Next Article in Journal
Gut Microbiome–Estrobolome Profile in Reproductive-Age Women with Endometriosis
Next Article in Special Issue
Effect of a Total Extract and Saponins from Astragalus glycyphyllos L. on Human Coronavirus Replication In Vitro
Previous Article in Journal
YKL-40 and the Cellular Metabolic Profile in Parkinson’s Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Kaempferol: A Review of Current Evidence of Its Antiviral Potential

by
Argyrios Periferakis
1,2,3,†,
Aristodemos-Theodoros Periferakis
1,3,†,
Lamprini Troumpata
1,
Konstantinos Periferakis
2,4,
Andreea-Elena Scheau
5,
Ilinca Savulescu-Fiedler
6,7,*,
Ana Caruntu
8,9,
Ioana Anca Badarau
1,
Constantin Caruntu
1,10,* and
Cristian Scheau
1,11
1
Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
2
Akadimia of Ancient Greek and Traditional Chinese Medicine, 16675 Athens, Greece
3
Elkyda, Research & Education Centre of Charismatheia, 17675 Athens, Greece
4
Pan-Hellenic Organization of Educational Programs (P.O.E.P), 17236 Athens, Greece
5
Department of Radiology and Medical Imaging, Fundeni Clinical Institute, 022328 Bucharest, Romania
6
Department of Internal Medicine, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
7
Department of Internal Medicine and Cardiology, Coltea Clinical Hospital, 030167 Bucharest, Romania
8
Department of Oral and Maxillofacial Surgery, “Carol Davila” Central Military Emergency Hospital, 010825 Bucharest, Romania
9
Department of Oral and Maxillofacial Surgery, Faculty of Dental Medicine, “Titu Maiorescu” University, 031593 Bucharest, Romania
10
Department of Dermatology, “Prof. N.C. Paulescu” National Institute of Diabetes, Nutrition and Metabolic Diseases, 011233 Bucharest, Romania
11
Department of Radiology and Medical Imaging, “Foisor” Clinical Hospital of Orthopaedics, Traumatology and Osteoarticular TB, 021382 Bucharest, Romania
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2023, 24(22), 16299; https://doi.org/10.3390/ijms242216299
Submission received: 16 October 2023 / Revised: 7 November 2023 / Accepted: 12 November 2023 / Published: 14 November 2023
(This article belongs to the Special Issue Antiviral Activities of Plant Extracts)

Abstract

:
Kaempferol and its derivatives are flavonoids found in various plants, and a considerable number of these have been used in various medical applications worldwide. Kaempferol and its compounds have well-known antioxidant, anti-inflammatory and antimicrobial properties among other health benefits. However, the antiviral properties of kaempferol are notable, and there is a significant number of experimental studies on this topic. Kaempferol compounds were effective against DNA viruses such as hepatitis B virus, viruses of the alphaherpesvirinae family, African swine fever virus, and pseudorabies virus; they were also effective against RNA viruses, namely feline SARS coronavirus, dengue fever virus, Japanese encephalitis virus, influenza virus, enterovirus 71, poliovirus, respiratory syncytial virus, human immunodeficiency virus, calicivirus, and chikungunya virus. On the other hand, no effectiveness against murine norovirus and hepatitis A virus could be determined. The antiviral action mechanisms of kaempferol compounds are various, such as the inhibition of viral polymerases and of viral attachment and entry into host cells. Future research should be focused on further elucidating the antiviral properties of kaempferol compounds from different plants and assessing their potential use to complement the action of antiviral drugs.

1. Introduction

Kaempferol (3,5,7-trihydroxy-2-(4-hydroxyphenyl)-4H-chromen-4-one) carries the name of Engelbert Kaempfer, a German doctor, naturalist, and historian [1]. It is a flavonoid which was initially discovered in Camelia sinensis [2] and is found in a great variety of plants [3]. The chemical compound has anticarcinogenic [4,5,6], anti-inflammatory [7], anti-adipogenic [8], hepatoprotective [9,10], cytoprotectivre/antioxidant [11], anti-venom [12], and antimicrobial effects [3]. Other polyphenols and phytochemicals share these types of effects (e.g., [13,14,15,16,17,18,19,20,21,22,23,24,25,26,27]), and plant phenolic extracts are under investigation for their medicinal uses [28,29,30,31,32,33].
The risk of viral diseases is ever increasing with the frequent emergence of new viral pathogens; meanwhile, effective antiviral treatments are in many cases non-existent or suboptimal [34]. The current state of antiviral treatment options reflects the need of novel solutions for combatting viral infections [35]. During the last few decades, the focus toward phytochemistry and phytomedicine has increased, as can be seen by the multitude of relevant scientific literature [36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59].
As mentioned, kaempferol and its derivatives show pronounced antibacterial effects along with a considerable antifungal and antiparasitic activity [3]. Coupled with the fact that flavonoids in general have been shown to exert a promising activity against viruses [60,61], a closer view on the antiviral activity of kaempferol is warranted. In this review, we will perform a thorough analysis of the available research on the antiviral properties of kaempferol and its derivatives (Figure 1) either in pure forms or as plant extract components. This approach aims to underline the contemporary perspective of this particular field and reveal potential avenues for future research.

2. Antiviral Activity against DNA Viruses

In general, it is believed that DNA viruses have been evolving for millions of years [62]. There exist 91 species of DNA viruses known, and of these, about 87% exhibit a degree of adaptation to the human host [63]. Well-known human pathogens, such as the herpes viruses, adenoviruses, poxviridae and human papillomaviruses, are included in this category. Despite vaccines being available, especially after the 1930s [64] against a number of DNA viruses, they nonetheless represent a significant health hazard. Research on the antiviral properties of kaempferol and its compounds is centred on African swine fever virus, hepatitis B virus, the alphaherpsevirinae, and pseudorabies virus (Table 1).

2.1. Antiviral Activity against Hepatitis B Virus (HBV)

HBV infection is a major healthcare concern worldwide [72,73,74] and is responsible for a considerable number of deaths [72,73,74]. The virus is transmitted through blood and other bodily fluids [73,74] and causes liver disfunction [75] and potentially chronic hepatitis [72,73,75]. There are several regimens available for treatment purposes [72,73] which significantly reduce the risk of occurrence of complications [72,73,74,75] though there is no evidence to suggest that treatment for the acute stage is effective. At any rate, tenofovir, entecavir, and pegylated interferon alfa-2a are the main agents applied against this pathogen [75]. As far as prevention is concerned, the vaccine currently in use has been successful in limiting the prevalence of the virus [73,74,75,76], and its effectiveness is particularly important for the main risk groups, young children and pregnant women [73,76].
Kaempferol has exhibited antiviral action against enveloped viruses, including hepatitis B [77]. Moreover, in an in vitro setting, Parvez et al. [66] noticed that anti-hepatitis B effects were displayed by the extract of the cold-adapted sea buckthorn Hippophae rhamnoides, which contains kaempferol. Kaempferol exerted these effects by inhibiting HBsAg and HBeAg synthesis (HBe is a marker for cccDNA replication and, by extension, the infectivity of the host). An in vitro and in silico molecular docking analysis proved the ability of quercetin, kaempferol and lamivudine to form stable complexes with HBV-polymerase binding-pocket amino acids and thus, the potential therapeutic potential of sea buckthorn was attributed to its extracts, quercetin, isorhamnetin and kaempferol [66]. The findings of Li et al. [65], which used the extract of Geranium carolinianum L., further support the anti-hepatitis B effects of kaempferol by displaying a reduction in intracellular viral DNA proportional to the administered dose.

2.2. Antiviral Activity against Alphaherpesvirinae

Herpes is among the oldest pathogens recorded, being known since the time of Ancient Greece [78,79]. The two serotypes HHV-1 and HHV-2 [79], alongside Varicella–Zoster virus, make up the Alphaherpesvirinae subfamily [79]. The virus is widely distributed [80,81], and it is responsible for a multitude of pathologies of varying severity [78,80,82,83,84]. The diseases most commonly associated with herpes simplex are facial and genital lesions [80,81] as well as a recurring and potentially eyesight-threatening keratitis [85], while the psychological implications of the aforementioned lesions constitute another important aspect that must be considered during treatment [80,81]. Recent advances like the use of PCR and the administration of acyclovir have enabled us to detect the infection reliably and help the host fend it off respectively [84]. Also, new studies have shown progress in the development of vaccines with the application of nanoparticles [86].
In the case of varicella-zoster, the primary infection causes a condition commonly known as chickenpox, with the virus establishing latency afterwards in the peripheral ganglia [87,88,89], with potential reactivation years later [87,88]. The ensuing condition, i.e., herpes zoster, is commonly accompanied by potent neuralgia and may lead to CNS complications [87]. PCR and acyclovir represent solid means of detecting and addressing the virus [88,90], and available vaccines against varicella-zoster virus provide protection from both the initial infection and its reactivation [88].
Based on the results of previous research, as summarised by Yang et al. [77], it was demonstrated that kaempferol has potent action against enveloped viruses, such as herpes. Two kaempferol derivatives, kaempferol 3-O-rutinoside and kaempferol 3-O-robinobioside, obtained from the extract of Ficus benjamina, were shown to have inhibiting action against both HHV-1 and HHV-2 but were not effective against varicella-zoster [67]. This effectiveness is corroborated by the findings of Behbahani et al. [68], which also display the potency of kaempferol and kaempferol-7-O-glucoside isolated from Securigera securidaca against HHV-1 due to the ability of the tested fractions to inhibit HSV-1 multiplication at a high rate. Interestingly, a flavonoid fraction from leaves of Ocotea notate, with kaempferol being one of its major compounds, exhibited notably antiherpetic action, particularly against HHV-2 by inhibiting the viral replication cycle at multiple levels [91].
In the case of varicella-zoster, Park et al. [69] looked into the potency of kaempferol compared to that of acyclovir; they found out that while the former could not inhibit the VZV entry into host cell and the activity of the VZV immediate-early promoter, it could block VZV DNA synthesis and VZV replication at similar time points. Since no blocking of early, late or immediate-early proteins synthesis was observed, the mechanism behind this is possibly attributed to inhibition of the viral DNA polymerase and/or cellular factors needed for the viral DNA replication.

2.3. Antiviral Activity against African Swine Fever Virus

African Swine Fever (ASF), a fatal disease in many cases, is a very contagious haemorrhagic disease [92] caused by the African Swine Fever Virus (ASFV) which originates in Africa but, due to the increased demands for swine consumption, it has found its way to other areas of the world [93]. Even though this virus poses no direct threat to human life, it is a very problematic factor for the pig industry, which in turn could have repercussions as far as the availability of pork meat is concerned as well as at an economic level [92]. Seeing as how vaccine development has yet to yield any noteworthy results [94], methods of treatment could be very useful in addressing the issue alongside the implementation of strict sanitary protocols.
ASFV targets monocytes and macrophages [93]. The virus enters the cells mainly via endocytosis, mediated by receptors, and via micropinocytosis [70]. Kaempferol’s potency in dealing with this pathogen stems from its ability to inhibit the endocytosis, thereby preventing the release of virions to the cell [70]. This results in the suppression of viral infection by an impressive amount of more than 90% [70]. The mechanism behind this has not been verified, but it may be related to the induction of autophagy in different cell lines due to the upregulation of p-AMP-activated protein kinase [70].

2.4. Antiviral Activity against Pseudorabies Virus

This virus, also known as Suid alphaherspesvirus 1, is the causative agent of Aujeszky’s disease, which is associated with significant financial losses in the pig industry [71,95,96,97]. Even though currently pigs are its only known reservoir, it exhibits notable recombination abilities [97] and can thus affect several other mammals [96,97] with a few cases of even human infection, in the form of endophthalmitis [98] and encephalitis [99], having been recorded. DIVA (differentiating infected from vaccinated animals) vaccines have been used to address this pathogen with impressive results [100]. However, the wild swine remain a possible reservoir, and research has been inconclusive as to the risk they pose in this regard [96]. It should also be noted that on an experimental level, this virus has been used alongside other neurotropic viruses to study brain organization, as a tracer of neural pathways, and the role that virally encoded proteins play in viral invasiveness and virulence [95,101].
Kaempferol could be potentially used to treat PRV infection as it has been shown to inhibit the virus’ replication in a dose-dependent manner when tested in vitro [71]. Specifically, at a concentration of 52.40 µmol/L, it decreased cell death caused by PRV by 90%, having an IC50 of 25.57 µmol/L [71]. Kaempferol also exhibited inhibitory action on the stage of viral penetration though at a lesser extent, reducing the viral loads by 4-fold when compared with the 30-fold reduction in the case of replication [71].

3. Antiviral Activity against RNA Viruses

The RNA viruses group comprises some of the most dangerous viral pathogens, such as dengue fever virus, poliomyelitis virus, and human immunodeficiency virus (HIV). The low fidelity of their polymerases and reverse transcriptase ensures a high mutagenicity, and thus, they exhibit much greater diversification compared to DNA viruses; to date, 158 species of RNA viruses are known to exist [102] of which a handful is pathogenic to humans [63].
While successful vaccines against some of these viruses do exist, it still remains important to explore alternative treatment options, given the existence of immunocompromised individuals and the potential for mutations. There is relevant literature data to support the antiviral properties of kaempferol and its compounds against RNA viruses (Table 2).

3.1. Antiviral Activity against Severe Acute Respiratory Syndrome-Related Coronaviruses

Severe acute respiratory syndrome coronaviruses (SARS-CoV) are extremely contagious [118,119,120,121], and SARS-CoV-2 was the cause for the most recent pandemic, which started on 2019 and escalated into a global crisis [122]. It gains entry to the cells of the respiratory system through the binding of protein S to the angiotensin-converting enzyme [118,119,123], but apart from its main target, it can spread to several other anatomical sites [124], such as the brain [125]. Consequently, the virus manifests in several different ways; from acute respiratory distress syndrome [119,123,124,125] to gastrointestinal symptoms and kidney involvement [123,124] as well as hepatic involvement, myocardial injury, and severe coagulopathy, and it even affects the CNS [123,125,126]. Having the largest genome among all RNA viruses, SARS-CoV-2 is equipped with the means of thriving in a wide range of environmental conditions and hosts [122]. Due to its high recombination potential [125], new ways of addressing the infection must be sought after, especially given that treatment options are still limited [122]. The vaccines which were used en masse may have been mostly safe and quite efficient [127]; however, there have been reported cases of vaccine-associated pathologies [128,129,130,131].
A number of studies have assessed the efficacy of kaempferol in combating SARS-CoV-2 [132,133]. In 2014, Schwarz et al. [103] determined that numerous kaempferol glycosides were effective in blocking the 3a channels of coronaviruses in concentrations as low as 20 μΜ. This mechanism is effective against the original SARS-COV-1 outbreak of 2002–2004 [43]. Both kaempferol and isokaempferide were evaluated for their antiviral potential against COVID-19 using a computational approach and were found to be acceptable as potential antiviral agents [134]; both compounds were extracted from Artemisia annua. A number of kaempferol compounds extracted from Salvadora persica exhibited promising molecular docking parameters on the N3 site in the SARS-CoV-2 main protease (Mpro) [135]. Equally promising molecular docking parameters of kaempferol extracted from Mollungo Nudicaulis were confirmed by Kanmani [136].

3.2. Antiviral Activity against Respiratory Syncytial Virus (RSV)

This pathogen is the main causative agent of pneumonia and bronchitis [137] as well as bronchiolitis [138,139], particularly in children and the elderly [139], and it can lead to long-term pulmonary complications [139,140]. Vaccine development was arduous with the first unsuccessful attempt being made more than fifty years ago [139], although the current research results seem promising, especially since reinfection has been well documented and treatment remains for the most part supportive to this day [138,139,141].
Both kaempferol-3-O-β-D-glucopyranosyl (12)-α-L-rhamnoside and kaempferol-3-O-α-L-rhamnoside, derived from Eucalyptus citriodora leaves, proved effective against this virus but with a slightly lower selectivity compared to ribavirin [104]. In addition, tested kaempferol derived from tea proved effective against RSV, being able to inhibit viral cytopathic effects by 50% at a concentration of 4.84 μg/mL [77].

3.3. Antiviral Activity against Influenza Virus

The influenza virus is a very contagious pathogen whose main target is the respiratory system [142]. One of this virus’ most notable traits is its antigenic variability, which is most prevalent in Type A, resulting in several pandemics [143] caused by the exhibited antigenic shift [144], while its antigenic drift forces us to update the vaccines against it on an annual basis [145]. A variety of antiviral drugs are in use in influenza infections [146]; however, not only do these drugs become less effective if they are not administered shortly after the onset of the infection [143], but the virus has developed resistance to several of them [143,146]. As such, the need for additional combative substances is ever present.
Kaempferol and a host of other flavonoids are effective viral neuraminidase inhibitors [147]. The subsequent research of Jeong et al. [105] on the extract from Rhodiola rosea roots determined that kaempferol was an effective antiviral agent with an EC50 of 18.50–32.50 μM depending on the viral strain. This neuraminidase inhibition was also noted by Yang et al. [77]. Interference with the hemagglutinin domains has also been noted [77].
Two kaempferol compounds from the extract of Eupatorium perfoliatum L. were found to be effective against influenza A, although their precise contribution to the total antiviral potential of the extract was unspecified [107]. The action mechanism of the extract consisted in obstructing viral attachment and entry into the cells. The earlier research of Park et al. [148] had indicated the antiviral potential of the extract of Aronia melanocarpa against influenza; while the extract contains kaempferol, its exact contribution to the antiviral properties of the extract is not known. Finally, the extensive research of Kai et al. [106] on kaempferol from Brazilian propolis indicated its effectiveness in supressing viral growth in the respiratory tract with an EC50 of 21.70–38.20 μM depending on the viral strain by exhibiting its potency in preventing mean body weight reduction and increasing the survival rate of infected mice overall; this effectiveness was not affected by whether the strain had developed drug resistance or not.

3.4. Antiviral Activity against Human Immunodeficiency Virus (HIV)

It is estimated that around 39 million people were living with HIV at the end of 2022 around the world [149]. The most detrimental effect of HIV is the eventual enfeeblement of the patient’s immune system, due to the mass depletion of CD4+ helper cells, which is a state commonly known as AIDS (Auto Immune Deficiency Syndrome) [150,151]. A problematic aspect of this pathogen is that being a “retrovirus”, HIV uses reverse transcriptase to copy its RNA genome into DNA, showing great variability allowing for viral strains to elude the immune response of the host and making it seemingly impossible to vaccinate against [150,152]. Lifelong treatment schemes called ART (Antiretroviral Therapy) [153,154] and prophylactic schemes are now available [154,155]; therefore, our ability to mitigate the consequences of infection with this pathogen have vastly improved [154], especially if the treatment begins during the early stages [153]. On the other hand, reduced adherence to therapy, for a variety of factors [156,157,158], reduces its effectiveness [159]. For this and the other reasons outlined above, it has proven impossible to completely cure HIV carriers with a few exceptions [160].
There is some evidence to suggest that kaempferol could be used effectively as an anti-HIV agent. Behbahani et al. [108] extracted kaempferol and kaempferol-7-O-glucoside from Securigera securidaca and determined that it potently inhibited the activity of the viral reverse transcriptase with the kaempferol glycoside being more potent in that regard. A more miscellaneous action of kaempferol against HIV is mentioned by Badshah et al. [43] and consists of its ability to inhibit the Vpu (Viral protein u)-mediated current of HIV-1 (an ion channel that is involved in virus release when activated) by a small amount of 10% at a concentration of 20 μM in a manner similar to that of genistein [113].

3.5. Antiviral Activity against Dengue Fever Virus (DFV)

This pathogen has a zoonotic transmission [161], and infections caused by it vary in severity; they can be subclinical and self-limiting or cause notable fever, which can be of haemorrhagic nature in the more severe cases, potentially leading to dengue shock syndrome [161,162]. There is no dedicated antiviral treatment; however, several vaccines have been developed for children and adolescents living in endemic areas as well as subjects previously confirmed with dengue fever [163,164,165,166]. Therefore, it is of great importance to conduct research with the purpose of identifying new substances which can be used against the virus.
Kaempferol 3-O-β-rutinoside seems to be effective in inhibiting dengue virus which was attributed to its ability to interfere with molecular docking [103]. Indicatively, even at a concentration of 10 μΜ, it achieved an inhibition of 55.60% and an inhibition of 77.7% at a concentration of 100 μΜ [110]. Conversely, the earlier research of Care at al. [109] had determined that not only kaempferol was ineffective against this particular virus, but it even increased infectivity in a particular cell line. This highlights a significant aspect, namely that of different interactions between kaempferol and presumably other flavonoids with the different molecular replication strategies of different viruses.

3.6. Antiviral Activity against Japanese Encephalitis Virus (JEV)

Japanese encephalitis is a zoonotic disease that is endemic in regions of the Eastern hemisphere [167] but is nevertheless of global concern due to its potential to spread in new regions; indicatively, its genetic material was recently found in mosquitos in northern Italy [168]. The causative agent is a neurotrophic virus of the flavivirus genus by the same name [169] which has five circulating genotypes [167] and leads to death of the infected individual in approximately one-third of cases, with half of the surviving patients suffering from neuronal complications long after the infection is over [170]. Initially, a mouse brain-derived inactivated vaccine was available, but it has been replaced by three cell culture-derived vaccines [171].
In general, flavonoids seem to be promising antiviral agents against JEV [70,111,172]. Kaempferol in particular was found to be able to bind with the frame-shift site RNA (fsRNA) in the JEV serogroup and thereby interfere with protein expression and consequently viral replication [111]. The antiviral action of kaempferol against this virus was also corroborated by Care et al. [109], even though higher concentrations of kaempferol were required, and this was above the toxicity limit for one of the two cell lines used.

3.7. Antiviral Activity against Enterovirus 71 (EV71)

Enterovirus 71 is the main causative agent of the so-called “hand, foot, and mouth disease” [173,174,175] alongside coxsackieviruses [174], which is a condition that affects mostly children [173,175,176], having neurological manifestations [173,175,176], and can lead to complications both neurologic [175] and systemic [173,175], like meningoencephalitis and pulmonary pathologies, respectively. Currently, there are formalin-inactivated (FI) EV71 vaccines which have undergone evaluation in human clinical trials in China, Taiwan, and Singapore and have been deemed safe and effective in eliciting neutralizing antibody responses against the virus [174].
Kaempferol can be potentially useful against enterovirus 71 infections because it hinders both its replication and its translation through its interaction with EV71 internal ribosome entry site (IRES) [112]. Indicatively, PCBP 1/2 and the viral polymerase precursor 3CD with poliovirus IRES stem-loop I RNA contribute significantly to the antiviral activity by forming a ternary complex required for the synthesis of negative-strand RNA while PCBP2 interacts with enterovirus IRES stem-loop IV RNA thus inhibiting IRES-mediated translation [112]. Kaempferol’s action against enterovirus 71 is made possible due to the contribution of cellular factors associated with the 50-untranslated region (50-UTR) of the EV71 genome [112].

3.8. Antiviral Activity against Hepatitis A Virus (HAV)

Hepatitis A is the most frequent type of viral hepatitis and is showing an increasing incidence especially in low and middle demographic index regions [177]. The faecal–oral transmission shows it is oftentimes linked to poor sanitary conditions [178]. The infection can have many clinical forms, from being short-lived and self-limiting, without displaying any symptoms [179] or displaying abdominal pain, hepatitis, hyperbilirubinemia, and jaundice [178] or only jaundice, to being seldom fatal [179]. Intramuscular anti-A gamma globulin is used for passive immune prophylactic purposes [179], but the most efficient preventive method is the available vaccine [178,179,180].
As mentioned in the research of Ohemu et al. [181], a variety of plant flavonoids, presumably including kaempferol, showed an antiviral effect against HAV. However, Orabi and Orabi [114] tested kaempferol-3-O-α-D-arabinopyranoside and kaempferol-3-O-β-D-galactopyranoside isolated from the extract of Ficus virens; their antiviral activity against HAV was found to be practically non-existent. Both compounds were tested up to their maximum non-toxic concentration on the tested cell line.

3.9. Antiviral Activity against Poliovirus

Poliovirus is a virus with three subtypes (1, 2 and 3), which is responsible for the condition known as poliomyelitis [182]. This enterovirus is highly contagious and can have either a paralytic form (most often spinal) or a bulbar form [182]. In addition to the dangerous nature of the acute phase, this virus remains a problem for the host’s health for a long time after the initial infection due to post-polio syndrome, which is a neurological disorder that significantly impairs his daily life [182,183,184], requiring long-term medical assistance to handle [183]. The vaccines of Salk and Sabin were a major step toward limiting the impact of this pathogen [182,184]. Apart from being effective, they were also relatively safe [185,186] even though some cases of vaccine-derived polio have been recorded [185].
Based on the research of Robin et al. [115], 3-methylkaempferol (also known as isokaempferide) and 3,4-dimethylkaempferol, two compounds derived from the leaves of Psiadia dentata, have been found to be very effective in preventing viral replication of the second subtype by inhibiting RNA synthesis of the plus-strand if administered shortly after the infection.

3.10. Antiviral Activity against Chikungunya Virus (CHIKV)

This pathogen has a zoonotic transmission and causes fever, often accompanied by a maculopapular rash, culminating into polyarthralgia of potentially chronic nature [187,188,189], which can even cause bone erosion [189], while it has also been associated with Guillain–Barré syndrome [190]. Although the disease is usually not life-threatening, some patients develop chronic joint pain possibly due to the migration of infected monocytes to the synovial tissues, perpetuating the inflammatory process [188,189]. Given that all attempts at creating a vaccine have not been successful thus far [189] and that there is no specific treatment available [187,188,190], with broad-spectrum antivirals like ribavirin currently comprising the most effective therapeutic scheme available [189,191], research has focused on finding natural compounds with action against this virus [190].
Kaempferol with the oxidation and cyclization of 1,3,5-trimethoxybenzene was found to inhibit replication after the entry of the virus at a concentration > 400 µM [191]. As such, kaempferol could be considered a possible CHIKV inhibitor for human use. On the other hand, based on the research of Lani et al. [116], kaempferol is not a good candidate as an antiviral agent against CHIKV, because even though it exhibits a degree of inhibition of post-viral replication [191], it does so at a very high concentration of 400 µM [116].

3.11. Antiviral Activity against Feline Calicivirus (FCV)

This pathogen is among the most common ones found in cats [192,193] causing sporadic outbreaks in that population [194]. It has a highly mutagenic nature with its great genomic plasticity making the diagnosis, treatment, and prevention quite challenging. Infecting the upper respiratory tract [193,194], it is commonly accompanied by oral ulcers and high fever [195], and it can even be fatal [192]. Treatment has only a supportive role [195] and, despite the effectiveness of the vaccine [193,195], the virus’ variability means that there are strains against which the protection granted is not as potent [192]. Although this particular virus is not pathogenic to humans, it belongs to the Caliciviridae family, which comprises notable human pathogens [196]. Therefore, the development of antiviral strategies against this virus may offer new insights against human pathogens.
The research of Seo et al. [117] shows that kaempferol was the most potent antiviral agent amongst a series of tested flavonoids; compared to the standard antiviral agent ribavirin, it proved by far less effective at a concentration of 50 μM and somewhat less effective at 100 μM. The inhibition by kaempferol was remarkably high at concentrations of 200 μM and 300 μM [117]. An important component of kaempferol’s effectiveness seems to be its antioxidant activity, which surpasses even that of ribavirin [110].

3.12. Antiviral Activity against Murine Norovirus (MNV)

This recently discovered pathogen affects mice, causing gastroenteritis [197]. There also is a strain which can affect humans, called human norovirus, which is the main causative agent of viral gastroenteritis worldwide [198,199]. Since the mechanisms of the human norovirus have yet to be fully elucidated, despite recent advances [198,199], the study of the murine norovirus could yield results that may culminate into a breakthrough, especially since the latter is unique among all the strains in its ability to replicate in a cell culture [197].
While belonging in the same family as FCV, murine norovirus was not inhibited by kaempferol at either 50 μM or 100 μM despite other flavonoids such as daidzein and quercetin being effective against it [117].

4. Effectiveness of Kaempferol Antiviral Activity

Another aspect we should consider is the relative effectiveness of kaempferol and its derivatives compared to existing antiviral drugs (Table 3). In a large number of cases, the compounds tested are of comparable effectiveness to the drugs used, which is important when considering the potential adverse effects of the antiviral drugs. Thus, it might be possible to partially supplant or supplement these drugs, with kaempferol either alone or in conjunction with other natural antiviral compounds, so as to reduce the dose and/or the length of administration of some antiviral drugs, especially in the case where serious side effects are manifested.

5. General Antiviral Activity and Natural Kaempferol Sources with Antiviral Effects

From all the aforementioned research results on the antiviral effects of kaempferol, it must become apparent that kaempferol and its derivatives are potent as antiviral agents against most of the viruses presented (Figure 2). In each experiment against DNA viruses, every compound tested was effective at reasonably low concentrations, which were below the toxicity limit for the cell lines tested by the same researchers. Regarding kaempferol toxicity, there is currently no consensus as to its safety profile in vivo [7]; in vitro studies [200,201,202] have found it to be genotoxic, but this effect was not replicated in vivo [203].
In the case of RNA viruses, the results are again encouraging, with kaempferol and its derivatives being able to either inhibit viral replication altogether or mitigate the cytopathic effects and inhibit the entry of viruses into the cells. Notable exceptions include the experiment of Care et al. [109] against dengue, the experiments against HAV [114] and the experiments of Sauter et al. [113] against HIV; in all cases, the compounds used were ineffective. In the case of dengue virus, it is important to explore the potential of other kaempferol compounds, given that it represents a notable public health hazard in certain countries, and vaccination was not yet successful [204]. It is worth noting that kaempferol seems to exhibit some sort of cell specificity [109]. In this study, kaempferol was even found to have a pro-viral effect, which was most probably associated with the presence of a functional IRES sequence in Dengue virus [205]. In the case of HIV, kaempferol elicited only a minor inhibition of the Vpu channel [113]. Finally, in the case of kaempferol compounds against HAV, the cause for the lack of any noticeable antiviral activity appears to be related to structural and molecular dynamics reasons [114]. The ability of kaempferol and its derivatives to inhibit viral replication was identified and described for both DNA and RNA viruses; an illustration of the reported effects is shown in Figure 3.
Kaempferol is abundant in nature, being found in many plants (e.g., [206,207,208,209,210,211,212,213,214,215,216,217]; for a comprehensive review, see the paper of Periferakis et al. [3]). It is synthesised via the shikimic acid pathway [218] as all flavonoids; this biosynthetic pathway may be metabolically engineered so that it can yield the desired flavonol amounts in the near future [219]. Indeed, the microbial synthesis of kaempferol, as an alternative to its purification from plants, has already been proposed by Duan et al. [220]. Regarding its absorption in the human body, kaempferol is usually absorbed as a glycoside, although the attached sugars impact its bioavailability [221]. The specific pharmacokinetics of kaempferol have been explored by a number of researchers [222,223,224,225,226,227,228,229,230].
A prominent detail readily apparent based on the data presented in this review is the importance of plants used in traditional and folk medicine systems since ancient times. The mentioned plants belong to one or several such medical traditions (Table 4), and most importantly, the majority already demonstrated antimicrobial/antiviral properties with the exception of Securigera securidaca. This highlights the fact that a persistent and detailed study of traditional remedies can yield very useful insights into bioactive compounds for a number of different pathologies.
The possibility of virus–host genetic recombination coupled with the viral adaptability may make it plausible that a number of viruses may yet circulate with the human population unnoticed until specific and characteristic symptoms are manifested [102]. Alongside the existing viruses against which no effective vaccines are available, the need for an intensification of research on the antiviral properties of natural compounds is evident. Kaempferol appears to be a strong candidate due to its effectiveness, variety of action mechanisms, and general availability. Moreover, in an effort to enhance tissue availability, selectivity and effectiveness, kaempferol and associated compounds could be combined with nanoparticles, which have promising medical applications [265,266,267]. Another interesting future research avenue would be to combine kaempferol with other phytochemicals with known antiviral properties such as catechins [268,269,270], quercetin [271,272,273], plant alkaloids [55,274,275,276], and other natural compounds [277,278,279].
A variety of zoonotic viruses have the potential to emerge and infect human populations; therefore, the surveillance of infections and nonhuman reservoirs is recommended [280]. Further research on the antiviral properties of kaempferol on viruses from these reservoirs could be beneficial in establishing its effectiveness and potential use alongside other antiviral drugs.

Author Contributions

Conceptualization, A.P., A.-T.P., L.T., K.P., C.C. and C.S.; resources, K.P., A.-E.S., I.S.-F., A.C. and I.A.B.; writing—original draft preparation, A.P., A.-T.P., L.T., K.P., A.-E.S., I.S.-F., A.C., I.A.B., C.C. and C.S.; writing—review and editing, A.P., A.-T.P., L.T., C.C. and C.S.; supervision, K.P., I.S.-F., C.C. and C.S. All authors have read and agreed to the published version of the manuscript.

Funding

Publication of this paper was supported by the University of Medicine and Pharmacy Carol Davila through the institutional program Publish not Perish.

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. Periferakis, A.; Periferakis, K. On the Dissemination of Acupuncture to Europe. JournalNX 2020, 6, 201–209. [Google Scholar]
  2. Farombi, E.O.; Akinmoladun, A.C.; Owumi, S.E. Anti-cancer Foods: Flavonoids. In Encyclopedia of Food Chemistry; Melton, L., Shahidi, F., Varelis, P., Eds.; Academic Press: Oxford, UK, 2019; pp. 224–236. [Google Scholar] [CrossRef]
  3. Periferakis, A.; Periferakis, K.; Badarau, I.A.; Petran, E.M.; Popa, D.C.; Caruntu, A.; Costache, R.S.; Scheau, C.; Caruntu, C.; Costache, D.O. Kaempferol: Antimicrobial Properties, Sources, Clinical, and Traditional Applications. Int. J. Mol. Sci. 2022, 23, 5054. [Google Scholar] [CrossRef] [PubMed]
  4. Neuhouser, M.L. Dietary flavonoids and cancer risk: Evidence from human population studies. Nutr. Cancer 2004, 50, 1–7. [Google Scholar] [CrossRef] [PubMed]
  5. Scheau, C.; Mihai, L.; Bădărău, I.; Caruntu, C. Emerging applications of some important natural compounds in the field of oncology. Farmacia 2020, 68, 984–991. [Google Scholar] [CrossRef]
  6. Weng, C.J.; Yen, G.C. Flavonoids, a ubiquitous dietary phenolic subclass, exert extensive in vitro anti-invasive and in vivo anti-metastatic activities. Cancer Metastasis Rev. 2012, 31, 323–351. [Google Scholar] [CrossRef]
  7. Alam, W.; Khan, H.; Shah, M.A.; Cauli, O.; Saso, L. Kaempferol as a Dietary Anti-Inflammatory Agent: Current Therapeutic Standing. Molecules 2020, 25, 4073. [Google Scholar] [CrossRef] [PubMed]
  8. Park, U.H.; Hwang, J.T.; Youn, H.; Kim, E.J.; Um, S.J. Kaempferol antagonizes adipogenesis by repressing histone H3K4 methylation at PPARγ target genes. Biochem. Biophys. Res. Commun. 2022, 617, 48–54. [Google Scholar] [CrossRef]
  9. Alkandahri, M.Y.; Pamungkas, B.T.; Oktoba, Z.; Shafirany, M.Z.; Sulastri, L.; Arfania, M.; Anggraeny, E.N.; Pratiwi, A.; Astuti, F.D.; Dewi, S.Y.; et al. Hepatoprotective Effect of Kaempferol: A Review of the Dietary Sources, Bioavailability, Mechanisms of Action, and Safety. Adv. Pharmacol. Pharm. Sci. 2023, 2023, 1387665. [Google Scholar] [CrossRef]
  10. Xiao, L.; Xu, G.; Chen, S.; He, Y.; Peng, F.; Yuan, C. Kaempferol ameliorated alcoholic liver disease through inhibiting hepatic bile acid synthesis by targeting intestinal FXR-FGF15 signaling. Phytomedicine 2023, 120, 155055. [Google Scholar] [CrossRef]
  11. Baňas, Š.; Benko, F.; Ďuračka, M.; Lukáč, N.; Tvrdá, E. Kaempferol Enhances Sperm Post-Thaw Survival by Its Cryoprotective and Antioxidant Behavior. Stresses 2023, 3, 687–700. [Google Scholar] [CrossRef]
  12. Ajisebiola, B.S.; Oladele, J.O.; Adeyi, A.O. Kaempferol from Moringa oleifera demonstrated potent antivenom activities via inhibition of metalloproteinase and attenuation of Bitis arietans venom–induced toxicities. Toxicon 2023, 233, 107242. [Google Scholar] [CrossRef] [PubMed]
  13. Nwankwo, J.O.; Tahnteng, J.G.; Emerole, G.O. Inhibition of aflatoxin B1 genotoxicity in human liver-derived HepG2 cells by kolaviron biflavonoids and molecular mechanisms of action. Eur. J. Cancer Prev. 2000, 9, 351–361. [Google Scholar] [CrossRef] [PubMed]
  14. Snijman, P.W.; Swanevelder, S.; Joubert, E.; Green, I.R.; Gelderblom, W.C.A. The antimutagenic activity of the major flavonoids of rooibos (Aspalathus linearis): Some dose–response effects on mutagen activation–flavonoid interactions. Mutat. Res./Genet. Toxicol. Environ. Mutagen. 2007, 631, 111–123. [Google Scholar] [CrossRef] [PubMed]
  15. Caruntu, C.; Negrei, C.; Ilie Ghita, M.; Caruntu, A.; Bădărău, A.I.; Ioan, B.; Boda, D.; Albu, A.; Brănişteanu, D. Capsaicin, a hot topic in skin pharmacology and physiology. Farmacia 2015, 63, 487–491. [Google Scholar]
  16. Farombi, E.O.; Shrotriya, S.; Surh, Y.-J. Kolaviron inhibits dimethyl nitrosamine-induced liver injury by suppressing COX-2 and iNOS expression via NF-κB and AP-1. Life Sci. 2009, 84, 149–155. [Google Scholar] [CrossRef]
  17. Scheau, C.; Caruntu, C.; Badarau, I.A.; Scheau, A.-E.; Docea, A.O.; Calina, D.; Caruntu, A. Cannabinoids and Inflammations of the Gut-Lung-Skin Barrier. J. Pers. Med. 2021, 11, 494. [Google Scholar] [CrossRef]
  18. Choudhary, N.; Bijjem, K.R.V.; Kalia, A.N. Antiepileptic potential of flavonoids fraction from the leaves of Anisomeles malabarica. J. Ethnopharmacol. 2011, 135, 238–242. [Google Scholar] [CrossRef]
  19. Olaleye, M.T.; Amobonye, A.E.; Komolafe, K.; Akinmoladun, A.C. Protective effects of Parinari curatellifolia flavonoids against acetaminophen-induced hepatic necrosis in rats. Saudi J. Biol. Sci. 2014, 21, 486–492. [Google Scholar] [CrossRef]
  20. Ghita, M.A.; Caruntu, C.; Rosca, A.E.; Caruntu, A.; Moraru, L.; Constantin, C.; Neagu, M.; Boda, D. Real-Time Investigation of Skin Blood Flow Changes Induced by Topical Capsaicin. Acta Dermatovenerol. Croat. 2017, 25, 223–227. [Google Scholar]
  21. Athira, K.V.; Madhana, R.M.; Lahkar, M. Flavonoids, the emerging dietary supplement against cisplatin-induced nephrotoxicity. Chem.-Biol. Interact. 2016, 248, 18–20. [Google Scholar] [CrossRef]
  22. Dumitrache, M.D.; Jieanu, A.S.; Scheau, C.; Badarau, I.A.; Popescu, G.D.A.; Caruntu, A.; Costache, D.O.; Costache, R.S.; Constantin, C.; Neagu, M.; et al. Comparative effects of capsaicin in chronic obstructive pulmonary disease and asthma (Review). Exp. Ther. Med. 2021, 22, 917. [Google Scholar] [CrossRef] [PubMed]
  23. Vázquez-Flores, L.F.; Casas-Grajales, S.; Hernández-Aquino, E.; Vargas-Pozada, E.E.; Muriel, P. Chapter 47—Antioxidant, Antiinflammatory, and Antifibrotic Properties of Quercetin in the Liver. In Liver Pathophysiology; Muriel, P., Ed.; Academic Press: Boston, MA, USA, 2017; pp. 653–674. [Google Scholar] [CrossRef]
  24. Popescu, G.D.A.; Scheau, C.; Badarau, I.A.; Dumitrache, M.D.; Caruntu, A.; Scheau, A.E.; Costache, D.O.; Costache, R.S.; Constantin, C.; Neagu, M.; et al. The Effects of Capsaicin on Gastrointestinal Cancers. Molecules 2020, 26, 94. [Google Scholar] [CrossRef] [PubMed]
  25. Cai, H.-D.; Su, S.-L.; Qian, D.-W.; Guo, S.; Tao, W.-W.; Cong, X.D.; Tang, R.; Duan, J.-A. Renal protective effect and action mechanism of Huangkui capsule and its main five flavonoids. J. Ethnopharmacol. 2017, 206, 152–159. [Google Scholar] [CrossRef] [PubMed]
  26. Yi, Y.-S. Regulatory Roles of Flavonoids in Caspase-11 Non-Canonical Inflammasome-Mediated Inflammatory Responses and Diseases. Int. J. Mol. Sci. 2023, 24, 10402. [Google Scholar] [CrossRef]
  27. Sitarek, P.; Kowalczyk, T.; Śliwiński, T.; Hatziantoniou, S.; Soulintzi, N.; Pawliczak, R.; Wieczfinska, J. Leonotis nepetifolia Transformed Root Extract Reduces Pro-Inflammatory Cytokines and Promotes Tissue Repair In Vitro. Int. J. Environ. Res. Public Health 2023, 20, 4706. [Google Scholar] [CrossRef]
  28. Dai, J.; Mumper, R.J. Plant phenolics: Extraction, analysis and their antioxidant and anticancer properties. Molecules 2010, 15, 7313–7352. [Google Scholar] [CrossRef]
  29. Tungmunnithum, D.; Thongboonyou, A.; Pholboon, A.; Yangsabai, A. Flavonoids and Other Phenolic Compounds from Medicinal Plants for Pharmaceutical and Medical Aspects: An Overview. Medicines 2018, 5, 93. [Google Scholar] [CrossRef]
  30. Mehmood, A.; Javid, S.; Khan, M.F.; Ahmad, K.S.; Mustafa, A. In vitro total phenolics, total flavonoids, antioxidant and antibacterial activities of selected medicinal plants using different solvent systems. BMC Chem. 2022, 16, 64. [Google Scholar] [CrossRef]
  31. Sun, W.; Shahrajabian, M.H. Therapeutic Potential of Phenolic Compounds in Medicinal Plants—Natural Health Products for Human Health. Molecules 2023, 28, 1845. [Google Scholar] [CrossRef]
  32. Islam, S.; Salekeen, R.; Ashraf, A. Computational screening of natural MtbDXR inhibitors for novel anti-tuberculosis compound discovery. J. Biomol. Struct. Dyn. 2023, 1–11. [Google Scholar] [CrossRef]
  33. Mejía-Méndez, J.L.; Bach, H.; Lorenzo-Leal, A.C.; Navarro-López, D.E.; López-Mena, E.R.; Hernández, L.R.; Sánchez-Arreola, E. Biological Activities and Chemical Profiles of Kalanchoe fedtschenkoi Extracts. Plants 2023, 12, 1943. [Google Scholar] [CrossRef] [PubMed]
  34. Kapoor, R.; Sharma, B.; Kanwar, S. Antiviral phytochemicals: An overview. Biochem. Physiol. 2017, 6, 7. [Google Scholar] [CrossRef]
  35. Biswas, D.; Nandy, S.; Mukherjee, A.; Pandey, D.K.; Dey, A. Moringa oleifera Lam. and derived phytochemicals as promising antiviral agents: A review. S. Afr. J. Bot. 2020, 129, 272–282. [Google Scholar] [CrossRef]
  36. Todorov, D.; Hinkov, A.; Shishkova, K.; Shishkov, S. Antiviral potential of Bulgarian medicinal plants. Phytochem. Rev. 2014, 13, 525–538. [Google Scholar] [CrossRef]
  37. Moyankova, D.; Hinkov, A.; Georgieva, D.; Shishkov, S.; Djilianov, D. Inhibitory effect of extracts from haberlea rhodopensis Friv. Against herpes simplex virus. Comptes Rendus L’académie Bulg. Sci. 2014, 67, 1369–1376. [Google Scholar]
  38. Todorov, D.; Shishkova, K.; Dragolova, D.; Hinkov, A.; Kapchina-Toteva, V.; Shishkov, S. Antiviral activity of medicinal plant Nepeta nuda. Biotechnol. Biotechnol. Equip. 2015, 29, S39–S43. [Google Scholar] [CrossRef]
  39. Angelova, P.; Tsvetkov, V.; Hinkov, A.; Todorov, D.; Shishkova, K.; Yordanova, Z.; Kapchina-Toteva, V.; Shishkov, S. Antiviral activity of Stachys Thracica Dav. extracts against Human Herpes virus type 1 and 2. BioDiscovery 2017, 20, e15022. [Google Scholar] [CrossRef]
  40. Ghildiyal, R.; Prakash, V.; Chaudhary, V.K.; Gupta, V.; Gabrani, R. Phytochemicals as Antiviral Agents: Recent Updates. In Plant-Derived Bioactives: Production, Properties and Therapeutic Applications; Swamy, M.K., Ed.; Springer: Singapore, 2020; pp. 279–295. [Google Scholar] [CrossRef]
  41. Petran, M.; Dragos, D.; Gilca, M. Historical ethnobotanical review of medicinal plants used to treat children diseases in Romania (1860s–1970s). J. Ethnobiol. Ethnomed. 2020, 16, 15. [Google Scholar] [CrossRef]
  42. Hinkov, A.; Angelova, P.; Marchev, A.; Hodzhev, Y.; Tsvetkov, V.; Dragolova, D.; Todorov, D.; Shishkova, K.; Kapchina-Toteva, V.; Blundell, R.; et al. Nepeta nuda ssp. nuda L. water extract: Inhibition of replication of some strains of human alpha herpes virus (genus simplex virus) in vitro, mode of action and NMR-based metabolomics. J. Herb. Med. 2020, 21, 100334. [Google Scholar] [CrossRef]
  43. Badshah, S.L.; Faisal, S.; Muhammad, A.; Poulson, B.G.; Emwas, A.H.; Jaremko, M. Antiviral activities of flavonoids. Biomed. Pharmacother. 2021, 140, 111596. [Google Scholar] [CrossRef]
  44. Oo, T.; Saiboonjan, B.; Srijampa, S.; Srisrattakarn, A.; Sutthanut, K.; Tavichakorntrakool, R.; Chanawong, A.; Lulitanond, A.; Tippayawat, P. Inhibition of Bacterial Efflux Pumps by Crude Extracts and Essential Oil from Myristica fragrans Houtt. (Nutmeg) Seeds against Methicillin-Resistant Staphylococcus aureus. Molecules 2021, 26, 4662. [Google Scholar] [CrossRef] [PubMed]
  45. Dragoș, D.; Petran, M.; Gradinaru, T.-C.; Gilca, M. Phytochemicals and Inflammation: Is Bitter Better? Plants 2022, 11, 2991. [Google Scholar] [CrossRef] [PubMed]
  46. Gilca, M.; Dragos, D.; Madalina, P.; Gradinaru, T.-C. PlantMolecularTasteDB: A Database of Taste Active Phytochemicals. Front. Pharmacol. 2022, 12, 751712. [Google Scholar] [CrossRef]
  47. Quenon, C.; Hennebelle, T.; Butaud, J.-F.; Ho, R.; Samaillie, J.; Neut, C.; Lehartel, T.; Rivière, C.; Siah, A.; Bonneau, N.; et al. Antimicrobial Properties of Compounds Isolated from Syzygium malaccense (L.) Merr. and L.M. Perry and Medicinal Plants Used in French Polynesia. Life 2022, 12, 733. [Google Scholar] [CrossRef]
  48. Jacquin, J.; Moureu, S.; Deweer, C.; Hakem, A.; Paguet, A.-S.; Bonneau, N.; Bordage, S.; Dermont, C.; Sahpaz, S.; Muchembled, J.; et al. Hop (Humulus lupulus L.) Specialized Metabolites: Extraction, Purification, Characterization in Different Plant Parts and In Vitro Evaluation of Anti-Oomycete Activities against Phytophthora infestans. Agronomy 2022, 12, 2826. [Google Scholar] [CrossRef]
  49. Ellatif, S.A.; Abdel Razik, E.S.; Abu-Serie, M.M.; Mahfouz, A.; Shater, A.F.; Saleh, F.M.; Hassan, M.M.; Alsanie, W.F.; Altalhi, A.; Daigham, G.E.; et al. Immunomodulatory Efficacy-Mediated Anti-HCV and Anti-HBV Potential of Kefir Grains; Unveiling the In Vitro Antibacterial, Antifungal, and Wound Healing Activities. Molecules 2022, 27, 2016. [Google Scholar] [CrossRef]
  50. Vuković, S.; Popović-Djordjević, J.; Kostić, A.; Pantelić, N.; Srećković, N.; Akram, M.; Laila, U.; Katanić Stanković, J. Allium Species in the Balkan Region-Major Metabolites, Antioxidant and Antimicrobial Properties. Horticulturae 2023, 9, 408. [Google Scholar] [CrossRef]
  51. Srivastava, A.; Jit, B.P.; Dash, R.; Srivastava, R.; Srivastava, S. Thuja occidentalis: An Unexplored Phytomedicine with Therapeutic Applications. Comb. Chem. High Throughput Screen. 2023, 26, 3–13. [Google Scholar] [CrossRef]
  52. Zhao, Z.J.; Sun, Y.L.; Ruan, X.F. Bornyl acetate: A promising agent in phytomedicine for inflammation and immune modulation. Phytomedicine 2023, 114, 154781. [Google Scholar] [CrossRef]
  53. Ross, S.M. Ashwagandha: An Effective Phytomedicine for Reducing Stress and Anxiety. Holist. Nurs. Pract. 2023, 37, 298–300. [Google Scholar] [CrossRef]
  54. Pfaar, O.; Beule, A.G.; Jobst, D.; Kraft, K.; Stammer, H.; Röschmann-Doose, K.I.L.; Wittig, T.; Stuck, B.A. Phytomedicine ELOM-080 in Acute Viral Rhinosinusitis: A Randomized, Placebo-Controlled, Blinded Clinical Trial. Laryngoscope 2023, 133, 1576–1583. [Google Scholar] [CrossRef] [PubMed]
  55. Periferakis, A.-T.; Periferakis, A.; Periferakis, K.; Caruntu, A.; Badarau, I.A.; Savulescu-Fiedler, I.; Scheau, C.; Caruntu, C. Antimicrobial Properties of Capsaicin: Available Data and Future Research Perspectives. Nutrients 2023, 15, 4097. [Google Scholar] [CrossRef] [PubMed]
  56. Kumar, N.; Jain, V. Kaempferol: A Key Emphasis on its Counter-Wired Potential. Int. J. Innov. Sci. Res. Technol. 2023, 8, 2534–2540. [Google Scholar]
  57. Hakem, A.; Desmarets, L.; Sahli, R.; Malek, R.B.; Camuzet, C.; François, N.; Lefèvre, G.; Samaillie, J.; Moureu, S.; Sahpaz, S.; et al. Luteolin Isolated from Juncus acutus L., a Potential Remedy for Human Coronavirus 229E. Molecules 2023, 28, 4263. [Google Scholar] [CrossRef]
  58. Moureu, S.; Jacquin, J.; Samaillie, J.; Deweer, C.; Rivière, C.; Muchembled, J. Antifungal Activity of Hop Leaf Extracts and Xanthohumol on Two Strains of Venturia inaequalis with Different Sensitivities to Triazoles. Microorganisms 2023, 11, 1605. [Google Scholar] [CrossRef]
  59. Shkondrov, A.; Hinkov, A.; Cvetkov, V.; Shishkova, K.; Todorov, D.; Shishkov, S.; Stambolov, I.; Yoncheva, K.; Krasteva, I. Astragalus glycyphyllos L.: Antiviral activity and tablet dosage formulation of a standardized dry extract. Biotechnol. Biotechnol. Equip. 2023, 37, 2221752. [Google Scholar] [CrossRef]
  60. Zakaryan, H.; Arabyan, E.; Oo, A.; Zandi, K. Flavonoids: Promising natural compounds against viral infections. Arch. Virol. 2017, 162, 2539–2551. [Google Scholar] [CrossRef]
  61. Chojnacka, K.; Skrzypczak, D.; Izydorczyk, G.; Mikula, K.; Szopa, D.; Witek-Krowiak, A. Antiviral Properties of Polyphenols from Plants. Foods 2021, 10, 2277. [Google Scholar] [CrossRef]
  62. Simmonds, P. Reconstructing the origins of human hepatitis viruses. Philos. Trans. R. Soc. Lond. B Biol. Sci. 2001, 356, 1013–1026. [Google Scholar] [CrossRef]
  63. Woolhouse, M.E.J.; Adair, K. The diversity of human RNA viruses. Future Virol. 2013, 8, 159–171. [Google Scholar] [CrossRef]
  64. Periferakis, A.; Bolocan, A.; Ion, D. A Review of Innovation in Medicine. Technol. Innov. Life Sci. 2022, 1, 42–48. [Google Scholar] [CrossRef]
  65. Li, J.; Huang, H.; Feng, M.; Zhou, W.; Shi, X.; Zhou, P. In vitro and in vivo anti-hepatitis B virus activities of a plant extract from Geranium carolinianum L. Antivir. Res. 2008, 79, 114–120. [Google Scholar] [CrossRef] [PubMed]
  66. Parvez, M.K.; Al-dosari, M.S.; Basudan, O.A.; Herqash, R.N. The anti-hepatitis B virus activity of sea buckthorn is attributed to quercetin, kaempferol and isorhamnetin. Biomed. Rep. 2022, 17, 89. [Google Scholar] [CrossRef] [PubMed]
  67. Yarmolinsky, L.; Huleihel, M.; Zaccai, M.; Ben-Shabat, S. Potent antiviral flavone glycosides from Ficus benjamina leaves. Fitoterapia 2012, 83, 362–367. [Google Scholar] [CrossRef]
  68. Behbahani, M.; Shanehsazzadeh, M.; Shokoohinia, Y.; Soltani, M. Evaluation of anti-herpetic activity of methanol seed extract and fractions of Securigera securidaca in vitro. J. Antivir. Antiretrovir. 2013, 5, 72–76. [Google Scholar]
  69. Park, S.; Kim, N.E.; Park, B.J.; Kwon, H.C.; Song, Y.J. Kaempferol Interferes with Varicella-Zoster Virus Replication in Human Foreskin Fibroblasts. Pharmaceuticals 2022, 15, 1582. [Google Scholar] [CrossRef]
  70. Arabyan, E.; Hakobyan, A.; Hakobyan, T.; Grigoryan, R.; Izmailyan, R.; Avetisyan, A.; Karalyan, Z.; Jackman, J.A.; Ferreira, F.; Elrod, C.C.; et al. Flavonoid Library Screening Reveals Kaempferol as a Potential Antiviral Agent Against African Swine Fever Virus. Front. Microbiol. 2021, 12, 736780. [Google Scholar] [CrossRef]
  71. Chen, X.; Chen, Y.-Q.; Yin, Z.-Q.; Wang, R.; Hu, H.-Y.; Liang, X.-X.; He, C.-L.; Yin, L.-Z.; Ye, G.; Zou, Y.-F.; et al. Kaempferol inhibits Pseudorabies virus replication in vitro through regulation of MAPKs and NF-κB signaling pathways. J. Integr. Agric. 2021, 20, 2227–2239. [Google Scholar] [CrossRef]
  72. Safioleas, M.; Lygidakis, N.J.; Manti, C. Hepatitis B today. Hepatogastroenterology 2007, 54, 545–548. [Google Scholar]
  73. Aspinall, E.J.; Hawkins, G.; Fraser, A.; Hutchinson, S.J.; Goldberg, D. Hepatitis B prevention, diagnosis, treatment and care: A review. Occup. Med. 2011, 61, 531–540. [Google Scholar] [CrossRef]
  74. Trépo, C.; Chan, H.L.; Lok, A. Hepatitis B virus infection. Lancet 2014, 384, 2053–2063. [Google Scholar] [CrossRef] [PubMed]
  75. Wilkins, T.; Sams, R.; Carpenter, M. Hepatitis B: Screening, Prevention, Diagnosis, and Treatment. Am. Fam. Physician 2019, 99, 314–323. [Google Scholar] [PubMed]
  76. Duff, P. Hepatitis in pregnancy. Semin. Perinatol. 1998, 22, 277–283. [Google Scholar] [CrossRef] [PubMed]
  77. Yang, Z.-F.; Bai, L.-P.; Huang, W.-B.; Li, X.-Z.; Zhao, S.-S.; Zhong, N.-S.; Jiang, Z.-H. Comparison of in vitro antiviral activity of tea polyphenols against influenza A and B viruses and structure–activity relationship analysis. Fitoterapia 2014, 93, 47–53. [Google Scholar] [CrossRef] [PubMed]
  78. Whitley, R.J.; Roizman, B. Herpes simplex virus infections. Lancet 2001, 357, 1513–1518. [Google Scholar] [CrossRef]
  79. Omarova, S.; Cannon, A.; Weiss, W.; Bruccoleri, A.; Puccio, J. Genital Herpes Simplex Virus-An Updated Review. Adv. Pediatr. 2022, 69, 149–162. [Google Scholar] [CrossRef]
  80. Landy, H.J.; Grossman, J.H., 3rd. Herpes simplex virus. Obstet. Gynecol. Clin. N. Am. 1989, 16, 495–515. [Google Scholar] [CrossRef]
  81. Fatahzadeh, M.; Schwartz, R.A. Human herpes simplex virus infections: Epidemiology, pathogenesis, symptomatology, diagnosis, and management. J. Am. Acad. Dermatol. 2007, 57, 737–763. [Google Scholar] [CrossRef]
  82. Madavaraju, K.; Koganti, R.; Volety, I.; Yadavalli, T.; Shukla, D. Herpes Simplex Virus Cell Entry Mechanisms: An Update. Front. Cell. Infect. Microbiol. 2020, 10, 617578. [Google Scholar] [CrossRef]
  83. James, S.H.; Kimberlin, D.W. Neonatal Herpes Simplex Virus Infection. Infect. Dis. Clin. N. Am. 2015, 29, 391–400. [Google Scholar] [CrossRef]
  84. Samies, N.L.; James, S.H.; Kimberlin, D.W. Neonatal Herpes Simplex Virus Disease: Updates and Continued Challenges. Clin. Perinatol. 2021, 48, 263–274. [Google Scholar] [CrossRef] [PubMed]
  85. Sibley, D.; Larkin, D.F.P. Update on Herpes simplex keratitis management. Eye 2020, 34, 2219–2226. [Google Scholar] [CrossRef] [PubMed]
  86. Agelidis, A.; Koujah, L.; Suryawanshi, R.; Yadavalli, T.; Mishra, Y.K.; Adelung, R.; Shukla, D. An Intra-Vaginal Zinc Oxide Tetrapod Nanoparticles (ZOTEN) and Genital Herpesvirus Cocktail Can Provide a Novel Platform for Live Virus Vaccine. Front. Immunol. 2019, 10, 500. [Google Scholar] [CrossRef] [PubMed]
  87. Nagel, M.A.; Gilden, D. Neurological complications of varicella zoster virus reactivation. Curr. Opin. Neurol. 2014, 27, 356–360. [Google Scholar] [CrossRef]
  88. Kennedy, P.G.E.; Gershon, A.A. Clinical Features of Varicella-Zoster Virus Infection. Viruses 2018, 10, 609. [Google Scholar] [CrossRef]
  89. Kennedy, P.G.E.; Mogensen, T.H.; Cohrs, R.J. Recent Issues in Varicella-Zoster Virus Latency. Viruses 2021, 13, 2018. [Google Scholar] [CrossRef]
  90. Grahn, A.; Studahl, M. Varicella-zoster virus infections of the central nervous system—Prognosis, diagnostics and treatment. J. Infect. 2015, 71, 281–293. [Google Scholar] [CrossRef]
  91. Garrett, R.; Romanos, M.T.V.; Borges, R.M.; Santos, M.G.; Rocha, L.; Silva, A.J.R.d. Antiherpetic activity of a flavonoid fraction from Ocotea notata leaves. Rev. Bras. Farmacogn. 2012, 22, 306–313. [Google Scholar] [CrossRef]
  92. Duan, X.; Ru, Y.; Yang, W.; Ren, J.; Hao, R.; Qin, X.; Li, D.; Zheng, H. Research progress on the proteins involved in African swine fever virus infection and replication. Front. Immunol. 2022, 13, 947180. [Google Scholar] [CrossRef]
  93. Njau, E.P.; Machuka, E.M.; Cleaveland, S.; Shirima, G.M.; Kusiluka, L.J.; Okoth, E.A.; Pelle, R. African Swine Fever Virus (ASFV): Biology, Genomics and Genotypes Circulating in Sub-Saharan Africa. Viruses 2021, 13, 2285. [Google Scholar] [CrossRef]
  94. Wang, F.; Zhang, H.; Hou, L.; Yang, C.; Wen, Y. Advance of African swine fever virus in recent years. Res. Vet. Sci. 2021, 136, 535–539. [Google Scholar] [CrossRef] [PubMed]
  95. Pomeranz, L.E.; Reynolds, A.E.; Hengartner, C.J. Molecular biology of pseudorabies virus: Impact on neurovirology and veterinary medicine. Microbiol. Mol. Biol. Rev. 2005, 69, 462–500. [Google Scholar] [CrossRef] [PubMed]
  96. Müller, T.; Hahn, E.C.; Tottewitz, F.; Kramer, M.; Klupp, B.G.; Mettenleiter, T.C.; Freuling, C. Pseudorabies virus in wild swine: A global perspective. Arch. Virol. 2011, 156, 1691–1705. [Google Scholar] [CrossRef] [PubMed]
  97. Bo, Z.; Li, X. A Review of Pseudorabies Virus Variants: Genomics, Vaccination, Transmission, and Zoonotic Potential. Viruses 2022, 14, 1003. [Google Scholar] [CrossRef] [PubMed]
  98. Ai, J.W.; Weng, S.S.; Cheng, Q.; Cui, P.; Li, Y.J.; Wu, H.L.; Zhu, Y.M.; Xu, B.; Zhang, W.H. Human Endophthalmitis Caused By Pseudorabies Virus Infection, China, 2017. Emerg. Infect. Dis. 2018, 24, 1087–1090. [Google Scholar] [CrossRef] [PubMed]
  99. Wong, G.; Lu, J.; Zhang, W.; Gao, G.F. Pseudorabies virus: A neglected zoonotic pathogen in humans? Emerg. Microbes Infect. 2019, 8, 150–154. [Google Scholar] [CrossRef]
  100. Pasick, J. Application of DIVA vaccines and their companion diagnostic tests to foreign animal disease eradication. Anim. Health Res. Rev. 2004, 5, 257–262. [Google Scholar] [CrossRef]
  101. Card, J.P. Pseudorabies virus neuroinvasiveness: A window into the functional organization of the brain. Adv. Virus Res. 2001, 56, 39–71. [Google Scholar] [CrossRef]
  102. Parvez, M.K.; Parveen, S. Evolution and Emergence of Pathogenic Viruses: Past, Present, and Future. Intervirology 2017, 60, 1–7. [Google Scholar] [CrossRef]
  103. Schwarz, S.; Sauter, D.; Wang, K.; Zhang, R.; Sun, B.; Karioti, A.; Bilia, A.R.; Efferth, T.; Schwarz, W. Kaempferol derivatives as antiviral drugs against the 3a channel protein of coronavirus. Planta Med. 2014, 80, 177–182. [Google Scholar] [CrossRef]
  104. Zhou, Z.-L.; Yin, W.-Q.; Zou, X.-P.; Huang, D.-Y.; Zhou, C.-L.; Li, L.-M.; Chen, K.-C.; Guo, Z.-Y.; Lin, S.-Q. Flavonoid glycosides and potential antivirus activity of isolated compounds from the leaves of Eucalyptus citriodora. J. Korean Soc. Appl. Biol. Chem. 2014, 57, 813–817. [Google Scholar] [CrossRef]
  105. Jeong, H.J.; Ryu, Y.B.; Park, S.J.; Kim, J.H.; Kwon, H.J.; Kim, J.H.; Park, K.H.; Rho, M.C.; Lee, W.S. Neuraminidase inhibitory activities of flavonols isolated from Rhodiola rosea roots and their in vitro anti-influenza viral activities. Bioorg. Med. Chem. 2009, 17, 6816–6823. [Google Scholar] [CrossRef] [PubMed]
  106. Kai, H.; Obuchi, M.; Yoshida, H.; Watanabe, W.; Tsutsumi, S.; Park, Y.K.; Matsuno, K.; Yasukawa, K.; Kurokawa, M. In vitro and in vivo anti-influenza virus activities of flavonoids and related compounds as components of Brazilian propolis (AF-08). J. Funct. Foods 2014, 8, 214–223. [Google Scholar] [CrossRef]
  107. Derksen, A.; Kühn, J.; Hafezi, W.; Sendker, J.; Ehrhardt, C.; Ludwig, S.; Hensel, A. Antiviral activity of hydroalcoholic extract from Eupatorium perfoliatum L. against the attachment of influenza A virus. J. Ethnopharmacol. 2016, 188, 144–152. [Google Scholar] [CrossRef]
  108. Behbahani, M.; Sayedipour, S.; Pourazar, A.; Shanehsazzadeh, M. In Vitro anti-HIV-1 activities of kaempferol and kaempferol-7-O-glucoside isolated from Securigera securidaca. Res. Pharm. Sci. 2014, 9, 463–469. [Google Scholar]
  109. Care, C.; Sornjai, W.; Jaratsittisin, J.; Hitakarun, A.; Wikan, N.; Triwitayakorn, K.; Smith, D. Discordant Activity of Kaempferol Towards Dengue Virus and Japanese Encephalitis Virus. Molecules 2020, 25, 1246. [Google Scholar] [CrossRef]
  110. Dwivedi, V.D.; Bharadwaj, S.; Afroz, S.; Khan, N.; Ansari, M.A.; Yadava, U.; Tripathi, R.C.; Tripathi, I.P.; Mishra, S.K.; Kang, S.G. Anti-dengue infectivity evaluation of bioflavonoid from Azadirachta indica by dengue virus serine protease inhibition. J. Biomol. Struct. Dyn. 2021, 39, 1417–1430. [Google Scholar] [CrossRef]
  111. Zhang, T.; Wu, Z.; Du, J.; Hu, Y.; Liu, L.; Yang, F.; Jin, Q. Anti-Japanese-Encephalitis-Viral Effects of Kaempferol and Daidzin and Their RNA-Binding Characteristics. PLoS ONE 2012, 7, e30259. [Google Scholar] [CrossRef]
  112. Tsai, F.J.; Lin, C.W.; Lai, C.C.; Lan, Y.C.; Lai, C.H.; Hung, C.H.; Hsueh, K.C.; Lin, T.H.; Chang, H.C.; Wan, L.; et al. Kaempferol inhibits enterovirus 71 replication and internal ribosome entry site (IRES) activity through FUBP and HNRP proteins. Food Chem. 2011, 128, 312–322. [Google Scholar] [CrossRef]
  113. Sauter, D.; Schwarz, S.; Wang, K.; Zhang, R.; Sun, B.; Schwarz, W. Genistein as antiviral drug against HIV ion channel. Planta Med. 2014, 80, 682–687. [Google Scholar] [CrossRef]
  114. Orabi, M.A.; Orabi, E.A. Antiviral and antioxidant activities of flavonoids of Ficus virens: Experimental and theoretical investigations. J. Pharmacogn. Phytochem. 2016, 5, 120–128. [Google Scholar]
  115. Robin, V.; Irurzun, A.; Amoros, M.; Boustie, J.; Carrasco, L. Antipoliovirus flavonoids from Psiadia dentata. Antivir. Chem. Chemother. 2001, 12, 283–291. [Google Scholar] [CrossRef] [PubMed]
  116. Lani, R.; Hassandarvish, P.; Chiam, C.W.; Moghaddam, E.; Chu, J.J.; Rausalu, K.; Merits, A.; Higgs, S.; Vanlandingham, D.; Abu Bakar, S.; et al. Antiviral activity of silymarin against chikungunya virus. Sci. Rep. 2015, 5, 11421. [Google Scholar] [CrossRef] [PubMed]
  117. Seo, D.J.; Jeon, S.B.; Oh, H.; Lee, B.-H.; Lee, S.-Y.; Oh, S.H.; Jung, J.Y.; Choi, C. Comparison of the antiviral activity of flavonoids against murine norovirus and feline calicivirus. Food Control 2016, 60, 25–30. [Google Scholar] [CrossRef]
  118. Santos-López, G.; Cortés-Hernández, P.; Vallejo-Ruiz, V.; Reyes-Leyva, J. SARS-CoV-2: Basic concepts, origin and treatment advances. Gac. Medica Mex. 2021, 157, 84–89. [Google Scholar] [CrossRef]
  119. 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]
  120. Kirtipal, N.; Bharadwaj, S.; Kang, S.G. From SARS to SARS-CoV-2, insights on structure, pathogenicity and immunity aspects of pandemic human coronaviruses. Infect. Genet. Evol. 2020, 85, 104502. [Google Scholar] [CrossRef]
  121. Rabaan, A.A.; Al-Ahmed, S.H.; Haque, S.; Sah, R.; Tiwari, R.; Malik, Y.S.; Dhama, K.; Yatoo, M.I.; Bonilla-Aldana, D.K.; Rodriguez-Morales, A.J. SARS-CoV-2, SARS-CoV, and MERS-COV: A comparative overview. Le Infez. Med. 2020, 28, 174–184. [Google Scholar]
  122. Yang, H.; Rao, Z. Structural biology of SARS-CoV-2 and implications for therapeutic development. Nat. Rev. Microbiol. 2021, 19, 685–700. [Google Scholar] [CrossRef]
  123. Bourgonje, A.R.; Abdulle, A.E.; Timens, W.; Hillebrands, J.L.; Navis, G.J.; Gordijn, S.J.; Bolling, M.C.; Dijkstra, G.; Voors, A.A.; Osterhaus, A.D.; et al. Angiotensin-converting enzyme 2 (ACE2), SARS-CoV-2 and the pathophysiology of coronavirus disease 2019 (COVID-19). J. Pathol. 2020, 251, 228–248. [Google Scholar] [CrossRef]
  124. Jackson, C.B.; Farzan, M.; Chen, B.; Choe, H. Mechanisms of SARS-CoV-2 entry into cells. Nat. Rev. Mol. Cell Biol. 2022, 23, 3–20. [Google Scholar] [CrossRef] [PubMed]
  125. Stein, S.R.; Ramelli, S.C.; Grazioli, A.; Chung, J.-Y.; Singh, M.; Yinda, C.K.; Winkler, C.W.; Sun, J.; Dickey, J.M.; Ylaya, K.; et al. SARS-CoV-2 infection and persistence in the human body and brain at autopsy. Nature 2022, 612, 758–763. [Google Scholar] [CrossRef] [PubMed]
  126. Daia, C.; Scheau, C.; Neagu, G.; Andone, I.; Spanu, A.; Popescu, C.; Stoica, S.I.; Verenca, M.C.; Onose, G. Nerve conduction study and electromyography findings in patients recovering from COVID-19—Case report. Int. J. Infect. Dis. 2020, 103, 420–422. [Google Scholar] [CrossRef] [PubMed]
  127. Fiolet, T.; Kherabi, Y.; MacDonald, C.J.; Ghosn, J.; Peiffer-Smadja, N. Comparing COVID-19 vaccines for their characteristics, efficacy and effectiveness against SARS-CoV-2 and variants of concern: A narrative review. Clin. Microbiol. Infect. 2022, 28, 202–221. [Google Scholar] [CrossRef]
  128. Cines, D.B.; Bussel, J.B. SARS-CoV-2 Vaccine-Induced Immune Thrombotic Thrombocytopenia. N. Engl. J. Med. 2021, 384, 2254–2256. [Google Scholar] [CrossRef]
  129. Zamfir, M.-A.; Moraru, L.; Dobrea, C.; Scheau, A.-E.; Iacob, S.; Moldovan, C.; Scheau, C.; Caruntu, C.; Caruntu, A. Hematologic Malignancies Diagnosed in the Context of the mRNA COVID-19 Vaccination Campaign: A Report of Two Cases. Medicina 2022, 58, 874. [Google Scholar] [CrossRef]
  130. Goldman, S.; Bron, D.; Tousseyn, T.; Vierasu, I.; Dewispelaere, L.; Heimann, P.; Cogan, E.; Goldman, M. Rapid Progression of Angioimmunoblastic T Cell Lymphoma Following BNT162b2 mRNA Vaccine Booster Shot: A Case Report. Front. Med. 2021, 8, 798095. [Google Scholar] [CrossRef]
  131. Brumfiel, C.M.; Patel, M.H.; DiCaudo, D.J.; Rosenthal, A.C.; Pittelkow, M.R.; Mangold, A.R. Recurrence of primary cutaneous CD30-positive lymphoproliferative disorder following COVID-19 vaccination. Leuk. Lymphoma 2021, 62, 2554–2555. [Google Scholar] [CrossRef]
  132. Anand, A.V.; Balamuralikrishnan, B.; Kaviya, M.; Bharathi, K.; Parithathvi, A.; Arun, M.; Senthilkumar, N.; Velayuthaprabhu, S.; Saradhadevi, M.; Al-Dhabi, N.A.; et al. Medicinal Plants, Phytochemicals, and Herbs to Combat Viral Pathogens Including SARS-CoV-2. Molecules 2021, 26, 1775. [Google Scholar] [CrossRef]
  133. Khazdair, M.; Anaeigoudari, A.; Agbor, G. Anti-viral and anti-inflammatory effects of kaempferol and quercetin and COVID-2019: A scoping review. Asian Pac. J. Trop. Biomed. 2021, 11, 327–334. [Google Scholar] [CrossRef]
  134. Johnson, A.P. Methicillin-resistant Staphylococcus aureus: The European landscape. J. Antimicrob. Chemother. 2011, 66 (Suppl. S4), iv43–iv48. [Google Scholar] [CrossRef] [PubMed]
  135. Owis, A.I.; El-Hawary, M.S.; El Amir, D.; Aly, O.M.; Abdelmohsen, U.R.; Kamel, M.S. Molecular docking reveals the potential of Salvadora persica flavonoids to inhibit COVID-19 virus main protease. RSC Adv. 2020, 10, 19570–19575. [Google Scholar] [CrossRef] [PubMed]
  136. Kanmani, R. Polyphenolics Screening in Mollungo Nudicaulis using UPLC-ESI-MS and Its Active Compound Kaempferol against SARs CoV2 Receptor. M.Sc. Thesis, JKK Nattaraja College of Pharmacy, Kumarapalayam, India, 2021. [Google Scholar]
  137. Qiu, X.; Xu, S.; Lu, Y.; Luo, Z.; Yan, Y.; Wang, C.; Ji, J. Development of mRNA vaccines against respiratory syncytial virus (RSV). Cytokine Growth Factor Rev. 2022, 68, 37–53. [Google Scholar] [CrossRef] [PubMed]
  138. Borchers, A.T.; Chang, C.; Gershwin, M.E.; Gershwin, L.J. Respiratory syncytial virus—A comprehensive review. Clin. Rev. Allergy Immunol. 2013, 45, 331–379. [Google Scholar] [CrossRef] [PubMed]
  139. Gatt, D.; Martin, I.; AlFouzan, R.; Moraes, T.J. Prevention and Treatment Strategies for Respiratory Syncytial Virus (RSV). Pathogens 2023, 12, 154. [Google Scholar] [CrossRef] [PubMed]
  140. Priante, E.; Cavicchiolo, M.E.; Baraldi, E. RSV infection and respiratory sequelae. Minerva Pediatr. 2018, 70, 623–633. [Google Scholar] [CrossRef]
  141. Jenkins, V.A.; Hoet, B.; Hochrein, H.; De Moerlooze, L. The Quest for a Respiratory Syncytial Virus Vaccine for Older Adults: Thinking beyond the F Protein. Vaccines 2023, 11, 382. [Google Scholar] [CrossRef]
  142. Labella, A.M.; Merel, S.E. Influenza. Med. Clin. N. Am. 2013, 97, 621–645. [Google Scholar] [CrossRef]
  143. Gaitonde, D.Y.; Moore, F.C.; Morgan, M.K. Influenza: Diagnosis and Treatment. Am. Fam. Physician 2019, 100, 751–758. [Google Scholar]
  144. Wu, N.C.; Wilson, I.A. Influenza Hemagglutinin Structures and Antibody Recognition. Cold Spring Harb. Perspect. Med. 2020, 10, a038778. [Google Scholar] [CrossRef]
  145. Webster, R.G.; Govorkova, E.A. Continuing challenges in influenza. Ann. N. Y. Acad. Sci. 2014, 1323, 115–139. [Google Scholar] [CrossRef] [PubMed]
  146. Świerczyńska, M.; Mirowska-Guzel, D.M.; Pindelska, E. Antiviral Drugs in Influenza. Int. J. Environ. Res. Public Health 2022, 19, 3018. [Google Scholar] [CrossRef] [PubMed]
  147. Liu, A.-L.; Wang, H.-D.; Lee, S.M.; Wang, Y.-T.; Du, G.-H. Structure–activity relationship of flavonoids as influenza virus neuraminidase inhibitors and their in vitro anti-viral activities. Bioorg. Med. Chem. 2008, 16, 7141–7147. [Google Scholar] [CrossRef] [PubMed]
  148. Park, S.; Kim, J.I.; Lee, I.; Lee, S.; Hwang, M.-W.; Bae, J.-Y.; Heo, J.; Kim, D.; Han, S.-Z.; Park, M.-S. Aronia melanocarpa and its components demonstrate antiviral activity against influenza viruses. Biochem. Biophys. Res. Commun. 2013, 440, 14–19. [Google Scholar] [CrossRef]
  149. World Health Organization. Global Situation and Trends of HIV. Available online: https://www.who.int/data/gho/data/themes/hiv-aids (accessed on 24 September 2023).
  150. Fanales-Belasio, E.; Raimondo, M.; Suligoi, B.; Buttò, S. HIV virology and pathogenetic mechanisms of infection: A brief overview. Ann. Ist. Super. Sanita 2010, 46, 5–14. [Google Scholar] [CrossRef]
  151. Lucas, S.; Nelson, A.M. HIV and the spectrum of human disease. J. Pathol. 2015, 235, 229–241. [Google Scholar] [CrossRef]
  152. Hu, W.S.; Hughes, S.H. HIV-1 reverse transcription. Cold Spring Harb. Perspect. Med. 2012, 2, a006882. [Google Scholar] [CrossRef]
  153. Sabin, C.A.; Lundgren, J.D. The natural history of HIV infection. Curr. Opin. HIV AIDS 2013, 8, 311–317. [Google Scholar] [CrossRef]
  154. Phanuphak, N.; Gulick, R.M. HIV treatment and prevention 2019: Current standards of care. Curr. Opin. HIV AIDS 2020, 15, 4–12. [Google Scholar] [CrossRef]
  155. Tsukada, K. HIV infection/AIDS. Nihon Rinsho 2016, 74, 1992–1997. [Google Scholar]
  156. Achappa, B.; Madi, D.; Bhaskaran, U.; Ramapuram, J.T.; Rao, S.; Mahalingam, S. Adherence to Antiretroviral Therapy Among People Living with HIV. N. Am. J. Med. Sci. 2013, 5, 220–223. [Google Scholar] [CrossRef] [PubMed]
  157. Dorcélus, L.; Bernard, J.; Georgery, C.; Vanessa, C. Factors associated with antiretroviral therapy adherence among people living with HIV in Haiti: A cross-sectional study. AIDS Res. Ther. 2021, 18, 81. [Google Scholar] [CrossRef] [PubMed]
  158. Kim, J.; Lee, E.; Park, B.-J.; Bang, J.H.; Lee, J.Y. Adherence to antiretroviral therapy and factors affecting low medication adherence among incident HIV-infected individuals during 2009–2016: A nationwide study. Sci. Rep. 2018, 8, 3133. [Google Scholar] [CrossRef] [PubMed]
  159. Chen, Y.; Chen, K.; Kalichman, S.C. Barriers to HIV Medication Adherence as a Function of Regimen Simplification. Ann. Behav. Med. 2017, 51, 67–78. [Google Scholar] [CrossRef] [PubMed]
  160. Jilg, N.; Li, J.Z. On the Road to a HIV Cure: Moving Beyond Berlin and London. Infect. Dis. Clin. N. Am. 2019, 33, 857–868. [Google Scholar] [CrossRef]
  161. Pang, X.; Zhang, R.; Cheng, G. Progress towards understanding the pathogenesis of dengue hemorrhagic fever. Virol. Sin. 2017, 32, 16–22. [Google Scholar] [CrossRef]
  162. Martina, B.E.; Koraka, P.; Osterhaus, A.D. Dengue virus pathogenesis: An integrated view. Clin. Microbiol. Rev. 2009, 22, 564–581. [Google Scholar] [CrossRef]
  163. Kariyawasam, R.; Lachman, M.; Mansuri, S.; Chakrabarti, S.; Boggild, A.K. A dengue vaccine whirlwind update. Ther. Adv. Infect. Dis. 2023, 10, 20499361231167274. [Google Scholar] [CrossRef]
  164. López-Medina, E.; Biswal, S.; Saez-Llorens, X.; Borja-Tabora, C.; Bravo, L.; Sirivichayakul, C.; Vargas, L.M.; Alera, M.T.; Velásquez, H.; Reynales, H.; et al. Efficacy of a Dengue Vaccine Candidate (TAK-003) in Healthy Children and Adolescents 2 Years after Vaccination. J. Infect. Dis. 2022, 225, 1521–1532. [Google Scholar] [CrossRef]
  165. Troost, B.; Smit, J.M. Recent advances in antiviral drug development towards dengue virus. Curr. Opin. Virol. 2020, 43, 9–21. [Google Scholar] [CrossRef]
  166. Taslem Mourosi, J.; Awe, A.; Jain, S.; Batra, H. Nucleic Acid Vaccine Platform for DENGUE and ZIKA Flaviviruses. Vaccines 2022, 10, 834. [Google Scholar] [CrossRef] [PubMed]
  167. Le Flohic, G.; Porphyre, V.; Barbazan, P.; Gonzalez, J.P. Review of climate, landscape, and viral genetics as drivers of the Japanese encephalitis virus ecology. PLoS Negl. Trop. Dis. 2013, 7, e2208. [Google Scholar] [CrossRef] [PubMed]
  168. Ravanini, P.; Huhtamo, E.; Ilaria, V.; Crobu, M.G.; Nicosia, A.M.; Servino, L.; Rivasi, F.; Allegrini, S.; Miglio, U.; Magri, A.; et al. Japanese encephalitis virus RNA detected in Culex pipiens mosquitoes in Italy. Eurosurveillance 2012, 17, 20221. [Google Scholar] [CrossRef]
  169. Yasui, K. Neuropathogenesis of Japanese encephalitis virus. J. Neurovirol. 2002, 8 (Suppl. S2), 112–114. [Google Scholar] [CrossRef] [PubMed]
  170. Sharma, K.B.; Vrati, S.; Kalia, M. Pathobiology of Japanese encephalitis virus infection. Mol. Asp. Med. 2021, 81, 100994. [Google Scholar] [CrossRef] [PubMed]
  171. Yun, S.I.; Lee, Y.M. Japanese encephalitis: The virus and vaccines. Hum. Vaccines Immunother. 2014, 10, 263–279. [Google Scholar] [CrossRef] [PubMed]
  172. Johari, J.; Kianmehr, A.; Mustafa, M.R.; Abubakar, S.; Zandi, K. Antiviral Activity of Baicalein and Quercetin against the Japanese encephalitis Virus. Int. J. Mol. Sci. 2012, 13, 16785–16795. [Google Scholar] [CrossRef]
  173. Solomon, T.; Lewthwaite, P.; Perera, D.; Cardosa, M.J.; McMinn, P.; Ooi, M.H. Virology, epidemiology, pathogenesis, and control of enterovirus 71. Lancet Infect. Dis. 2010, 10, 778–790. [Google Scholar] [CrossRef]
  174. Chong, P.; Liu, C.C.; Chow, Y.H.; Chou, A.H.; Klein, M. Review of enterovirus 71 vaccines. Clin. Infect. Dis. 2015, 60, 797–803. [Google Scholar] [CrossRef]
  175. Nayak, G.; Bhuyan, S.K.; Bhuyan, R.; Sahu, A.; Kar, D.; Kuanar, A. Global emergence of Enterovirus 71: A systematic review. Beni Suef Univ. J. Basic Appl. Sci. 2022, 11, 78. [Google Scholar] [CrossRef]
  176. de Crom, S.C.; Rossen, J.W.; van Furth, A.M.; Obihara, C.C. Enterovirus and parechovirus infection in children: A brief overview. Eur. J. Pediatr. 2016, 175, 1023–1029. [Google Scholar] [CrossRef]
  177. Cao, G.; Jing, W.; Liu, J.; Liu, M. The global trends and regional differences in incidence and mortality of hepatitis A from 1990 to 2019 and implications for its prevention. Hepatol. Int. 2021, 15, 1068–1082. [Google Scholar] [CrossRef]
  178. Abutaleb, A.; Kottilil, S. Hepatitis A: Epidemiology, Natural History, Unusual Clinical Manifestations, and Prevention. Gastroenterol. Clin. N. Am. 2020, 49, 191–199. [Google Scholar] [CrossRef]
  179. Pereira, F.E.; Gonçalves, C.S. Hepatitis A. Rev. Soc. Bras. Med. Trop. 2003, 36, 387–400. [Google Scholar] [CrossRef] [PubMed]
  180. Jeong, S.H.; Lee, H.S. Hepatitis A: Clinical manifestations and management. Intervirology 2010, 53, 15–19. [Google Scholar] [CrossRef] [PubMed]
  181. Ohemu, T.; Agunu, A.; Chollom, S.; Okwori, V.; Dafam, D.; Olotu, P. Preliminary Phytochemical Screening and Antiviral Potential of Methanol Stem Bark Extract of Enantia chlorantha Oliver (Annonaceae) and Boswellia dalzielii Hutch (Burseraceae) against Newcastle Disease In Ovo. Eur. J. Med. Plants 2018, 25, 1–8. [Google Scholar] [CrossRef]
  182. Kidd, D.; Williams, A.J.; Howard, R.S. Poliomyelitis. Postgrad. Med. J. 1996, 72, 641–647. [Google Scholar] [CrossRef] [PubMed]
  183. Lo, J.K.; Robinson, L.R. Post-polio syndrome and the late effects of poliomyelitis: Part 2. treatment, management, and prognosis. Muscle Nerve 2018, 58, 760–769. [Google Scholar] [CrossRef] [PubMed]
  184. Shapiro, L.T.; Sherman, A.L. Medical Comorbidities and Complications Associated with Poliomyelitis and Its Sequelae. Phys. Med. Rehabil. Clin. N. Am. 2021, 32, 591–600. [Google Scholar] [CrossRef]
  185. Goodrick, S. Preventing polio. Lancet Neurol. 2014, 13, 653. [Google Scholar] [CrossRef]
  186. Bandyopadhyay, A.S.; Garon, J.; Seib, K.; Orenstein, W.A. Polio vaccination: Past, present and future. Future Microbiol. 2015, 10, 791–808. [Google Scholar] [CrossRef]
  187. Suhrbier, A.; Jaffar-Bandjee, M.C.; Gasque, P. Arthritogenic alphaviruses—An overview. Nat. Rev. Rheumatol. 2012, 8, 420–429. [Google Scholar] [CrossRef]
  188. Caglioti, C.; Lalle, E.; Castilletti, C.; Carletti, F.; Capobianchi, M.R.; Bordi, L. Chikungunya virus infection: An overview. New Microbiol. 2013, 36, 211–227. [Google Scholar]
  189. Burt, F.J.; Chen, W.; Miner, J.J.; Lenschow, D.J.; Merits, A.; Schnettler, E.; Kohl, A.; Rudd, P.A.; Taylor, A.; Herrero, L.J.; et al. Chikungunya virus: An update on the biology and pathogenesis of this emerging pathogen. Lancet Infect. Dis. 2017, 17, e107–e117. [Google Scholar] [CrossRef]
  190. Martins, D.O.S.; Santos, I.A.; de Oliveira, D.M.; Grosche, V.R.; Jardim, A.C.G. Antivirals against Chikungunya Virus: Is the Solution in Nature? Viruses 2020, 12, 272. [Google Scholar] [CrossRef]
  191. Ghildiyal, R.; Gabrani, R. Antiviral therapeutics for chikungunya virus. Expert. Opin. Ther. Pat. 2020, 30, 467–480. [Google Scholar] [CrossRef]
  192. Radford, A.D.; Coyne, K.P.; Dawson, S.; Porter, C.J.; Gaskell, R.M. Feline calicivirus. Vet. Res. 2007, 38, 319–335. [Google Scholar] [CrossRef]
  193. Spiri, A.M. An Update on Feline Calicivirus. Schweiz Arch. Tierheilkd. 2022, 164, 225–241. [Google Scholar] [CrossRef]
  194. Bordicchia, M.; Fumian, T.M.; Van Brussel, K.; Russo, A.G.; Carrai, M.; Le, S.J.; Pesavento, P.A.; Holmes, E.C.; Martella, V.; White, P.; et al. Feline Calicivirus Virulent Systemic Disease: Clinical Epidemiology, Analysis of Viral Isolates and In Vitro Efficacy of Novel Antivirals in Australian Outbreaks. Viruses 2021, 13, 2040. [Google Scholar] [CrossRef]
  195. Radford, A.D.; Addie, D.; Belák, S.; Boucraut-Baralon, C.; Egberink, H.; Frymus, T.; Gruffydd-Jones, T.; Hartmann, K.; Hosie, M.J.; Lloret, A.; et al. Feline calicivirus infection. ABCD guidelines on prevention and management. J. Feline Med. Surg. 2009, 11, 556–564. [Google Scholar] [CrossRef]
  196. Hofmann-Lehmann, R.; Hosie, M.J.; Hartmann, K.; Egberink, H.; Truyen, U.; Tasker, S.; Belák, S.; Boucraut-Baralon, C.; Frymus, T.; Lloret, A.; et al. Calicivirus Infection in Cats. Viruses 2022, 14, 937. [Google Scholar] [CrossRef]
  197. Henderson, K.S. Murine norovirus, a recently discovered and highly prevalent viral agent of mice. Lab Anim. 2008, 37, 314–320. [Google Scholar] [CrossRef]
  198. Karst, S.M.; Tibbetts, S.A. Recent advances in understanding norovirus pathogenesis. J. Med. Virol. 2016, 88, 1837–1843. [Google Scholar] [CrossRef]
  199. Graziano, V.R.; Wei, J.; Wilen, C.B. Norovirus Attachment and Entry. Viruses 2019, 11, 495. [Google Scholar] [CrossRef]
  200. MacGregor, J.T.; Jurd, L. Mutagenicity of plant flavonoids: Structural requirements for mutagenic activity in Salmonella typhimurium. Mutat. Res. 1978, 54, 297–309. [Google Scholar] [CrossRef]
  201. Francis, A.R.; Shetty, T.K.; Bhattacharya, R.K. Modifying role of dietary factors on the mutagenicity of aflatoxin B1: In Vitro effect of plant flavonoids. Mutat. Res. 1989, 222, 393–401. [Google Scholar] [CrossRef]
  202. Niering, P.; Michels, G.; Wätjen, W.; Ohler, S.; Steffan, B.; Chovolou, Y.; Kampkötter, A.; Proksch, P.; Kahl, R. Protective and detrimental effects of kaempferol in rat H4IIE cells: Implication of oxidative stress and apoptosis. Toxicol. Appl. Pharmacol. 2005, 209, 114–122. [Google Scholar] [CrossRef]
  203. Takanashi, H.; Aiso, S.; Hirono, I.; Matsushima, T.; Sugimura, T. Carcinogenicity test of quercetin and kaempferol in rats by oral administration. J. Food Saf. 1983, 5, 55–60. [Google Scholar] [CrossRef]
  204. Halstead, S.B. Dengvaxia sensitizes seronegatives to vaccine enhanced disease regardless of age. Vaccine 2017, 35, 6355–6358. [Google Scholar] [CrossRef]
  205. Song, Y.; Mugavero, J.; Stauft, C.B.; Wimmer, E. Dengue and Zika Virus 5′ Untranslated Regions Harbor Internal Ribosomal Entry Site Functions. mBio 2019, 10, 1110–1128. [Google Scholar] [CrossRef]
  206. Calzada, F.; Cedillo-Rivera, R.; Mata, R. Antiprotozoal activity of the constituents of Conyza filaginoides. J. Nat. Prod. 2001, 64, 671–673. [Google Scholar] [CrossRef]
  207. Arot Manguro, L.O.; Ugi, I.; Hermann, R.; Lemmen, P. Flavonol and drimane-type sesquiterpene glycosides of Warburgia stuhlmannii leaves. Phytochemistry 2003, 63, 497–502. [Google Scholar] [CrossRef]
  208. Krauze-Baranowska, M. Flavonoids from the genus Taxus. Z. Naturforschung C J. Biosci. 2004, 59, 43–47. [Google Scholar] [CrossRef]
  209. Nguemeving, J.R.; Azebaze, A.G.B.; Kuete, V.; Eric Carly, N.N.; Beng, V.P.; Meyer, M.; Blond, A.; Bodo, B.; Nkengfack, A.E. Laurentixanthones A and B, antimicrobial xanthones from Vismia laurentii. Phytochemistry 2006, 67, 1341–1346. [Google Scholar] [CrossRef]
  210. Jiang, R.-W.; Zhou, J.-R.; Hon, P.-M.; Li, S.-L.; Zhou, Y.; Li, L.-L.; Ye, W.-C.; Xu, H.-X.; Shaw, P.-C.; But, P.P.-H. Lignans from Dysosma versipellis with Inhibitory Effects on Prostate Cancer Cell Lines. J. Nat. Prod. 2007, 70, 283–286. [Google Scholar] [CrossRef]
  211. Marín, C.; Boutaleb-Charki, S.; Díaz, J.G.; Huertas, O.; Rosales, M.J.; Pérez-Cordon, G.; Guitierrez-Sánchez, R.; Sánchez-Moreno, M. Antileishmaniasis activity of flavonoids from Consolida oliveriana. J. Nat. Prod. 2009, 72, 1069–1074. [Google Scholar] [CrossRef]
  212. Calzada, F.; Correa-Basurto, J.; Barbosa, E.; Mendez-Luna, D.; Yepez-Mulia, L. Antiprotozoal Constituents from Annona cherimola Miller, a Plant Used in Mexican Traditional Medicine for the Treatment of Diarrhea and Dysentery. Pharmacogn. Mag. 2017, 13, 148–152. [Google Scholar]
  213. Maas, M.; Hensel, A.; Costa, F.B.; Brun, R.; Kaiser, M.; Schmidt, T.J. An unusual dimeric guaianolide with antiprotozoal activity and further sesquiterpene lactones from Eupatoriumperfoliatum. Phytochemistry 2011, 72, 635–644. [Google Scholar] [CrossRef]
  214. Prasasty, V.D.; Cindana, S.; Ivan, F.X.; Zahroh, H.; Sinaga, E. Structure-based discovery of novel inhibitors of Mycobacterium tuberculosis CYP121 from Indonesian natural products. Comput. Biol. Chem. 2020, 85, 107205. [Google Scholar] [CrossRef]
  215. Tagne, M.; Nougang, M.; Metsopkeng, C.; Bricheux, G.; Donnadieu, F.; Nana, P.-A.; Ripoche, I.; Donfagsiteli Tchinda, N.; Agbor, G.; Chalard, P.; et al. Effects of aqueous and hydro-ethanolic Moringa oleifera Lam leaf extracts on the cultivability of 2 Bacillus strains isolated from rainwater. J. Food Stab. 2023, 6, 1–19. [Google Scholar]
  216. Khan, M.; Khan, T.; Wahab, S.; Aasim, M.; Sherazi, T.A.; Zahoor, M.; Yun, S.-I. Solvent based fractional biosynthesis, phytochemical analysis, and biological activity of silver nanoparticles obtained from the extract of Salvia moorcroftiana. PLoS ONE 2023, 18, e0287080. [Google Scholar] [CrossRef] [PubMed]
  217. Youl, O.; Moné-Bassavé, B.R.H.; Yougbaré, S.; Yaro, B.; Traoré, T.K.; Boly, R.; Yaméogo, J.B.G.; Koala, M.; Ouedraogo, N.; Kabré, E.; et al. Phytochemical Screening, Polyphenol and Flavonoid Contents, and Antioxidant and Antimicrobial Activities of Opilia amentacea Roxb. (Opiliaceae) Extracts. Appl. Biosci. 2023, 2, 493–512. [Google Scholar] [CrossRef]
  218. Santos-Buelga, C.; Feliciano, A.S. Flavonoids: From Structure to Health Issues. Molecules 2017, 22, 477. [Google Scholar] [CrossRef]
  219. Wu, S.; Chen, W.; Lu, S.; Zhang, H.; Yin, L. Metabolic Engineering of Shikimic Acid Biosynthesis Pathway for the Production of Shikimic Acid and Its Branched Products in Microorganisms: Advances and Prospects. Molecules 2022, 27, 4779. [Google Scholar] [CrossRef]
  220. Duan, L.; Ding, W.; Liu, X.; Cheng, X.; Cai, J.; Hua, E.; Jiang, H. Biosynthesis and engineering of kaempferol in Saccharomyces cerevisiae. Microb. Cell Factories 2017, 16, 165. [Google Scholar] [CrossRef]
  221. Viskupicova, J.; Ondrejovič, M.; Sturdik, E. Bioavailability and metabolism of flavonoids. J. Food Nutr. Res. 2008, 47, 151–162. [Google Scholar]
  222. Nielsen, S.E.; Kall, M.; Justesen, U.; Schou, A.; Dragsted, L.O. Human absorption and excretion of flavonoids after broccoli consumption. Cancer Lett. 1997, 114, 173–174. [Google Scholar] [CrossRef]
  223. de Vries, J.H.; Hollman, P.C.; Meyboom, S.; Buysman, M.N.; Zock, P.L.; van Staveren, W.A.; Katan, M.B. Plasma concentrations and urinary excretion of the antioxidant flavonols quercetin and kaempferol as biomarkers for dietary intake. Am. J. Clin. Nutr. 1998, 68, 60–65. [Google Scholar] [CrossRef]
  224. Németh, K.; Plumb, G.W.; Berrin, J.G.; Juge, N.; Jacob, R.; Naim, H.Y.; Williamson, G.; Swallow, D.M.; Kroon, P.A. Deglycosylation by small intestinal epithelial cell beta-glucosidases is a critical step in the absorption and metabolism of dietary flavonoid glycosides in humans. Eur. J. Nutr. 2003, 42, 29–42. [Google Scholar] [CrossRef]
  225. Terao, J. Dietary flavonoids as antioxidants. Forum. Nutr. 2009, 61, 87–94. [Google Scholar] [CrossRef]
  226. Chen, A.Y.; Chen, Y.C. A review of the dietary flavonoid, kaempferol on human health and cancer chemoprevention. Food Chem. 2013, 138, 2099–2107. [Google Scholar] [CrossRef]
  227. Cassidy, A.; Minihane, A.M. The role of metabolism (and the microbiome) in defining the clinical efficacy of dietary flavonoids. Am. J. Clin. Nutr. 2017, 105, 10–22. [Google Scholar] [CrossRef]
  228. Williamson, G.; Kay, C.D.; Crozier, A. The Bioavailability, Transport, and Bioactivity of Dietary Flavonoids: A Review from a Historical Perspective. Compr. Rev. Food Sci. Food Saf. 2018, 17, 1054–1112. [Google Scholar] [CrossRef]
  229. Dabeek, W.M.; Marra, M.V. Dietary Quercetin and Kaempferol: Bioavailability and Potential Cardiovascular-Related Bioactivity in Humans. Nutrients 2019, 11, 2288. [Google Scholar] [CrossRef]
  230. Bangar, S.P.; Chaudhary, V.; Sharma, N.; Bansal, V.; Ozogul, F.; Lorenzo, J.M. Kaempferol: A flavonoid with wider biological activities and its applications. Crit. Rev. Food Sci. Nutr. 2022, 63, 9580–9604. [Google Scholar] [CrossRef]
  231. Ćavar, S.; Maksimović, M.; Vidic, D.; Parić, A. Chemical composition and antioxidant and antimicrobial activity of essential oil of Artemisia annua L. from Bosnia. Ind. Crops Prod. 2012, 37, 479–485. [Google Scholar] [CrossRef]
  232. Mueller, M.S.; Karhagomba, I.B.; Hirt, H.M.; Wemakor, E. The potential of Artemisia annua L. as a locally produced remedy for malaria in the tropics: Agricultural, chemical and clinical aspects. J. Ethnopharmacol. 2000, 73, 487–493. [Google Scholar] [CrossRef]
  233. Kumar, V.S.; Navaratnam, V. Neem (Azadirachta indica): Prehistory to contemporary medicinal uses to humankind. Asian Pac. J. Trop. Biomed. 2013, 3, 505–514. [Google Scholar] [CrossRef]
  234. Alzohairy, M.A. Therapeutics Role of Azadirachta indica (Neem) and Their Active Constituents in Diseases Prevention and Treatment. Evid. Based Complement. Altern. Med. 2016, 2016, 7382506. [Google Scholar] [CrossRef]
  235. Islas, J.F.; Acosta, E.; G-Buentello, Z.; Delgado-Gallegos, J.L.; Moreno-Treviño, M.G.; Escalante, B.; Moreno-Cuevas, J.E. An overview of Neem (Azadirachta indica) and its potential impact on health. J. Funct. Foods 2020, 74, 104171. [Google Scholar] [CrossRef]
  236. Gbekley, H.E.; Katawa, G.; Karou, S.D.; Anani, S.; Tchadjobo, T.; Ameyapoh, Y.; Batawila, K.; Simpore, J. Ethnobotanical Study of Plants Used to Treat Asthma in the Maritime Region in Togo. Afr. J. Tradit. Complement. Altern. Med. 2017, 14, 196–212. [Google Scholar] [CrossRef] [PubMed]
  237. Tolba, H.; Moghrani, H.; Benelmouffok, A.; Kellou, D.; Maachi, R. Essential oil of Algerian Eucalyptus citriodora: Chemical composition, antifungal activity. J. Mycol. Méd. 2015, 25, e128–e133. [Google Scholar] [CrossRef] [PubMed]
  238. Seyoum, A.; Pålsson, K.; Kung’a, S.; Kabiru, E.W.; Lwande, W.; Killeen, G.F.; Hassanali, A.; Knots, B.G.J. Traditional use of mosquito-repellent plants in western Kenya and their evaluation in semi-field experimental huts against Anopheles gambiae: Ethnobotanical studies and application by thermal expulsion and direct burning. Trans. R. Soc. Trop. Med. Hyg. 2002, 96, 225–231. [Google Scholar] [CrossRef] [PubMed]
  239. Muanza, D.; Kim, B.; Euler, K.; Williams, L. Antibacterial and antifungal activities of nine medicinal plants from Zaire. Int. J. Pharmacogn. 1994, 32, 337–345. [Google Scholar] [CrossRef]
  240. Stammel, H.J. Die Apotheke Manitous—Das Medizinische Wissen der Indianer und Ihre Heilpflanzen; Rowolth Verlag GmbH: Reinbek bei Hamburg, Germany, 1986. [Google Scholar]
  241. Moerman, D.A. Native American Ethnobotany; Timber Press: Portland, OR, USA, 1998. [Google Scholar]
  242. Hensel, A.; Maas, M.; Sendker, J.; Lechtenberg, M.; Petereit, F.; Deters, A.; Schmidt, T.; Stark, T. Eupatorium perfoliatum L.: Phytochemistry, traditional use and current applications. J. Ethnopharmacol. 2011, 138, 641–651. [Google Scholar] [CrossRef]
  243. Imran, M.; Rasool, N.; Rizwan, K.; Zubair, M.; Riaz, M.; Zia-Ul-Haq, M.; Rana, U.A.; Nafady, A.; Jaafar, H.Z. Chemical composition and Biological studies of Ficus benjamina. Chem. Cent. J. 2014, 8, 12. [Google Scholar] [CrossRef]
  244. Parajuli, S.P. Ethnobotanical study at Khandbari Municipality of Sankhuwasabha District, Nepal. Banko Janakari 2000, 10, 29–34. [Google Scholar] [CrossRef]
  245. Kanaujia, V.K.; Rirchhaiya, H.; Kailasiya, S.; Verma, M.; Yadav, R.; Shivhare, D. Evaluation of hepatoprotective activity on the leaves of Ficus benjamina Linn. J. Nat. Prod. Plant 2011, 1, 59–69. [Google Scholar]
  246. Sirisha, N.; Sreenivasulu, M.; Sangeeta, K.; Chetty, C.M. Antioxidant properties of Ficus species-a review. Int. J. PharmTech Res. 2010, 2, 2174–2182. [Google Scholar]
  247. Patel, M.H.; Desai, B.; Jha, S.; Patel, D.; Mehta, A.; Garde, Y. Phytochemical, pharmacognosy and ethnobotanical importance of the Ficus virens Aiton. Pharma Innov. J. 2023, 12, 4017–4021. [Google Scholar]
  248. Li, Y.; Ye, Y.; Wang, S.-J.; Xia, W.; Rahman, K.; Yue, W.; Zhang, H. Analgesic, anti-inflammatory and antipyretic activities of the aqueous extract of Geranium carolinianum L. Afr. J. Tradit. Complement. Altern. Med. 2016, 13, 105–113. [Google Scholar] [CrossRef]
  249. Bal, L.M.; Meda, V.; Naik, S.N.; Satya, S. Sea buckthorn berries: A potential source of valuable nutrients for nutraceuticals and cosmoceuticals. Food Res. Int. 2011, 44, 1718–1727. [Google Scholar] [CrossRef]
  250. Guliyev, V.B.; Gul, M.; Yildirim, A. Hippophae rhamnoides L.: Chromatographic methods to determine chemical composition, use in traditional medicine and pharmacological effects. J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 2004, 812, 291–307. [Google Scholar] [CrossRef] [PubMed]
  251. Vogl, S.; Picker, P.; Mihaly-Bison, J.; Fakhrudin, N.; Atanasov, A.G.; Heiss, E.H.; Wawrosch, C.; Reznicek, G.; Dirsch, V.M.; Saukel, J.; et al. Ethnopharmacological in vitro studies on Austria’s folk medicine--an unexplored lore in vitro anti-inflammatory activities of 71 Austrian traditional herbal drugs. J. Ethnopharmacol. 2013, 149, 750–771. [Google Scholar] [CrossRef]
  252. Pundir, S.; Garg, P.; Dviwedi, A.; Ali, A.; Kapoor, V.K.; Kapoor, D.; Kulshrestha, S.; Lal, U.R.; Negi, P. Ethnomedicinal uses, phytochemistry and dermatological effects of Hippophae rhamnoides L.: A review. J. Ethnopharmacol. 2021, 266, 113434. [Google Scholar] [CrossRef]
  253. Costa, I.F.d.J.B.; Simão, T.L.B.V.; Calixto, S.D.; Pereira, R.V.; Konno, T.U.P.; Pinto, S.C.; Tinoco, L.W.; Lasunskaia, E.; Leal, I.C.R.; Muzitano, M.F. Anti-mycobacterial and immunomodulatory activity of n-hexane fraction and spathulenol from Ocotea notata leaves. Rodriguésia 2021, 72. [Google Scholar] [CrossRef]
  254. Fortin, H.; Tomasi, S.; Jaccard, P.; Robin, V.; Boustie, J. A Prenyloxycoumarin from Psiadia dentata. Chem. Pharm. Bull. 2001, 49, 619–621. [Google Scholar] [CrossRef]
  255. Mahadeo, K.; Grondin, I.; Kodja, H.; Soulange Govinden, J.; Jhaumeer Laulloo, S.; Frederich, M.; Gauvin-Bialecki, A. The genus Psiadia: Review of traditional uses, phytochemistry and pharmacology. J. Ethnopharmacol. 2018, 210, 48–68. [Google Scholar] [CrossRef]
  256. Ivanova Stojcheva, E.; Quintela, J.C. The Effectiveness of Rhodiola rosea L. Preparations in Alleviating Various Aspects of Life-Stress Symptoms and Stress-Induced Conditions-Encouraging Clinical Evidence. Molecules 2022, 27, 3902. [Google Scholar] [CrossRef]
  257. Chen, Y.; Tang, M.; Yuan, S.; Fu, S.; Li, Y.; Li, Y.; Wang, Q.; Cao, Y.; Liu, L.; Zhang, Q. Rhodiola rosea: A Therapeutic Candidate on Cardiovascular Diseases. Oxid. Med. Cell. Longev. 2022, 2022, 1348795. [Google Scholar] [CrossRef]
  258. Panossian, A.; Wikman, G.; Sarris, J. Rosenroot (Rhodiola rosea): Traditional use, chemical composition, pharmacology and clinical efficacy. Phytomedicine 2010, 17, 481–493. [Google Scholar] [CrossRef] [PubMed]
  259. Khatak, M.; Khatak, S.; Siddqui, A.A.; Vasudeva, N.; Aggarwal, A.; Aggarwal, P. Salvadora persica. Pharmacogn. Rev. 2010, 4, 209–214. [Google Scholar] [CrossRef] [PubMed]
  260. Aljarbou, F.; Almobarak, A.; Binrayes, A.; Alamri, H.M. Salvadora persica’s Biological Properties and Applications in Different Dental Specialties: A Narrative Review. Evid. Based Complement. Alternat. Med. 2022, 2022, 8667687. [Google Scholar] [CrossRef] [PubMed]
  261. Abdelbagi, M.E.M.; Al-Mazaideh, G.M.; Ahmed, A.E.; Al-Rimawi, F.; Ayyal Salman, H.; Almutairi, A.; Abuilaiwi, F.A.; Wedian, F. Drug Formulation of Securigera securidaca Seed Extracts. Processes 2023, 11, 1955. [Google Scholar] [CrossRef]
  262. Nasehi, Z.; Kheiripour, N.; Taheri, M.A.; Ardjmand, A.; Jozi, F.; Aghadavod, E.; Doustimotlagh, A.H.; Shahaboddin, M.E. The Protective Effects of Securigera securidaca Seed Extract on Liver Injury Induced by Bile Duct Ligation in Rats. BioMed Res. Int. 2022, 2022, 6989963. [Google Scholar] [CrossRef]
  263. He, X.; Bai, Y.; Zhao, Z.; Wang, X.; Fang, J.; Huang, L.; Zeng, M.; Zhang, Q.; Zhang, Y.; Zheng, X. Local and traditional uses, phytochemistry, and pharmacology of Sophora japonica L.: A review. J. Ethnopharmacol. 2016, 187, 160–182. [Google Scholar] [CrossRef]
  264. Chen, H.-N.; Hsieh, C.-L. Effects of Sophora japonica flowers (Huaihua) on cerebral infarction. Chin. Med. 2010, 5, 34. [Google Scholar] [CrossRef]
  265. Matei, A.-M.; Caruntu, C.; Tampa, M.; Georgescu, S.R.; Matei, C.; Constantin, M.M.; Constantin, T.V.; Calina, D.; Ciubotaru, D.A.; Badarau, I.A.; et al. Applications of Nanosized-Lipid-Based Drug Delivery Systems in Wound Care. Appl. Sci. 2021, 11, 4915. [Google Scholar] [CrossRef]
  266. Wilczewska, A.Z.; Niemirowicz, K.; Markiewicz, K.H.; Car, H. Nanoparticles as drug delivery systems. Pharmacol. Rep. 2012, 64, 1020–1037. [Google Scholar] [CrossRef]
  267. Liu, R.; Luo, C.; Pang, Z.; Zhang, J.; Ruan, S.; Wu, M.; Wang, L.; Sun, T.; Li, N.; Han, L. Advances of nanoparticles as drug delivery systems for disease diagnosis and treatment. Chin. Chem. Lett. 2023, 34, 107518. [Google Scholar] [CrossRef]
  268. Song, J.M.; Lee, K.H.; Seong, B.L. Antiviral effect of catechins in green tea on influenza virus. Antivir. Res. 2005, 68, 66–74. [Google Scholar] [CrossRef]
  269. Isaacs, C.E.; Wen, G.Y.; Xu, W.; Jia, J.H.; Rohan, L.; Corbo, C.; Di Maggio, V.; Jenkins, E.C., Jr.; Hillier, S. Epigallocatechin gallate inactivates clinical isolates of herpes simplex virus. Antimicrob. Agents Chemother. 2008, 52, 962–970. [Google Scholar] [CrossRef]
  270. Panara, A.; Gikas, E.; Tzavellas, I.; Thomaidis, N.S. Comprehensive HRMS Chemical Characterization of Pomegranate-Based Antioxidant Drinks via a Newly Developed Suspect and Target Screening Workflow. Molecules 2023, 28, 4986. [Google Scholar] [CrossRef]
  271. Zandi, K.; Teoh, B.-T.; Sam, S.-S.; Wong, P.-F.; Mustafa, M.R.; AbuBakar, S. Antiviral activity of four types of bioflavonoid against dengue virus type-2. Virol. J. 2011, 8, 560. [Google Scholar] [CrossRef]
  272. Bachmetov, L.; Gal-Tanamy, M.; Shapira, A.; Vorobeychik, M.; Giterman-Galam, T.; Sathiyamoorthy, P.; Golan-Goldhirsh, A.; Benhar, I.; Tur-Kaspa, R.; Zemel, R. Suppression of hepatitis C virus by the flavonoid quercetin is mediated by inhibition of NS3 protease activity. J. Viral Hepat. 2012, 19, e81–e88. [Google Scholar] [CrossRef]
  273. Ganesan, S.; Faris, A.N.; Comstock, A.T.; Wang, Q.; Nanua, S.; Hershenson, M.B.; Sajjan, U.S. Quercetin inhibits rhinovirus replication in vitro and in vivo. Antivir. Res. 2012, 94, 258–271. [Google Scholar] [CrossRef]
  274. Ieven, M.; Vlietinck, A.J.; Vanden Berghe, D.A.; Totte, J.; Dommisse, R.; Esmans, E.; Alderweireldt, F. Plant antiviral agents. III. Isolation of alkaloids from Clivia miniata Regel (Amaryllidaceae). J. Nat. Prod. 1982, 45, 564–573. [Google Scholar] [CrossRef]
  275. Ren, G.; Ding, G.; Zhang, H.; Wang, H.; Jin, Z.; Yang, G.; Han, Y.; Zhang, X.; Li, G.; Li, W. Antiviral activity of sophoridine against enterovirus 71 in vitro. J. Ethnopharmacol. 2019, 236, 124–128. [Google Scholar] [CrossRef]
  276. Zhang, X.; Hung, T.M.; Phuong, P.T.; Ngoc, T.M.; Min, B.S.; Song, K.S.; Seong, Y.H.; Bae, K. Anti-inflammatory activity of flavonoids from Populus davidiana. Arch. Pharm. Res. 2006, 29, 1102–1108. [Google Scholar] [CrossRef]
  277. Periferakis, A.; Caruntu, A.; Periferakis, A.T.; Scheau, A.E.; Badarau, I.A.; Caruntu, C.; Scheau, C. Availability, Toxicology and Medical Significance of Antimony. Int. J. Environ. Res. Public Health 2022, 19, 4669. [Google Scholar] [CrossRef]
  278. Thomas, E.; Stewart, L.E.; Darley, B.A.; Pham, A.M.; Esteban, I.; Panda, S.S. Plant-Based Natural Products and Extracts: Potential Source to Develop New Antiviral Drug Candidates. Molecules 2021, 26, 6197. [Google Scholar] [CrossRef] [PubMed]
  279. Frediansyah, A.; Sofyantoro, F.; Alhumaid, S.; Al Mutair, A.; Albayat, H.; Altaweil, H.I.; Al-Afghani, H.M.; AlRamadhan, A.A.; AlGhazal, M.R.; Turkistani, S.A.; et al. Microbial Natural Products with Antiviral Activities, Including Anti-SARS-CoV-2: A Review. Molecules 2022, 27, 4305. [Google Scholar] [CrossRef] [PubMed]
  280. Woolhouse, M.E.; Brierley, L.; McCaffery, C.; Lycett, S. Assessing the Epidemic Potential of RNA and DNA Viruses. Emerg. Infect. Dis. 2016, 22, 2037–2044. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Chemical structures of kaempferol and common derivatives mentioned in the text.
Figure 1. Chemical structures of kaempferol and common derivatives mentioned in the text.
Ijms 24 16299 g001
Figure 2. Summary of kaempferol antiviral effects on DNA and RNA viral infections.
Figure 2. Summary of kaempferol antiviral effects on DNA and RNA viral infections.
Ijms 24 16299 g002
Figure 3. Illustration of the action mechanisms of kaempferol and its derivatives on viral entry, replication and effects. A. Direct effect on viral molecules; B. Inhibition of viral attachment; C. Inhibition of polymerase activity; D. Inhibition of reverse transcription; E. Inhibition of viral proteins transcription; F. Inhibition of viral proteins activity; G. Down-regulation of the PI3K-Akt pathway; H. Up-regulation of AMPK signalling. DNA = deoxyribonucleic acid, RNA = ribonucleic acid, DNAP = deoxyribonucleic acid polymerase, RT = reverse transcriptase, AKT = protein kinase B, AMPK = adenosine monophosphate-activated protein kinase.
Figure 3. Illustration of the action mechanisms of kaempferol and its derivatives on viral entry, replication and effects. A. Direct effect on viral molecules; B. Inhibition of viral attachment; C. Inhibition of polymerase activity; D. Inhibition of reverse transcription; E. Inhibition of viral proteins transcription; F. Inhibition of viral proteins activity; G. Down-regulation of the PI3K-Akt pathway; H. Up-regulation of AMPK signalling. DNA = deoxyribonucleic acid, RNA = ribonucleic acid, DNAP = deoxyribonucleic acid polymerase, RT = reverse transcriptase, AKT = protein kinase B, AMPK = adenosine monophosphate-activated protein kinase.
Ijms 24 16299 g003
Table 1. In vitro studies on the potential antiviral actions and effects of kaempferol and its derivatives against DNA viruses.
Table 1. In vitro studies on the potential antiviral actions and effects of kaempferol and its derivatives against DNA viruses.
FamilyGenusExtract fromCompound TestedToxicity LimitConcentration (Type of Effect)MechanismYear of ResearchReference
HepadnaviridaeHepatitis B virus (HBV)Extract of Geranium carolianum L.kaempferol160.79 μg/mL47.54 mg/kg (ED50)Decrease in HBsAg and HBeAg and of viral DNA synthesis2008[65]
Leaf ethanol extract of Hippophae rhamnoideskaempferol32.58 μg/mL (CC50 for SB-Chl)
150 μg/mL (SB-Eac, SB-But and SB-Aqu)
10 μg/mL (stable concentration used in the experiment)Inhibition of HBsAg and HBeAg expression2022[66]
Orthoherpesviridae (Alphaherpesvirinae)Human herpesvirus 1virus 1 (HHV-1)Extract from Ficus benjaminakaempferol
3-O-rutinoside
300 ± 2.79 μmol/L3.00 ± 0.97 μmol/L (EC50)Unknown2012[67]
kaempferol 3-O-robinobioside600 ± 10.45 μmol/L0.90 ± 0.23 μmol/L
(EC50)
Extracts of dried and powdered Securigera securidaca seedskaempferol60.0 ± 5.0 μg/mL (CC50)0.20 ± 0.12 μg/mL
(EC50)
Inhibition of viral attachment and entry into cells; inhibition of viral polymerase2014[68]
kaempferol-7-O-glycoside250 ± 1.7 μg/mL (CC50)0.20 ± 0.01 μg/mL
(EC50)
Human herpesvirus 2 (HHV-2)Extract from Ficus benjaminakaempferol
3-O-rutinoside
Unknownn/an/a2012[67]
kaempferol 3-O-robinobiosideUnknown
Varicella-Zoster Virus (VZV)Pure compoundkaempferolNo cytotoxicity detected6.36 ± 0.73 μg/m (IC50)Blockade of viral DNA synthesis and viral replication2022[69]
AsfarviridaeAfrican Swine Fever Virus (ASFV)Pure compoundkaempferol93.10 μg/mL (CC50)2.20 μg/mL (IC50)Unknown2021[70]
OrthoherpesviridaePseudorabies Virus (PRV)Pure compoundkaempferol254.97 ± 1.86 μmol/L (CC50)25.57 ± 0.74 μmol/L (IC50)Regulation of the MAPK and NF-κB pathways2021[71]
Table 2. In vitro studies on the potential antiviral actions and effects of kaempferol and its derivatives against RNA viruses.
Table 2. In vitro studies on the potential antiviral actions and effects of kaempferol and its derivatives against RNA viruses.
FamilyGenusExtract FromCompound TestedToxicity LimitConcentration (Type)MechanismYear of ResearchReference
CoronaviridaeSARS coronavirus (SARS-COV-2)Pure compoundsNumerous kaempferol glycosidesNot calculated20 μΜ (minimum effective concentration)Inhibition of the 3a membrane channel2014[103]
PneumoviridaeRespiratory syncytial virus (RSV)Extract from Eucalyptus citriodoraKaempferol-3-O-β-D-glucopyranosyl (12)-α-L-rhamnoside137.60 μg/mL
(CC50)
57.30 μg/mL (IC50)Reduction in virus multiplication2014[104]
Kaempferol-3-O-α-L-rhamnoside258.1 μg/mL
(CC50)
56.90 μg/mL (IC50)
Extract from Sophora japonica flowersKaempferol143.79 μg/mL
(TC50)
4.84 μg/mL (IC50)Reduction in viral cytopathic effects2014[77]
OrthomyxoviridaeInfluenza virusExtract from Rhodiola rosea rootsKaempferol>300 μΜ18.50–30.20 μM (EC50 depending on viral strain)Inhibition of neuraminidase2009[105]
Extract from Brazilian propolisKaempferol>100 μg/mL21.70–38.20 μM (depending on viral strain)Limitation of infection symptoms2014[106]
Extract from Eupatorium perfoliatum L.Kaempferol-3-O-β-D-galactoside (trifolin)Not calculated for individual compoundsVarious effective concentrationsPrevention of viral attachment and entry into the cells2016[107]
Kaempferol-3-O-β-D-glucoside (astragalin)
RetroviridaeHuman immunodeficiency virus (HIV)Extract from Securigera securidacaKaempferol320 μg/mL50 μg/mL (IC50)Inhibition of reverse transcriptase2014[108]
Kaempferol-7-O-glycoside2500 μg/mL32 μg/mL (IC50)
FlaviviridaeDengue fever virus (DFV)Pure compoundKaempferol228.50 Μμ (HEK293Τ/17 cells); 139.70 Μμ (BHK-21 cells)None (not effective at tested concentration)-2020[109]
Extract from Azadirachta indicaKaempferol-3-O-rutinosideNot significant10 μΜ (minimum tested concentration)Inhibition of viral protease2021[110]
Japanese encephalitis virusPure compoundKaempferol230 μΜ12.6–21.5 μM (depending on experimental conditions)Inhibition of viral protein expression2012[111]
Pure compoundKaempferol228.50 Μμ (HEK293Τ/17 cells); 139.70 Μμ (BHK-21 cells)66.33 μM (EC50)Probably inhibition of cap-dependent translation2020[109]
PicornaviridaeEnterovirus 71Pure compoundKaempferol50 μΜ<>35 μM (standard concentration used)Inhibition of translation and replication2011[112]
Hepatitis A virus (HAV)Pure compoundKaempferolNot determinedNone (not effective at tested concentrations)-2014[113]
Extract from Ficus virensKaempferol-3-O-α-D-arabinopyranoside329.9 ± 5.3 μg/mLNone (not effective at tested concentrations)-2016[114]
Kaempferol-3-O-β-D-galactopyranoside313.3 ± 1.19 μg/mLNone (not effective at tested concentrations)
PoliovirusExtract from Psiadia dentata3-Methylkaempferol107 μΜVarious tested concentration under different settingsInhibition of the replication2001[115]
3,4′-Dimethylkaempferol197 μΜ
TogaviridaeChikungunya virus (CHIKV)Pure compoundKaempferol>1000 μg/mL (CC50 Vero cells); 537.30 μg/mL (CC50 BHK-21 cells)400 μΜ (concentration necessary for a degree of inhibition)Inhibition of post-entry replication2015[116]
CaliciviridaeFeline calicivirus (FCV)Pure compoundKaempferol>300 μΜ50 μΜ (minimum effective concentration)Unknown2016[117]
Murine norovirus (MNV)None (not effective at tested concentrations)-
Table 3. Effectives of kaempferol and its derivatives relative to common antiviral drugs.
Table 3. Effectives of kaempferol and its derivatives relative to common antiviral drugs.
VirusCompound TestedCompound Effectiveness (Concentration)Reference DrugDrug Effectiveness (Concentration)Reference
Hepatitis B virusKaempferol10 μg/mL (62.3% viral inhibition of HBsAg synthesis)Lamivudine2 μΜ (87.4% viral inhibiton of HBsAg synthesis)[66]
Human herpesvirus 1Kaempferol0.20 ± 0.01 μg/mL (EC50)Acyclovir0.10 ± 0.01 μg/mL (EC50)[68]
Kaempferol-7-O glycoside0.10 ± 0.01 μg/mL (EC50)
Varicella-zosterKaempferol6.36 ± 0.73 µg/mL (IC50)Acyclovir0.54 ± 0.12 µM (IC50) [69]
Pseudorabies virusKaempferol25.57 μg/mL (IC50)Acyclovir54.97 μg/mL (IC50)[71]
Feline calicivirusKaempferol50–100 μΜ (tested concentrations)RibavirinHigher effectiveness at the same concentrations[117]
Influenza virusKaempferol21.70–38.20 μg/mL (EC50)Ribavirin19.20 ± 7.5 μg/mL (EC50)[106]
Respiratory syncytial virusKaempferol-3-O-β-D-glucopyranosyl (12)-α-L-rhamnoside57.30 μg/mL (IC50)Ribavirin2.60 μg/mL (IC50)[104]
Kaempferol-3-O-α-L-rhamnoside56.90 μg/mL (IC50)Ribavirin2.60 μg/mL (IC50)
HIVKaempferol50 μg/mL (IC50)Zidovudine1 μg/mL (IC50)[108]
Kaempferol-7-O-glycoside32 μg/mL (IC50)
Table 4. Traditional medical uses of plants mentioned in this review.
Table 4. Traditional medical uses of plants mentioned in this review.
PlantMedical TraditionTraditional/Ethnobotanical UsesReferences
Atermisia annuaTraditional Chinese medicineAnti-hyperlipidaemic, anti-plasmodial, anti-convulsant, anti-inflammatory, antimicrobial, anti-cholesterolaemic and antiviral properties[231,232]
Azadirachta indicaTraditional Chinese medicine, Ayurvedic medicine, Unani medicineAntimicrobial and anti-inflammatory uses[233,234,235]
Eucalyptus citriodoraAfrican folk medicine (various geographical areas)Anti-asthmatic, antifungal, general antimicrobial[236,237,238,239]
Eupatorium perfoliatumNative American folk medicine, European medical traditionsAntipyretic, antirheumatic agent and treatment of colds, anti-malarial agent and use as an antiviral agent[240,241,242]
Ficus benjaminaNumerous local remedies in Asia, Africa, the Pacific islands and the AmericasAntimicrobial, antinociceptive, antipyretic and hypotensive uses; anti-dysentery remedy[243,244,245,246]
Ficus virensTraditional Indian medicine and AyurvedaPrevention and treatment of diseases and various other reported medicinal effects[247]
Geranium carolinianum L.Traditional Chinese medicineAntimicrobial, anti-inflammatory, and antipyretic uses[248]
Hippophae rhamnoidesLocal medical traditions in Russia and Asia, Austrian folk medicineTreatment of hypertension, oedema, inflammation; tissue regeneration; treatment of burns, wounds, and ulcers[249,250,251,252]
Ocotea notataFolk medicine of South AmericaTreatment of chest pain, rheumatism wounds and viral infections[253]
Psiadia dentataLocal African medical traditions, folk medicine of the Mascarene islandsTreatment of abdominal pains, colds, fevers, bronchitis, asthma, rheumatoid arthritis, skin infections and liver disorders[254,255]
Rhodiola roseaTraditional Chinese medicine, Viking folk medicine, various local medical traditions of Asian and European countriesNervous system stimulation, stress and fatigue alleviation, treatment for gastrointestinal complaints, anaemia, infections, and impotence[256,257,258]
Salvadora persicaTraditional Indian medicine, African folk medicine, medical tradition of Saudi ArabiaAntidote to poison, prevention of scurvy, treatment of rheumatism, anti-inflammatory use, treatment of skin conditions, purgative, treatment of gastrointestinal disorders, antimicrobial properties[259,260]
Securigera securidacaTraditional Iranian medicine, traditional Egyptian medicine, traditional Indian medicineAnti-epileptic, anticonvulsant, and blood lipid-lowering actions[261,262]
Sophora japonicaTraditional Chinese medicine, traditional Japanese medicine, traditional Korean medicineTreatment of haemorrhoids, haematochezia, haematuria, hematemesis, haemorrhinia, uterine or intestinal haemorrhage, arteriosclerosis, headache, hypertension, dysentery, dizziness, and pyoderma[263,264]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Periferakis, A.; Periferakis, A.-T.; Troumpata, L.; Periferakis, K.; Scheau, A.-E.; Savulescu-Fiedler, I.; Caruntu, A.; Badarau, I.A.; Caruntu, C.; Scheau, C. Kaempferol: A Review of Current Evidence of Its Antiviral Potential. Int. J. Mol. Sci. 2023, 24, 16299. https://doi.org/10.3390/ijms242216299

AMA Style

Periferakis A, Periferakis A-T, Troumpata L, Periferakis K, Scheau A-E, Savulescu-Fiedler I, Caruntu A, Badarau IA, Caruntu C, Scheau C. Kaempferol: A Review of Current Evidence of Its Antiviral Potential. International Journal of Molecular Sciences. 2023; 24(22):16299. https://doi.org/10.3390/ijms242216299

Chicago/Turabian Style

Periferakis, Argyrios, Aristodemos-Theodoros Periferakis, Lamprini Troumpata, Konstantinos Periferakis, Andreea-Elena Scheau, Ilinca Savulescu-Fiedler, Ana Caruntu, Ioana Anca Badarau, Constantin Caruntu, and Cristian Scheau. 2023. "Kaempferol: A Review of Current Evidence of Its Antiviral Potential" International Journal of Molecular Sciences 24, no. 22: 16299. https://doi.org/10.3390/ijms242216299

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

Article Metrics

Back to TopTop