Open access peer-reviewed chapter

Neurological Complications in COVID-19: Implications on International Health Security and Possible Interventions of Phytochemicals

Written By

Johnson Olaleye Oladele, Oluwaseun Titilope Oladele, Oyedotun M. Oyeleke and Adenike T. Oladiji

Submitted: 02 December 2020 Reviewed: 15 January 2021 Published: 10 March 2021

DOI: 10.5772/intechopen.96039

From the Edited Volume

Contemporary Developments and Perspectives in International Health Security - Volume 2

Edited by Stanislaw P. Stawicki, Thomas J. Papadimos, Sagar C. Galwankar, Andrew C. Miller and Michael S. Firstenberg

Chapter metrics overview

476 Chapter Downloads

View Full Metrics

Abstract

Global health security or international health security (IHS) includes any natural or man-made phenomenon that challenged human health and well-being including emerging infectious diseases such as the current global pandemic: COVID-19. Since the sudden outburst of COVID-19 pandemic in 2019, many COVID-19 patients have exhibited neurological symptoms and signs. Till now, there is no known effective established drug against the highly contagious COVID-19 infection despite the frightening associated mortality rate. This chapter aims to present the mechanism of action of coronavirus-2 (SARS-CoV-2), the clinical neurological manifestations displayed by COVID-19 patients, impact on the global health system and present phytochemicals with neuroprotective ability that can offer beneficial effects against COVID-19 mediated neuropathology. Reports from COVID-19 clinical studies, case reports, and other related literature were evaluated. Neurological complications of COVID-19 include anosmia, acute cerebrovascular disease, acute disseminated post-infectious encephalomyelitis, encephalitis, etc. Also, SARS-CoV-2 соuld be a neurotropic vіruѕ due to its iѕоlаtіоn from сеrеbrоѕріnаl fluіd. Multірlе nеurоlоgісаl dаmаgе displayed by COVID-19 patients might be due to hyperinflammation associated with SARS-CoV-2 infections. Kolaviron, resveratrol, vernodalin, vernodalol, and apigenin are natural phytochemicals with proven anti-inflammatory and therapeutic properties that could extenuate the adverse effects of COVID-19. The phytochemicals have been documented to suppress JNK and MAPK pathways which are essential in the pathogenesis of COVID-19. They also showed significant inhibitory activities against SARS-CoV-2 main protease. Taken together, these phytochemicals may offer neuroprotective benefits against COVID-19 mediated neuropathology and suppress the burden of the pandemic on IHS.

Keywords

  • international health security
  • Neuropathology
  • COVID-19
  • Resveratrol
  • Kolaviron
  • Apigenin
  • Phytochemicals
  • SARS-CoV-2

1. Introduction

International health security (IHS) is a sum total of any natural, simulated or synthetic phenomenons that pose major threats on human health and well-being including emerging infectious diseases (EIDs) such as the current global pandemic: COVID-19. COVID-19, novel coronavirus pneumonia is ranked amidst the nine most deadly global pandemic ever occurred in the world. It was first recorded in 2019 at Wuhan, a Chinese city and since its first outbreak, the pandemic has dispersed wide to every region of the globe having critical negative impact on many countries both developed and developing nations. This severe acute respiratory disease is highly contagious and transmissible via a pathogenic virus called SARS-CoV-2 to humans and animals. Reports by the WHO team on COVID-19 pandemic as of 25th November 2020 showed that COVID-19 has really inflicted great havoc on human health and constitutes a major danger to global public health. It was reported that over 57.8 million cases of SARS-CoV-2 infections have been recorded with over 1.3 million deaths globally [1, 2]. In Nigeria, the most populous country in Africa, over 66,000 cases had been confirmed and more than 1,160 mortalities recorded [1, 2]. This statistic reveals the impact of this pandemic on the global health system capacity. The emergence of pathogens represents a significant threat to public health, including both high-income regions and low/middle-income regions [3, 4, 5].

COVID-19 has an average incubation period of 3 days [6]. The most prevalent medical manifestations of COVID-19 (such as cough, fever, shortness of breath, fatigue, and other complications) are nearly the same to those of other viral pneumonia; multiple organ failures and death were documented in critical and severe cases [7]. These indications are prominently expressed in aged persons perhaps owing to lingering and chronic underlying diseases such as diabetes, hypertension, neurodegenerative disorders, or heart diseases [8]. The spread of the virus (SARS-CoV-2) amid individuals happens when there is an infiltration of infected aerosols from cough, sneeze, or respiratory droplets into the lungs through inhalation in the nose or mouth.

Clinical case reports have documented a spectrum of neuropathological features displayed by COVID-19 patients. These neurological manifestations include anosmia, acute cerebrovascular disease, acute disseminated post-infectiousencephalomyelitis, Encephalitis, Guillain–Barré syndrome, acutedisseminated post-infectiousencephalitis, and viral meningitis [9]. Presence or confirmation of SARS-CоV-2 in сеrеbrоѕріnаl fluіd ѕuggеѕt thаt it соuld invade and infect the central nervous system (CNS) as a neurotropic vіruѕ inducing multірlе nеurоlоgісаl impairments [9].

Advertisement

2. Methods

This chapter presents the pathogenic mechanism of SARS-CoV-2 and neurological complications of COVID-19. Furthermore, we present the possible intervention of potential anti-COVID-19 phytochemicals in the treatment of neuropathology associated with COVID-19. The literature search for this article was done on Medline, Google Scholar, and PubMed Central using keywords Clinical features, Coronavirus, SARSCOV-2, COVID-19, and complications.

2.1 Neurological damage associated with Covid-19

Several mechanisms have been projected for the neuropathology linked to SARS-CoV-2 in reference to clinical manifestations displayed by COVID-19 patients. Mao et al. [10] documented hyposmia and anosmia in COVID-19 patients. This indicates that SARS-CoV-2 may be spread directly from the cribriform plate near the olfactory bulb to brain regions [11]. SARS-CoV-2 can diffuse to the CNS via enteric nerve and sympathetic afferent mediated by gastrointestinaltract infection [12]. Furthermore, anterograde and retrograde transmission can mediate neuro-invasion of SARS-CoV-2 through the sensory and motor nerve endings [13], coupled with involvement of motor proteins (dynein and kinesins), in particular through the vagus nerve from the lungs [14].

Brains are more vulnerable to oxidative and neuroinflammation insults due to the low level of cytoprotective endogenous enzymes. The cytokine storm syndrome (hyper-inflammation) accompanying SARS-CoV-2 infections may be one of the causes of the neurological impairments observed in COVID-19 patients. Viral infections have been documented as one of the chief agents that induces secondary haemophagocytic lymphohistiocytosis (sHLH) [15]. sHLH similarly referred to as Macrophage Activation Syndrome (MAS) is a severe health disorder which includes diverse group of hyper-inflammatory condition arisen after an infringement in the interaction between genetic predisposition and initiators such as infections. One of the features of sHLH is an abrupt and severe hyper-cytokinaemia due to inapt persistence of histiocytes and cytotoxic T-lymphocytes which eventually leads to multi-organ failure, haemophagocytosis, and mortality [16]. Other features of sHLH includes persistent fever, cytopenias, and hyper-ferritinaemia; pulmonary involvement occurs in approximately 50% of patients [17].

In the brain, activation of glial cells caused brain damage and severe inflammation with the secretion of pro-inflammatory cytokines including TNF-alpha, interleukin-2, and interleukin-5 [18]. Neuro-invasion of SARS-CoV-2 can activate macrophage via CD4+ cells to produce interleukin-6 which is a principal constituent of cytokine storm syndrome via granulocyte-macrophage colony-stimulating factor. Thus, causing damage to the neuronal cells.

2.2 Modes of action of SARS-CoV-2

Prospective EIDs which are major factor in IHS can emanate from vector-borne, vaccine-preventable, epidemic-prone, food-borne, zoonotic, and/or antibiotic-resistant pathogens or from a lack of access to safe water and sanitation. The experiences had and measures taken from prior outbreaks can enhance interventions and improve future responses to emerging infectious diseases [19, 20]. The genetic investigation on SARS-CoV-2 showed that the comprehensive genome sequence recognition rates of bat SARS coronavirus (SARSr-CoV-RaTG13) and SARS-CoV were 96.2% and 79.5%, respectively [21]. Comparing with other coronaviruses, SARS-CoV-2 proteins for viral replication, spikes formation, and nucleocapsid are initiated in specific genes in ORF1 [22]. The virus (SARS-CoV-2) gain entrance into the host cell and invade it via series of cellular alterations and modifications like other types of beta-coronaviruses. Subsequently, SARS-CoV-2 binds to the Angiotensin-Converting Enzyme 2 (ACE2) receptor in the human and/or host’s alveoli of the lungs and respiratory epithelium via theRBM of the S protein [23, 24]. Similar type of receptors has been documented in the viral genome of SARS-CoV and SARS-CoV-2, particularly, the receptor binding motif (RBM) and the receptor binding domain (RBD) [25, 26, 27]. Attachment of SARS-CoV to the receptor leads to the recruitment of cellular proteases to split the S protein into S1 and S2 domains. Transmembrane protease serine 2 (TMPRSS2), human airway trypsin-like protease (HAT) and cathepsins are the cellular proteases that cleave the spike protein and enhance additional penetration modifications [28, 29]. The splitting of S protein facilitates the activation of S2 via a conformational modification thereby allowing the insertion of the internal fusion protein (FP) into the membrane which facilitate the entry of the virus into the host.

There is a prospect that SARS-CoV-2 utilized mechanism similar to that of SARS-CoV as its receptor-binding domain (RBD) binding motif comprises the nucleotides connected to ACE2. Once SARS-CoV-2 enter into its host cell, ACE2 is shed and ADAM metallopeptidase domain 17 (ADAM17) exuviate it into the extra membrane space. This resulted into high concentration of angiotensin II from the transition of angiotensin I to angiotensin II by ACE2 and concomitant respiratory distress because angiotensin II negatively regulates the renin-angiotensin pathway consequently, damage the alveoli by increasing pulmonary vascular permeability [30]. Subsequent to SARS-CoV-2 proteins translation in the host, ORF3a protein is synthesized which codes for a SARS-CoV-2 related calcium (Ca2+) ion channel. It reacts with TNF receptor associated factor 3 (TRAF3) and initiates the transcription of Nuclear Factor kappa-light-chain-enhancer of activated B-cells (NF-kB) pathway, resulting to the secretion of the pro-IL-1B gene [31], ORF3a together with TRAF3 can mobilize the inflammasome complex which includes caspase 1, Nod-like receptor protein 3 (NLRP3) and apoptosis-associated speck-like protein containing a CARD (ASC). Another signaling which include caspases activation, mitochondrial damage, ROS production, and Ca2+ influx activates pro-IL-1B to interleukin 1 beta (IL-1B) which enhance cytokine production. Furthermore, ORF8b protein through NLRP3 facilitates the inflammasome pathway. ORF8b protein is longer in SARS-CoV-2 [31]. Further studies are needful to ascertain the benefit or significance of the extra-nucleotides as contained in SARS-CoV-2. The E protein that forms an ion channel is also implicated in the cytokine’s over-secretion (an occurrence referred to as cytokine storm syndromes which has been reported to be one of the major causes of respiratory distress in COVID-19) via NLRP3 inflammasome pathway (Figure 1) [32].

Figure 1.

Proposed mechanism of SARS-CoV-2.

c-Jun N- terminal kinase (JNK) pathway is also one of the vital SARS-CoV pathogenic pathways. It is activated by ORF3a, ORF3b, and ORF7a. and results in pro-inflammatory cytokines over-secretion. These over-secretions of inflammatory cytokines have deleterious effects on lung and can accelerate lungs damage [33]. Secondary haemophagocytic lymphohistiocytosis (sHLH) is a cytokine profile with a hyperinflammatory syndrome described by an abrupt hypercytokinaemia with multiorgan failure is related to COVID-19 severity. This also features increased granulocyte-colony stimulating factor, interferon-γ inducible protein 10, tumor necrosis factor-α, interleukin (IL)-2, macrophage inflammatory protein 1-α, IL-7, and monocyte chemoattractant protein 1 [33].

Additionally, SARS-CoV-2 exhibited higher infectivity and transmissibility but lower mortality rate when compared with other types of respiratory syndrome coronaviruses: severe acute respiratory syndrome coronavirus (SARS-CoV) and Middle East respiratory syndrome coronavirus (MERS-CoV). The noted increase in virulence of SARS-CoV-2 may be owing to great intensity and affinity at which SARS-CoV-2 attached to ACE2 and noted mutation in its genome sequence. The reported modifications on the SARS-CoV-2 gene include shorter 3b segments, alteration on Nsp 2 and 3 proteins, absent 8a, differences in orf8 and orf10 proteins, and longer 8b [34, 35, 36, 37].

Advertisement

3. Phytochemicals with possible SARS-CoV-2 inhibitory and neuroprotective activities

Prevention is one of the predefined frameworks to effectively approach health security threats. To prevent the emergence or re-emergence of potentially life-threatening diseases, necessary measures must be initiated and these measures must be accessible, affordable and effective. Moreover, transmission of IHS threats was able to increase at an accelerating rate due to an overburdening of local health-care systems and widespread poverty where people lacked access to adequate water and waste management infrastructure. Therefore, the preventive and/or curative measures must be affordable by the populace. Furthermore, the remedy must be such that will be generally accepted by all; this will enhance the response and hasten the curb of the pandemic.

Different therapeutic аррrоасhеѕ are being used ѕіnсе time іmmеmоrіаl for mаnу hеаlth ailments араrt frоm the рhаrmасоlоgісаl trеаtmеnts. Aррrоxіmаtеlу еіghtу реrсеnt оf thе Wоrld population still depends upon thе uѕе of herbal rеmеdіеѕ for thеіr hеаlth care. This trаdіtіоnаl mеthоd оf trеаtіng ailment іѕ trаnѕfеrrеd from one gеnеrаtіоn tо thе оthеr аll оvеr the wоrld. Dереndеnсе on рlаntѕ uѕаgе has been аttrіbutеd tо thеіr аffоrdаbіlіtу, еffесtіvеnеѕѕ, ѕаfеtу, сulturаl preferences, аnd аmрlе accessibility аt all tіmеѕ аnd when it іѕ nееdеd. Glоbаllу, traditional healers are using various medicinal plants fоr thе trеаtmеnt оf COVID-19. Wе thеrеfоrе рrеѕеnt ѕоmе of thе phytochemicals wіth thеrареutіс аbіlіtіеѕ whісh mау ѕеrvе аѕ еffесtіvе treatment for COVID-19 duе to thеіr аntіvіrаl, аntі-іnflаmmаtоrу, аntіоxіdаnt, аntіруrеtіс, immunomodulatory аnd суtорrоtесtіvе рrореrtіеѕ.

3.1 Kolaviron from Garcinia kola (Guttiferae)

Sіnсе time іmmеmоrіаl, medicinal рlаntѕ have become a ѕоurсе оf novel and affordable drug соmроundѕ аѕ plant-derived mеdісіnеѕ have made ѕіgnіfісаnt impacts tо humаn health аnd wеll-bеіng [38, 39]. Garcinia kоlа (bіttеr kоlа) іѕ a mеdісіnаl рlаnt аnd a mеmbеr of the Guttіfеrае fаmіlу. It іѕ аn evergreen tree lаrgеlу сultіvаtеd аnd hіghlу esteemed fоr its еdіblе nuts in Wеѕt аnd Cеntrаl Africa. Garcinia kоlа is соmmоnlу uѕеd bу thе реорlе duе tо its аbіlіtу to іmрrоvе mоuth оdоur аnd cause nеrvоuѕ alertness. In African traditional medicine, bіttеr kоlа is employed in the treatment and management оf lаrуngіtіѕ, thrоаt іnfесtіоnѕ, brоnсhіtіѕ, іnflаmmаtоrу dіѕоrdеrѕ, аnd аѕ an аntіbасtеrіаl, antiparasitic, аnd аntірurgаtіvе. Thе ѕееdѕ have аlѕо bееn uѕеd іn thе trеаtmеnt of сhrоnіс hераtіtіѕ and сhоlаngіtіѕ with ѕіgnіfісаnt іmрrоvеmеnt оf lіvеr funсtіоnѕ. Sіmіlаrlу, Garcinia kola ѕееdѕ are uѕеd a gеnеrаl tоnіс tо bооѕt the іmmunе ѕуѕtеm [40, 41].

Mаnу еxреrіmеntаl findings hаvе established thе trаdіtіоnаl medicinal uѕеѕ of Gаrсіnіа kola. Kоlаvіrоn, thе bіflаvаnоnе оf Gаrсіnіа kоlа (Figure 2) hаvе bееn dосumеntеd tо protect аgаіnѕt оxіdаtіvе stress аnd hераtоtоxісіtу іnduсеd bу many xenobiotics which іnсludеѕ аflаtоxіn, 2-асеtуlаmіnоfluоrеnе, саrbоn tеtrасhlоrіdе, dіmеthуlnіtrоѕаmіnе, раrасеtаmоl, рhаllоіdіn іn аnіmаl ѕtudіеѕ [42, 43, 44, 45]. Futhermore, thе рhаrmасоlоgісаllу асtіvities of bіflаvаnоnе оf Gаrсіnіа kola hаvе been ѕhоwn with many pharmacokinetic preferences оvеr basic mоnоmеrіс flаvоnоіdѕ as they pull through fіrѕt-раѕѕ mеtаbоlіѕm which incapacitates mоѕt flаvоnоіdѕ [40].

Figure 2.

2D structure of kolaviron (Kolaflavanone).

Nеurорrоtесtіvе аbіlіtіеѕ оf kolaviron has been rероrtеd іn many neuronal сеll lines. Abаrіkwu et аl. [46] dосumеntеd the рrоtесtіvе rоlеѕ оf kоlаvіrоn against atrazine іnduсеd toxic іnѕult іn human dораmіnеrgіс SH-SY5Y сеllѕ. Thе fіndіngѕ rеvеаlеd thаt thе аntіарорtоtіс and аntіоxіdаtіvе properties оf Kolaviron mаkе it effective to prevent agаіnѕt atrazine-іnduсеd tоxісіties. Sіmіlаrlу, kolaviron wаѕ rероrtеd tо рrоtесt аgаіnѕt арорtоtіс сеll death in pheochromocytoma derived (PC12) сеllѕ еxроѕеd to Atrаzіnе [47]. Igаdо еt al. [48] reported thе bіосhеmісаl and morphological examination on thе potential рrоtесtіvе effects оf kоlаvіrоn in vanadium-induced neuronal damage in rats. Kolaviron has bееn ѕhоwn tо suppress nеurоіnflаmmаtіоn іn BV2 mісrоglіа vіа thе Nrf2/ARE аntіоxіdаnt рrоtесtіvе mесhаnіѕm [49]. Alѕо, Olаjіdе еt al. [50] rероrtеd multіdіrесtіоnаl suppression оf соrtісо-hірросаmраl nеurоdеgеnеrаtіоn bу kоlаvіrоn. In аnоthеr ѕtudу, Omоtоѕо et аl. [51] reported thаt kolaviron ameliorated сuрrіzоnе-іnduсеd multiple ѕсlеrоѕіѕ in the brain of experimental animals.

In a recent study, wе rероrtеd thе neuroprotective effects оf kоlаvіrоn іn ѕtrіаtаl oxidative stress and neuroіnflаmmаtіоn аѕѕосіаtеd wіth rоtеnоnе mоdеl оf neurodegenerative dіѕеаѕе [52]. In thе ѕtudу, we showed that kolaviron rеstored rоtеnоnе-аѕѕосіаtеd еxрlоrаtоrу dеfісіtѕ, mоtоr/nеurоmuѕсulаr іnсоmреtеnсе and locomotor іmраіrmеnt. Also, kоlаvіrоn еffесtіvеlу аmеlіоrаtеd thе nеurоbіосhеmісаl іmbаlаnсе, ѕtrіаtаl neurodegeneration, nеurоіnflаmmаtіоn and аltеrеd аntіоxіdаnt dеfеnсе ѕуѕtеm іn thе brаіn of the nеurоdеgеnеrаtіvе rats. Kolaviron dіѕрlауеd a potential capacity tо enhance еffісіеnt gait wіth minimal severity аnd improved сооrdіnаtіоn. Thіѕ ѕhоwѕ thаt kоlаvіrоn соuld bе a prospective drug fоr the еffесtіvе management and/or trеаtmеnt of Parkinson’s disease.

Kolaviron has been noted to be a potential anti-COVID-19 drug candidate in a computational experimental study aimed to screen phytochemicals in drug repurposing approach to combat COVID-19 [53]. The study employed USCF Chimera in virtual screening and molecular docking for possible inhibitors of SARS-CoV-2. Kolaviron was observed to exhibited a higher docked score with the SARS-CoV-2 major protease (6 LU7) above remdesivir, a recommended drug for the treatment of COVID-19. This showed that kolaviron could offer a better inhibitory effect on SARS-CoV-2 and be a more effective drug candidate in the treatment of COVID-19.

3.2 Apigenin

Apigenin (4′,5,7-trihydroxyflavone) іѕ one of the most explored рhеnоlісѕ аnd the mоѕt commonlу disseminated flаvоnоіdѕ іn many рlаnt species (Figure 3). It is predominantly рrеѕеnt in hеrbѕ (оrеgаnо, thуmе, bаѕіl, chamomile), рhytochemical-bаѕеd beverages (tea, bееr, and wіnе), in vegetables (раrѕlеу, сеlеrу, onions), and fruіtѕ (оrаngеѕ). It іѕ аlѕо fоund extensively in Mаtrісаrіа, Achillea, Artеmіѕіа, and Tаnасеtum [54]. Aріgеnіn hаѕ bееn documented to have аntісаnсеr activities аѕ wеll аѕ theurapeutic effects on depression, Alzhеіmеr’ѕ dіѕеаѕе, аmnеѕіа, and іnѕоmnіа trеаtmеnt [54]. Thе dietary availability оf apigenin could fасіlіtаtе an efficacious іntеrvеntіоn tо inhibit activation of mісrоglіаl аnd рrevеnt onset of Alzhеіmеr’ѕ dіѕеаѕе.

Figure 3.

2D structure of apigenin.

Aftеr absorption, аріgеnіn саn easily bе trаnѕроrtеd through the сіrсulаtоrу ѕуѕtеm, сrоѕѕіng tо thе blооd-brаіn barrier tо the brain, where іt асts оn thе CNS and exhibits interaction wіth thе GABAA-rесерtоr [55, 56]. Sloley еt аl. [57] rероrtеd thе inhibitory activity of аріgеnіn оn neuronal mоnоаmіnе оxіdаѕеѕ. Unrеgulаtеd activities оf mоnоаmіnе oxidases may be one of the causes of some psychiatric cases аnd neurological dіѕоrdеrѕ. However, mоnоаmіnе oxidases іnhіbіtоrѕ such as аріgеnіn showed efficacy as antidepressant аnd аnxiolytic аgеntѕ.

Thе рrоtесtіvе roles оf аріgеnіn іn thе аmуlоіd рrесurѕоr рrоtеіn dоublе trаnѕgеnіс Alzhеіmеr’ѕ dіѕеаѕе mоuѕе has bееn reроrtеd bу Zhао еt аl. [58]. Apigenin іѕ аlѕо a роtеnt соgnіtіоn-еnhаnсіng, аntі-аmуlоіdоgеnіс, аntіоxіdаnt, nеurорrоtесtіvе, and аntі-іnflаmmаtоrу agent with efficacy іn thе prevention and/or trеаtmеnt оf nеurоdеgеnеrаtіvе dіѕеаѕеѕ [58]. Nаbаvі еt al. [59] іn a rеvіеw аrtісlе еmрhаѕіѕеd thе thеrареutіс роtеntіаlѕ оf аріgеnіn іn ѕоmе humаn сlіnісаl trials аnd еxреrіmеntаl animal mоdеlѕ. Furthеrmоrе, аріgеnіn’ѕ chemical ѕtruсturе, mеtаbоlіѕm of action, and рhаrmасоkіnеtісѕ were elucidated in relation to its medicinal usefulness in dерrеѕѕіоn, Pаrkіnѕоn’ѕ and Alzhеіmеr’ѕ dіѕеаѕеs [59].

Aріgеnіn hаѕ аlѕо dеmоnѕtrаtеd strong аntі-іnflаmmаtоrу рrореrtу in lіророlуѕассhаrіdе -induced mасrорhаgеѕ bу rеduсіng thе lеvеl of interleukin 6 (IL-6) {a pro-inflammatory суtоkіnе}. It also іnhіbіtеd tumоur nесrоѕіѕ fасtоr (TNF-α), interleukin 6, and cluster of dіffеrеntіаtіоn 40 (CD40) рrоduсtіоn vіа ѕuррrеѕѕіоn of іntеrfеrоn gamma-mediated STAT1 (ѕіgnаl transducers аnd асtіvаtоrѕ оf transcription 1) рhоѕрhоrуlаtіоn іn microglia [60]. An experimental study hаѕ еѕtаblіѕhеd thе іnhіbіtоrу ability оf аріgеnіn on nuclear factor kappa-light-chain-enhancer (NF-kB), fасіlіtаtеd by inhibition оf lіророlуѕассhаrіdе-mediated рhоѕрhоrуlаtіоn of thе р65 subunit [61]. Aріgеnіn also ѕuррrеѕѕеd thе activities of аdhеѕіоn molecules whісh іѕ vеrу еѕѕеntіаl tо mіtіgаtе оxіdаtіvе stress аnd рrеvеnt oxidative dаmаgе [62].

Aріgеnіn promotes thе rеlеаѕе оf суtорrоtесtіvе enzymes ѕuсh as glutаthіоnе-ѕ-trаnѕfеrаѕе, superoxide dismutase, аnd саtаlаѕе to inhibit аnd neutralize сеllulаr оxіdаtіvе. Similarly, аріgеnіn еnhаnсеѕ асtіvаtіоn оf Nrf-2 ѕіgnаlіng pathway leading tо іnсrеаѕе іn рhаѕе II еnzуmеѕ рrоduсtіоn [63, 64]. Anticancer рrореrtу of аріgеnіn in humаn сеll сulturе mоdеlѕ has bееn rероrtеd to bе via suppression оf аngіоgеnеѕіѕ and metastasis bу іntеrfеrіng wіth thе mаіn ѕіgnаlіng molecules іn mіtоgеn-асtіvаtеd protein kіnаѕе (MAPK) раthwауѕ which іnсludе с-Jun N-tеrmіnаl kіnаѕеѕ (JNK), еxtrасеllulаr-ѕіgnаl-rеgulаtеd kinase (ERK), аnd р38 [65].

Apigenin has been documented to interact with both S 1 and S2 domains of the spike protein of SARS-COV-2 with substantial binding energies thus unsettling viral attachment and internalization into the host [66]. Similarly, in silico study in our laboratory revealed that apigenin displayed a significant binding affinity with the SARS-CoV-2 major protease (6 LU7). The result also suggested that apigenin could be a potential inhibitor of SARS-COV-2 [53].

3.3 Phytochemicals from Vernonia amygdalina

Bitter leaf (Vernonia amygdalina) is an indigenous African plant with a number of scientific proven medicinal importance [67, 68, 69, 70]. Recent study from our laboratory has examined the possible inhibitory activity of selected phytochemical constituents of the leaf extract of Vernonia amygdalina (hydroxyvernolide, vernodalin, vernodalol, vernolide, and veronicoside) (Figure 4) against SARS-COV2 major protease (6 LU7) [71]. The phytochemicals exhibited significant binding affinity to the binding pocket of SARS-COV2 major protease suggesting them as potential molecules that could mitigate/inhibit SARS-COV2. Binding of these phytochemicalsto SARS-COV2 could inhibit or interfere the pathogenesis of COVID-19 thereby preventing its cellular entryand proliferation.

Figure 4.

2D structures of (A) Vernodalin, (B) Vernodalol, (C) Vernolide, (D) Veronicoside A.

Veronicoside has been reported to have radical scavenging and antioxidant activities. It has also been documented to have cytotoxicity activities againstHep-2 (human larynx epidermoidcarcinoma), RD (humanrhabdomyosarcoma), andL-20B (transgenic murine cells) cell lines [72]. Severalspecies of plants containing veronicoside are being used in traditional medicine to treat influenza, respiratory diseases, hernia, cough, laryngopharyngitis, cancer, hemoptysis, and are also used as an antiscorbutic and expectorant [73].

Vernodalinandvernolidehave been reported to exhibit antiproliferativeactivities [74] against lung A549 (adenocarcinomic human alveolar basal epithelial cells), HeLa, and MDA-MB-23 (human breast cancer) celllines and induced apoptosis on HepG2 cells with G2/Mphase cell cycle arrest [75]. They have potential to be usedas lead compounds in the development of a therapeutic natural product for treatment of cancers in the lungs, breast or liver. These phytochemicals may also offer help in inhibiting the proliferative activities of SARS-COV2 in the host thereby mitigate the pathogenesis of COVID-19.

Sinisi et al [76] has reported vernodalol has a good activator of Nrf2. NF-E2-related factor-2 (NRF2) is a transcriptional factor that bindsto and facilitates the activation of the ARE-dependent gene. Under basal conditions, NRF2 is sequestered in the cytoplasmand its expression is maintained to be low due to constantpolyubiquitination. In response to different kinds of stress, NRF2is significantly induced andtranslocates into the nucleus, where it activates the antioxidant responseelement (ARE)-dependent gene expression in association with small Maf proteins and other coactivators. Thus, causing the release of phase IIcytoprotective enzymes such as γ-glutamylcysteine ligase (γ-GCS), NAD[P]H:quinone oxidoreductase-1 (NQO1), heme oxygenase-1 (HO-1), and glutathioneS-transferase (GST) which protect the cells against the attack of the stress. Since oxidative stress has been reported as one of the features of COVID-19, vernodalol can help to extenuate it by activation of NRF2.

Nuclear Factor kappa-light-chain-enhancer of activated B-cells (NF-kB) pathway has been implicated in the mode of actions of SARS-COV2 [77, 78] leading to a cytokine profile resembling secondary haemophagocyticlymphohistiocytosis (sHLH) with a hyperinflammatory syndrome characterized by a fulminant and severe hypercytokinaemia with multiorgan failure. This is characterized by increased tumor necrosis factor-α, interleukin (IL)-2, IL-7, interferon-γ inducible protein 10, granulocyte-colony stimulating factor, macrophage inflammatory protein 1-α, and monocyte chemoattractant protein 1 [79]. However, vernolide and vernodalol have been documented to show marked inhibitory activity onSTAT3/NF-κB [76]. Therefore, vernolide and vernodalol could protect against COVID-19-induced multiorgan failure by suppressing the hyperinflammatory syndrome via inhibition of NF-kB.

3.4 Resveratrol

Resveratrol (3,5,4′-trihydroxystilbene) is a naturally occurring lipophilic and phenolic phytochemical found abundantly in edible plants and easily cross the plasma membrane after oral absorption [80, 81, 82]. it is a polyphenolic phytoalexin which comprises two aromatic rings linked by a styrene double bond which permit its trans- and cis-isomers formation (Figure 5) [83, 84]. Resveratrol has been reported as a possible reason accountable for the French paradox [85, 86], a phenomenon described by an epidemiological study that the French population displayed a comparatively low rate of coronary heart disease, in spite of their high consumption of saturated fat diet [87, 88]. A number of preclinical studies proposes that resveratrol has the capability to influence a variety of human diseases, this is due to its cardioprotective [89, 90] antiviral [91, 92], antiapoptotic [93, 94], anti-inflammatory [95, 96] antidiabetic [97, 98], and antioxidative [97, 99] properties.

Figure 5.

2D structure of resveratrol.

Evidences from experimental studies has established the neuroprotective properties of resveratrol which may be beneficial in combating neurological disorders showed in COVID-19 patients. Resveratrol enhances enzymes that are responsible in stress response, for instance quinone reductase 2 (QR2), a cytosolic enzyme which influences the release of destructive activated quinone and ROS, thus, exhibiting a pivotal role in the cellular response [100]. Previous report has showed that QR2 is overproduced in the hippocampus of rat’s brain in a model of learning deficits. Hippocampus is a brain region which is seriously affected in Alzheimer disease and it is primarily responsible for memory and learning. This indicates that the overproduction of this enzyme initiates memory impairments [101]. Similarly, neuroprotective effect of resveratrol has been documented to includes inhibition of microglia-mediated neuroinflammation [102]. Resveratrol has been demonstrated to inhibit the activation of NF-κB signaling pathways and mitogen-activated protein kinases (MAPKs) in lipopolysaccharides-induced dopaminergic neuronal death [102].

Activation of microglia is the hallmark of neuroinflammation and play a critical role in the pathogenesis of neurological diseases [103, 104]. Microglia is the neuronal immune cells that perform a vital role in the homeostasis in the central nervous system, and act as the first line of defense during cellular assaults, oxidative damage or progression of neurological diseases in the brain [105]. During microglial activation (microgliosis), different kinds of proinflammatory markers such as chemokines, prostaglandins, reactive nitrogen species, and cytokines are release. The overproduction and accumulation of these proinflammatory factors leads to damage of the neuronal cells and ultimately cause release of soluble factors and debris [106]. Many experimental studies have demonstrated the neuroprotective ability of resveratrol to inhibit the activation microglia [107, 108, 109]. Resveratrol has been reported to suppress upsurge expression of IL-1β, nitric oxide and TNFα that accompanied activation of microglia which mediate phosphorylation of p38 and NF-κB signaling [109, 110]. Resveratrol inhibited secretion of TNFα, IL-1β and reactive nitrogen species, and activation of microglia in the ischemic cortex [111].

Anti-covid-19 potentials of resveratrol has been reported in an in-silico study designed for drug development targeting SARS-CoV-2 Spike Protein of COVID-19 [66]. The study reported that resveratrol displayed a strong binding ability with the S2 domain of SARS-CoV-2spike protein. This spike glycoprotein, located on the surface of the virus (SARS-CoV-2), is a class I fusion protein which enhance the initial attachment of the virus with ACE2 receptor and itsconsecutive fusion with the host cells [112]. The ability of resveratrol to bind to this spike protein indicates that resveratrol may inhibit or alter the mechanism by which the virus gain entrance into its host. Furthermore, since resveratrol has been reported to modulate phosphoinositide3-kinase (PI3-k), NF-κB signaling and mitogen-activated protein kinases pathways whose end products release cytokines; It may provide beneficial effects in COVID-19 via these pathways to inhibit the over-secretion of inflammatory cytokines which resulted to cytokine storm syndromes that accelerate lungs damage and multiorgan failure is related to COVID-19.

3.5 Quercetin

Quercetin, 3,3′,4′5,7-pentahydroxyflavone (Figure 6), is a broadly disseminated plant polyphenol, found as conjugates with residualsugars (quercetin glycosides) in many grains, fruits, seeds, leaves, and vegetables (capers, onions, berries, and apples) [113]. The highest levels of quercetin among vegetables were found in red leaf lettuce, asparagus (Asparagus officinalis L.), and onions (Allium cepa L.), while peas, green peppers, broccoli, and tomatoes contained lower levels. Quercetin arabinoside, quercetin galactoside, and quercetin glucoside are examples of quercetin glycosides present in vegetables, fruits and other food items. They are first are deglycosylated by gut microbiota-derived betaglucosidase or lactase phlorizin hydrolaseto quercetin aglycone before passive absorption in the small intestine [114]. The quercetin aglycone produced then go through series of metabolic reactions to form methylated, sulphated, and glucuronidated metabolites, signifying participation of the phase II enzymes COMT (catechol-O-methyltransferase), SULT (sulfotransferase) and UGT (uridine 5-diphosphoglucuronosyltransferase) respectively.

Figure 6.

2D structure of quercetin.

Similar toother polyphenols, Studies have reported that quercetin exhibited anti-inflammatory, and immunoprotective [115], antioxidant [116], and antiviral [117], effects. It’s medicinal effects on cancer, nervous system disorders, gastrointestinal tract function, infections, inflammatory processes, diabetes, and cardiovascular diseases has been documented [118, 119, 120]). Previous findings have documented the inhibitory activities of quercetin against reverse transcriptase [121], proteases [122], and polymerases [123]. Also, it has been studied in modelsof viral infection to bind to viral capsidproteins and inhibit DNA gyrase [124, 125].

During viral infection, entrance of virus into the host cell is a vital step and has been targeted as a possible point of intervention in antiviral treatments [126, 127, 128]. Quercetin has been reported to inhibit H1N1 and H3N2 influenza infection ofMDCK cells through binding to hemagglutinin proteins which is accountablefor membrane fusion during virus entry and virus-mediatedhaemolysis [129]. Furthermore, quercetin has been studied to interfere with DNA and RNA polymerases in viral infections. During adenoviruses (ADV-3,−8,−11) and herpesviruses (HSV-1, 2) infections, quercetin was reported to suppress viral DNA and RNA polymerase [123, 130, 131] and inhibit the early stage of viral replication [125, 132]. Li et al. [133] also reported antiviral activities of quercetin against HIV via its ability to suppress protease, integrase and reversetranscriptase. Quercetin upregulated IL-13 and suppressed the levels of Long Terminal Repeat (LTR) gene expression, TNF-α, p24 in HIV infection [115].

Possible antiviral effect of quercetin on many types of Coronaviruses has been described by Yi et al. [134]. Quercetin metabolite have been documented to bind to SARS-Cov 3CL protease and suppressed itsproteolytic activity [135]. Quercetin has been studied through computational studies to interact with the S2 domain of spike protein of SARS-CoV-2, thus altering the virus entry process [66]. The obstruction of virus entrance into the host cell signifies a vital approach in antiviral therapy and quercetin hinders viral membrane fusion for SARS-Cov and influenza in vitro [134].

Advertisement

4. Conclusion

International health security is multifaceted phenomenon that threatened the peace existence of man including emerging infectious diseases such as the current global pandemic: COVID-19. COVID-19 is a highly infectious and severe acute respiratory disorder induced by a morbific virus referred to as SARS-CoV-2. Many COVID-19 patients have displayed neurological symptoms and signs which include anosmia, acute cerebrovascular disease, acute disseminated post-infectiousencephalomyelitis, encephalitis, etc. The underlying mechanisms of pathogenic actions of SARS-CoV-2 includes activated by ORF3a, ORF3b, and ORF7a via the JNK pathway which induces lung damage; reduction of ACE2 to enhance pulmonary vascular permeability and damage the alveoli; immunosuppression; hyper-inflammation characterized by a fulminant and fatal hyper-cytokinaemia with multi-organ failure. Prevention is one of the predefined frameworks to effectively approach health security threats. To prevent the emergence IHS threats, effective measures must be initiated. Moreover, transmission of IHS threats was able to increase at an accelerating rate due to an overburdening of local health-care systems. Therefore, the preventive and/or curative measures must be affordable by the populace. Dереndеnсе on рlаntѕ uѕаgе has been аttrіbutеd tо thеіr аffоrdаbіlіtу, еffесtіvеnеѕѕ, ѕаfеtу, сulturаl preferences, аnd аmрlе accessibility аt all tіmеѕ аnd when it іѕ nееdеd. Kolaviron, hydroxyvernolide, vernodalin, vernodalol, vernolide, and veronicoside, resveratrol, quercetin and apigenin are phytochemicals and natural products from medicinal plants with proven аntіvіrаl, аntіруrеtіс, аntі-іnflаmmаtоrу, суtорrоtесtіvе, аntіоxіdаnt, immunomodulatory, and pharmacological activities that can inhibit SARS-CoV-2 and mitigate COVID-19. The phytochemicals have been documented to suppress JNK and MAPK pathways which are essential in the pathogenesis of COVID-19. Taken together, these phytochemicals could be potential drug candidates in the treatment/management of COVID-19 mediated neuropathology.

References

  1. 1. WHO COVID-19 weekly epidemiological update- 24th November 2020. https://who.int/publications/m/item/weekly-epidermiological-update---24-november-2020.
  2. 2. NCDC. Coronavirus COVID-19. https://covid19.ncdc.gov.ng
  3. 3. Nicole K. Le, Manish Garg, Ricardo Izurieta, Sona M. Garg, Thomas J. Papadimos, Bonnie Arquilla, Andrew C. Miller, Abbas M. Khan, Tamara Worlton, Michael S. Firstenberg, Sagar C. Galwankar, Sunil Raina, Harry L. Anderson III, Rebecca Jeanmonod, Donald Jeanmonod, Ijeoma Nnodim Opara, Kristiana Kaufmann, Juan A. Asensio and Stanislaw P. Stawicki. International Health Security: A Summative Assessment by ACAIM Consensus Group. 2020. DOI: http://dx.doi.org/10.5772/intechopen.93214
  4. 4. De Cock KM et al. The new global health. Emerging Infectious Diseases. 2013; 19 (8):1192–1197
  5. 5. Sikka V et al. The emergence of Zika virus as a global health security threat: A review and a consensus statement of the INDUSEM Joint Working Group (JWG). Journal of Global Infectious Diseases. 2016; 8(1):3–15
  6. 6. Guan W-J, Ni Z-Y, Hu Y, Liang W-H, Ou C-Q, He J-X, et al. Clinical characteristics of 2019 novel coronavirus infection in China. N Engl J Med 2020. doi: 10.1056/NEJMoa2002032.
  7. 7. World Health Organization (WHO). Q&A on coronaviruses (COVID-19);2020. Available from: https://www.who.int/news-room/q-a-detail/q-a-coronaviruses. Accessed March 6, 2020.
  8. 8. Chen N, Zhou M, Dong X, Qu J, Gong F, Han Y, et al. Epidemiological and clini- cal characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: a descriptive study. Lancet 2020; 395:507–513.
  9. 9. Ahmed MU, Hanif M, Ali MJ,Haider MA, Kherani D, Memon GM,Karim AH andSattar A (2020)Neurological Manifestations ofCOVID-19 (SARS-CoV-2): A Review.Front. Neurol. 11:518.doi: 10.3389/fneur.2020.00518
  10. 10. Mao L, Jin H, Wang M, Hu Y, Chen S, He Q, et al. Neurologicmanifestations of hospitalized patients with coronavirus disease 2019 inWuhan, China. JAMA Neurol. (2020). e201127. doi: 10.1001/jamaneurol.2020.1127
  11. 11. Baig AM, Khaleeq A, Ali U, Syeda H. Evidence of the COVID-19virus targeting the CNS: tissue distribution, host–virus interaction, andproposed neurotropic mechanisms. ACS Chem Neurosci. (2020) 11:995–998.doi: 10.1021/acschemneuro.0c00122
  12. 12. Wong SH, Lui RN, Sung JJ. Covid-19 and the digestive system. J GastroenterolHepatol. (2020) 35:744–748. doi: 10.1111/jgh.15047
  13. 13. Swanson PA 2nd, McGavern DB. Viral diseases of the central nervous system.CurrOpinVirol. (2015) 11:44–54. doi: 10.1016/j.coviro.2014.12.009
  14. 14. Li YC, BaiWZ,Hashikawa T. The neuroinvasive potential of SARS-CoV2 mayplay a role in the respiratory failure of COVID-19 patients. JMedVirol. (2020)92:552–5. doi: 10.1002/jmv.25728
  15. 15. Ramos-Casals M, Brito-Zeron P, Lopez-Guillermo A, Khamashta MA, Bosch X. Adult haemophagocytic syndrome. Lancet 2014; 383: 1503–1516.
  16. 16. Henter JI, Samuelsson-Harne A, Arico M, Egeler RM, Elinder G, Filipovich AH, et al. Treatment of haemophagocyticlymphohistiocytosis with HLH-94 immunochemotherapy and bone marrow transplantation. Blood. 2002; 100: 2367–2373.
  17. 17. Seguin A, Galicier L, Boutboul D, Lemiale V, Azoulay E. Pulmonary involvement in patients with hemophagocytic lymphohistiocytosis. Chest 2016; 149: 1294–1301.
  18. 18. Bohmwald K, Gálvez NMS, Ríos M, Kalergis AM. Neurologic alterationsdue to respiratory virus infections. Front Cell Neurosci. (2018) 12:386.doi: 10.3389/fncel.2018.00386
  19. 19. Kalra S et al. The emergence of Ebola as a global health security threat: From ‘lessons learned’to coordinated multilateral containment efforts. Journal of Global Infectious Diseases. 2014; 6(4):164
  20. 20. Abramowitz S et al. The opposite of denial: Social learning at the onset of the Ebola emergency in Liberia. Journal of Health Communication. 2017; 22 (suppl 1):59–65.
  21. 21. Chen N, Zhou M, Dong X, Qu J, Gong F, Han Y, et al. Epidemiological and clini- cal characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: a descriptive study. Lancet 2020;395(10223):507–513.
  22. 22. van Boheemen S, de Graaf M, Lauber C, Bestebroer TM, Raj VS, Zaki AM, et al. Genomic characterization of a newly discovered coronavirus associated with acute respiratory distress syndrome in humans. MBio 2012;3(6): e00473–e0e512.
  23. 23. Phan, T., 2020. Novel coronavirus: from discovery to clinical diagnostics. Infect. Genet.Evo. 79, 104211. https://doi.org/10.1016/j.meegid.2020.104211.
  24. 24. Liu, Z., Xiao, X.,Wei, X., Li, J., Yang, J., Tan, H., Zhu, J., Zhang, Q.,Wu, J., Liu, L., 2020b. Composition and divergence of coronavirus spike proteins and host ACE2 receptors predict potential intermediate hosts of SARS-CoV-2. J. Med. Virol. https://doi.org/10.1002/jmv.25726.
  25. 25. Yin, Y., Wunderink, R.G., 2018.MERS, SARS and other coronaviruses as causes of pneumonia. Respirology 23, 130–137. https://doi.org/10.1111/resp.13196.
  26. 26. Zhao, Y., Zhao, Z.,Wang, Y., Zhou, Y., Ma, Y., Zuo,W., 2020. Single-cell RNA expressionprofilingof ACE2, the putative receptor of Wuhan 2019-nCov. BioRxivhttps://doi.org/10.1101/2020.01.26.919985.
  27. 27. Tai, W., He, L., Zhang, X., Pu, J., Voronin, D., Jiang, S., Zhou, Y., Du, L., 2020.Characterizationof the receptor-binding domain (RBD) of 2019 novel coronavirus: implication for developmentof RBD protein as a viral attachment inhibitor and vaccine. Cell. Mol.Immunol., 1–8 https://doi.org/10.1038/s41423-020-0400-4.
  28. 28. Glowacka I, Bertram S, Müller MA, Allen P, Soilleux E, Pfefferle S, et al. Evidence that TMPRSS2 activates the severe acute respiratory syndrome coronavirus spike protein for membrane fusion and reduces viral control by the humoral immune response. J Virol 2011;85(9):4122–4134.
  29. 29. Bertram S, Glowacka I, Müller MA, Lavender H, Gnirss K, Nehlmeier I, et al. Cleavage and activation of the severe acute respiratory syndrome coronavirus spike protein by human airway trypsin-like protease. J Virol 2011;85 (24):13363–13372.
  30. 30. Chan JF, To KK, Tse H, Jin DY, Yuen KY. Interspecies transmission and emergence of novel viruses: lessons from bats and birds. Trends Microbiol 2013; 21:544–555.
  31. 31. Siu, K.L., Yuen, K.S., Castaño-Rodriguez, C., Ye, Z.W., Yeung, M.L., Fung, S.Y., Yuan, S., Chan, C.P., Yuen, K.Y., Enjuanes, L., 2019. Severe acute respiratory syndrome coronavirus ORF3a protein activates the NLRP3 inflammasome by promoting TRAF3-dependent ubiquitination of ASC. FASEB J. 33, 8865–8877. https://doi.org/10.1096/fj.201802418R.
  32. 32. Nieto-Torres, J.L., Verdiá-Báguena, C., Jimenez-Guardeño, J.M., Regla-Nava, J.A., Castaño-Rodriguez, C., Fernandez-Delgado, R., Torres, J., Aguilella, V.M., Enjuanes, L., 2015. Severe acute respiratory syndrome coronavirus E protein transports calcium ions and activates the NLRP3 inflammasome. Virology 485, 330–339. https://doi.org/10.1016/j.virol.2015.08.010.
  33. 33. Huang C, Wang Y, Li X, et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet 2020; 395: 497–506.
  34. 34. Wu, A., Peng, Y., Huang, B., Ding, X.,Wang, X., Niu, P., Meng, J., Zhu, Z., Zhang, Z.,Wang, J., 2020a. Genome composition and divergence of the novel coronavirus (2019-nCoV) originating in China. Cell Host Microbe 27, 325–328. https://doi.org/10.1016/j.chom.2020.02.001.
  35. 35. Wu, C., Liu, Y., Yang, Y., Zhang, P., Zhong,W.,Wang, Y.,Wang, Q., Xu, Y., Li, M., Li, X., 2020b. Analysis of therapeutic targets for SARS-CoV-2 and discovery of potential drugs by computational methods. Acta Pharm. Sin. B https://doi.org/10.1016/j.apsb.2020.02.008.
  36. 36. Xu, J., Zhao, S., Teng, T., Abdalla, A.E., Zhu,W., Xie, L.,Wang, Y., Guo, X., 2020a. Systematic comparison of two animal-to-human transmitted human coronaviruses: SARS-CoV-2 and SARS-CoV. Viruses 12, 244. https://doi.org/10.3390/v12020244.
  37. 37. Xu, X., Yu, C., Qu, J., Zhang, L., Jiang, S., Huang, D., Chen, B., Zhang, Z., Guan, W., Ling, Z., 2020b. Imaging and clinical features of patients with 2019 novel coronavirus SARSCoV-2. Eur. J. Nucl. Med. and Mol. Imaging, 1–6 https://doi.org/10.1007/s00259-020-04735-9.
  38. 38. Oladele, J.O., Oyewole, O.I., Bello, O.K., Oladele, O.T. (2017). Hepatoprotective Effect of Aqueous Extract of Telfairia occidentalis on Cadmium Chloride-Induced Oxidative Stress and Hepatotoxicity in Rats. Journal of Drug Design andMedicinal Chemistry. 3(3): 32–36.
  39. 39. Oyewole, O.I., Oladele, J.O., and Oladele, O.T. (2017). Methanolic leaf extract of Ficus Exasperata Leaf attenuates Arsenate-Mediated hepatic and renal oxidative stress in rats. Res. J. of Health Sci. 5(2): 115–123.
  40. 40. Iwu MM (1985). Antihepatotoxic constituents of Garcinia kola seeds. Experientia 41: 699–670.
  41. 41. Taiwo, O.; Xu, H.-X.; Lee, S.F. (1999). Antibacteria activities of extracts from Nigerian chewing sticks. Phytother. Res., 13, 675–679.
  42. 42. Farombi EO, Tahnteng JG, Agboola AO, Nwankwo JO, and Emerole GO (2000). Chemoprevention of 2-acetylaminofluorene-induced hepatotoxicity and lipid peroxidation in rats by kolaviron-a Garcinia kola seed extract. Food Chem Toxicol; 38: 535–541.
  43. 43. Farombi, E O Adepoju, B F Ola-Davies O E and Emerole, G O. (2005). Chemoprevention of aflatoxin B1-induced genotoxicity and hepatic oxidative damage in rats by kolaviron, a natural biflavonoid of Garcinia kola seeds. European Journal of Cancer Prevention, 14, No. 3:207–214.
  44. 44. Farombi, E. O. (2000). Mechanisms for the hepatoprotective action of kolaviron: studies on hepatic enzymes, microsomal lipids and lipid peroxidation in carbontetrachloride-treated rats Pharmacological Research, Vol. 42, No. 1, 2000.
  45. 45. Farombi, E. O., Shrotriya, S., and Surh, Y. J. (2009). Kolaviron inhibits dimethyl nitrosamine-induced liver injury by suppressing COX-2 and iNOS expression via NF-κB and AP-1. Life Sciences. Volume 84, Issues 5–6, Pages 149–155.
  46. 46. Abarikwu, S O., Farombi E. O, Kashyap, M. P., and Pant, A. B. (2011). Kolaviron protects apoptotic cell death in PC12 cells exposed to Atrazine Free Radical Research, September 2011; 45(9): 1061–1073
  47. 47. Abarikwu, S.O., Farombi, E.O., and Pant, A.B. (2011)b.Biflavanone-kolaviron protects human dopaminergic SH-SY5Y cells against atrazine induced toxic insult Toxicology in Vitro 25 848–858.
  48. 48. Igado, O. O, Olopade, J O, Adesida A, Aina, O. O., and Farombi, E. O. (2012). Morphological and biochemical investigation into the possible neuroprotective effects of kolaviron (Garcinia kola bioflavonoid) on the brains of rats exposed to vanadium. Drug and Chemical Toxicology, 35(4): 371–380.
  49. 49. Onasanwo, S.A., Velagapudi, R., El-Bakoush, A., Olajide, O.A., 2016. Inhibition ofneuroinflammationin BV2 microglia by the biflavonoid kolaviron is dependent on theNrf2/ARE antioxidant protective mechanism. Mol. Cell. Biochem. 414, 23–26.
  50. 50. Olajide, O.J., Asogwa, N.T., Moses, B.O., Oyegbola, C.B., 2017. Multidirectional inhibitionof cortico-hippocampal neurodegeneration by kolaviron treatment in rats.Metab. Brain Dis. https://doi.org/10.1007/s11011-017-0012-6.
  51. 51. Omotoso, G.O., Ukwubile, I.I., Arietarhire, L., Sulaimon, F., Gbadamosi, I.T., 2018.Kolaviron Protects the brain in Cuprizone-induced Model of Experimental MultipleSclerosis viaenhancement of intrinsic antioxidant mechanisms: Possible TherapeuticApplications?Pathophysiology 25, 299–306. https://doi.org/10.1016/j.pathophys.2018.04.004.
  52. 52. Farombi EO, Awogbindin IO, Farombi TH, Oladele JO, Izomoh ER, Aladelokun OB, Ezekiel IO, Adebambo OI, Abah VO (2019) Neuroprotective role of kolaviron in striatal redo-inflammation associated with rotenone model of Parkinson’s disease. Neurotoxicology 73:132–141. https://doi.org/10.1016/j.neuro.2019.03.005
  53. 53. Oladele JO, Oyeleke OM, Oladele OT, Olowookere BD, Oso BJ, Oladiji AT. (2020). Kolaviron (Kolaflavanone), apigenin, fisetin as potential Coronavirus inhibitors: In silico investigation
  54. 54. Dewick, P.M. Chimica, Biosintesi e BioattivitàdelleSostanzeNaturali; Piccin: Roma, Italy, 2001.
  55. 55. Campbell, E.L.; Chebib, M.; Johnston, G.A.R. The dietary flavonoids apigenin and (−)-epigallocatechingallate enhance the positive modulation by diazepam of the activation by GABA of recombinant GABA(A)receptors. Biochem. Pharmacol. 2004, 68, 1631–1638.
  56. 56. Jäger, A.K.; Krydsfeldt, K.; Rasmussen, H.B. Bioassay-guided isolation of apigenin with GABAbenzodiazepineactivity from Tanacetum parthenium. Phytother. Res. 2009, 23, 1642–1644.
  57. 57. Sloley, B.D.; Urichuk, L.J.; Morley, P.; Durkin, J.; Shan, J.J.; Pang, P.K.T.; Coutts, R.T. Identification ofkaempferol as a monoamine oxidase inhibitor and potential neuroprotectant in extracts of Ginkgo bilobaleaves. J. Pharm. Pharmacol. 2000, 52, 451–459.
  58. 58. Zhao, L.;Wang, J.; Liu, R.; Li, X.X.; Li, J.; Zhang, L. Neuroprotective, anti-amyloidogenic and neurotrophiceffects of apigenin in an Alzheimer’s disease mouse model. Molecules 2013, 18, 9949–9965.
  59. 59. Nabavi, S.F.; Khan, H.; D’onofrio, G.; Šamec, D.; Shirooie, S.; Dehpour, A.R.; Argüelles, S.; Habtemariam, S.;Sobarzo-Sanchez, E. Apigenin as neuroprotective agent: Of mice and men. Pharm. Res. 2018, 128, 359–365.
  60. 60. Rezai-Zadeh, K.; Ehrhart, J.; Bai, Y.; Sanberg, P.R.; Bickford, P.; Tan, J.; Douglas, R.D.Apigenin and luteolinmodulate microglial activation via inhibition of STAT1-induced CD40expression. J. Neuroinflamm. 2008, 5,41–51.
  61. 61. Nicholas, C.; Batra, S.; Vargo, M.A.; Voss, O.H.; Gavrilin, M.A.;Wewers, M.D.; Guttridge, D.C.; Grotewold, E.;Doseff, A.I. Apigenin blocks lipopolysaccharide-induced lethality in vivo and proinflammatory cytokinesexpression by inactivating NF-kappaB through the suppression of p65 phosphorylation. J. Immunol. 2007,179, 7121–7127.
  62. 62. Myhrstad, M.C.W.; Carlsen, H.; Nordström, O.; Blomhoff, R.; Moskaug, J.Ø. Flavonoids increase theintracellular glutathione level by transactivation of the -glutamylcysteine synthetase catalytical subunitpromoter. Free Radic. Biol. Med. 2002, 32, 386–393.
  63. 63. Paredes-Gonzalez, X.; Fuentes, F.; Jeffery, S.; Saw, C.L.L.; Shu, L.; Su, Z.Y.; Kong, A.N.T. Induction ofNRF2-mediated gene expression by dietary phytochemical flavones apigenin and luteolin. Biopharm. DrugDispos. 2015, 36, 440–451.
  64. 64. Huang, C.S.; Lii, C.K.; Lin, A.H.; Yeh, Y.W.; Yao, H.T.; Li, C.C.;Wang, T.S.; Chen, H.W. Protection by chrysin,apigenin, and luteolin against oxidative stress is mediated by the Nrf2-dependent up-regulation of hemeoxygenase 1 and glutamate cysteine ligase in rat primary hepatocytes. Arch. Toxicol. 2013, 87, 167–178.
  65. 65. Peng, Q.; Deng, Z.; Pan, H.; Gu, L.; Liu, O.; Tang, Z. Mitogen-activated protein kinase signaling pathway inoral cancer. Oncol. Lett. 2017, 15, 1379–1388.
  66. 66. Jitendra S. R., Aroni C., Abhijeet K., Shashikant R. (2020): Targeting SARS-CoV-2 spike protein of COVID-19 with naturally occurring phytochemicals: An in silico study for drug development. ChemRxiv. Preprint. https://doi.org/10.26434/chemrxiv.12094203.v1.
  67. 67. Oladele J.O.,Oyeleke O.M., Oladele O.T., Babatope O.D., Awosanya O.O. (2020). Nitrobenzene-induced hormonal disruption, alteration of steroidogenic pathway, and oxidative damage in rat: protective effects of Vernonia amygdalina. Clinical Phytoscience.
  68. 68. Oladele JO.,Oyeleke OM., Oladele OT. Olaniyan MD. (2020). Neuroprotective mechanism of Vernonia amygdalina in a rat model of neurodegenerative diseases.Toxicology report. https://doi.org/10.1016/j.toxrep.2020.09.005.
  69. 69. Oladele JO.,Oyeleke OM.,Akindolie BO., Olowookere BD., Oladele OT. (2021). Vernonia amygdalina abates oxidative hepatic damage and inflammation associated with nitrobenzene in rat. Jordan Journal of Biological Sciences. in press.
  70. 70. Oladele JO.,Oyeleke OM.,Olowookere BD., Babatope OD., Olaniyan MD., Akindolie BO., Oladele OT. (2020). Bitter leaf (Vernonia amygdalina) modulates nitrobenzene-induced renal damage in rats via suppression of oxido-inflammatory activities. Serbian Journal of Experimental and Clinical Research. in press.
  71. 71. Oladele JO, Oyeleke OM, Oladele OT, Oladiji AT. (2020).Covid-19 treatment: Investigation on the phytochemical constituents of Vernonia amygdalina as potential Coronavirus-2 inhibitors. Computational toxicology.
  72. 72. IclalSaracoglu, F. Handan Oztunca, AkitoNagatsu, and U. SebnemHarput. Iridoid content and biological activities of Veronica cuneifoliasubsp. cuneifoliaand V. CymbalariaPharmaceutical Biology, 2011; 49(11): 1150–1157
  73. 73. Graham JG, Quinn ML, Fabricant DS, Farnsworth NR. (2000). Plantsused against cancer - an extension of the work of JonathanHartwell. j Ethnopharmacol, 73, 347–377.
  74. 74. T. Ito, S. Aimaiti, N.N. Win, T. Kodama, H. Morita, New sesquiterpene lactones,vernonilides A and B, from the seeds of Vernonia anthelminticain Uyghur and theirantiproliferative, Bioorg. Med. Chem. Lett. 26 (2016) 3608–3611, https://doi.org/10.1016/j.bmcl.2016.06.009.
  75. 75. SanitThongnesta, PornsudaChawengrumb, SiripornKeeratichamroen,KriengsakLirdprapamongkol, ChatchakornEurtivong, JutatipBoonsombat,PrasatKittakoop, JisnusonSvasti, SomsakRuchirawata. Vernodalidimer L, a sesquiterpene lactone dimer from Vernonia extensa andanti-tumor effects of vernodalin, vernolepin, and vernolide on HepG2 livercancer cellsBioorganic Chemistry 92 (2019) 103197.
  76. 76. Annamaria Sinisi,Estrella Millán,Solomon M. Abay,Annette Habluetzel, GiovanniAppendino,Eduardo Muñoz,and OrazioTaglialatela-Scafati. Poly-Electrophilic Sesquiterpene Lactones from Vernoniaamygdalina: New Members and Differences in Their Mechanism ofThiol Trapping and in Bioactivity. Journal of Natural ProductsDOI: 10.1021/acs.jnatprod.5b00179
  77. 77. Siu, K.L., Yuen, K.S., Castaño-Rodriguez, C., Ye, Z.W., Yeung, M.L., Fung, S.Y., Yuan, S., Chan, C.P., Yuen, K.Y., Enjuanes, L., 2019. Severe acute respiratory syndrome coronavirus ORF3a protein activates the NLRP3 inflammasome by promoting TRAF3-dependent ubiquitination of ASC. FASEB J, 33: 8865–8877.
  78. 78. Oladele JO, Ajayi EIO, Oyeleke OM, Oladele OT, Olowookere BD, Adeniyi BM, Oyewole OI(2020) Curative potentials of Nigerian medicinal plants in COVID-19 treatment: A Mechanistic approach. Jordan Journal of Biological Science. in press.
  79. 79. Huang C, Wang Y, Li X, et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet 2020; 395: 497–506.
  80. 80. Jeandet, P., Douillet-Breuil, A. C., Bessis, R., Debord, S., Sbaghi,M., and Adrian, M. (2002) Phytoalexins from the Vitaceae:Biosynthesis, phytoalexin gene expression in transgenic plants,antifungal activity, and metabolism. J. Agric. Food Chem. 50, 2731–2741.
  81. 81. de Santi, C., Pietrabissa, A., Mosca, F., and Pacifici, G. M.(2000) Glucuronidation of resveratrol, a natural product present ingrape and wine, in the human liver. Xenobiotica 30, 1047–1054.
  82. 82. De Santi, C., Pietrabissa, A., Spisni, R., Mosca, F., and Pacifici, G.M. (2000) Sulphation of resveratrol, a natural compound present inwine, and its inhibition by natural flavonoids. Xenobiotica 30, 857–866.
  83. 83. Ribeiro de Lima, M. T., Waffo-Teguo, P., Teissedre, P. L.,Pujolas, A., Vercauteren, J., Cabanis, J. C., and Merillon, J. M. (1999)Determination of stilbenes (trans-astringin, cis- and trans-piceid, andcis- and trans-resveratrol) in Portuguese wines. J. Agric. Food Chem. 47,2666–2670.
  84. 84. BerzasNevado, J. J., Contento Salcedo, A. M., and CastanedaPenalvo, G. (1999) Simultaneous determination of cis- and transresveratrolin wines by capillary zone electrophoresis. Analyst 124, 61–66.
  85. 85. Kopp, P. (1998) Resveratrol, a phytoestrogen found in red wine.A possible explanation for the conundrum of the ‘French paradox’?Eur. J. Endocrinol. 138, 619–620.
  86. 86. Hengst, J. A., and Yun, J. K. (2012) Sphingosine kinase: A key tosolving the ‘French Paradox’? Br. J. Pharmacol. 166, 1603–1604.
  87. 87. Ferrieres, J. (2004) The French paradox: Lessons for othercountries. Heart 90, 107–111.
  88. 88. Constant, J. (1997) Alcohol, ischemic heart disease, and theFrench paradox. Coron. Artery Dis. 8, 645–649.
  89. 89. Wang, Z., Zou, J., Cao, K., Hsieh, T. C., Huang, Y., and Wu, J.M. (2005) Dealcoholized red wine containing known amounts ofresveratrol suppresses atherosclerosis in hypercholesterolemic rabbitswithout affecting plasma lipid levels. Int. J. Mol. Med. 16, 533–540.
  90. 90. Szmitko, P. E., and Verma, S. (2005) Cardiology patient pages.Red wine and your heart. Circulation 111, e10–e11.
  91. 91. Berardi, V., Ricci, F., Castelli, M., Galati, G., and Risuleo, G.(2009) Resveratrol exhibits a strong cytotoxic activity in cultured cellsand has an antiviral action against polyomavirus: potential clinical use.J. Exp. Clin. Cancer Res. 28, 96.
  92. 92. Clouser, C. L., Chauhan, J., Bess, M. A., Oploo, J. L., Zhou, D.,Dimick-Gray, S., Mansky, L. M., and Patterson, S. E. (2012) Anti-HIV-1 activity of resveratrol derivatives and synergistic inhibition of HIV-1by the combination of resveratrol and decitabine. Bioorg. Med. Chem.Lett. 22, 6642–6646.
  93. 93. Nicolini, G., Rigolio, R., Miloso, M., Bertelli, A. A., and Tredici,G. (2001) Anti-apoptotic effect of trans-resveratrol on paclitaxelinducedapoptosis in the human neuroblastoma SH-SY5Y cell line.Neurosci. Lett. 302, 41–44.
  94. 94. Baarine, M., Thandapilly, S. J., Louis, X. L., Mazue, F., Yu, L.,Delmas, D., Netticadan, T., Lizard, G., and Latruffe, N. (2011) Proapoptoticversus anti-apoptotic properties of dietary resveratrol ontumoral and normal cardiac cells. Genes Nutr. 6, 161–169.
  95. 95. Udenigwe, C. C., Ramprasath, V. R., Aluko, R. E., and Jones, P. J.(2008) Potential of resveratrol in anticancer and anti-inflammatorytherapy. Nutr. Rev. 66, 445–454.
  96. 96. Chen, G., Shan, W., Wu, Y., Ren, L., Dong, J., and Ji, Z. (2005)Synthesis and anti-inflammatory activity of resveratrol analogs. Chem.Pharm. Bull. (Tokyo) 53, 1587–1590.
  97. 97. Chang, C. C., Chang, C. Y., Huang, J. P., and Hung, L. M.(2012) Effect of resveratrol on oxidative and inflammatory stress inliver and spleen of streptozotocin-induced type 1 diabetic rats. Chin. J.Physiol. 55, 192–201.
  98. 98. Dao, T. M., Waget, A., Klopp, P., Serino, M., Vachoux, C.,Pechere, L., Drucker, D. J., Champion, S., Barthelemy, S., Barra, Y.,Burcelin, R., and Seree, E. (2011) Resveratrol increases glucoseinduced GLP-1 secretion in mice: A mechanism which contributes tothe glycemic control. PLoS One 6, No. e20700.
  99. 99. Spanier, G., Xu, H., Xia, N., Tobias, S., Deng, S., Wojnowski, L.,Forstermann, U., and Li, H. (2009) Resveratrol reduces endothelialoxidative stress by modulating the gene expression of superoxidedismutase 1 (SOD1), glutathione peroxidase 1 (GPx1) and NADPHoxidase subunit (Nox4). J. Physiol. Pharmacol. 60 (Suppl 4), 111–116.
  100. 100. F. Vella, G. Ferry, P. Delagrange, J.A. Boutin, NRH:quinone reductase 2: an enzymeof surprises and mysteries, Biochem. Pharmacol. 71 (2005) 1–12.
  101. 101. C.E. Benoit, S. Bastianetto, J. Brouillette, Y. Tse, J.A. Boutin, P. Delagrange, T.Wong, P.Sarret, R. Quirion, Loss of quinone reductase 2 function selectively facilitateslearning behaviors, J. Neurosci. 30 (2010) 12690–12700.
  102. 102. F. Zhang, J. Liu, J.S. Shi, Anti-inflammatory activities of resveratrol in the brain: roleof resveratrol in microglial activation, Eur. J. Pharmacol. 636 (2010) 1–7.
  103. 103. Block, M. L., and Hong, J. S. (2007) Chronic microglialactivation and progressive dopaminergic neurotoxicity. Biochem. Soc.Trans. 35, 1127–1132.
  104. 104. Gao, H. M., Liu, B., Zhang, W., and Hong, J. S. (2003) Novelanti-inflammatory therapy for Parkinson’s disease. Trends Pharmacol.Sci. 24, 395–401.
  105. 105. Ransohoff, R. M., and Perry, V. H. (2009) Microglialphysiology: Unique stimuli, specialized responses. Annu. Rev. Immunol.27, 119−145.
  106. 106. Zhang, F., Liu, J., and Shi, J. S. (2010) Anti-inflammatoryactivities of resveratrol in the brain: role of resveratrol in microglialactivation. Eur. J. Pharmacol. 636, 1–7.
  107. 107. Candelario-Jalil, E., de Oliveira, A. C., Graf, S., Bhatia, H. S.,Hull, M., Munoz, E., and Fiebich, B. L. (2007) Resveratrol potentlyreduces prostaglandin E2 production and free radical formation inlipopolysaccharide-activated primary rat microglia. J. Neuroinflammation4, 25.
  108. 108. Lorenz, P., Roychowdhury, S., Engelmann, M., Wolf, G., andHorn, T. F. (2003) Oxyresveratrol and resveratrol are potentantioxidants and free radical scavengers: Effect on nitrosative andoxidative stress derived from microglial cells. Nitric Oxide 9, 64–76.
  109. 109. Bi, X. L., Yang, J. Y., Dong, Y. X., Wang, J. M., Cui, Y. H.,Ikeshima, T., Zhao, Y. Q., and Wu, C. F. (2005) Resveratrol inhibitsnitric oxide and TNF-alpha production by lipopolysaccharide-activatedmicroglia. Int. Immunopharmacol. 5, 185–193.
  110. 110. Bureau, G., Longpre, F., and Martinoli, M. G. (2008)Resveratrol and quercetin, two natural polyphenols, reduce apoptoticneuronal cell death induced by neuroinflammation. J. Neurosci. Res. 86,403–410.
  111. 111. Shin, J. A., Lee, H., Lim, Y. K., Koh, Y., Choi, J. H., and Park, E.M. (2010) Therapeutic effects of resveratrol during acute periodsfollowing experimental ischemic stroke. J. Neuroimmunol. 227, 93–100.
  112. 112. Oladele JO, Ajayi EIO, Oyeleke OM, Oladele OT, Olowookere BD, Adeniyi BM, Oyewole OI, Oladiji AT.(2020). Asystematic review on COVID-19 pandemic with special emphasis on Curative potentials ofmedicinal plants. Heliyon. https://doi.org/10.1016/j.heliyon.2020.e04897.
  113. 113. Li Y, Yao J, Han C, Yang J, Chaudhry MT, Wang S, et al.Quercetin, inflammation and immunity. Nutrients. (2016)8:167. doi: 10.3390/nu8030167
  114. 114. Y. Guo andR. S. Bruno, “Endogenous and exogenousmediatorsof quercetin bioavailability,” The Journal of Nutritional Biochemistry,vol. 26, no. 3, pp. 201–210, 2015.
  115. 115. Nair MP, Kandaswami C, Mahajan S, Chadha KC, Chawda R,NairandSchwartz SAH. The flavonoid, quercetin, differentially regulatesTh-1 (IFNgamma) and Th-2 (IL4) cytokine gene expression by normalperipheral blood mononuclear cells. BiochimBiophys Acta. (2002)1593:29–36. doi: 10.1016/S0167-4889(02)00328-2
  116. 116. Robaszkiewicz A, Balcerczyk A, Bartosz G. Antioxidative and prooxidativeeffects of quercetin on A549 cells. Cell Biol Int. (2007) 31:1245–1250. doi: 10.1016/j.cellbi.2007.04.009
  117. 117. Uchide N, Toyoda H. Antioxidant therapy as a potential approachto severe influenza-associated complications. Molecules. (2011) 16:2032–2052. doi: 10.3390/molecules16032032
  118. 118. G. S. Kelly, “Quercetin. Monograph,” Alternative MedicineReview, vol. 16, no. 2, pp. 172–194, 2011.
  119. 119. M. Russo, C. Spagnuolo, I. Tedesco, S. Bilotto, and G. L. Russo,“The flavonoid quercetin in disease prevention and therapy:facts and fancies,” Biochemical Pharmacology, vol. 83, no. 1, pp.6–15, 2012.
  120. 120. A.W. Boots, G. R. M. M.Haenen, and A. Bast, “Health effects ofquercetin: from antioxidant to nutraceutical,” European Journalof Pharmacology, vol. 585, no. 2–3, pp. 325–337, 2008.
  121. 121. Spedding G, Ratty A, Middleton E Jr. Inhibition of reversetranscriptases by flavonoids. Antiviral Res. (1989) 12:99–110. doi: 10.1016/0166-3542(89)90073-9
  122. 122. Bachmetov L, Gal-Tanamy M, Shapira A, Vorobeychik M, Giterman-GalamT, Sathiyamoorthy P, et al. Suppression of hepatitis C virus by the flavonoidquercetin is mediated by inhibition of NS3 protease activity. J Viral Hepat.(2012) 19:e81–8. doi: 10.1111/j.1365-2893.2011.01507.x
  123. 123. Shinozuka K, Kikuchi Y, Nishino C, Mori A, Tawata S. Inhibitory effectof flavonoids on DNA-dependent DNA and RNA polymerases. Experientia.(1988) 44:882–885. doi: 10.1007/BF01941188
  124. 124. Cushnie TP, Lamb AJ. Antimicrobial activity of flavonoids. Int J AntimicrobAgents. (2005) 26:343–356. doi: 10.1016/j.ijantimicag.2005.09.002
  125. 125. Debiaggi M, Tateo F, Pagani L, Luini M, Romero E. Effects ofpropolis flavonoids on virus infectivity and replication. Microbiologica.(1990) 13:207–213.
  126. 126. Liu S, Wu S, Jiang S. HIV entry inhibitors targeting gp41: frompolypeptides to small-molecule compounds. CurrPharmDes. (2007) 13:143–162. doi: 10.2174/138161207779313722
  127. 127. Yang J, Li M, Shen X, Liu S. Influenza A virus entry inhibitors targeting thehemagglutinin. Viruses. (2013) 5:352–373. doi: 10.3390/v5010352
  128. 128. Xia S, Liu Q, Wang Q, Sun Z, Su S, Duand L, et al. Middle East respiratorysyndrome coronavirus (MERS-CoV) entry inhibitors targeting spike protein.Virus Res. (2014) 194:200–210. doi: 10.1016/j.virusres.2014.10.007
  129. 129. Wu W, Li R, Li X, He J, Jiang S, Liu S, et al. Quercetin as anantiviral agent inhibits Influenza A Virus (IAV) entry. Viruses. (2015)8:6. doi: 10.3390/v8010006
  130. 130. Ono K, Nakane H. Mechanisms of inhibition of various cellular DNAand RNA polymerases by several flavonoids. J Biochem. (1990) 108:609–613. doi: 10.1093/oxfordjournals.jbchem.a123251
  131. 131. Ono K, Nakane H, Fukushima M, Chermann JC, Barré-Sinoussi F.Differential inhibitory effects of various flavonoids on the activities of reversetranscriptase and cellular DNA and RNA polymerases. Eur J Biochem. (1990)190:469–76. doi: 10.1111/j.1432-1033.1990.tb15597.x
  132. 132. Chiang LC, Chiang W, Liu MC, Lin CC. In vitro antiviral activities ofCaesalpinia pulcherrima and its related flavonoids. J Antimicrob Chemother.(2003) 52:194–198. doi: 10.1093/jac/dkg291
  133. 133. Li BW, Zhang FH, Serrao E, Chen H, Sanchez TW, Yang LM, et al. Designand discovery of flavonoid-based HIV-1 integrase inhibitors targeting boththe active site and the interaction with LEDGF/p75. BioorgMed Chem. (2014)22:3146–58. doi: 10.1016/j.bmc.2014.04.016
  134. 134. Yi L, Li Z, Yuan K, Qu X, Chen J, Wang G, et al. Small molecules blockingthe entry of severe acute respiratory syndrome coronavirus into host cells. JVirol. (2004) 78:11334. doi: 10.1128/JVI.78.20.11334-11339.2004
  135. 135. Chen L, Li J, Luo C, Liu H, Xu W, Chen G, et al. Bindinginteraction of quercetin-3-beta-galactoside and its synthetic derivativeswith SARS-CoV 3CL(pro): structure-activity relationship studies revealsalient pharmacophore features. Bioorg Med Chem. (2006) 14:8295–8306. doi: 10.1016/j.bmc.2006.09.014

Written By

Johnson Olaleye Oladele, Oluwaseun Titilope Oladele, Oyedotun M. Oyeleke and Adenike T. Oladiji

Submitted: 02 December 2020 Reviewed: 15 January 2021 Published: 10 March 2021