their effectiveness; a review. Sci. Total Environ. 2007, 384, 1–35. [Google Scholar] [CrossRef] [PubMed]
  754. Destandau, F.; Imfeld, G.; Rozan, A. Regulation of diffuse pesticide pollution: Combining point source reduction and mitigation in stormwater wetland (Rouffach, France). Ecol. Eng. 2013, 60, 299–308. [Google Scholar] [CrossRef]
  755. Jin, S.; Bluemling, B.; Mol, A.P. Mitigating land pollution through pesticide packages–The case of a collection scheme in Rural China. Sci. Total Environ. 2018, 622, 502–509. [Google Scholar] [CrossRef] [PubMed]
  756. Quaglia, G.; Joris, I.; Broekx, S.; Desmet, N.; Koopmans, K.; Vandaele, K.; Seuntjens, P. A spatial approach to identify priority areas for pesticide pollution mitigation. J. Environ. Manag. 2019, 246, 583–593. [Google Scholar] [CrossRef]
  757. Sahin, C.; Karpuzcu, M.E. Mitigation of organophosphate pesticide pollution in agricultural watersheds. Sci. Total Environ. 2020, 710, 136261. [Google Scholar] [CrossRef]
  758. Intisar, A.; Ramzan, A.; Sawaira, T.; Kareem, A.T.; Hussain, N.; Din, M.I.; Bilal, M.; Iqbal, H.M. Occurrence, toxic effects, and mitigation of pesticides as emerging environmental pollutants using robust nanomaterials–A review. Chemosphere 2022, 293, 133538. [Google Scholar] [CrossRef]
  759. Anastopoulos, I.; Robalds, A.; Tran, H.; Mitrogiannis, D.; Giannakoudakis, D.A.; Hosseini-Bandegharaei, A.; Dotto, G. Removal of heavy metals by leaves-derived biosorbents. Environ. Chem. Lett. 2018, 17, 755–766. [Google Scholar] [CrossRef]
  760. Mitrogiannis, D.; Psychoyou, M.; Kornaros, M.; Tsigkou, K.; Brulé, M.; Koukouzas, N.; Alexopoulos, D.; Palles, D.; Kamitsos, E.; Oikonomou, G.; et al. Calcium-modified clinoptilolite as a recovery medium of phosphate and potassium from anaerobically digested olive mill wastewater. Environ. Sci. Pollut. Res. 2020, 27, 2977–2991. [Google Scholar] [CrossRef]
  761. Mitrogiannis, D.; Psychoyou, M.; Baziotis, I.; Mavrogonatos, C.; Koukouzas, N.; Anastopoulos, I.; Fyrillas, M.; Inglezakis, V. Phosphate removal by Ca(OH)2-treated natural minerals: Experimental and modeling studies. Colloids Surf. A Physicochem. Eng. Asp. 2022, 660, 130805. [Google Scholar] [CrossRef]
  762. Koitabashi, R.; Suzuki, T.; Kawazu, T.; Sakai, A.; Kuroiwa, H.; Kuroiwa, T. 1,8-Cineole inhibits root growth and DNA synthesis in the root apical meristem of Brassica campestris L. J. Plant Res. 1997, 110, 1–6. [Google Scholar] [CrossRef]
  763. De Feo, V.; De Simone, F.; Senatore, F. Potential allelochemicals from the essential oil of Ruta graveolens. Phytochemistry 2002, 61, 573–578. [Google Scholar] [CrossRef] [PubMed]
  764. Singh, H.P.; Batish, D.R.; Kaur, S.; Arora, K.; Kohli, R.K. α-Pinene Inhibits Growth and Induces Oxidative Stress in Roots. Ann. Bot. 2006, 98, 1261–1269. [Google Scholar] [CrossRef]
  765. Koutsaviti, K.; Giatropoulos, A.; Pitarokili, D.; Papachristos, D.; Michaelakis, A.; Tzakou, O. Greek Pinus essential oils: Larvicidal activity and repellency against Aedes albopictus (Diptera: Culicidae). Parasitol. Res. 2015, 114, 583–592. [Google Scholar] [CrossRef] [PubMed]
  766. Sakulpanich, A.; Attrapadung, S.; Gritsanapan, W. Insecticidal activity of Stemona collinsiae root extract against Parasarcophaga ruficornis (Diptera: Sarcophagidae). Acta Trop. 2017, 173, 62–68. [Google Scholar] [CrossRef] [PubMed]
  767. Sakulpanich, A.; Attrapadung, S.; Gritsanapan, W. Larvicidal activity of Stemona collinsiae root extract against Musca domestica and Chrysomya megacephala. Sci. Rep. 2023, 13, 15689. [Google Scholar] [CrossRef]
  768. Directive 2009/128/EC of the European Parliament and of the Council of 21 October 2009 Establishing a Framework for Community Action to Achieve the Sustainable Use of Pesticides; Official Journal of the European Union: Luxembourg, 2009; pp. 71–86.
  769. Regulation (EC) No 1107/2009 of the European Parliament and of the Council of 21 October 2009 Concerning the Placing of Plant Protection Products on the Market and Repealing Council Directives 79/117/EEC and 91/414/EEC; European Parliament, Ed.; Official Journal of the European Union: Luxembourg, 2009; pp. 1–50. [Google Scholar]
  770. Karamfilova, E. Revision of Directive 2009/128/EC on the sustainable use of pesticides. In Briefing Implementation Appraisal; European Parliamentary Research Service PE 730; European Parliamentary Research Service: Brussels, Belgium, 2022; pp. 1–12. [Google Scholar]
  771. Dinu, A.; Karamfilova, E. Regulation (EC) 1107/2009 on the Placing of Plant Protection Products on the Market; European Parliamentary Research Service: Brussels, Belgium, 2018; pp. 1–70. [Google Scholar]
  772. Moagar-Poladian, S.; Folea, V.; Paunica, M. Competitiveness of EU member states in attracting EU funding for research and innovation. Rom. J. Econ. Forecast. 2017, 20, 150–167. [Google Scholar]
  773. Schebesta, H.; Candel, J.J.L. Game-changing potential of the EU’s Farm to Fork Strategy. Nat. Food 2020, 1, 586–588. [Google Scholar] [CrossRef]
  774. Billen, G.; Aguilera, E.; Einarsson, R.; Garnier, J.; Gingrich, S.; Grizzetti, B.; Lassaletta, L.; Le Noë, J.; Sanz-Cobena, A. Beyond the Farm to Fork Strategy: Methodology for designing a European agro-ecological future. Sci. Total Environ. 2024, 908, 168160. [Google Scholar] [CrossRef]
  775. Communication from The Commission to The European Parliament, The European Council, The Council, The European Economic and Social Committee and the Committee of the Regions. In The European Green Deal; European Commission: Brussels, Belgium, 2019.
  776. Salt, D.E.; Smith, R.; Raskin, I. Phytoremediation. Annu. Rev. Plant Biol. 1998, 49, 643–668. [Google Scholar] [CrossRef]
  777. Pilon-Smits, E. Phytoremediation. Annu. Rev. Plant Biol. 2005, 56, 15–39. [Google Scholar] [CrossRef]
  778. Arthur, E.L.; Rice, P.J.; Rice, P.J.; Anderson, T.A.; Baladi, S.M.; Henderson, K.L.; Coats, J.R. Phytoremediation—An overview. Crit. Rev. Plant Sci. 2005, 24, 109–122. [Google Scholar] [CrossRef]
  779. Assembly, G. Transforming our world: The 2030 Agenda for Sustainable Development Resolutions/Recommendations/Declarations/Decisions, A/RES/70/1; General Assembly of the United Nations: New York, NY, USA, 2015. [Google Scholar]
  780. Treaty on European Union; Official Journal of the European Communities: Luxembourg, 1992.
  781. Directive 2000/60/EC of the European Parliament and of the Council of 23 October 2000 Establishing a Framework for Community Action in the Field of Water Policy; O J L 327; European Parliament: Strasbourg, France, 2008; pp. 1–73.
  782. Directive 2008/98/EC of the European Parliament and of the Council of 19 November 2008 on Waste and Repealing Certain Directives; 312; Official Journal of the European Union: Legislation Series, Ed.; European Parliament: Strasbourg, France, 2008; pp. 3–30. [Google Scholar]
  783. Directive, E. Council Directive of 21. May 1991 concerning urban waste water treatment (91/271/EEC). J. Eur. Commun. 1991, 34, 40. [Google Scholar]
  784. Preisner, M.; Smol, M.; Szołdrowska, D. Trends, insights and effects of the Urban Wastewater Treatment Directive (91/271/EEC) implementation in the light of the Polish coastal zone eutrophication. Environ. Manag. 2021, 67, 342–354. [Google Scholar] [CrossRef] [PubMed]
  785. Coric, V.; Bojovic, A.K.; Glintic, M. Thirtieth Anniversary of the Urban Waste Water Treatment Directive 91/271/EEC: Consideration of the Need for Its Amendments. Rev. Eur. L. 2021, 23, 73. [Google Scholar]
  786. Sandström, E. The LIFE-Fund of the European Union as a Means to Support the Development of the Implementation of the Swedish Forest Policy. In Financial Instruments of Forest Policy; European Forest Institute, Gummerus Printing: Saarijärvi, Finland, 2002; pp. 77–86. [Google Scholar]
  787. de Sadeleer, N. The Principle of Subsidiarity and Its Impact on EU Environmental Law and Policy. Droit Soc. 2012, 80, 73. [Google Scholar] [CrossRef]
  788. Summary of the Federal Insecticide, Fungicide, and Rodenticide Act. In 7 U.S.C. §136 et seq; United States Environmental Protection Agency, (U.S. EPA) (website); 1996. Available online: https://www.epa.gov/laws-regulations/summary-federal-insecticide-fungicide-and-rodenticide-act (accessed on 12 February 2025).
  789. The Food Quality Protection Act (FQPA), or H.R.1627. In Public Law 104-170; The U.S. National Archives and Records Administration: College Park, MD, USA, 1996.
  790. The National Environmental Policy Act (NEPA). In Public Law 91-70; The U.S. National Archives and Records Administration: College Park, MD, USA, 1969.
  791. The Toxic Substances Control Act (TSCA). In 15 U.S.C. §2601 et seq; The U.S. National Archives and Records Administration: College Park, MD, USA, 1976.
  792. Agricultural Chemicals Regulation Act, Act No. 82 of 1948 (Japan), as Revised by the Act No. 53 of 2018. 2018. [Agricultural Chemicals Regulation Act—English—Japanese Law Translation]. Available online: https://www.japaneselawtranslation.go.jp/en/laws/view/3507/en (accessed on 12 February 2025).
  793. Act on Promotion of Organic Agriculture, Act No. 112 of 2006 (Japan), as Revised by the Act No. 105 of 2011. 2011. [Act on Promotion of Organic Agriculture—English—Japanese Law Translation]. Available online: https://www.japaneselawtranslation.go.jp/en/laws/view/3728/en (accessed on 12 February 2025).
  794. Basic Act on the Environment, Act No. 91 of 1993 (Japan) Domestic Law. 1993. [The Basic Environment Law—Ministry of the Environment, Government of Japan]. Available online: https://www.env.go.jp/en/laws/policy/basic/index.html (accessed on 12 February 2025).
  795. Framework Act on Environmental Policy, as revised by Act No. 18918. 10 June 2022. Available online: https://faolex.fao.org/docs/pdf/kor51838.pdf (accessed on 12 February 2025).
  796. Kim, S.; Seol, T.K. Environmental law in South Korea. Lexology. Available online: https://www.lexology.com/library/detail.aspx?g=4b3fbe13-1c55-48df-b34b-14f1dd35b0bb (accessed on 12 January 2025).
  797. Agrochemicals Control Act (Act No. 445 of August 28, 1957, as Last Amended by Act No. 9658 of May 8, 2009). 2009. [Agrochemicals Control Act (Act No. 445 of August 28, 1957, as last amended by Act No. 9658 of May 8, 2009), Republic of Korea, WIPO Lex]. Available online: https://www.wipo.int/wipolex/en/legislation/details/12867 (accessed on 12 February 2025).
  798. Consolidated Act No. 11459 of 1 June 2012, as Amended Last by Act No. 18026 of 13 April 2021. 2021. [Act on the Promotion of Environment-Friendly Agriculture and Fisheries and the Management of and Support for Organic Foods, etc.—UNEP Law and Environment Assistance Platform]. Available online: https://leap.unep.org/en/countries/kr/national-legislation/act-promotion-environment-friendly-agriculture-and-fisheries-and?fbclid=IwY2xjawJDx_JleHRuA2FlbQIxMAABHckAEC6NAN5ePGRtzw9qLTOifzu1UMmoasP1Dcq1GDRZuwUXHv3xtn8jCQ_aem_AZ0Iv7bD6rwPj-FkMSRdhA (accessed on 12 February 2025).
Figure 1. Pinosylvin and its most common derivatives. Glu = glucose.
Figure 1. Pinosylvin and its most common derivatives. Glu = glucose.
Cimb 47 00204 g001
Figure 2. Schematic representation of biosynthetic (phenylpropanoid) pathways and chemical pathways (beige overlay) for pinosylvin. PAL = phenylalanine ammonia-lyase, 4CL = 4-cumaroyl: CoA-lyase, C4H = cinnamate-4-hydroxylase, STS = stilbene synthase.
Figure 2. Schematic representation of biosynthetic (phenylpropanoid) pathways and chemical pathways (beige overlay) for pinosylvin. PAL = phenylalanine ammonia-lyase, 4CL = 4-cumaroyl: CoA-lyase, C4H = cinnamate-4-hydroxylase, STS = stilbene synthase.
Cimb 47 00204 g002
Figure 3. Summary of key biological activities and known action mechanisms involved. Nrf2 = nuclear factor E2-related factor 2; SOD = superoxide dismutase; GPx = glutathione peroxidase; HO-1 = heme-oxygenase 1; iNOS = inducible nitric oxide synthase; NADPH = nicotinamide adenine dinucleotide phosphate; NF-κB = nuclear factor kappa B; TNF = tumour necrosis factor; IL = interleukin; COX = cyclooxygenase; LOX = lipoxygenase; TRP = transient receptor potential; MMP = matrix metalloproteinase; VEGF = vascular endothelial growth factor; ROS = reactive oxygen species; CDK = cyclin-dependent kinase.
Figure 3. Summary of key biological activities and known action mechanisms involved. Nrf2 = nuclear factor E2-related factor 2; SOD = superoxide dismutase; GPx = glutathione peroxidase; HO-1 = heme-oxygenase 1; iNOS = inducible nitric oxide synthase; NADPH = nicotinamide adenine dinucleotide phosphate; NF-κB = nuclear factor kappa B; TNF = tumour necrosis factor; IL = interleukin; COX = cyclooxygenase; LOX = lipoxygenase; TRP = transient receptor potential; MMP = matrix metalloproteinase; VEGF = vascular endothelial growth factor; ROS = reactive oxygen species; CDK = cyclin-dependent kinase.
Cimb 47 00204 g003
Table 1. Taxonomical classification of pinosylvin-producing plants (listed alphabetically by species).
Table 1. Taxonomical classification of pinosylvin-producing plants (listed alphabetically by species).
OrderFamilyGenusSpeciesReference
FabalesFabaceaeArachisA. hypogaea[51]
CajanusC. cajan[53]
RosalesRhamnaceaeHoveniaH. dulcis Thunb.[54]
LauralesLauraceaeLinderaL. reflexa Hemsl.[55]
MyrtalesMyrtaceaeAgonisA. flexuosa[56]
PinalesPinaceaePiceaP. abies[57]
P. glauca[58]
PinusP. banksiana[59,60,61]
P. brutia Hen.[62]
P. caribaea[63]
P. cembra[64]
P. contorta[64]
P. densiflora[65,66]
P. halepensis Mill.[67,68]
P. merkusii[69]
P. nigra Arn.[70]
P. palustris[71]
P. pinaster[72,73,74]
P. resinosa[60]
P. roxburghii Sargent[75]
P. strobus[71]
P. sibirica[64]
P. sylvestris[76]
P. taeda[77]
PandanalesStemonaceaeStemonaS. cf. peirrei[78]
S. collinsae[79,80]
S. tuberosa[81]
Table 3. Antibacterial actions of pinosylvin and its derivatives based on current research (reported alphabetically based on genera).
Table 3. Antibacterial actions of pinosylvin and its derivatives based on current research (reported alphabetically based on genera).
GenusSpeciesTested SubstanceExtract OriginEffectivenessYearReference
AchromobacterA. xylosoxidansPinosylvin, pinosylvin monomethyl etherP. banksiana, P. contorta, P. resinosa, P. sylvestris11–20 (and more) mm 12004[60]
ArcobacterA. butzleriPinosylvinn/a (pure compound)128 μg/mL (MIC) 2019[139]
BacillusB. cereusPinosylvin, pinosylvin monomethyl ether, dihydropinosylvin monomethyl etherP. strobus, P. sylvestris101 ± 6, 92 ± 4, 82 ± 3 (% of inhibition)2007[140]
B. coagulansPinosylvin, pinosylvin monomethyl etherP. contorta, P. banksiana, P. resinosa, P. sylvestris16–20 (and more) mm 12004[60]
B. subtilisPinosylvinn/a (laboratory synthesis)64 μg/mL (MIC)2016[141]
Pinosylvinn/a (pure compound)99.2% 22019[142]
BurkholderiaB. multivoransPinosylvin, pinosylvin monomethyl etherP. contorta, P. banksiana, P. resinosa, P. sylvestris11–20 (and more) mm 12004[60]
CampylobacterC. coli, C. jejuniPinosylvinn/a (pure compound)25–50 μg/mL (MIC)2015[143]
Pinosylvinn/a (pure compound)Multiple values of inhibition haloes based on experimental parameters2018[144]
EscherichiaE. coliPinosylvinn/a (pure compound)250 μg/mL (MIC)2005[145]
Pinosylvin, pinosylvin monomethyl ether, dihydropinosylvin monomethyl etherP. strobus, P. sylvestris54 ± 8, 71 ± 7, 18 ± 2 (% of inhibition)2007[140]
Pinosylvinn/a (laboratory synthesis)64 μg/mL (MIC)2016[141]
Pinosylvinn/a (pure compound)58.9% 22019[142]
ListeriaL. monocytogenesPinosylvin, pinosylvin monomethyl ether, dihydropinosylvin monomethyl etherP. strobus, P. sylvestris62 ± 15, 100 ± 7, 64 ± 12 (% of inhibition)2007[140]
Pinosylvinn/a (laboratory synthesis)93.2 ± 0.4 (% of inhibition at concentration of 0.5 mM)2013[146]
Pinosylvinn/a (pure compound)97.9% 22019[142]
ProteusP. vulgarisPinosylvinn/a (laboratory synthesis)>128 μg/mL (MIC)2016[141]
PseudomonasP. aeruginosaPinosylvinn/a (laboratory synthesis)>128 μg/mL (MIC)2016[141]
Pinosylvinn/a (pure compound)8.9% 22019[142]
P. fluorescensPinosylvin, pinosylvin monomethyl ether, dihydropinosylvin monomethyl etherP. strobus, P. sylvestris50 ± 15, 35 ± 2, 22 ± 6 (% of inhibition)2007[140]
SalmonellaS. infantisPinosylvin, pinosylvin monomethyl ether, dihydropinosylvin monomethyl etherP. strobus, P. sylvestris42 ± 20, 40 ± 7, 14 ± 2 (% of inhibition)2007[140]
S. enteritidisPinosylvinn/a (laboratory synthesis)80.6% 22019[142]
StaphylococcusS. aureusPinosylvinn/a (pure compound)250 μg/mL (MIC)2005[145]
Pinosylvin, pinosylvin monomethyl ether, dihydropinosylvin monomethyl etherP. strobus, P. sylvestris76 ± 2, 105 ± 12, 76 ± 4 (% of inhibition)2007[140]
Pinosylvinn/a (laboratory synthesis)75 μg/mL MIC 2013[146]
Pinosylvinn/a (laboratory synthesis)64 μg/mL (MIC) (higher for some MRSA strains)2016[141]
PinosylvinA. hypogea≤100 μg/mL (MIC)2018[147]
Pinosylvinn/a (pure compound)100% 22019[142]
S. epidermidisPinosylvinn/a (laboratory synthesis)128 μg/mL (MIC)2016[141]
1 inhibition zone diameter—different diameters depending on extract source; 2 bacterial load difference. MIC = minimum inhibitory concentration.
Table 4. Antifungal actions of pinosylvin and its derivatives based on current research (reported alphabetically based on genera).
Table 4. Antifungal actions of pinosylvin and its derivatives based on current research (reported alphabetically based on genera).
GenusSpeciesTested SubstanceExtract OriginEffectivenessYearReference
AspergillusA. fumigatusPinosylvin, pinosylvin monomethyl ether, dihydropinosylvin monomethyl etherP. strobus, P. sylvestris14 ± 1/ 12 ± 3 12007[140]
CandidaC. albicansPinosylvinn/a (pure compound)62.5 μg/mL (MIC)2005[145]
Pinosylvin, pinosylvin monomethyl ether, dihydropinosylvin monomethyl etherP. strobus, P. sylvestris85 ± 5/ 80 ± 4 12007[140]
CladosporiumC. herbarumPinosylvin, 4′-methylpinosylvinS. collinsaen/a (only structurally similar stilbenoids were tested)2002[79]
Pinosylvin, dihydropinosylvinS. cf. pierrei10mg/mL (EC50)2004[78]
PlasmoparaP. viticolaPinosylvin, pinosylvin monomethyl etherP. pinaster23, 18 μΜ (available as IC50)2017[73]
PenicilliumP. brevicompactumPinosylvin, pinosylvin monomethyl ether, dihydropinosylvin monomethyl etherP. strobus, P. sylvestris15 ± 1/ 14 ± 2 12007[140]
SaccharomycesS. cerevisiaePinosylvinn/a (pure compound)125 μg/mL (MIC)2005[145]
Pinosylvin, pinosylvin monomethyl ether, dihydropinosylvin monomethyl etherP. strobus, P. sylvestris82 ± 13/ 35 ± 21 12007[140]
1 percentage of inhibition zone compared to control.
Table 7. Experiments on the anticancer properties of pinosylvin and its derivatives.
Table 7. Experiments on the anticancer properties of pinosylvin and its derivatives.
CompoundPlantTested onMechanismEffectConcentration/
Administration
YearReference
Pinosylvinn/a (laboratory synthesis)In vitro—Raji and Molt human lymphoblastoid cell linesInhibition of protein uptakeAntiproliferativeDirect addition in culture—15, 30 μg/ml1986[339]
PinosylvinP. sylvestrisIn vitro—MCF-7 or T-47D breast cancer cellsIncrease in oestrogen expressionPro-proliferativeDilution in ethanol and culture for 7 days—1 pM to 1 μΜ 1996[344]
PinosylvinP. sylvestrisIn vitro—RAW264.7 cells and HT-9 human colon cancer cellsCOX-2 inhibition, free radicals scavenging and xanthine oxidase activity inhibitionAntiproliferativen/a2003[359]
Pinosylvin, pinosylvin monomethyl ether, pinosylvin dimethyl etherP. resinosa Ait.In vitro—lung
carcinoma cells A549 lung
carcinoma cells, DLD-1 human colorectal adenocarcinoma cells, WS1
healthy cells
UnknownCytotoxicIncubation for 48 h—different concentrations (IC50 = ~41–130 μg/mL depending on cell line; value for extract)2008[356]
Pinosylvinn/a (laboratory synthesis)In vitro—HT1080 human fibrosarcoma cellsDecreased expression of matrix metalloproteinasesAntimetastaticTreatment of cells at almost confluency, with pinosylvin—12.5, 25 and 50 μM2012[347]
In vivo—miceDownregulation of COX-2 expression, decreased ERK1/2 and Akt phosphorylation and reduced metalloproteinase expressionIntraperitoneal administration—10 mg/kg per body weight
Pinosylvinn/a (laboratory synthesis)In vitro—HTC 116 human colorectal cancer cellsSrc/ERK and GSK-3/β-catenin signalling suppressionAntiproliferativeIncubation for different periods—various concentrations (depending on different experimental protocols); IC50 = 48.2 μM (for 24 h)2013[340]
Pinosylvin methyl ethern/a (pure compound)In vitro—LNCaP prostate cancer cells, RWPE-1, and EP156T non-malignant cancer cellsAlteration of cell cycle-related genes, modification of steroid and cholesterol biosynthesis and androgen-signallingAntiproliferativeIncubation over a span of 3 days (EC50 = 250 nM) 2016[341]
Pinosylvinn/a (pure compound)In vitro—THP-1 and U937 human monocytic cell linesCaspace-3 activation, flipflop of phosphatidylserine, p62 degradation and LC3-II accumulation, downregulation of AMPKa expressionCytotoxicPretreatment with pinosylvin—0 to 100 μmol/L (IC50 = ~20–30 μmol/L)2018[357]
Pinosylvinn/a (pure compound)In vitro—SAS, SCC-9, and HSC-3 cellsInhibition of MMP-2, upregulation of TIMP-2 expression, and downregulation of the ERK1/2 signalling pathwayAntiproliferative and anti-metastaticDirect addition in culture medium—20, 40, 80 μΜ2019[263]
Pinosylvinn/a (pure compound)In vitro—NPC-039, NPC-BM and RPMI 2650 cells (nasopharyngeal carcinoma cells)Inhibition of MMP-2, and decreased expression of MMP-2 and MMP-9; decrease in vimentin and N-cadherin and E-cadherin expression and of ZO-1AntimetastaticTreatment of cells with pinosylvin—20, 40, 80 μΜ 2021[348]
Pinosylvinn/a (pure compound)In vitro—ECA109 and TE1
oesophageal cancer cells
Reduction in syntaxin-6 and integrin α3 expression and enhanced vasodilator-stimulated phosphoprotein
expression
AntimetastaticTreatment of cells with pinosylvin for 24 h—0, 10, 20, 40, and 80 μM2023[354]
Table 8. Experiments on the neuroprotective effects of pinosylvin and its derivatives.
Table 8. Experiments on the neuroprotective effects of pinosylvin and its derivatives.
CompoundPlantTested onMechanismEffectConcentration and AdministrationYearReference
Pinosylvinn/a (pure compound)In vitro—OGD/R-damaged PC12
cells
Upregulation of PINK1/Parkin-regulated mitophagy, upregulation of the Nrf2 pathwayNeuroprotectionPretreatment of cells with pinosylvin for 24 h—10 μΜ2021[364]
In vivo—ratsReduction in infarct size and of neural cell deathDecrease in brain function lossIntraperitoneal injection—50 mg/kg
Table 9. Traditional ethnobotanical uses of pinosylvin-producing plants and experimentally verified actions.
Table 9. Traditional ethnobotanical uses of pinosylvin-producing plants and experimentally verified actions.
PlantTraditional/
Ethnobotanical Uses
Medical SystemTested ActionsRegion/
Populations
References
Tax. NameCommon Name
A. hypogaeaPeanutTreatment of sleep disorders, antidiabetic, antilipidemic, weight loss, anticancer use, cardiovascular pathologies and haemorrhages, treatment of bronchitis, antibacterialTraditional Chinese medicine, Nigerian folk medicineTreatment of sleep disorders, antimicrobial, antioxidantChina, Nigeria/Chinese, local population[381,382,383]
C. cajanPigeon peaAntidiabetic, stimulant, analgesic, treatment of haemorrhage, oral pathologies, laxative, lactation inductionBangladesh medical tradition, Trinidad and Tobacco medical tradition, traditional Chinese medicine, AyurvedaAntimicrobial, antidyslipidaemic, antidiabetic, antioxidant, anticancer, hepatoprotectiveBangladesh, Trinidad and Tobacco, China, India[384,385,386,387]
H. dulcisJapanese raisin treeAlcohol detoxification (perhaps hepatoprotective)Traditional Chinese medicine, traditional Japanese medicine, traditional Korean medicineAlcohol detoxification, hepatoprotective effect, antioxidant, antidiabetic, antimicrobial, antilipidaemic, anti-allergic, anti-inflammatory, prokineticChina, Japan, Korea/Chinese, Japanese, Koreans[388,389]
P. glaucaWhite spruceAntibacterial, treatment for sore mouth and strep throat, sinusitis, haemoptysis, wound treatment, chronic pain treatmentCarrier people’s medical practices, Canadian Boreal Forest peopleAntimicrobial, organoprotective, antioxidantCanada/Carrier people, indigenous populations[390,391,392,393]
P. brutia Ten.Turkish red pineAntitussive, antidiabetic, treatment of gastrointestinal complains, tonicTurkish folk medicineAntifungal, antibacterial, (potential) treatment of acute lung injury, (potential) antioxidant action, anti-inflammatoryTurkey/local population[394,395,396,397,398,399]
P. densiflora Sieb. et Zucc.Korean red pineAntihypertensive, anti-atherosclerotic, treatment of strokes, diabetes, cancer, and baldingTraditional Korean medicinen/aKorea/Korean people[400]
P. halepensis Mill.Aleppo pineRespiratory pathologies, wound treatments, anti-inflammatory, urinary problems, GI ulcers, prostate infections, infertility, antiseptic, adrenal gland stimulant, antimicrobial, toothache, baldnessLocal medical traditionsAntibacterial, antifungal, antioxidant, cytoprotective, anticancer (cytotoxic), anti-coagulant, anti-haemolytic, anti-inflammatoryItaly, Spain, Algeria, Morocco/Berber people, local populations[68]
P. nigraBlack pineColds, cough, treatment of respiratory pathologies, furuncles, warts, treatment of teeth decay, digestive complaintsLocal medical traditions and Austro-Hungarian pharmacopoeiaAntimicrobial, antioxidant (weak), food supplement, antiproliferativeRomania/local people of Transylvania[401,402,403,404]
P. mugo TuraDwarf mountain pine, scrub mountain pine, Swiss mountain pineExpectorantLocal medical traditionsn/aItaly[405,406]
P. roxburghii SargentChir pine, long-leaved pineAntiseptic, diarrhetic, diaphoretic, tonic, vermifuge (anthelmintic), rubefacient, spasmolytic, antioxidant, anti-inflammatory, treatment for ocular and ear pathologies, ulcer treatment, bronchitis treatment, treatment of skin diseases, treatment of blood diseases, treatment of snake and scorpion bitesAyurveda, various local medical traditionsAntibacterial, antioxidant, anticonvulsant, anti-asthmatic, analgesic activityHimalayas, Hindu Kush/local tribes[407,408,409,410]
P. sylvestrisScots pine, Baltic pine, European red pineTreatment of asthma, cough and respiratory complaints, treatment of rheumatisms and varicose veinsLocal medical traditions and Austro-Hungarian pharmacopoeiaAntioxidant, anti-inflammatory, food supplementRomania/local people of Transylvania[403,404,411]
P. virginiana MillerVirginia pineTreatment of skin ulcers and sores, baths for painful joints, and treatment of cold and feverNative American traditional medicinen/aCherokee Native Americans[412]
Table 10. Non-medical applications of the properties of pinosylvin-containing plants.
Table 10. Non-medical applications of the properties of pinosylvin-containing plants.
Plant Part UsedPurpose/ActivityMechanismYearReference
Bark of P. roxburghiiWater purificationRemoval of Cr(VI) via adsorption2005[579]
Pine barkOrganochlorine pesticides removal2011[722]
Pine bark acting as a substrate for biofilm formationPhycoremediation2020[721]
Essential oils of different Pinus spp.Insecticidal activityRepellent and larvicidal activity against A. albopticus mosquito larvae2015[765]
Ethanolic extract from S. collinsiae rootsElimination of P. ruficornis in larval and adult stages2017[766]
Ethanolic extract from S. collinsiae rootsElimination of C. megacephala flies at the larval stage2023[767]
Cr(VI) = hexavalent chromium.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Periferakis, A.; Periferakis, A.-T.; Troumpata, L.; Periferakis, K.; Georgatos-Garcia, S.; Touriki, G.; Dragosloveanu, C.D.M.; Caruntu, A.; Savulescu-Fiedler, I.; Dragosloveanu, S.; et al. Pinosylvin: A Multifunctional Stilbenoid with Antimicrobial, Antioxidant, and Anti-Inflammatory Potential. Curr. Issues Mol. Biol. 2025, 47, 204. https://doi.org/10.3390/cimb47030204

AMA Style

Periferakis A, Periferakis A-T, Troumpata L, Periferakis K, Georgatos-Garcia S, Touriki G, Dragosloveanu CDM, Caruntu A, Savulescu-Fiedler I, Dragosloveanu S, et al. Pinosylvin: A Multifunctional Stilbenoid with Antimicrobial, Antioxidant, and Anti-Inflammatory Potential. Current Issues in Molecular Biology. 2025; 47(3):204. https://doi.org/10.3390/cimb47030204

Chicago/Turabian Style

Periferakis, Argyrios, Aristodemos-Theodoros Periferakis, Lamprini Troumpata, Konstantinos Periferakis, Spyrangelos Georgatos-Garcia, Georgia Touriki, Christiana Diana Maria Dragosloveanu, Ana Caruntu, Ilinca Savulescu-Fiedler, Serban Dragosloveanu, and et al. 2025. "Pinosylvin: A Multifunctional Stilbenoid with Antimicrobial, Antioxidant, and Anti-Inflammatory Potential" Current Issues in Molecular Biology 47, no. 3: 204. https://doi.org/10.3390/cimb47030204

APA Style

Periferakis, A., Periferakis, A.-T., Troumpata, L., Periferakis, K., Georgatos-Garcia, S., Touriki, G., Dragosloveanu, C. D. M., Caruntu, A., Savulescu-Fiedler, I., Dragosloveanu, S., Scheau, A.-E., Badarau, I. A., Caruntu, C., & Scheau, C. (2025). Pinosylvin: A Multifunctional Stilbenoid with Antimicrobial, Antioxidant, and Anti-Inflammatory Potential. Current Issues in Molecular Biology, 47(3), 204. https://doi.org/10.3390/cimb47030204

Article Metrics

Back to TopTop