Next Article in Journal
The Development and Evaluation of a Prediction Model for Kidney Transplant-Based Pneumocystis carinii Pneumonia Patients Based on Hematological Indicators
Next Article in Special Issue
Mitochondria-Targeted Catalase Does Not Suppress Development of Cellular Senescence during Aging
Previous Article in Journal
Transcriptome-Wide N6-Methyladenosine Alternations in Pulmonary Arteries of Monocrotaline-Induced Pulmonary Arterial Hypertension in Rats and Novel Therapeutic Targets
Previous Article in Special Issue
Suppression of Bcl3 Disrupts Viability of Breast Cancer Cells through Both p53-Dependent and p53-Independent Mechanisms via Loss of NF-κB Signalling
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Plant Monoterpenes and Essential Oils as Potential Anti-Ageing Agents: Insights from Preclinical Data

by
Mónica Zuzarte
1,2,3,*,
Cátia Sousa
4,5,
Jorge Alves-Silva
1,2,3 and
Lígia Salgueiro
1,6
1
Univ Coimbra, Faculty of Pharmacy, Azinhaga de S. Comba, 3000-548 Coimbra, Portugal
2
Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Azinhaga de S. Comba, 3000-548 Coimbra, Portugal
3
Clinical Academic Centre of Coimbra (CACC), 3000-548 Coimbra, Portugal
4
iNOVA4HEALTH, NOVA Medical School, Faculdade de Ciências Médicas (NMS/FCM), Universidade Nova de Lisboa, 1159-056 Lisboa, Portugal
5
Centro Clínico e Académico de Lisboa, 1156-056 Lisboa, Portugal
6
Univ Coimbra, Chemical Engineering and Renewable Resources for Sustainability (CERES), Department of Chemical Engineering, 3030-790 Coimbra, Portugal
*
Author to whom correspondence should be addressed.
Biomedicines 2024, 12(2), 365; https://doi.org/10.3390/biomedicines12020365
Submission received: 28 December 2023 / Revised: 25 January 2024 / Accepted: 26 January 2024 / Published: 4 February 2024
(This article belongs to the Special Issue Cellular Senescence: Recent Advances and Discoveries)

Abstract

:
Ageing is a natural process characterized by a time-dependent decline of physiological integrity that compromises functionality and inevitably leads to death. This decline is also quite relevant in major human pathologies, being a primary risk factor in neurodegenerative diseases, metabolic disorders, cardiovascular diseases and musculoskeletal disorders. Bearing this in mind, it is not surprising that research aiming at improving human health during this process has burst in the last decades. Importantly, major hallmarks of the ageing process and phenotype have been identified, this knowledge being quite relevant for future studies towards the identification of putative pharmaceutical targets, enabling the development of preventive/therapeutic strategies to improve health and longevity. In this context, aromatic plants have emerged as a source of potential bioactive volatile molecules, mainly monoterpenes, with many studies referring to their anti-ageing potential. Nevertheless, an integrated review on the current knowledge is lacking, with several research approaches studying isolated ageing hallmarks or referring to an overall anti-ageing effect, without depicting possible mechanisms of action. Herein, we aim to provide an updated systematization of the bioactive potential of volatile monoterpenes on recently proposed ageing hallmarks, and highlight the main mechanisms of action already identified, as well as possible chemical entity–activity relations. By gathering and categorizing the available scattered information, we also aim to identify important research gaps that could help pave the way for future research in the field.

1. Introduction

The number and proportion of people aged 60 years or older is increasing at an unprecedented rate and, by 2050, will reach up to 2.1 billion, as stated by WHO [1]. Indeed, population ageing is the most global demographic trend, conveying several public challenges primarily in the health sector. The ageing process is associated with an impairment of several body functions and an increase in age-related disorders, including neurodegenerative, cardiovascular, cancer and metabolic diseases [2]. Several factors including genetics, lifestyle and environmental features are key in this inevitable gradual process, and interventions that promote health and longevity, thus increasing lifespan, are a matter of current interest and research.
In the last years, several strategies have been proposed to delay ageing, namely caloric restriction, nutritional interventions and microbiota transplantation [3,4]. In addition, specific treatments including depletion of senescent cells, stem cell therapy, antioxidant and anti-inflammatory treatments, and hormone replacement therapy have been used in order to promote healthy ageing and longevity [5]. The identification of potential drugs for age-related diseases such as metformin, rapamycin, resveratrol and senolytics is also a hot topic in the field, although several recognized limitations and side effects raise some concerns that need further clarification [2].
The main cellular and molecular mechanisms involved in biological ageing enabled the identification, in 2013, of ageing hallmarks [6]. Later, in 2022, a research symposium held in Copenhagen considered the addition of new hallmarks to the existing list, bearing in mind their role in the ageing process [7]. The accepted current list now includes 12 ageing hallmarks that comprise genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, compromised autophagy, deregulated nutrient sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion, altered intercellular communication, chronic inflammation and dysbiosis [8]. These hallmarks have proven to be very useful in the identification of intervention targets, primarily in age-related diseases with promising results in preclinical models [9,10] and validations in clinical trials [11]. Among the tested compounds, natural compounds have attracted researchers’ interest, with recent studies pointing out their effect on critical ageing pathways such as mammalian target of rapamycin (mTOR), adenosine monophosphate-activated kinase (AMPK), Sirtuin 1 (SIRT1) and tumor protein p53, as reviewed elsewhere [12]. Nevertheless, the majority of the studies resort to in vitro approaches, lacking in-depth analysis of individual compounds and specific effects on these pathways [12]. Bearing this in mind, we sought to review the potential of volatile monoterpenes, a group of compounds highly prevalent in aromatic plants. These compounds are valued by several industries due to their bioactive potential, namely antimicrobial, antioxidant, anti-inflammatory, analgesic, antinociceptive and anticancer effects, as reviewed elsewhere [13]. Importantly, many monoterpenes are considered safe for consumer use and have a generally recognized as safe (GRAS) status by the Food and Drug Administration (FDA), being included in many foods and beverages.
In the present review, an up-to-date overview of the effect of monoterpenes is provided, considering the current hallmarks of ageing. In addition, whenever known, the main mechanisms of action underlying the observed effects are stated, and a possible chemical entity–bioactivity relation is attempted. Importantly, the main gaps in the current knowledge are pointed out to help guide future research in the field.

2. Aromatic Plants and Volatile Compounds

The term aromatic plants refers to plants that produce volatile compounds. These compounds can be extracted from the plant source as a volatile mixture called essential oil. To comply with regulatory requirements, in particular those from the International Standard Organization on Essential Oils [14] and the European Pharmacopoeia [15], essential oils are obtained from the plant raw material by hydrodistillation, steam distillation or dry distillation, or by a suitable mechanical process in the case of Citrus fruits. Several monographs on essential oils are available, for example, in the European Pharmacopoeia [15] or the European Medicine Agency [16], which highlights their importance in the pharmaceutical industry. The main types of compounds present in essential oils are monoterpenes and sesquiterpenes. Monoterpenes are isoprene dimers with carbon and hydrogen atoms (C10H16), such as α-pinene, limonene, camphene, myrcene and p-cymene. Monoterpenoids include in their chemical structure functional groups like alcohols (e.g., linalool, geraniol, menthol), esters (e.g., linalyl acetate, geranyl acetate, neryl acetate), ketones (e.g., camphor, menthone, carvone), aldehydes (e.g., geranial, neral), ethers (e.g., 1,8-cineole) or phenols (e.g., carvacrol, thymol) [17]. Monoterpenes are synthetized from geranyl pyrophosphate by monoterpene synthases and, according to their structure, can be classified as acyclic, monocyclic or bicyclic. These volatile compounds generally present a pleasant aroma and can be found in fruits, vegetables, spices and herbs, as well as in different plant parts like leaves, flowers, seeds and roots. They are appreciated as aromatic and flavoring agents in cosmetic, pharmaceutical and food industries. Also, emerging evidence shows that monoterpenes present several biological properties including antimicrobial, antioxidant, anti-inflammatory and cardioprotective, as previously reviewed [18]. These findings have led to the incorporation of monoterpenes in distinct products, such as diet supplementation, animal feed additives, food packaging and biopesticides, among others. Interestingly, modifications of these compounds have also been considered with growing evidence on the biological and medical application of monoterpene derivatives [19].

3. Anti-Ageing Potential of Low-Molecular-Weight Terpenes

3.1. Ageing Hallmarks

Ageing is characterized by a progressive loss of physiological integrity and impaired function that increases vulnerability to pathology or age-associated diseases, such as neurodegenerative diseases, metabolic disorders, cardiovascular diseases and musculoskeletal disorders [6]. Ageing per se does not cause these diseases but favours their clinical manifestation [20], since the mechanisms driving ageing and those driving age-associated diseases largely overlap [21].
In 2013, López-Otín and colleagues proposed nine hallmarks of ageing, namely genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, deregulated nutrient sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion and altered intercellular communication. Accordingly, to be considered a hallmark, these processes should manifest during ageing; their aggravation, resorting to experimental models, should accelerate the ageing process, and experimental improvement should delay the ageing process [6]. During the last years, extensive research in the ageing field and its hallmarks has been carried out, leading to an update of the initially proposed list [8]. Thus, three new hallmarks were added, namely disabled macroautophagy, chronic inflammation and dysbiosis. Indeed, López-Otín and colleagues considered that macroautophagy does not only affect proteins but also targets organelles and non-proteinaceous macromolecules, thereby being separated from the hallmark loss of proteostasis. Moreover, the authors also suggested that altered cellular communication was a vast hallmark, so chronic inflammation and dysbiosis were separated from it [8].
Independently of the number of hallmarks considered, there is an established hierarchy among them [6,8]. Thus, the hallmarks are categorized as: primary, which reflects the damage, inflicted to the genome, telomeres, epigenome, proteome and organelles; antagonistic, which reflects the response to the damage and includes deregulated nutrient sensing, mitochondrial dysfunction and cellular senescence; and integrative, which considers the end result, leading to stem cell exhaustion, intercellular communication alterations, chronic inflammation and dysbiosis. Consequently, the integrative hallmarks are responsible for the functional decline associated with ageing [6,8].
Interestingly, the hallmarks of ageing are not independent, implying that experimental accentuation or attenuation of one specific hallmark will consequently affect the others [8]. This is an interesting aspect to bear in mind, regarding the discovery and/or development of new drugs to tackle ageing and related diseases.

3.2. Monoterpenes and Essential Oils in Ageing Hallmarks

In this section, a compilation of the main studies reporting the effect of volatile monoterpenes on ageing hallmarks is presented. The search was carried out in the PubMed database combining the words “monoterpene” and each hallmark of ageing, without date restrictions. The results obtained are presented in the following subsections. However, for inflammation, due to the high number of studies on this topic, a more targeted search was carried out, as detailed in Section 3.2.3. For each hierarchized hallmark, a brief overview is provided followed by a table summarizing the studies organized in alphabetic order of the compound name. Information on the in vitro or in vivo study model used and the main effects reported is shown. In addition, evidence regarding the effect of essential oils on these hallmarks is also provided.

3.2.1. Primary Hallmarks

Genomic Instability

Genomic instability is generally characterized by genetic lesions, which include point mutations, deletions, translocations, telomere shortening, single and double strand breaks, chromosomal rearrangements, defects in nuclear architecture and gene disruption. These lesions can be caused by extrinsic (e.g., chemical, physical and biological agents) and intrinsic (e.g., DNA replication errors, chromosome segregation defects, oxidative processes and spontaneous hydrolytic reactions) sources. To maintain the genomic integrity and stability, organisms have a complex network of DNA repair mechanisms. Nevertheless, excessive DNA damage or insufficient repair favours the ageing process [6,8]. Different tools have been developed to detect and analyse genomic instability, particularly in cancer models, and include DNA damage, micronuclei development, chromatin and anaphase bridge formation, as well as aneuploidy [22]. Studies reporting the in vitro effect of monoterpenes on genomic instability are summarized in Table 1.
Although the primary hallmarks of ageing are the primary causes of cell damage, only two in vitro studies reported the negative effects of monoterpenes on genomic instability, namely α-pinene and D-limonene. These monoterpenes are quite frequent in several essential oils, α-pinene being characteristic of Pinus spp. (pine) essential oils and D-limonene highly prevalent in Citrus spp. essential oils. The results presented in Table 1 suggest that α-pinene and D-limonene favour genomic instability. Indeed, α-pinene induces genomic instability by promoting mitotic alterations and reactive oxygen species (ROS) production, which causes DNA damage [23]. On the other hand, D-limonene affects the cell division by preventing the assembly of mitotic spindle microtubules, which then affect chromosome segregation and cytokinesis, consequently leading to aneuploidy [24].
Regarding other primary hallmarks, a study on the effect of Thymus vulgaris (thyme) essential oil on blood telomere attrition, in chronologically aged C57BL/6J mice, was carried out [25]. Interestingly, the authors reported a beneficial effect and demonstrated that mice supplemented with this essential oil (0.2% w/w) showed higher survival rates and significantly longer blood telomere lengths than the control mice. Although the study did not assess isolated compounds, the authors suggest that the phenolic monoterpene thymol and the monoterpene hydrocarbon p-cymene, present in high amounts in the oil, may primarily contribute to the observed telomere-protecting effect, due to their antioxidant and anti-inflammatory potentials [25]. Thus, it seems that the aromatic ring might be responsible for the different outcomes observed in these primary hallmarks, particularly genomic instability and telomere attrition. However, further studies are needed to verify this hypothesis.

3.2.2. Antagonistic Hallmarks

Mitochondrial Dysfunction

Mitochondria are organelles that play a vital role in homeostasis maintenance [26]. Deterioration in mitochondria function increases with ageing due to the occurrence of diverse mechanisms, namely accumulation of mitochondrial DNA mutations and changes in their dynamics. These alterations might compromise the mitochondria contribution to cellular bioenergetics, increasing the production of ROS and, consequently, promoting the mitochondria membranes permeabilisation. The latter phenomenon leads to inflammation and cell death [8].
Mitochondrial dysfunction can be assessed both in vitro and in vivo. For example, isolated mitochondria can be used to assess mitochondria respiratory capacity, defined as the increase in respiration rate in response to ADP. In cells, studies include the measurement of several parameters such as ATP production rate, proton leak rate, coupling efficiency, maximum respiratory rate, respiratory capacity ratio and spare respiratory capacity [27]. Table 2 compiles both in vitro and in vivo studies on the effect of monoterpenes on mitochondrial dysfunction.
Combining both in vitro and in vivo studies, 1,8-cineole (synonym of eucalyptol), carvacrol, geraniol and perillyl alcohol are the most studied compounds. These compounds can be found in the essential oils of several aromatic plants; for example, 1,8-cineole is very common in Eucalyptus globulus, while carvacrol is characteristic of Origanum species, geraniol is abundant in Rosa spp. and perillyl alcohol occurs in different Mentha species. A closer analysis of the information presented in Table 2 shows that the studied monoterpenes have a beneficial effect, thus inhibiting mitochondrial dysfunction in both in vitro and in vivo models. The majority of the tested compounds present in their structure a hydroxyl group or a phenol group that might contribute to the observed effects. In general, the tested monoterpenes decreased ROS production, thus showing a beneficial effect, by restoring the mitochondrial membrane potential as shown for 1,8-cineole [29,31] and geraniol [34]. The increase in mitochondrial membrane potential is also beneficial as ATP synthase activity is enhanced [51]. Indeed, most studies reporting an increase/restoration of mitochondrial membrane potential also demonstrate an increase in ATP production, such as geraniol [34] and carvacrol [33]. The activity of the mitochondrial respiratory chain complexes can also contribute to the maintenance of the mitochondrial membrane potential through proton flux [51]. These effects were reported, for example, for geraniol [34] and linalool [37].
Besides monoterpenes, several studies have reported the effects of essential oils on mitochondrial dysfunction, although in distinct contexts not directly related to ageing. Indeed, the majority of the studies are carried out in cancer models, thus showing an overall beneficial effect but through an enhancement of mitochondrial dysfunction. For example, Schisandrae semen essential oil induced ROS- and caspase-dependent cell death involving mitochondrial dysfunction and nuclear translocation of mitochondrial pro-apoptosis proteins in human leukaemia U937 cells [52]. The essential oil of Alpinia officinarum promoted lung cancer regression by triggering mitochondrial membrane potential dysfunction and activating caspase-3 cleavage, inducing cell apoptosis [53]. Cinnamomum cassia essential oil showed anti-oral cancer activity in HSC-3 cells, through a significant increase in ROS production [54]. The cytotoxic potential of Croton heliotropiifolius on different human cancer cell lines, namely leukaemia, colon, melanoma and glioblastoma, was also related to oxidative stress, culminating in mitochondrial respiratory dysfunction and DNA damage, triggering cell death via apoptosis [55]. In the context of cancer, some monoterpenes like carvacrol [56] and hinokitiol (synonym of β-thujaplicin) [57] are also able to potentiate mitochondrial dysfunction.
In addition to essential oils’ anticancer effects, their antimicrobial properties have also been related to mitochondrial dysfunction, being the mitochondrial impairment referred to as the mechanism of action that compromises infection. These effects were shown, for example, in Rosmarinus officinalis essential oil against Candida albicans [58], Piper nigrum essential oil against Aspergillus flavus [59], and Anethum graveolens essential oil against both A. flavus [60] and C. albicans [61]. Moreover, insecticidal properties of essential oils through mitochondrial impairment in insects have been reported, for example, for Eugenia uniflora essential oil in Drosophila melanogaster [62] and Melaleuca alternifolia, that prevented bioenergetics dysfunction in the spleen of silver catfish naturally infected with Ichthyophthirius multifiliis [63].

Cellular Senescence

Cellular senescence is a state of permanent cell cycle arrest occurring when telomere length decreases below a critical size, in response to different damaging stimuli, such as oxidative stress and DNA damage [64,65]. This is an important barrier mechanism to tumorigenesis as it limits the growth of potentially oncogenic cells [65]. Moreover, cellular senescence plays a key role in some physiological processes such as embryogenesis, tissue remodelling and repair [65]. The senescent phenotype is characterized by chronic activation of the DNA damage response, upregulation of Cyclin-Dependent Kinase (CDK) inhibitors (e.g., p16INK4a, p15INK4b and p21CIP), apoptosis resistance, altered metabolic rates, endoplasmic reticulum stress and increasing secretion of pro-inflammatory and tissue remodelling factors, known as the Senescence-Associated Secretory Phenotype (SASP) or senescence-messaging secretome (SMS) [64]. The SASP is quite important to ensure the efficient growth arrest by autocrine signalling, in particular immediately after senescence induction, to signal senescent cells for clearance by the immune system and, consequently, for tissue repair and remodelling [66,67]. Moreover, the SASP is highly heterogeneous [64] and can include a wide range of cytokines, chemokines, proteases, growth factors [66] and non-macromolecular elements [65]. Although various signalling pathways are involved in the regulation of SASP components, most of them converge to NF-κB activation [65,66]. Morphologically, senescent cells show structural aberrations, including an enlarged and more flattened shape; modified composition of the plasma membrane due to an upregulation of caveolin-1; increased lysosomal content which is directly correlated with higher senescence-associated β-galactosidase activity (pH 6.0); accumulation of mitochondria and nuclear changes like loss of Lamin B1 and formation of senescence-associated heterochromatin foci [64]. Excessive and anomalous accumulation of senescent cells in tissues has a negative impact on homeostasis mainly via SASP [67]. This phenomenon can be detrimental to regenerative capacities and generate a disruptive pro-inflammatory environment that is favourable for the onset and progression of a variety of age-related diseases [64,67]. The accumulation of these cells can occur during ageing [65].
González-Gualda and colleagues proposed that, at least, three different types of markers that confirm (i) cell cycle arrest, (ii) increased lysosomal compartment or another global senescent-related structural change and (iii) an additional trait specific for the particular type of senescence being assessed (such as SASP or DNA damage features) should be considered to assess senescence [68]. Monoterpenes known to possess anti-senescent effects are summarized in Table 3.
Regarding the effect of monoterpenes on cellular senescence, only five compounds have been tested for their senolytic or senomorphic effects. Senolytic compounds are those that are able to effectively kill senescent cells, by modulating several associated pathways, such as p21/p53 and by inducing apoptosis, whereas senomorphic compounds are responsible for inhibiting SASP, through modulation of associated pathways, such as NF-κB, mTOR and JAK/STAT [73]. For instance, α-pinene can be considered a senolytic compound due to its effect on the p21/p53 axis [71], thus increasing DNA repair, preventing the entrance in cell cycle arrest and, consequently, senescence [74]. D-Limonene also acts as a senolytic, probably by inducing cell death by apoptosis as it prevents the nuclear translocation of p53 [35], which is known to induce mitochondria-dependent apoptosis [75]. Myrcene, on the other hand, showed a senomorphic effect through a direct decrease of IL-6 secretion [39]. Secretion of matrix-degrading mediators, such as metalloproteinases, is also characteristic of SASP [73]. Having this in mind, α-pinene [72], camphor [69], myrcene [39] and hinokitiol [70] are all considered senomorphics due to their inhibitory effect in the synthesis or secretion of metalloproteinases. The compounds, referred to in Table 3, are present in several essential oils. Camphor is typically encountered in Cinnamomum camphora, hinokitiol is present in Chamaecyparis spp., D-limonene is characteristic of Citrus fruit peels, myrcene occurs in Cannabis sativa and in several other essential oils and α-pinene is one of the main compounds in Pinus spp.
Interestingly, other studies assessed the effect of monoterpenes on features related to senescence but in a cancer context, similar to that reported in the previous section. In these cases, an enhancement in cellular senescence is the beneficial effect as it negatively affects the cancer cell growth. For example, a nanoemulsion of carvacrol induced cell senescence leading to cell cycle arrest by reducing CDK2, CDK4, CDK6, Cyclin E, Cyclin D1 and enhancing p21 protein expressions. Also, an increase in SA-β-galactosidase activity was observed [56]. Hinokitiol decreased cell proliferation and increased DNA damage and phase S cell cycle arrest in osteosarcoma cells U2OS and MG63 [76], while increasing γH2AX-positive cells in a xenograft mice model [77]. Other monoterpenes assessed in the context of cancer include 1,8-cineole that was effective in increasing phase G0/G1 cell cycle arrest, while decreasing CDK2, CDK4 and CDK6 and cyclin A protein levels. This compound also increased p27 and cyclin D1 protein levels and SA-β-galactosidase activity [78]. Geraniol increased phase sub G1 cell cycle arrest in prostate cancer cells PC3 [79]. Menthol and α-phellandrene increased p53 levels in the leukaemia cell lines NB4 and Molt 4 [80] and nuclei levels of p53 while decreasing its cytoplasmic levels in the hepatocellular cell line J5, respectively [81].
Regarding essential oils, several studies have assessed their effect on cellular senescence. For example, Thymbra capitata decreased SA-β-galactosidase activity in fibroblasts treated with the senescence inducer etoposide [82]. Similar results were reported for Santolina rosmarinifolia [83], Salvia aurea [84] and Ferulago lutea [85]. For the latter, additional senescence features (p21/p53 protein levels and the nuclear accumulation of γH2AX) were also decreased following essential oil treatment [85]. In addition, Eucalyptus globulus essential oil showed anti-senescent effects in both etoposide-stimulated keratinocytes (HaCaT) and fibroblasts (NIH/3T3). Thus, in the presence of the oil, the number of senescent cells decreased as well as the levels of p53 [86]. Other essential oils showed beneficial properties in vitro, senescence being induced by distinct agents. For example, several Cistus spp. oils significantly attenuated UVB-induced cellular senescence in a keratinocyte cell line (HaCaT) [87], while Cymbopogon nardus and Cymbopogon citratus oils showed cytoprotective properties on doxorubicin-induced senescence in the fibroblast-like kidney cell line (Vero) and fibroblasts (NIH/3T3) [88].
An in vivo study was also carried out in diabetic mice with Alpiniae zerumbet administered orally, ameliorating vascular endothelial cell senescence by activating PPAR-γ signalling [89].

3.2.3. Integrative Hallmarks

Disabled Macroautophagy

Autophagy is a physiological cellular mechanism whereby cytoplasmic material is delivered to the lysosome for degradation [90]. Macroauthophagy is the most well characterized and involves the sequestration of cytoplasmic material, including soluble macromolecules and organelles, into a double- or multi-membrane-bound structure, named autophagosome. These structures will then fuse with the lysosome, promoting degradation and recycling of its content [90]. This section will focus only on macroautophagy, henceforth referred to as autophagy. Under normal conditions, constitutive autophagy occurs as a housekeeping mechanism [91]. However, autophagy can be activated during starvation and stress conditions such as hypoxia, increased ROS production, DNA damage, protein aggregates, damaged organelles or intracellular pathogens, in order to restore homeostasis [92]. Moreover, it is well established that autophagy declines with age [6,93].
Several approaches have been developed to assess autophagy, namely immunoblotting of canonical markers like LC3 and p62, detection of autophagosomes by fluorescence microscopy or autophagosome maturation resorting to mRFP-GFP fluorescence microscopy, although a combined use of several methods is recommended [94]. Table 4 includes studies on the effect of monoterpenes on autophagy.
In what concerns the effect of monoterpenes on autophagy, both in vitro and in vivo studies are available and, similarly to that shown in the mitochondrial dysfunction section, the majority of the tested compounds present in their structure a hydroxyl group or a phenol group that might contribute to the observed effects. For this hallmark, several compounds have been tested in vivo: for example, borneol that occurs in Salvia rosmarinus; carvacrol present in Origanum spp.; 1,8-cineole characteristic of Eucalyptus spp.; citronellol found in Cymbopogon nardus; geraniol abundant in several flowers, such as Rosa spp.; myrcene present, for example, in Cannabis sativa; and thymol frequent in Thymus spp.
Regarding their effect on autophagy, monoterpenes can be divided into those that promote autophagy and those that decrease it. Although these effects seem contradictory, it is important to refer that the disease model used differs between the studies, and therefore, the monoterpenes increase or decrease autophagy in order to restore homeostasis. For instance, carvacrol [43,95], 1,8-cineole [29,31], hinokitiol [98], menthol [100] and borneol [102] induced an increase in autophagy, by enhancing the ratio LC3B-II/LC3B-I, LC3 and/or Beclin-1 levels, whereas decreasing that of p62. On the other hand, some studies showed the opposite effect. For example, 1,8-cineole [96,97], hinokitiol [99], thymol [101], citronellol [103], geraniol [104] and myrcene [105] decreased the ratio of LC3B-II/I, LC3 and/or Beclin-1 levels, while increasing that of p62 and/or p-mTOR levels, consequently reducing autophagy.
Besides the studies highlighted in Table 4, the effect of monoterpenes on autophagy has also been reported in cancer cells. The majority of the compounds tested in this setting increase autophagy, thus suppressing tumorigenesis by inhibiting cell survival and inducing cell death. For example, hinokitiol was largely assessed and increased autophagy in vitro in hepatoblastoma cell line HepG2 [57], HeLa cervical cancer cells [107], U2OS and MG63 osteosarcoma cells [76] and H1975 lung cancer cells [77], through an increase in LC3 levels, Beclin-1 levels and/or autophagosome formation, among other autophagy markers. In addition, α-phellandrene induced autophagy in human hepatocellular carcinoma (J5) cells by regulating mTOR and LC3-II expression, as well as p53 signalling. Indeed, an increase in autophagic vacuoles formation was observed together with a decrease in PI3K, Akt and mTOR protein levels and an increase in Beclin-1 and LC3-II [81]. In addition, α-thujone induced oxidative stress and autophagy in a glioblastoma cell line (T98G), as shown by an increase in autophagolysosome formation, an increase in LC3-II/LC3-I ratio, as well as an increase in the levels of LC3-II, Beclin-1, Atg3, Atg5 and Atg7 [108]. Furthermore, terpinen-4-ol induced an accumulation of LC3-I/II, Atg5 and Beclin-1, as well as regulatory proteins required for autophagy in human leukemic HL-60 cells [109]. In vivo studies also pointed out the effects of hinokitiol on the inhibition of xenograft tumour growth in association with DNA damage and autophagy, as shown by the expression of γ-H2AX and LC3 in the tumour tissue [77].
Essential oils also affect autophagy, and have been assessed in distinct contexts, namely cancer and cell toxicity, among others. Concerning anticancer effects, for example, the essential oil of Origanum montana inhibited colony growth of human HT-29 colorectal cancer cells by inducing protective autophagy, associated with downregulation of the mTOR/p70S6K pathway and apoptotic cells death, via the activation of the p38 MAPK signalling pathway [110]. In what concerns cell toxicity, the essential oil of Schisandra chinensis was able to attenuate acetaminophen-induced liver injury by alleviating oxidative stress and activating autophagy, showing an upregulation of hepatic LC3-II and a decrease in p62 in mice with an overdose of acetaminophen [111]. Moreover, the essential oil of Acorus tatarinowii ameliorated Aβ-induced toxicity in Caenorhabditis elegans by maintaining protein homeostasis through the autophagy pathway regulated partly by hsf-1 and sir-2.1 genes [112].

Inflammation

According to Antonelli and Kushner (2017), inflammation is defined as “an innate immune response to harmful stimuli such as pathogens, injury and metabolic stress that aims to restore homeostasis” [113]. In basal conditions, tissues are maintained in the homeostatic state with the aid of tissue-resident macrophages, when it is required. Nevertheless, in noxious conditions, tissues undergo stress and can malfunction. In the case of considerable changes, adaptation to these conditions requires the help of tissue-resident macrophages or the recruitment of other macrophages and may require small-scale delivery of additional leukocytes and plasma proteins. This response has characteristics that are intermediate between basal and inflammatory states and was termed para-inflammation [114]. Para-inflammation is not a classic form of inflammation triggered by exogenous tissue injury or infection, but it is switched on by tissue malfunction in order to promote its adaptation to a harmful environment and to maintain its adequate functionality. This state is characterized by a low-grade/subclinical immune reaction [114]. Nevertheless, if the tissue malfunction is present for a sustained period, para-inflammation progresses into a chronic low-grade inflammation (pathophysiological para-inflammation) [115,116]. Ageing and related diseases are characterized by the presence of chronic low-grade inflammation [8,114]. Moreover, chronic inflammation might be a consequence of alterations in other hallmarks of ageing and vice-versa, representing a vicious cycle [8,117]. This hallmark is, by far, the most addressed with a PubMed bibliographic search on “monoterpenes” and “inflammation” encountering more than 1500 studies. Therefore, only the studies performed in the last 4 years that assessed the mechanism of action associated to the observed effect were considered, being summarized in Table 5. The anti-inflammatory potential of monoterpenes is usually studied by evaluating the expression of pro-inflammatory mediators (e.g., IL-1β, IL-6, TNF-α, iNOS, NO and COX-2), anti-inflammatory mediators (e.g., IL-10) and the impact on signalling pathways relevant to the inflammation process, particularly NF-κB, MAPKs and Nrf2.
As shown in Table 5, monoterpenes with different chemical structures are able to impact inflammation. Previously, a standardized analysis of the anti-inflammatory activity of p-menthane-derived monoterpenes, namely (R)-(+)-limonene, (S)-(−)-limonene, (S)-(+)-carvone and (R)-(−)-carvone, was carried out by Sousa and colleagues. Considering the carbon numbering relative to the common precursor (limonene), the authors highlighted that the presence of an oxygenated group at C6 conjugated to a double bond at C1 and an isopropenyl group and S configuration at C4 are the major chemical features relevant for activity and potency [164]. Nevertheless, further studies are needed to clarify the structure–activity relation of these molecules using similar models and endpoints.
Most of the studies herein reported assess the effect of monoterpenes on the NF-κB signalling pathway as this transcription factor is the master regulator of inflammation [117]. For instance, carvacrol [118], citral [122], geraniol [126], perillyl alcohol [129,130], thymol [132], among others (Table 5) interfered with the canonical activation of NF-κB, blocking its nuclear translocation and, consequently, its transcriptional activity. On the other hand, some monoterpenes did not interfere directly with NF-κB activation but promoted the activation of other molecules, namely SIRT1 and Nrf2, which antagonize the transcriptional activity of NF-κB [119,120,121,131]. Examples of these compounds include (S)-(+)-carvone [119], (R)-(−)-carvone [120], 1,8-cineole [121] and perillaldehyde [131]. Other signaling pathways were also evaluated, namely MAPK and inflammasome NLRP3 that include menthol [128] and hinokitiol [127], respectively.
Aside from the studies presented in Table 5, many others were found in the bibliographic search reporting the anti-inflammatory activity of monoterpenes by evaluating the expression of pro-inflammatory and/or anti-inflammatory mediators, as these are the most common endpoints found in the inflammatory response. Examples include carvacrol [165,166,167,168], 1,8-cineole [169,170], citral [171,172], isopulegol [173], linalool [174,175], linalyl acetate [174] and thymol [176,177,178].
Similar to monoterpenes, many studies also report the anti-inflammatory potential of essential oils. Indeed, the combined search of “essential oil” and “chronic inflammation” brings up 37 reviews in PubMed. Regarding original studies, the essential oil of Thymus vulgaris was assessed in ageing-induced brain inflammation in chronologically aged C57BL/6J mice and showed a significantly lower gene expression of the pro-inflammatory cytokine Il6 in the hippocampus and lower Il1b expression in the liver and cerebellum, [25]. Many other studies have been performed not directly related to ageing, with some genera standing out as the most studied. These include essential oils from Cinnamomum spp. [179,180], Lavandula spp. [181,182], Thymus spp. [183,184] and Citrus spp. [185,186,187].

Dysbiosis

Microbiota includes the microorganisms (e.g., bacteria, fungi, viruses and parasites) that inhabit all parts of our body and can affect human health. The composition and tissue colonization by microbiota are affected by external and internal factors. The former include mainly infant-related factors, such as the mode of delivery, the gestation time and the feeding type, as well as the type of diet and use of antibiotics. The latter includes host internal factors, particularly genetics and the immune system [188]. Importantly, alterations in the microbiome have been recently associated with the emergence of several chronic diseases such as cancer, diabetes, cardiovascular diseases, as well as some psychological disorders, for example, schizophrenia [189]. Moreover, ageing also affects microbiota, particularly gut microbiota, and is associated with microbiome disturbance, named dysbiosis, which is characterized by a shift in microbiota populations and the loss of diversity [8,190]. Indeed, ageing-induced dysbiosis has been associated with the development of several diseases such as lung diseases [191], cardiovascular diseases [192], glaucoma [193] and neurological disorders [194].
Microbiome can be assessed resorting to a plethora of technologies, particularly gene marker analysis, shotgun metagenomics, metabolomics, metaproteomics and metatranscriptomics [195]. Although gene marker analysis that resorts to a targeted sequencing method encompassing the 16S ribosomal RNA gene for bacteria and internal transcribed spacer (ITS) region for fungi is the most common technique, it fails to include other microorganisms, such as virus. Having that in mind, shotgun metagenomics was developed, which resorts to untargeted sequencing methods that allow the capture of the full repertoire of genetic information from a sample, detecting the presence of several microbes. The remaining technologies rely on the detection of expressed microbiome-related genes in the sample (metatranscriptomics), on the small metabolites and their interaction with the host and vice versa (metabolomics) and on the proteins present in the sample (metaproteomics) [195]. The keyword combination of “dysbiosis” and “monoterpenes” only gave back one result, as shown in Table 6. The authors of the study resorted to gene marker analysis by sequencing the 16S ribosomal gene in order to identify the main bacteria found in the mice belonging to the Bacteriodetes and Firmicutes divisions.
The study herein reported shows that geraniol is able to prevent colitis-associated dysbiosis in the dextran sulphate sodium-induced colitis mouse model, when administrated orally or through enema [196].
Essential oils have also shown promising effects on dysbiosis. For example, several essential oils were tested against species of intestinal bacteria mostly found in the human gastrointestinal tract. Overall, Carum carvi, Lavandula angustifolia, Trachyspermum copticum and Citrus aurantium var. amara essential oils were the most effective in inhibiting pathogens growth without affecting beneficial bacteria [197]. Another study showed that Zanthoxylum bungeanum essential oil showed a beneficial effect on chronic unpredictable stress-induced anxiety behaviour in rats, by restoring gut microbiota dysbiosis, namely by increasing the Sobs and Chao indexes, inhibiting Lachnospiraceae, facilitating Bacteroidales_S24-7_group, Lactobacillaceae and Prevotellaceae [198]. Essential oil emulsions of Satureja hortensis, Petroselinum crispum and Rosmarinus officinalis were also administered to humanized mice harbouring gut microbiota derived from patients with ischemic heart disease and Type 2 diabetes mellitus. Overall, the essential oil emulsions in mice supplemented with L-carnitine showed prebiotic effects on beneficial commensal bacteria, mainly the Lactobacillus genus [199]. Furthermore, a review carried out on the modulation of gut microbiota by essential oils and inorganic nanoparticles showed that essential oils are widely studied for their gastrointestinal interference, being potent modulators for gut inflammation, metabolic reactions and microbiota diversity [200].

4. Discussion and Future Perspectives

Studies on monoterpenes’ effect on ageing hallmarks have been carried out with many compounds presenting in their chemical structure a hydroxyl group or a phenol group that might justify the observed effects. However, an accurate chemical structure–activity relation is difficult to establish due to the multitude of study models used, concentrations tested and observed effects that hamper direct comparisons. Figure 1 shows the chemical structures of the compounds summarized in Table 1, Table 2, Table 3, Table 4, Table 5 and Table 6. Importantly, some monoterpenes like carvacrol, 1,8-cineole, limonene, perillyl alcohol and thymol have been largely assessed, showing relevant effects on different ageing hallmarks. Overall, inflammation is the hallmark mostly addressed with more than 1500 studies identified in a PubMed bibliographic search combining “monoterpenes” and “inflammation”. On the other hand, some hallmarks remain elusive, lacking studies on the effect of these compounds, namely epigenetic alterations, loss of proteostasis, deregulated nutrient sensing, stem cell exhaustion and altered intercellular communication (Figure 2).
As shown by the review herein presented, monoterpenes seem to be very promising anti-ageing compounds, but it is important to highlight that the majority of the studies assess the hallmarks individually and not in an ageing context. Indeed, several compounds are studied in a cancer setting or are performed to justify the mechanism of action associated with other properties of these compounds, namely antimicrobial or insecticidal. In these situations, monoterpenes also exerted a beneficial effect but through an enhancement of the studied hallmark, as cancer cell or microbe eradication is aimed. In addition, regarding senescence studies, the majority of the studies do not address, as recommended, at least three markers. Additional studies resorting to ageing models are, therefore, needed and could include monoterpenes with anti-inflammatory properties, as these will most likely exert a senomorphic effect by modulating components of the SASP. Moreover, essential oils have also shown very promising effects, and one cannot neglect possible synergistic effects between several compounds, including monoterpenes, present in the mixture. Therefore, further studies are still required to elucidate the real potential of monoterpenes to boost natural defences during ageing and extend healthy lifespan in the elderly.
The majority of the studies include in vitro and in vivo approaches, and, in line with this, clinical trials have also been carried out, although in much lesser extent. Interestingly, carvacrol and geraniol are the monoterpenes mostly included in clinical trials. Once again, several features of ageing hallmarks are assessed but not in an age-related context. Indeed, many of the trials are conducted in patients with lung disorders [201,202,203] or intestinal diseases like irritable bowel disease [204,205]. Furthermore, clinical translation of these compounds remains a challenge mainly due to their volatility and low water solubility and stability. Moreover, pharmacokinetic studies are sparse, and more information on the absorption, metabolism, distribution and elimination of volatile monoterpenes and essential oils is required for safe and effective use in humans. To overcome these issues, encapsulation and targeting strategies have been considered to improve bioavailability and efficacy, as reviewed elsewhere [206]. Furthermore, more large-scale and well-controlled human clinical trials taking into account the heterogeneity of ageing need to be conducted to validate the potential of these compounds and develop innovative anti-ageing strategies.
Overall, as ageing is not a disease per se, strategies that promote healthy ageing and longevity and decrease the incidence and development of ageing-related diseases are in the spotlight. In this context, monoterpenes and/or essential oils emerge as promising compounds with beneficial effects on the drivers of ageing-related diseases. These compounds can also be considered for prevention strategies, thus meeting the new developments in geriatric medicine that is gradually focusing on pre-intervention rather than post-disease treatment.

Author Contributions

Conceptualization, M.Z.; validation, C.S. and L.S.; investigation, M.Z., J.A.-S. and C.S.; writing—original draft preparation, M.Z., J.A.-S. and C.S.; writing—review and editing, M.Z., J.A.-S., C.S. and L.S.; funding acquisition, M.Z. All authors have read and agreed to the published version of the manuscript.

Funding

National Funds via FCT (Foundation for Science and Technology) through the Strategic Projects UIDB/04539/2020 and UIDP/04539/2020 (CIBB) and the Exploratory Project PATCH (022.05810.PTDC) and BPI/La Caixa Foundation and FCT under the project PLANTS4AGEING (PD21-00003).

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are contained within the article.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. World Health Organization Ageing. Available online: https://www.who.int/health-topics/ageing#tab=tab_2 (accessed on 20 December 2023).
  2. Li, Z.; Zhang, Z.; Ren, Y.; Wang, Y.; Fang, J.; Yue, H.; Ma, S.; Guan, F. Aging and age-related diseases: From mechanisms to therapeutic strategies. Biogerontology 2021, 22, 165–187. [Google Scholar] [CrossRef] [PubMed]
  3. Ros, M.; Carrascosa, J.M. Current nutritional and pharmacological anti-aging interventions. Biochim. Biophys. Acta Mol. Basis Dis. 2020, 1866, 165612. [Google Scholar] [CrossRef] [PubMed]
  4. Donati Zeppa, S.; Agostini, D.; Ferrini, F.; Gervasi, M.; Barbieri, E.; Bartolacci, A.; Piccoli, G.; Saltarelli, R.; Sestili, P.; Stocchi, V. Interventions on gut microbiota for healthy aging. Cells 2022, 12, 34. [Google Scholar] [CrossRef] [PubMed]
  5. Guo, J.; Huang, X.; Dou, L.; Yan, M.; Shen, T.; Tang, W.; Li, J. Aging and aging-related diseases: From molecular mechanisms to interventions and treatments. Signal Transduct. Target. Ther. 2022, 7, 391. [Google Scholar] [CrossRef]
  6. López-Otín, C.; Blasco, M.A.; Partridge, L.; Serrano, M.; Kroemer, G. The hallmarks of aging. Cell 2013, 153, 1194–1217. [Google Scholar] [CrossRef] [PubMed]
  7. Schmauck-Medina, T.; Molière, A.; Lautrup, S.; Zhang, J.; Chlopicki, S.; Madsen, H.B.; Cao, S.; Soendenbroe, C.; Mansell, E.; Vestergaard, M.B.; et al. New hallmarks of ageing: A 2022 Copenhagen ageing meeting summary. Aging 2022, 14, 6829–6839. [Google Scholar] [CrossRef]
  8. López-Otín, C.; Blasco, M.A.; Partridge, L.; Serrano, M.; Kroemer, G. Hallmarks of aging: An expanding universe. Cell 2023, 186, 243–278. [Google Scholar] [CrossRef]
  9. Xu, M.; Pirtskhalava, T.; Farr, J.N.; Weigand, B.M.; Palmer, A.K.; Weivoda, M.M.; Inman, C.L.; Ogrodnik, M.B.; Hachfeld, C.M.; Fraser, D.G.; et al. Senolytics improve physical function and increase lifespan in old age. Nat. Med. 2018, 24, 1246–1256. [Google Scholar] [CrossRef]
  10. Baar, M.P.; Brandt, R.M.C.; Putavet, D.A.; Klein, J.D.D.; Derks, K.W.J.; Bourgeois, B.R.M.; Stryeck, S.; Rijksen, Y.; van Willigenburg, H.; Feijtel, D.A.; et al. Targeted apoptosis of senescent cells restores tissue homeostasis in response to chemotoxicity and aging. Cell 2017, 169, 132–147.e16. [Google Scholar] [CrossRef]
  11. Justice, J.N.; Nambiar, A.M.; Tchkonia, T.; LeBrasseur, N.K.; Pascual, R.; Hashmi, S.K.; Prata, L.; Masternak, M.M.; Kritchevsky, S.B.; Musi, N.; et al. Senolytics in idiopathic pulmonary fibrosis: Results from a first-in-human, open-label, pilot study. EBioMedicine 2019, 40, 554–563. [Google Scholar] [CrossRef]
  12. Gao, L.; Liu, X.; Luo, X.; Lou, X.; Li, P.; Li, X.; Liu, X. Antiaging effects of dietary supplements and natural products. Front. Pharmacol. 2023, 14, 1192714. [Google Scholar] [CrossRef] [PubMed]
  13. Potocka, W.; Assy, Z.; Bikker, F.J.; Laine, M.L. Current and potential applications of monoterpenes and their derivatives in oral health care. Molecules 2023, 28, 7178. [Google Scholar] [CrossRef] [PubMed]
  14. ISO 9235:2013; Aromatic Natural Raw Materials—Vocabulary. ISO: Geneva, Switzerland, 2013.
  15. European Directorate for the Quality of Medicines & HealthCare (EDQM). European Pharmacopoeia, 11th ed.; Council of Europe: Strasbourg, France, 2022. [Google Scholar]
  16. European Medicines Agency European Union Monographs and List Entries. Available online: https://www.ema.europa.eu/en/human-regulatory-overview/herbal-medicinal-products/european-union-monographs-and-list-entries (accessed on 20 December 2023).
  17. Zuzarte, M.; Salgueiro, L. Essential Oils Chemistry. In Bioactive Essential Oils and Cancer; Springer International Publishing: Cham, Switzerland, 2015; pp. 19–61. [Google Scholar]
  18. Salakhutdinov, N.F.; Volcho, K.P.; Yarovaya, O.I. Monoterpenes as a renewable source of biologically active compounds. Pure Appl. Chem. 2017, 89, 1105–1117. [Google Scholar] [CrossRef]
  19. Zielińska-Błajet, M.; Feder-Kubis, J. Monoterpenes and their derivatives—Recent development in biological and medical applications. Int. J. Mol. Sci. 2020, 21, 7078. [Google Scholar] [CrossRef]
  20. Fulop, T.; Witkowski, J.M.; Olivieri, F.; Larbi, A. The integration of inflammaging in age-related diseases. Semin. Immunol. 2018, 40, 17–35. [Google Scholar] [CrossRef]
  21. Sierra, F. The emergence of geroscience as an interdisciplinary approach to the enhancement of health span and life span. Cold Spring Harb. Perspect. Med. 2016, 6, a025163. [Google Scholar] [CrossRef]
  22. Gantchev, J.; Ramchatesingh, B.; Berman-Rosa, M.; Sikorski, D.; Raveendra, K.; Amar, L.; Xu, H.H.; Villarreal, A.M.; Ordaz, D.J.G.; Litvinov, I.V. Tools used to assay genomic instability in cancers and cancer meiomitosis. J. Cell Commun. Signal. 2022, 16, 159–177. [Google Scholar] [CrossRef]
  23. Catanzaro, I.; Caradonna, F.; Barbata, G.; Saverini, M.; Mauro, M.; Sciandrello, G. Genomic instability induced by α-pinene in Chinese hamster cell line. Mutagenesis 2012, 27, 463–469. [Google Scholar] [CrossRef]
  24. Mauro, M.; Catanzaro, I.; Naselli, F.; Sciandrello, G.; Caradonna, F. Abnormal mitotic spindle assembly and cytokinesis induced by D-Limonene in cultured mammalian cells. Mutagenesis 2013, 28, 631–635. [Google Scholar] [CrossRef] [PubMed]
  25. Warman, D.J.; Jia, H.; Kato, H. Effects of Thyme (Thymus vulgaris L.) Essential oil on aging-induced brain inflammation and blood telomere attrition in chronologically aged C57BL/6J mice. Antioxidants 2023, 12, 1178. [Google Scholar] [CrossRef] [PubMed]
  26. Cherry, A.D.; Piantadosi, C.A. Regulation of mitochondrial biogenesis and its intersection with inflammatory responses. Antioxid. Redox Signal. 2015, 22, 965–976. [Google Scholar] [CrossRef] [PubMed]
  27. Brand, M.D.; Nicholls, D.G. Assessing mitochondrial dysfunction in cells. Biochem. J. 2011, 435, 297–312. [Google Scholar] [CrossRef] [PubMed]
  28. Alves-Silva, J.M.; Zuzarte, M.; Marques, C.; Viana, S.; Preguiça, I.; Baptista, R.; Ferreira, C.; Cavaleiro, C.; Domingues, N.; Sardão, V.A.; et al. 1,8-Cineole ameliorates right ventricle dysfunction associated with pulmonary arterial hypertension by restoring connexin43 and mitochondrial homeostasis. Pharmacol. Res. 2022, 180, 106151. [Google Scholar] [CrossRef]
  29. Chen, L.; Tao, D.; Yu, F.; Wang, T.; Qi, M.; Xu, S. Cineole regulates Wnt/β-catenin pathway through Nrf2/keap1/ROS to inhibit bisphenol A-induced apoptosis, autophagy inhibition and immunosuppression of grass carp hepatocytes. Fish Shellfish Immunol. 2022, 131, 30–41. [Google Scholar] [CrossRef]
  30. Sun, W.; Liu, H.; Zhu, H.; Gao, M.; Xu, S. Eucalyptol antagonized the apoptosis and immune dysfunction of grass carp hepatocytes induced by tetrabromobisphenol A by regulating ROS/ASK1/JNK pathway. Environ. Toxicol. 2023, 38, 820–832. [Google Scholar] [CrossRef]
  31. Liu, H.; Sun, W.; Zhu, H.; Guo, J.; Liu, M.; Xu, S. Eucalyptol relieves the toxicity of diisobutyl phthalate in Ctenopharyngodon idellus kidney cells through Keap1/Nrf2/HO-1 pathway: Apoptosis-autophagy crosstalk and immunoregulation. Fish Shellfish Immunol. 2022, 130, 490–500. [Google Scholar] [CrossRef] [PubMed]
  32. Baek, S.; Kim, J.; Moon, B.S.; Park, S.M.; Jung, D.E.; Kang, S.Y.; Lee, S.J.; Oh, S.J.; Kwon, S.H.; Nam, M.H.; et al. Camphene attenuates skeletal muscle atrophy by regulating oxidative stress and lipid metabolism in rats. Nutrients 2020, 12, 3731. [Google Scholar] [CrossRef]
  33. Chenet, A.L.; Duarte, A.R.; de Almeida, F.J.S.; Andrade, C.M.B.; de Oliveira, M.R. Carvacrol depends on heme oxygenase-1 (HO-1) to exert antioxidant, anti-inflammatory, and mitochondria-related protection in the human neuroblastoma SH-SY5Y cells line exposed to hydrogen peroxide. Neurochem. Res. 2019, 44, 884–896. [Google Scholar] [CrossRef]
  34. Rekha, K.R.; Inmozhi Sivakamasundari, R. Geraniol protects against the protein and oxidative stress induced by rotenone in an in vitro model of Parkinson’s Disease. Neurochem. Res. 2018, 43, 1947–1962. [Google Scholar] [CrossRef]
  35. Kumar, K.J.S.; Vani, M.G.; Wang, S. Limonene protects human skin keratinocytes against UVB-induced photodamage and photoaging by activating the Nrf2-dependent antioxidant defense system. Environ. Toxicol. 2022, 37, 2897–2909. [Google Scholar] [CrossRef]
  36. Suh, K.S.; Chon, S.; Choi, E.M. Limonene protects osteoblasts against methylglyoxal-derived adduct formation by regulating glyoxalase, oxidative stress, and mitochondrial function. Chem. Biol. Interact. 2017, 278, 15–21. [Google Scholar] [CrossRef]
  37. Sabogal-Guáqueta, A.M.; Hobbie, F.; Keerthi, A.; Oun, A.; Kortholt, A.; Boddeke, E.; Dolga, A. Linalool attenuates oxidative stress and mitochondrial dysfunction mediated by glutamate and NMDA toxicity. Biomed. Pharmacother. 2019, 118, 109295. [Google Scholar] [CrossRef] [PubMed]
  38. Kumar, S.; Acharya, T.K.; Halder, R.R.; Mahapatra, P.; Chang, Y.-T.; Goswami, C. Menthol causes mitochondrial Ca2+-influx, affects structure-function relationship and cools mitochondria. Life Sci. 2023, 331, 122032. [Google Scholar] [CrossRef] [PubMed]
  39. Hwang, E.; Ngo, H.T.T.; Park, B.; Seo, S.-A.; Yang, J.-E.; Yi, T.-H. Myrcene, an aromatic volatile compound, ameliorates human skin extrinsic aging via regulation of MMPs production. Am. J. Chin. Med. 2017, 45, 1113–1124. [Google Scholar] [CrossRef]
  40. Ahmed, S.; Panda, S.R.; Kwatra, M.; Sahu, B.D.; Naidu, V. Perillyl alcohol attenuates NLRP3 inflammasome activation and rescues dopaminergic neurons in experimental in vitro and in vivo models of Parkinson’s Disease. ACS Chem. Neurosci. 2022, 13, 53–68. [Google Scholar] [CrossRef]
  41. Anis, E.; Zafeer, M.F.; Firdaus, F.; Islam, S.N.; Fatima, M.; Mobarak Hossain, M. Evaluation of phytomedicinal potential of perillyl alcohol in an in vitro Parkinson’s Disease model. Drug Dev. Res. 2018, 79, 218–224. [Google Scholar] [CrossRef]
  42. Zafeer, M.F.; Firdaus, F.; Ahmad, F.; Ullah, R.; Anis, E.; Waseem, M.; Ali, A.; Hossain, M.M. Perillyl alcohol alleviates amyloid-β peptides-induced mitochondrial dysfunction and cytotoxicity in SH-SY5Y cells. Int. J. Biol. Macromol. 2018, 109, 1029–1038. [Google Scholar] [CrossRef] [PubMed]
  43. Arruri, V.K.; Gundu, C.; Kalvala, A.K.; Sherkhane, B.; Khatri, D.K.; Singh, S.B. Carvacrol abates NLRP3 inflammasome activation by augmenting Keap1/Nrf-2/p62 directed autophagy and mitochondrial quality control in neuropathic pain. Nutr. Neurosci. 2022, 25, 1731–1746. [Google Scholar] [CrossRef]
  44. Prasad, S.N.; Muralidhara. Neuroprotective effect of geraniol and curcumin in an acrylamide model of neurotoxicity in Drosophila melanogaster: Relevance to neuropathy. J. Insect Physiol. 2014, 60, 7–16. [Google Scholar] [CrossRef]
  45. Prasad, S.N.; Muralidhara. Protective effects of geraniol (a monoterpene) in a diabetic neuropathy rat model: Attenuation of behavioral impairments and biochemical perturbations. J. Neurosci. Res. 2014, 92, 1205–1216. [Google Scholar] [CrossRef]
  46. Prasad, S.N.; Muralidhara. Mitigation of acrylamide-induced behavioral deficits, oxidative impairments and neurotoxicity by oral supplements of geraniol (a monoterpene) in a rat model. Chem. Biol. Interact. 2014, 223, 27–37. [Google Scholar] [CrossRef] [PubMed]
  47. Eddin, L.B.; Azimullah, S.; Jha, N.K.; Nagoor Meeran, M.F.; Beiram, R.; Ojha, S. Limonene, a monoterpene, mitigates rotenone-induced dopaminergic neurodegeneration by modulating neuroinflammation, Hippo signaling and apoptosis in rats. Int. J. Mol. Sci. 2023, 24, 5222. [Google Scholar] [CrossRef] [PubMed]
  48. Anis, E.; Zafeer, M.F.; Firdaus, F.; Islam, S.N.; Khan, A.A.; Hossain, M.M. Perillyl alcohol mitigates behavioural changes and limits cell death and mitochondrial changes in unilateral 6-OHDA lesion model of Parkinson’s Disease through alleviation of oxidative stress. Neurotox. Res. 2020, 38, 461–477. [Google Scholar] [CrossRef] [PubMed]
  49. Morselli, M.B.; Baldissera, M.D.; Souza, C.F.; Reis, J.H.; Baldisserotto, B.; Sousa, A.A.; Zimmer, F.; Lopes, D.L.A.; Petrolli, T.G.; Da Silva, A.S. Effects of thymol supplementation on performance, mortality and branchial energetic metabolism in grass carp experimentally infected by Aeromonas hydrophila. Microb. Pathog. 2020, 139, 103915. [Google Scholar] [CrossRef] [PubMed]
  50. Nagoor Meeran, M.F.; Jagadeesh, G.S.; Selvaraj, P. Thymol, a dietary monoterpene phenol abrogates mitochondrial dysfunction in β-adrenergic agonist induced myocardial infarcted rats by inhibiting oxidative stress. Chem. Biol. Interact. 2016, 244, 159–168. [Google Scholar] [CrossRef] [PubMed]
  51. Zorova, L.D.; Popkov, V.A.; Plotnikov, E.Y.; Silachev, D.N.; Pevzner, I.B.; Jankauskas, S.S.; Babenko, V.A.; Zorov, S.D.; Balakireva, A.V.; Juhaszova, M.; et al. Mitochondrial membrane potential. Anal. Biochem. 2018, 552, 50–59. [Google Scholar] [CrossRef]
  52. Yu, G.J.; Choi, I.-W.; Kim, G.-Y.; Hwang, H.J.; Kim, B.W.; Kim, C.M.; Kim, W.-J.; Yoo, Y.H.; Choi, Y.H. Induction of reactive oxygen species–mediated apoptosis by purified Schisandrae semen essential oil in human leukemia U937 cells through activation of the caspase cascades and nuclear relocation of mitochondrial apoptogenic factors. Nutr. Res. 2015, 35, 910–920. [Google Scholar] [CrossRef]
  53. Li, N.; Zhang, Q.; Jia, Z.; Yang, X.; Zhang, H.; Luo, H. Volatile oil from Alpinia officinarum promotes lung cancer regression in vitro and in vivo. Food Funct. 2018, 9, 4998–5006. [Google Scholar] [CrossRef]
  54. Chang, W.; Cheng, F.; Wang, S.; Chou, S.; Shih, Y. Cinnamomum cassia essential oil and its major constituent cinnamaldehyde induced cell cycle arrest and apoptosis in human oral squamous cell carcinoma HSC-3 cells. Environ. Toxicol. 2017, 32, 456–468. [Google Scholar] [CrossRef]
  55. Cavalcanti, B.C.; Magalhães, I.L.; Rocha, D.D.; Stefânio Barreto, F.; de Andrade Neto, J.B.; Magalhães, H.I.F.; dos Santos, C.C.; de Moraes, M.O. In vitro evaluation of cytotoxic potential of essential oil extracted from leaves of Croton heliotropiifolius Kunth in human tumor cells. J. Toxicol. Environ. Health Part A 2024, 87, 91–107. [Google Scholar] [CrossRef]
  56. Khan, I.; Bahuguna, A.; Bhardwaj, M.; Pal Khaket, T.; Kang, S.C. Carvacrol nanoemulsion evokes cell cycle arrest, apoptosis induction and autophagy inhibition in doxorubicin resistant-A549 cell line. Artif. Cells Nanomed. Biotechnol. 2018, 46, 664–675. [Google Scholar] [CrossRef]
  57. Zhang, G.; He, J.; Ye, X.; Zhu, J.; Hu, X.; Shen, M.; Ma, Y.; Mao, Z.; Song, H.; Chen, F. β-Thujaplicin induces autophagic cell death, apoptosis, and cell cycle arrest through ROS-mediated Akt and p38/ERK MAPK signaling in human hepatocellular carcinoma. Cell Death Dis. 2019, 10, 255. [Google Scholar] [CrossRef] [PubMed]
  58. Shahina, Z.; Al Homsi, R.; Price, J.D.W.; Whiteway, M.; Sultana, T.; Dahms, T.E.S. Rosemary essential oil and its components 1,8-cineole and α-pinene induce ROS-dependent lethality and ROS-independent virulence inhibition in Candida albicans. PLoS ONE 2022, 17, e0277097. [Google Scholar] [CrossRef]
  59. Zhang, C.; Zhao, J.; Famous, E.; Pan, S.; Peng, X.; Tian, J. Antioxidant, hepatoprotective and antifungal activities of black pepper (Piper nigrum L.) essential oil. Food Chem. 2021, 346, 128845. [Google Scholar] [CrossRef] [PubMed]
  60. Tian, J.; Ban, X.; Zeng, H.; He, J.; Chen, Y.; Wang, Y. The mechanism of antifungal action of essential oil from dill (Anethum graveolens L.) on Aspergillus flavus. PLoS ONE 2012, 7, e30147. [Google Scholar] [CrossRef] [PubMed]
  61. Chen, Y.; Zeng, H.; Tian, J.; Ban, X.; Ma, B.; Wang, Y. Antifungal mechanism of essential oil from Anethum graveolens seeds against Candida albicans. J. Med. Microbiol. 2013, 62, 1175–1183. [Google Scholar] [CrossRef]
  62. de Carvalho, N.R.; Rodrigues, N.R.; Macedo, G.E.; Bristot, I.J.; Boligon, A.A.; de Campos, M.M.; Cunha, F.A.B.; Coutinho, H.D.; Klamt, F.; Merritt, T.J.S.; et al. Eugenia uniflora leaf essential oil promotes mitochondrial dysfunction in Drosophila melanogaster through the inhibition of oxidative phosphorylation. Toxicol. Res. 2017, 6, 526–534. [Google Scholar] [CrossRef]
  63. Baldissera, M.D.; Souza, C.F.; Baldisserotto, B. Melaleuca alternifolia essential oil prevents bioenergetics dysfunction in spleen of silver catfish naturally infected with Ichthyophthirius multifiliis. Microb. Pathog. 2018, 123, 47–51. [Google Scholar] [CrossRef]
  64. Hernandez-Segura, A.; Nehme, J.; Demaria, M. Hallmarks of cellular senescence. Trends Cell Biol. 2018, 28, 436–453. [Google Scholar] [CrossRef]
  65. Calcinotto, A.; Kohli, J.; Zagato, E.; Pellegrini, L.; Demaria, M.; Alimonti, A. Cellular senescence: Aging, cancer, and injury. Physiol. Rev. 2019, 99, 1047–1078. [Google Scholar] [CrossRef]
  66. Freund, A.; Orjalo, A.V.; Desprez, P.-Y.; Campisi, J. Inflammatory networks during cellular senescence: Causes and consequences. Trends Mol. Med. 2010, 16, 238–246. [Google Scholar] [CrossRef]
  67. Soto-Gamez, A.; Demaria, M. Therapeutic interventions for aging: The case of cellular senescence. Drug Discov. Today 2017, 22, 786–795. [Google Scholar] [CrossRef]
  68. González-Gualda, E.; Baker, A.G.; Fruk, L.; Muñoz-Espín, D. A guide to assessing cellular senescence in vitro and in vivo. FEBS J. 2021, 288, 56–80. [Google Scholar] [CrossRef]
  69. Tran, T.A.; Ho, M.T.; Song, Y.W.; Cho, M.; Cho, S.K. Camphor induces proliferative and anti-senescence activities in human primary dermal fibroblasts and inhibits UV-induced wrinkle formation in mouse skin. Phyther. Res. 2015, 29, 1917–1925. [Google Scholar] [CrossRef] [PubMed]
  70. Cherng, J.Y.; Chen, L.Y.; Shih, M.F. Preventive Effects of β-thujaplicin against UVB-induced MMP-1 and MMP-3 mRNA expressions in skin fibroblasts. Am. J. Chin. Med. 2012, 40, 387–398. [Google Scholar] [CrossRef] [PubMed]
  71. Karthikeyan, R.; Kanimozhi, G.; Prasad, N.R.; Agilan, B.; Ganesan, M.; Srithar, G. Alpha pinene modulates UVA-induced oxidative stress, DNA damage and apoptosis in human skin epidermal keratinocytes. Life Sci. 2018, 212, 150–158. [Google Scholar] [CrossRef] [PubMed]
  72. Karthikeyan, R.; Kanimozhi, G.; Madahavan, N.R.; Agilan, B.; Ganesan, M.; Prasad, N.R.; Rathinaraj, P. Alpha-pinene attenuates UVA-induced photoaging through inhibition of matrix metalloproteinases expression in mouse skin. Life Sci. 2019, 217, 110–118. [Google Scholar] [CrossRef] [PubMed]
  73. Lagoumtzi, S.M.; Chondrogianni, N. Senolytics and senomorphics: Natural and synthetic therapeutics in the treatment of aging and chronic diseases. Free Radic. Biol. Med. 2021, 171, 169–190. [Google Scholar] [CrossRef] [PubMed]
  74. Smith, M.L.; Ford, J.M.; Hollander, M.C.; Bortnick, R.A.; Amundson, S.A.; Seo, Y.R.; Deng, C.-X.; Hanawalt, P.C.; Fornace, A.J. p53-Mediated DNA repair responses to UV radiation: Studies of mouse cells lacking p53, p21, and/or gadd45 genes. Mol. Cell. Biol. 2000, 20, 3705–3714. [Google Scholar] [CrossRef] [PubMed]
  75. Mihara, M.; Erster, S.; Zaika, A.; Petrenko, O.; Chittenden, T.; Pancoska, P.; Moll, U.M. p53 has a direct apoptogenic role at the mitochondria. Mol. Cell 2003, 11, 577–590. [Google Scholar] [CrossRef] [PubMed]
  76. Yang, S.-C.; Chen, H.-Y.; Chuang, W.-L.; Wang, H.-C.; Hsieh, C.-P.; Huang, Y.-F. Different cell responses to hinokitiol treatment result in senescence or apoptosis in Hhuman osteosarcoma cell lines. Int. J. Mol. Sci. 2022, 23, 1632. [Google Scholar] [CrossRef]
  77. Li, L.-H.; Wu, P.; Lee, J.-Y.; Li, P.-R.; Hsieh, W.-Y.; Ho, C.-C.; Ho, C.-L.; Chen, W.-J.; Wang, C.-C.; Yen, M.-Y.; et al. Hinokitiol induces DNA damage and autophagy followed by cell cycle arrest and senescence in gefitinib-resistant lung adenocarcinoma cells. PLoS ONE 2014, 9, e104203. [Google Scholar] [CrossRef]
  78. Rodenak-Kladniew, B.; Castro, A.; Stärkel, P.; Galle, M.; Crespo, R. 1,8-Cineole promotes G0/G1 cell cycle arrest and oxidative stress-induced senescence in HepG2 cells and sensitizes cells to anti-senescence drugs. Life Sci. 2020, 243, 117271. [Google Scholar] [CrossRef]
  79. Jeon, J.-H. Geraniol induces cooperative interaction of apoptosis and autophagy to elicit cell death in PC-3 prostate cancer cells. Int. J. Oncol. 2011, 40, 1683–1690. [Google Scholar] [CrossRef]
  80. Naksawat, M.; Norkaew, C.; Charoensedtasin, K.; Roytrakul, S.; Tanyong, D. Anti-leukemic effect of menthol, a peppermint compound, on induction of apoptosis and autophagy. PeerJ 2023, 11, e15049. [Google Scholar] [CrossRef] [PubMed]
  81. Hsieh, L.-C.; Hsieh, S.-L.; Chen, C.-T.; Chung, J.-G.; Wang, J.-J.; Wu, C.-C. Induction of α-Pphellandrene on autophagy in human liver tumor cells. Am. J. Chin. Med. 2015, 43, 121–136. [Google Scholar] [CrossRef] [PubMed]
  82. Alves-Silva, J.M.; Pedreiro, S.; Cavaleiro, C.; Cruz, M.T.; Figueirinha, A.; Salgueiro, L. Effect of Thymbra capitata (L.) Cav. on inflammation, senescence and cell migration. Nutrients 2023, 15, 1930. [Google Scholar] [CrossRef] [PubMed]
  83. Alves-Silva, J.M.; Gonçalves, M.J.; Silva, A.; Cavaleiro, C.; Cruz, M.T.; Salgueiro, L. Chemical profile, anti-microbial and anti-inflammaging activities of Santolina rosmarinifolia L. essential oil from Portugal. Antibiotics 2023, 12, 179. [Google Scholar] [CrossRef] [PubMed]
  84. Alves-Silva, J.M.; Maccioni, D.; Cocco, E.; Gonçalves, M.J.; Porcedda, S.; Piras, A.; Cruz, M.T.; Salgueiro, L.; Maxia, A. Advances in the phytochemical characterisation and bioactivities of Salvia aurea L. Essential Oil. Plants 2023, 12, 1247. [Google Scholar] [CrossRef] [PubMed]
  85. Alves-Silva, J.M.; Moreira, P.; Cavaleiro, C.; Pereira, C.; Cruz, M.T.; Salgueiro, L. Effect of Ferulago lutea (Poir.) Grande essential oil on molecular hallmarks of skin aging. Plants 2023, 12, 3741. [Google Scholar] [CrossRef] [PubMed]
  86. Moreira, P.; Sousa, F.J.; Matos, P.; Brites, G.S.; Gonçalves, M.J.; Cavaleiro, C.; Figueirinha, A.; Salgueiro, L.; Batista, M.T.; Branco, P.C.; et al. Chemical composition and effect against skin alterations of bioactive extracts obtained by the hydrodistillation of Eucalyptus globulus Leaves. Pharmaceutics 2022, 14, 561. [Google Scholar] [CrossRef]
  87. Ledrhem, M.; Nakamura, M.; Obitsu, M.; Hirae, K.; Kameyama, J.; Bouamama, H.; Gadhi, C.; Katakura, Y. Essential oils derived from Cistus species activate mitochondria by inducing SIRT1 expression in human keratinocytes, leading to senescence inhibition. Molecules 2022, 27, 2053. [Google Scholar] [CrossRef]
  88. Salsabila, D.; Wardani, R.; Hasanah, N.; Tafrihani, A.; Zulfin, U.; Ikawati, M.; Meiyanto, E. Cytoprotective properties of citronella oil (Cymbopogon nardus (L.) Rendl.) and lemongrass oil (Cymbopogon citratus (DC.) Stapf) through attenuation of senescent-induced chemotherapeutic agent doxorubicin on Vero and NIH-3T3 Cells. Asian Pac. J. Cancer Prev. 2023, 24, 1667–1675. [Google Scholar] [CrossRef]
  89. Liao, J.; Fu, L.; Tai, S.; Xu, Y.; Wang, S.; Guo, L.; Guo, D.; Du, Y.; He, J.; Yang, H.; et al. Essential oil from Fructus Alpiniae zerumbet ameliorates vascular endothelial cell senescence in diabetes by regulating PPAR-γ signalling: A 4D label-free quantitative proteomics and network pharmacology study. J. Ethnopharmacol. 2024, 321, 117550. [Google Scholar] [CrossRef]
  90. Galluzzi, L.; Baehrecke, E.H.; Ballabio, A.; Boya, P.; Bravo-San Pedro, J.M.; Cecconi, F.; Choi, A.M.; Chu, C.T.; Codogno, P.; Colombo, M.I.; et al. Molecular definitions of autophagy and related processes. EMBO J. 2017, 36, 1811–1836. [Google Scholar] [CrossRef]
  91. Netea-Maier, R.T.; Plantinga, T.S.; van de Veerdonk, F.L.; Smit, J.W.; Netea, M.G. Modulation of inflammation by autophagy: Consequences for human disease. Autophagy 2016, 12, 245–260. [Google Scholar] [CrossRef] [PubMed]
  92. Qian, M.; Fang, X.; Wang, X. Autophagy and inflammation. Clin. Transl. Med. 2017, 6, 24. [Google Scholar] [CrossRef] [PubMed]
  93. Leidal, A.M.; Levine, B.; Debnath, J. Autophagy and the cell biology of age-related disease. Nat. Cell Biol. 2018, 20, 1338–1348. [Google Scholar] [CrossRef] [PubMed]
  94. Zhang, Z.; Singh, R.; Aschner, M. Methods for the detection of autophagy in mammalian cells. Curr. Protoc. Toxicol. 2016, 69, 20.12.1–20.12.26. [Google Scholar] [CrossRef]
  95. Joshi, S.; Kundu, S.; Priya, V.V.; Kulhari, U.; Mugale, M.N.; Sahu, B.D. Anti-inflammatory activity of carvacrol protects the heart from lipopolysaccharide-induced cardiac dysfunction by inhibiting pyroptosis via NLRP3/Caspase1/Gasdermin D signaling axis. Life Sci. 2023, 324, 121743. [Google Scholar] [CrossRef] [PubMed]
  96. Chen, M.; Hu, Q.; Wang, S.; Tao, L.; Hu, X.; Shen, X. 1,8-Cineole ameliorates endothelial injury and hypertension induced by L-NAME through regulation of autophagy via PI3K/mTOR signaling pathway. Eur. J. Pharmacol. 2023, 954, 175863. [Google Scholar] [CrossRef] [PubMed]
  97. Reis, R.; Orak, D.; Yilmaz, D.; Cimen, H.; Sipahi, H. Modulation of cigarette smoke extract-induced human bronchial epithelial damage by eucalyptol and curcumin. Hum. Exp. Toxicol. 2021, 40, 1445–1462. [Google Scholar] [CrossRef] [PubMed]
  98. Moon, J.-H.; Lee, J.-H.; Lee, Y.-J.; Park, S.-Y. Hinokitiol protects primary neuron cells against prion peptide-induced toxicity via autophagy flux regulated by hypoxia inducing factor-1. Oncotarget 2016, 7, 29944–29957. [Google Scholar] [CrossRef] [PubMed]
  99. Xiao, H.; Liang, S.; Cai, Q.; Liu, J.; Jin, L.; Yang, Z.; Chen, X. Hinokitiol protects cardiomyocyte from oxidative damage by inhibiting GSK3β-mediated autophagy. Oxid. Med. Cell. Longev. 2022, 2022, 2700000. [Google Scholar] [CrossRef] [PubMed]
  100. Liu, S.; Guo, W.; Jia, Y.; Ye, B.; Liu, S.; Fu, S.; Liu, J.; Hu, G. Menthol targeting AMPK alleviates the inflammatory response of bovine mammary epithelial cells and restores the synthesis of milk fat and milk protein. Front. Immunol. 2021, 12, 782989. [Google Scholar] [CrossRef] [PubMed]
  101. Guo, C.; Zheng, L.; Chen, S.; Liang, X.; Song, X.; Wang, Y.; Hua, B.; Qiu, L. Thymol ameliorates ethanol-induced hepatotoxicity via regulating metabolism and autophagy. Chem. Biol. Interact. 2023, 370, 110308. [Google Scholar] [CrossRef]
  102. Yu, B.; Ruan, M.; Liang, T.; Huang, S.-W.; Yu, Y.; Cheng, H.-B.; Shen, X.-C. The synergic effect of tetramethylpyrazine phosphate and borneol for protecting against ischemia injury in cortex and hippocampus regions by modulating apoptosis and autophagy. J. Mol. Neurosci. 2017, 63, 70–83. [Google Scholar] [CrossRef]
  103. Jayaraj, R.L.; Azimullah, S.; Parekh, K.A.; Ojha, S.K.; Beiram, R. Effect of citronellol on oxidative stress, neuroinflammation and autophagy pathways in an in vivo model of Parkinson’s disease. Heliyon 2022, 8, e11434. [Google Scholar] [CrossRef]
  104. Younis, N.S.; Abduldaium, M.S.; Mohamed, M.E. Protective Effect of geraniol on oxidative, inflammatory and apoptotic alterations in isoproterenol-induced cardiotoxicity: Role of the Keap1/Nrf2/HO-1 and PI3K/Akt/mTOR pathways. Antioxidants 2020, 9, 977. [Google Scholar] [CrossRef]
  105. Azimullah, S.; Jayaraj, R.L.; Meeran, M.F.N.; Jalal, F.Y.; Adem, A.; Ojha, S.; Beiram, R. Myrcene salvages rotenone-induced loss of dopaminergic neurons by inhibiting oxidative stress, inflammation, apoptosis, and autophagy. Molecules 2023, 28, 685. [Google Scholar] [CrossRef]
  106. Dou, X.; Yan, D.; Liu, S.; Gao, L.; Shan, A. Thymol alleviates LPS-induced liver inflammation and apoptosis by inhibiting NLRP3 inflammasome activation and the AMPK-mTOR-autophagy pathway. Nutrients 2022, 14, 2809. [Google Scholar] [CrossRef]
  107. Wang, C.-C.; Chen, B.-K.; Chen, P.-H.; Chen, L.-C. Hinokitiol induces cell death and inhibits epidermal growth factor-induced cell migration and signaling pathways in human cervical adenocarcinoma. Taiwan J. Obstet. Gynecol. 2020, 59, 698–705. [Google Scholar] [CrossRef]
  108. Pudełek, M.; Catapano, J.; Kochanowski, P.; Mrowiec, K.; Janik-Olchawa, N.; Czyż, J.; Ryszawy, D. Therapeutic potential of monoterpene α-thujone, the main compound of Thuja occidentalis L. essential oil, against malignant glioblastoma multiforme cells in vitro. Fitoterapia 2019, 134, 172–181. [Google Scholar] [CrossRef]
  109. Banjerdpongchai, R.; Khaw-on, P. Terpinen-4-ol induces autophagic and apoptotic cell death in human leukemic HL-60 Cells. Asian Pac. J. Cancer Prev. 2013, 14, 7537–7542. [Google Scholar] [CrossRef]
  110. Athamneh, K.; Alneyadi, A.; Alsamri, H.; Alrashedi, A.; Palakott, A.; El-Tarabily, K.A.; Eid, A.H.; Al Dhaheri, Y.; Iratni, R. Origanum majorana essential oil triggers p38 MAPK-mediated protective autophagy, apoptosis, and caspase-dependent cleavage of P70S6K in colorectal cancer cells. Biomolecules 2020, 10, 412. [Google Scholar] [CrossRef]
  111. Zhao, J.; Ding, K.; Hou, M.; Li, Y.; Hou, X.; Dai, W.; Li, Z.; Zhao, J.; Liu, W.; Bai, Z. Schisandra chinensis essential oil attenuates acetaminophen-induced liver injury through alleviating oxidative stress and activating autophagy. Pharm. Biol. 2022, 60, 958–967. [Google Scholar] [CrossRef]
  112. Chen, X.; Liao, D.; Sun, M.; Cui, X.; Wang, H. Essential oil of Acorus tatarinowii Schott ameliorates Aβ-induced toxicity in Caenorhabditis elegans through an autophagy pathway. Oxid. Med. Cell. Longev. 2020, 2020, 3515609. [Google Scholar] [CrossRef] [PubMed]
  113. Antonelli, M.; Kushner, I. It’s time to redefine inflammation. FASEB J. 2017, 31, 1787–1791. [Google Scholar] [CrossRef] [PubMed]
  114. Medzhitov, R. Origin and physiological roles of inflammation. Nature 2008, 454, 428–435. [Google Scholar] [CrossRef]
  115. Nita, M.; Grzybowski, A.; Ascaso, F.J.; Huerva, V. Age-related macular degeneration in the aspect of chronic low-grade inflammation (Pathophysiological paraInflammation). Mediators Inflamm. 2014, 2014, 930671. [Google Scholar] [CrossRef] [PubMed]
  116. Robinson, W.H.; Lepus, C.M.; Wang, Q.; Raghu, H.; Mao, R.; Lindstrom, T.M.; Sokolove, J. Low-grade inflammation as a key mediator of the pathogenesis of osteoarthritis. Nat. Rev. Rheumatol. 2016, 12, 580–592. [Google Scholar] [CrossRef]
  117. Baechle, J.J.; Chen, N.; Makhijani, P.; Winer, S.; Furman, D.; Winer, D.A. Chronic inflammation and the hallmarks of aging. Mol. Metab. 2023, 74, 101755. [Google Scholar] [CrossRef]
  118. Zhao, W.; Deng, C.; Han, Q.; Xu, H.; Chen, Y. Carvacrol may alleviate vascular inflammation in diabetic db/db mice. Int. J. Mol. Med. 2020, 46, 977–988. [Google Scholar] [CrossRef]
  119. Sousa, C.; Neves, B.M.; Leitão, A.J.; Mendes, A.F. Elucidation of the mechanism underlying the anti-inflammatory properties of (S)-(+)-Carvone identifies a novel class of Sirtuin-1 activators in a murine macrophage cell line. Biomedicines 2021, 9, 777. [Google Scholar] [CrossRef] [PubMed]
  120. Sousa, C.; Neves, B.M.; Leitão, A.J.; Mendes, A.F. Molecular mechanisms underlying the anti-inflammatory properties of (R)-(-)-Carvone: Potential roles of JNK1, Nrf2 and NF-κB. Pharmaceutics 2023, 15, 249. [Google Scholar] [CrossRef] [PubMed]
  121. Yin, C.; Liu, B.; Wang, P.; Li, X.; Li, Y.; Zheng, X.; Tai, Y.; Wang, C.; Liu, B. Eucalyptol alleviates inflammation and pain responses in a mouse model of gout arthritis. Br. J. Pharmacol. 2020, 177, 2042–2057. [Google Scholar] [CrossRef] [PubMed]
  122. Li, Q.; Yu, C.; Chen, Y.; Liu, S.; Azevedo, P.; Gong, J.; Karmin, O.; Yang, C. Citral alleviates peptidoglycan-induced inflammation and disruption of barrier functions in porcine intestinal epithelial cells. J. Cell. Physiol. 2022, 237, 1768–1779. [Google Scholar] [CrossRef] [PubMed]
  123. Oliveira, H.B.M.; das Neves Selis, N.; Brito, T.L.S.; Sampaio, B.A.; de Souza Bittencourt, R.; Oliveira, C.N.T.; Júnior, M.N.S.; Almeida, C.F.; Almeida, P.P.; Campos, G.B.; et al. Citral modulates human monocyte responses to Staphylococcus aureus infection. Sci. Rep. 2021, 11, 22029. [Google Scholar] [CrossRef] [PubMed]
  124. Horváth, G.; Horváth, A.; Reichert, G.; Böszörményi, A.; Sipos, K.; Pandur, E. Three chemotypes of thyme (Thymus vulgaris L.) essential oil and their main compounds affect differently the IL-6 and TNFα cytokine secretions of BV-2 microglia by modulating the NF-κB and C/EBPβ signalling pathways. BMC Complement. Med. Ther. 2021, 21, 148. [Google Scholar] [CrossRef] [PubMed]
  125. Ma, J.; Xu, Y.; Zhang, M.; Li, Y. Geraniol ameliorates acute liver failure induced by lipopolysaccharide/D-galactosamine via regulating macrophage polarization and NLRP3 inflammasome activation by PPAR-γ methylation Geraniol alleviates acute liver failure. Biochem. Pharmacol. 2023, 210, 115467. [Google Scholar] [CrossRef] [PubMed]
  126. Ammar, R.B.; Mohamed, M.E.; Alfwuaires, M.; Abdulaziz Alamer, S.; Bani Ismail, M.; Veeraraghavan, V.P.; Sekar, A.K.; Ksouri, R.; Rajendran, P. Anti-inflammatory activity of geraniol isolated from lemon grass on Ox-LDL-stimulated endothelial cells by upregulation of Heme Oxygenase-1 via PI3K/Akt and Nrf-2 signaling pathways. Nutrients 2022, 14, 4817. [Google Scholar] [CrossRef]
  127. Hiyoshi, T.; Domon, H.; Maekawa, T.; Yonezawa, D.; Kunitomo, E.; Tabeta, K.; Terao, Y. Protective effect of hinokitiol against periodontal bone loss in ligature-induced experimental periodontitis in mice. Arch. Oral Biol. 2020, 112, 104679. [Google Scholar] [CrossRef]
  128. Du, J.; Liu, D.; Zhang, X.; Zhou, A.; Su, Y.; He, D.; Fu, S.; Gao, F. Menthol protects dopaminergic neurons against inflammation-mediated damage in lipopolysaccharide (LPS)-Evoked model of Parkinson’s disease. Int. Immunopharmacol. 2020, 85, 106679. [Google Scholar] [CrossRef]
  129. Sudha Yalamarthi, S.; Puppala, E.R.; Abubakar, M.; Saha, P.; Challa, V.S.; NP, S.; USN, M.; Gangasani, J.K.; Naidu, V.G.M. Perillyl alcohol inhibits keratinocyte proliferation and attenuates imiquimod-induced psoriasis like skin-inflammation by modulating NF-κB and STAT3 signaling pathways. Int. Immunopharmacol. 2022, 103, 108436. [Google Scholar] [CrossRef]
  130. Puppala, E.R.; Aochenlar, S.L.; Shantanu, P.; Ahmed, S.; Jannu, A.K.; Jala, A.; Yalamarthi, S.S.; Borkar, R.M.; Tripathi, D.M.; Naidu, V.G.M. Perillyl alcohol attenuates chronic restraint stress aggravated dextran sulfate sodium-induced ulcerative colitis by modulating TLR4/NF-κB and JAK2/STAT3 signaling pathways. Phytomedicine 2022, 106, 154415. [Google Scholar] [CrossRef]
  131. Fan, Y.; Li, C.; Peng, X.; Jiang, N.; Hu, L.; Gu, L.; Zhu, G.; Zhao, G.; Lin, J. Perillaldehyde ameliorates Aspergillus fumigatus keratitis by activating the Nrf2/HO-1 signaling Ppathway and inhibiting dectin-1-mediated inflammation. Investig. Opthalmology Vis. Sci. 2020, 61, 51. [Google Scholar] [CrossRef] [PubMed]
  132. Chen, J.; Li, D.-L.; Xie, L.-N.; Ma, Y.; Wu, P.-P.; Li, C.; Liu, W.-F.; Zhang, K.; Zhou, R.-P.; Xu, X.-T.; et al. Synergistic anti-inflammatory effects of silibinin and thymol combination on LPS-induced RAW264.7 cells by inhibition of NF-κB and MAPK activation. Phytomedicine 2020, 78, 153309. [Google Scholar] [CrossRef] [PubMed]
  133. Bansod, S.; Chilvery, S.; Saifi, M.A.; Das, T.J.; Tag, H.; Godugu, C. Borneol protects against cerulein-induced oxidative stress and inflammation in acute pancreatitis mice model. Environ. Toxicol. 2021, 36, 530–539. [Google Scholar] [CrossRef] [PubMed]
  134. Riaz, M.; Al Kury, L.T.; Atzaz, N.; Alattar, A.; Alshaman, R.; Shah, F.A.; Li, S. Carvacrol alleviates hyperuricemia-induced oxidative stress and Iinflammation by modulating the NLRP3/NF-κB pathway. Drug Des. Devel. Ther. 2022, 16, 1159–1170. [Google Scholar] [CrossRef] [PubMed]
  135. Yıldız, M.O.; Çelik, H.; Caglayan, C.; Genç, A.; Doğan, T.; Satıcı, E. Neuroprotective effects of carvacrol against cadmium-induced neurotoxicity in rats: Role of oxidative stress, inflammation and apoptosis. Metab. Brain Dis. 2022, 37, 1259–1269. [Google Scholar] [CrossRef] [PubMed]
  136. Yesildag, K.; Gur, C.; Ileriturk, M.; Kandemir, F.M. Evaluation of oxidative stress, inflammation, apoptosis, oxidative DNA damage and metalloproteinases in the lungs of rats treated with cadmium and carvacrol. Mol. Biol. Rep. 2022, 49, 1201–1211. [Google Scholar] [CrossRef] [PubMed]
  137. Zheng, K.; Wu, S.; Lv, Y.; Pang, P.; Deng, L.; Xu, H.; Shi, Y.; Chen, X. Carvacrol inhibits the excessive immune response induced by influenza virus A via suppressing viral replication and TLR/RLR pattern recognition. J. Ethnopharmacol. 2021, 268, 113555. [Google Scholar] [CrossRef] [PubMed]
  138. Lee, B.; Yeom, M.; Shim, I.; Lee, H.; Hahm, D. Inhibitory effect of carvacrol on lipopolysaccharide-induced memory impairment in rats. Korean J. Physiol. Pharmacol. 2020, 24, 27. [Google Scholar] [CrossRef] [PubMed]
  139. Cerrah, S.; Ozcicek, F.; Gundogdu, B.; Cicek, B.; Coban, T.A.; Suleyman, B.; Altuner, D.; Bulut, S.; Suleyman, H. Carvacrol prevents acrylamide-induced oxidative and inflammatory liver damage and dysfunction in rats. Front. Pharmacol. 2023, 14, 1161448. [Google Scholar] [CrossRef]
  140. Alvi, A.M.; Al Kury, L.T.; Alattar, A.; Ullah, I.; Muhammad, A.J.; Alshaman, R.; Shah, F.A.; Khan, A.U.; Feng, J.; Li, S. Carveol attenuates seizure severity and neuroinflammation in pentylenetetrazole-kindled epileptic rats by regulating the Nrf2 signaling pathway. Oxid. Med. Cell. Longev. 2021, 2021, 9966663. [Google Scholar] [CrossRef]
  141. Asle-Rousta, M.; Amini, R.; Aghazadeh, S. Carvone suppresses oxidative stress and inflammation in the liver of immobilised rats. Arch. Physiol. Biochem. 2023, 129, 597–602. [Google Scholar] [CrossRef]
  142. Dai, M.; Wu, L.; Yu, K.; Xu, R.; Wei, Y.; Chinnathambi, A.; Alahmadi, T.A.; Zhou, M. D-Carvone inhibit cerebral ischemia/reperfusion induced inflammatory response TLR4/NLRP3 signaling pathway. Biomed. Pharmacother. 2020, 132, 110870. [Google Scholar] [CrossRef]
  143. Mohamed, M.E.; Younis, N.S. Ameliorative Effect of D-carvone against hepatic ischemia-reperfusion-induced injury in rats. Life 2022, 12, 1502. [Google Scholar] [CrossRef]
  144. Xu, G.; Guo, J.; Sun, C. Eucalyptol ameliorates early brain injury after subarachnoid haemorrhage via antioxidant and anti-inflammatory effects in a rat model. Pharm. Biol. 2021, 59, 112–118. [Google Scholar] [CrossRef]
  145. Venkataraman, B.; Almarzooqi, S.; Raj, V.; Bhongade, B.A.; Patil, R.B.; Subramanian, V.S.; Attoub, S.; Rizvi, T.A.; Adrian, T.E.; Subramanya, S.B. Molecular docking identifies 1,8-Cineole (Eucalyptol) as a novel PPARγ agonist that alleviates colon inflammation. Int. J. Mol. Sci. 2023, 24, 6160. [Google Scholar] [CrossRef]
  146. Shi, C.; Jin, T.; Guo, D.; Zhang, W.; Yang, B.; Su, D.; Xia, X. Citral attenuated intestinal inflammation induced by Cronobacter sakazakii in newborn Mice. Foodborne Pathog. Dis. 2020, 17, 243–252. [Google Scholar] [CrossRef]
  147. Formiga, R.d.O.; Alves Júnior, E.B.; Vasconcelos, R.C.; Guerra, G.C.B.; Antunes de Araújo, A.; de Carvalho, T.G.; Garcia, V.B.; de Araújo Junior, R.F.; Gadelha, F.A.A.F.; Vieira, G.C.; et al. p-Cymene and rosmarinic acid ameliorate TNBS-induced intestinal inflammation upkeeping ZO-1 and MUC-2: Role of antioxidant system and immunomodulation. Int. J. Mol. Sci. 2020, 21, 5870. [Google Scholar] [CrossRef] [PubMed]
  148. Nawaz, S.; Irfan, H.M.; Alamgeer; Arshad, L.; Jahan, S. Attenuation of CFA-induced chronic inflammation by a bicyclic monoterpene fenchone targeting inducible nitric oxide, prostaglandins, C-reactive protein and urea. Inflammopharmacology 2023, 31, 2479–2491. [Google Scholar] [CrossRef]
  149. Zou, G.; Wan, J.; Balupillai, A.; David, E.; Ranganathan, B.; Saravanan, K. Geraniol enhances peroxiredoxin-1, and prevents isoproterenol-induced oxidative stress and inflammation associated with myocardial infarction in experimental animal models. J. Biochem. Mol. Toxicol. 2022, 36, e23098. [Google Scholar] [CrossRef] [PubMed]
  150. Fu, Y.; Duan, X.; Cheng, K.; Fei, Y.Y.; Liu, L.; Duan, H.; Hu, Q.; Xia, S.; Wang, X.; Cheng, Z. Geraniol relieves mycoplasma pneumonia infection-induced lung injury in mice through the regulation of ERK/JNK and NF-κB signaling pathways. J. Biochem. Mol. Toxicol. 2022, 36, e22984. [Google Scholar] [CrossRef] [PubMed]
  151. Mahmoud, N.M.; Elshazly, S.M.; Rezq, S. Geraniol protects against cyclosporine A-induced renal injury in rats: Role of Wnt/β-catenin and PPARγ signaling pathways. Life Sci. 2022, 291, 120259. [Google Scholar] [CrossRef] [PubMed]
  152. Malik, M.N.H.; Tahir, M.N.; Alsahli, T.G.; Tusher, M.M.H.; Alzarea, S.I.; Alsuwayt, B.; Jahan, S.; Gomaa, H.A.M.; Shaker, M.E.; Ali, M.; et al. Geraniol suppresses oxidative stress, inflammation, and interstitial collagenase to protect against inflammatory arthritis. ACS Omega 2023, 8, 37128–37139. [Google Scholar] [CrossRef] [PubMed]
  153. Babaeenezhad, E.; Hadipour Moradi, F.; Rahimi Monfared, S.; Fattahi, M.D.; Nasri, M.; Amini, A.; Dezfoulian, O.; Ahmadvand, H. D-Limonene alleviates acute kidney injury following gentamicin administration in rats: Role of NF-κB pathway, mitochondrial apoptosis, oxidative stress, and PCNA. Oxid. Med. Cell. Longev. 2021, 2021, 6670007. [Google Scholar] [CrossRef] [PubMed]
  154. Han, X.; Qi, H.; Niu, J. L-limonene reduces aortic artery atherosclerosis by inhibiting oxidative stress/inflammatory responses in diabetic rats fed high-fat diet. Chin. J. Physiol. 2023, 66, 129. [Google Scholar] [CrossRef]
  155. Zhu, S.-M.; Xue, R.; Chen, Y.-F.; Zhang, Y.; Du, J.; Luo, F.-Y.; Ma, H.; Yang, Y.; Xu, R.; Li, J.-C.; et al. Antidepressant-like effects of L-menthol mediated by alleviating neuroinflammation and upregulating the BDNF/TrkB signaling pathway in subchronically lipopolysaccharide-exposed mice. Brain Res. 2023, 1816, 148472. [Google Scholar] [CrossRef]
  156. Yang, L.; Liao, M. Influence of myrcene on inflammation, matrix accumulation in the kidney tissues of streptozotocin-induced diabetic rat. Saudi J. Biol. Sci. 2021, 28, 5555–5560. [Google Scholar] [CrossRef] [PubMed]
  157. Islam, A.U.S.; Hellman, B.; Nyberg, F.; Amir, N.; Jayaraj, R.L.; Petroianu, G.; Adem, A. Myrcene attenuatesrenal inflammation and oxidative stress in the adrenalectomized rat model. Molecules 2020, 25, 4492. [Google Scholar] [CrossRef] [PubMed]
  158. Rathinam, A.; Pari, L.; Venkatesan, M.; Munusamy, S. Myrtenal attenuates oxidative stress and inflammation in a rat model of streptozotocin-induced diabetes. Arch. Physiol. Biochem. 2022, 128, 175–183. [Google Scholar] [CrossRef] [PubMed]
  159. Xuemei, L.; Qiu, S.; Chen, G.; Liu, M. Myrtenol alleviates oxidative stress and inflammation in diabetic pregnant rats via TLR4/MyD88/NF-κB signaling pathway. J. Biochem. Mol. Toxicol. 2021, 35, e22904. [Google Scholar] [CrossRef] [PubMed]
  160. Zhang, B.; Wang, H.; Yang, Z.; Cao, M.; Wang, K.; Wang, G.; Zhao, Y. Protective effect of alpha-pinene against isoproterenol-induced myocardial infarction through NF-κB signaling pathway. Hum. Exp. Toxicol. 2020, 39, 1596–1606. [Google Scholar] [CrossRef]
  161. Khan, R.; Jori, C.; Ansari, M.M.; Ahmad, A.; Nadeem, A.; Siddiqui, N.; Sultana, S. α-Terpineol mitigates dextran sulfate sodium-induced colitis in rats by attenuating inflammation and apoptosis. ACS Omega 2023, 8, 29794–29802. [Google Scholar] [CrossRef]
  162. Abd-Elhakim, Y.M.; Saber, T.M.; Metwally, M.M.M.; Abd-Allah, N.A.; Mohamed, R.M.S.M.; Ahmed, G.A. Thymol abates the detrimental impacts of imidacloprid on rat brains by lessening oxidative damage and apoptotic and inflammatory reactions. Chem. Biol. Interact. 2023, 383, 110690. [Google Scholar] [CrossRef]
  163. Hussein, R.M.; Arafa, E.-S.A.; Raheem, S.A.; Mohamed, W.R. Thymol protects against bleomycin-induced pulmonary fibrosis via abrogation of oxidative stress, inflammation, and modulation of miR-29a/TGF-β and PI3K/Akt signaling in mice. Life Sci. 2023, 314, 121256. [Google Scholar] [CrossRef]
  164. Sousa, C.; Leitão, A.J.; Neves, B.M.; Judas, F.; Cavaleiro, C.; Mendes, A.F. Standardised comparison of limonene-derived monoterpenes identifies structural determinants of anti-inflammatory activity. Sci. Rep. 2020, 10, 7199. [Google Scholar] [CrossRef] [PubMed]
  165. Daldal, H.; Nazıroğlu, M. Carvacrol protects the ARPE19 retinal pigment epithelial cells against high glucose-induced oxidative stress, apoptosis, and inflammation by suppressing the TRPM2 channel signaling pathways. Graefe’s Arch. Clin. Exp. Ophthalmol. 2022, 260, 2567–2583. [Google Scholar] [CrossRef] [PubMed]
  166. Wijesundara, N.; Lee, S.; Davidson, R.; Cheng, Z.; Rupasinghe, H. Carvacrol suppresses inflammatory biomarkers production by lipoteichoic acid- and peptidoglycan-stimulated human tonsil epithelial cells. Nutrients 2022, 14, 503. [Google Scholar] [CrossRef]
  167. Nouri, A.; Izak-Shirian, F.; Fanaei, V.; Dastan, M.; Abolfathi, M.; Moradi, A.; Khaledi, M.; Mirshekari-Jahangiri, H. Carvacrol exerts nephroprotective effect in rat model of diclofenac-induced renal injury through regulation of oxidative stress and suppression of inflammatory response. Heliyon 2021, 7, e08358. [Google Scholar] [CrossRef]
  168. Amin, F.; Memarzia, A.; Kazemi Rad, H.; Shakeri, F.; Boskabady, M.H. Systemic inflammation and oxidative stress induced by inhaled paraquat in rat improved by carvacrol, possible role of PPARγ receptors. BioFactors 2021, 47, 778–787. [Google Scholar] [CrossRef] [PubMed]
  169. Cui, Y.; Zhang, Q.; Yin, K.; Song, N.; Wang, B.; Lin, H. DEHP-induce damage in grass carp hepatocytes and the remedy of Eucalyptol. Ecotoxicol. Environ. Saf. 2020, 206, 111151. [Google Scholar] [CrossRef] [PubMed]
  170. Rui, Y.; Han, X.; Jiang, A.; Hu, J.; Li, M.; Liu, B.; Qian, F.; Huang, L. Eucalyptol prevents bleomycin-induced pulmonary fibrosis and M2 macrophage polarization. Eur. J. Pharmacol. 2022, 931, 175184. [Google Scholar] [CrossRef] [PubMed]
  171. Guo, D.; Bai, F.; Zhan, X.; Zhang, W.; Jin, T.; Wang, Y.; Xia, X.; Shi, C. Citral mitigates inflammation of Caco-2 cells induced by Cronobacter sakazakii. Food Funct. 2022, 13, 3540–3550. [Google Scholar] [CrossRef] [PubMed]
  172. Emílio-Silva, M.T.; Rodrigues, V.P.; Bueno, G.; Ohara, R.; Martins, M.G.; Horta-Júnior, J.A.C.; Branco, L.G.S.; Rocha, L.R.M.; Hiruma-Lima, C.A. Hypothermic Effect of acute citral treatment during LPS-induced systemic inflammation in obese mice: Reduction of serum TNF-α and leptin levels. Biomolecules 2020, 10, 1454. [Google Scholar] [CrossRef] [PubMed]
  173. Kang, B.; Li, N.; Liu, S.; Qi, S.; Mu, S. Protective effect of isopulegol in alleviating neuroinflammation in lipopolysaccharide-induced BV-2 cells and in Parkinson Disease model induced with MPTP. J. Environ. Pathol. Toxicol. Oncol. 2021, 40, 75–85. [Google Scholar] [CrossRef]
  174. Rai, V.K.; Sinha, P.; Yadav, K.S.; Shukla, A.; Saxena, A.; Bawankule, D.U.; Tandon, S.; Khan, F.; Chanotiya, C.S.; Yadav, N.P. Anti-psoriatic effect of Lavandula angustifolia essential oil and its major components linalool and linalyl acetate. J. Ethnopharmacol. 2020, 261, 113127. [Google Scholar] [CrossRef]
  175. Nawaz, S.; Muhammad Irfan, H.; Alamgeer; Akram, M.; Jahan, S. Linalool: Monoterpene alcohol effectiveness in chronic synovitis through lowering Interleukin-17, spleen and thymus indices. Int. Immunopharmacol. 2023, 121, 110517. [Google Scholar] [CrossRef]
  176. Wang, W.; Wang, H.; Zhao, Z.; Huang, X.; Xiong, H.; Mei, Z. Thymol activates TRPM8-mediated Ca2+ influx for its antipruritic effects and alleviates inflammatory response in Imiquimod-induced mice. Toxicol. Appl. Pharmacol. 2020, 407, 115247. [Google Scholar] [CrossRef]
  177. Şehitoğlu, M.H.; Öztopuz, R.Ö.; Kılınç, N.; Ovalı, M.A.; Büyük, B.; Gulcin, İ. Thymol regulates the Endothelin-1 at gene expression and protein synthesis levels in septic rats. Chem. Biol. Interact. 2023, 375, 110426. [Google Scholar] [CrossRef] [PubMed]
  178. Tada, A.; Nakayama-Imaohji, H.; Yamasaki, H.; Elahi, M.; Nagao, T.; Yagi, H.; Ishikawa, M.; Shibuya, K.; Kuwahara, T. Effect of thymoquinone on Fusobacterium nucleatum-associated biofilm and inflammation. Mol. Med. Rep. 2020, 22, 643–650. [Google Scholar] [CrossRef] [PubMed]
  179. Xiao, S.; Yu, H.; Xie, Y.; Guo, Y.; Fan, J.; Yao, W. The anti-inflammatory potential of Cinnamomum camphora (L.) J. Presl essential oil in vitro and in vivo. J. Ethnopharmacol. 2021, 267, 113516. [Google Scholar] [CrossRef] [PubMed]
  180. Sun, L.; Zong, S.-B.; Li, J.-C.; Lv, Y.-Z.; Liu, L.-N.; Wang, Z.-Z.; Zhou, J.; Cao, L.; Kou, J.-P.; Xiao, W. The essential oil from the twigs of Cinnamomum cassia Presl alleviates pain and inflammation in mice. J. Ethnopharmacol. 2016, 194, 904–912. [Google Scholar] [CrossRef] [PubMed]
  181. Zuzarte, M.; Sousa, C.; Cavaleiro, C.; Cruz, M.T.; Salgueiro, L. The anti-inflammatory rresponse of Lavandula luisieri and Lavandula pedunculata essential oils. Plants 2022, 11, 370. [Google Scholar] [CrossRef] [PubMed]
  182. Zuzarte, M.; Francisco, V.; Neves, B.; Liberal, J.; Cavaleiro, C.; Canhoto, J.; Salgueiro, L.; Cruz, M.T. Lavandula viridis L’Hér. essential oil inhibits the inflammatory response in macrophages through blockade of NF-KB signaling cascade. Front. Pharmacol. 2022, 12, 695911. [Google Scholar] [CrossRef] [PubMed]
  183. Roxo, M.; Zuzarte, M.; Gonçalves, M.J.; Alves-Silva, J.M.; Cavaleiro, C.; Cruz, M.T.; Salgueiro, L. Antifungal and anti-inflammatory potential of the endangered aromatic plant Thymus albicans. Sci. Rep. 2020, 10, 18859. [Google Scholar] [CrossRef] [PubMed]
  184. Zuzarte, M.; Alves-Silva, J.M.; Alves, M.; Cavaleiro, C.; Salgueiro, L.; Cruz, M.T. New insights on the anti-inflammatory potential and safety profile of Thymus carnosus and Thymus camphoratus essential oils and their main compounds. J. Ethnopharmacol. 2018, 225, 10–17. [Google Scholar] [CrossRef]
  185. Shen, C.-Y.; Jiang, J.-G.; Zhu, W.; Ou-Yang, Q. Anti-inflammatory effect of essential oil from Citrus aurantium L. var. amara Engl. J. Agric. Food Chem. 2017, 65, 8586–8594. [Google Scholar] [CrossRef]
  186. Maurya, A.K.; Mohanty, S.; Pal, A.; Chanotiya, C.S.; Bawankule, D.U. The essential oil from Citrus limetta Risso peels alleviates skin inflammation: In-vitro and in-vivo study. J. Ethnopharmacol. 2018, 212, 86–94. [Google Scholar] [CrossRef]
  187. Lombardo, G.E.; Cirmi, S.; Musumeci, L.; Pergolizzi, S.; Maugeri, A.; Russo, C.; Mannucci, C.; Calapai, G.; Navarra, M. Mechanisms Underlying the anti-inflammatory activity of bergamot essential oil and its antinociceptive effects. Plants 2020, 9, 704. [Google Scholar] [CrossRef]
  188. Muthaiyan, A. Determinants of the Gut Microbiota. In Gut Microbiome and Its Impact on Health and Diseases; Springer International Publishing: Cham, Switzerland, 2020; pp. 19–62. [Google Scholar]
  189. El-Sayed, A.; Aleya, L.; Kamel, M. Microbiota’s role in health and diseases. Environ. Sci. Pollut. Res. 2021, 28, 36967–36983. [Google Scholar] [CrossRef]
  190. Holmannova, D.; Borsky, P.; Parova, H.; Stverakova, T.; Vosmik, M.; Hruska, L.; Fiala, Z.; Borska, L. Non-genomic hallmarks of aging—The Review. Int. J. Mol. Sci. 2023, 24, 15468. [Google Scholar] [CrossRef]
  191. Malik, J.A.; Zafar, M.A.; Lamba, T.; Nanda, S.; Khan, M.A.; Agrewala, J.N. The impact of aging-induced gut microbiome dysbiosis on dendritic cells and lung diseases. Gut Microbes 2023, 15, 2290643. [Google Scholar] [CrossRef]
  192. Liu, S.; He, Y.; Zhang, Y.; Zhang, Z.; Huang, K.; Deng, L.; Liao, B.; Zhong, Y.; Feng, J. Targeting gut microbiota in aging-related cardiovascular dysfunction: Focus on the mechanisms. Gut Microbes 2023, 15, 2290331. [Google Scholar] [CrossRef] [PubMed]
  193. Huang, L.; Hong, Y.; Fu, X.; Tan, H.; Chen, Y.; Wang, Y.; Chen, D. The role of the microbiota in glaucoma. Mol. Asp. Med. 2023, 94, 101221. [Google Scholar] [CrossRef] [PubMed]
  194. Wang, Y.; Du, W.; Hu, X.; Yu, X.; Guo, C.; Jin, X.; Wang, W. Targeting the blood–brain barrier to delay aging-accompanied neurological diseases by modulating gut microbiota, circadian rhythms, and their interplays. Acta Pharm. Sin. B 2023, 13, 4667–4687. [Google Scholar] [CrossRef] [PubMed]
  195. Galloway-Peña, J.; Hanson, B. Tools for analysis of the microbiome. Dig. Dis. Sci. 2020, 65, 674–685. [Google Scholar] [CrossRef] [PubMed]
  196. De Fazio, L.; Spisni, E.; Cavazza, E.; Strillacci, A.; Candela, M.; Centanni, M.; Ricci, C.; Rizzello, F.; Campieri, M.; Valerii, M.C. Dietary geraniol by oral or enema administration strongly reduces dysbiosis and systemic inflammation in dextran sulfate sodium-treated mice. Front. Pharmacol. 2016, 7, 38. [Google Scholar] [CrossRef] [PubMed]
  197. Hawrelak, J.A.; Cattley, T.; Myers, S.P. Essential oils in the treatment of intestinal dysbiosis: A preliminary in vitro study. Altern. Med. Rev. 2009, 14, 380–384. [Google Scholar]
  198. Wei, D.; Zhao, Y.; Zhang, M.; Zhu, L.; Wang, L.; Yuan, X.; Wu, C. The volatile oil of Zanthoxylum bungeanum pericarp improved the hypothalamic-pituitary-adrenal axis and gut microbiota to attenuate chronic unpredictable stress-induced anxiety behavior in rats. Drug Des. Devel. Ther. 2021, 15, 769–786. [Google Scholar] [CrossRef]
  199. Sánchez-Quintero, M.J.; Delgado, J.; Medina-Vera, D.; Becerra-Muñoz, V.M.; Queipo-Ortuño, M.I.; Estévez, M.; Plaza-Andrades, I.; Rodríguez-Capitán, J.; Sánchez, P.L.; Crespo-Leiro, M.G.; et al. Beneficial effects of essential oils from the Mediterranean diet on gut microbiota and their metabolites in ischemic heart disease and Type-2 Diabetes Mellitus. Nutrients 2022, 14, 4650. [Google Scholar] [CrossRef] [PubMed]
  200. Lazar, V.; Holban, A.-M.; Curutiu, C.; Ditu, L.M. Modulation of gut microbiota by essential oils and inorganic nanoparticles: Impact in nutrition and health. Front. Nutr. 2022, 9, 920413. [Google Scholar] [CrossRef] [PubMed]
  201. Alavinezhad, A.; Khazdair, M.R.; Boskabady, M.H. Possible therapeutic effect of carvacrol on asthmatic patients: A randomized, double blind, placebo-controlled, Phase II clinical trial. Phyther. Res. 2018, 32, 151–159. [Google Scholar] [CrossRef] [PubMed]
  202. Khazdair, M.R.; Alavinezhad, A.; Boskabady, M.H. Carvacrol ameliorates haematological parameters, oxidant/antioxidant biomarkers and pulmonary function tests in patients with sulphur mustard-induced lung disorders: A randomized double-blind clinical trial. J. Clin. Pharm. Ther. 2018, 43, 664–674. [Google Scholar] [CrossRef] [PubMed]
  203. Ghorani, V.; Alavinezhad, A.; Rajabi, O.; Boskabady, M.H. Carvacrol improves pulmonary function tests, oxidant/antioxidant parameters and cytokine levels in asthmatic patients: A randomized, double-blind, clinical trial. Phytomedicine 2021, 85, 153539. [Google Scholar] [CrossRef] [PubMed]
  204. Rizzello, F.; Ricci, C.; Scandella, M.; Cavazza, E.; Giovanardi, E.; Valerii, M.C.; Campieri, M.; Comparone, A.; De Fazio, L.; Candela, M.; et al. Dietary geraniol ameliorates intestinal dysbiosis and relieves symptoms in irritable bowel syndrome patients: A pilot study. BMC Complement. Altern. Med. 2018, 18, 338. [Google Scholar] [CrossRef] [PubMed]
  205. Ricci, C.; Rizzello, F.; Valerii, M.C.; Spisni, E.; Gionchetti, P.; Turroni, S.; Candela, M.; D’Amico, F.; Spigarelli, R.; Bellocchio, I.; et al. Geraniol treatment for irritable bowel syndrome: A double-blind randomized clinical trial. Nutrients 2022, 14, 4208. [Google Scholar] [CrossRef]
  206. Zuzarte, M.; Vitorino, C.; Salgueiro, L.; Girão, H. Plant nanovesicles for essential oil delivery. Pharmaceutics 2022, 14, 2581. [Google Scholar] [CrossRef]
Figure 1. Chemical structures of monoterpenes with effects on ageing hallmarks.
Figure 1. Chemical structures of monoterpenes with effects on ageing hallmarks.
Biomedicines 12 00365 g001
Figure 2. Hallmarks of ageing. Hallmarks with studies on the effect of monoterpenes are highlighted on the top part, whereas on the bottom part are shown the hallmarks lacking studies. Created with BioRender.com.
Figure 2. Hallmarks of ageing. Hallmarks with studies on the effect of monoterpenes are highlighted on the top part, whereas on the bottom part are shown the hallmarks lacking studies. Created with BioRender.com.
Biomedicines 12 00365 g002
Table 1. Effect of monoterpenes on genomic instability.
Table 1. Effect of monoterpenes on genomic instability.
CompoundStudy ModelObserved EffectsRef.
In vitro
α-PineneChinese hamster cell line—V79-Cl3
(25–50 µM; 1 h)
↑ Apoptic cells (40 and 50 µM); Multipolar or incorrectly localized spindles; ↑ Hypodiploid metaphases;
↑ Endoreduplicated cells; ↑ Chromosome breaks;
↑ Kinetochore-negative micronuclei; ↑ DNA lesions;
↑ ROS production
[23]
D-LimoneneChinese hamster cell line—V79
(0.1–2.5 mM; 1 h)
At 2–2.5 mM: ↑ Nuclear abnormalities; ↑ Aberrant spindles; Cytokinesis failure[24]
↑—increase; ROS—reactive oxygen species.
Table 2. Effect of monoterpenes on mitochondrial dysfunction.
Table 2. Effect of monoterpenes on mitochondrial dysfunction.
CompoundStudy ModelEffectRef.
In vitro
1,8-CineoleCardiomyoblast cell line—H9c2 and primary neonatal cardiomiocytes
(400 µg/mL; 24 h in cells submitted to high pressure—80 mmHg)
↓ Mitophagy; ↓ Mitochondrial fission;
Restored complexes III and V protein levels
[28]
Grass carp hepatocytes—L8824
(20 µM, in the presence of 200 nM of BPA; 24 h)
↓ ROS production; ↑ Mitochondrial membrane potential[29]
Grass carp hepatocytes—L8824
(20 µM, in the presence of 8 µg/mL of TPPBA; 24 h)
↓ ROS production; ↑ Mitochondrial membrane potential[30]
Grass carp kidney cells—CIK (20 µM, in the presence of 27.8 µg/mL of DiBP; 24 h)↓ ROS production[31]
CampheneMyoblast cell line—L6 (300 µM in serum-free medium; 48 h)Prevented mitochondrial shape alteration[32]
CarvacrolNeuroblastoma cell line—SH-SY5Y (100 µM; 4 h before the addition of
300 µM of H2O2; 24 h)
↑ Complexes I and V activities; ↑ ATP levels; ↑ Mitochondrial membrane potential; ↑ aconitase, α-KGDH, and SDH activities[33]
GeraniolNeuroblastoma cell line—SK-N-SH
(60 nM; 2 h followed by 24 h with
100 nM of rotenone)
↓ ROS production; ↑ Mitochondrial membrane potential; ↑ Complex I activity; ↑ ATP production; ↑ Mitophagy of damaged mitochondria[34]
LimoneneSkin epidermal keratinocytes cell line—HaCaT (25, 50 and 100 µM; 2 h before UVB irradation; 30 min to 24 h)↓ ROS production; ↑ HO-1, NQO-1 and γ-GCLC protein levels; ↑ Nrf2 expression[35]
MG-induced damage in osteoblast—MC3T3-E1 (0.01—1 µM for 1 h prior to 48 h stimulation with 400 µM of MG)↓ Mitochondrial superoxide levels;
↓ Cardiolipin peroxidation; ↑ ATP production; ↑ Mitochondrial membrane potential
[36]
LinaloolHippocampal neuronal cell line—
HT-22 (100 µM in the presence of
6 mM of glutamate; Respiratory capacity assays: 100 and 200 µM; 24 h)
↓ Mitochondrial fragmentation;
↓ Mitochondrial ROS production;
↑ Mitochondrial membrane potential;
↓ Mitochondrial Ca2+ levels;
↑ Mitochondrial respiratory capacity
[37]
MentholOsteosarcoma cell line—Saos2
(100 µM; 6 h)
↑ Intracellular and mitochondrial Ca2+ levels; ↑ Mitochondrial circularity;
↑ ATP production; ↓ Mitochondrial membrane potential; ↑ Mitochondrial ROS and cardiolipin levels; ↑ ER–mitochondria contact sites; Restored mitochondrial membrane potential in the presence of CCCP
[38]
MyrceneDermal fibroblasts—primary cultures (0.1, 1 and 10 μM; 24 h or 72 h after UVB irradiation)↓ ROS production[39]
Perillyl alcoholHuman microglial cell line—HMC-3 (100 and 200 µM in the presence of LPS/H2O2; 24 h)↑ Mitochondrial membrane potential;
↓ Mitochondrial ROS production; ↓ H2O2 release; ↑ Parkin translocation to damaged mitochondria
[40]
Neuroblastoma cell line—SH-SY5Y
(10 and 20 µM; 1 h prior to an overnight incubation with 150 µM of
6-OHDA)
↓ Intracellular ROS production;
↑ Mitochondrial membrane potential
[41]
Neuroblastoma cell line—SH-SY5Y
(10 and 20 µM; 1 h followed by 24 h with 40 µM of β-amyloid 25–35)
↓ ROS production; ↑ Mitochondrial membrane potential (20 µM); ↑ mPTP opening[42]
In vivo
1,8-CineoleMonocrotaline-induced pulmonary arterial hypertension rat model
(25 mg/Kg transdermally; daily for
3 weeks)
↓ Mitophagy; ↓ Mitochondrial fission [28]
CarvacrolNeuropathic pain animal model
(30 and 60 mg/Kg p.o.; 14 days)
↓ NO production; ↑ ATP production;
↑ NRF-1, TFAM, PGC-1α, complex I and ATP synthase C protein levels; ↓ Drp1 and Fis1 protein levels
[43]
GeraniolACR-induced neurotoxicity in Drosophila melanogaster (5 and 10 µM in culture medium; 7 days)↓ ROS production; ↑ MTT reduction;
↑ SDH and CS activity
[44]
STZ-induced diabetic neuropathy in rats (100 mg/Kg/day p.o.; 8 weeks)↓ ROS production in sciatic nerve and brain; ↓ NO levels; ↑ MTT reduction;
↑ Complexes I–III, SDH, CS activity
[45]
ACR-induced neuropathy in rats
(100 mg/Kg/day p.o. and simultaneous administration of ACR, 50 mg/Kg i.p.; twice a week for 4 weeks)
↓ ROS production in sciatic nerve and brain regions; ↓ Mitochondrial ROS production in cortex and cerebellum;
↑ MTT reduction in cortex and cerebellum; ↑ Complexes I–III, complexes II and III and CS activities
[46]
LimoneneRotenone-induced dopaminergic neurodegeneration in rats (50 mg/Kg p.o. and Rot 2.5 mg/Kg, i.p.; 5 days a week for 28 days)↑ Mitochondrial respiratory capacity; ↑ Complex I levels[47]
Perillyl alcohol6-OHDA-induced Parkinson’s disease rat model (100 mg/Kg p.o.; 7-day pre- and 7-day post-surgery)↓ Intracellular ROS production;
↑ PGC-1α mRNA and protein levels in striatum; ↑ Complexes I and IV levels;
↓ Bax and Drp1 protein levels;
↑ Nuclear accumulation of Nrf2 and PGC-1α
[48]
MPTP-induced Parkinson’s disease rat model (100 and 200 mg/Kg/day p.o.;
14 days following 5 consecutive days of 25 mg/Kg of MPTP)
↑ ATP levels[40]
ThymolGrass carp (100–300 mg/Kg before a
7-day infection with Aeromonas hydrophylla; 68 days)
↑ Cytosolic and mitochondrial CK
(100 mg/Kg); ↑ Branchial AK activity
(100 and 300 mg/Kg); ↑ Branchial ATP levels; ↓ Branchial ROS production
[49]
ISO-induced myocardial infarction rat model (7.5 mg/Kg intragastric; 7 days with ISO (100 mg/Kg), s.c.,
administered on days 6 and 7)
↑ Mitochondrial enzymes activity;
↓ Mitochondrial Ca2+ levels; ↑ ATP production; Maintained mitochondrial architecture; ↓ Mitochondrial swelling
[50]
↓—decrease; ↑—increase; 6-OHDA—6-hydroxydopamine; ACR—acrylamide; AK—Adenylate Kinase; ATP—adenosine triphosphate; Bax—BCL-2-associated protein X; BPA—bisphenol A; Ca2+—calcium; CCCP—carbonyl cyanide m-chlorophenyl hydrazone; CK—Creatine Kinase; CS—Citrate Synthase; DiBP—diisobutyl phthalate; GCLC—Glutamate–Cysteine Ligase Catalytic subunit; Drp1—Dynamin-related protein 1; Fis1—Mitochondrial Fission 1 protein; H2O2—hydrogen peroxide; HO-1—HemeOxygenase-1; i.p.—intraperitoneally; ISO—isoproterenol; MG—methylglyoxal; MPTP—1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine; mPTP—mitochondrial permeability transition pore; mRNA—messenger ribonucleic acid; MTT—3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide; NRF-1—Nuclear Respiratory Factor 1; Nrf2 –Nuclear factor erythroid 2-Related Factor 2; NO—nitric oxide; NQO-1—NAD(P)H Quinone Dehydrogenase 1; LPS—lipopolysaccharide; p.o.—per os (orally); PGC-1α—Peroxisome proliferator-activated receptor-gamma Coactivator-1alpha; ROS—reactive oxygen species; ROT—rotenone; s.c. –subcutaneously; SDH—Succinate Dehydrogenase; TBBPA—tetrabromobisphenol A; TFAM—Mitochondrial Transcription Factor A; UVB—Ultraviolet B; α-KGDH—alpha-KetoGlutarate DeHydrogenase.
Table 3. Effect of monoterpenes on cellular senescence.
Table 3. Effect of monoterpenes on cellular senescence.
CompoundStudy ModelEffectRef.
In vitro
CamphorPrimary cultures of dermal fibroblasts (65, 130 and 260 µM;
24 h and 260 µM; 6, 12 or 24 h)
↓ Elastase activity; ↑ Elastin and collagen productions; ↓ SA-β-galactosidase activity (in the presence of H2O2)[69]
HinokitiolSkin fibroblast cell line—966SK following UVB irradiation (4, 8 and 12 µM; 24 h)↓ Secreted MMP-1 levels; ↓ MMP-1 and MMP-3 mRNA levels; ↑ Procollagen mRNA levels[70]
D-LimoneneSkin epidermal keratinocytes cell line—HaCaT (25, 50 and 100 µM for 2 h before UVB irradation; 30 min to 24 h)↓ α-MSH intracellular levels; ↓ POMC mRNA; ↓ Phosphorylated levels of p53; Maintained skin barrier function[35]
MyrceneNormal human dermal fibroblasts (0.1, 1 and 10 μM; 1, 4, 24 or 72 h after UVB irradiation)↓ ROS production; ↓ MMP-1 and MMP-3 secretion; ↓ IL-6 secretion; ↑ Procollagen-1 and TGF-1β secretion; ↓ MMP-1 mRNA levels;
↑ Procollagen mRNA levels; ↓ MAPK activation; ↓ AP-1 activation
[39]
α-PineneSkin epidermal keratinocytes cell line—HaCaT (30 µM; 30 min before UVA irradiation, then cultured for 24 h)↓ Single-strand DNA damage; ↓ Pyrimidine dimers formation; ↑ NER pathway-associated proteins expression; ↑ p53 and p21 protein levels[71]
In vivo
CamphorUV-induced wrinkle formation in mice (26 and 52 mM, topical; for
2 weeks after 4 weeks of UV irradiation)
↑ Collagen 1, collagen II and elastin production; ↓ MMP-1 protein levels;
↓ Epidermis and subcutaneous fat layer thickness
[69]
α-PineneUVA-induced photoageing mice model (100 mg/Kg, topical; 1 h prior to irradiation)↑ Collagen staining; ↓ MMP-2 staining;
↓ MMP-9 and MMP-13 mRNA expression;
↓ MMP-1 and MMP-9 protein levels
[72]
—decrease; —increase; AP-1—Activator Protein 1; IL—InterLeukin; MAPK—Mitogen-Activated Protein Kinase; MMP—Matrix MetalloProteinase; NER—Nucleotide excision repair; POMC—pro-opiomelanocortin; SA—senescence-associated; TGF-1β—Transforming Growth Factor 1β; UV—Ultraviolet; UVA—Ultraviolet A; UVB—Ultraviolet B; α-MSH—α-Melanocyte-Stimulating Hormone.
Table 4. Effect of monoterpenes on autophagy.
Table 4. Effect of monoterpenes on autophagy.
CompoundStudy ModelEffectRef.
In vitro
CarvacrolCardiomyoblasts—H9c2 cells
(2.5, 5 and 10 µg/mL; 6 h followed by
1 µg/mL of LPS; 18 h)
↑ Beclin-1 protein levels; ↓ p62 protein levels[95]
1,8-CineoleGrass carp hepatocytes—L8824 (20 µM and 200 nM of BPA; 24 h)↑ LC3B fluorescence; ↓ p62 fluorescence; ↑ LC3, Atg5 and beclin-1 mRNA and protein levels; ↓ p62 mRNA and protein levels[29]
Human umbilical vein endothelial cells—HUVEC (5 and 50 µM; 1.5 h followed by 300 µM of L-NAME; 24 h)↓ LC3-II/LC3-I ratio; ↑ p62 mRNA and protein levels; ↓ LC3 fluorescence; ↑ PI3K and mTOR phosphorylated levels[96]
Human bronchial epithelial cell line—BEAS-2B (6.25–200 µM; 2 h, before 4 h with CSE)↓ LC3-II/LC3-I ratio[97]
Ctenopharyngodon idellus kidney cells (20 µM and 27.8 µg/mL of DiBP; 24 h)↑ Autophagosome formation; ↑ Atg5, beclin-1 and LC3 mRNA levels; ↓ p62 mRNA and protein levels; ↑ LC3 and beclin-1 protein levels[31]
HinokitiolNeuroblastoma cell line—SK-N-SH
(2, 4 and 8 µM; 6 h followed by 100 µM of PrP; 6 h)
↑ LC3-II/LC3-I ratio; ↑ LC3 puncta;
↑ Autophagosome formation; ↑ p62 mRNA and protein levels
[98]
Cardiomyocyte cell line—AC16
(20 µM; 30 min followed by 1 mM of H2O2; 2 h)
↑ Phosphorylated form of mTOR and p62 protein levels; ↓ Beclin-1 protein levels and LC3-II/LC3-I ratio; ↓ LC3-positive cells[99]
MentholBovine mammary epithelial cells
(200 µM; 1 h followed by 5 µg/mL of LPS; 12 h)
↓ p62 protein levels; ↑ LC3-II protein levels; ↑ Autophagosomes and autophagolysosome formation[100]
ThymolMouse liver cell line—AML12
(100 µM in the presence of 100 mM of EtOH; 48 h)
↓ LC3-II and p62 protein levels; ↓ LC3-II/LC3-I ratio[101]
In vivo
BorneolCerebral ischemia/reperfusion rat model (0.16 mg/Kg/day p.o.; 7 days)Cortex: ↑ ULK1 protein levels and LC3-II/LC3-I ratio
Hippocampus: ↑ ULK1 protein levels and LC3-II/LC3-I ratio; ↓ mTOR
[102]
CarvacrolLPS-induced cardiac dysfunction mice model (50 and 100 mg/Kg i.p.; 2 h prior to 10 mg/Kg of LPS; 12 h)↑ Beclin-1 protein levels; ↓ p62 protein levels[95]
Chronic constriction injury of sciatic-nerve-induced neuropathic pain in rats (30 and 60 mg/Kg/day p.o.; 14 days)↑ Beclin-1, LC3-II, Atg7 and Atg16 protein levels; ↓ p62 protein levels and immunostaining[43]
1,8-CineoleL-NAME-induced hypertension rat model (20 and 40 mg/Kg/day, gavage on weeks 5 to 8 during an 8-week administration of L-NAME
40 mg/Kg/day)
↓ LC3-II/LC3-I ratio; ↑ p62 mRNA and protein levels; ↓ LC3 immunohistochemical staining in rat blood vessels; ↑ PI3K and mTOR phosphorylated levels[96]
CitronellolRotenone-induced Parkinson’s disease rat model (25 mg/Kg p.o.; 30 min before 2.5 mg/Kg i.p. of Rot; 4 weeks)↓ LC3 and p62 protein levels[103]
GeraniolISO-induced myocardial infarction (100 and 200 mg/Kg/day p.o. followed by 100 mg/Kg s.c. of ISO on days
13 and 14)
↑ PI3K, Akt and mTOR mRNA levels;
↑ Phosphorylated levels of PI3K, Akt and mTOR
[104]
MyrceneRotenone-induced Parkinson’s disease rat model (25 mg/Kg p.o.; 30 min before 2.5 mg/Kg i.p. of Rot; 5 days a week for 28 days)↓ Beclin-1, p62 and LC3B protein levels;
↑ p-mTOR/mTOR ratio
[105]
ThymolLPS-induced liver inflammation mouse model (80 mg/Kg/day, gavage, for 34 days followed by 10 mg/Kg, i.p. of LPS; 4 h)↓ p62 mRNA levels; ↑ Atg7 mRNA levels; ↑ phosphorylated levels of AMPK[106]
—decrease; —increase; Akt—Protein kinase B; AMPK—5′Adenosine Monophosphate-activated Protein Kinase; Atg—autophagy-related; BPA—bisphenol A; CSE—cigarette smoke extract; DiBP—diisobutyl phthalate; EtOH—ethanol; H2O2—hydrogen peroxide; i.p.—intraperitoneally; ISO—isoproterenol; L—NAME—N(ω)-nitro-L-arginine methyl ester; LC3—Microtubule-associated protein Light Chain 3; LPS—lipopolysaccharide; mRNA—messenger ribonucleic acid; mTOR—mammalian Target Of Rapamycin; p.o.—per os (orally); PrP—Prion Protein; p62/SQSTM1—Sequestosome-1; PI3K—PhosphoInositide 3-Kinases; Rot—rotenone; ULK1—Unc-51-Like Kinase 1.
Table 5. Effect of monoterpenes on inflammation.
Table 5. Effect of monoterpenes on inflammation.
CompoundStudy ModelObserved EffectsRef.
In vitro
CarvacrolHuman umbilical vein endothelial cells—HUVEC (10 mg/Kg; 24 h in HG medium)↓ IKK, NALP3, NF-κB and TLR4 mRNA and protein levels[118]
(S)-(+)-CarvoneMouse leukemic cell line—RAW 264.7 (666 µM; 5 min and 1 h, before 1 µg/mL of LPS)↓ p-JNK 1; ↓ Ac-p65; ↓ IκB-α resynthesis;
↑ SIRT 1 activity (in chemico)
[119]
(R)-(-)-CarvoneMouse leukemic cell line—RAW 264.7 (666 µM; 5 min, 1 h and 18 h before 1 µg/mL of LPS)↓ p-JNK 1; ↓ Ac-p65 (tendency); ↓ IκB-α resynthesis; ↑ Nrf2 translocation (tendency); ↑ HO-1 protein level (tendency)[120]
1,8-CineoleHuman bronchial epithelial cell line—BEAS-2B (6.25–200 µM; 2 h, before 4 h with CSE)↑ Nrf2 nuclear translocation; ↓ ROS and
IL-6 levels
[97]
Mouse leukemic cell line—RAW 264.7 (0.1, 1, 5 and 10 µM; 20 min, before 0.5 mg/mL of MSU for 4 h)↑ Nrf2 protein levels; ↓ TNF-α, IL-6, CXCL1 and CXCL2 mRNA expression[121]
CitralPorcine jejunal epithelial cell line—IPEC-J2 (PGN-stimulated cells)↓ Cytokine expression; ↓ TLR2 protein levels; ↓ TLR2/NF-κB signaling pathway[122]
Human peripheral blood mononuclear cells—PBMC (4, 2 and 1%; 30 min before and after innoculation with S. aureus for 6 h—TI/IT, respectively)TI: ↓ IL-1β, IL-6, IL-12p70, IL-23, IFN-γ and TNF-α levels
IT: ↓ IL-1β, TNF-α and IL-6 (only 4%)
[123]
GeraniolMicroglial cell line—BV-2
(200-, 500- and 1000-fold concentrations; 24 h pretreatment with geraniol followed by 24 h with
1 µg/mL of LPS. Pretreatment with LPS; 24 h, followed by 24 h with geraniol, co-stimulation with LPS and geraniol for 24 h)
Geraniol pretreatment: ↓ TNF-α and IL-6 mRNA and secretion; ↓ p50 and p-C/EBP-β chromatin-bound protein levels
LPS pretreatment: ↓ TNF-α and IL-6 mRNA and secretion; ↓ p50 and p-C/EBP-β chromatin-bound protein levels
Co-treatment: ↓ TNF-α and IL-6 mRNA and secretion; ↓ p50 and p65 chromatin-bound protein levels
[124]
Primary cultures of macrophages
(5, 25 and 50 µM; 12 h followed by
1 µg/mL of LPS for 24 h)
↓ IL-1β protein levels; ↓ IL-1β and NLRP3 mRNA expression; ↓ TNF-α mRNA expression; ↑ IL-4 and IL-10 mRNA expressions[125]
Human umbilical vein endothelial cells—HUVEC (0–100 µmol/mL; 2 h before 100 µg/mL of OxLDL for 72 h)↓ TNF-α, IL-6, IL-1β, ICAM-1, VCAM-1 and TGF-β protein levels; ↓ IL-6, VCAM-1 and ICAM-1 mRNA levels; ↓ p-IκBα and p-p65 protein levels[126]
HinokitiolMouse leukemic cell line—RAW 264.7 (5, 10 and 20 µg/mL; 1 h, before 100 ng/mL of LPS for 3 h)↓ IL-6, IL-1β, TNF-α and NLRP3 mRNA expression[127]
LinaloolMicroglial cell line—BV-2
(200-, 500- and 1000-fold concentrations; 24 h pretreatment with linalool followed by 24 h with 1 µg/mL of LPS. Pretreatment with LPS; 24 h, followed by 24 h with linalool, co-stimulation with LPS and linalool for 24 h)
Linalool pretreatment: ↓ TNF-α and IL-6 mRNA and secretion; ↓ p50, p65 and
p-C/EBP-β chromatin-bound protein levels
LPS pretreatment: ↓ TNF-α mRNA and secretion; ↓ IL-6 secretion; ↓ p50 and
p-C/EBP-β chromatin-bound protein levels
Co-treatment: ↓ TNF-α secretion
[124]
MentholMicroglial cell line—BV-2 (5–40 µM;
2 h, before 1 µg/mL of LPS for 2 or
12 h)
↓ iNOS and COX-2 mRNA and protein levels; ↓ IL-1β, IL-6 and TNF-α mRNA and protein expression; ↓ p-p65/p65, p-Akt/Akt,
p-ERK/ERK, p-JNK/JNK and p-p38/p38 ratios
[128]
MyrceneDermal fibroblasts—primary cultures (0.1, 1 and 10 μM; 24 h or 72 h after UVB irradiation)↓ IL-6 release[39]
Perillyl alcoholHuman keratinocytes cell line—HaCaT (50 and 100 µM; 2 h prior to
1 µg/mL of LPS for 22 h)
↓ p-p65 and p-STAT3 nuclear accumulation[129]
Human embryonic kidney—HEK293 (25 and 50 µM; 24 h following 30 ng/mL of TNF-α for 2 h)↓ NF-κB activation; ↓ p-NF-κB and p-IκBα immunoreactivity[130]
PerillaldehydeHuman corneal epithelial cell line—HCEC (0.6 mM; 4 h, followed by 8 h stimulation with A. fumigatus)↓ IL-1β, IL-6, TNF-α and IL-8 mRNA expression; ↓ IL-6, TNF-α and IL-8 secretion; ↑ Nrf2 and HO-1 mRNA and protein expression; ↓ Dectin-1 mRNA and protein levels[131]
α-PineneSkin epidermal keratinocytes cell line—HaCaT (30 µM; 30 min before UVA irradiation, cultured for an additional 24 h)↓ NF-κB, IL-6 and TNF-α protein levels[71]
ThujanolMicroglial cell line—BV-2
(200-, 500- and 1000-fold concentrations; 24 h pretreatment followed by 24 h with
1 µg/mL of LPS. Pretreatment with LPS; 24 h, followed by 24 h with thujanol, co-stimulation with LPS and thujanol for 24 h)
Thujanol pretreatment: ↓ TNF-α and mRNA and secretion;
LPS pretreatment: ↓ TNF-α secretion;
↓ p50, p65 and p-C/EBP-β chromatin-bound protein levels. Co-treatment: ↓ p50 chromatin-bound protein levels
[124]
ThymolMouse leukemic cell line—RAW 264.7 (30–150 µM; 2 h before 1 µg/mL of LPS for 24 h)↓ NO production; ↓ TNF-α and IL-6 release (120 µM); ↓ ROS production;
↓ COX-2 protein expression; ↓ p65 nuclear translocation
[132]
In vivo
BorneolCerulein-induced acute pancreatitis mice model (100 and 300 mg/Kg, p.o.; 7 days, on day 7, 6 injections of cerulein
(50 µg/Kg, i.p.) were given 1 h apart, sacrifice 6 h after last injection)
↓ NO, IL-1β and IL-6 levels; ↓ MPO activity; ↓ iNOS, IL-1β, p-NF-κB, TNF-α and IL-6 protein levels; ↓ Inflammatory cells infiltration[133]
CarvacrolHyperuricemia-induced inflammation rat model (20 and 50 mg/Kg i.p.;
7 days, 1 h after PO administration)
↓ Monourate crystals; ↓ Lymphocyte infiltration; ↓ TNFα and NRLP3 levels;
↓ TNF-α and p-NF-κB staining
[134]
CdCl2-induced neurotoxicity rat model (25 and 50 mg/mL wth 25 mg/Kg p.o. of CdCl2; daily for 7 days)↓ NF-κB release; ↓ Brain COX-2, MPO and PGE2 levels; ↓ Brain nNOS, iNOS, GFAP and MAO levels; ↓ TNF-α, IL-1β, MMP-9 and MMP-13 mRNA levels[135]
CdCl2-induced lung injury rat model (25 and 50 mg/Kg, p.o.; 30 min after
25 mg/Kg, p.o. of CdCl2 for 7 days)
↓ Lung NF-κB, iNOS, COX-2, MPO and PGE2 levels; ↓ TNF-α, IL-1β, MMP-2 and MMP-9 mRNA expression[136]
T2DM db/db mice model
(5 and 10 mg/Kg, gavage; 6 weeks)
↓ IL-1β, IL-6, IL-18 and TNF-α secretion;
↓ IKK, NALP3, NF-κB and TLR4 mRNA and protein levels
[118]
FM1-induced viral infection mice model (50 mg/Kg, intranasally; 5 days post viral infection)↑ % of Treg cells; ↓ Th1/Th2 and Th17/Treg ratio; ↓ IFN-γ, IL-2, IL-4, IL-12, TNF-α, IL-1, IL-10 and IL-6 secretion; ↓ RIG-I, MyD88, NF-κB mRNA and protein levels[137]
LPS-induced memory impairment rat model (25, 50 and 100 mg/Kg, i.p.; until 21 post-LPS sacrifice at day 28
post-LPS)
↓ IL-1β, IL-6, TNF-α, COX-2 and NF-κB levels; ↓ iNOS, TLR4 and BDNF mRNA expression[138]
ACR-induced liver damage rat model (50 mg/Kg, i.p., followed by 20 mg/Kg, p.o. of ACR; 30 days)↓ TNF-α, IL-1β and NF-κB protein levels[139]
CarveolPTZ-kindled epileptic rat model
(10 and 20 mg/Kg 30 min before PTZ; repeated every 48 h for 15 days)
↓ TNF-α, p-p65 and COX-2 protein levels[140]
S-CarvoneStress-induced liver damage rat model (20 mg/Kg, gavage, with restraint for
6 h; daily for 21 days)
↓ TNF-α, IL-6, IL-1β and NF-κB mRNA expression; ↓ Inflammatory cells infiltration[141]
Cerebral I/R-induced neuroinflammation rat model
(10 and 20 mg/Kg, i.p.; 15 min before reperfusion daily for 15 days)
↓ Serum and brain IL-1β and TNF-α levels; ↓ IL-6 and IL-4 levels; ↑ IL-10 levels;
↓ NLRP3, ASC, TLR4, IL-1β, TNF-α mRNA levels
[142]
Hepatic I/R-induced injury rat model (25 and 50 mg/Kg, gavage; 3 weeks before I/R)↓ HMGB1, TLR4, NF-κB and NLRP3 mRNA expression; ↓ TLR4 and NF-κB immunoreactivity; ↓ ICAM-1, MPO, IL-1β, IL-6 and TNF-α protein levels; ↑ IL-10 protein levels[143]
1,8-CineoleSAH-induced early brain injury rat model (100 mg/Kg, i.p.; 1 h before SAH and 30 min after)↓ Iba-1 and p65 protein levels; ↓ TNF-α, IL-1β and IL-6 mRNA levels[144]
MSU-induced gout arthritis mice model (30–600 mg/Kg, i.p.; 1 h before and 5, 23 and 47 h after 0.5 mg/20 µL of MSU, intra-articular)↓ Ankle edema; ↓ cell infiltration;
↓ MPO activity; ↓ NLRP3 and IL-1β mRNA and protein levels
[121]
DSS-induced ulcerative colitis mice model (100 and 200 mg/Kg with
2% DSS; 8 days)
↓ IL-6, IL-1β, TNF-α, IL-17A protein and mRNA levels; ↓ iNOS and COX-2 mRNA and protein levels; ↓ p-p65/p65 ratio[145]
CitralC. sakazakii-induced intestinal inflammation in newborn mice
(0.54 mg/mL, gavage; once per day for 8 days, starting on day 3 postnatal, at day 7, C. sakazakii p.o. was given, sacrifice at day 10)
↓ IL-1β, TNF-α, PAF receptor, IL-6, IFN-γ, NF-κB p65 and iNOS mRNA levels; ↓ IL-6 and TNF-α levels; ↓ NF-κB p65 protein levels; ↑ IκBα protein levels[146]
p-CymeneTNBS-induced intestinal inflammation rat model (25, 50, 100 and 200 mg/Kg, p.o.; 48 h, 24 h and 1 h before and 24 h after 10 mg/animal TNBS)↓ Inflammatory lesions; ↓ MPO activity;
↓ IL-1β and TNF-α secretion; ↑ IL-10 secretion;↓ COX-2, IFN-γ, iNOS, p65 and SOCS3 mRNA expression
[147]
FenchoneFCA-induced arthritis rat model
(100, 200 and 400 mg/Kg; daily for 28 days post FCA injection)
↓ Paw volume and arthritis severity;
↓ iNOS, IL-17, COX-2, IL-1β, TNF-α and IL-6 mRNA levels; ↑ IL-10 levels; ↓ NO and PGE2 production
[148]
GeraniolISO-induced myocardial infarction rat model (100 mg/Kg/day p.o.; 14 days,
85 mg/Kg of ISO i.p., in the last 2 days)
↓ TNF-α, IL-6 and NF-κB protein levels;
↓ MMP-9 and MMP-2 mRNA levels
[149]
Mycoplasma pneumoniae-induced pneumonia mice model (20 mg/Kg;
3 days, after 2-day infection by M. pneumoniae)
↓ IL-1, IL-6, IL-8, TNF-α and TGF levels;
↓ ERK1/2 and NF-κB mRNA expression
[150]
Cyclosporine A-induced renal injury rat model (50, 100 and 200 mg/Kg, intragastric, 1 h prior to 30 mg/Kg of cyclosporine; daily for 28 days)↓ NF-κB mRNA levels; ↓ Renal IL-18 and ICAM-1 levels; ↓ Renal TGF-β levels;
↓ MMP-9 mRNA expression
[151]
FCA-induced arthritis rat model
(25, 50 and 100 mg/Kg, i.p.; days 7 to 35 post FCA injection, on alternate days)
↓ Paw edema; ↓ Arthritis severity;
↓ NF-κB, IL-1β, TNF-α, COX-2, mPGES-1, PTGDS and MMP-1 mRNA levels
[152]
HinokitiolTooth-ligation-induced periodontitis mice model (2 mg/mL injected into the palatal gingiva; daily for 7 days)Prevented bone loss; ↓ IL-6, IL-1β, TNF-α and NLRP3 mRNA expression;[127]
LimoneneGentamycin-induced acute kidney injury rat model (100 mg/Kg p.o.; 1 h before 100 mg/Kg, i.p., of gentamycin; daily for 12 days)↓ NO production; ↓ Renal TNF-α, IL-6 and NF-κB mRNA levels; ↓ TNF-α immunoreactivity[153]
HFD-induced atherosclerosis in diabetic rat model (200 mg/Kg, gavage on day 30, under HFD; for 8 weeks)↓ TNF-α and IL-6 protein levels; ↑ IL-10 protein levels; ↑ p-AMPK/APMK ratio;
↓ p-p65/p65 ratio
[154]
MentholLPS-induced Parkinson’s disease rat model (10 and 20 mg/Kg, gavage;
28 days post LPS injection)
↓ Iba-1-positive cells; ↓ OX-42 protein levels; ↓ iNOS and COX-2 protein levels;
↓ IL-1β, IL-6, TNF-α, COX-2 and iNOS mRNA expression; ↓ p-p65/p65, p-Akt/Akt, p-ERK/ERK, p-JNK/JNK and p-p38/p38 ratios
[128]
LPS-induced neuroinflammation mice model (26 and 52 mg/Kg, p.o. with
0.2 mg/Kg, i.p., of LPS; daily for 12 days)
↓ Iba1-positive microglia; ↓ TNF-α, IL-1β and IL-6 protein levels; ↑ BDNF, TrkB protein levels and p-CREB/CREB ratio[155]
MyrceneSTZ-induced diabetic rat
(25 mg/Kg, 72 h after diabetes induction; for 45 days)
↓ TNF-α and IL-6 serum levels; ↓ TGF-1β, TNF-α and NF-κB levels; ↓ MCP-1, VCAM-1 and ICAM-1 levels; ↑ MMP-2 protein levels; ↓ TIMP1- protein levels[156]
ADX-induced renal inflammation rat mode (100 mg/Kg; for 14 days post-surgery)↓ IL-1β, IL-6, TNF-α, p65, and IL-4 levels;
↑ IFN-γ and IL-10 levels; ↓ iNOS and
COX-2 protein levels
[157]
MyrtenalSTZ-induced diabetes rat model
(80 mg/Kg, p.o.; daily for 4 weeks)
↓ TNF-α, IL-6 and NF-κB liver and pancreas mRNA and protein levels[158]
MyrtenolSTZ-induced diabetes in pregnant rat model (50 mg/Kg, p.o.; daily for
2 weeks, starting on gestational day 4)
↓ TLR4, MyD88, p65 and NRLP3 mRNA levels; ↓ TNF-α and IL-1β production[159]
Perillyl alcoholImiquimod-induced psoriasis-like skin inflammation (100 and 200 mg/Kg, topical; days 3 to 7 post 62.5 mg/Kg imiquimod ↓ Psoriasis severity (scaling, epidermal thickness, erythema); ↓ Skin NO, IL-1β, IL-6, IL-12/IL-23p40 and TNF-α levels;
↓ Skin TLR7, TLR8, IL-23, IL-17 and IL-22 mRNA expression; ↓ Skin iNOS, IL-17A, TNF-α, p-p65, COX-2, IL-22 and IL-10 protein levels
[129]
DSS-induced ulcerative colitis mice model (100 and 200 mg/Kg, p.o.; from day 15 to day 28, chronic restraint stress days 1 to 28 and 2.5% DSS days
8 to 14)
↓ NO, IL-1β, IL-6, TNF-α release; ↓ IL-1β, TNF-α, TLR4 and NF-κB mRNA expression;
↓ MPO activity; ↓ IL-6, p-IκBα and TNF-α protein levels; ↓ p-NF-κB/NF-κB ratio
[130]
α-PineneUVA-induced photoageing mice model (100 mg/Kg, topical; 1 h prior to irradiation)↓ COX-2, NF-κB, iNOS, VEGF and CD34 expression in mouse skin; ↓ COX-2, iNOS, VEGF protein levels; ↓ Nuclear translocation of NF-κB[72]
ISO-induced myocardial infarction rat model (50 mg/Kg, p.o.; 21 days with
85 mg/Kg, s.c., of ISO, on days 20 and 21)
↓ TNF-α and IL-6 secretion;
↓ TNF-α, IL-6 and NF-κB protein levels
[160]
α-TerpineolDSS-induced ulcerative colitis
(50 mg/Kg, p.o.; 14 days, DSS in water from days 7 to 14)
↓ NO and MPO content; ↓ Mast cell infiltration; ↓ NF-κB p65, COX-2 and iNOS immunoreactivity;[161]
ThymolIMID-induced brain damage rat model (30 mg/Kg, p.o., 1 h before 22.5 mg/Kg, p.o., of IMID; 56 days)↓ NO and MPO content; ↓ NF-κB immunoreactivity[162]
Bleomycin-induced pulmonary fibrosis mice model (50 and 100 mg/Kg, p.o., with 15 mg/Kg, i.p., of bleomycin; daily for 4 weeks)↓ Inflammatory infiltrate and edema;
↓ TNF-α, IL-1β, IL-6 and NF-κB protein levels
[163]
—decrease; —increase; Ac—acetylated; ACR—acrylamide; ADX—adrenalectomy; Akt—Protein kinase B; AMPK—5′Adenosine Monophosphate-activated Protein Kinase; ASC—Apoptosis-associated speck-like protein containing a caspase recruitment domain; BDNF—Brain-Derived Neurotrophic Factor; CD35—cluster of differentiation 35; C/EBP-β—CCAAT/Enhancer-Binding Protein beta; CdCl2—cadmium chloride; COX—Cyclooxygenase; CREB—cAMP Response Element-Binding protein; CSE—cigarette smoke extract; CXCL—chemokine (C-X-C motif) ligand; DSS—dextran sulphate sodium; ERK—Extracellular signal-Regulated Kinases; FCA—Freund’s complete adjuvant; GFAP—Glial Fibrillary Acidic Protein; HFD—high-fat diet; HG—high glucose; HMGB1—High-Mobility Group Box 1 protein; HO-1—HemeOxygenase-1; I/R –ischemia/reperfusion; Iba-1—ionized calcium-binding adaptor molecule 1; ICAM—InterCellular Adhesion Molecule; IKK –Nuclear Factor kappa B Inhibitor kinase; IL—InterLeukin; IMID—imidacloprid; INF-γ—Interferon gamma; iNOS—inducible Nitric Oxide synthase; ISO—isoproterenol; i.p.—intraperitoneally; IκBα –Nuclear Factor kappa B Inhibitor; JNK—c-Jun N-terminal Kinase; LPS—lipopolysaccharide; MAO—MonoAmine Oxidases; MCP-1—Monocyte Chemoattract Protein-1; MMP—Matrix MetalloProteinase; mPGES-1—microsomal ProstaGlandin E synthase-1; MPO—MyeloPerOxidase; mRNA—messenger ribonucleic acid; MSU—monosodium urate; MyD88—Myeloid Differentiation primary response 88; NALP3—NACHT, LRR and PYD domains-containing protein 3; NF-κB—Nuclear Factor kappa B; nNOS—neuronal Nitric Oxide synthase; NO –nitric oxide; Nrf2—Nuclear factor erythroid 2-Related Factor 2; NRLP3—NLR family pyrin domain-containing 3; oxLDL—oxidized Low-Density Lipoprotein; OX-42—CD11a/b; p50—nuclear factor NF-kappa-B p50 subunit; p65—nuclear factor NF-kappa-B p65 subunit; PAF—Platelet-Activating Factor; PGE 2—ProstaGlandin E 2; PGN—Peptidoglycan; p.o.—per os (orally); PO –Potassium monooxonate; PTGDS—ProstaGlandin D2 synthase; PTZ—Pentylenetetrazole; RIG-I—Retinoic acid-Inducible Gene I; ROS—reactive oxygen species; SAH –subarachnoid heamorrhage; SIRT1—Sirtuin-1; SOCS3—Suppressor Of Cytokine Signaling 3; STAT3—Signal Transducer and Activator of Transcription 3; STZ—Streptozotocin; T2DM—Type 2 diabetes mellitus; TGF-β—Transforming Growth Factor beta; TIMP—Tissue Inhibitors of MetalloProteinases: TLR—Toll-Like Receptor; TNBS—Trinitrobenzenesulfonic acid; TNF—Tumor Necrosis Factor; TrkB—Tropomyosin receptor Kinase B; UVA—Ultraviolet A; UVB—Ultraviolet B; VCAM—Vascular Cell Adhesion Molecule; VEGF—Vascular Endothelial Growth Factor.
Table 6. Effect of monoterpenes on dysbiosis.
Table 6. Effect of monoterpenes on dysbiosis.
CompoundStudy ModelObserved EffectsRef.
GeraniolDSS-induced colitis mouse model
(30 and 120 mg/Kg, p.o., for 17 days (days 8–24), DSS (1.5%) was given in tap water for 7 days (days 17–23); total time 37 days
Maintained a microbiota composition similar to healthy mice (120 mg/Kg);
Bacteriodetes population;
↑ Lactobacillaceae population higher than control (120 mg/Kg, day 25)
[196]
DSS-induced colitis mouse model
(120 mg/Kg, enema administration on days 19, 21, 23 and 25, DSS (1.5%) was given in tap water for 7 days
(days 17–23); total time 37 days
Maintained a microbiota composition similar to healthy mice; ↑ Bacteriodetes population;
↑ Lactobacillaceae population higher than control (day 25)
[196]
—increase; DSS—dextran sulphate sodium; p.o.—per os (orally).
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

Zuzarte, M.; Sousa, C.; Alves-Silva, J.; Salgueiro, L. Plant Monoterpenes and Essential Oils as Potential Anti-Ageing Agents: Insights from Preclinical Data. Biomedicines 2024, 12, 365. https://doi.org/10.3390/biomedicines12020365

AMA Style

Zuzarte M, Sousa C, Alves-Silva J, Salgueiro L. Plant Monoterpenes and Essential Oils as Potential Anti-Ageing Agents: Insights from Preclinical Data. Biomedicines. 2024; 12(2):365. https://doi.org/10.3390/biomedicines12020365

Chicago/Turabian Style

Zuzarte, Mónica, Cátia Sousa, Jorge Alves-Silva, and Lígia Salgueiro. 2024. "Plant Monoterpenes and Essential Oils as Potential Anti-Ageing Agents: Insights from Preclinical Data" Biomedicines 12, no. 2: 365. https://doi.org/10.3390/biomedicines12020365

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop