Next Article in Journal
Beta-Caryophyllene Enhances the Anti-Tumor Activity of Cisplatin in Lung Cancer Cell Lines through Regulating Cell Cycle and Apoptosis Signaling Molecules
Next Article in Special Issue
The Effect of Combined Treatment of Psilocybin and Eugenol on Lipopolysaccharide-Induced Brain Inflammation in Mice
Previous Article in Journal
A Polyoxometalate-Encapsulated Metal–Organic Framework Nanoplatform for Synergistic Photothermal–Chemotherapy and Anti-Inflammation of Ovarian Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Tackling the Future Pandemics: Broad-Spectrum Antiviral Agents (BSAAs) Based on A-Type Proanthocyanidins

by
Massimo E. Maffei
1,
Cristiano Salata
2 and
Giorgio Gribaudo
3,*
1
Department of Life Sciences and Systems Biology, University of Turin, Via Quarello 15/a, 10135 Turin, Italy
2
Department of Molecular Medicine, University of Padua, 35121 Padua, Italy
3
Department of Life Sciences and Systems Biology, University of Torino, Via Accademia Albertina 13, 10123 Turin, Italy
*
Author to whom correspondence should be addressed.
Molecules 2022, 27(23), 8353; https://doi.org/10.3390/molecules27238353
Submission received: 22 October 2022 / Revised: 19 November 2022 / Accepted: 25 November 2022 / Published: 30 November 2022

Abstract

:
A-type proanthocyanidins (PAC-As) are plant-derived natural polyphenols that occur as oligomers or polymers of flavan-3-ol monomers, such as (+)-catechin and (−)-epicatechin, connected through an unusual double A linkage. PAC-As are present in leaves, seeds, flowers, bark, and fruits of many plants, and are thought to exert protective natural roles against microbial pathogens, insects, and herbivores. Consequently, when tested in isolation, PAC-As have shown several biological effects, through antioxidant, antibacterial, immunomodulatory, and antiviral activities. PAC-As have been observed in fact to inhibit replication of many different human viruses, and both enveloped and non-enveloped DNA and RNA viruses proved sensible to their inhibitory effect. Mechanistic studies revealed that PAC-As cause reduction of infectivity of viral particles they come in contact with, as a result of their propensity to interact with virion surface capsid proteins or envelope glycoproteins essential for viral attachment and entry. As viral infections and new virus outbreaks are a major public health concern, development of effective Broad-Spectrum Antiviral Agents (BSAAs) that can be rapidly deployable even against future emerging viruses is an urgent priority. This review summarizes the antiviral activities and mechanism of action of PAC-As, and their potential to be deployed as BSAAs against present and future viral infections.

Graphical Abstract

1. Introduction

One of the most unsettling lessons that the Coronavirus disease 2019 (COVID-19) pandemic has taught the world is its general unpreparedness for tackling a new respiratory virus pandemic by a therapeutic approach [1]. Notwithstanding that the Severe Acute Respiratory Syndrome Corona Virus 1 (SARS-CoV-1) (2003) [2], and the Middle East Respiratory Syndrome Coronavirus (MERS-CoV) (2012) [3] outbreaks proved the risk of the emergence of new zoonotic coronaviruses, the lack of already available and effective Broad-Spectrum Antiviral Agents (BSAAs), rapidly deployable against the new SARS-CoV-2, made it difficult in the beginning to reduce hospitalizations and deaths, as well as to slow down the spread of COVID-19 [4]. Thus, new BSAAs that can be rapidly deployed against future emerging respiratory viruses in humans, such as coronavirus and influenza virus, are urgently needed. Such BSAAs might allow antiviral treatments to begin immediately after the virus emergence, thus gaining time for the development of the new virus specific vaccines and therapeutics [5].
BSAAs are compounds that inhibit the replication of a wide range of viruses, since different viruses share similar biochemical pathways to synthetize their components and/or exploit the same cellular molecules and pathways to replicate in the host [6]. Given the substantial diversity in viral structures and replication strategies, the development of effective BSAAs has proven to be more difficult than that of the most approved antiviral drugs (i.e., Direct-Acting Antivirals, DAAs), that inhibit only a specific virus-encoded target, such as a polymerase or a protease. Nonetheless, in the last two decades the increasing number of new viral outbreaks in humans has restated the critical need for molecules able to implement the “one drug, multiple virus” paradigm, that is the inhibition of viruses from different families by the same molecule. Thus, effective BSAAs could constitute an essential weapon in the ultimate arsenal of available antiviral options, as they could provide immediate therapeutic intervention against emerging and re-emerging viral threats.
Based on the target, BSAAs can then be categorized into two main types: (1) compounds that target viral structures or enzyme activities, and thus belong to the more general category of DAAs; and (2) compounds that affect host factors or cellular biochemical pathways essential for viral replication, and therefore defined as host-targeted antivirals (HTAs) [7].
The main advantage of BSAAs over the most current approved virus-specific DAAs that are likely inadequate for treating new emerging viruses, consists in their activity not only against viruses belonging to different families, but also towards different genotypes of the same virus species; therefore, they are potentially effective also against viruses not yet emerged in humans. Because of this, BSAAs are suitable as first-line treatments for emerging respiratory virus outbreaks or new sexually transmitted infections, thanks to their rapid repositioning from one pandemic event to the next emerging one. Moreover, host-targeted BSAAs have the inherent edge of a high barrier to the emergence and development of viral drug resistance [6,7]. However, HTAs are burdened with the potential high risk of cellular toxicity, as well as a poor in vitro-to-in vivo translation caused by the systemic compensation of the effects of the blockage of a specific cellular pathway [6]. On the other hand, BSAAs that directly target virus components, such as some DAA nucleoside analogues, although endowed with a lesser potential for host cell toxicity compared to host-targeted BSAA, are prone to the selection of drug-resistant strains [7]. Obviously, the balancing, at least theoretically, of these disadvantages could be accepted in the design of new BSAA-based intervention strategies depending on the threat posed by an emerging viral infection, the characteristics of the causative agent, and the length of treatment, with the final goal of increasing the BSAA’s therapeutic window [6].
Because the development of effective BSAAs remains a challenging task in drug discovery, natural products have been considered as a unique source of chemical complexity and diversity within which antiviral activities can be identified. Indeed, an increasing body of evidence, based on robust molecular, biochemical and pharmacological studies, indicates that a wide-range of natural products derived from plants show inhibitory effects on the replication of many different viruses, thus having the potential to be deployed as BSAAs against both current viruses and new emerging viral threats [8,9,10]. The bioactive components of some of these plant extracts endowed with antiviral activity have been identified as specific polyphenols, flavonoids, glucosides, terpenes, and alkaloids, and the mechanisms of action and molecular targets for some of these molecules have been elucidated [11,12]. Among the large category of natural polyphenols, proanthocyanidins (PACs) characterized by unusual double A linkages of the component catechins monomers (A-type PACs, PAC-As), have been reported to recapitulate the antibacterial and antiviral effects of several plant extracts from which they were isolated and characterized [13,14,15].
This review focuses on A-type PACs as the bioactive chemical components of plant extracts with antiviral activities, with emphasis on their structure, natural origin, mechanism of the antiviral actions including the molecular targets, and their potential to be developed as BSAAs.

2. A-Type Proanthocyanidins’ Chemistry and Where They Can Be Found

It is known that plants biosynthesize bioactive molecules to fulfil their physiological needs, such as defense against herbivores and pathogens, as well as for the interspecific allelopathic competition with other plants [16]. Among these molecules, PACs are interesting bioactive polyphenols that derive from at least two or more 2-phenyl-3,4-dihydro-2H-chromen-3-ol (flavan-3-ol) units which can be condensed through a single (B-type) or a double (A-type) bond [17] (Figure 1). Flavan-3-ols have a saturated A-ring which makes PACs non-planar molecules [18]. PACs are quite complex molecules with a variety of structures because of the stereochemistry of flavanol heterocycle, the type of linkage among the different units, and more importantly, the number and position of the hydroxyl groups linked to the aromatic rings [19]. C–O bonds between the oxygen in position 7 (O7) of one flavan-3-ol unit and the carbon in position 2 (C2) of another unit generate A-type PACs. Because the hydroxyl group linked to the C-ring of each flavan-3-ol can be in either S or in R configuration, different typologies of A-type PACs can be formed.
The biosynthesis, transport and polymerization of PACs, as well as the synthetic efforts made to obtain both naturally occurring A-type PACs and their structurally simplified analogues have been recently reviewed [19].
The distribution of natural PACs in plants is quite large and many red fruits contain different amounts of these bioactive molecules. However, one of the major problems in PAC quantification in plant extracts is the variability of used methods (e.g., the classical gravimetric methods, colorimetric methods based on acid butanol, the ethanol/butanol method, the vanillin assay, and the Brunswick Laboratories 4-dimethylaminocinnamaldehyde or BL-DMAC assay), which represents a bottleneck in the classification of natural sources containing these active polyphenols. From an analytical point of view, HPLC coupled to mass spectrometry, the Matrix-Assisted Laser Desorption/Ionization (MALDI), and ion-mobility mass spectrometry are the best methods for the characterization of both the type of bonds and degree of polymerization [20,21,22,23]. The BL-DMAC method is actually the most widely used method, and although originally developed for the quantification of PACs in cranberry extracts, it proved to be a reliable method also for quantification of PAC in different plant sources [24,25,26,27,28,29,30].
Restricting the natural sources of PACs to those characterized through the BL-DMAC method, Mannino and co-workers [19] performed a meta-analysis that made it possible to identify plant species belonging to 35 different families. The results showed that PACs occur primarily in fruits and seeds, and less in fruit skins (with the exception of peanut skins), leaves and resins. As expected, the meta-analysis showed that the most represented plant family is the Ericaceae, and especially the genus Vaccinium with 10 different species characterized by high contents of PACs. Other high-PAC species such as Styrax ramirezii (fam. Styracaceae) and Carya illinoinensis (fam. Juglandaceae) were also identified. Owing to the biological activities of A-Type PACs, we extended the search for plant sources that mostly contain this type of PACs, and listed their source, biological activity, and chemical properties in Table 1 below.
A-type PACs from different natural sources have been observed to exert different biological effects, such as antiviral (see below), antioxidant, antibacterial, and immunomodulatory activities. Based on the data summarized in Table 1 and Table 2 (see below), we performed a Principal Component Analysis (PCA) that correlates the presence (1) or the absence (0) of different PAC-A degrees of polymerization with three main biological activity (antiviral, antibacterial and antioxidant) by using a single linkage method, with Pearson distances and a varimax rotation. The results of this PCA analysis that are depicted in Figure 2 show that the antiviral activity of PAC-As is correlated primarily to the presence of low-degrees of A-type PAC polymerization (from monomers to tetramers), whereas the antibacterial activity (primarily against bacteria that cause Urinary Tract Infections, UTI) was associated with the presence of dimeric and trimeric PAC-A. Most of the data summarized in Table 1 show that plants that possess polymeric A-type PACs (from pentamers to dodecamers) display antioxidant activity. This observation is confirmed by our PCA analysis (Figure 2).

3. The Broad-Spectrum Antiviral Activity of A-Type PACs

In this section we focus on the activity of PACs as BSAAs, with particular reference to A-type PACs. The studies that have identified A-type PACs as specific components responsible for the antiviral activities of several natural extracts are summarized below, taking into consideration the human viruses for which the inhibitory activity has been characterized. PAC-As with antiviral activity are listed in Table 2 and described in the following paragraphs.

3.1. Herpes Simplex Virus

Herpes Simplex Virus type 1 (HSV-1) and type 2 (HSV-2) cause lifelong infections with periodic reactivations that are highly prevalent worldwide [198,199]. A wide range of diseases result from HSV infections, from the most common cold sores and genital herpes, to recurrent keratitis, and even life-threatening systemic infections and encephalitis [198,199]. Antiviral intervention is therefore needed for the therapy of these diseases. However, the currently available DAAs cannot eliminate an established latent infection, and their prolonged administration may lead to the occurrence of viral resistant strains as well as toxicity [200]. Therefore, the development of new anti-HSV agents that may even prevent the establishment of an HSV infection is a significant medical need [200].
In this regard, many different small molecules from plant extracts, such as polyphenols, terpenes, and flavonoids have been described as exerting an anti-HSV activity in vitro [201,202,203]. Among polyphenols, PACs have been identified as the bioactive anti-HSV agents through chemical and biological characterization of fractions derived from several plant extracts [175,204,205,206,207,208]. In some studies, the antiviral activity of PACs against both HSV-1 and HSV-2, was observed to stem from the ability of PACs to inhibit the virus attachment to the cell surface and the subsequent entry into host cells [209,210]. Especially, Gescher et al. [204] observed that the epicatechin-3-O-gallate-(4→8)-epicatechin-3-O-gallate, a dimeric B-type PAC, isolated from an acetone-water extract obtained from the aerial parts of Rumex acetosa, interacted directly with purified HSV-1 particles and provoked the oligomerization of gD, an essential envelope glycoprotein required for the virus binding to cellular receptors [211]. It was concluded that the R. acetosa-derived PAC-B2 inhibited HSV-1 replication as a result of its ability to bind infectious viral particles and tampering with gD, thus preventing efficient interactions with cell surface receptors [204].
As regards the anti-HSV activity of A-type PACs, in an early study, a series of PAC dimers was isolated from an extract of a byproduct in cocoa production, and then tested for antiviral activity by De Bruyne et al. [124]. Among the different PAC dimers examined, the PAC-A1 or epicatechin-(4β→8, 2β→O→7)-catechin, and the PAC-A2 or epicatechin-(4β→8, 2β→O→7)-epicatechin, were observed to exert the most potent inhibitory activity against in vitro replication of HSV, inasmuch a 4-log reduction in viral titer was measured in the presence of 100 µg/mL of either PAC-A1 or PAC-A2 compared to untreated controls (16). Later, Xu et al., [181] isolated seven A-type PACs from an alcoholic extract of lychee (Litchi chinensis) seeds, that were then examined for antioxidants and antiviral activities. In this study, an anti-HSV-1 activity of a lychee-derived PAC-A2 was determined in Vero cells, with an EC50 of 18.9 µg/mL, and a Specificity Index (SI) of 3.0 [181].
In a subsequent study, oligomeric A-type PACs fractionated from an extract of Chamaecrista nictitans were observed to be related to the overall anti-HSV activity of the extract [175].
More recently, we examined the suitability of a cranberry extract as a direct-acting anti-HSV agent [188]. Analysis of the anti-HSV activity of purified fractions revealed that the ability of the whole cranberry extract to hinder HSV replication was due to its high content of type-A PACs. In fact, only the fraction that contained PAC-A dimers and small amounts of trimers exerted an antiviral activity against HSV-1 and HSV-2 replication in Vero cells, with EC50 of 19.2 and 6.8 µg/mL and SI of 9.5 and 27.6, respectively [188]. Then, mechanistic investigations highlighted that the whole extract or its PACs-A-containing fraction interacted with the envelope glycoproteins gD and gB, the fusion protein of the HSV machinery for entry that carries out membrane fusion [211], thus causing a loss of infectivity of HSV particles [188].
It is therefore possible to recapitulate a common mechanism of action of the anti-HSV activity of both PAC-A and -B [188,209] that could depend from their ability to interact with viral envelope glycoproteins. These interactions, in turn, may affect the functions of those glycoproteins required for HSV attachment and entry, such as gD and gB, thus preventing these initial phases of the HSV replication cycle.

3.2. Human Immunodeficiency Virus (HIV)

Acquired Immunodeficiency Syndrome (AIDS), caused by the Human Immunodeficiency Virus 1 (HIV-1), is an immunosuppressive disease that creates susceptibility to lethal opportunistic infections and malignancies [212]. Although many drugs have been approved and increase the quality of the life of infected people, the high costs and the life-long treatments makes therapy a hard goal in low-income countries. In addition, viral drug resistance prompts researchers to develop new antiviral agents. In this context, several Authors investigated plant extracts to identify anti-HIV-1 activities [213]. Among bioactive compounds able to interfere with HIV-1 infection, PAC-As have been identified as candidates for new antivirals development. In 1999, De Bruyne and co-workers [124] evaluated the biological effects and antiviral activity of PAC-As and related polyphenols. They reported that PAC-A1 and PAC-A2 were the most potent antiviral compounds, reducing the HIV-1 cytopathic effects (CPE) in infected cells with EC50 of 14 and 5.8 µg/mL and SI of 10 and 24, respectively [124]. The anti-HIV-1 mechanism of PAC-As was then elucidated by Fink and coworkers [176]. They observed that elderberry and cinnamon extracts incubated with the virus during the infection step significantly reduced the number of foci of infected cells, with an EC50 from 0.5 to 201 µg/mL for four different HIV-1 types. A direct binding assay coupled with a mass spectrometry approach then showed that PAC-As interacted with viral particles, thereby reducing the virus infectivity. The interaction of PAC-As with HIV-1 particles followed a stoichiometric pattern, thus suggesting HIV-1 envelope glycoproteins as the specific viral target [176]. Furthermore, PAC-As also showed a synergistic effect with the antiretroviral drug enfuvirtide, a drug interacting with the envelope gp41 subunit that blocks the fusion of the HIV-1 to target cells. Indeed, the PAC-As-mediated antiviral activity, being not competitive with enfuvirtide, was suggested as being most likely to target the gp120 subunit [176].
More recently, the anti-HIV-1 activity of the cinnamon-derived compound IND02, that contains A-type PAC trimers and pentamers, was reported [177]. Using surface plasmon resonance, the authors showed that IND02 and IND02-trimer bind to gp120 of HIV-1 types that use CXCR4 (X4, lympho-tropic strain) or CCR5 (R5, macrophage-tropic strain) as co-receptors [177]. Because HIV-1 infection requires multiple interactions of the gp120 with host molecules, such as heparansulfate (HS), the viral receptor CD4, and the R5/X4 co-receptors, the potential of IND02 to interfere with different interaction stages of HIV-1 attachment and entry was investigated. IND02 and IND02 trimer were observed to inhibit the gp120-HS binding in a concentration-dependent manner, while only IND02 affected the gp120-CD4 interaction, as well as the binding of gp120 of R5- and X4-tropic viruses. These results suggested that IND02 could interact with the gp120’s CD4 binding domain of both R5 and X4-tropic viruses, probably to the protein domains involved in interactions with co-receptors. Finally, the antiviral activity of IND02 was confirmed in a biological assay by its addition during the infection step of activated human peripheral blood mononuclear cells (PBMCs) with a panel of clinically relevant primary strains, for which low micromolar EC50 values were observed [177].
Of note, Suedee and colleagues reported a new anti-HIV-1 mechanism PAC-As. With the aim to investigate the anti-integrase (IN) activity of some Thai medicinal plant extracts, they discovered that PAC-A2 derived from a leaf extract of Pometia pinnata inhibited the HIV-1 enzyme with an IC50 value of 30.1 µM. However, this result was obtained from an in vitro enzymatic assay, and no evidence of this PAC-A2 activity in the context of HIV-1 infection was reported [184]. Moreover, Tietjen et al. [180] identified the ixoratannin A-2 as HIV-1 inhibitor with an EC50 value of 35 µM. Ixoratannin A-2 is a doubly linked A-type PAC trimer isolated from the Ixora coccinea shrub collected in western Nigeria, and it was suggested that ixoratannin A-2 might inhibit the ion channel activity of the viral protein Vpu [180]. More recently, and relevant to this hypothesis, a computational study indicated that ixoratannin A-2 might interact with several human and viral proteins, included Vpu [214], thus supporting Tietjen et al.’s hypothesis [180].
Additional studies reported that plant-derived procyanidins other than PAC-As can affect HIV-1 infection. For example, Nair and co-workers [197], reported that grape seed extract-derived PACs inhibited HIV-1 infection by downregulating the co-receptors on the surface of PBMCs [197]. In addition, Feng et al. [182] reported that a procyanidin-rich extract from French maritime pine not only affected HIV-1 virus entry but also its genome replication. Because the prominent biochemical alteration induced in target cells by the French maritime pine consists of an overexpression of the Mn-superoxide dismutase, an intracellular antioxidant protein, its involvement was suggested in the overall anti-HIV-1 activity [182]. Thus, the modulation of some stress-induced cellular pathways by PACs may represent an additional strategy to counteract HIV-1 infections.

3.3. Chronic Hepatitis Viruses

Hepatitis B virus (HBV) and Hepatitis C virus (HCV) are a major cause of liver disease worldwide. HBV is an enveloped double stranded DNA virus (Hepadnaviridae) while HCV is a enveloped positive-strand RNA virus (Flaviviridae), both characterized by hepatic tropism. Transmission happens through intra-family contacts among infants, by sexual or parenteral contact or by the vertical route. For both viruses, after a possible acute phase, viral infection may progress in chronicity. During chronic infection, viral cytopathic effects combined with the cell damage due to the immune response may promote liver cirrhosis and hepatocellular carcinoma [215]. Although effective therapies for the treatment of HBV and HCV infection are available, with a clear improvement of patient treatments and the cure of the infection, at least for HCV, the high costs of the therapy and the risk of drug failure still prompt to the search and development of new drugs.
Many efforts have been addressed to the identification of natural products as cheaper and more accessible sources of new anti-HBV agents [216]. With the aim of discovering potential anti-HBV molecules, Tsukuda and co-workers [217] identified PACs as HBV inhibitors. PACs inhibited HBV infection both in cell lines and in primary human hepatocytes by blocking viral particles’ attachment to target cells (EC50 of ~8 μM) without any effect on viral genome replication and cell viability. Using biochemical assays, it was observed that PACs interacted with the preS1 region of the viral glycoprotein. In addition, PACs showed an anti-HBV effect against multiple viral genotypes and one viral isolate resistant to the approved antiviral drug entecavir. In contrast to other known molecules that interfere with the HBV life cycle, the antiviral activity of PACs directly targets the viral particle, thus acting as a virucidal agent.
Concerning HCV, Takeshika et al. [196,218] reported that purified PACs (PAC-B primarily) from blueberry leaves inhibited HCV RNA replication (EC50 0.087 µg/mL, SI 212). This antiviral activity was evaluated using an HCV subgenomic expression system, while the adhesion/internalization stages of viral particles were not investigated. However, it was observed that blueberry leaf-derived PACs interacted with the heterogeneous nuclear ribonucleoprotein A2/B1 that is indispensable for HCV subgenome expression. Moreover, the anti-HCV activity was found dependent on the polymerization level of PACs, reaching the maximum efficacy with a polymerization degree between 8 and 9 [196,218]. Similarly, Li and coworkers [219] reported that PAC-B1 purified from a cinnamon bark extract inhibited HCV RNA synthesis in a concentration-dependent manner in Huh-7 cells, but it did not interfere with viral entry or receptor expression [219]. As for HIV-1, a French maritime pine extract was reported to inhibit HCV. Since oxidative stress has been identified as a key mechanism of HCV-induced pathogenesis, Ezzikouri et al. [183] evaluated the antiviral properties of a French maritime pine extract in both in vitro and in vivo models. Using HCV replicon cell lines, the authors reported both the inhibition of the HCV replication (EC50 ~ 40 μg/mL) and the reduction of ROS [183]. In addition, treatment of infected chimeric mice with the same extract suppressed HCV replication and showed a synergistic effect with interferon-alpha [183].
Regarding PAC-A, the cinnamon-derived compound IND02 was tested in Huh7.5.1 cells and primary human hepatocytes (PHH) by using HCV and HCV pseudoparticles [178]. IND02, added to target cells for one hour before viral infection, showed a concentration-dependent inhibitory effect against both the wild type virus and a difficult-to-treat HCV strain, characterized by enhanced cell entry efficiency and poor neutralization by neutralizing antibodies [178]. To shed light on the stage of the HCV replication cycle affected by IND02, the authors used HCV pseudoparticles and a subgenomic replicon system to investigate IND02 activity on virus entry and replication, respectively. The results showed that IND02 markedly inhibited the first stage of infection in a way that overlapped with the inhibitory activity of the anti-CD81 antibody that targets the HCV cell entry factor CD81. Then, a kinetics experiment showed that IND02 inhibited HCV infection when added after HCV attachment to target cells, thus suggesting an interference with the internalization of adsorbed virus particles or with the membrane fusion step [178].
Finally, the anti-HIV-1 ixoratannin A-2, a PAC-A trimer from the Ixora coccinea [180], was observed to also inhibit HCV replication in Huh-7 cells with an EC50 of 23.0 μM.
Interestingly, PAC-A and PAC-B seem to target different stages of the HCV replication cycle by acting mainly at the level of viral entry or viral RNA replication, respectively.

3.4. Enteric Viruses

Enteric viruses are a major cause of morbidity and mortality, especially among children in developing countries [220]. Different families of human viruses include agents that target the gastrointestinal tract to cause gastroenteritis, diarrhea, and hepatitis [220]. Members of the Picornaviridae (e.g., enterovirus, hepatitis A virus, Aichi virus), Reoviridae (e.g., rotavirus), Caliciviridae (e.g., norovirus), Astroviridae (e.g., astrovirus), Hepeviridae (e.g., hepatitis E virus) and Adenoviridae (e.g., adenovirus 40 and 41) are in fact major enteric viral pathogens [220,221,222]. These viruses represent a major public health concern worldwide, as they are transmitted through contaminated water or food, shed in high amounts within feces, and remain stable for a long time in the environment [220,221,222].
Accordingly, natural extracts of fruits, such as grapes and berries, have been tested extensively against enteric viruses to identify antiviral activities that may be exploited to develop new preventive or therapeutic agents, and thus to alleviate the burden of foodborne gastrointestinal viral diseases [223]. However, only for a few enteric viruses A-type PACs were observed to reproduce the antiviral activity of fruit extracts in which they have been characterized [191,224].
Rotavirus is a genus of non-enveloped, segmented double-stranded RNA viruses of the Reoviridae family. They are the major cause of acute gastroenteritis (AGE) in infants and young children worldwide, and the leading cause of viral diarrheal mortality with about 200,000 children under the age of 5 each year [225,226]. Even though implementation of rotavirus vaccination, as part of the routine childhood immunization program, proved to be effective in reducing AGE in countries where vaccines are used routinely, millions of children in several high-burden countries still lack access to rotavirus vaccine. Therefore, in the absence of effective control measures or treatment strategies, food extracts and juices endowed with anti-rotavirus activity may be of interest to control the infection and spread of AGE in those countries [223].
In this regard, in an early study, a cranberry juice was investigated for antiviral activity against the simian rotavirus SA-11 and found to protect monkey epithelial MA-104 cells from lytic infection. This antiviral effect was associated with the juice-mediated inhibition of the SA-11 hemagglutination activity, thus suggesting an interference of the juice’s components with adsorption of the rhesus rotavirus to the surface of host cells [192]. Subsequently, the same authors observed that dimeric and polymeric A-type PACs isolated from the cranberry extract indeed determined the loss of SA-11 viral capsid integrity in cell-free suspension, as measured by quantitative antigen capture assay of the virion VP6 protein [193]. Ultrastructural studies by transmission electron microscopy (TEM) then allowed visualization of a direct interaction of the A-type PACs with SA11 viral particles that were observed to be aggregated by the addition of PAC-As. It was therefore suggested that A-type PACs, by binding to and damaging rotavirus capsid proteins, affected the virus’ ability to attach to the host epithelial cell receptors, and in doing so they determined a reduction of viral infectivity [193].
Human noroviruses (HuNoVs) are non-enveloped single-stranded RNA positive viruses belonging to the Caliciviridae family [227]. HuNovs spread through the fecal-oral route and are the leading causative agent of AGE worldwide, with about 700 million cases and 200,000 deaths per year, and the second leading cause of AGE in children after rotavirus [228,229]. HuNoV infections therefore represent a major public health concern with considerable societal and economic outcomes. At present, however, no vaccines or antiviral agents have been licensed for prevention or treatment of HuNoV infections [230].
As for rotavirus, fruit extracts from different plants, such as cranberry, blueberry, pomegranate, and grape have been tested for anti-HuNoV activity. However, given the lack of a robust and reproducible cell system for in vitro HuNoV cultivation, surrogate caliciviruses, such as the feline calicivirus-F9 (FCV-F9) and the murine norovirus-1 (MNV-1) have been used extensively for investigating NoVs replication and pathogenesis, as well as in antiviral assays [231]. Using these animal caliciviruses, cranberry, blueberry and raspberry juices were observed to reduce infectivity of both FCV-F9 and MNV-1 as tested in virucidal assays [189,190,195,232,233]. As reported above, cranberries mainly contain A-type PAC, while blueberries contain mostly B-type PACs [145]; therefore, the two PAC types purified from the corresponding fruits were tested for anti-calicivirus activity, and found to reproduce the inhibitory activity of the corresponding juice, thus indicating that PACs characterized by both A-type and B-type linkages exerted antiviral activity against human enteric viral surrogates [189,190,233]. Especially, TEM analysis on FCV-9 particles exposed to PAC-As revealed major morphological alterations of capsid structure, thus suggesting the ability of cranberry’s type-A PACs to bind to the capsid proteins and altering virion structure in a manner such that the viral infectivity was compromised [190].
Taken together, the available data on the mechanism of action of A-type PACs against enteric viruses confirm the ability of these polyphenols to interact with proteins of the viral surface, thus causing alterations that, in turn, affect severely the virus’ ability to attach and/or enter into target cells. It is therefore tempting to envisage that PAC-As could potentially be exploited for the treatment and/or prevention of foodborne viral diseases.

3.5. Respiratory Viruses

The ongoing COVID-19 pandemic is proving that respiratory viral infections are a leading cause of morbidity and mortality worldwide, and a major societal and healthcare problem [234,235]. In fact, respiratory viruses replicate within the respiratory apparatus causing a broad range of respiratory tract infection (RTI) outcomes, ranging from asymptomatic to acute life-threatening diseases. These viruses spread through the respiratory secretions from an infected individual with three different mechanisms: direct/indirect contact, droplet spray, or aerosol (airborne transmission) [236]. RNA viruses are the predominant cause of RTIs in humans and include: influenza viruses (IV), parainfluenza viruses (PIV), metapneumoviruses (MPV), respiratory syncytial viruses (RSV), human rhinoviruses (hRV), enteroviruses, and human coronaviruses (hCoV). Among DNA viruses, adenoviruses (AdV), human bocavirus (hBoV), and reactivating herpesviruses in immunosuppressed individuals, can cause RTIs [234,235,236].
Although inhibitory activities against several of the above respiratory viruses have been described for many medicinal plant-derived extracts [237,238,239], PACs have been identified as the bioactive antiviral agents only in a few studies in which their inhibitory activity has been characterized against IVs and hCoVs.
Influenza remains a major public health challenge and, every year worldwide, IVs cause around 1 billion infections, 3–5 million of severe RTIs, and 290,000–650,000 respiratory deaths [240,241,242]. Even though seasonal vaccines represent the most effective measure for prevention and control of IV infections, antiviral agents are beneficial to reduce the burden of complications and case-fatality rates. However, the limited arsenal of anti-influenza drugs brings about challenges in the therapeutic management of influenza [241]. Therefore, new anti-influenza agents, effective against different IVs resulting from antigen variation, are urgently required and therefore intensely investigated.
To meet this need, PAC-enriched extracts derived from fruits and herbs have been examined in recent years as direct-acting anti-IV compounds [173,174,186,187,243,244]. Accordingly, oligomeric PAC-A and PAC-B were identified as the main antiviral principle of plant extracts. In a study, the dimeric proanthocyanidin epicatechin-3-O-gallate-(4b→8)-epicatechin-3′-O-gallate (procyanidin B2-di-gallate) was identified as the primary antiviral compound of an extract of garden sorrel (Rumex acetosa) able to inhibit influenza A viruses (IAV) H1N1, both laboratory strains and clinical isolates. Procyanidin B2-di-gallate was then proved to physically interact with the envelope hemagglutinin (HA) glycoprotein as alterations of electrophoretic mobility and immunoreactivity were observed [243]. The envelope of IAV contains two major glycoproteins, IAV hemagglutinin (HA) and neuraminidase (NA), that are essential for efficient infection and viral release from host cells. It was therefore suggested that PAC-B2 may interfere with the receptor binding pocket of HA and consequently affect the IV attachment to host cells. Specific penetration assays indeed confirmed that the PAC-B2 interfered with IAV entry [243]. In silico docking studies then indicated that procyanidin B2-di-gallate was predicted to interact with the receptor binding site of HA. In subsequent studies, PACs isolated from an extract of Alpinia zerumbet, an aromatic and medicinal plant, were observed to reduce the infectivity of the IAV H1N1 laboratory strain PR/8/34 in virucidal assays, thus indicating a direct interaction with viral particles [173]. The composition of A. zerumbet-derived PAC (AzPAC) was then determined and PAC-B2 and -B5 were identified as its major antiviral components. Interestingly, AzPAC was observed in quenching assays to strongly interact with recombinant HA and NA, and to affect the secondary structure of these viral glycoproteins in circular dichroism experiments [235]. It was therefore suggested that the impairment of IV replication caused by AzPAC was due to its direct interaction with IV envelope proteins in a manner that affected their function, thus preventing the attachment phase of the IV infection [174].
Regarding the anti-IV activity of A-type PACs, we have observed that a cranberry extract containing a high content of A-type dimers and trimers, potently inhibited the in vitro replication of both IAV and influenza B virus (IBV) [186]. Mechanistic studies revealed that this cranberry extract blocked the attachment and entry phases of IAV and IBV into target cells and exerted a virucidal activity against both IVs. These biological effects resulted from the ability of the extract to interact with the HA1 ectodomain of HA, as demonstrated by alteration of recombinant HA1 electrophoretic mobility with the occurrence of high molecular weight aggregates. Then, a detailed in silico docking simulation analysis indicated that among the different components of the chemical profile of the cranberry extract, PAC-A2 exhibited the best docking propensity to bind the HA protein with an affinity below 10 nM [186]. Subsequent docking simulation tests predicted the ability of PAC-A2 to bind first within the internal grooves of the HA structure by forming hydrogen bonds with phenylalanine and tryptophan residues, and then to other residues on the HA surface. These in silico predictions were then verified by fluorescence spectroscopy experiments that confirmed a direct interaction between the recombinant HA1 protein and purified PAC-A2. Importantly, purified AC-A2 was observed to potently inhibit both IAV and IVB replication with at about 5-log of reduction in viral titers, and to cause a complete loss of infectivity of IV particles in virucidal assays [186]. These antiviral assays therefore confirmed PAC-A2 as the major active anti-IV component of the cranberry extract. We concluded that the interactions of PAC-A2 with HA and the subsequent alterations in the viral protein function, determined the loss of infectivity of IV particles, thus preventing infection [186].
The disastrous consequence of COVID-19 indeed confirm that emerging coronaviruses are an indisputable major health threat, as proved by more than 600 million cases of COVID-19, including 6.5 million deaths worldwide [245]. In the last two decades, in fact, in addition to the endemic hCoVs (OC43, HKU1, 229E, and NL63), three highly-pathogenic hCoV, namely the severe acute respiratory syndrome coronavirus (SARS-CoV) in 2003, the Middle East respiratory coronavirus (MERS-CoV) in 2012, and SARS-CoV-2 in 2019 emerged as a result of zoonotic outbreaks [246]. These facts proved the urgent need for BSAAs that could be deployed against future hCoVs that could emerge in the future. For this purpose, plant extracts can be evaluated as sources of new anti-hCoV agents.
In an early study, Zhuang et al. [179] observed that a fraction of a cinnamon bark extract reduced the in vitro SARS-CoV replication so that the virus was exposed to the extract before cell infection. Among the chemicals purified from this fraction, PAC-A2 exerted the most potent inhibitory activity on SARS-CoV replication; thus, a virucidal effect was hypothesized as the event responsible for inhibition of infection [179].
With regard to SARS-CoV-2, a PAC-A-rich fraction from the leaves of rabbit-eye blueberry (Vaccinium virgatum Aiton) has been reported recently by Sugamoto et al. [194] to potently inhibit SARS-CoV-2 replication in Vero E6 cells (EC50 1 µg/mL). For SARS-CoV-2, the maximum inhibitory effect was measured when the virus was treated with the PAC-A-rich fraction prior to infection, thus indicating that it could be administered as preventative treatment [194]. Interestingly, the PAC-A-rich fraction was observed to inhibit also the enzymatic activity of both the angiotensin-converting enzyme 2 (ACE2) receptor, which is the essential cell receptor for SARS-CoV-2, and the viral main protease chymotrypsin-like cysteine protease (3CLpro or Mpro), which is fundamental for processing viral polyproteins [246]. These findings suggest different inhibitory effects of the PAC-A-rich fraction against SARS-CoV-2. Indeed, the interference of PAC-As with viral particles and the cell surface receptor could result in the prevention of infection, while their direct-acting activity against Mpro may contribute to the overall antiviral activity after infection [194].
In addition to the thoroughly studied IV and hCoVs, the antiviral activity of PAC-As, in particular of the PAC-A2 dimer, was also observed for respiratory viruses of veterinary interest. In a first study, PAC-A2 purified from the bark of Aesculus hippocastanum was found to inhibit the in vitro replication of the Canine distemper virus (CDV), a Morbillivirus of the family of Paramyxoviridae that affects domestic and wild canines and other carnivores, and causes respiratory and systemic infections [247]. Time-of-addition experiments indicated the ability of PAC-A2 to exert its inhibitory activity during both early and late phases of the CDV replication cycle [248]. In a following investigation, lychee seeds-derived PAC-A2 was observed to exert a potent antiviral activity against the Porcine reproductive and respiratory syndrome virus (PRRSV) in alveolar macrophages, that represent the primary in vivo target cell type of PRRSV infection [249]. PRRSV is a single-stranded positive-sense RNA virus of the family of Arteriviridae and an endemic swine pathogen that causes pneumonia in piglets and growing pigs, thus determining one of the most economically costly diseases in the pig industry [250]. Although the specific mechanism of action against PRRSV was not detailed, PAC-A2 was monitored to prevent PRRSV replication by affecting both viral entry and progeny virus release. It was therefore suggested that PAC-A2 could be used to develop preventative and/or treatment interventions for PRRSV infections [250].

3.6. Non-Respiratory Emerging and Highly Pathogenic Viruses

Emerging viral infections represent a major concern for public health caused by both respiratory viruses and other zoonotic viral agents, as proved by the number of outbreaks and epidemics/pandemics occurring since the year 2000 [251,252]. Natural products have been tested against a number of such emerging viruses with the aim of developing control strategies; however, very limited information is available regarding the antiviral activity of PACs, and in particular PAC-A, as reported below.
One of the most deadly emerging viral diseases is the Ebola virus disease (EVD), caused by infection with Ebola virus (EBOV), an enveloped single-stranded RNA negative virus belonging to Filoviridae family [253]. EVD is characterized by hemorrhagic fever, shock from fluid loss and multi-organ failure with a high case fatality rate. However, there are no approved small molecules-based drugs for its effective treatment [254]. Several efforts have been advanced for the development of anti-EBOV agents targeting viral entry or viral genome replication [255,256]. The most advanced small molecule to treat EVD is remdesivir, a high cost broad-spectrum RdRp inhibitor available only for intravenous use, and thus difficult to use in the context of the low-income countries affected by EVD. Regarding natural products able to inhibit EBOV infection, a few reports have demonstrated the efficacy of PACs and their monomeric flavan-3-ols. In particular, the flavan-3-ol monomers gallic acid and epigallocatechin-3-gallate (EGCG) were reported to inhibit EBOV entry [257]. Time-of-addition assay in fact revealed that gallic acid likely interfered with the GP-mediated fusion in the late endosomes, while EGCG was found to inhibit the endoplasmic reticulum chaperone HSPA5, a host protein required for Ebola virus replication [257,258].
More recently, the screening of more than 500 extracts of medicinal plants collected in China allowed the identification of an anti-EBOV activity in a Maesa perlarius extract. Dimeric PAC and several flavan-3-ol monomers within this extract were found to be potent EBOV entry inhibitors at low micromolar concentrations. By docking analysis and microscale thermophoresis technology, the authors determined that these compounds exhibited virucidal potency by interacting with EBOV glycoprotein, and the most efficient antiviral compound was the PAC-B2 [259].
Dengue fever is caused by a flavivirus and represents one of the major public health concerns affecting almost 400 million people worldwide; it is endemic in at least 100 countries in the tropics and subtropics [260] The Dengue virus (DENV) is transmitted through the bite of female Aedes aegypti or Aedes albopictus mosquitoes. Human infection can range from asymptomatic cases to a severe disease characterized by severe plasma leakage leading to shock, bleeding or organ impairment [260]. Neither vaccines nor specific antivirals are available. Plant extracts were proposed as source of antivirals to treat Dengue fever [261]. In particular, Kimmel and co-workers [261] evaluated the antiviral effect of oligomeric PACs derived from unripe apple peels (rich in PAC-B) using cultured human PBMC derived from healthy subjects. Addition of purified oligomeric PACs (trimers and tetramers), immediately after the infection, reduced viral titer of 1.5 log. It was also observed that these PACs directly interacted with DENV particles, thus reducing virus infectivity. Finally, the authors reported that unripe apple peels-derived PACs also modulated the innate immune response in infected PBMCs, likely contributing to the overall inhibition of DENV replication in target cells [261].
Mayaro virus (MAYV) is an emerging mosquito-borne alphavirus (Togaviridae) affecting individuals in permanent contact with forested areas in tropical South America. This enveloped virus with single-stranded, positive-sense RNA genome causes nonspecific febrile illness and long-lasting arthritis/arthralgia [262]. MAYV diffusion is increasing and is a potential candidate to cause large-scale epidemics; therefore, the design and development of candidates for anti-MAYN viral drugs are urgently needed [263]. To this end, many strategies have been applied to identify antiviral molecules, including the use of plant extracts [263]. In particular, PACs obtained from methanol extraction of Maytenus imbricata (Celastraceae) roots showed a concentration-dependent virucidal effect on MAYV. This compound acted directly in MAYV particles and not on host cells as their treatment before infection did not show any antiviral effect. Interestingly, experiments with dialyzed virus suggested an irreversible inhibition of viral infectivity upon PACs treatment, thus suggesting a strong interaction between PACs and viral envelope or physical damage of the virion [264].
Crimean-Congo hemorrhagic fever virus (CCHFV) is an enveloped single-stranded negative sense RNA virus with a tri-segmented genome belonging to the Nairoviridae family. CCHFV causes an emerging tick-borne viral disease widely distributed across Africa, Southern Europe, the Middle East and Asia. Human infections can present as a spectrum from the absence of symptoms through mild signs, to severe hemorrhagic illness with a fatality rate up to 30% [265]; there is no FDA-approved vaccine or specific antiviral [266]. In 2018, CCHF was included in the WHO Blueprint list of priority diseases to promote the research for vaccines and drugs [267]. Nevertheless, to date very few papers reporting the discovery of anti-CCHF agents have been published [268]. In this regard, we have recently reported that a cranberry (V. macrocarpon Aiton) extract rich in PAC-As inhibits CCHFV infection [185]. To investigate the antiviral mechanism of this cranberry extract, we used the Hazara virus, a nairovirus model of CCHFV that can be handled in Biosafety Level (BSL)-2 Laboratories, instead of BSL-4 required for CCHFV. Time-of-addition experiments showed that the cranberry extract inhibited viral infection by targeting early stages of the replication cycle. In particular, specific viral attachment assays indicated that the main antiviral mechanism is the inhibition of virus attachment to target cells, thus suggesting interactions between bioactive PAC-As and Hazara virus glycoproteins. This hypothesis was further supported by the observation of a virucidal activity of the extract when incubated with HAZV particles before the infection of cells [185].
Overall, PACs, and in some cases PAC-As, have been reported to inhibit non-respiratory emerging viruses by affecting primarily the early step of viral replication cycles, likely as a consequence of alterations of the functions of viral proteins required for attachment and/or entry into host cells.

4. Biological Activities of PAC-As Other than the Antiviral Effects

In many cases, the antiviral action of PAC-A has been associated also with other important properties of these polyphenols, such as the antioxidant, antibacterial, antidiabetic, antihypoglycemic, cardioprotectant, and immunomodulatory activities. Therefore, the following paragraphs will summarize these properties as a compendium of the biological activity of PAC-A.

4.1. Antioxidant Activity

In general, the antioxidant activity of a PAC-A-containing plant extract increases with increasing degrees of A-type PAC polymerization [269], as we noticed in our PCA analysis (Figure 2). A-type PACs may reduce oxidative stress by acting as free radical scavengers, and by affecting signaling pathways associated with cellular oxidative stress homeostasis [18]. In the Malvaceae family, a radical-scavenging effect was shown in Adansonia digitata pericarp (fruit wall) fruits, which contained an A-type PAC trimer [31], whereas in cocoa (Theobroma cacao) epicatechin-containing dimers showed a strong antioxidant power [124]. In the Sapindaceae family, Aesculus turbinata polyphenol polymers with doubly linked A-type interflavans linkages exhibit potent antioxidant activities [32], while in Litchi chinensis A-type dimers and trimers qualify the fruit stones and the pericarp of this plant as a raw material for polyphenol extracts exerting significant antioxidant properties [78,79,80,81]. In the same family, Dimocarpus longan contains PAC trimers-octamers that show promising antioxidant activities which could be applied as potential functional food components [62]. In the Fabaceae family, peanut (Arachis hypogaea) skin A-type PACs were effective against H2O2-induced oxidative stress damage in prostate cancer DU145 cells [35], and thus they have been proposed as an inexpensive source of antioxidants for use as functional ingredients in foods or dietary supplements [42]. In the same family, Spatholobus suberectus fractions enriched in PAC monomers and oligomers exerted antioxidant activity in MCF-7 breast cancer cells [120]. In the Rosaceae, apple (Malus domestica) extracts showed a high antioxidant potential using 2,2-diphenyl-1-picrylhidrazyl (DPPH) and oxygen radical absorbance capacity (ORAC) methods, whereas in plum (Prunus domestica) the antioxidant activity was even higher [86]. In the Ericaceae, cranberry (Vaccinium macrocarpon) radical scavenging and antioxidant activities were attributable to their composition of PACs [151], while in Gaultheria procumbens the leaf antioxidant activity was found to change according to the harvesting season [66,67]. Finally, in grapevine (Vitis vinifera, Vitaceae) glial cultures pretreated with grape seed-derived type-A PACs showed improved viability after H2O2-induced oxidative stress [160].

4.2. Antibacterial Activity

Cranberry (Vaccinium macrocarpon, Ericaceae)-derived PACs are unique in their structure with a higher percentage of A-type bonds, compared with PACs from other commonly consumed fruits [270]. It is well known that cranberry extracts and juices have an anti-bacterial effect and are thus traditionally used to treat cystitis and UTIs [15,137,138,139,140,141,147], such as those caused by uropathogenic Escherichia coli [271]. In the same plant genus, V. myrtillus-derived A2-type PACs contained in juices were effective against bacterial strains of Asaia lannensis and Asaia bogorensis [117]. The same effects were found also in Ribes nigrum (Grossulariaceae family) [117]. A. hypogaea skin extracts display anti-microbial activity due to its A-type PAC content, able to prevent pathogen infection [36]. A-type PACs of Cinnamomum zeylanicum (Lauraceae family) were effective against uropathogenic E. coli multidrug-resistant strains and showed a marked antibiofilm activity [50]. Adhesion of Streptococcus pyogenes to human airway epithelial (HEp-2) cells was found to be inhibited by A-type PACs contained in Pelargonium sidoides (Geraniaceae family) extracts [97], whereas Pinus pinaster (Pinaceae family) bark extracts containing A-type PAC dimers showed bactericidal actions against Staphylococcus aureus and E. coli [102].

4.3. Antidiabetic and Hypoglycemic Activity

A recent meta-analysis revealed that there is a significant effect of PAC supplementation on blood glucose levels and, once in the liver, PACs oligomers may modulate hepatocyte functions and interfere with glucose uptake and metabolism [19]. In the family Lauraceae, the genus Cinnamomum contains A-type PACs that exerted hypoglycemic effects. In C. cassia, the main A-type PAC oligomers could reverse palmitic acid-induced dysfunction of glucose-stimulated insulin secretion in primary cultured islets, improved the insulin concentration in the blood and pancreas, and (as C. japonica) improved insulin sensitivity in type 2 diabetes mellitus [46,47,48]. A-type PAC oligomers of C. tamala improved the insulin concentration in the blood and pancreas [47], whereas C. zeylanicum A-type PACs potentiated insulin action, and may be beneficial in the control of glucose intolerance and diabetes [56,57]. In the Ericaceae family, the A-type doubly linked PAC trimers of V. corymbosum and V. myrtillus acted as antidiabetic substances [127], whereas in Areca catechu (Arecaceae family) the presence of A-type PACs ameliorates the streptozocin-induced hyperglycemia by regulating gluconeogenesis [43]. Excellent inhibitory effects on α-glucosidase were found in extracts of Pyracantha fortuneana (Rosaceae family), and these effects were due to the alteration of the active site catalytic configuration of the enzyme in such a manner as to reduce substrate binding affinity [110].

4.4. Lipid Lowering Effects and Cardiovascular Protection

PACs can interfere with lipid metabolism affecting intestinal absorption of lipids [272] and liver secretion of chylomicrons and lipoproteins [19]. For instance, PAC-A2 significantly reduces cellular lipid accumulation and restricts ox-LDL-induced cellular oxidative stress and inflammation [273]. Grape seed (V. vinifera, Vitaceae)-derived PACs can regulate lipid metabolism and significantly decreased the expression of pro-inflammatory cytokines, thus exerting hypolipidemic and potential anti-inflammatory effects in the liver [164]. The consumption of grape seed PACs has been related to lower oxidized low-density lipoprotein particles and LDL cholesterol [161,163], to improve dyslipidemia associated with a high-fat diet, mainly by repressing lipogenesis and VLDL assembly in the liver [162]. Furthermore, grape seed PACs exerted a pronounced effect on the cholesterol and triglyceride levels [165] and, by inhibiting oxidation of LDL, showed an antiatherosclerotic activity [166]. On the other hand, A-type PACs of peanut (A. hypogaea) skin extracts exert protection against hepatic steatosis induced in rats fed with a high-fat diet by inhibiting the absorption of dietary lipid and chylomicron secretion by enterocytes [34]. Litchi (L. chinensis, Sapindaceae) pericarp-derived extracts rich in PAC-As have cardioprotection effects on myocardial ischemia injury and lower serum malondialdehyde contents in high-fat/cholesterol-dietary hamsters [74,75]. Mandevilla moricandiana (Apocynaceae family) A-type PAC trimers have been observed to induce a concentration-dependent vasodilation on aortic rings through the NO pathway, with the involvement of histamine H1 and estrogen ER alpha receptors [87]. Finally, A-type PACs of cranberry (V. macrocarpon) have been reported to inhibit platelet aggregation and adhesion, to inhibit enzymes involved in lipid and lipoprotein metabolism, to induce endothelium-dependent vasorelaxation, and to increase reverse cholesterol transport and decrease total and LDL cholesterol [152].

4.5. Immunomodulatory Activity

Proanthocyanidin rich foods can markedly influence the immune responses to enteric infections. Mechanistic studies have demonstrated that dietary PACs exert direct modulatory effects on immune cell signaling, by boosting the recruitment of immune cells and suppressing the amount of pro-inflammatory cytokines. Some anti-inflammatory effects of PAC stem from a direct modulation of mucosal immune cells [274]. The prebiotic effect of PAC has been speculated to be primarily responsible for their anti-inflammatory and immunomodulatory activity [275,276]. Some pathologies, like psoriasis, involve inflammatory mechanisms that interact with immune homeostasis and prevent autoimmune diseases by suppressing immune responses [167]. Grape seed (V. vinifera) extracts containing PACs have been reported to act on the immune system by regulating the differentiation of inflammatory T cells and possess the ability of multidirectional regulation of immunity by maintaining the dynamic balance of immunity in psoriasis [168,277]. Dietary V. vinifera-derived PACs promote the DNA repair-dependent stimulation of the immune system following the functional activation of dendritic cells and effector T cells [169,170,171,172], whereas V. macrocarpon PACs improve immune function and modify cytokine and signal transduction pathways [153]. Grape seed PACs were found to attenuate TNF-alpha and IL-1 beta-induced IL-6 production, and decreased IL-17-stimulated ERK 1/2, p38, and JNK MAPK activities in A549 human pulmonary epithelial cells [278]. Grape seed extracts were also found to inhibit the NF-kappa B pathway in human prostate carcinoma DU145 cells [279]. Finally, transcription of inflammatory factors such as myeloperoxidase, interleukin (IL)-1 beta, IL-6, and tumor necrosis factor alpha (TNF-alpha) was also down-regulated in lung tissue by grape seed PACs [280]. Overall, these data indicate a potential immunomodulatory effect of PACs.

5. Conclusions

The devastating consequence of COVID-19 is indisputable evidence of the need for BSAAs effective also against viruses that may emerge from future zoonoses. Indeed, the availability of an antiviral arsenal that includes such BSAAs would make it possible to immediately protect human populations from an emerging viral disease, while waiting for the development of the new virus specific vaccines and DAAs.
The exploitation of natural products to derive BSAAs can meet this urgent need. However, some critical issues must be faced, such as the production of highly active and standardized extracts, the identification of the bioactive components responsible for the antiviral activity, and the characterization of the mechanism(s) of action, which is often related to a synergistic cooperation among different components. The studies we have reviewed here suggest that A-type PACs can overcome these hurdles, and highlight facts that sustain the feasibility of PAC-As as BSAAs candidates.
Firstly, the main mechanism of the antiviral action of PAC-As appears to be the same for most of the viruses examined regardless of whether they are non-enveloped or enveloped DNA or RNA viruses, thus making PAC-As suitable for interventions against new or hitherto unrecognized viruses. Indeed, when examined in detail, the BSAA activity of PAC-As has proven to originate from the inhibition of the virus attachment to the surface of target cells. In many studies, this anti-adhesive effect of PAC-As has been associated with their ability to interact directly with those virion capsid proteins or glycoproteins that are essential for attachment and entry, thus preventing access to their normal binding partners on target cells. This general mechanism of the antiviral activity of PAC-As could result from the natural propensity of polyphenols to bind and aggregate proteins [281,282]. In this regard, it has been proposed that different types of chemical interactions, such as hydrogen bonding, van der Waals and electrostatic interactions, or even covalent linkages may contribute to the formation of protein-polyphenol complexes [282]. The endurance of electrophoretic mobility alterations induced by PAC-A dimers and trimers on HSV gD and gB, as well as on IV HA1, upon boiling of protein samples in SDS sample buffer, sustains the view that the exposure of purified viral glycoproteins to PAC-As results in the formation of covalent linkages between PAC-As molecules and viral proteins [186,188]. These covalent interactions may result in protein-protein crosslinking, as most PACs have two or more reactive quinone moieties [283]; this would explain the smearing and disappearance of glycoprotein bands that we and others have observed in electrophoretic mobility shift assay experiments with purified viral glycoproteins [186,188,204,215,243].
Taken as a whole, the PAC-As-protein interactions may lead to alterations of viral capsid or envelope protein structures and functions, or to masking/blocking their binding sites to cellular receptors, eventually resulting in the inhibition of binding of the viral particles to cell receptors (Figure 3). Accordingly, this mechanism of action advocates the potential application of PAC-As-containing agents as BSAAs in the treatment or prevention of current viral infections, as well as in the preparedness for future emerging viral threats.
Secondly, given the virucidal activity of PAC-As, it is possible to envisage that PAC-As-containing formulations would allow inactivation of a broad range of infecting viruses and therefore prevention of many viral diseases. For example, considering the significant global incidence, morbidity, and mortality rates of both viral RTIs and sexually transmitted infections (STIs), the development of new, safe, attachment/entry inhibitors based on PAC-A-containing agents could provide a realistic method of antiviral intervention, as well as blocking virus shedding and transmission by close personal contact. Regarding RTIs, local application of formulations rich in PAC-As in the upper respiratory tract, administered as tablets or chewing gums or through inhaling devices, would allow the inactivation of infecting virus and thus prevention of infection [283]. Likewise, PAC-As-containing topical microbicides to be applied directly to the genital tract would prevent the establishment of a viral STI, such as HSV or HIV-1 [284,285]. Furthermore, the topical use of PAC-As-based formulations (as aerosolized suspensions or creams) would overcome limitations that might occur due to unsatisfactory PAC-As levels in the blood following systemic treatments. However, such formulations must satisfy the two fundamental requirements of efficacy and safety. Regarding the toxicity of PAC-As, we have recently observed that when tested for effects on the viability of human cells, different purified PAC-As were found to be safe with noteworthy low cytotoxicity values [286]. Moreover, from this perspective, the widespread use of different formulations of dried cranberry extracts, naturally rich in PAC-As [146], for the prevention of urinary tract infections (UTIs) sustains the high safety profile of PAC-As-containing products to develop broad-spectrum antiviral agents of natural origin suitable to prevent infections [287,288]. Interestingly, the antibacterial activity of PAC-As could be valuable also for the prevention of bacterial superinfections of the respiratory tract that may follow viral RTIs (due to both virus- and immune-mediated damage of the respiratory mucosa).
Thirdly, in many low- and middle-income countries, antiviral drugs are often beyond the reach of the people who need them most or are unavailable. Therefore, for those health systems a reliable, affordable, and high-quality supply of low cost antivirals could be essential to control viral infections [289]. Economic models indicate that in developing countries manufacturers of pharmaceuticals and phytopharmaceuticals should be able to charge substantially lower prices with respect to industrialized countries, without impairing their profits and with no reduction of the therapeutic power. Although in middle-income and developing countries prices are already substantially discounted, compared to developed countries, an economic foundation for fair antiviral drug pricing could be based on widely available BSAAs with low costs of production [290]. Low-cost production PAC-A-based BSAAs could be therefore of particular interest to those low-income countries where viral RTIs or sexually transmitted diseases still have a high incidence. In this perspective, the development of PAC-A-rich phytocomplexes as new BSAAs, could not only be advantageous from an economic point of view compared to more expensive purification procedures or chemical synthesis of specific PAC-A molecules, but could allow better exploitation of the synergistic and holistic effects of different bioactive PACs naturally present in a plant-derived extract [19,27]. Thus, a PAC-A-enhanced phytocomplex could be the most suitable candidate for both preclinical and clinical development of PAC-A-based BSAA, owing to the presence of the most active components that contribute to the overall antiviral activity of the plant extract.
Taken together, these considerations support the suitability of A-type PACs to constitute the antiviral active agent of plant-derived formulations for the development of effective BSAAs, that can be rapidly deployable against current viral infections and future emerging viruses.

Author Contributions

Conceptualization, M.E.M., C.S. and G.G.; validation, M.E.M., C.S. and G.G.; writing—original draft preparation, M.E.M., C.S. and G.G.; writing—review and editing, M.E.M., C.S. and G.G.; supervision, G.G. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Piedmont Region (PAR FSC INFRA-P2 B COVID) to G.G.; NATO SPS Grant. No. G5937 to G.G.; the University of Torino (Ricerca Locale) to M.E.M. and G.G.; and the University of Padova (DOR-PRID) to CS.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. To, K.K.; Sridhar, S.; Chiu, K.H.; Hung, D.L.; Li, X.; Hung, I.F.; Tam, A.R.; Chung, T.W.; Chan, J.F.; Zhang, A.J.; et al. Lessons learned 1 year after SARS-CoV-2 emergence leading to COVID-19 pandemic. Emerg. Microbes Infect. 2021, 10, 507–535. [Google Scholar] [CrossRef] [PubMed]
  2. Drosten, C.; Gunther, S.; Preiser, W.; van der Werf, S.; Brodt, H.R.; Becker, S.; Rabenau, H.; Panning, M.; Kolesnikova, L.; Fouchier, R.A.; et al. Identification of a novel coronavirus in patients with severe acute respiratory syndrome. N. Engl. J. Med. 2003, 348, 1967–1976. [Google Scholar] [CrossRef] [PubMed]
  3. Chafekar, A.; Fielding, B.C. Mers-cov: Understanding the latest human coronavirus threat. Viruses 2018, 10, 93. [Google Scholar] [CrossRef] [PubMed]
  4. Adalja, A.; Inglesby, T. Broad-spectrum antiviral agents: A crucial pandemic tool. Expert Rev. Anti Infect. Ther. 2019, 17, 467–470. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Dolgin, E. Stocking the shelves for the next pandemic. Nature 2021, 592, 340–343. [Google Scholar] [CrossRef]
  6. Geraghty, R.J.; Aliota, M.T.; Bonnac, L.F. Broad-spectrum antiviral strategies and nucleoside analogues. Viruses 2021, 13, 667. [Google Scholar] [CrossRef]
  7. Andersen, P.I.; Ianevski, A.; Lysvand, H.; Vitkauskiene, A.; Oksenych, V.; Bjørås, M.; Telling, K.; Lutsar, I.; Dumpis, U.; Irie, Y.; et al. Discovery and development of safe-in-man broad-spectrum antiviral agents. Int. J. Infect. Dis. 2020, 93, 268–276. [Google Scholar] [CrossRef]
  8. Owen, L.; Laird, K.; Shivkumar, M. Antiviral plant-derived natural products to combat RNA viruses: Targets throughout the viral life cycle. Lett. Appl. Microbiol. 2022, 75, 476–499. [Google Scholar] [CrossRef]
  9. Newman, D.J.; Cragg, G.M. Natural products as sources of new drugs over the 30 years from 1981 to 2010. J. Nat. Prod. 2012, 75, 311–335. [Google Scholar] [CrossRef] [Green Version]
  10. Martinez, J.P.; Sasse, F.; Brönstrup, M.; Diez, J.; Meyerhans, A. Antiviral drug discovery: Broad-spectrum drugs from nature. Nat. Prod. Rep. 2015, 32, 29–48. [Google Scholar] [CrossRef]
  11. Mohan, S.; Elhassan Taha, M.M.; Makeen, H.A.; Alhazmi, H.A.; Al Bratty, M.; Sultana, S.; Ahsan, W.; Najmi, A.; Khalid, A. Bioactive natural antivirals: An updated review of the available plants and isolated molecules. Molecules 2020, 25, 4878. [Google Scholar] [CrossRef] [PubMed]
  12. Musarra-Pizzo, M.; Pennisi, R.; Ben-Amor, I.; Mandalari, G.; Sciortino, M.T. Antiviral activity exerted by natural products against human viruses. Viruses 2021, 13, 828. [Google Scholar] [CrossRef] [PubMed]
  13. Pappas, E.; Schaich, K.M. Phytochemicals of cranberries and cranberry products: Characterization, potential health effects, and processing stability. Crit. Rev. Food Sci. Nutr. 2009, 49, 741–781. [Google Scholar] [CrossRef] [PubMed]
  14. Foo, L.Y.; Lu, Y.; Howell, A.B.; Vorsa, N. The structure of cranberry proanthocyanidins which inhibit adherence of uropathogenic p-fimbriated Escherichia coli in vitro. Phytochemistry 2000, 54, 173–181. [Google Scholar] [CrossRef] [PubMed]
  15. Foo, L.Y.; Lu, Y.R.; Howell, A.B.; Vorsa, N. A-type proanthocyanidin trimers from cranberry that inhibit adherence of uropathogenic p-fimbriated Escherichia coli. J. Nat. Prod. 2000, 63, 1225–1228. [Google Scholar] [CrossRef]
  16. Maffei, M.E. Plant Bioactive Molecules; Cambridge Scholars Publishing: Newcastle upon Tyne, UK, 2018. [Google Scholar]
  17. Alejo-Armijo, A.; Salido, S.; Altarejos, J. Synthesis of a-type proanthocyanidins and their analogues: A comprehensive review. J. Agric. Food Chem. 2020, 68, 8104–8118. [Google Scholar] [CrossRef]
  18. Mannino, G.; Gentile, C.; Ertani, A.; Serio, G.; Bertea, C.M. Anthocyanins: Biosynthesis, distribution, ecological role, and use of biostimulants to increase their content in plant foods—A review. Agriculture 2021, 11, 212. [Google Scholar] [CrossRef]
  19. Mannino, G.; Chinigò, G.; Serio, G.; Genova, T.; Gentile, C.; Munaron, L.; Bertea, C.M. Proanthocyanidins and where to find them: A meta-analytic approach to investigate their chemistry, biosynthesis, distribution, and effect on human health. Antioxidants 2021, 10, 1229. [Google Scholar] [CrossRef]
  20. Feliciano, R.P.; Shea, M.P.; Shanmuganayagam, D.; Krueger, C.G.; Howell, A.B.; Reed, J.D. Comparison of isolated cranberry (Vaccinium macrocarpon ait.) proanthocyanidins to catechin and procyanidins a2 and b2 for use as standards in the 4-(dimethylamino)cinnamaldehyde assay. J. Agric. Food Chem. 2012, 60, 4578–4585. [Google Scholar] [CrossRef]
  21. Rue, E.A.; Glinski, J.A.; Glinski, V.B.; van Breemen, R.B. Ion mobility-mass spectrometry for the separation and analysis of procyanidins. J. Mass Spectrom. 2020, 55, e4377. [Google Scholar] [CrossRef]
  22. Reed, J.D.; Krueger, C.G.; Vestling, M.M. Maldi-tof mass spectrometry of oligomeric food polyphenols. Phytochemistry 2005, 66, 2248–2263. [Google Scholar] [CrossRef] [PubMed]
  23. Krueger, C.G.; Reed, J.D.; Feliciano, R.P.; Howell, A.B. Quantifying and characterizing proanthocyanidins in cranberries in relation to urinary tract health. Anal. Bioanal. Chem. 2013, 405, 4385–4395. [Google Scholar] [CrossRef] [PubMed]
  24. Gentile, C.; Mannino, G.; Palazzolo, E.; Gianguzzi, G.; Perrone, A.; Serio, G.; Farina, V. Pomological, sensorial, nutritional and nutraceutical profile of seven cultivars of cherimoya (Annona cherimola mill). Foods 2021, 10, 35. [Google Scholar] [CrossRef] [PubMed]
  25. Zhu, F. Proanthocyanidins in cereals and pseudocereals. Crit. Rev. Food Sci. Nutr. 2019, 59, 1521–1533. [Google Scholar] [CrossRef]
  26. de Carvalho, M.V.O.; de Oliveira, L.d.L.; Costa, A.M. Effect of training system and climate conditions on phytochemicals of Passiflora setacea, a wild passiflora from Brazilian savannah. Food Chem. 2018, 266, 350–358. [Google Scholar] [CrossRef]
  27. Mannino, G.; Maffei, M.E. Metabolomics-based profiling, antioxidant power, and uropathogenic bacterial anti-adhesion activity of sp4(tm), a formulation with a high content of type-a proanthocyanidins. Antioxidants 2022, 11, 1234. [Google Scholar] [CrossRef]
  28. Cáceres-Mella, A.; Peña-Neira, Á.; Narváez-Bastias, J.; Jara-Campos, C.; López-Solís, R.; Canals, J.M. Comparison of analytical methods for measuring proanthocyanidins in wines and their relationship with perceived astringency. Int. J. Food Sci. Technol. 2013, 48, 2588–2594. [Google Scholar] [CrossRef]
  29. Li, H.-J.; Deinzer, M.L. Tandem mass spectrometry for sequencing proanthocyanidins. Anal. Chem. 2007, 79, 1739–1748. [Google Scholar] [CrossRef]
  30. Krueger, C.G.; Chesmore, N.; Chen, X.; Parker, J.; Khoo, C.; Marais, J.P.J.; Shanmuganayagam, D.; Crump, P.; Reed, J.D. Critical reevaluation of the 4-(dimethylamino)cinnamaldehyde assay: Cranberry proanthocyanidin standard is superior to procyanidin a2 dimer for accurate quantification of proanthocyanidins in cranberry products. J. Funct. Foods 2016, 22, 13–19. [Google Scholar] [CrossRef]
  31. Shahat, A.A.; Ahmed, H.H.; Hassan, R.A.; Hussein, A.A. Antooxidant activity of proanthocyanidins from adansonia digitata fruit. Asian Pac. J. Trop. Med. 2008, 1, 55–59. [Google Scholar]
  32. Ogawa, S.; Kimura, H.; Niimi, A.; Katsube, T.; Jisaka, M.; Yokota, K. Fractionation and structural characterization of polyphenolic antioxidants from seed shells of Japanese horse chestnut (Aesculus turbinata blume). J. Agric. Food Chem. 2008, 56, 12046–12051. [Google Scholar] [CrossRef] [PubMed]
  33. Karonen, M.; Bin Imran, I.; Engstrom, M.T.; Salminen, J.P. Characterization of natural and alkaline-oxidized proanthocyanidins in plant extracts by ultrahigh-resolution uhplc-ms/ms. Molecules 2021, 26, 1873. [Google Scholar] [CrossRef] [PubMed]
  34. Bansode, R.R.; Randolph, P.; Ahmedna, M.; Williams, L.L.; Yu, J.M. Bioavailability and hypolipidemic effects of peanut skin polyphenols. J. Med. Food 2015, 18, 265–272. [Google Scholar] [CrossRef] [PubMed]
  35. Yan, F.F.; Chen, L.; Chen, W.B.; Zhao, L.; Lu, Q.; Liu, R. Protective effect of procyanidin A-type dimers against H2O2-induced oxidative stress in prostate DU145 cells through the MAPKs signaling pathway. Life Sci. 2021, 266, 118908. [Google Scholar] [CrossRef] [PubMed]
  36. Enomoto, H.; Nirasawa, T. Localization of flavan-3-ol species in peanut testa by mass spectrometry imaging. Molecules 2020, 25, 2373. [Google Scholar] [CrossRef] [PubMed]
  37. Rush, M.D.; Rue, E.A.; Wong, A.; Kowalski, P.; Glinsk, J.A.; van Breemen, R.B. Rapid determination of procyanidins using maldi-tof/tof mass spectrometry. J. Agric. Food Chem. 2018, 66, 11355–11361. [Google Scholar] [CrossRef]
  38. Appeldoorn, M.M.; Sanders, M.; Vincken, J.P.; Cheynier, V.; Le Guerneve, C.; Hollman, P.C.H.; Gruppen, H. Efficient isolation of major procyanidin a-type dimers from peanut skins and b-type dimers from grape seeds. Food Chem. 2009, 117, 713–720. [Google Scholar] [CrossRef]
  39. Appeldoorn, M.M.; Vincken, J.P.; Sanders, M.; Hollman, P.C.H.; Gruppen, H. Combined normal-phase and reversed-phase liquid chromatography/esi-ms as a tool to determine the molecular diversity of a-type procyanidins in peanut skins. J. Agric. Food Chem. 2009, 57, 6007–6013. [Google Scholar] [CrossRef]
  40. Yu, J.M.; Ahmedna, M.; Goktepe, P. Peanut skin phenolics: Extraction, identification, antioxidant activity, and potential applications, Symposium on Antioxidant Measurement and Applications. In Proceedings of the 229th ACS National Meeting, San Diego, CA, USA, 13–17 March 2005; Amer Chemical Soc.: San Diego, CA, USA, 2007; pp. 226–241. [Google Scholar]
  41. Dudek, M.K.; Glinski, V.B.; Davey, M.H.; Sliva, D.; Kazmierski, S.; Glinski, J.A. Trimeric and tetrameric a-type procyanidins from peanut skins. J. Nat. Prod. 2017, 80, 415–426. [Google Scholar] [CrossRef]
  42. Yu, J.M.; Ahmedna, M.; Goktepe, I.; Dai, J.A. Peanut skin procyanidins: Composition and antioxidant activities as affected by processing. J. Food Compos. Anal. 2006, 19, 364–371. [Google Scholar] [CrossRef]
  43. Huang, P.-L.; Chi, C.-W.; Liu, T.-Y. Areca nut procyanidins ameliorate streptozocin-induced hyperglycemia by regulating gluconeogenesis. Food Chem. Toxicol. 2013, 55, 137–143. [Google Scholar] [CrossRef]
  44. Ropiak, H.M.; Ramsay, A.; Mueller-Harvey, I. Condensed tannins in extracts from European medicinal plants and herbal products. J. Pharm. Biomed. Anal. 2016, 121, 225–231. [Google Scholar] [CrossRef] [PubMed]
  45. Chen, L.; Yuan, P.L.; Chen, K.X.; Jia, Q.; Li, Y.M. Oxidative conversion of b- to a-type procyanidin trimer: Evidence for quinone methide mechanism. Food Chem. 2014, 154, 315–322. [Google Scholar] [CrossRef]
  46. Wang, T.; Sun, P.; Chen, L.; Huang, Q.; Chen, K.X.; Jia, Q.; Li, Y.M.; Wang, H.Y. Cinnamtannin d-1 protects pancreatic beta-cells from palmitic acid-induced apoptosis by attenuating oxidative stress. J. Agric. Food Chem. 2014, 62, 5038–5045. [Google Scholar] [CrossRef] [PubMed]
  47. Chen, L.; Sun, P.; Wang, T.; Chen, K.X.; Jia, Q.; Wang, H.Y.; Li, Y.M. Diverse mechanisms of antidialbetic effects of the different procyanidin oligomer types of two different cinnamon species on db/db mice. J. Agric. Food Chem. 2012, 60, 9144–9150. [Google Scholar] [CrossRef]
  48. Lu, Z.L.; Jia, Q.; Wang, R.; Wu, X.M.; Wu, Y.C.; Huang, C.G.; Li, Y.M. Hypoglycemic activities of a- and b-type procyanidin oligomer-rich extracts from different cinnamon barks. Phytomedicine 2011, 18, 298–302. [Google Scholar] [CrossRef] [PubMed]
  49. Wang, X.Y.; Zhu, B.R.; Jia, Q.; Li, Y.M.; Wang, T.; Wang, H.Y. Cinnamtannin d1 protects pancreatic beta-cells from glucolipotoxicity-induced apoptosis by enhancement of autophagy in vitro and in vivo. J. Agric. Food Chem. 2020, 68, 12617–12630. [Google Scholar] [CrossRef] [PubMed]
  50. Vasudevan, S.; Selvan, G.T.; Bhaskaran, S.; Hari, N.; Solomon, A.P. Reciprocal cooperation of type a procyanidin and nitrofurantoin against multi-drug resistant (mdr) upec: A ph-dependent study. Front. Cell. Infect. Microbiol. 2020, 10, 421. [Google Scholar] [CrossRef]
  51. Kandhare, A.D.; Aswar, U.M.; Mohan, V.; Thakurdesai, P.A. Ameliorative effects of type-a procyanidins polyphenols from cinnamon bark in compound 48/80-induced mast cell degranulation. Anat. Cell Biol. 2017, 50, 275–283. [Google Scholar] [CrossRef] [Green Version]
  52. Lin, W.L.; Guu, S.Y.; Tsai, C.C.; Prakash, E.; Viswaraman, M.; Chen, H.B.; Chang, C.F. Derivation of cinnamon blocks leukocyte attachment by interacting with sialosides. PLoS ONE 2015, 10, e0130389. [Google Scholar] [CrossRef] [Green Version]
  53. Aswar, U.M.; Kandhare, A.D.; Mohan, V.; Thakurdesai, P.A. Anti-allergic effect of intranasal administration of type-a procyanidin polyphenols based standardized extract of cinnamon bark in ovalbumin sensitized balb/c mice. Phytother. Res. 2015, 29, 423–433. [Google Scholar] [CrossRef] [PubMed]
  54. Panickar, K.S.; Polansky, M.M.; Graves, D.J.; Urban, J.F.; Anderson, R.A. A procyanidin type a trimer from cinnamon extract attenuates glial cell swelling and the reduction in glutamate uptake following ischemia-like injury in vitro. Neuroscience 2012, 202, 87–98. [Google Scholar] [CrossRef] [PubMed]
  55. Lu, J.M.; Zhang, K.Q.; Nam, S.; Anderson, R.A.; Jove, R.; Wen, W. Novel angiogenesis inhibitory activity in cinnamon extract blocks vegfr2 kinase and downstream signaling. Carcinogenesis 2010, 31, 481–488. [Google Scholar] [CrossRef] [PubMed]
  56. Anderson, R.A.; Broadhurst, C.L.; Polansky, M.M.; Schmidt, W.F.; Khan, A.; Flanagan, V.P.; Schoene, N.W.; Graves, D.J. Isolation and characterization of polyphenol type-a polymers from cinnamon with insulin-like biological activity. J. Agric. Food Chem. 2004, 52, 65–70. [Google Scholar] [CrossRef]
  57. Chen, L.; Chen, L.; Wang, T.; Yuan, P.L.; Chen, K.X.; Jia, Q.; Wang, H.Y.; Li, Y.M. Preparation of methylated products of a-type procyanidin trimers in cinnamon bark and their protective effects on pancreatic beta-cell. J. Food Sci. 2016, 81, C1062–C1069. [Google Scholar] [CrossRef]
  58. Xu, Q.; Chen, Z.Y.; Zhu, B.R.; Wang, G.R.; Jia, Q.; Li, Y.M.; Wu, X.J. A-type cinnamon procyanidin oligomers protect against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced neurotoxicity in mice through inhibiting the p38 mitogen-activated protein kinase/p53/bcl-2 associated x protein signaling pathway. J. Nutr. 2020, 150, 1731–1737. [Google Scholar] [CrossRef]
  59. Wong-Paz, J.E.; Guyot, S.; Aguilar-Zarate, P.; Muniz-Marquez, D.B.; Contreras-Esquivel, J.C.; Aguilar, C.N. Structural characterization of native and oxidized procyanidins (condensed tannins) from coffee pulp (Coffea arabica) using phloroglucinolysis and thioglycolysis-hplc-esi-ms. Food Chem. 2021, 340, 127830. [Google Scholar] [CrossRef]
  60. Chai, W.M.; Chen, C.M.; Gao, Y.S.; Feng, H.L.; Ding, Y.M.; Shi, Y.; Zhou, H.T.; Chen, Q.X. Structural analysis of proanthocyanidins isolated from fruit stone of chinese hawthorn with potent antityrosinase and antioxidant activity. J. Agric. Food Chem. 2014, 62, 123–129. [Google Scholar] [CrossRef]
  61. Soong, Y.Y.; Barlow, P.J. Isolation and structure elucidation of phenolic compounds from longan (Dimocarpus longan lour.) seed by high-performance liquid chromatography-electrospray ionization mass spectrometry. J. Chromatogr. A 2005, 1085, 270–277. [Google Scholar] [CrossRef]
  62. Fu, C.L.; Yang, X.N.; Lai, S.J.; Liu, C.; Huang, S.R.; Yang, H.S. Structure, antioxidant and alpha-amylase inhibitory activities of longan pericarp proanthocyanidins. J. Funct. Food. 2015, 14, 23–32. [Google Scholar] [CrossRef]
  63. Lin, L.C.; Kuo, Y.C.; Chou, C.J. Immunomodulatory proanthocyanidins from ecdysanthera utilis. J. Nat. Prod. 2002, 65, 505–508. [Google Scholar] [CrossRef]
  64. Yoshimura, M.; Amakura, Y.; Hyuga, S.; Hyuga, M.; Nakamori, S.; Maruyama, T.; Oshima, N.; Uchiyama, N.; Yang, J.; Oka, H.; et al. Quality evaluation and characterization of fractions with biological activity from ephedra herb extract and ephedrine alkaloids-free ephedra herb extract. Chem. Pharm. Bull. 2020, 68, 140–149. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Orejola, J.; Matsuo, Y.; Saito, Y.; Tanaka, T. Characterization of proanthocyanidin oligomers of ephedra sinica. Molecules 2017, 22, 1308. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Michel, P.; Owczarek, A.; Kosno, M.; Gontarek, D.; Matczak, M.; Olszewska, M.A. Variation in polyphenolic profile and in vitro antioxidant activity of eastern teaberry (Gaultheria procumbens L.) leaves following foliar development. Phytochem. Lett. 2017, 20, 356–364. [Google Scholar] [CrossRef]
  67. Michel, P.; Dobrowolska, A.; Kicel, A.; Owczarek, A.; Bazylko, A.; Granica, S.; Piwowarski, J.P.; Olszewska, M.A. Polyphenolic profile, antioxidant and anti-inflammatory activity of eastern teaberry (Gaultheria procumbens L.) leaf extracts. Molecules 2014, 19, 20498–20520. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Idowu, T.O.; Ogundaini, A.O.; Salau, A.O.; Obuotor, E.M.; Bezabih, M.; Abegaz, B.M. Doubly linked, a-type proanthocyanidin turner and other constituents of ixora coccinea leaves and their antioxidant and antibacterial properties. Phytochemistry 2010, 71, 2092–2098. [Google Scholar] [CrossRef]
  69. Alejo-Armijo, A.; Ortega-Vidal, J.; Salido, S.; Altarejos, J. Recovery and seasonal variation of cinnamtannin b-1 from laurel (Laurus nobilis L.) pruning wood wastes. Chem. Biodivers. 2022, 19, e202100807. [Google Scholar] [CrossRef]
  70. Sui, Y.; Zheng, Y.; Li, X.P.; Li, S.Y.; Xie, B.J.; Sun, Z.D. Characterization and preparation of oligomeric procyanidins from litchi chinensis pericarp. Fitoterapia 2016, 112, 168–174. [Google Scholar] [CrossRef]
  71. Li, W.; Liang, H.; Zhang, M.W.; Zhang, R.F.; Deng, Y.Y.; Wei, Z.C.; Zhang, Y.; Tang, X.J. Phenolic profiles and antioxidant activity of litchi (Litchi chinensis Sonn.) fruit pericarp from different commercially available cultivars. Molecules 2012, 17, 14954–14967. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Li, S.Y.; Xiao, J.; Chen, L.; Hu, C.L.; Chen, P.; Xie, B.J.; Sun, Z.D. Identification of a-series oligomeric procyanidins from pericarp of litchi chinensis by ft-icr-ms and lc-ms. Food Chem. 2012, 135, 31–38. [Google Scholar] [CrossRef]
  73. Su, D.; Luo, M.; Liu, H.; Qi, X.; Zeng, Q.; He, S.; Fen, S.; Zhang, J. The effect of simulated digestion on the composition of phenolic compounds and antioxidant activities in lychee pulp of different cultivars. Int. J. Food Sci. Technol. 2019, 54, 3042–3050. [Google Scholar] [CrossRef]
  74. Chen, Y.F.; Li, H.Q.; Zhang, S.; Yang, C.Y.; Mai, Z.Y.; Hu, X.Y.; Gao, Z.H.; Deng, H. Anti-myocardial ischemia effect and components of litchi pericarp extracts. Phytother. Res. 2017, 31, 1384–1391. [Google Scholar] [CrossRef] [PubMed]
  75. Yang, D.J.; Chang, Y.Y.; Hsu, C.L.; Liu, C.W.; Wang, Y.; Chen, Y.C. Protective effect of a litchi (Litchi chinensis Sonn.)-flower-water-extract on cardiovascular health in a high-fat/cholesterol-dietary hamsters. Food Chem. 2010, 119, 1457–1464. [Google Scholar] [CrossRef]
  76. Li, S.Y.; Liu, Y.; Liu, G.; He, J.R.; Qin, X.G.; Yang, H.C.; Hu, Z.Z.; Lamikanra, O. Effect of the a-type linkage on the pharmacokinetics and intestinal metabolism of litchi pericarp oligomeric procyanidins. J. Agric. Food Chem. 2017, 65, 1893–1899. [Google Scholar] [CrossRef]
  77. Wu, Q.; Li, S.Y.; Xiao, J.; Sui, Y.; Xie, B.J.; Sun, Z.D. Analysis of distribution and pharmacokinetics of litchi pericarp procyanidins in rat plasma and organs by using liquid chromatography-tandem mass spectrometry. Eur. Food Res. Technol. 2017, 243, 167–176. [Google Scholar] [CrossRef]
  78. Lv, Q.; Luo, F.L.; Zhao, X.Y.; Liu, Y.; Hu, G.B.; Sun, C.D.; Li, X.; Chen, K.S. Identification of proanthocyanidins from litchi (Litchi chinensis Sonn.) pulp by lc-esi-q-tof-ms and their antioxidant activity. PLoS ONE 2015, 10, e0120480. [Google Scholar] [CrossRef] [Green Version]
  79. Reichel, M.; Triani, R.; Wellhofer, J.; Sruamsiri, P.; Carle, R.; Neidhart, S. Vital characteristics of litchi (Litchi chinensis Sonn.) pericarp that define postharvest concepts for Thai cultivars. Food Bioprocess Technol. 2013, 6, 1191–1206. [Google Scholar] [CrossRef]
  80. Zhou, H.C.; Lin, Y.M.; Li, Y.Y.; Li, M.; Wei, S.D.; Chai, W.M.; Tam, N.F.Y. Antioxidant properties of polymeric proanthocyanidins from fruit stones and pericarps of litchi chinensis Sonn. Food Res. Int. 2011, 44, 613–620. [Google Scholar] [CrossRef]
  81. Liu, L.; Xie, B.J.; Cao, S.Q.; Yang, E.N.; Xu, X.; Guo, S.S. A-type procyanidins from litchi chinensis pericarp with antioxidant activity. Food Chem. 2007, 105, 1446–1451. [Google Scholar] [CrossRef]
  82. Li, S.Y.; Sui, Y.; Xiao, J.; Wu, Q.; Hu, B.; Xie, B.J.; Sun, Z.D. Absorption and urinary excretion of a-type procyanidin oligomers from litchi chinensis pericarp in rats by selected ion monitoring liquid chromatography-mass spectrometry. Food Chem. 2013, 138, 1536–1542. [Google Scholar] [CrossRef]
  83. Li, S.Y.; Chen, L.; Yang, T.; Wu, Q.; Lv, Z.J.; Xie, B.J.; Sun, Z.D. Increasing antioxidant activity of procyanidin extracts from the pericarp of litchi chinensis processing waste by two probiotic bacteria bioconversions. J. Agric. Food Chem. 2013, 61, 2506–2512. [Google Scholar] [CrossRef] [PubMed]
  84. Engemann, A.; Hubner, F.; Rzeppa, S.; Humpf, H.U. Intestinal metabolism of two a-type procyanidins using the pig cecum model: Detailed structure elucidation of unknown catabolites with fourier transform mass spectrometry (ftms). J. Agric. Food Chem. 2012, 60, 749–757. [Google Scholar] [CrossRef]
  85. Sivakumaran, S.; Rumball, W.; Lane, G.A.; Fraser, K.; Foo, L.Y.; Yu, M.; Meagher, L.P. Variation of proanthocyanidins in lotus species. J. Chem. Ecol. 2006, 32, 1797–1816. [Google Scholar] [CrossRef]
  86. Navarro, M.; Moreira, I.; Arnaez, E.; Quesada, S.; Azofeifa, G.; Vargas, F.; Alvarado, D.; Chen, P. Polyphenolic characterization and antioxidant activity of malus domestica and prunus domestica cultivars from costa rica. Foods 2018, 7, 15. [Google Scholar] [CrossRef] [Green Version]
  87. Ferreira, L.; Leao, V.D.; de Melo, C.M.; Machado, T.D.; Amaral, A.C.F.; da Silva, L.L.; Simas, N.K.; Muzitano, M.F.; Leal, I.C.R.; Raimundo, J.M. Ethyl acetate fraction and isolated phenolics derivatives from mandevilla moricandiana identified by uhplc-dad-esi-msn with pharmacological potential for the improvement of obesity-induced endothelial dysfunction. Pharmaceutics 2021, 13, 1173. [Google Scholar] [CrossRef] [PubMed]
  88. Chen, W.B.; Zhang, L.; Zhao, L.; Yan, F.F.; Zhu, X.L.; Lu, Q.; Liu, R. Metabolomic profiles of a-type procyanidin dimer and trimer with gut microbiota in vitro. J. Funct. Food. 2021, 85, 104637. [Google Scholar] [CrossRef]
  89. Yang, S.Y.; Zhang, Y.Y.; Li, W.; You, B.Y.; Yu, J.W.; Huang, X.X.; Yang, R.L. Gut microbiota composition affects procyanidin a2-attenuated atherosclerosis in apoe(-/-) mice by modulating the bioavailability of its microbial metabolites. J. Agric. Food Chem. 2021, 69, 6989–6999. [Google Scholar] [CrossRef] [PubMed]
  90. Alejo-Armijo, A.; Glibota, N.; Frias, M.P.; Altarejos, J.; Galvez, A.; Salido, S.; Ortega-Morente, E. Synthesis and evaluation of antimicrobial and antibiofilm properties of a-type procyanidin analogues against resistant bacteria in food. J. Agric. Food Chem. 2018, 66, 2151–2158. [Google Scholar] [CrossRef]
  91. Ge, Z.Z.; Dong, X.Q.; Zhu, W.; Zhang, Y.; Li, C.M. Metabolites and changes in antioxidant activity of a-type and b-type proanthocyanidin dimers after incubation with rat intestinal microbiota. J. Agric. Food Chem. 2015, 63, 8991–8998. [Google Scholar] [CrossRef]
  92. Ou, K.Q.; Sarnoski, P.; Schneider, K.R.; Song, K.J.; Khoo, C.; Gu, L.W. Microbial catabolism of procyanidins by human gut microbiota. Mol. Nutr. Food Res. 2014, 58, 2196–2205. [Google Scholar] [CrossRef]
  93. Machado, K.N.; Barbosa, A.D.; de Freitas, A.A.; Alvarenga, L.F.; de Padua, R.M.; Faraco, A.A.G.; Braga, F.C.; Vianna-Soares, C.D.; Castilho, R.O. Tnf-alpha inhibition, antioxidant effects and chemical analysis of extracts and fraction from Brazilian guarana seed powder. Food Chem. 2021, 355, 129563. [Google Scholar] [CrossRef] [PubMed]
  94. da Silva, G.S.; Canuto, K.M.; Ribeiro, P.R.V.; de Brito, E.S.; Nascimento, M.M.; Zocolo, G.J.; Coutinho, J.P.; de Jesus, R.M. Chemical profiling of guarana seeds (Paullinia cupana) from different geographical origins using uplc-qtof-ms combined with chemometrics. Food Res. Int. 2017, 102, 700–709. [Google Scholar] [CrossRef] [PubMed]
  95. Spiegler, V. Anthelmintic a-type procyanidins and further characterization of the phenolic composition of a root extract from Paullinia pinnata. Molecules 2020, 25, 2287. [Google Scholar] [CrossRef] [PubMed]
  96. Spiegler, V.; Liebau, E.; Peppler, C.; Raue, K.; Werne, S.; Strube, C.; Heckendorn, F.; Agyare, C.; Stark, T.; Hofmann, T.; et al. A hydroalcoholic extract from Paullinia pinnata L. Roots exerts anthelmintic activity against free-living and parasitic nematodes. Planta Med. 2016, 82, 1173–1179. [Google Scholar] [CrossRef] [Green Version]
  97. Janecki, A.; Conrad, A.; Engels, I.; Frank, U.; Kolodziej, H. Evaluation of an aqueous-ethanolic extract from pelargonium sidoides (eps((r)) 7630) for its activity against group a-streptococci adhesion to human hep-2 epithelial cells. J. Ethnopharmacol. 2011, 133, 147–152. [Google Scholar] [CrossRef]
  98. Lopez-Cobo, A.; Gomez-Caravaca, A.M.; Pasini, F.; Caboni, M.F.; Segura-Carretero, A.; Fernandez-Gutierrez, A. Hplc-dad-esi-qtof-ms and hplc-fld-ms as valuable tools for the determination of phenolic and other polar compounds in the edible part and by-products of avocado. LWT Food Sci. Technol. 2016, 73, 505–513. [Google Scholar] [CrossRef]
  99. Gu, L.W.; Kelm, M.A.; Hammerstone, J.F.; Beecher, G.; Holden, J.; Haytowitz, D.; Prior, R.L. Screening of foods containing proanthocyanidins and their structural characterization using lc-ms/ms and thiolytic degradation. J. Agric. Food Chem. 2003, 51, 7513–7521. [Google Scholar] [CrossRef]
  100. Sirisena, S.; Zabaras, D.; Ng, K.; Ajlouni, S. Characterization of date (Deglet Nour) seed free and bound polyphenols by high-performance liquid chromatography-mass spectrometry. J. Food Sci. 2017, 82, 333–340. [Google Scholar] [CrossRef]
  101. Zhou, B.; Alania, Y.; Reis, M.C.; McAlpine, J.B.; Bedran-Russo, A.K.; Pauli, G.F.; Chen, S.N. Rare a-type, spiro-type, and highly oligomeric proanthocyanidins from Pinus massoniana. Org. Lett. 2020, 22, 5304–5308. [Google Scholar] [CrossRef]
  102. Ramos, P.A.B.; Pereira, C.; Gomes, A.P.; Neto, R.T.; Almeida, A.; Santos, S.A.O.; Silva, A.M.S.; Silvestre, A.J.D. Chemical characterisation, antioxidant and antibacterial activities of Pinus pinaster ait. And Pinus pinea L. Bark polar extracts: Prospecting forestry by-products as renewable sources of bioactive compounds. Appl. Sci. 2022, 12, 784. [Google Scholar] [CrossRef]
  103. Glavnik, V.; Vovk, I. High performance thin-layer chromatography-mass spectrometry methods on diol stationary phase for the analyses of flavan-3-ols and proanthocyanidins in invasive Japanese knotweed. J. Chromatogr. A 2019, 1598, 196–208. [Google Scholar] [CrossRef] [PubMed]
  104. Li, S.Y.; Yang, Y.J.; Li, J.S.; Zhu, Z.Z.; Lorenzo, J.M.; Barba, F.J. Increasing yield and antioxidative performance of litchi pericarp procyanidins in baked food by ultrasound-assisted extraction coupled with enzymatic treatment. Molecules 2018, 23, 2089. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Nunes, C.; Guyot, S.; Marnet, N.; Barros, A.S.; Saraiva, J.A.; Renard, C.; Coimbra, M.A. Characterization of plum procyanidins by thiolytic depolymerization. J. Agric. Food Chem. 2008, 56, 5188–5196. [Google Scholar] [CrossRef] [Green Version]
  106. Prodanov, M.; Garrido, I.; Vacas, V.; Lebron-Aguilar, R.; Duenas, M.; Gomez-Cordoves, C.; Bartolome, B. Ultrafiltration as alternative purification procedure for the characterization of low and high molecular-mass phenolics from almond skins. Anal. Chim. Acta 2008, 609, 241–251. [Google Scholar] [CrossRef] [PubMed]
  107. Bolling, B.W.; Chen, C.Y.; McKay, D.L.; Blumberg, J.B. Tree nut phytochemicals: Composition, antioxidant capacity, bioactivity, impact factors. A systematic review of almonds, brazils, cashews, hazelnuts, macadamias, pecans, pine nuts, pistachios and walnuts. Nutr. Res. Rev. 2011, 24, 244–275. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Irizar, A.C.; Fernandez, M.F.; Gonzalez, A.G.; Ravelo, A.G. Constituents of prunus-spinosa. J. Nat. Prod. 1992, 55, 450–454. [Google Scholar] [CrossRef]
  109. Pham, H.N.; Michalet, S.; Bodillis, J.; Nguyen, T.D.; Nguyen, T.K.O.; Le, T.P.Q.; Haddad, M.; Nazaret, S.; Dijoux-Franca, M.G. Impact of metal stress on the production of secondary metabolites in pteris Vittata L. and associated rhizosphere bacterial communities. Environ. Sci. Pollut. Res. 2017, 24, 16735–16750. [Google Scholar] [CrossRef]
  110. Wei, M.K.; Chai, W.M.; Yang, Q.; Wang, R.; Peng, Y.Y. Novel insights into the inhibitory effect and mechanism of proanthocyanidins from Pyracantha fortuneana fruit on alpha-glucosidase. J. Food Sci. 2017, 82, 2260–2268. [Google Scholar] [CrossRef]
  111. Jeong, D.E.; Cho, J.Y.; Lee, Y.G.; Jeong, H.Y.; Lee, H.J.; Moon, J.H. Isolation of five proanthocyanidins from pear (Pyrus pyrifolia nakai) fruit peels. Food Sci. Biotechnol. 2017, 26, 1209–1215. [Google Scholar] [CrossRef]
  112. Zhang, L.L.; Wang, Y.M.; Wu, D.M.; Xu, M.; Chen, J.H. Study on the structure of mangrove polyflavonoid tannins with maldi-tof mass spectrometry and NMR. In Proceedings of the 2nd International Conference on Chemical Engineering and Advanced Materials (CEAM 2012), Guangzhou, China, 13–15 July 2012; Trans Tech Publications Ltd.: Stafa-Zurich, Switzerland, 2012; pp. 1988–1993. [Google Scholar]
  113. Kandil, F.E.; Grace, M.H.; Seigler, D.S.; Cheeseman, J.M. Polyphenolics in Rhizophora mangle L. Leaves and their changes during leaf development and senescence. Trees Struct. Funct. 2004, 18, 518–528. [Google Scholar] [CrossRef]
  114. Louis, A.; Petereit, F.; Lechtenberg, M.; Deters, A.; Hensel, A. Phytochemical characterization of rhododendron ferrugineum and in vitro assessment of an aqueous extract on cell toxicity. Planta Med. 2010, 76, 1550–1557. [Google Scholar] [CrossRef] [PubMed]
  115. Way, T.D.; Tsai, S.J.; Wang, C.M.; Jhan, Y.L.; Ho, C.T.; Chou, C.H. Cinnamtannin d1 from rhododendron formosanum induces autophagy via the inhibition of akt/mtor and activation of erk1/2 in non-small-cell lung carcinoma cells. J. Agric. Food Chem. 2015, 63, 10407–10417. [Google Scholar] [CrossRef]
  116. Wang, C.M.; Hsu, Y.M.; Jhan, Y.L.; Tsai, S.J.; Lin, S.X.; Su, C.H.; Chou, C.H. Structure elucidation of procyanidins isolated from rhododendron formosanum and their anti-oxidative and anti-bacterial activities. Molecules 2015, 20, 12787–12803. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Antolak, H.; Czyzowska, A.; Kregiel, D. Black currant (Ribes nigrum L.) and bilberry (Vaccinium myrtillus L.) fruit juices inhibit adhesion of Asaia spp. Biomed Res. Int. 2016, 2016, 3671306. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Carvalho, E.; Franceschi, P.; Feller, A.; Herrera, L.; Palmieri, L.; Arapitsas, P.; Riccadonna, S.; Martens, S. Discovery of a-type procyanidin dimers in yellow raspberries by untargeted metabolomics and correlation based data analysis. Metabolomics 2016, 12, 144. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Spencer, P.; Sivakumaran, S.; Fraser, K.; Foo, L.Y.; Lane, G.A.; Edwards, P.J.B.; Meagher, L.P. Isolation and characterisation of procyanidins from Rumex obtusifolius. Phytochem. Anal. 2007, 18, 193–203. [Google Scholar] [CrossRef]
  120. Li, W.T.; Liu, J.; Guan, R.G.; Chen, J.P.; Yang, D.P.; Zhao, Z.M.; Wang, D.M. Chemical characterization of procyanidins from Spatholobus suberectus and their antioxidative and anticancer activities. J. Funct. Food. 2015, 12, 468–477. [Google Scholar] [CrossRef]
  121. Hurst, W.J.; Stanley, B.; Glinski, J.A.; Davey, M.; Payne, M.J.; Stuart, D.A. Characterization of primary standards for use in the hplc analysis of the procyanidin content of cocoa and chocolate containing products. Molecules 2009, 14, 4136–4146. [Google Scholar] [CrossRef] [Green Version]
  122. De Taeye, C.; Caullet, G.; Evina, V.J.E.; Collin, S. Procyanidin a2 and its degradation products in raw, fermented, and roasted cocoa. J. Agric. Food Chem. 2017, 65, 1715–1723. [Google Scholar] [CrossRef]
  123. Appeldoorn, M.M.; Vincken, J.P.; Gruppen, H.; Hollman, P.C.H. Procyanidin dimers a1, a2, and b2 are absorbed without conjugation or methylation from the small intestine of rats. J. Nutr. 2009, 139, 1469–1473. [Google Scholar] [CrossRef] [Green Version]
  124. De Bruyne, T.; Pieters, L.; Witvrouw, M.; De Clercq, E.; Vanden Berghe, D.; Vlietinck, A.J. Biological evaluation of proanthocyanidin dimers and related polyphenols. J. Nat. Prod. 1999, 62, 954–958. [Google Scholar] [CrossRef] [PubMed]
  125. Matsuo, Y.; Fujita, Y.; Ohnishi, S.; Tanaka, T.; Hirabaru, H.; Kai, T.; Sakaida, H.; Nishizono, S.; Kouno, I. Chemical constituents of the leaves of rabbiteye blueberry (Vaccinium ashei) and characterisation of polymeric proanthocyanidins containing phenylpropanoid units and a-type linkages. Food Chem. 2010, 121, 1073–1079. [Google Scholar] [CrossRef] [Green Version]
  126. Esquivel-Alvarado, D.; Munoz-Arrieta, R.; Alfaro-Viquez, E.; Madrigal-Carballo, S.; Krueger, C.G.; Reed, J.D. Composition of anthocyanins and proanthocyanidins in three tropical vaccinium species from costa rica. J. Agric. Food Chem. 2020, 68, 2872–2879. [Google Scholar] [CrossRef] [PubMed]
  127. Toomik, P.; Pussa, T.; Raal, A. Variability of procyanidin type a- and -b trimers content in aerial parts of some vaccinium species and cultivars. Nat. Prod. Commun. 2014, 9, 815–816. [Google Scholar] [CrossRef] [Green Version]
  128. Sintara, M.; Wang, Y.F.; Li, L.; Liu, H.Y.; Cunningham, D.G.; Prior, R.R.; Chen, P.; Chang, T.; Wu, X.L. Quantification of cranberry proanthocyanidins by normal-phase high-performance liquid chromatography using relative response factors. Phytochem. Anal. 2020, 31, 874–883. [Google Scholar] [CrossRef]
  129. Gardana, C.; Scialpi, A.; Fachechi, C.; Simonetti, P. Identification of markers for the authentication of cranberry extract and cranberry-based food supplements. Heliyon 2020, 6, e03863. [Google Scholar] [CrossRef]
  130. Gao, C.; Cunningham, D.G.; Liu, H.Y.; Khoo, C.; Gu, L.W. Development of a thiolysis hplc method for the analysis of procyanidins in cranberry products. J. Agric. Food Chem. 2018, 66, 2159–2167. [Google Scholar] [CrossRef]
  131. van Dooren, I.; Foubert, K.; Theunis, M.; Naessens, T.; Pieters, L.; Apers, S. Advantages of a validated uplc-ms/ms standard addition method for the quantification of a-type dimeric and trimeric proanthocyanidins in cranberry extracts in comparison with well-known quantification methods. J. Pharm. Biomed. Anal. 2018, 148, 32–41. [Google Scholar] [CrossRef]
  132. Wang, Y.F.; Singh, A.P.; Hurst, W.J.; Glinski, J.A.; Koo, H.; Vorsa, N. Influence of degree-of-polymerization and linkage on the quantification of proanthocyanidins using 4-dimethylaminocinnamaldehyde (dmac) assay. J. Agric. Food Chem. 2016, 64, 2190–2199. [Google Scholar] [CrossRef]
  133. Carpenter, J.L.; Caruso, F.L.; Tata, A.; Vorsa, N.; Neto, C.C. Variation in proanthocyanidin content and composition among commonly grown north American cranberry cultivars (Vaccinium macrocarpon). J. Sci. Food Agric. 2014, 94, 2738–2745. [Google Scholar] [CrossRef]
  134. Lee, J. Proanthocyanidin a2 purification and quantification of American cranberry (vaccinium macrocarpon ait.) products. J. Funct. Food. 2013, 5, 144–153. [Google Scholar] [CrossRef]
  135. Jungfer, E.; Zimmermann, B.F.; Ruttkat, A.; Galensa, R. Comparing procyanidins in selected vaccinium species by uhplc-ms2 with regard to authenticity and health effects. J. Agric. Food Chem. 2012, 60, 9688–9696. [Google Scholar] [CrossRef] [PubMed]
  136. White, B.L.; Howard, L.R.; Prior, R.L. Release of bound procyanidins from cranberry pomace by alkaline hydrolysis. J. Agric. Food Chem. 2010, 58, 7572–7579. [Google Scholar] [CrossRef] [PubMed]
  137. Sanchez-Patan, F.; Bartolome, B.; Martin-Alvarez, P.J.; Anderson, M.; Howell, A.; Monagas, M. Comprehensive assessment of the quality of commercial cranberry products. Phenolic characterization and in vitro bioactivity. J. Agric. Food Chem. 2012, 60, 3396–3408. [Google Scholar] [CrossRef] [PubMed]
  138. Severo, C.; Anjos, I.; Souza, V.G.L.; Canejo, J.P.; Bronze, M.R.; Fernando, A.L.; Coelhoso, I.; Bettencourt, A.F.; Ribeiro, I.A.C. Development of cranberry extract films for the enhancement of food packaging antimicrobial properties. Food Packag. Shelf Life 2021, 28, 100646. [Google Scholar] [CrossRef]
  139. Botto, H.; Neuzillet, Y. Effectiveness of a cranberry (Vaccinium macrocarpon) preparation in reducing asymptomatic bacteriuria in patients with an ileal enterocystoplasty. Scand. J. Urol. Nephrol. 2010, 44, 165–168. [Google Scholar] [CrossRef] [PubMed]
  140. Baranowska, M.; Bartoszek, A. Antioxidant and antimicrobial properties of bioactive phytochemicals from cranberry. Postep. Hig. Med. Dosw. 2016, 70, 1460–1468. [Google Scholar] [CrossRef] [PubMed]
  141. Faggian, M.; Bernabe, G.; Valente, M.; Francescato, S.; Baratto, G.; Brun, P.; Castagliuolo, I.; Dall’Acqua, S.; Peron, G. Characterization of pacs profile and bioactivity of a novel nutraceutical combining cranberry extracts with different pac-a oligomers, d-mannose and ascorbic acid: An in vivo/ex vivo evaluation of dual mechanism of action on intestinal barrier and urinary epithelium. Food Res. Int. 2021, 149, 110649. [Google Scholar]
  142. Jepson, R.G.; Williams, G.; Craig, J.C. Cranberries for preventing urinary tract infections. Cochrane Database Syst. Rev. 2012, 10, CD001321. [Google Scholar] [CrossRef] [Green Version]
  143. Ermel, G.; Georgeault, S.; Inisan, C.; Besnard, M. Inhibition of adhesion of uropathogenic Escherichia coli bacteria to uroepithelial cells by extracts from cranberry. J. Med. Food 2012, 15, 126–134. [Google Scholar] [CrossRef]
  144. Fu, Z.; Liska, D.; Talan, D.; Chung, M. Cranberry reduces the risk of urinary tract infection recurrence in otherwise healthy women: A systematic review and meta-analysis. J. Nutr. 2017, 147, 2282–2288. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Howell, A.B.; Reed, J.D.; Krueger, C.G.; Winterbottom, R.; Cunningham, D.G.; Leahy, M. A-type cranberry proanthocyanidins and uropathogenic bacterial anti-adhesion activity. Phytochemistry 2005, 66, 2281–2291. [Google Scholar] [CrossRef] [PubMed]
  146. Nemzer, B.V.; Al-Taher, F.; Yashin, A.; Revelsky, I.; Yashin, Y. Cranberry: Chemical composition, antioxidant activity and impact on human health: Overview. Molecules 2022, 27, 1503. [Google Scholar] [CrossRef] [PubMed]
  147. González de Llano, D.; Moreno-Arribas, M.V.; Bartolomé, B. Cranberry polyphenols and prevention against urinary tract infections: Relevant considerations. Molecules 2020, 25, 3523. [Google Scholar]
  148. Jiao, J.J.; Wei, Y.; Chen, J.N.; Chen, X.Y.; Zhang, Y. Anti-aging and redox state regulation effects of a-type proanthocyanidins-rich cranberry concentrate and its comparison with grape seed extract in mice. J. Funct. Food. 2017, 30, 63–73. [Google Scholar] [CrossRef]
  149. Liu, H.W.; Zou, T.; Gao, J.M.; Gu, L.W. Depolymerization of cranberry procyanidins using (+)-catechin, (-)-epicatechin, and (-)-epigallocatechin gallate as chain breakers. Food Chem. 2013, 141, 488–494. [Google Scholar] [CrossRef]
  150. Ou, K.Q.; Percival, S.S.; Zou, T.; Khoo, C.; Gu, L.W. Transport of cranberry a-type procyanidin dimers, trimers, and tetramers across monolayers of human intestinal epithelial caco-2 cells. J. Agric. Food Chem. 2012, 60, 1390–1396. [Google Scholar] [CrossRef]
  151. Maatta-Riihinen, K.R.; Kahkonen, M.P.; Torronen, A.R.; Heinonen, I.M. Catechins and procyanidins in berries of vaccinium species and their antioxidant activity. J. Agric. Food Chem. 2005, 53, 8485–8491. [Google Scholar] [CrossRef]
  152. Reed, J. Cranberry flavonoids, atherosclerosis and cardiovascular health. Crit. Rev. Food Sci. Nutr. 2002, 42, 301–316. [Google Scholar] [CrossRef]
  153. Weh, K.M.; Clarke, J.; Kresty, L.A. Cranberries and cancer: An update of preclinical studies evaluating the cancer inhibitory potential of cranberry and cranberry derived constituents. Antioxidants 2016, 5, 27. [Google Scholar] [CrossRef]
  154. Zhao, Y.; Wu, X.L.; Yu, L.L.; Chen, P. Retention of polyphenols in blueberries (Vaccinium corymbosum) after different cooking methods, using uhplc-dad-ms based metabolomics. J. Food Compos. Anal. 2017, 56, 55–66. [Google Scholar] [CrossRef]
  155. Merghem, R.; Jay, M.; Brun, N.; Voirin, B. Qualitative analysis and hplc isolation and identification of procyanidins from vicia faba. Phytochem. Anal. 2004, 15, 95–99. [Google Scholar] [CrossRef] [PubMed]
  156. Passos, C.P.; Cardoso, S.M.; Domingues, M.R.M.; Domingues, P.; Silva, C.M.; Coimbra, M.A. Evidence for galloylated type-a procyanidins in grape seeds. Food Chem. 2007, 105, 1457–1467. [Google Scholar] [CrossRef]
  157. Zhao, L.; Wen, L.M.; Lu, Q.; Liu, R. Interaction mechanism between alpha-glucosidase and a-type trimer procyanidin revealed by integrated spectroscopic analysis techniques. Int. J. Biol. Macromol. 2020, 143, 173–180. [Google Scholar] [CrossRef] [PubMed]
  158. Vaid, M.; Prasad, R.; Singh, T.; Katiyar, S.K. Dietary grape seed proanthocyanidins inactivate regulatory t cells by promoting ner-dependent DNA repair in dendritic cells in uvb-exposed skin. Oncotarget 2017, 8, 49625–49636. [Google Scholar] [CrossRef] [Green Version]
  159. Zhou, D.Y.; Du, Q.; Li, R.R.; Huang, M.; Zhang, Q.; Wei, G.Z. Grape seed proanthocyanidin extract attenuates airway inflammation and hyperresponsiveness in a murine model of asthma by downregulating inducible nitric oxide synthase. Planta Med. 2011, 77, 1575–1581. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  160. Roychowdhury, S.; Wolf, G.; Keilhoff, G.; Bagchi, D.; Horn, T. Protection of primary glial cells by grape seed proanthocyanidin extract against nitrosative/oxidative stress. Nitric Oxide 2001, 5, 137–149. [Google Scholar] [CrossRef] [PubMed]
  161. Natella, F.; Belelli, F.; Gentili, V.; Ursini, F.; Scaccini, C. Grape seed proanthocyanidins prevent plasma postprandial oxidative stress in humans. J. Agric. Food Chem. 2002, 50, 7720–7725. [Google Scholar] [CrossRef]
  162. Quesada, H.; del Bas, J.M.; Pajuelo, D.; Diaz, S.; Fernandez-Larrea, J.; Pinent, M.; Arola, L.; Salvado, M.J.; Blade, C. Grape seed proanthocyanidins correct dyslipidemia associated with a high-fat diet in rats and repress genes controlling lipogenesis and vldl assembling in liver. Int. J. Obes. 2009, 33, 1007–1012. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Razavi, S.M.; Gholamin, S.; Eskandari, A.; Mohsenian, N.; Ghorbanihaghjo, A.; Delazar, A.; Rashtchizadeh, N.; Keshtkar-Jahromi, M.; Argani, H. Red grape seed extract improves lipid profiles and decreases oxidized low-density lipoprotein in patients with mild hyperlipidemia. J. Med. Food 2013, 16, 255–258. [Google Scholar] [CrossRef]
  164. Lu, R.H.; Qin, C.B.; Yang, F.; Zhang, W.Y.; Zhang, Y.R.; Yang, G.K.; Yang, L.P.; Meng, X.L.; Yan, X.; Nie, G.X. Grape seed proanthocyanidin extract ameliorates hepatic lipid accumulation and inflammation in grass carp (Ctenopharyngodon idella). Fish Physiol. Biochem. 2020, 46, 1665–1677. [Google Scholar] [CrossRef]
  165. Vinson, J.A.; Mandarano, M.A.; Shuta, D.L.; Bagchi, M.; Bagchi, D. Beneficial effects of a novel ih636 grape seed proanthocyanidin extract and a niacin-bound chromium in a hamster atherosclerosis model. Mol. Cell. Biochem. 2002, 240, 99–103. [Google Scholar] [CrossRef] [PubMed]
  166. Yamakoshi, J.; Kataoka, S.; Koga, T.; Ariga, T. Proanthocyanidin-rich extract from grape seeds attenuates the development of aortic atherosclerosis in cholesterol-fed rabbits. Atherosclerosis 1999, 142, 139–149. [Google Scholar] [CrossRef] [PubMed]
  167. Zhong, L.; Luo, N.; Zhong, X.; Xu, T.; Hao, P. The immunoregulatory effects of natural products on psoriasis via its action on th17 cells versus regulatory t cells balance. Int. Immunopharmacol. 2022, 110, 109032. [Google Scholar] [CrossRef] [PubMed]
  168. Park, M.-K.; Park, J.-S.; Cho, M.-L.; Oh, H.-J.; Heo, Y.-J.; Woo, Y.-J.; Heo, Y.-M.; Park, M.-J.; Park, H.-S.; Park, S.-H.; et al. Grape seed proanthocyanidin extract (gspe) differentially regulates foxp3+ regulatory and il-17+ pathogenic t cell in autoimmune arthritis. Immunol. Lett. 2011, 135, 50–58. [Google Scholar] [CrossRef] [PubMed]
  169. Katiyar, S.K.; Pal, H.C.; Prasad, R. Dietary proanthocyanidins prevent ultraviolet radiation-induced non-melanoma skin cancer through enhanced repair of damaged DNA-dependent activation of immune sensitivity. Semin. Cancer Biol. 2017, 46, 138–145. [Google Scholar] [CrossRef]
  170. Katiyar, S.K. Dietary proanthocyanidins inhibit uv radiation-induced skin tumor development through functional activation of the immune system. Mol. Nutr. Food Res. 2016, 60, 1374–1382. [Google Scholar] [CrossRef] [Green Version]
  171. Katiyar, S.K. Proanthocyanidins from grape seeds inhibit uv-radiation-induced immune suppression in mice: Detection and analysis of molecular and cellular targets. Photochem. Photobiol. 2015, 91, 156–162. [Google Scholar] [CrossRef] [Green Version]
  172. Percival, S.S. Grape consumption supports immunity in animals and humans. J. Nutr. 2009, 139, 1801S–1805S. [Google Scholar] [CrossRef] [Green Version]
  173. Narusaka, M.; Hatanaka, T.; Narusaka, Y. Inactivation of plant and animal viruses by proanthocyanidins from alpinia zerumbet extract. Plant Biotechnol. 2021, 38, 453–455. [Google Scholar] [CrossRef]
  174. Morimoto, H.; Hatanaka, T.; Narusaka, M.; Narusaka, Y. Molecular investigation of proanthocyanidin from alpinia zerumbet against the influenza a virus. Fitoterapia 2022, 158, 105141. [Google Scholar] [CrossRef]
  175. Mateos-Martín, M.L.; Fuguet, E.; Jiménez-Ardón, A.; Herrero-Uribe, L.; Tamayo-Castillo, G.; Torres, J.L. Identification of polyphenols from antiviral chamaecrista nictitans extract using high-resolution lc-esi-ms/ms. Anal. Bioanal. Chem. 2014, 406, 5501–5506. [Google Scholar] [CrossRef] [PubMed]
  176. Fink, R.C.; Roschek, B., Jr.; Alberte, R.S. Hiv type-1 entry inhibitors with a new mode of action. Antivir. Chem. Chemother. 2009, 19, 243–255. [Google Scholar] [CrossRef]
  177. Connell, B.J.; Chang, S.Y.; Prakash, E.; Yousfi, R.; Mohan, V.; Posch, W.; Wilflingseder, D.; Moog, C.; Kodama, E.N.; Clayette, P.; et al. A cinnamon-derived procyanidin compound displays anti-hiv-1 activity by blocking heparan sulfate- and co-receptor- binding sites on gp120 and reverses t cell exhaustion via impeding tim-3 and pd-1 upregulation. PLoS ONE 2016, 11, e0165386. [Google Scholar] [CrossRef] [Green Version]
  178. Fauvelle, C.; Lambotin, M.; Heydmann, L.; Prakash, E.; Bhaskaran, S.; Vishwaraman, M.; Baumert, T.F.; Moog, C. A cinnamon-derived procyanidin type a compound inhibits hepatitis c virus cell entry. Hepatol. Int. 2017, 11, 440–445. [Google Scholar] [CrossRef]
  179. Zhuang, M.; Jiang, H.; Suzuki, Y.; Li, X.; Xiao, P.; Tanaka, T.; Ling, H.; Yang, B.; Saitoh, H.; Zhang, L.; et al. Procyanidins and butanol extract of cinnamomi cortex inhibit SARS-CoV infection. Antivir. Res. 2009, 82, 73–81. [Google Scholar] [CrossRef] [PubMed]
  180. Tietjen, I.; Ntie-Kang, F.; Mwimanzi, P.; Onguéné, P.A.; Scull, M.A.; Idowu, T.O.; Ogundaini, A.O.; Meva’a, L.M.; Abegaz, B.M.; Rice, C.M.; et al. Screening of the pan-african natural product library identifies ixoratannin a-2 and boldine as novel hiv-1 inhibitors. PLoS ONE 2015, 10, e0121099. [Google Scholar] [CrossRef] [Green Version]
  181. Xu, X.; Xie, H.; Wang, Y.; Wei, X. A-type proanthocyanidins from lychee seeds and their antioxidant and antiviral activities. J. Agric. Food Chem. 2010, 58, 11667–11672. [Google Scholar] [CrossRef] [PubMed]
  182. Feng, W.Y.; Tanaka, R.; Inagaki, Y.; Saitoh, Y.; Chang, M.O.; Amet, T.; Yamamoto, N.; Yamaoka, S.; Yoshinaka, Y. Pycnogenol, a procyanidin-rich extract from french maritime pine, inhibits intracellular replication of hiv-1 as well as its binding to host cells. Jpn. J. Infect. Dis. 2008, 61, 279–285. [Google Scholar]
  183. Ezzikouri, S.; Nishimura, T.; Kohara, M.; Benjelloun, S.; Kino, Y.; Inoue, K.; Matsumori, A.; Tsukiyama-Kohara, K. Inhibitory effects of pycnogenol® on hepatitis c virus replication. Antivir. Res. 2015, 113, 93–102. [Google Scholar] [CrossRef] [Green Version]
  184. Suedee, A.; Tewtrakul, S.; Panichayupakaranant, P. Anti-hiv-1 integrase compound from pometia pinnata leaves. Pharm. Biol. 2013, 51, 1256–1261. [Google Scholar] [CrossRef] [PubMed]
  185. Mirandola, M.; Salvati, M.V.; Rodigari, C.; Appelberg, K.S.; Mirazimi, A.; Maffei, M.E.; Gribaudo, G.; Salata, C. Cranberry (Vaccinium macrocarpon) extract impairs nairovirus infection by inhibiting the attachment to target cells. Pathogens 2021, 10, 1025. [Google Scholar] [CrossRef] [PubMed]
  186. Luganini, A.; Terlizzi, M.E.; Catucci, G.; Gilardi, G.; Maffei, M.E.; Gribaudo, G. The cranberry extract oximacro(®) exerts in vitro virucidal activity against influenza virus by interfering with hemagglutinin. Front. Microbiol. 2018, 9, 1826. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Turmagambetova, A.S.; Sokolova, N.S.; Bogoyavlenskiy, A.P.; Berezin, V.E.; Lila, M.A.; Cheng, D.M.; Dushenkov, V. New functionally-enhanced soy proteins as food ingredients with anti-viral activity. Virusdisease 2015, 26, 123–132. [Google Scholar] [CrossRef] [PubMed]
  188. Terlizzi, M.E.; Occhipinti, A.; Luganini, A.; Maffei, M.E.; Gribaudo, G. Inhibition of herpes simplex type 1 and type 2 infections by oximacro(®), a cranberry extract with a high content of a-type proanthocyanidins (pacs-a). Antivir. Res. 2016, 132, 154–164. [Google Scholar] [CrossRef] [Green Version]
  189. Su, X.W.; Howell, A.B.; D’Souza, D.H. Antiviral effects of cranberry juice and cranberry proanthocyanidins on foodborne viral surrogates—A time dependence study in vitro. Food Microbiol. 2010, 27, 985–991. [Google Scholar] [CrossRef]
  190. Su, X.; Howell, A.B.; D’Souza, D.H. The effect of cranberry juice and cranberry proanthocyanidins on the infectivity of human enteric viral surrogates. Food Microbiol. 2010, 27, 535–540. [Google Scholar] [CrossRef]
  191. Lipson, S.M.; Cohen, P.; Zhou, J.; Burdowski, A.; Stotzky, G. Cranberry cocktail juice, cranberry concentrates, and proanthocyanidins reduce reovirus infectivity titers in african green monkey kidney epithelial cell cultures. Mol. Nutr. Food Res. 2007, 51, 752–758. [Google Scholar] [CrossRef]
  192. Lipson, S.M.; Sethi, L.; Cohen, P.; Gordon, R.E.; Tan, I.P.; Burdowski, A.; Stotzky, G. Antiviral effects on bacteriophages and rotavirus by cranberry juice. Phytomedicine 2007, 14, 23–30. [Google Scholar] [CrossRef]
  193. Lipson, S.M.; Ozen, F.S.; Karthikeyan, L.; Gordon, R.E. Effect of ph on anti-rotavirus activity by comestible juices and proanthocyanidins in a cell-free assay system. Food Environ. Virol. 2012, 4, 168–178. [Google Scholar] [CrossRef]
  194. Sugamoto, K.; Tanaka, Y.L.; Saito, A.; Goto, Y.; Nakayama, T.; Okabayashi, T.; Kunitake, H.; Morishita, K. Highly polymerized proanthocyanidins (pac) components from blueberry leaf and stem significantly inhibit SARS-CoV-2 infection via inhibition of ace2 and viral 3clpro enzymes. Biochem. Biophys. Res. Commun. 2022, 615, 56–62. [Google Scholar] [CrossRef]
  195. Joshi, S.S.; Howell, A.B.; D’Souza, D.H. Reduction of enteric viruses by blueberry juice and blueberry proanthocyanidins. Food Environ. Virol. 2016, 8, 235–243. [Google Scholar] [CrossRef] [PubMed]
  196. Takeshita, M.; Ishida, Y.; Akamatsu, E.; Ohmori, Y.; Sudoh, M.; Uto, H.; Tsubouchi, H.; Kataoka, H. Proanthocyanidin from blueberry leaves suppresses expression of subgenomic hepatitis c virus RNA. J. Biol. Chem. 2009, 284, 21165–21176. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  197. Nair, M.P.; Kandaswami, C.; Mahajan, S.; Nair, H.N.; Chawda, R.; Shanahan, T.; Schwartz, S.A. Grape seed extract proanthocyanidins downregulate hiv-1 entry coreceptors, ccr2b, ccr3 and ccr5 gene expression by normal peripheral blood mononuclear cells. Biol. Res. 2002, 35, 421–431. [Google Scholar] [CrossRef]
  198. McCormick, I.; James, C.; Welton, N.J.; Mayaud, P.; Turner, K.M.E.; Gottlieb, S.L.; Foster, A.; Looker, K.J. Incidence of herpes simplex virus keratitis and other ocular disease: Global review and estimates. Ophthalmic Epidemiol. 2022, 29, 353–362. [Google Scholar] [CrossRef] [PubMed]
  199. James, C.; Harfouche, M.; Welton, N.J.; Turner, K.M.; Abu-Raddad, L.J.; Gottlieb, S.L.; Looker, K.J. Herpes simplex virus: Global infection prevalence and incidence estimates, 2016. Bull. World Health Organ. 2020, 98, 315–329. [Google Scholar] [CrossRef] [PubMed]
  200. Whitley, R.; Baines, J. Clinical management of herpes simplex virus infections: Past, present, and future. F1000Research 2018, 7, F1000. [Google Scholar] [CrossRef] [Green Version]
  201. Son, M.; Lee, M.; Sung, G.H.; Lee, T.; Shin, Y.S.; Cho, H.; Lieberman, P.M.; Kang, H. Bioactive activities of natural products against herpesvirus infection. J. Microbiol. 2013, 51, 545–551. [Google Scholar] [CrossRef]
  202. Hassan, S.T.; Masarčíková, R.; Berchová, K. Bioactive natural products with anti-herpes simplex virus properties. J. Pharm. Pharmacol. 2015, 67, 1325–1336. [Google Scholar] [CrossRef]
  203. Khan, M.T.; Ather, A.; Thompson, K.D.; Gambari, R. Extracts and molecules from medicinal plants against herpes simplex viruses. Antivir. Res. 2005, 67, 107–119. [Google Scholar] [CrossRef]
  204. Gescher, K.; Hensel, A.; Hafezi, W.; Derksen, A.; Kühn, J. Oligomeric proanthocyanidins from rumex acetosa l. Inhibit the attachment of herpes simplex virus type-1. Antivir. Res. 2011, 89, 9–18. [Google Scholar] [CrossRef]
  205. Gescher, K.; Kühn, J.; Lorentzen, E.; Hafezi, W.; Derksen, A.; Deters, A.; Hensel, A. Proanthocyanidin-enriched extract from myrothamnus flabellifolia welw. Exerts antiviral activity against herpes simplex virus type 1 by inhibition of viral adsorption and penetration. J. Ethnopharmacol. 2011, 134, 468–474. [Google Scholar] [CrossRef] [PubMed]
  206. Freitas, A.M.; Almeida, M.T.R.; Andrighetti-Fröhner, C.R.; Cardozo, F.T.G.S.; Barardi, C.R.M.; Farias, M.R.; Simões, C.M.O. Antiviral activity-guided fractionation from araucaria angustifolia leaves extract. J. Ethnopharmacol. 2009, 126, 512–517. [Google Scholar] [CrossRef] [PubMed]
  207. Shahat, A.A.; Cos, P.; De Bruyne, T.; Apers, S.; Hammouda, F.M.; Ismail, S.I.; Azzam, S.; Claeys, M.; Goovaerts, E.; Pieters, L.; et al. Antiviral and antioxidant activity of flavonoids and proanthocyanidins from Crataegus sinaica. Planta Med. 2002, 68, 539–541. [Google Scholar] [CrossRef] [PubMed]
  208. Erdelmeier, C.A.; Cinatl, J., Jr.; Rabenau, H.; Doerr, H.W.; Biber, A.; Koch, E. Antiviral and antiphlogistic activities of hamamelis virginiana bark. Planta Med. 1996, 62, 241–245. [Google Scholar] [CrossRef] [PubMed]
  209. Danaher, R.J.; Wang, C.; Dai, J.; Mumper, R.J.; Miller, C.S. Antiviral effects of blackberry extract against herpes simplex virus type 1. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. Endodontol. 2011, 112, e31–e35. [Google Scholar] [CrossRef] [Green Version]
  210. Cheng, H.Y.; Lin, C.C.; Lin, T.C. Antiviral properties of prodelphinidin b-2 3’-o-gallate from green tea leaf. Antivir. Chem. Chemother. 2002, 13, 223–229. [Google Scholar] [CrossRef] [Green Version]
  211. Connolly, S.A.; Jardetzky, T.S.; Longnecker, R. The structural basis of herpesvirus entry. Nature reviews. Microbiology 2021, 19, 110–121. [Google Scholar]
  212. Waymack, J.R.; Sundareshan, V. Acquired Immune Deficiency Syndrome; StatPearls Publishing LLC.: Tampa, FL, USA, 2022. [Google Scholar]
  213. Laila, U.; Akram, M.; Shariati, M.A.; Hashmi, A.M.; Akhtar, N.; Tahir, I.M.; Ghauri, A.O.; Munir, N.; Riaz, M.; Akhter, N.; et al. Role of medicinal plants in hiv/aids therapy. Clin. Exp. Pharmacol. Physiol. 2019, 46, 1063–1073. [Google Scholar] [CrossRef] [Green Version]
  214. Olubiyi, O.O.; Idowu, T.O.; Ogundaini, A.O.; Orhuah, G. Computational prospecting for the pharmacological mechanism of activity: Hiv-1 inhibition by ixoratannin a-2. Curr. Comput. Aided Drug Des. 2020, 16, 376–388. [Google Scholar] [CrossRef]
  215. Zai, W.; Hu, K.; Ye, J.; Ding, J.; Huang, C.; Li, Y.; Fang, Z.; Wu, M.; Wang, C.; Chen, J.; et al. Long-term hepatitis b virus infection induces cytopathic effects in primary human hepatocytes, and can be partially reversed by antiviral therapy. Microbiol. Spectr. 2022, 10, e0132821. [Google Scholar] [CrossRef]
  216. Jose-Abrego, A.; Rivera-Iñiguez, I.; Torres-Reyes, L.A.; Roman, S. Anti-hepatitis b virus activity of food nutrients and potential mechanisms of action. Ann. Hepatol. 2022, 100766. [Google Scholar] [CrossRef] [PubMed]
  217. Tsukuda, S.; Watashi, K.; Hojima, T.; Isogawa, M.; Iwamoto, M.; Omagari, K.; Suzuki, R.; Aizaki, H.; Kojima, S.; Sugiyama, M.; et al. A new class of hepatitis b and d virus entry inhibitors, proanthocyanidin and its analogs, that directly act on the viral large surface proteins. Hepatology 2017, 65, 1104–1116. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  218. Ishida, Y.; Takeshita, M.; Kataoka, H. Functional foods effective for hepatitis c: Identification of oligomeric proanthocyanidin and its action mechanism. World J. Hepatol. 2014, 6, 870–879. [Google Scholar] [CrossRef]
  219. Li, S.; Kodama, E.N.; Inoue, Y.; Tani, H.; Matsuura, Y.; Zhang, J.; Tanaka, T.; Hattori, T. Procyanidin b1 purified from cinnamomi cortex suppresses hepatitis c virus replication. Antivir. Chem. Chemother. 2010, 20, 239–248. [Google Scholar] [CrossRef] [PubMed]
  220. Bishop, R.; Kirkwood, C. Enteric viruses. In Encyclopedia of Virology, 3rd ed.; Mahy, B.W.J., Van Regenmortel, M.H.V., Eds.; Academic Press: London, UK, 2008; pp. 116–123. [Google Scholar]
  221. Eckardt, A.J.; Baumgart, D.C. Viral gastroenteritis in adults. Recent Pat. Anti Infect. Drug Discov. 2011, 6, 54–63. [Google Scholar] [CrossRef] [PubMed]
  222. Fong, T.T.; Lipp, E.K. Enteric viruses of humans and animals in aquatic environments: Health risks, detection, and potential water quality assessment tools. Microbiol. Mol. Biol. Rev. 2005, 69, 357–371. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  223. Bernier, C.; Goetz, C.; Jubinville, E.; Jean, J. The new face of berries: A review of their antiviral proprieties. Foods 2021, 11, 102. [Google Scholar] [CrossRef]
  224. Iwasawa, A.; Niwano, Y.; Mokudai, T.; Kohno, M. Antiviral activity of proanthocyanidin against feline calicivirus used as a surrogate for noroviruses, and coxsackievirus used as a representative enteric virus. Biocontrol Sci. 2009, 14, 107–111. [Google Scholar] [CrossRef] [Green Version]
  225. Clark, A.; Black, R.; Tate, J.; Roose, A.; Kotloff, K.; Lam, D.; Blackwelder, W.; Parashar, U.; Lanata, C.; Kang, G.; et al. Estimating global, regional and national rotavirus deaths in children aged < 5 years: Current approaches, new analyses and proposed improvements. PLoS ONE 2017, 12, e0183392. [Google Scholar]
  226. Esona, M.D.; Gautam, R. Rotavirus. Clin. Lab. Med. 2015, 35, 363–391. [Google Scholar] [CrossRef]
  227. Green, K.Y. Caliciviridae. In The Noroviruses, 6th ed.; Wolters Kluwer Health/Lippincott Williams & Wilkins: Philadelphia, PA, USA, 2013; Volume 1. [Google Scholar]
  228. Lopman, B.A.; Steele, D.; Kirkwood, C.D.; Parashar, U.D. The vast and varied global burden of norovirus: Prospects for prevention and control. PLoS Med. 2016, 13, e1001999. [Google Scholar] [CrossRef]
  229. de Graaf, M.; van Beek, J.; Koopmans, M.P. Human norovirus transmission and evolution in a changing world. Nat. Reviews. Microbiol. 2016, 14, 421–433. [Google Scholar] [CrossRef] [PubMed]
  230. Karst, S.M.; Wobus, C.E.; Goodfellow, I.G.; Green, K.Y.; Virgin, H.W. Advances in norovirus biology. Cell Host Microbe 2014, 15, 668–680. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  231. Estes, M.K.; Ettayebi, K.; Tenge, V.R.; Murakami, K.; Karandikar, U.; Lin, S.C.; Ayyar, B.V.; Cortes-Penfield, N.W.; Haga, K.; Neill, F.H.; et al. Human norovirus cultivation in nontransformed stem cell-derived human intestinal enteroid cultures: Success and challenges. Viruses 2019, 11, 638. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  232. Lee, J.H.; Bae, S.Y.; Oh, M.; Seok, J.H.; Kim, S.; Chung, Y.B.; Gowda, K.G.; Mun, J.Y.; Chung, M.S.; Kim, K.H. Antiviral effects of black raspberry (Rubus coreanus) seed extract and its polyphenolic compounds on norovirus surrogates. Biosci. Biotechnol. Biochem. 2016, 80, 1196–1204. [Google Scholar] [CrossRef]
  233. Joshi, S.; Howell, A.B.; D’Souza, D.H. Blueberry proanthocyanidins against human norovirus surrogates in model foods and under simulated gastric conditions. Food Microbiol. 2017, 63, 263–267. [Google Scholar] [CrossRef] [PubMed]
  234. Kutter, J.S.; Spronken, M.I.; Fraaij, P.L.; Fouchier, R.A.; Herfst, S. Transmission routes of respiratory viruses among humans. Curr. Opin. Virol. 2018, 28, 142–151. [Google Scholar] [CrossRef]
  235. Hodinka, R.L. Respiratory RNA viruses. Microbiol. Spectr. 2016, 4. [Google Scholar] [CrossRef] [Green Version]
  236. Moriyama, M.; Hugentobler, W.J.; Iwasaki, A. Seasonality of respiratory viral infections. Annu. Rev. Virol. 2020, 7, 83–101. [Google Scholar] [CrossRef]
  237. Roschek, B., Jr.; Fink, R.C.; McMichael, M.D.; Li, D.; Alberte, R.S. Elderberry flavonoids bind to and prevent h1n1 infection in vitro. Phytochemistry 2009, 70, 1255–1261. [Google Scholar] [CrossRef]
  238. Ikuta, K.; Hashimoto, K.; Kaneko, H.; Mori, S.; Ohashi, K.; Suzutani, T. Anti-viral and anti-bacterial activities of an extract of blackcurrants (Ribes nigrum L.). Microbiol. Immunol. 2012, 56, 805–809. [Google Scholar] [CrossRef]
  239. Lin, L.T.; Hsu, W.C.; Lin, C.C. Antiviral natural products and herbal medicines. J. Tradit. Complement. Med. 2014, 4, 24–35. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  240. Global Influenza Strategy 2019–2030; World Health Organization: Geneva, Switzerland, 2019.
  241. Krammer, F.; Smith, G.J.D.; Fouchier, R.A.M.; Peiris, M.; Kedzierska, K.; Doherty, P.C.; Palese, P.; Shaw, M.L.; Treanor, J.; Webster, R.G.; et al. Influenza. Nat. Rev. Dis. Prim. 2018, 4, 3. [Google Scholar] [CrossRef] [PubMed]
  242. World Health Organization. Available online: https://www.Who.Int/en/news-room/fact-sheets/detail/influenza-(seasonal) (accessed on 22 September 2022).
  243. Derksen, A.; Hensel, A.; Hafezi, W.; Herrmann, F.; Schmidt, T.J.; Ehrhardt, C.; Ludwig, S.; Kühn, J. 3-o-galloylated procyanidins from rumex acetosa l. Inhibit the attachment of influenza a virus. PLoS ONE 2014, 9, e110089. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  244. Sekizawa, H.; Ikuta, K.; Mizuta, K.; Takechi, S.; Suzutani, T. Relationship between polyphenol content and anti-influenza viral effects of berries. J. Sci. Food Agric. 2013, 93, 2239–2241. [Google Scholar] [CrossRef]
  245. World Health Organization. Available online: https://covid19.Who.Int (accessed on 23 September 2022).
  246. Hu, B.; Guo, H.; Zhou, P.; Shi, Z.L. Characteristics of SARS-CoV-2 and COVID-19. Nat. Rev. Microbiol. 2021, 19, 141–154. [Google Scholar] [CrossRef]
  247. Martella, V.; Elia, G.; Buonavoglia, C. Canine distemper virus. Vet. Clin. N. Am. Small Anim. Pract. 2008, 38, 787–797. [Google Scholar] [CrossRef]
  248. Gallina, L.; Dal Pozzo, F.; Galligioni, V.; Bombardelli, E.; Scagliarini, A. Inhibition of viral RNA synthesis in canine distemper virus infection by proanthocyanidin a2. Antivir. Res. 2011, 92, 447–452. [Google Scholar] [CrossRef]
  249. Lunney, J.K.; Fang, Y.; Ladinig, A.; Chen, N.; Li, Y.; Rowland, B.; Renukaradhya, G.J. Porcine reproductive and respiratory syndrome virus (prrsv): Pathogenesis and interaction with the immune system. Annu. Rev. Anim. Biosci. 2016, 4, 129–154. [Google Scholar] [CrossRef]
  250. Zhang, M.; Wu, Q.; Chen, Y.; Duan, M.; Tian, G.; Deng, X.; Sun, Y.; Zhou, T.; Zhang, G.; Chen, W.; et al. Inhibition of proanthocyanidin a2 on porcine reproductive and respiratory syndrome virus replication in vitro. PLoS ONE 2018, 13, e0193309. [Google Scholar] [CrossRef]
  251. Salata, C.; Calistri, A.; Parolin, C.; Palù, G. Coronaviruses: A paradigm of new emerging zoonotic diseases. Pathog. Dis. 2019, 77, ftaa006. [Google Scholar] [CrossRef] [Green Version]
  252. Salata, C.; Calistri, A.; Parolin, C.; Baritussio, A.; Palù, G. Antiviral activity of cationic amphiphilic drugs. Expert Rev. Anti Infect. Ther. 2017, 15, 483–492. [Google Scholar] [CrossRef] [Green Version]
  253. Salata, C.; Calistri, A.; Alvisi, G.; Celestino, M.; Parolin, C.; Palù, G. Ebola virus entry: From molecular characterization to drug discovery. Viruses 2019, 11, 274. [Google Scholar] [CrossRef] [Green Version]
  254. Liu, C.H.; Hu, Y.T.; Wong, S.H.; Lin, L.T. Therapeutic strategies against ebola virus infection. Viruses 2022, 14, 579. [Google Scholar] [CrossRef]
  255. Hansen, F.; Feldmann, H.; Jarvis, M.A. Targeting ebola virus replication through pharmaceutical intervention. Expert Opin. Investig. Drugs 2021, 30, 201–226. [Google Scholar] [CrossRef]
  256. Du, R.; Cui, Q.; Caffrey, M.; Rong, L. Ebola virus entry inhibitors. Adv. Exp. Med. Biol. 2022, 1366, 155–170. [Google Scholar]
  257. Cui, Q.; Du, R.; Anantpadma, M.; Schafer, A.; Hou, L.; Tian, J.; Davey, R.A.; Cheng, H.; Rong, L. Identification of ellagic acid from plant rhodiola rosea l. As an anti-ebola virus entry inhibitor. Viruses 2018, 10, 152. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  258. Reid, S.P.; Shurtleff, A.C.; Costantino, J.A.; Tritsch, S.R.; Retterer, C.; Spurgers, K.B.; Bavari, S. Hspa5 is an essential host factor for ebola virus infection. Antivir. Res. 2014, 109, 171–174. [Google Scholar] [CrossRef]
  259. Tsang, N.Y.; Li, W.F.; Varhegyi, E.; Rong, L.; Zhang, H.J. Ebola entry inhibitors discovered from Maesa perlarius. Int. J. Mol. Sci. 2022, 23, 2620. [Google Scholar] [CrossRef]
  260. Altamish, M.; Khan, M.; Baig, M.S.; Pathak, B.; Rani, V.; Akhtar, J.; Khan, A.A.; Ahmad, S.; Krishnan, A. Therapeutic potential of medicinal plants against dengue infection: A mechanistic viewpoint. ACS Omega 2022, 7, 24048–24065. [Google Scholar] [CrossRef]
  261. Kimmel, E.M.; Jerome, M.; Holderness, J.; Snyder, D.; Kemoli, S.; Jutila, M.A.; Hedges, J.F. Oligomeric procyanidins stimulate innate antiviral immunity in dengue virus infected human PBMCS. Antivir. Res. 2011, 90, 80–86. [Google Scholar] [CrossRef] [Green Version]
  262. Andreolla, A.P.; Borges, A.A.; Bordignon, J.; Duarte Dos Santos, C.N. Mayaro virus: The state-of-the-art for antiviral drug development. Viruses 2022, 14, 1787. [Google Scholar] [CrossRef]
  263. Mello, M.V.P.; Domingos, T.F.S.; Ferreira, D.F.; Ribeiro, M.M.J.; Ribeiro, T.P.; Rodrigues, C.R.; Souza, A.M.T. Antiviral drug discovery and development for mayaro fever—What do we have so far? Mini Rev. Med. Chem. 2020, 20, 921–928. [Google Scholar] [CrossRef]
  264. Ferraz, A.C.; Almeida, L.T.; da Silva Caetano, C.C.; da Silva Menegatto, M.B.; Souza Lima, R.L.; de Senna, J.P.N.; de Oliveira Cardoso, J.M.; Perucci, L.O.; Talvani, A.; Geraldo de Lima, W.; et al. Hepatoprotective, antioxidant, anti-inflammatory, and antiviral activities of silymarin against mayaro virus infection. Antivir. Res. 2021, 194, 105168. [Google Scholar] [CrossRef]
  265. Kuehnert, P.A.; Stefan, C.P.; Badger, C.V.; Ricks, K.M. Crimean-congo hemorrhagic fever virus (cchfv): A silent but widespread threat. Curr. Trop. Med. Rep. 2021, 8, 141–147. [Google Scholar] [CrossRef]
  266. Shahhosseini, N.; Wong, G.; Babuadze, G.; Camp, J.V.; Ergonul, O.; Kobinger, G.P.; Chinikar, S.; Nowotny, N. Crimean-congo hemorrhagic fever virus in Asia, Africa and Europe. Microorganisms 2021, 9, 1907. [Google Scholar] [CrossRef]
  267. Mehand, M.S.; Al-Shorbaji, F.; Millett, P.; Murgue, B. The who r&d blueprint: 2018 review of emerging infectious diseases requiring urgent research and development efforts. Antivir. Res. 2018, 159, 63–67. [Google Scholar]
  268. Dai, S.; Deng, F.; Wang, H.; Ning, Y. Crimean-congo hemorrhagic fever virus: Current advances and future prospects of antiviral strategies. Viruses 2021, 13, 1195. [Google Scholar] [CrossRef]
  269. Harding, S.A. Condensed tannins: Arbiters of abiotic stress tolerance? Tree Physiol. 2019, 39, 341–344. [Google Scholar] [CrossRef]
  270. Liu, H.Y.; Howell, A.B.; Zhang, D.J.; Khoo, C. A randomized, double-blind, placebo-controlled pilot study to assess bacterial anti-adhesive activity in human urine following consumption of a cranberry supplement. Food Funct. 2019, 10, 7645–7652. [Google Scholar] [CrossRef] [Green Version]
  271. Terlizzi, M.E.; Gribaudo, G.; Maffei, M.E. Uropathogenic Escherichia coli (upec) infections: Virulence factors, bladder responses, antibiotic, and non-antibiotic antimicrobial strategies. Front. Microbiol. 2017, 8, 1566. [Google Scholar] [CrossRef] [Green Version]
  272. Bladé, C.; Arola, L.; Salvadó, M.-J. Hypolipidemic effects of proanthocyanidins and their underlying biochemical and molecular mechanisms. Mol. Nutr. Food Res. 2010, 54, 37–59. [Google Scholar] [CrossRef]
  273. Zhang, Y.Y.; Li, X.L.; Li, T.Y.; Li, M.Y.; Huang, R.M.; Li, W.; Yang, R.L. 3-(4-hydroxyphenyl)propionic acid, a major microbial metabolite of procyanidin a2, shows similar suppression of macrophage foam cell formation as its parent molecule. RSC Adv. 2018, 8, 6242–6250. [Google Scholar] [CrossRef] [Green Version]
  274. Andersen-Civil, A.I.S.; Arora, P.; Williams, A.R. Regulation of enteric infection and immunity by dietary proanthocyanidins. Front. Immunol. 2021, 12, 637603. [Google Scholar] [CrossRef]
  275. Martinez-Micaelo, N.; González-Abuín, N.; Terra, X.; Richart, C.; Ardèvol, A.; Pinent, M.; Blay, M. Omega-3 docosahexaenoic acid and procyanidins inhibit cyclo-oxygenase activity and attenuate nf-κb activation through a p105/p50 regulatory mechanism in macrophage inflammation. Biochem. J. 2011, 441, 653–663. [Google Scholar] [CrossRef] [Green Version]
  276. Anhê, F.F.; Varin, T.V.; Le Barz, M.; Desjardins, Y.; Levy, E.; Roy, D.; Marette, A. Gut microbiota dysbiosis in obesity-linked metabolic diseases and prebiotic potential of polyphenol-rich extracts. Curr. Obes. Rep. 2015, 4, 389–400. [Google Scholar] [CrossRef]
  277. Zheng, Y.Y.; Viswanathan, B.; Kesarwani, P.; Mehrotra, S. Dietary agents in cancer prevention: An immunological perspective. Photochem. Photobiol. 2012, 88, 1083–1098. [Google Scholar] [CrossRef]
  278. Kim, H.; Kim, J.Y.; Song, H.S.; Park, K.U.; Mun, K.C.; Ha, E. Grape seed proanthocyanidin extract inhibits interleukin-17-induced interleukin-6 production via mapk pathway in human pulmonary epithelial cells. Naunyn Schmiedebergs Arch. Pharmacol. 2011, 383, 555–562. [Google Scholar] [CrossRef]
  279. Dhanalakshmi, A.; Agarwal, R.; Agarwal, C. Inhibition of nf-kappa b pathway in grape seed extract-induced apoptotic death of human prostate carcinoma du145 cells. Int. J. Oncol. 2003, 23, 721–727. [Google Scholar]
  280. Chen, F.Z.; Wang, H.; Zhao, J.; Yan, J.J.; Meng, H.Y.; Zhan, H.L.; Chen, L.W.; Yuan, L.B. Grape seed proanthocyanidin inhibits monocrotaline-induced pulmonary arterial hypertension via attenuating inflammation: In vivo and in vitro studies. J. Nutr. Biochem. 2019, 67, 72–77. [Google Scholar] [CrossRef] [PubMed]
  281. Charlton, A.J.; Baxter, N.J.; Khan, M.L.; Moir, A.J.; Haslam, E.; Davies, A.P.; Williamson, M.P. Polyphenol/peptide binding and precipitation. J. Agric. Food Chem. 2002, 50, 1593–1601. [Google Scholar] [CrossRef] [PubMed]
  282. Ebraihimnejad, H.; Burkholz, T.; Jacob, C. Flavanols and proanthocyanidins. In Recent Advances in Redox Active Plant and Microbial Products; Jacob, C., Kirsch, G., Slusarenko, A., Winyard, P.G., Burkholz, T., Eds.; Springer: London, UK, 2014; pp. 211–234. [Google Scholar]
  283. Ali, S.; Alam, M.; Khatoon, F.; Fatima, U.; Elasbali, A.M.; Adnan, M.; Islam, A.; Hassan, M.I.; Snoussi, M.; De Feo, V. Natural products can be used in therapeutic management of COVID-19: Probable mechanistic insights. Biomed. Pharmacother. 2022, 147, 112658. [Google Scholar] [CrossRef] [PubMed]
  284. Karim, Q.A.; Baxter, C. Microbicides for the prevention of sexually transmitted HIV infection. Expert Rev. Anti Infect. Ther. 2013, 11, 13–23. [Google Scholar] [CrossRef] [PubMed]
  285. Obiero, J.; Mwethera, P.G.; Wiysonge, C.S. Topical microbicides for prevention of sexually transmitted infections. Cochrane Database Syst. Rev. 2012, 3, CD007961. [Google Scholar] [CrossRef]
  286. Sibille, G.; Pavan, M.; Mannino, G.; Frasson, I.; Salata, C.; Luganini, A.; Maffei, M.E.; Gribaudo, G. Manuscript in preparation. 2022. [Google Scholar]
  287. Howell, A.B.; Botto, H.; Combescure, C.; Blanc-Potard, A.B.; Gausa, L.; Matsumoto, T.; Tenke, P.; Sotto, A.; Lavigne, J.P. Dosage effect on uropathogenic Escherichia coli anti-adhesion activity in urine following consumption of cranberry powder standardized for proanthocyanidin content: A multicentric randomized double blind study. BMC Infect. Dis. 2010, 10, 94. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  288. Blumberg, J.B.; Camesano, T.A.; Cassidy, A.; Kris-Etherton, P.; Howell, A.; Manach, C.; Ostertag, L.M.; Sies, H.; Skulas-Ray, A.; Vita, J.A. Cranberries and their bioactive constituents in human health. Adv. Nutr. 2013, 4, 618–632. [Google Scholar] [CrossRef] [Green Version]
  289. Silverman, R.; Keller, J.M.; Glassman, A.; Chalkidou, K. Tackling the Triple Transition in Global Health Procurement. Available online: https://www.Cgdev.Org/better-health-procurement (accessed on 12 October 2022).
  290. Schweitzer, S.O.; Comanor, W.S. Prices of pharmaceuticals in poor countries are much lower than in wealthy countries. Health Aff. 2011, 30, 1553–1561. [Google Scholar] [CrossRef]
Figure 1. Structure of proanthocyanidins showing A-type and B-type interflavanic bonds and the position of A and C rings along with the numbers of carbons involved in the C–O bounds.
Figure 1. Structure of proanthocyanidins showing A-type and B-type interflavanic bonds and the position of A and C rings along with the numbers of carbons involved in the C–O bounds.
Molecules 27 08353 g001
Figure 2. Factor loading plot from the Principal Component Analysis (PCA) performed on data summarized in Table 1 and Table 2 by considering the different degrees of PAC-A polymerization and the three main biological effects. Antiviral activity is associated mainly with A-type PAC monomers, dimers, trimers and tetramers. Antioxidant activity is correlated with A-type PAC polymers with a degree of polymerization > 5. Antibacterial activity is correlated primarily to PAC-A dimers and trimers. Varimax rotation; total variance explained by the three factors: 26% factor (1), 24% factor (2) and 17% factor (3).
Figure 2. Factor loading plot from the Principal Component Analysis (PCA) performed on data summarized in Table 1 and Table 2 by considering the different degrees of PAC-A polymerization and the three main biological effects. Antiviral activity is associated mainly with A-type PAC monomers, dimers, trimers and tetramers. Antioxidant activity is correlated with A-type PAC polymers with a degree of polymerization > 5. Antibacterial activity is correlated primarily to PAC-A dimers and trimers. Varimax rotation; total variance explained by the three factors: 26% factor (1), 24% factor (2) and 17% factor (3).
Molecules 27 08353 g002
Figure 3. A proposed mechanism of action for the broad-spectrum antiviral activity of type-A PACs. (A) Tampering with virion capsid or envelope glycoproteins. (B) Masking/blocking virion binding sites to cellular receptors.
Figure 3. A proposed mechanism of action for the broad-spectrum antiviral activity of type-A PACs. (A) Tampering with virion capsid or envelope glycoproteins. (B) Masking/blocking virion binding sites to cellular receptors.
Molecules 27 08353 g003
Table 1. Natural sources of A-type PACs and biological properties.
Table 1. Natural sources of A-type PACs and biological properties.
Natural SourceA-Type PACsPropertiesReferences
Adansonia digitatadimersantioxidant[31]
Aesculus turbinataprocyanidins *antioxidant[32]
Aglaonema commutatum var. maculatumtrimerschemical composition[33]
Aglaonema crispumdimerschemical composition[33]
Arachis hypogaeadimers, trimerscardiovascular diseases, dyslipidemia
antioxidant
prevention of pathogen infection
chemical composition
anti-inflammatory
antioxidant
[34]
[35]
[36]
[21,37,38,39,40]
[41]
[42]
Areca catechudimerhypoglycemic[43]
Calluna vulgarisdimers, trimerschemical composition[44]
Cinnamomum cassiadimers, oligomersoxidative conversion of B- to A-procyanidins
antidiabetic
[45]
[46,47,48]
Cinnamomum japonicaoligomershypoglycemic[48]
Cinnamomum tamalaoligomersantidiabetic[47,49]
Cinnamomum zeylanicumtrimers, tetramersmultidrug resistance, biofilm inhibitory activity
antiasthmatic, antiallergic
anti-inflammatory
antiallergic
attenuation of the reduction in glutamate uptake
anti- vascular endothelial growth factor (VEGF)
antidiabetic
prevention of neurodegeneration
[50]
[51]
[52]
[53]
[54]
[55]
[56,57]
[58]
Coffea arabicatrimerschemical composition[59]
Crataegus pinnatifida var. majorprocyanidinsantioxidant[60]
Dimocarpus longandimerhealth-beneficial bioactivity[61]
procyanidinsantioxidant[62]
Ecdysanthera utilismonomers, dimersimmunomodulator[63]
Ephedra equisetinaprocyanidinschemical composition[64]
Ephedra intermediaprocyanidinschemical composition[64]
Ephedra sinicaprocyanidinschemical composition[64,65]
Gaultheria procumbenstrimersantioxidant[66,67]
Ixora coccineadimersantioxidant, antibacterial[68]
Laurus nobilistrimersantioxidant[69]
Litchi chinensisdimers, trimerschemical composition
antioxidant
cardioprotection
alteration of oligomers in the gastrointestinal system
bioavailability
antioxidant
absorption and urinary excretion
bacterial bioconversions
[70,71,72]
[73]
[74,75]
[76]
[77]
[78,79,80,81]
[82]
[83,84]
Lotus americanusprocyanidinschemical composition[85]
Malus domesticadimersantioxidant[86]
Mandevilla moricandianatrimersantioxidant[87]
Microbiota (faecal and gut)procyanidinsInability to cleave A-type linkages
preventing of biofilm formation
antioxidant
degradation by human gut microbiota
[88,89]
[90]
[91]
[92]
Paullinia cupanatrimersAnti-inflammatory, antioxidant[93,94]
Paullinia pinnatatrimers, tetramersantihelminthic[95,96]
Pelargonium sidoidestrimersantiadhesive[97]
Persea americanadimers, trimers, tetramers, procyanidinschemical composition[98,99]
Pheonix dactyliferadimerschemical composition[100]
Pinus massonianatrimers, tetramersincreased modulus of elasticity of dentin[101]
Pinus pinasterdimersantioxidant, bactericidal[102]
Polygonum cuspidatumdimerschemical composition[103,104]
Prunus domesticadimerschemical composition
antioxidant
[99,105]
[86]
Prunus dulcisprocyanidinschemical composition[106,107]
Prunus spinosadimers, trimerschemical composition[44,108]
Pteris vittataprocyanidinsantioxidant[109]
Pyracantha fortuneanaprocyanidinsantidiabetic[110]
Pyrus pyrifoliatrimerschemical composition[111]
Rhizophora apiculatamonomerschemical composition[112]
Rhizophora mangleprocyanidinschemical composition[113]
Rhododendron ferrugineumtrimersvitality and the proliferation rates of epithelial HaCaT keratinocytes[114]
Rhododendron formosanumtrimersinduction of autophagy
antioxidant
[115]
[116]
Ribes nigrumdimersbacterial growth and cell adhesion[117]
Rubus idaeusprocyanidinschemical composition[118]
Rumex obtusifoliustrimerschemical composition[119]
Spatholobus suberectusprocyanidinsantioxidants, inhibitor of breast cancer[120]
Tectaria macrodontatrimerschemical composition[33]
Theobroma cacaoprocyanidinschemical composition
absorption
antioxidant
[37,121,122]
[123]
[124]
Vaccinium asheidimers, dodecamerschemical composition[125]
Vaccinium consanguineum,monomers, dimers, trimers, tetramers, procyanidinschemical composition[126]
Vaccinium corymbosumtrimersantidiabetic[127]
Vaccinium floribundummonomers, dimers, trimers, tetramers, procyanidinschemical composition[126]
Vaccinium macrocarponmonomers, dimers, trimers, tetramers, procyanidinschemical composition
urinary tract infections (UTIs)
antiaging
bioavailability
transported across Caco-2 cells
antioxidant
cardiovascular health
immune system
[30,128,129,130,131,132,133,134,135,136]
[14,15,23,137,138,139,140,141,142,143,144,145,146,147]
[148]
[149]
[150]
[151]
[152]
[153]
Vaccinium myrtillusdimers, trimersantidiabetic
chemical composition
bacterial growth and cell adhesion
[127]
[44,154]
[117]
Vaccinium oxycoccusmonomers, dimers, trimers, tetramers, procyanidinschemical composition[135]
Vaccinium poasanummonomers, dimers, trimers, tetramers, procyanidinschemical composition[126]
Vaccinium vitis-idaeadimers, trimerschemical composition[44,135]
Vicia fabadimerschemical composition[155]
Vitis viniferadimers, trimers, tetramers, procyanidinschemical composition
inhibition of alpha-glucosidase
promotion of DNA repair in dendritic cells in UVB-exposed skin.
decreases the progression of airway inflammation
antioxidant
control of lipid metabolism
immune system
[156]
[157]
[158]
[159]
[160]
[161,162,163,164,165,166]
[167,168,169,170,171,172]
* The term procyanidins indicates A-type PACs with degrees of polymerization (DP) 5 < DP < 12.
Table 2. Antiviral activity of plants-derived PAC-As.
Table 2. Antiviral activity of plants-derived PAC-As.
Natural SourceA-Type PACsVirus and Mechanism of ActionReferences
Alpinia zerumbetprocyanidins *influenza A virus, inhibition of attachment, virucidal[173,174]
Chamaecrista nictitansprocyanidinsHSV-1 and HSV-2, NA [175]
Cinnamomum cassiadimers, oligomersHIV-1, interaction with envelope glycoproteins [176]
Cinnamomum zeylanicumtrimers, tetramersHIV-1, inhibition of attachment
HCV, inhibition of attachment
SARS-CoV, virucidal
[177]
[178]
[179]
Ixora coccineatrimersHIV-1, inhibition of Vpu activity; HCV, NA[180]
Litchi chinensisdimers, trimersHSV-1 and Coxsackie virus B3, NA[181]
Pinus maritimaprocyanidinsHIV-1, inhibition of entry and replication
HCV, inhibition of replication
[182]
[183]
Pomelia pinnatadimersHIV-1, inhibition of integrase activity[184]
Sambucus nigradimersHIV-1, interaction with envelope glycoproteins[176]
Theobroma cacaodimersHSV and HIV, NA[124]
Vaccinium macrocarponmonomers, dimers, trimers, tetramers, procyanidinsnairovirus, inhibition of attachment[185]
influenza A and B virus, inhibition of attachment[186,187]
and entry, virucidal
HSV-1 and HSV-2, inhibition of entry[188]
human norovirus surrogates: murine norovirus (MNV-1), feline calicivirus (FCV-F9), virucidal[189,190]
reovirus, NA[191]
rotavirus, inhibition of attachment, interaction
with capsid proteins
[192,193]
Vaccinium myrtillusdimers, trimersSARS-CoV-2, inhibition of entry and replication[194]
HA, NA[195]
HCV, inhibition of replication[196]
Vitis viniferadimers, trimers, tetramers, procyanidinsrotavirus, affecting virion integrity
HIV-1, inhibition of entry by down-modulation of
co-receptors
[193]
[197]
* The term procyanidins indicates A-type PACs with degrees of polymerization (DP) 5 < DP < 12; NA: not available.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Maffei, M.E.; Salata, C.; Gribaudo, G. Tackling the Future Pandemics: Broad-Spectrum Antiviral Agents (BSAAs) Based on A-Type Proanthocyanidins. Molecules 2022, 27, 8353. https://doi.org/10.3390/molecules27238353

AMA Style

Maffei ME, Salata C, Gribaudo G. Tackling the Future Pandemics: Broad-Spectrum Antiviral Agents (BSAAs) Based on A-Type Proanthocyanidins. Molecules. 2022; 27(23):8353. https://doi.org/10.3390/molecules27238353

Chicago/Turabian Style

Maffei, Massimo E., Cristiano Salata, and Giorgio Gribaudo. 2022. "Tackling the Future Pandemics: Broad-Spectrum Antiviral Agents (BSAAs) Based on A-Type Proanthocyanidins" Molecules 27, no. 23: 8353. https://doi.org/10.3390/molecules27238353

Article Metrics

Back to TopTop