Next Article in Journal
Non-Coding RNAs in the Regulation of Hippocampal Neurogenesis and Potential Treatment Targets for Related Disorders
Previous Article in Journal
Use of Insect Promoters in Genetic Engineering to Control Mosquito-Borne Diseases
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Bioactivity and In Silico Studies of Isoquinoline and Related Alkaloids as Promising Antiviral Agents: An Insight

by
Divya Sharma
1,
Neetika Sharma
2,
Namish Manchanda
1,
Satyendra K. Prasad
3,
Prabodh Chander Sharma
1,
Vijay Kumar Thakur
4,5,*,
M. Mukhlesur Rahman
6 and
Mahaveer Dhobi
1,*
1
School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, Sector-III, Pushp Vihar, New Delhi 110017, India
2
Delhi Institute of Pharmaceutical Sciences and Research, Delhi Pharmaceutical Sciences and Research University, Sector-III, Pushp Vihar, New Delhi 110017, India
3
Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur 440033, India
4
Biorefining and Advanced Materials Research Centre, Scotland’s Rural College (SRUC), Kings Buildings, 11 West Mains Road, Edinburgh EH9 3JG, UK
5
School of Engineering, University of Petroleum & Energy Studies (UPES), Dehradun 248007, India
6
Pharmaceutical and Natural Products Chemistry, School of Health, Sports and Bioscience, University of East London, Stratford Campus, London E15 4LZ, UK
*
Authors to whom correspondence should be addressed.
Biomolecules 2023, 13(1), 17; https://doi.org/10.3390/biom13010017
Submission received: 14 November 2022 / Revised: 10 December 2022 / Accepted: 15 December 2022 / Published: 21 December 2022

Abstract

:
Viruses are widely recognized as the primary cause of infectious diseases around the world. The ongoing global pandemic due to the emergence of SARS-CoV-2 further added fuel to the fire. The development of therapeutics becomes very difficult as viruses can mutate their genome to become more complex and resistant. Medicinal plants and phytocompounds could be alternative options. Isoquinoline and their related alkaloids are naturally occurring compounds that interfere with multiple pathways including nuclear factor-κB, mitogen-activated protein kinase/extracellular-signal-regulated kinase, and inhibition of Ca2+-mediated fusion. These pathways play a crucial role in viral replication. Thus, the major goal of this study is to comprehend the function of various isoquinoline and related alkaloids in viral infections by examining their potential mechanisms of action, structure-activity relationships (SAR), in silico (particularly for SARS-CoV-2), in vitro and in vivo studies. The current advancements in isoquinoline and related alkaloids as discussed in the present review could facilitate an in-depth understanding of their role in the drug discovery process.

1. Introduction

Human health has been impacted for decades by a variety of life-threatening viruses such as the hepatitis C virus (HCV), influenza virus, herpes simplex virus (HSV), hepatitis B virus (HBV), dengue virus (DENV), human immunodeficiency virus (HIV), human coronaviruses (HCoV), human cytomegalovirus (HCMV), and Zika virus. Viral infections have become a serious global concern for health workers due to their uncontrollable morbidity and death rate. The recent SARS-CoV-2 (severe acute respiratory syndrome-corona virus-2) has affected approximately 631 million people with 65 lakh deaths as per the WHO dashboard, on 14 November 2022. COVID-19 or SARS-CoV-2 is a novel coronavirus that shares 79 percent of its DNA with the SARS-CoV (severe acute respiratory syndrome-corona virus) and 50% of its DNA with the Middle East Respiratory Syndrome (MERS) virus [1,2,3]. Similarly, HIV is also a major health concern worldwide. In 2021, approximately 6 lakh deaths occurred due to HIV-related causes, and approx. 1.5 million people have been diagnosed with HIV viral infection [4]. It is estimated that at the end of 2021, people living with HIV infection reached up to 38.4 million. The global prevalence of HBV infection was 296 million in 2019 with 1.5 million new infections each year. Furthermore, it is reported that 2.7 million HBV-infected people are HIV-positive [5]. Due to the social behavior of humans, several viral diseases such the HIV, HBV, and HCV continue to have a significant negative impact on some geographical areas. Certain viral infections are also involved in the etiology of complex diseases such as hyperglycemia, Alzheimer’s disease, and cancer [6,7,8]. Various synthetic approaches are being developed by researchers including triazoles [9], peptides [10], and thiadiazole [11] to combat viral infections. However, the safest method to protect the community against viral infections is vaccination. However, due to viral mutation, vaccinations may frequently lose their effectiveness. Alternatively, they have not yet been developed; for example, in the Zika virus [12]. Recent mutating variants of COVID-19 such as alpha, beta, delta, and omicron also pose great challenges to the development of novel therapeutics [13]. There are currently no authorized treatments for several viruses, and immunization is only limited to the hepatitis A virus, mumps, and varicella [14]. Moreover, increased urbanization and worldwide travel have also resulted in pandemic outbreaks and ultimately made day-to-day life riskier. Thus, there is an urgent need to explore highly potent and cost-effective medicine to control newly emerged mutating variants of viruses.
Medicinal plants and their phytochemicals continue to play an important role in treating viral infection to inhibit virus entry, multiplication, and release [15,16,17]. Moreover, phytoconstituents also act as immune boosters to protect against viral infections [18,19]. Certain reports strongly suggest using drugs of natural origin in the treatment of complex viral infections including HIV and SARS-CoV-2 [20,21]. Recently, the role of phytochemicals in anti-inflammatory activity has also got considerable scientific attention for possible intervention in viral infections including SARS-CoV-2 [22,23]. Researchers are also exploring natural products to find the potentiality against newly emerged SARS-CoV-2 viral infection [24]. Numerous in silico, in vitro, and in vivo investigations are going on to find potential candidates against viral infections. The current review comprises in silico studies (particularly SARS-CoV-2) followed by in vitro and in vivo investigations of different categories of isoquinoline alkaloids and their related moieties against viral strains. The structure–activity relationship of a few significant subcategories of isoquinoline and related alkaloids are also included in this study. Some recent reports on isoquinoline alkaloids suggest their role in complex diseases such as Alzheimer’s, parkinsonism, cancer, and viral diseases [25,26].

2. Isoquinoline and Related Alkaloids: A New Ray of Hope

Alkaloids are natural organic compounds containing nitrogen atom/atoms in their heterocyclic ring and are mainly derived from amino acids. These basic nitrogenous compounds represent numerous medicinal properties such as anti-inflammatory [27,28], analgesic [29], antibacterial [30], anti-cancer [31], and anti-fungal [32]. For decades, the antiviral activity of naturally obtained alkaloids against different strains of viruses has been well documented [33,34,35].
The second-largest category of alkaloids after indole alkaloids is isoquinoline alkaloids [36] which are mainly derived from L-tyrosine and phenylalanine. In the 19th century, morphine was the first isoquinoline alkaloid isolated from Papaver somniferum [37]. There is no structural homogeneity among the isoquinoline alkaloids [25,38,39]. Due to this reason, the classification of these alkaloids is a bit puzzling. However, based on varying levels of oxygenation, rearrangements at the intramolecular level, distribution, shared biosynthetic routes, and the presence of extra rings linked to the main moiety, isoquinoline alkaloids may be divided into 13 classes including benzyltetrahydro-isoquinoline, benzylisoquinolines, aporphines, protoberberine, benzophenanthridine, phthalideisoquinoline, morphinans, pavines, cularines, bisbenzylisoquinolines (BBI), naphythylisoquinoline, promorphinans, and ipecac alkaloids [26,39,40]. Interestingly, the primary precursor for biosynthesizing the other varieties of isoquinoline alkaloids in plants is the benzyltetrahydroisoquinoline type alkaloid (Figure 1). These alkaloids possess numerous biological activities including antifungal, antidiabetic, anticancer, anti-inflammatory, antibacterial, and antiviral. The possible mechanism by which isoquinoline-related alkaloids could show antiviral activity is illustrated in Figure 2 [26,40,41].
Recent research on viral infections has confirmed the potential role of various categories of isoquinoline and related alkaloids in the prevention of diseases such as SARS-CoV and SARS-CoV-2 [42,43,44].

3. Therapeutic Targets for SARS-CoV-2 Inhibition: In Silico Approaches

One of the main advantages of in silico drug designing process is its cost-effective nature in the research and development process. Molecular modeling and in silico methodologies have gained lots of attention nowadays. Thus, these approaches have been extremely useful in identifying targets and predicting the efficacy of new drugs in recent pandemics. In the recent SARS-CoV-2 infection cycle, angiotensin-converting enzyme (ACE2) fuses with SARS-CoV-2 spike protein to promote and facilitate the virus’s entry. Natural compounds inhibiting ACE2 will directly help in the management of COVID-19 as it restricts the entry of SARS-CoV-2 to the host cell. Similarly, inhibition of other target proteins such as spike protein, main proteases (Mpro/3CLpro), RNA-dependent RNA polymerase (RdRp), and non-structural proteins (NS) may suppress the SARS-CoV-2 infection [45]. Isoquinoline and their related alkaloids effectively bind with all these above-mentioned targets of SARS-CoV-2 enlisted in Table 1.

3.1. Angiotensin-Converting Enzyme-2 (ACE2) and Spike(S) Protein

A docking study of 17 natural products including alkaloids such as thebaine and berberine (BBR) showed satisfactory binding with ACE2 and S-protein of SARS-CoV-2. Thebaine (−100.77 kcal/mol) and BBR (−97.54 kcal/mol) showed the strongest binding affinities toward ACE2. While binding with S-protein, thebaine interacted with Gln314, Arg765, and Thr768 residues demonstrating a binding score of −104.56 and −103.58 kcal/mol respectively, whereas, BBR showed interactions with Arg765, Asn317, and Ser316 with a binding score of −99.93 kcal/mol [46]. In one study, previously reported anti-coronavirus alkaloids were assessed through in silico study and a Libdock score was calculated. A BBI alkaloid cepharanthrine (CEP) was the second-highest alkaloid exhibiting a −106.74 kcal/mol binding score after homorringtonine. Other isoquinoline alkaloids such as lycorine (−86.92), tetrandrine (−72.96), and fangchinoline (−92.66) were effectively bound with the S1 subunit of SARS-CoV-2 [42].

3.2. Main Protease (Mpro) or 3-Chemotrypsin-like Protease (3CLpro)

Apart from ACE2, the bis-benzylisoquinoline alkaloidal category shows potential binding affinity against Mpro/3CLpro, PLpro, RNA-dependent RNA polymerase (RdRp), and other NS proteins. Berbamine and oxyacanthine are also bis-benzylisoquinoline alkaloids isolated from Berberis asiatica Roxb. ex DC. Docking of these compounds using AutoDock Vina demonstrated maximum binding affinity with the binding free energy of −20.79 kcal/mol and −33.35 kcal/mol against Mpro [48]. CEP obtained from Stephania cepharantha Hayata has been discovered to have a critical role in the prevention and management of COVID-19 [56]. This phytoconstituent and its natural analogues showed satisfactory results when evaluated collectively by in silico and in vitro methods. Moreover, tetrandrine was also assessed in this research but showed slightly lower activity than CEP [57]. CEP showed strong binding interaction against 3CLpro (−8.5 kcal/mol) and TMPRSS-2 (−7.4 kcal/mol) when docked with the help of Swiss PDB viewer, PyRx, and PyMol software [52]. These studies indicate that bis-benzylisoquinoline alkaloids should be evaluated more for the prevention of SARS-CoV-2 infection.
An in silico and molecular dynamic investigation on three primary alkaloids namely BBR, choline, and tetrahydropalmatine from Tinospora cordifolia (Willd.) Miers was conducted with the help of Auto Dock and MG Tools of Auto DockVina software. BBR (−7.3 kcal/mole) had the highest affinity for the SARS-CoV-2 protein 3CLpro [47]. Natural compound noscapine mainly present in the opium poppy plant also possesses high Mpro inhibition activity exhibiting a very high docking score (−292.42 kJ/mol) [49]. Further molecular dynamic (MD) simulation studies also confirmed the stability of noscapine with Mpro [58]. The Mpro inhibition of palmatine obtained from T. cordifolia was also performed with the help of the SwissDock server. The binding score of palmatine and other natural compounds such as gingerol, and BBR with SwissDock-based docking software EADock DSS showed greater than −8 kcal/mol binding affinity toward the Mpro [50]. Molecular docking of a bis-benzylisoquinoline alkaloid oxycanthine showed a binding affinity of −10.99 kcal/mol via Auto Dock which ultimately suggested that it could be a successful candidate in the treatment or prevention of SARS-CoV-2. BBR and capsaicin also showed slightly good binding energy (−7.910 kcal/mol and −5.510 kcal/mol respectively) than caffeine [59]. A recent in silico study, using Autodock (version 4.2) on selected isoquinoline alkaloids including cephaeline, coptisine, galanthamine, glaucine, drotaverine, chelidonine, hydrastine, boldine, fumaricin stated that coptisine had the best binding affinity (−9.15 kcal/mol) toward Mpro. MD simulation study of coptisine depicted a stable complex of N3-Mpro with a binding energy of −8.17 kcal/mol [54].

3.3. RNA-Dependent RNA Polymerase (RdRp)

RdRp is an important target to cure RNA-based viral diseases such as MERS, SARS, and SARS-CoV-2 as it is responsible for viral RNA replication. In SARS-CoV-2 infection, RNA genome replication is a prominent step to spread the infection; however, RdRp inhibitors prevent this step and treat the infection. Molecular docking of 38 Chinese patent drugs including phytocompounds, flavonoids and alkaloids, was studied for a molecular docking study against RdRp, ACE2, and Mpro. The in silico study, with the help of AutoDock Vina, the findings demonstrated that morphine, codeine, indirubin, and BBR showed effective binding capacity (>−6.0 kcal/mol) against Mpro; however, their binding affinity against RdRp was much higher (>−8.1 kcal/mol) than that of Mpro [51]. CEP also showed binding with RdRp protein to inhibit viral replication [43]. A study includes 143 isoquinoline compounds for in silico screening using Molegro Virtual Docker (version 6.0, Molegro ApS, Aarhus, Denmark) against RdRp of the Zika virus. Molecular docking studies also demonstrated that Cassiarin D possessed a high binding score against RdRp (−150.7 kJ/mol) [53].

4. Antiviral Actions of Isoquinoline Related Alkaloids

Natural isoquinoline and its related alkaloids have been widely explored in recent years for their antiviral potential. For instance, BBR is a protoberberine alkaloid extracted from various species of the Berberis genus exerting antiviral activity against a diverse range of viruses including HIV, HSV, HCMV, and human papillomavirus (HPV) [27]. BBR interferes with certain pathways such as nuclear factor-kappa B (NF-κB), mitogen-activated protein kinase/extracellular-signal-regulated kinase (MEK/ERK), and mammalian target of rapamycin and the adenosine monophosphate-activated protein kinase (AMPK/mTOR) to resist viral replication, thereby, providing antiviral action. Other antiviral activities of isoquinoline and related alkaloids are summarized in Table 2.

4.1. Protoberberine Alkaloids

Protoberberine alkaloids contain a tetracycline ring system and are mainly derived from the benzyltetrahydroisoquinoline by oxidation of the phenolic group and coupling with the N-methyl group of isoquinoline [36]. Natural protoberberine alkaloids have multiple pharmacological actions such as antiseptic, sedative, stomachic, analgesic action, and antiviral action [112].
Protoberberine alkaloids show prominent effects against HSV, HBV, SARS-CoV, SARS-CoV-2, and HCMV. A protoberberine alkaloid berberine (BBR) isolated from various species of Berberis represents antiviral against HIV by inhibiting tumor necrosis factor (TNF-α) and IL-6 in macrophage of cultured mouse J774A [61]. Inhibition of the NF-κB pathway and c-Jun N-terminal kinase (JNK) phosphorylation is the main mechanism behind the anti-HSV action of BBR [60]. A complex of ZnO/BBR complex was also tested against SARS-CoV-2 by performing a plaque reduction assay and Vero E6 cell anti-viral assay. Molecular docking of this complex indicates the potential to inhibit papain-like protease (PLpro), spike protein, and spike-receptor-binding domain (RBD). Further, an in vitro study concludes that ZnO/BBR at various concentrations showed PLpro inhibition in the range of 70–98% and thus, suggested its role in SARS-CoV-2 infection. The complex inhibits S protein-ACE2 binding as compared to single BBR and ZnO-NP [64]. A review also showed the antiviral potential of BBR in different strains of coronaviruses [27].
Recently, a nanomedicine containing BBR (NIT-X) was investigated against SARS-CoV and SARS-CoV-2. This nano-oral formulation increased CD8+ cells and interferon-gamma (IFN-γ), thus showing immunomodulatory activity [62]. A study on Vero E6 cells evaluated the effectiveness of BBR and chloroquine along with synthetic drugs like lopinavir, remdesivir, and cyclosporine. IC50 values of chloroquine (1.38 μM) and BBR (10.58 μM) suggested their anti-SARS-CoV-2 activity [63]. Moreover, in vitro studies on calu-3 cells demonstrated the potential of this nanoformulation to inhibit ACE2, TMPRSS2, IL-1α, IL-8, and IL-6 [62]. Furthermore, inhibiting pro-inflammatory mediators and improving immunomodulation would be an excellent therapy against SARS-CoV-2. A recent study included BBR and obatoclox for the assessment of anti-SARS-CoV-2 activity in Vero E6 cells and nasal epithelial cells, which showed EC50 values of BBR is 9.1 µM, thus confirming the role of BBR in reducing the viral replication that justifies its promising effects against SARS-CoV-2 [65]. BBR was also found to be effective against severe post-COVID conditions such as pulmonary fibrosis and reduced inflammation during COVID-19 pneumonia [113]. The in vivo study of BBR shows activity against influenza viral pneumonia. It can cause reduction in pulmonary edema and lung index in mice and ultimately suppress lung hemorrhage [114]. BBR also showed effectiveness against chikungunya virus through decreasing viral load in wild-type C57BL6/J mice. This isoquinoline alkaloid significantly reduced joint swelling in mice [67]. Moreover, BBR also shows another anti-influenza activity by decreasing viral titers in the mouse lungs and thus eventually cause reduction in the mortality rate of mice [115].
Other protoberberine alkaloids such as dehydrocavidine, BBR, dehydroapocavidine, dehydroisoapocavidine, and dehydroisocorypalmine were derived from Corydalis saxicola Bunting plant and proved to possess anti-HBV activity. The in vitro assay performed on the 2.2.15 cell lines showed that dehydrocavidine and dehydroapocavidine represent 51% and 54% inhibition against HBeAg respectively [71,116]. Similarly, a study performed on the isoquinoline alkaloids namely saxicolalines A and N-methylnarceimicine and other alkaloids of C. saxicola showed anti-HBV activity. Along with these two isoquinoline alkaloids, dihydrochelerythzrine (IC50 < 0.05 µM) also demonstrated potent activity against HBV [71]. An anti-HBV study conducted on 18 isoquinoline alkaloids obtained from the various plant species also demonstrated potent in vitro activity against HepG2.2.15 cell lines [117]. A few alkaloids such as columbamine iodide and jatorrhizine chloride also showed activity against HIV with IC50 values of 58 and 71 µg/mL respectively [118].
BBR can inhibit Toll-like receptor-7 (TLR7), NF-κB, and myeloid differentiation primary response 88 (MyD88) in the TLR7 signaling pathway and thus restrict viral copies in mice, which is attributed to the treatment of influenza viral infection (H1N1) [69]. A study indicates that BBR can inhibit DNA polymerase and immediate-early (IE-3) proteins. BBR has also been evaluated against different strains of HCMV and exerted broad-spectrum anti- HCMV activity due to its suppressive nature against murine CMV (MCMV) showing EC50 value within the range of 1.30 to 2.70 µM [70]. Another study performed on cell lines indicated that BBR suppresses viral replication of influenza A virus by interfering the MEK/ERK pathway. Different cell lines studies of BBR including human alveolar basal epithelial cell (A549), lung epithelial type 1 (LET1), primary human airway epithelial cells (HAE), and Madin–Darby canine kidney (MDCK) represented IC50 values of 17 µM, 4 µM, 16 µM, and 52 µM respectively [68]. BBR is able to suppress IL-6, IL-1β, IL-1α, and TNF-α by inhibiting Nod-like receptor protein 3 (NLRP-3) pathway [119].
The structure–activity relationship of protoberberine alkaloids indicates that linkage between C2 and C3, a quaternary nitrogen atom and substitution with hexyl and methyl is important for anti-HIV, anti-HBV, and anti-polio activities [26,71,116,120,121] (Figure 3).

4.2. Aporphine Alkaloids

Aporphine alkaloids have been known for their antioxidant, anticancer, anthelmintic, antibacterial, antimalarial, and antiviral activity [122]. The in vitro activities of these alkaloids show potential action against HIV, HCV, and HSV viruses. A study of ixoratannin A-2 and aporphine alkaloid boldine on CEM-GXR cells showed anti-HIV activity with EC50 values of 34.4 and 50.2 µM. This in vitro study demonstrated the anti-HCV effect of ixoratannin A-2 and boldine [73]. Similarly, another category such as aporphine alkaloids also exerted anti-HIV action. For example, twelve compounds were isolated from the plant Dasymaschalon rostratum Merr. and Chun from which raymarine A, 3-methoxyoxoputerine-N-oxide, and dasymaroine B showed EC50 values in the range of 1.93 to 9.70 µM and exerted potent activity against HIV [75]. Moreover, marine alkaloids such as lamellarins alkaloids also exhibited anti-HIV action [123,124]. A natural steroidal alkaloid obtained from marine sponge didehydro-cortistatin has shown its potential against HIV-mediated inflammation [125]. An antiviral study included didehydro-cortistatin and highlighted ‘‘Block-and-lock’’ approach for the treatment of HIV. Didehydro-cortistatin A causes a reduction in HIV transcription and suppression of viral rebound in bone marrow-liver-thymus mice [126].
Two aporphine alkaloids namely magnoflorine and langinosine from the plant Magnolia grandiflora L. possessed strong HIV inhibition potential. Moreover, further cytotoxic assays were carried out using tumor cell lines to determine anti-HSV activity. The results showed anti-HSV action of M. lanuginosine methanolic extract with 76.7% inhibition and 47% inhibition against polio-virus. Both the alkaloids magnoflorine and lanuginosine represent potent cytotoxic activity also with IC50 values of 0.4 and 2.5 µg/mL [127]. Nineteen aporphine alkaloids were also investigated on Vero cells of monkey kidneys against HSV, and results indicated the selection of three alkaloids namely oliverine HCl, pachystaudine, and oxostephanine as potent inhibitors of HSV [77]. The structure–activity relationship of aporphine alkaloids is demonstrated in Figure 4.

4.3. Benzyltetrahydroisoquinoline and Benzylisoquinoline

These type II isoquinoline alkaloids contain a three-ring system in which the B ring is reduced at C1-C2 and C3-C4. These alkaloids are the main precursor for the biosynthesis of many naturally occurring alkaloids including BBI, protoberberine, protopine, phthalideisoquinoline, and many others [36,128]. The chemical structures of several isoquinoline related alkaloids are given in Figure 5. An anti-viral study of 22 traditional medicinal plants including Cimicifuga racemosa (L.) Nutt, Coptidis rhizoma, Meliae cortex, Phellodendron cortex, and Sophora subprostrata Chun and T. Chen was found to be effective against coronavirus activity [129]. Major phytocompounds such as tylophorine, reserpine, lycorine, myricetin, scutellarin, apigenin, luteolin, emetine, emodin, aescin, and betulonic acid possess antiviral action, particularly against SARS-CoV [130,131,132,133,134,135]. Lycorine and emetine showed in vivo antiviral activity against HCoV-OC43 and MERS-CoV in mice model [111].
Two isoquinoline alkaloids namely (+)-1(R)-coclaurine and (+)-1(S)-norcoclaurine from Nelumbo nucifera Gaertn showed EC50 values of 0.8 and >0.8 µg/mL respectively against HIV [72]. Synthetic tetrahydroisoquinolines alkaloids were also developed and evaluated for antiviral activity in this study. The in vitro assay showed that a non-halogenated isoquinoline compound ethyl-1-benzyl-7-methoxyisoquinoline-4-carboxylate (6d) showed potent inhibitory activity against HIV [136].
The in vitro examination of papaverine hydrochloride against HIV with the help of human T-cell line such as MT4 cells showed effective ED50 values of 5.8 µM at a dose of 30 µM [79]. A similar in vitro study conducted on papaverine using H9 cells in peripheral blood mononuclear cell (PBMC) culture indicated potent antiviral activity against HIV at a concentration of 10 µg/mL [137]. The anti-HIV activity of salsolinol, tetrahydropapaveroline, and N, N-dimethylsalsolinol was performed on Raji cells. These three isoquinoline alkaloids also showed activity against Epstein-Barr virus early antigen (EBV-EA) [138]. Dimethoxy-3,4-dihydro isoquinoline and dihydroxyisoquinolinium salts were also reported effective against HIV with IC50 values of 2.07 µg/mL and 23.6 µg/mL respectively. EC50 values of both the compounds were found to be >0.10 µg/mL [139]. Thirty-three isoquinoline alkaloids selected from Corydalis and Fumaria species represented antiviral activity against herpes simplex and para-influenza virus. The in vitro study of alkaloids against anti-HSV and anti-PI3 activity using Vero and madine-darby bovine kidney (MDBK) cell lines represents effective results [76].

4.4. Bisbenzylisoquinoline (BBI)

BBI comprises two benzyltetrahydroisoquinoline that is combined via phenolic oxidation to form bisbenzylisoquinoline [36]. However, the main precursor for the BBI alkaloids is tyrosine. These alkaloids are known to have antibacterial, antifungal, antiviral, and antimalarial activity [140].
This famous subcategory of isoquinoline alkaloids shows a potential role in viral infections. In a study, methanolic extract from Indonesian plants was investigated against HSV-1. Later the same researcher evaluated subtypes of isoquinoline alkaloids obtained from water and methanolic extract of Chinese medicinal plants by subjecting them to plaque reduction assay to investigate the anti-HSV activity. All BBI alkaloids included in the study were found to be effective and showed IC50 values in the range of 14.8–43.2 µg/mL. Results confirmed that homoarmoline, isotetrandrine, berbamine, thalrugosine, and obamegine demonstrated anti-HSV-1 and HSV-2 potential with IC50 in the range of 16.3 to 24.9 µg/mL [93,102].
CEP, a plant alkaloid found in Stephania cepharantha from the family Menispermaeceae showed antiviral activity against HIV in U1 cells through inhibition of the NF-κB pathway [94]. A study revealed another mechanism of CEP against HIV by decreasing plasma membrane fluidity [96]. CEP also inhibits pro-inflammatory mediators to prove its anti-inflammatory action [95,99]. The synthetic analog of CEP demonstrated effectiveness against HIV-1 [99]. Two alkaloids from the plant S. cepharantha roots namely aromoline and di-O-acetylsinococuline indicate HIV inhibitory activity with CC0 values of 62.5 and 15.6 µg/mL, and IC100 value of 31.3 and 7.8 µg/mL [80]. Synthetic derivatives of cepharanoline were investigated against HIV in U1 cells. Out of all the analogues of cepharanoline, five compounds were selected and showed more potent inhibition than CEP [97]. Furthermore, CEP in combination with the 8-difluoromethoxy-1-ethyl-6-fluoro-1,4-dihydro-7-[4-(2-methoxyphenyl)-1-piperazinyl]-4-oxoquinoline-3-carboxylic acid (K-12) also reduced viral infection by inhibiting NF-κB and TNF-α in U1 cells [141]. In vitro activity of CEP showed potent activity against HSV also with an IC50 value of 0.835 µg/mL [142]. The antiviral activity of some important alkaloids including tetrandrine, CEP, neferine, and hernandezine on HEK 293T cells expressing ACE2 (293T-ACE2 cells) exhibited half-maximal effective concentrations (EC50) less than 10 µM [85], indicating inhibition of viral entry against SARS-CoV-2. Another in vitro assay was performed to identify CEP and nelfinavir (NFV) as anti-viral agents. N-protein expression and RNA levels were significantly decreased by NFV and CEP which suggested them to be potent drugs for COVID-19 [101].
It has been reported that CEP possessed anti-inflammatory and immunomodulatory action by suppressing cytokine release, NF-κB, nitric oxide (NO), and cyclooxygenase (COX). A newly established cell-culture model GX-P2V (pangolin coronavirus) such as SARS-CoV-2 model is being used to identify therapeutic drugs against COVID-19. CEP has been evaluated by this novel cell culture model and showed the highest inhibition against GX-P2V with an EC50 value of 0.98 µmol/L. In addition, CEP also inhibited viral entry and multiplication in the host cell and exerted maximum antiviral activity [143]. Similar research performed on the same cell culture model along with transcriptome analysis showed the potential of CEP in inhibiting viruses through various pathways including cellular stress responses and autophagy [144]. Thus, considering the above facts, CEP could be beneficial for treating COVID-19. The in vivo study of CEP demonstrated inhibition of porcine epidemic diarrhea virus (PEDV) through reduction in viral load at a dose of 11.1 mg/kg of b.w. [145]. Another bisbenzylisoquinoline alkaloid dauricine, isolated from Menispermum dauricum in combination with clindamycin could inhibit NF-κB activation in mice to treat influenza viral H5N1 infection [146].
Two bis-benzyl isoquinoline alkaloids including tetrandrine and isotetrandrine isolated from the roots of Mahonia bealei (Fortune) Pynaert possessed anti-influenza activity at a concentration of 0.25 mg/mL [147]. Tetrandrine (30 mg/kg, i.p.) in combination with acyclovir (120 mg/kg i.p.) proved to have anti-HSV activity in the Bragg albino mouse model (BALB). This BBI alkaloid was found responsible to inhibit IL-6 in the cornea of mice and thus exerts inhibition against HSV [84]. It was observed that tetrandrine suppressed delayed-type hypersensitivity and produces inflammatory responses to the virus [148]. In addition to tetrandrine two other alkaloids including fangchinoline, and CEP were investigated against human coronavirus OC43 (HCoV-OC43) using a Medical Research Council cell strain 5 (MRC-5) fibroblast lung cell line. Results demonstrated dose-dependent inhibition of tetrandrine, fangchinoline and CEP with IC50 values of 0.33, 1.01, and 0.83 µM respectively [83].
BBI alkaloid neferine was also assessed for anti-SARS-CoV-2 activity along with other 29 compounds. huh7, HEK293/hACE2 cell lines were used to check the potential of molecules toward SARS-CoV-2. This bis-benzylisoquinoline alkaloid possessed significant inhibition (approx. 75%) against SARS-CoV-2 pseudovirus and showed an EC50 value of 0.36 µM. It blocked the viral entry by inhibiting Ca2+-mediated fusion and thus could be a promising agent to treat viral infections including newly emerged SARS-SoV-2 [88]. Cycleanine, a BBI alkaloid, possessed potent anti-HIV efficacy with an EC50 value of 1.83 µg/mL [81]. Another BBI alkaloid, fangchinoline, also showed its effectiveness against various strains of HIV, showing EC50 value in the range of 0.8 to 1.7 µM [82].
Berbamine, isolated from Berberis amurensis Rupr., demonstrated its anti-COVID-19 activity by causing a reduction in ACE2 and dipeptidyl peptidase-4 (DPP-IV) levels in the plasma membrane. A study conducted on Huh7-cells indicates that berbamine caused inhibition of Ca2+ influx, which ultimately led to the inhibition of TRPMLs, which later resulted in the intervention of virus entry [91]. Moreover, berbamine also showed inhibition of the 2-E channel with an IC50 value of 111.50 µM, thus preventing SARS-CoV-2 infection. The EC50 value of this BBI alkaloid confirmed its inhibitory action against other flaviviruses as well as Japanese encephalitis virus (JEV) (1.62 mM) and Zika virus (2.17 mM) [91]. The β-carboline and isoquinoline alkaloids such as dictamine, cinchonine, and skimmianine showed inhibition against SARS-CoV. The quinine derivative, chloroquine also showed potent inhibition toward SARS-CoV-2 [149]. Multiple compounds were studied in vitro to investigate action against SARS-CoV-2 on the human epithelial colorectal adenocarcinoma cell line (Caco-2 cells). Top hits included synthetic drugs as well as phytocompound present in plants namely lopinavir, camostat, nafamostat, mefloquine, papaverine, and cetylpridinium [78].
The BBI alkaloids category seems to have an important role in the treatment and prevention of SARS-CoV-2 and other viral infections. Some alkaloids categorized under this division were evaluated for various viral targets and produced effective results. The structure–activity relationship of this category is depicted in Figure 6.

4.5. Ipecac Alkaloids

A natural alkaloid, emetine present in the plant Psychotria ipecacuanhae (Brot.) Standl. has been reported against HIV with 80% inhibition [104]. Emetine showed inhibition of Zika virus nonstructural protein-5 (NS5) polymerase activity and ebolavirus (EBOV) with effective IC50 values. They disrupted the entry of lysosomal function and exerted anti-viral action [150]. The semi-synthetic derivative of emetine known as emetine dihydrochloride hydrate is found to be effective against SARS and MERS with EC50 values of 0.051 and 0.014 respectively [103]. A study identified four alkaloids including emetine as potential antiviral agents against echovirus (EV-1), and herpes simplex virus (HSV-2) on retinal pigment epithelial (RPE) cells [151].
The salts of ipecac alkaloids such as psychotrine dihydrogen oxalate and o-methylpsychotrine sulfate heptahydrate also exhibited potential inhibitory activity towards reverse transcriptase of HIV [152]. Some other alkaloids such as o-methylpsychotrine sulfate and papaverine also demonstrated anti-HIV activity [153]. One study investigated the effect of seven compounds including lycorine and emetine on HCoV-OC43 and MERS-CoV. Findings stated that lycorine suppressed the viral load in the BALB mice model and represented anti-HCoV-OC43 potential. However, emetine was found effective against MERS-CoV [111].

4.6. Naphythylisoquinoline Alkaloids

Michellamines alkaloids from Ancistrocladus congolensis J. were identified for their anti-viral action and found effective against HIV-1 and HIV-2 [104,105,154,155,156]. A study indicated that michellamine A2, A3, A4, and michellamine B showed IC50 values of 29.6, 15.2, 35.9, and 20.4 µM respectively, and thus exerted anti-HIV action [106]. Another study isolated korupensamine E along with michellamine B, D, E, and F from the same plant and was investigated for in vitro HIV activity. Results confirmed that all michellamine possessed antiviral activity against HIV with EC50 values in the range of 17–188 µM [157]. A bis-benzylisoquinoline alkaloid neferine and their desmethyl and didesmethyl analogues such as isoliensinine and liensinine also showed effective activity against HIV [89].

4.7. Pavine Alkaloids

A pavine alkaloid (−)-thalimonine reduces the apoptosis caused by viral proteins and possessed anti-HSV action on wild-type cells [107]. However, Cherylline inhibits DENV and Zika virus replication with an EC50 value of 8.8 µM and 20 µM [158].

4.8. Morphinan and Promorphinan Alkaloids

The skeleton of morphinan alkaloids is very similar to aporphine alkaloids except for one extra ring closure [41]. Morphine inhibits interferon (IFN) in peripheral blood mononuclear cells (PBMC) cells and thus demonstrated anti-HIV activity [159]. A study includes the anti-HSV activity of multiple morphinan alkaloids such as FK-3000, cephakicine, sinoacutine, cephasamine, cephamonine, sinomenine, 14-episinomenine, and tannagine. Among all, the two main morphinan alkaloids namely FK-3000 and cephakicine demonstrated effective results against HSV-1 with IC50 value of 7.8 and 44.4 µg/mL respectively. However, in addition, FK-3000 also showed its notable activity against HSV-1 TK- and HSV-2 demonstrating 9.9 and 8.7 IC50 values respectively [93].

4.9. Benzophenanthridines

The higher plants mainly consist of benzophenanthridine isoquinoline compounds exhibit a wide range of pharmacological activities. A study investigated 2000 synthetic and natural compound which include sanguinarine as a HIV-protease inhibitor with IC50 value of 4.3 µg/mL [160]. An anti-HIV-1 and HIV-2 reverse transcriptase (RT) study included fangronine chloride and nitidine chloride to determine the antiviral potential. The IC50 value of fangronine chloride (8.5 µg/mL) and nitidine chloride (7.4 µg/mL) suggest the efficacy of these two compounds against HIV-RT. Similar results were shown in the case of HIV-2 RT where fangronine chloride displayed a 9.5 µg/mL IC50 value, whereas, nitidine chloride displayed a 7.1 µg/mL IC50 value [109]. Anti-HCV and PI-3 (para-influenza) activity of norsanguinarine displayed moderate action against HCV and PI-3 with minimum inhibitory concentration (MIC) range from 16 to 32 µg/mL [76].

5. Inflammation Inhibition

Anti-inflammatory phytochemicals might be viable therapeutic candidates against various viruses, including SARS-CoV2, as they have direct antiviral effects and can alleviate the status of inflammatory diseases [22,23]. Moreover, it is evident from recent reports that severe COVID-19 patients cause elevation of inflammatory pro-markers which eventually lead to multiorgan failure [161]. Thus, researchers are now focusing on natural products such as medicinal plants that possess anti-inflammatory as well as anti-viral potential. Tetrandrine from the category of bis-benzylisoquinoline alkaloids of isoquinoline alkaloids has been reported for immunomodulation and anti-inflammatory activities. It has anti-inflammatory action against croton oil-induced ear edema in mice and demonstrates 95% inhibition at 12.5 µM [162]. This isoquinoline alkaloid also showed inhibition against proinflammatory mediators of cytokines, iNOS, and cyclooxygenase (COX-2) in human monocytic cells [163]. In vitro lipopolysaccharide assay suggested the potential of tetrandrine to inhibit TNF-α, IL-6, and NO release in lipopolysaccharide (LPS)-induced microglial activation [164]. Similarly, in vivo studies performed on BBR, which is another isoquinoline alkaloid at the dose of 50 mg/kg, indicated a reduction of IL-6, IL-10, IL-1β, and IFN-γ caused by thioacetamide injection [165]. Noscapine is also found to be responsible for showing inhibitory action against cytokine storm in COVID-19 [166]. Recently, an in vitro study indicated that litcubanine, an isoquinoline alkaloid greatly reduced the activation of inflammatory macrophages by LPS via the NF-κB pathway, which would reduce the levels of inflammatory mediators such iNOS, TNF-α, and IL-1β [167].

6. Clinical Findings

The efficacy of isoquinoline and related alkaloids in various scientific reports including in silico, in vitro, and in vivo evaluation justified a slew of human clinical trials to assess the safety, pharmacokinetics, and pharmacodynamic effectiveness in a variety of viral pathologies. Few recent clinical trials also justify the role of isoquinoline and related alkaloids in viral infections. For example, clinicians investigated the effects of BBR (NCT04479202) on intestinal function, serum concentrations of inflammatory biomarkers, and organ function in severe SARS-CoV-2-infected patients in a prospective randomized controlled clinical trial supported by the Chinese medical association. An herbal formulation of COVIDEX also contains BBR (NCT05228626) targeting SARS-CoV-2 in a clinical trial sponsored by Makerere University in Uganda. Tetrandrine (NCT04308317) in combination with standard drug improves prognosis and reduces the incidence of pulmonary fibrosis during rehabilitation in SARS-CoV-2 patients. Even though there are only a few clinical trials (enlisted in Table 3) in the literature that include the use of isoquinoline and related alkaloids with various viral infections, these moieties are expected to have a greater impact on viral load, making these molecules the alternative option for improving the limited clinical efficacy and expanding the field of use to other viral pathologies.

7. Conclusions and Future Perspective

The emerging variants of powerful viruses have made the task of clinical practitioners and scientists more challenging to find an effective solution against viral infections. However, naturally occurring isoquinoline and related alkaloids could be an alternative option for treating these rapidly mutating viruses. From research findings, it can be concluded that these alkaloids exert their antiviral action majorly by interfering with the signaling pathways such as NF-κB, and MEK/ERK that eventually restricts the entry and replication of the virus. Furthermore, inhibition of Ca2+-mediated fusion and SandN protein expression could also facilitate the antiviral actions. Isoquinoline and their related alkaloids also resist pro-inflammatory markers such as IL-6, IL-10, and IL-1β and exert anti-inflammatory action. Additionally, an increase in CD8+ cells and IFN-γ production by some of the isoquinoline alkaloids represent immunomodulatory effects.
The structure–activity relationship has clearly shown that the quaternary nitrogen atom is essential for the antiviral activity of protoberberine and benzophenanthridine alkaloids. However, substitution with a methyl group at benzyltetrahydroisoquinoline and BBI moieties potentiate the antiviral action. From the in vitro and in vivo studies, it is concluded that isoquinoline and related alkaloids including berbamine, CEP, tetrandrine, neferine and lycorine from the BBI category of isoquinoline alkaloids represent broad-spectrum activities against HSV, HIV, SARS-CoV, and SARS-CoV-2 infection. Thus, BBIalkaloids should be further structurally explored for the treatment of newly emerged viral strains.

Author Contributions

Conceptualization: N.S. and M.D.; original draft preparation, writing and editing: D.S. and N.M.; review of structure–activity relationships: P.C.S.; draft revision and editing: M.D., N.S., and V.K.T.; final approval: S.K.P., M.M.R., and V.K.T. All authors have read and agreed to the published version of the manuscript.

Funding

This study received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We thank the Department of Science and Technology, Government of India. for providing DST-Inspire fellowship to Divya Sharma for conducting her doctoral research work.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Caldaria, A.; Conforti, C.; Di Meo, N.; Dianzani, C.; Jafferany, M.; Lotti, T.; Zalaudek, I.; Giuffrida, R. COVID-19 and SARS: Differences and similarities. Dermatol. Ther. 2020, 33, e13395. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. 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. Infez Med 2020, 28, 174–184. [Google Scholar] [PubMed]
  3. Chen, Z.; Boon, S.S.; Wang, M.H.; Chan, R.W.Y.; Chan, P.K.S. Genomic and evolutionary comparison between SARS-CoV-2 and other human coronaviruses. J. Virol. Methods 2021, 289, 114032. [Google Scholar] [CrossRef] [PubMed]
  4. Available online: https://www.who.int/news-room/fact-sheets/detail/hiv-aids#:~:text=HIV%20continues%20to%20be%20a,no%20cure%20for%20HIV%20infection (accessed on 9 November 2022).
  5. Available online: https://www.who.int/news-room/fact-sheets/detail/hepatitis-b#:~:text=HBV%2DHIV%20coinfection&text=Conversely%2C%20the%20global%20prevalence%20of,%2Dinfected%20persons%20is%207.4%25 (accessed on 24 June 2022).
  6. Ball, M.J.; Lukiw, W.J.; Kammerman, E.M.; Hill, J.M. Intracerebral propagation of Alzheimer’s disease: Strengthening evidence of a herpes simplex virus etiology. Alzheimer’s Dement. 2013, 9, 169–175. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Hober, D.; Sane, F.; Jaidane, H.; Riedweg, K.; Goffard, A.; Desailloud, R. Immunology in the clinic review series; focus on type 1 diabetes and viruses: Role of antibodies enhancing the infection with Coxsackievirus-B in the pathogenesis of type 1 diabetes. Clin. Exp. Immunol. 2012, 168, 47–51. [Google Scholar] [CrossRef]
  8. Morgan, R.L.; Baack, B.; Smith, B.D.; Yartel, A.; Pitasi, M.; Falck-Ytter, Y. Eradication of hepatitis C virus infection and the development of hepatocellular carcinoma: A meta-analysis of observational studies. Ann. Intern. Med. 2013, 158 Pt 1, 329–337. [Google Scholar] [CrossRef] [Green Version]
  9. Kharb, R.; Shahar Yar, M.; Sharma, P.C. Recent advances and future perspectives of triazole analogs as promising antiviral agents. Mini Rev. Med. Chem. 2011, 11, 84–96. [Google Scholar] [CrossRef]
  10. Parra, A.L.; Bezerra, L.P.; Shawar, D.E.; Neto, N.A.; Mesquita, F.P.; Silva, G.O.D.; Souza, P.F. Synthetic antiviral peptides: A new way to develop targeted antiviral drugs. Future Virol. 2022, 17, 577–591. [Google Scholar] [CrossRef]
  11. Serban, G. Synthetic Compounds with 2-Amino-1,3,4-Thiadiazole Moiety Against Viral Infections. Molecules 2020, 25, 942. [Google Scholar] [CrossRef] [Green Version]
  12. Wang, Y.; Ling, L.; Zhang, Z.; Marin-Lopez, A. Current Advances in Zika Vaccine Development. Vaccines 2022, 10, 1816. [Google Scholar] [CrossRef]
  13. Burki, T. The origin of SARS-CoV-2 variants of concern. Lancet Infect. Dis. 2022, 22, 174–175. [Google Scholar] [CrossRef] [PubMed]
  14. Nováková, L.; Pavlík, J.; Chrenková, L.; Martinec, O.; Červený, L. Current antiviral drugs and their analysis in biological materials–Part II: Antivirals against hepatitis and HIV viruses. J. Pharm. Biomed. Anal. 2018, 147, 378–399. [Google Scholar] [CrossRef] [PubMed]
  15. Ganjhu, R.K.; Mudgal, P.P.; Maity, H.; Dowarha, D.; Devadiga, S.; Nag, S.; Arunkumar, G. Herbal plants and plant preparations as remedial approach for viral diseases. Virus Dis. 2015, 26, 225–236. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Kitazato, K.; Wang, Y.; Kobayashi, N. Viral infectious disease and natural products with antiviral activity. Drug Discov. 2007, 1, 14–22. [Google Scholar]
  17. Adhikari, B.; Marasini, B.P.; Rayamajhee, B.; Bhattarai, B.R.; Lamichhane, G.; Khadayat, K.; Adhikari, A.; Khanal, S.; Parajuli, N. Potential roles of medicinal plants for the treatment of viral diseases focusing on COVID-19: A review. Phytother. Res. 2021, 35, 1298–1312. [Google Scholar] [CrossRef]
  18. Wang, K.; Conlon, M.; Ren, W.; Chen, B.B.; Bączek, T. Natural Products as Targeted Modulators of the Immune System. J. Immunol. Res. 2018, 2018, 7862782. [Google Scholar] [CrossRef]
  19. Khanna, K.; Kohli, S.K.; Kaur, R.; Bhardwaj, A.; Bhardwaj, V.; Ohri, P.; Sharma, A.; Ahmad, A.; Bhardwaj, R.; Ahmad, P. Herbal immune-boosters: Substantial warriors of pandemic Covid-19 battle. Phytomed. Int. J. Phytother. Phytopharm. 2021, 85, 153361. [Google Scholar] [CrossRef]
  20. Sharma, P.C.; Sharma, O.; Vasudeva, N.; Mishra, D.; Singh, S. Anti-HIV substances of natural origin–An updated account. Nat. Prod. Radiance 2006, 70–78. [Google Scholar]
  21. Patel, B.; Sharma, S.; Nair, N.; Majeed, J.; Goyal, R.K.; Dhobi, M. Therapeutic opportunities of edible antiviral plants for COVID-19. Mol Cell Biochem 2021, 476, 2345–2364. [Google Scholar] [CrossRef]
  22. Mrityunjaya, M.; Pavithra, V.; Neelam, R.; Janhavi, P.; Halami, P.; Ravindra, P. Immune-boosting, antioxidant and anti-inflammatory food supplements targeting pathogenesis of COVID-19. Front. Immunol. 2020, 2337. [Google Scholar] [CrossRef]
  23. Zhang, D.-H.; Wu, K.-L.; Zhang, X.; Deng, S.-Q.; Peng, B. In silico screening of Chinese herbal medicines with the potential to directly inhibit 2019 novel coronavirus. J. Integr. Med. 2020, 18, 152–158. [Google Scholar] [CrossRef] [PubMed]
  24. Omrani, M.; Keshavarz, M.; Nejad Ebrahimi, S.; Mehrabi, M.; Mcgaw, L.J.; Ali Abdalla, M.; Mehrbod, P. Potential Natural Products Against Respiratory Viruses: A Perspective to Develop Anti-COVID-19 Medicines. Front. Pharmacol. 2021, 11. [Google Scholar] [CrossRef] [PubMed]
  25. Erika, P.; Avila M, M.C.; Muñoz, D.R.; Cuca S, L.E. Natural isoquinoline alkaloids: Pharmacological features and multi-target potential for complex diseases. Pharmacol. Res. 2022, 177, 106126. [Google Scholar] [CrossRef]
  26. Qing, Z.-X.; Yang, P.; Tang, Q.; Cheng, P.; Liu, X.-B.; Zheng, Y.-J.; Liu, Y.-S.; Zeng, J.-G. Isoquinoline Alkaloids and Their Antiviral, Antibacterial, and Antifungal Activities and Structure-activity Relationship. Curr. Org. Chem. 2017, 21, 1920–1934. [Google Scholar] [CrossRef]
  27. Warowicka, A.; Nawrot, R.; Goździcka-Józefiak, A. Antiviral activity of berberine. Arch. Virol. 2020, 165, 1935–1945. [Google Scholar] [CrossRef]
  28. Souto, A.L.; Tavares, J.F.; Da Silva, M.S.; Diniz, M.D.F.F.M.; De Athayde-Filho, P.F.; Barbosa Filho, J.M. Anti-inflammatory activity of alkaloids: An update from 2000 to 2010. Molecules 2011, 16, 8515–8534. [Google Scholar] [CrossRef]
  29. Bribi, N. Pharmacological activity of Alkaloids: A Review. Asian J. Bot. 2018, 1–6. [Google Scholar] [CrossRef]
  30. Yan, Y.; Li, X.; Zhang, C.; Lv, L.; Gao, B.; Li, M. Research Progress on Antibacterial Activities and Mechanisms of Natural Alkaloids: A Review. Antibiotics 2021, 10, 318. [Google Scholar] [CrossRef]
  31. Mondal, A.; Gandhi, A.; Fimognari, C.; Atanasov, A.G.; Bishayee, A. Alkaloids for cancer prevention and therapy: Current progress and future perspectives. Eur. J. Pharm. 2019, 858, 172472. [Google Scholar] [CrossRef]
  32. Khan, H.; Mubarak, M.S.; Amin, S. Antifungal Potential of Alkaloids As An Emerging Therapeutic Target. Curr Drug Targets 2017, 18, 1825–1835. [Google Scholar] [CrossRef]
  33. Farnsworth, N.R.; Svoboda, G.H.; Blomster, R.N. Antiviral Activity of Selected Catharanthus Alkaloids. J. Pharm. Sci. 1968, 57, 2174–2175. [Google Scholar] [CrossRef] [PubMed]
  34. Renard-Nozaki, J.; Kim, T.; Imakura, Y.; Kihara, M.; Kobayashi, S. Effect of alkaloids isolated from Amaryllidaceae on herpes simplex virus. Res. Virol. 1989, 140, 115–128. [Google Scholar] [CrossRef] [PubMed]
  35. Topcu, G.S.H.; Toraman, G.O.A.; Altan, V.M. Natural alkaloids as potential anti-coronavirus compounds. (Special Issue: Therapeutic options and potential vaccine studies against COVID-19. Bezmialem Sci. 2020, 8, 131–139. [Google Scholar] [CrossRef]
  36. Ramawat, K.G. Biodiversity and Chemotaxonomy; Sustainable Development and Biodiversity; Ramawat, K.G., Ed.; Springer: Berlin/Heidelberg, Germany, 2019; Volume 24. [Google Scholar]
  37. Santoro, D.; Bellinghieri, G.; Savica, V. Development of the concept of pain in history. J. Nephrol. 2011, 24 (Suppl. 17), S133–S136. [Google Scholar] [CrossRef] [PubMed]
  38. Kukula-Koch, W.A.; Widelski, J. Chapter 9—Alkaloids, Pharmacognosy; Badal, S., Delgoda, R., Eds.; Academic Press: Boston, MA, USA, 2017; pp. 163–198. [Google Scholar]
  39. Qing, Z.; Xu, Y.; Yu, L.; Liu, J.; Huang, X.; Tang, Z.; Cheng, P.; Zeng, J. Investigation of fragmentation behaviours of isoquinoline alkaloids by mass spectrometry combined with computational chemistry. Sci. Rep. 2020, 10, 733. [Google Scholar] [CrossRef] [Green Version]
  40. Qing, Z.-X.; Huang, J.-L.; Yang, X.-Y.; Liu, J.-H.; Cao, H.-L.; Xiang, F.; Cheng, P.; Zeng, J.-G. Anticancer and Reversing Multidrug Resistance Activities of Natural Isoquinoline Alkaloids and their Structure-activity Relationship. Curr. Med. Chem. 2018, 25, 5088–5114. [Google Scholar] [CrossRef]
  41. Shang, X.-F.; Yang, C.-J.; Morris-Natschke, S.L.; Li, J.-C.; Yin, X.-D.; Liu, Y.-Q.; Guo, X.; Peng, J.-W.; Goto, M.; Zhang, J.-Y.; et al. Biologically active isoquinoline alkaloids covering 2014–2018. Med. Res. Rev. 2020, 40, 2212–2289. [Google Scholar] [CrossRef]
  42. Fielding, B.C.; Da Silva Maia Bezerra Filho, C.; Ismail, N.S.M.; Sousa, D.P.D. Alkaloids: Therapeutic Potential against Human Coronaviruses. Molecules 2020, 25, 5496. [Google Scholar] [CrossRef]
  43. Majnooni, M.B.; Fakhri, S.; Bahrami, G.; Naseri, M.; Farzaei, M.H.; Echeverría, J. Alkaloids as Potential Phytochemicals against SARS-CoV-2: Approaches to the Associated Pivotal Mechanisms. Evid. -Based Complement. Altern. Med. 2021, 2021, 6632623. [Google Scholar] [CrossRef]
  44. Abookleesh, F.L.; Al-Anzi, B.S.; Ullah, A. Potential Antiviral Action of Alkaloids. Molecules 2022, 27, 903. [Google Scholar] [CrossRef]
  45. Goyal, R.K.; Majeed, J.; Tonk, R.; Dhobi, M.; Patel, B.; Sharma, K.; Apparsundaram, S. Current targets and drug candidates for prevention and treatment of SARS-CoV-2 (COVID-19) infection. Rev. Cardiovasc. Med. 2020, 21, 365–384. [Google Scholar] [CrossRef] [PubMed]
  46. Maurya, V.K.; Kumar, S.; Prasad, A.K.; Bhatt, M.L.; Saxena, S.K. Structure-based drug designing for potential antiviral activity of selected natural products from Ayurveda against SARS-CoV-2 spike glycoprotein and its cellular receptor. Virusdisease 2020, 31, 179–193. [Google Scholar] [CrossRef] [PubMed]
  47. Chowdhury, P. In silico investigation of phytoconstituents from Indian medicinal herb ‘Tinospora cordifolia (giloy)’against SARS-CoV-2 (COVID-19) by molecular dynamics approach. J. Biomol. Struct. Dyn. 2021, 39, 6792–6809. [Google Scholar] [CrossRef] [PubMed]
  48. Joshi, T.; Bhat, S.; Pundir, H.; Chandra, S. Identification of Berbamine, Oxyacanthine and Rutin from Berberis asiatica as anti-SARS-CoV-2 compounds: An in silico study. J. Mol. Graph. Model. 2021, 109, 108028. [Google Scholar] [CrossRef] [PubMed]
  49. Kumar, N.; Sood, D.; Van Der Spek, P.J.; Sharma, H.S.; Chandra, R. Molecular binding mechanism and pharmacology comparative analysis of noscapine for repurposing against SARS-CoV-2 protease. J. Proteome Res. 2020, 19, 4678–4689. [Google Scholar] [CrossRef] [PubMed]
  50. Jadhav, V.; Ingle, S.; Ahmed, R. Inhibitory activity of palmatine on main protease complex (mpro) of SARS-CoV-2. Rom. J. Biophys. 2021, 31, 27–40. [Google Scholar] [CrossRef]
  51. Yan, J.; Shen, X.; Cao, Y.; Zhang, J.; Wang, Y.; Cheng, Y. Discovery of Anti-2019-nCoV Agents from 38 Chinese Patent Drugs toward Respiratory Diseases via Docking Screening. Preprints 2020. [Google Scholar] [CrossRef] [Green Version]
  52. Jain, D.; Hossain, R.; Khan, R.; Dey, D.; Toma, T.R.; Islam, M.; Pracheta, D.; Khalid, R.; Hakeem, K. Computer-aided Evaluation of Anti-SARS-CoV-2 (3-chymotrypsin-like Protease and Transmembrane Protease Serine 2 Inhibitors) Activity of Cepharanthine: An In silico Approach. Biointerface Res. Appl. Chem. 2021, 12, 768–780. [Google Scholar]
  53. Byler, K.G.; Ogungbe, I.V.; Setzer, W.N. Setzer In-silico screening for anti-Zika virus phytochemicals. J. Mol. Graph. Model. 2016, 69, 78–91. [Google Scholar] [CrossRef]
  54. Sadeghi, M.; Miroliaei, M. Inhibitory effects of selected isoquinoline alkaloids against main protease (Mpro) of SARS-CoV-2, in silico study. Silico Pharmacol. 2022, 10, 1–8. [Google Scholar] [CrossRef]
  55. Garg, S.; Roy, A. In silico analysis of selected alkaloids against main protease (Mpro) of SARS-CoV-2. Chem.-Biol. Interact. 2020, 332, 109309. [Google Scholar] [CrossRef] [PubMed]
  56. Rogosnitzky, M.; Okediji, P.; Koman, I. Cepharanthine: A review of the antiviral potential of a Japanese-approved alopecia drug in COVID-19. Pharmacol. Rep. 2020, 72, 1509–1516. [Google Scholar] [CrossRef] [PubMed]
  57. Hijikata, A.; Shionyu-Mitsuyama, C.; Nakae, S.; Shionyu, M.; Ota, M.; Kanaya, S.; Hirokawa, T.; Nakajima, S.; Watashi, K.; Shirai, T. Evaluating cepharanthine analogues as natural drugs against SARS-CoV-2. FEBS Open Bio 2022, 12, 285–294. [Google Scholar] [CrossRef] [PubMed]
  58. Kumar, D.; Kumari, K.; Jayaraj, A.; Kumar, V.; Kumar, R.V.; Dass, S.K.; Chandra, R.; Singh, P. Understanding the binding affinity of noscapines with protease of SARS-CoV-2 for COVID-19 using MD simulations at different temperatures. J. Biomol. Struct. Dyn. 2021, 39, 2659–2672. [Google Scholar] [CrossRef]
  59. Agrawal, A.; Jain, N.K.; Kumar, N.; Kulkarni, G.T. Molecular docking study to identify a potential inhibitor of covid-19 main protease enzyme: An in-silico approach. ChemRxiv 2020. Cambridge: Cambridge Open Engage; This content is a preprint and has not been peer-reviewed. [Google Scholar] [CrossRef]
  60. Song, S.; Qiu, M.; Chu, Y.; Chen, D.; Wang, X.; Su, A.; Wu, Z. Downregulation of cellular c-Jun N-terminal protein kinase and NF-κB activation by berberine may result in inhibition of herpes simplex virus replication. Antimicrob. Agents Chemother. 2014, 58, 5068–5078. [Google Scholar] [CrossRef] [Green Version]
  61. Zha, W.; Liang, G.; Xiao, J.; Studer, E.J.; Hylemon, P.B.; Pandak, W.M., Jr.; Wang, G.; Li, X.; Zhou, H. Berberine Inhibits HIV Protease Inhibitor-Induced Inflammatory Response by Modulating ER Stress Signaling Pathways in Murine Macrophages. PLoS ONE 2010, 5, e9069. [Google Scholar] [CrossRef] [Green Version]
  62. Wang, Z.Z.; Li, K.; Maskey, A.R.; Huang, W.; Toutov, A.A.; Yang, N.; Srivastava, K.; Geliebter, J.; Tiwari, R.; Miao, M. A small molecule compound berberine as an orally active therapeutic candidate against COVID-19 and SARS: A computational and mechanistic study. FASEB J. 2021, 35, e21360. [Google Scholar] [CrossRef]
  63. Pizzorno, A.; Padey, B.; Dubois, J.; Julien, T.; Traversier, A.; Dulière, V.; Brun, P.; Lina, B.; Rosa-Calatrava, M.; Terrier, O. In vitro evaluation of antiviral activity of single and combined repurposable drugs against SARS-CoV-2. Antivir. Res. 2020, 181, 104878. [Google Scholar] [CrossRef]
  64. Ghareeb, D.A.; Saleh, S.R.; Seadawy, M.G.; Nofal, M.S.; Abdulmalek, S.A.; Hassan, S.F.; Khedr, S.M.; Abdelwahab, M.G.; Sobhy, A.A.; Yassin, A.M. Nanoparticles of ZnO/Berberine complex contract COVID-19 and respiratory co-bacterial infection in addition to elimination of hydroxychloroquine toxicity. J. Pharm. Investig. 2021, 51, 735–757. [Google Scholar] [CrossRef]
  65. Varghese, F.S.; Van Woudenbergh, E.; Overheul, G.J.; Eleveld, M.J.; Kurver, L.; Van Heerbeek, N.; Van Laarhoven, A.; Miesen, P.; Den Hartog, G.; De Jonge, M.I. Berberine and obatoclax inhibit SARS-CoV-2 replication in primary human nasal epithelial cells in vitro. Viruses 2021, 13, 282. [Google Scholar] [CrossRef] [PubMed]
  66. Cecil, C.E.; Davis, J.M.; Cech, N.B.; Laster, S.M. Inhibition of H1N1 influenza A virus growth and induction of inflammatory mediators by the isoquinoline alkaloid berberine and extracts of goldenseal (Hydrastis canadensis). Int. Immunopharmacol. 2011, 11, 1706–1714. [Google Scholar] [CrossRef] [PubMed]
  67. Varghese, F.S.; Thaa, B.; Amrun, S.N.; Simarmata, D.; Rausalu, K.; Nyman, T.A.; Merits, A.; Mcinerney, G.M.; Ng, L.F.P.; Ahola, T. The Antiviral Alkaloid Berberine Reduces Chikungunya Virus-Induced Mitogen-Activated Protein Kinase Signaling. J. Virol. 2016, 90, 9743–9757. [Google Scholar] [CrossRef] [PubMed]
  68. Botwina, P.; Owczarek, K.; Rajfur, Z.; Ochman, M.; Urlik, M.; Nowakowska, M.; Szczubiałka, K.; Pyrc, K. Berberine hampers influenza a replication through inhibition of MAPK/ERK pathway. Viruses 2020, 12, 344. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Yan, Y.Q.; Fu, Y.J.; Wu, S.; Qin, H.Q.; Zhen, X.; Song, B.M.; Weng, Y.S.; Wang, P.C.; Chen, X.Y.; Jiang, Z.Y. Anti-influenza activity of berberine improves prognosis by reducing viral replication in mice. Phytother. Res. 2018, 32, 2560–2567. [Google Scholar] [CrossRef]
  70. Luganini, A.; Mercorelli, B.; Messa, L.; Palù, G.; Gribaudo, G.; Loregian, A. The isoquinoline alkaloid berberine inhibits human cytomegalovirus replication by interfering with the viral Immediate Early-2 (IE2) protein transactivating activity. Antivir. Res. 2019, 164, 52–60. [Google Scholar] [CrossRef]
  71. Wu, Y.R.; Ma, Y.B.; Zhao, Y.X.; Yao, S.Y.; Zhou, J.; Zhou, Y.; Chen, J.J. Two new quaternary alkaloids and anti-hepatitis B virus active constituents from Corydalis saxicola. Planta Med. 2007, 73, 787–791. [Google Scholar] [CrossRef] [Green Version]
  72. Kashiwada, Y.; Aoshima, A.; Ikeshiro, Y.; Chen, Y.-P.; Furukawa, H.; Itoigawa, M.; Fujioka, T.; Mihashi, K.; Cosentino, L.M.; Morris-Natschke, S.L. Anti-HIV benzylisoquinoline alkaloids and flavonoids from the leaves of Nelumbo nucifera, and structure–activity correlations with related alkaloids. Bioorganic Med. Chem. 2005, 13, 443–448. [Google Scholar] [CrossRef]
  73. Tietjen, I.; Ntie-Kang, F.; Mwimanzi, P.; Onguéné, P.A.; Scull, M.A.; Idowu, T.O.; Ogundaini, A.O.; Meva’a, L.M.; Abegaz, B.M.; Rice, C.M. Screening of the Pan-African natural product library identifies ixoratannin A-2 and boldine as novel HIV-1 inhibitors. PLoS ONE 2015, 10, e0121099. [Google Scholar] [CrossRef] [Green Version]
  74. Boustie, J.; Stigliani, J.-L.; Montanha, J.; Amoros, M.; Payard, M.; Girre, L. Antipoliovirus structure− activity relationships of some aporphine alkaloids. J. Nat. Prod. 1998, 61, 480–484. [Google Scholar] [CrossRef]
  75. Yu, Z.; Han, C.; Song, X.; Chen, G.; Chen, J. Bioactive aporphine alkaloids from the stems of Dasymaschalon rostratum. Bioorganic Chem. 2019, 90, 103069. [Google Scholar] [CrossRef] [PubMed]
  76. Orhana, I.; Ozçelik, B.; Karaoğlu, T.; Sener, B. Antiviral and antimicrobial profiles of selected isoquinoline alkaloids from Fumaria and Corydalis species. Z. Nat. C J. Biosci. 2007, 62, 19–26. [Google Scholar] [CrossRef] [PubMed]
  77. Montanha, J.; Amoros, M.; Boustie, J.; Girre, L. Anti-herpes virus activity of aporphine alkaloids. Planta Med. 1995, 61, 419–424. [Google Scholar] [CrossRef] [PubMed]
  78. Ellinger, B.; Bojkova, D.; Zaliani, A.; Cinatl, J.; Claussen, C.; Westhaus, S.; Keminer, O.; Reinshagen, J.; Kuzikov, M.; Wolf, M. A SARS-CoV-2 cytopathicity dataset generated by high-content screening of a large drug repurposing collection. Sci. Data 2021, 8, 1–10. [Google Scholar] [CrossRef]
  79. Nokta, M.; Albrecht, T.; Pollard, R. Papaverine hydrochloride: Effects on HIV replication and T-lymphocyte cell function. Immunopharmacology 1993, 26, 181–185. [Google Scholar] [CrossRef]
  80. Ma, C.M.; Nakamura, N.; Miyashiro, H.; Hattori, M.; Komatsu, K.; Kawahata, T.; Otake, T. Screening of Chinese and Mongolian herbal drugs for anti-human immunodeficiency virus type 1 (HIV-1) activity. Phytother. Res. 2002, 16, 186–189. [Google Scholar] [CrossRef]
  81. Otshudi, A.L.; Apers, S.; Pieters, L.; Claeys, M.; Pannecouque, C.; De Clercq, E.; Van Zeebroeck, A.; Lauwers, S.; Frederich, M.; Foriers, A. Biologically active bisbenzylisoquinoline alkaloids from the root bark of Epinetrum villosum. J. Ethnopharmacol. 2005, 102, 89–94. [Google Scholar] [CrossRef]
  82. Wan, Z.; Lu, Y.; Liao, Q.; Wu, Y.; Chen, X. Fangchinoline inhibits human immunodeficiency virus type 1 replication by interfering with gp160 proteolytic processing. PLoS ONE 2012, 7, e39225. [Google Scholar] [CrossRef] [Green Version]
  83. Kim, D.E.; Min, J.S.; Jang, M.S.; Lee, J.Y.; Shin, Y.S.; Song, J.H.; Kim, H.R.; Kim, S.; Jin, Y.H.; Kwon, S. Natural Bis-Benzylisoquinoline Alkaloids-Tetrandrine, Fangchinoline, and Cepharanthine, Inhibit Human Coronavirus OC43 Infection of MRC-5 Human Lung Cells. Biomolecules 2019, 9, 696. [Google Scholar] [CrossRef] [Green Version]
  84. Hu, M.; Dutt, J.; Arrunategui-Correa, V.; Baltatzis, S.; Foster, C.S. Cytokine mRNA in BALB/c mouse corneas infected with herpes simplex virus. Eye 1999, 13, 309–313. [Google Scholar] [CrossRef] [Green Version]
  85. He, C.-L.; Huang, L.-Y.; Wang, K.; Gu, C.-J.; Hu, J.; Zhang, G.-J.; Xu, W.; Xie, Y.-H.; Tang, N.; Huang, A.-L. Identification of bis-benzylisoquinoline alkaloids as SARS-CoV-2 entry inhibitors from a library of natural products. Signal Transduct. Target. Ther. 2021, 6, 131. [Google Scholar] [CrossRef] [PubMed]
  86. Heister, P.M.; Poston, R.N. Pharmacological hypothesis: TPC2 antagonist tetrandrine as a potential therapeutic agent for COVID-19. Pharmacol. Res. Perspect. 2020, 8, e00653. [Google Scholar] [CrossRef] [PubMed]
  87. Glebov, O.O. Understanding SARS-CoV-2 endocytosis for COVID-19 drug repurposing. FEBS J. 2020, 287, 3664–3671. [Google Scholar] [CrossRef] [PubMed]
  88. Yang, Y.; Yang, P.; Huang, C.; Wu, Y.; Zhou, Z.; Wang, X.; Wang, S. Inhibitory effect on SARS-CoV-2 infection of neferine by blocking Ca2+-dependent membrane fusion. J. Med. Virol. 2021, 93, 5825–5832. [Google Scholar] [CrossRef] [PubMed]
  89. Zhou, H.; Jiang, H.; Yao, T.; Zeng, S. Fragmentation study on the phenolic alkaloid neferine and its analogues with anti-HIV activities by electrospray ionization tandem mass spectrometry with hydrogen/deuterium exchange and its application for rapid identification of in vitro microsomal metabolites of neferine. Rapid. Commun. Mass Spectrom. 2007, 21, 2120–2128. [Google Scholar]
  90. Huang, L.; Li, H.; Ye, Z.; Xu, Q.; Fu, Q.; Sun, W.; Qi, W.; Yue, J. Berbamine inhibits Japanese encephalitis virus (JEV) infection by compromising TPRMLs-mediated endolysosomal trafficking of low-density lipoprotein receptor (LDLR). Emerg. Microbes Infect. 2021, 10, 1257–1271. [Google Scholar] [CrossRef]
  91. Huang, L.; Li, H.; Yuen, T.T.-T.; Ye, Z.; Fu, Q.; Sun, W.; Xu, Q.; Yang, Y.; Chan, J.F.-W.; Zhang, G. Berbamine inhibits the infection of SARS-CoV-2 and flaviviruses by compromising TPRMLs-mediated endolysosomal trafficking of viral receptors. Signal Transduct. Target. Ther. 2020, 6, 1–3. [Google Scholar] [CrossRef]
  92. Xia, B.; Shen, X.; He, Y.; Pan, X.; Liu, F.-L.; Wang, Y.; Yang, F.; Fang, S.; Wu, Y.; Duan, Z. SARS-CoV-2 envelope protein causes acute respiratory distress syndrome (ARDS)-like pathological damages and constitutes an antiviral target. Cell Res. 2021, 31, 847–860. [Google Scholar] [CrossRef]
  93. Nawawi, A.; Ma, C.; Nakamura, N.; Hattori, M.; Kurokawa, M.; Shiraki, K.; Kashiwaba, N.; Ono, M. Anti-herpes simplex virus activity of alkaloids isolated from Stephania cepharantha. Biol Pharm Bull 1999, 22, 268–274. [Google Scholar] [CrossRef] [Green Version]
  94. Okamoto, M.; Ono, M.; Baba, M. Potent inhibition of HIV type 1 replication by an antiinflammatory alkaloid, cepharanthine, in chronically infected monocytic cells. AIDS Res. Hum. Retrovir. 1998, 14, 1239–1245. [Google Scholar] [CrossRef]
  95. Furusawa, S.; Wu, J. The effects of biscoclaurine alkaloid cepharanthine on mammalian cells: Implications for cancer, shock, and inflammatory diseases. Life Sci. 2007, 80, 1073–1079. [Google Scholar] [CrossRef] [PubMed]
  96. Matsuda, K.; Hattori, S.; Komizu, Y.; Kariya, R.; Ueoka, R.; Okada, S. Cepharanthine inhibited HIV-1 cell–cell transmission and cell-free infection via modification of cell membrane fluidity. Bioorganic Med. Chem. Lett. 2014, 24, 2115–2117. [Google Scholar] [CrossRef] [PubMed]
  97. Baba, M.; Okamoto, M.; Kashiwaba, N.; Ono, M. Anti-HIV-1 activity and structure-activity relationship of cepharanoline derivatives in chronically infected cells. Antivir. Chem. Chemother. 2001, 12, 307–312. [Google Scholar] [CrossRef] [PubMed]
  98. Liu, X.; Wang, Y.; Zhang, M.; Li, G.; Cen, Y. Study on the inhibitory effect of cepharanthine on herpes simplex type-1 virus (HSV-1) in vitro. J. Chin. Med. Mater. 2004, 27, 107–110. [Google Scholar]
  99. Okamoto, M.; Ono, M.; Baba, M. Suppression of cytokine production and neural cell death by the anti-inflammatory alkaloid cepharanthine: A potential agent against HIV-1 encephalopathy. Biochem. Pharm. 2001, 62, 747–753. [Google Scholar] [CrossRef]
  100. Zhang, C.H.; Wang, Y.F.; Liu, X.J.; Lu, J.H.; Qian, C.W.; Wan, Z.Y.; Yan, X.G.; Zheng, H.Y.; Zhang, M.Y.; Xiong, S.; et al. Antiviral activity of cepharanthine against severe acute respiratory syndrome coronavirus in vitro. Chin. Med. J. (Engl.) 2005, 118, 493–496. [Google Scholar]
  101. Ohashi, H.; Watashi, K.; Saso, W.; Shionoya, K.; Iwanami, S.; Hirokawa, T.; Shirai, T.; Kanaya, S.; Ito, Y.; Kim, K.S. Multidrug treatment with nelfinavir and cepharanthine against COVID-19. BioRxiv 2020. [Google Scholar] [CrossRef] [Green Version]
  102. Nawawi, A.; Nakamura, N.; Hattori, M.; Kurokawa, M.; Shiraki, K. Inhibitory effects of Indonesian medicinal plants on the infection of herpes simplex virus type 1. Phytother. Res. 1999, 13, 37–41. [Google Scholar] [CrossRef]
  103. Dyall, J.; Coleman, C.M.; Venkataraman, T.; Holbrook, M.R.; Kindrachuk, J.; Johnson, R.F.; Olinger Jr, G.G.; Jahrling, P.B.; Laidlaw, M.; Johansen, L.M. Repurposing of clinically developed drugs for treatment of Middle East respiratory syndrome coronavirus infection. Antimicrob. Agents Chemother. 2014, 58, 4885–4893. [Google Scholar] [CrossRef] [Green Version]
  104. Boyd, M.R.; Hallock, Y.F.; Cardellina, J.H., 2nd; Manfredi, K.P.; Blunt, J.W.; Mcmahon, J.B.; Buckheit, R.W., Jr.; Bringmann, G.; Schaffer, M.; Cragg, G.M.; et al. Anti-HIV michellamines from Ancistrocladus korupensis. J. Med. Chem. 1994, 37, 1740–1745. [Google Scholar] [CrossRef]
  105. McMahon, J.B.; Currens, M.J.; Gulakowski, R.J.; Buckheit, R.W., Jr.; Lackman-Smith, C.; Hallock, Y.F.; Boyd, M.R. Michellamine B a novel plant alkaloid, inhibits human immunodeficiency virus-induced cell killing by at least two distinct mechanisms. Antimicrob. Agents Chemother. 1995, 39, 484–488. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Bringmann, G.; Steinert, C.; Feineis, D.; Mudogo, V.; Betzin, J.; Scheller, C. HIV-inhibitory michellamine-type dimeric naphthylisoquinoline alkaloids from the Central African liana Ancistrocladus congolensis. Phytochemistry 2016, 128, 71–81. [Google Scholar] [CrossRef] [PubMed]
  107. Angelova, A.; Tenev, T.; Varadinova, T. Expression of cellular proteins Bcl-X (L), XIAP and Bax involved in apoptosis in cells infected with herpes simplex virus 1 and effect of pavine alkaloid (-)-thalimonine on virus-induced suppression of apoptosis. Acta Virol. 2004, 48, 193–196. [Google Scholar] [PubMed]
  108. Vanquelef, E.; Amoros, M.; Boustie, J.; Lynch, M.A.; Waigh, R.D.; Duval, O. Synthesis and antiviral effect against herpes simplex type 1 of 12-substituted benzo [c] phenanthridinium salts. J. Enzym. Inhib. Med. Chem. 2004, 19, 481–487. [Google Scholar] [CrossRef] [PubMed]
  109. Tan, G.T.; Miller, J.F.; Kinghorn, A.D.; Hughes, S.H.; Pezzuto, J.M. HIV-1 and HIV-2 reverse transcriptases: A comparative study of sensitivity to inhibition by selected natural products. Biochem. Biophys. Res. Commun. 1992, 185, 370–378. [Google Scholar] [CrossRef] [PubMed]
  110. Cheng, T.-J.; Goodsell, D.S.; Kan, C.-C. Identification of sanguinarine as a novel HIV protease inhibitor from high-throughput screening of 2,000 drugs and natural products with a cell-based assay. Lett. Drug Des. Discov. 2005, 2, 364–371. [Google Scholar] [CrossRef]
  111. Shen, L.; Niu, J.; Wang, C.; Huang, B.; Wang, W.; Zhu, N.; Deng, Y.; Wang, H.; Ye, F.; Cen, S. High-throughput screening and identification of potent broad-spectrum inhibitors of coronaviruses. J. Virol. 2019, 93, e00023-19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Leitao Da-Cunha, E.V.; Fechine, I.M.; Guedes, D.N.; Barbosa-Filho, J.M.; Sobral Da Silva, M. Protoberberine Alkaloids; The Alkaloids: Chemistry and Biology; Cordell, G.A., Ed.; Academic Press: Cambridge, MA, USA, 2005; Volume 62, pp. 1–75. [Google Scholar]
  113. Babalghith, A.O.; Al-Kuraishy, H.M.; Al-Gareeb, A.I.; De Waard, M.; Al-Hamash, S.M.; Jean-Marc, S.; Negm, W.A.; Batiha, G.E.-S. The role of berberine in Covid-19: Potential adjunct therapy. Inflammopharmacology 2022, 3(6), 2003–2016. [Google Scholar] [CrossRef]
  114. Liu, H.; You, L.; Wu, J.; Zhao, M.; Guo, R.; Zhang, H.; Su, R.; Mao, Q.; Deng, D.; Hao, Y. Berberine suppresses influenza virus-triggered NLRP3 inflammasome activation in macrophages by inducing mitophagy and decreasing mitochondrial ROS. J. Leukoc. Biol. 2020, 108, 253–266. [Google Scholar] [CrossRef]
  115. Wu, Y.; Li, J.-Q.; Kim, Y.-J.; Wu, J.; Wang, Q.; Hao, Y. In vivo and in vitro antiviral effects of berberine on influenza virus. Chin. J. Integr. Med. 2011, 17, 444–452. [Google Scholar] [CrossRef]
  116. Li, H.L.; Han, T.; Liu, R.H.; Zhang, C.; Chen, H.S.; Zhang, W.D. Alkaloids from Corydalis saxicola and their anti-hepatitis B virus activity. Chem. Biodivers. 2008, 5, 777–783. [Google Scholar] [CrossRef] [PubMed]
  117. Aljofan, M.; Netter, H.J.; Aljarbou, A.N.; Hadda, T.B.; Orhan, I.E.; Sener, B.; Mungall, B.A. Anti-hepatitis B activity of isoquinoline alkaloids of plant origin. Arch. Virol. 2014, 159, 1119–1128. [Google Scholar] [CrossRef] [PubMed]
  118. Ng, T.B.; Huang, B.; Fong, W.P.; Yeung, H.W. Anti-human immunodeficiency virus (anti-HIV) natural products with special emphasis on HIV reverse transcriptase inhibitors. Life Sci. 1997, 61, 933–949. [Google Scholar] [CrossRef] [PubMed]
  119. Yao, M.; Fan, X.; Yuan, B.; Takagi, N.; Liu, S.; Han, X.; Ren, J.; Liu, J. Berberine inhibits NLRP3 Inflammasome pathway in human triple-negative breast cancer MDA-MB-231 cell. BMC Complement. Altern. Med. 2019, 19, 1–11. [Google Scholar] [CrossRef] [Green Version]
  120. Yang, L.M.; Wang, R.; Li, J.J.; Li, N.S.; Zheng, Y.T. Anti-HIV-1 activities of four berberine compounds in vitro. Chin. J. Nat. Med. 2007, 5, 225–228. [Google Scholar]
  121. Kaur, R.; Sharma, P.; Gupta, G.K.; Ntie-Kang, F.; Kumar, D. Structure-Activity-Relationship and Mechanistic Insights for Anti-HIV Natural Products. Molecules 2020, 25, 2070. [Google Scholar] [CrossRef]
  122. Ge, Y.-C.; Wang, K.-W. New analogues of aporphine alkaloids. Mini Rev. Med. Chem. 2018, 18, 1590–1602. [Google Scholar] [CrossRef]
  123. Reddy, M.V.; Rao, M.R.; Rhodes, D.; Hansen, M.S.; Rubins, K.; Bushman, F.D.; Venkateswarlu, Y.; Faulkner, D.J. Lamellarin alpha 20-sulfate, an inhibitor of HIV-1 integrase active against HIV-1 virus in cell culture. J. Med. Chem. 1999, 42, 1901–1907. [Google Scholar] [CrossRef]
  124. Fukuda, T.; Ishibashi, F.; Iwao, M. Lamellarin Alkaloids: Isolation, Synthesis, and Biological Activity. Alkaloids Chem. Biol. 2020, 83, 1–112. [Google Scholar]
  125. Mediouni, S.; Chinthalapudi, K.; Ekka, M.K.; Usui, I.; Jablonski, J.A.; Clementz, M.A.; Mousseau, G.; Nowak, J.; Macherla, V.R.; Beverage, J.N. Didehydro-cortistatin A inhibits HIV-1 by specifically binding to the unstructured basic region of Tat. mBio 2019, 10, e02662-18. [Google Scholar] [CrossRef] [Green Version]
  126. Kessing, C.F.; Nixon, C.C.; Li, C.; Tsai, P.; Takata, H.; Mousseau, G.; Ho, P.T.; Honeycutt, J.B.; Fallahi, M.; Trautmann, L.; et al. In Vivo Suppression of HIV Rebound by Didehydro-Cortistatin A, a "Block-and-Lock" Strategy for HIV-1 Treatment. Cell Rep. 2017, 21, 600–611. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Mohamed, S.M.; Hassan, E.M.; Ibrahim, N.A. Cytotoxic and antiviral activities of aporphine alkaloids of Magnolia grandiflora L. Nat. Prod. Res. 2010, 24, 1395–1402. [Google Scholar] [CrossRef] [PubMed]
  128. Avci, F.G.; Atas, B.; Gulsoy Toplan, G.; Gurer, C.; Sariyar Akbulut, B. Chapter 4—Antibacterial and Antifungal Activities of Isoquinoline Alkaloids of the Papaveraceae and Fumariaceae Families and Their Implications in Structure–Activity Relationships; Studies in Natural Products Chemistry; Atta ur, R., Ed.; Elsevier: Amsterdam, The Netherlands, 2021; Volume 70, pp. 87–118. [Google Scholar]
  129. Kim, H.-Y.; Shin, H.-S.; Park, H.; Kim, Y.-C.; Yun, Y.G.; Park, S.; Shin, H.-J.; Kim, K. In vitro inhibition of coronavirus replications by the traditionally used medicinal herbal extracts, Cimicifuga rhizoma, Meliae cortex, Coptidis rhizoma, and Phellodendron cortex. J. Clin. Virol. 2008, 41, 122–128. [Google Scholar] [CrossRef] [PubMed]
  130. Khan, T.; Khan, M.A.; Ullah, N.; Nadhman, A. Therapeutic potential of medicinal plants against COVID-19: The role of antiviral medicinal metabolites. Biocatal. Agric. Biotechnol. 2021, 31, 101890. [Google Scholar] [CrossRef]
  131. Pham, L.V.; Ngo, H.T.; Lim, Y.-S.; Hwang, S.B. Hepatitis C virus non-structural 5B protein interacts with cyclin A2 and regulates viral propagation. J. Hepatol. 2012, 57, 960–966. [Google Scholar] [CrossRef]
  132. Li, S.-Y.; Chen, C.; Zhang, H.-Q.; Guo, H.-Y.; Wang, H.; Wang, L.; Zhang, X.; Hua, S.-N.; Yu, J.; Xiao, P.-G. Identification of natural compounds with antiviral activities against SARS-associated coronavirus. Antivir. Res. 2005, 67, 18–23. [Google Scholar] [CrossRef]
  133. Cao, J.; Forrest, J.C.; Zhang, X. A screen of the NIH Clinical Collection small molecule library identifies potential anti-coronavirus drugs. Antivir. Res. 2015, 114, 1–10. [Google Scholar] [CrossRef]
  134. Yang, C.-W.; Lee, Y.-Z.; Kang, I.-J.; Barnard, D.L.; Jan, J.-T.; Lin, D.; Huang, C.-W.; Yeh, T.-K.; Chao, Y.-S.; Lee, S.-J. Identification of phenanthroindolizines and phenanthroquinolizidines as novel potent anti-coronaviral agents for porcine enteropathogenic coronavirus transmissible gastroenteritis virus and human severe acute respiratory syndrome coronavirus. Antivir. Res. 2010, 88, 160–168. [Google Scholar] [CrossRef]
  135. Jahan, I.; Ahmet, O. Potentials of plant-based substance to inhabit and probable cure for the COVID-19. Turk. J. Biol. 2020, 44, 228–241. [Google Scholar] [CrossRef]
  136. George, A.; Gopi Krishna Reddy, A.; Satyanarayana, G.; Raghavendra, N.K. 1,2,3,4-Tetrahydroisoquinolines as inhibitors of HIV-1 integrase and human LEDGF/p75 interaction. Chem. Biol. Drug Des. 2018, 91, 1133–1140. [Google Scholar] [CrossRef]
  137. Turano, A.S.G.; Caruso a Bonfanti, C.; Luzzati, R.; Bassetti, D.; Manca, N. Inhibitory effect of papaverine on HIV replication in vitro. AIDS Res. Hum. Retrovir. 1989, 5, 183–192. [Google Scholar] [CrossRef]
  138. Nishiyama, Y.; Iwasa, K.; Okada, S.; Takeuchi, S.; Moriyasu, M.; Kamigauchi, M.; Koyama, J.; Takeuchi, A.; Tokuda, H.; Kim, H.S. Geranyl derivatives of isoquinoline alkaloids show increased biological activities. Heterocycles 2010, 81, 1193–1229. [Google Scholar] [CrossRef]
  139. Iwasa, K.; Moriyasu, M.; Tachibana, Y.; Kim, H.-S.; Wataya, Y.; Wiegrebe, W.; Bastow, K.F.; Cosentino, L.M.; Kozuka, M.; Lee, K.-H. Simple Isoquinoline and Benzylisoquinoline Alkaloids as Potential Antimicrobial, Antimalarial, Cytotoxic, and Anti-HIV Agents. Bioorganic Med. Chem. 2001, 9, 2871–2884. [Google Scholar] [CrossRef] [PubMed]
  140. Weber, C.; Opatz, T. Chapter One—Bisbenzylisoquinoline Alkaloids, The Alkaloids: Chemistry and Biology; Knölker, H.-J., Ed.; Academic Press: Cambridge, MA, USA, 2019; pp. 1–114. [Google Scholar]
  141. Okamoto, M.; Okamoto, T.; Baba, M. Inhibition of human immunodeficiency virus type 1 replication by combination of transcription inhibitor K-12 and other antiretroviral agents in acutely and chronically infected cells. Antimicrob. Agents Chemother. 1999, 43, 492–497. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Liu, Y.; Tang, Q.; Rao, Z.; Fang, Y.; Jiang, X.; Liu, W.; Luan, F.; Zeng, N. Inhibition of herpes simplex virus 1 by cepharanthine via promoting mcellular autophagy through up-regulation of STING/TBK1/P62 pathway. Antivir. Res. 2021, 193, 105143. [Google Scholar] [CrossRef] [PubMed]
  143. Fan, H.-H.; Wang, L.-Q.; Liu, W.-L.; An, X.-P.; Liu, Z.-D.; He, X.-Q.; Song, L.-H.; Tong, Y.-G. Repurposing of clinically approved drugs for treatment of coronavirus disease 2019 in a 2019-novel coronavirus-related coronavirus model. Chin. Med. J. 2020, 133, 1051–1056. [Google Scholar] [CrossRef]
  144. Li, S.; Liu, W.; Chen, Y.; Wang, L.; An, W.; An, X.; Song, L.; Tong, Y.; Fan, H.; Lu, C. Transcriptome analysis of cepharanthine against a SARS-CoV-2-related coronavirus. Brief. Bioinform. 2021, 22, 1378–1386. [Google Scholar] [CrossRef]
  145. Dong, S.; Yu, R.; Wang, X.; Chen, B.; Si, F.; Zhou, J.; Xie, C.; Li, Z.; Zhang, D. Bis-Benzylisoquinoline Alkaloids Inhibit Porcine Epidemic Diarrhea Virus In Vitro and In Vivo. Viruses 2022, 14, 1231. [Google Scholar] [CrossRef]
  146. Li, H.; Chen, X.; Zhou, S.-J. Dauricine combined with clindamycin inhibits severe pneumonia co-infected by influenza virus H5N1 and Streptococcus pneumoniae in vitro and in vivo through NF-κB signaling pathway. J. Pharmacol. Sci. 2018, 137, 12–19. [Google Scholar] [CrossRef]
  147. Zeng, X.; Lao, B.; Dong, X.; Sun, X.; Dong, Y.; Sheng, G.; Fu, J. Study on anti-influenza effect of alkaloids from roots of Mahonia bealei in vitro. Zhong Yao Cai 2003, 26, 29–30. [Google Scholar]
  148. Hu, S.; Dutt, J.; Zhao, T.; Foster, C.S. Stephen Foster Tetrandrine potently inhibits herpes simplex virus type-1-induced keratitis in BALB/c mice. Ocul. Immunol. Inflamm. 1997, 5. [Google Scholar] [CrossRef] [PubMed]
  149. Wink, M. Potential of DNA intercalating alkaloids and other plant secondary metabolites against SARS-CoV-2 causing COVID-19. Diversity 2020, 12, 175. [Google Scholar] [CrossRef]
  150. Yang, S.; Xu, M.; Lee, E.M.; Gorshkov, K.; Shiryaev, S.A.; He, S.; Sun, W.; Cheng, Y.; Hu, X.; Tharappel, A.M.; et al. Emetine inhibits Zika and Ebola virus infections through two molecular mechanisms: Inhibiting viral replication and decreasing viral entry. Cell Discov. 2018, 4, 31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  151. Andersen, P.I.; Krpina, K.; Ianevski, A.; Shtaida, N.; Jo, E.; Yang, J.; Koit, S.; Tenson, T.; Hukkanen, V.; Anthonsen, M.W.; et al. Novel Antiviral Activities of Obatoclax, Emetine, Niclosamide, Brequinar, and Homoharringtonine. Viruses 2019, 11, 964. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Tan, G.T.; Kinghorn, A.D.; Hughes, S.H.; Pezzuto, J.M. Psychotrine and its O-methyl ether are selective inhibitors of human immunodeficiency virus-1 reverse transcriptase. J. Biol.Chem. 1991, 266, 23529–23536. [Google Scholar] [CrossRef] [PubMed]
  153. Tan, G.T.; Pezzuto, J.M.; Kinghorn, A.D.; Hughes, S.H. Evaluation of natural products as inhibitors of human immunodeficiency virus type 1 (HIV-1) reverse transcriptase. J. Nat. Prod. 1991, 54, 143–154. [Google Scholar] [CrossRef]
  154. Manfredi, K.P.; Blunt, J.W.; Cardellina, J.H., 2nd; Mcmahon, J.B.; Pannell, L.L.; Cragg, G.M.; Boyd, M.R. Novel alkaloids from the tropical plant Ancistrocladus abbreviatus inhibit cell killing by HIV-1 and HIV-2. J. Med. Chem. 1991, 34, 3402–3405. [Google Scholar] [CrossRef]
  155. White, E.L.; Chao, W.R.; Ross, L.J.; Borhani, D.W.; Hobbs, P.D.; Upender, V.; Dawson, M.I. Michellamine alkaloids inhibit protein kinase C. Arch. Biochem. Biophys. 1999, 365, 25–30. [Google Scholar] [CrossRef]
  156. Supko Jg, M.L. Pharmacokinetics of michellamine B, a naphthylisoquinoline alkaloid with in vitro activity against human immunodeficiency virus types 1 and 2, in the mouse and dog. Antimicrob. Agents Chemother. 1995, 39, 9–14. [Google Scholar] [CrossRef]
  157. Hallock, Y.F.; Manfredi, K.P.; Dai, J.-R.; Cardellina, J.H.; Gulakowski, R.J.; Mcmahon, J.B.; Schäffer, M.; Stahl, M.; Gulden, K.-P.; Bringmann, G. Michellamines D− F, new HIV-inhibitory dimeric naphthylisoquinoline alkaloids, and korupensamine E, a new antimalarial monomer, from Ancistrocladus korupensis. J. Nat. Prod. 1997, 60, 677–683. [Google Scholar] [CrossRef]
  158. Ka, S.; Merindol, N.; Sow, A.A.; Singh, A.; Landelouci, K.; Plourde, M.B.; Pépin, G.; Masi, M.; Di Lecce, R.; Evidente, A. Amaryllidaceae alkaloid cherylline inhibits the replication of dengue and Zika viruses. Antimicrob. Agents Chemother. 2021, 65, e00398-21. [Google Scholar] [CrossRef] [PubMed]
  159. Homan, J.W.; Steele, A.D.; Martinand-Mari, C.; Rogers, T.J.; Henderson, E.E.; Charubala, R.; Pfleiderer, W.; Reichenbach, N.L.; Suhadolnik, R.J. Inhibition of morphine-potentiated HIV-1 replication in peripheral blood mononuclear cells with the nuclease-resistant 2-5A agonist analog, 2-5A(N6B). JAIDS J. Acquir. Immune Defic. Syndr. 2002, 30, 9–20. [Google Scholar] [CrossRef] [PubMed]
  160. Sun, W.W. Structural Modification, Bioactivity of Derivatives of Three Protopines and Their Structure-Activity Relationship. Postgraduate Thesis, Northwest A&F University, ShanXi, China.
  161. Huang, Q.; Wu, X.; Zheng, X.; Luo, S.; Xu, S.; Weng, J. Targeting inflammation and cytokine storm in COVID-19. Pharmacol. Res. 2020, 159, 105051. [Google Scholar] [CrossRef] [PubMed]
  162. Choi, H.S.; Kim, H.S.; Min, K.R.; Kim, Y.; Lim, H.K.; Chang, Y.K.; Chung, M.W. Anti-inflammatory effects of fangchinoline and tetrandrine. J. Ethnopharmacol. 2000, 69, 173–179. [Google Scholar] [CrossRef]
  163. Wu, S.-J.; Ng, L.-T. Tetrandrine Inhibits Proinflammatory Cytokines, iNOS and COX-2 Expression in Human Monocytic Cells. Biol. Pharm. Bull. 2007, 30, 59–62. [Google Scholar] [CrossRef] [Green Version]
  164. Xue, Y.; Wang, Y.; Feng, D.-C.; Xiao, B.-G.; Xu, L.-Y. Tetrandrine suppresses lipopolysaccharide-induced microglial activation by inhibiting NF-κB pathway. Acta Pharmacol. Sin. 2008, 29, 245–251. [Google Scholar] [CrossRef]
  165. Eissa, L.A.; Kenawy, H.I.; El-Karef, A.; Elsherbiny, N.M.; El-Mihi, K.A. Antioxidant and anti-inflammatory activities of berberine attenuate hepatic fibrosis induced by thioacetamide injection in rats. Chem. -Biol. Interact. 2018, 294, 91–100. [Google Scholar] [CrossRef]
  166. Ebrahimi, S.A. Noscapine, a possible drug candidate for attenuation of cytokine release associated with SARS-CoV-2. Drug Dev. Res. 2020, 81, 765–767. [Google Scholar] [CrossRef]
  167. Xia, H.; Liu, Y.; Xia, G.; Liu, Y.; Lin, S.; Guo, L. Novel isoquinoline alkaloid litcubanine A-a potential anti-inflammatory candidate. Front. Immunol. 2021, 1987. [Google Scholar] [CrossRef]
Figure 1. Some important L-tyrosine-derived isoquinoline and their related alkaloids.
Figure 1. Some important L-tyrosine-derived isoquinoline and their related alkaloids.
Biomolecules 13 00017 g001
Figure 2. Various steps involved in the pathology of viral infections that might harm cells. MEK/ERK: Mitogen-activated protein kinase/extracellular-signal-regulated kinase, NF-κB: nuclear factor kappa B, TRPML: transient receptor potential, TLR7: Toll-like receptor (TLR)-7, MyD88: myeloid differentiation primary response protein, TNF-α: tumor necrosis factor alpha, IL-6: interleukin-6.
Figure 2. Various steps involved in the pathology of viral infections that might harm cells. MEK/ERK: Mitogen-activated protein kinase/extracellular-signal-regulated kinase, NF-κB: nuclear factor kappa B, TRPML: transient receptor potential, TLR7: Toll-like receptor (TLR)-7, MyD88: myeloid differentiation primary response protein, TNF-α: tumor necrosis factor alpha, IL-6: interleukin-6.
Biomolecules 13 00017 g002
Figure 3. Structure–activity relationship of protoberberine alkaloids.
Figure 3. Structure–activity relationship of protoberberine alkaloids.
Biomolecules 13 00017 g003
Figure 4. Structure–activity relationship of aporphine alkaloids.
Figure 4. Structure–activity relationship of aporphine alkaloids.
Biomolecules 13 00017 g004
Figure 5. Chemical structures of isoquinoline and related alkaloids falling under various categories.
Figure 5. Chemical structures of isoquinoline and related alkaloids falling under various categories.
Biomolecules 13 00017 g005
Figure 6. Structure–activity relationship of benzylisoquinoline (A) and bisbenzyl-isoquinoline alkaloids (B).
Figure 6. Structure–activity relationship of benzylisoquinoline (A) and bisbenzyl-isoquinoline alkaloids (B).
Biomolecules 13 00017 g006
Table 1. In silico binding affinities of isoquinoline and related alkaloids against SARS-CoV-2.
Table 1. In silico binding affinities of isoquinoline and related alkaloids against SARS-CoV-2.
Sr. No.AlkaloidIsoquinoline and Related AlkaloidStructureTargetBinding Score/AffinityDocking Software References
1.Thebaineaporphine alkaloidBiomolecules 13 00017 i001ACE2, S-protein−100.77 and −103.58 kcal/molMolegro Virtual Docker 3.0.0[46]
2.BerberineProtoberberine alkaloidBiomolecules 13 00017 i002ACE2, S-protein, 3CLpro−97.54,
−99.93 and
−7.3 kcal/mol
Auto Dock;
Auto Dock Vina
[46,47]
3.LycorineTetrahydroisoquinolinesBiomolecules 13 00017 i003S1 subunit; SARS-CoV-2−86.92 kcal/molDiscovery Studio 2.5[42]
4.Tylophorinephenanthraindolizidine alkaloidBiomolecules 13 00017 i004S1 subunit; SARS-CoV-2−89.77 kcal/molDiscovery Studio 2.5[42]
5.Tetrandrinebis-benzylisoquinoline alkaloidBiomolecules 13 00017 i005S1 subunit; SARS-CoV-2−72.96 kcal/molDiscovery Studio 2.5[42]
6.Fangchinolinebis-benzylisoquinolineBiomolecules 13 00017 i006S1 subunit; SARS-CoV-2−92.66 kcal/molDiscovery Studio 2.5[42]
7.Berbaminebis-benzylisoquinolineBiomolecules 13 00017 i007Mpro−20.79 kcal/molPyRx[48]
8.Noscapinephthalideisoquinoline alkaloidBiomolecules 13 00017 i008Mpro−292.42 kJ/molHex 8.0[49]
9.PalmatineprotoberberineBiomolecules 13 00017 i009Mpro>8 kcal/molSwiss Dock;
EADock DSS
[50]
10.Morphine aporphine alkaloidBiomolecules 13 00017 i010Mpro>−6.0 kcal/molAutoDock Vina v.1.0.2.[51]
11.Codeineaporphine alkaloidBiomolecules 13 00017 i011Mpro>−6.0 kcal/molAutoDock Vina v.1.0.2.[51]
12.Cepharanthinebiscoclaurine alkaloidBiomolecules 13 00017 i0123CLpro, S1-RBD, and TMPRSS-2−8.5, −106.74, −7.4 kcal/mol Auto Dock Vina v.1.0.2;
Discovery Studio 2.5
[42,52]
13.Cassiarin Disoquinoline alkaloidBiomolecules 13 00017 i013RdRP−150.7 kJ/molMolegro Virtual Docker (version 6.0, Molegro ApS, Aarhus, Denmark))[53]
14.CoptisineprotoberberineBiomolecules 13 00017 i014Mpro, N3-Mpro−9.15 and −8.17 kcal/molAutodock (version 4.2)[54]
15.EmetineIpecac alkaloidsBiomolecules 13 00017 i015Mpro−10.17 kcal/molAutodock version 4.2.6[55]
Table 2. In vitro activities of isoquinoline and related alkaloids against several viral infections.
Table 2. In vitro activities of isoquinoline and related alkaloids against several viral infections.
Sr. No.Active CompoundCell Lines/modelIC50/CC50 ValueEC50/ED50 ValueMechanism/ActivityAntiviral ActivityReferences
Protoberberine Alkaloids
1.BerberineHEC-1-A cells HEK293T165.7 µM-suppression of HSV-induced NF-κB activation, IκB-α degradation and p65 nuclear translocationHSV[60]
J774A.1 cells--inhibition of HIV-PI induced TNF-α and IL-6 in macrophages HIV[61]
Calu-3 cells--increases CD8+ cells and IFN-γ to produce immunomodulatory activitySARS-CoV and SARS-CoV-2[62]
Vero E6 cells10.58 µΜ-drug antagonism was seen (remdesivir + BBR)SARS-CoV-2[63]
Vero E6 cell-16.70 μMsuppression of SARS-CoV-2 target proteinsSARS-CoV-2[64]
VeroE6 cells and nasal epithelial cells-9.1 µMinhibition of SARS-CoV-2 replicationSARS-CoV-2[65]
RAW 264.7 macrophage and A549 cells0.44 μM-inhibition of viral growth of influenza and suppresses production of TNF-α and PGE2H1N1[66]
human osteosarcoma cell (HOS)-12.2 µMinhibits MAPK/PI3K-AKt signaling pathwaysChikungunya virus[67]
A549 cells and in vivo (mice)--BBR suppresses TLR7, MyD88, and NF-κB to resist viral replication in mice H1N1[68,69]
HFF, HELF, and NIH 3T3-2.65 µMinhibition of DNA polymerase and immediate-early (IE-3) proteins HCMV[70]
A549,
LET1,
HAE, and
MDCK
17 µM,
4 µM,
16 µM, and
52 µM
- interfere MEK/ERK pathway to inhibit export of the viral ribonucleoproteinH1N1[68]
2.Saxicolalines AHep G 2.2.15 cell line2.19 and >2.81 against HBsAg b* and HBeAg c* respectively--HBV[71]
3.N-methylnarceimicineHep G 2.2.15 cell line1.24 µM and 1.86 µM against HBsAg b and HBeAg c respectively--HBV[71]
Aporphine Alkaloids
1.RoemerineH9 cells-0.84 μg/mLinhibition of HIV replication by 50%HIV-1[72]
2.Nuciferine0.8 μg/mL
3.Nornuciferine<0.8 μg/mL
4.BoldinePMBMCs, CEM-GXR cells, Vero cells207.7, 250 µM50.2 µMinhibit HIV-1 replication, reduction of the viral titerHIV-1, poliovirus[73,74]
5.Dasymaroine AC8166 cell line-1.93 to 9.70 µMreduce viral replication by 50%HIV-1[75]
6.Dasymaroine B
7.LaurolitsineVero cells95 µM 62 µMreduction of the viral titerPoliovirus[74]
8.Isoboldine HCL217 µM−15 µM
9.Glaucine fumarate142 µM9 µM
10.N-acetylnorglaucine342 µM50 µM
11.N-methyllaurotetanine250 µM15 µM
12.laurotetanine, HCl165 µM31 µM
13.(+)-bulbocapnineMDBK and Vero cell lines---PI-3[76]
14.PachystaudineVero cells68 µM−31 µMreduction of the viral titer and viral replicationPoliovirus and HSV[74,77]
15.Oxostephanine28 µM-
16.Oliverine HClVero cells9 µM-reduction of the viral titerPoliovirus[74]
Benzylisoquinoline and
Benzyltetrahydroisoquinoline Alkaloids
1.(+)-1(R)-coclaurineH9 cells>100 µg/mL 0.8 µg/mLinhibition of HIV replication by 50%HIV-1[72]
2.(−)-1(R)-N-methylcoclaurinre1.45 µg/mL<0.8 µg/mL
3.(−)-1(S)-norcoclaurine20 µg/mL<0.8 µg/mL
4.Armepavine1.77 µg/mL<0.8 µg/mL
5.Lotusine>100 µg/mL20.7 µg/mL
6.PapaverineVero E6, Caco-2, and BHK-21 cells1.1 µM-reduce SARS-CoV-2 cytotoxic effectSARS-CoV-2[78]
7.Papaverine hydrochlorideMT4 cells 5.8 µMinhibition of viral replicationHIV[79]
Bis-benzylisoquinoline Alkaloids (BBI)
1.AromolineMT-4 cells-IC100 31.3µg/mLcytopathic effect of HIV is inhibitedHIV-1[80]
2.CycleanineHCT-116-1.83 µg/mL-HIV-2[81]
3.FangchinolineMT-4, PM1, and 293T cells-0.8 to 1.7 µMinterfering with gp160 proteolytic cleavage thus inhibit viral replicationHIV-1[82]
MRC-5 fibroblast lung cell line1.01 µM-inhibition of S and N protein expression as well as HCoV-OC43 replicationHCoV-OC43[83]
4.TetrandrineBALB--IL-6 inhibition in the corneal inflammation HSV[84]
HEK 293T cells expressing ACE2->10 µMsuppress viral entry by inhibiting Ca2+-mediated fusionSARS-CoV-2[85]
MRC-5 cells0.33 μmol/L-two-pore channel 2 inhibitionSARS-CoV-2[86,87]
MRC-5 fibroblast lung cell line0.33 µM-inhibition of S and N protein expression as well as HCoV-OC43 replicationHCoV-OC43[83]
5.NeferineHEK293/hACE2 and HuH7 cell lines-0.13–0.41 µΜblockage of viral entry by inhibiting Ca2+-mediated fusionHCOV OC43[88]
Dog hepatic microsomes--N-demethylation and O-demethylationHIV[89]
HEK 293T cells expressing ACE2->10 µMsuppress viral entry by inhibiting Ca2+−-mediated fusionSARS-CoV-2[85]
Huh7, HEK293/hACE2 cell lines-0.36 µMblockage of Ca2+ dependent membrane fusionSARS-CoV-2[88]
6.BerbamineHela cells, A549 cells-JEV (1.62 µM) ZIKV (2.17 µM) inhibiting Ca2+ channel transient receptor potential membrane channel mucolipin (TRPML)JEV andZIKV[90]
Huh7-cells-~2.4 μMinhibition of Ca2+ influx and TRPMLsSARS-CoV-2[91]
Vero E6 cells5.79 µM0.94 µMinhibition to the 2-E channelSARS-CoV-2[92]
Vero cells16.3–24.9 µg/mL--HSV-1 TK and HSV-2[93]
7.CepharanthineU1 and ACH-2-0.016 µg/mLinhibition of NF-κB pathway HIV-1[94]
U937 cells4.6 µg/mL-inhibition of inflammatory cytokinesHIV-1[95]
PHA-blast cells from PBMC, HEK 293T, MOLT4 cellsCC50—10.0 µg/mL-reduce plasma membrane fluidity to block the NF-κB pathway and HIV-1 entranceHIV-1[96]
U1 cells-0.0041 µg/mLinhibition of NF-κB pathwayHIV-1[94,97]
Vero cells and Hela cells0.835 μg/mL-inhibition of NF-κBHSV[98,99]
MRC-5 fibroblast lung cell line0.83 µM -inhibition of viral S and N protein expression as well as HCoV-OC43 replicationHCoV-OC43[83]
Vero E6 cells6.0 µg/mL–9.5 µg/mL--SARS-CoV[100]
HEK 293T cells expressing ACE2 ->10 µMsuppression of viral entry through inhibition of Ca2+-mediated fusionSARS-CoV-2[85]
Vero E6 cells0.91 µM-combination of NFV and CEP reduces viral RNA levelsSARS-CoV-2[101]
8.di-O-acetylsinococuline (FK-3000)MT-4 cellsCC0 15.6 µg/mL-inhibition of HIV cytopathic effect HIV-1[80]
9.12-O-ethylpiperazinyl cepharanolineU1 cells-0.028 µg/mLinhibition of NF-κB pathwayHIV-1[97]
10.HomoaromolineVero cells16.3–24.9 µg/mL--HSV-1 TK and HSV-2[102]
11.Isotetrandrine
12.Thalrugosine
13.Obamegine
14.HernandezineHEK 293T cells ->10 µMinhibition of Ca2+-mediated fusion SARS-CoV-2[85]
Ipecac Alkaloids
1.EmetineHEK293 cells93–52.9 nM-inhibit MERS-CoV entryMERS-CoV[103]
SNB-19 cells29.8 nM-
Vero E6 cells8.74 nM-
BHK21 cells0.30 µM
Vero E6, Caco-2, and BHK-21 cells0.52 µM-inhibit cytotoxic effect of SARS-CoV-2SARS-CoV-2[78]
2.Emetine dihydrochloride hydrateVero E6 cells-0.051 µMinhibition of viral replication -SARS[103]
0.014 µMMERS
Naphthylisoquinoline Alkaloids
1.Michellamine AMT-2 target, CEM-SS cells-EC50~20 µMinhibition of viral replicationHIV-1 and HIV-2[104]
2.Michellamine BMT-2 target, CEM-SS cells-EC50~20 µMinhibition of viral replicationHIV-1 and HIV-2[104,105]
CEM-SS and MT-2-18 µg/mLinhibition of viral replicationHIV-1 and HIV-2[104]
H9 Cells20.4 µM-
3.Michellamine A2H9 Cells29.6 µM-inhibition of HIV replicationHIV-1[106]
4.Michellamine A3H9 Cells15.2 µM-HIV-1[106]
5.Michellamine A4 H9 Cells35.9 µM-HIV-1[106]
Pavine Alkaloid
1.(−)-ThalimonineMDBK cells--restoration of apoptosis during viral replicationHSV[107]
Morphinan and Promorphinan
1.FK-3000Vero cells7.8 µg/mL-reduction in plaque formationHSV[93]
2.Cephakicine44.4 µg/mL
3.Sinoacutine>50 µg/mL
4.Cephasamine
5.Cephamonine
6.Sinomenine
7.14-episinomenine
8.Tannagine
Benzophenanthridines
1.FagaronineVero cellsCC50 > 0.3 mM-inhibition of topoisomerases I and IIHSV[108]
2.Fagaronine chlorideHIV-1 and HIV-2 RT assaysIC50—9.5 µg/mL-inhibit RNA and DNA polymerizing enzymesHIV-2 RT[109]
3.Nitidine chlorideIC50—7.1 µg/mL-
4.SanguinarineHTS (E. coli-based assay)IC50—12.82 µM-inhibits proteolytic activityHIV-1[110]
5.ChelerythrineIC50—38.71 µM
6.LycorineBHK21 cells-0.15 µMviral load suppressionHCoV-OC43[111]
*HBsAg b: hepatitis B virus surface antigen and HBeAgc: hepatitis B virus e antigen
Table 3. Clinical trials of isoquinoline alkaloids against viral infections.
Table 3. Clinical trials of isoquinoline alkaloids against viral infections.
Sr. No.TitlePhaseViral InfectionStudyCompletion of StudyOriginNCT Number
1The Effect of Berberine on Intestinal Function and Inflammatory Mediators in Severe Patients with Covid-19 (BOIFIM)Phase 4COVID-19InterventionalApril 2020Chinese medical associationNCT04479202
2Effect of Berberine on Metabolic Syndrome, Efficacy and Safety in Combination with Antiretroviral Therapy in PLWH. (BERMESyH)Phase 3HIV-1InterventionalJuly 2022Hospital Civil de GuadalajaraNCT04860063
3Tetrandrine Tablets Used in the Treatment of COVID-19 (TT-NPC)Phase 4COVID-19InterventionalMay 2021Henan Provincial People’s HospitalNCT04308317
4Study of Oral High/Low-dose Cepharanthine Compared with Placebo in Non-Hospitalized Adults with COVID-19Phase 2COVID-19InterventionalAugust 2022Hai Li, Shanghai Jiao Tong University School of MedicineNCT05398705
5Measurement of the Efficacy of MORPHINE in the Early Management of Dyspnea in COVID-19 Positive Patients (CODYS)NACOVID-19ObservationalFebruary 2021Hospices Civils de LyonNCT04522037
6Safety and Efficacy of COVIDEX™ Therapy in Management of Adult COVID-19 Patients in Uganda. (COT)Phase 2COVID-19InterventionalDecember 2022College of Health Sciences, Makerere UniversityNCT05228626
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

Sharma, D.; Sharma, N.; Manchanda, N.; Prasad, S.K.; Sharma, P.C.; Thakur, V.K.; Rahman, M.M.; Dhobi, M. Bioactivity and In Silico Studies of Isoquinoline and Related Alkaloids as Promising Antiviral Agents: An Insight. Biomolecules 2023, 13, 17. https://doi.org/10.3390/biom13010017

AMA Style

Sharma D, Sharma N, Manchanda N, Prasad SK, Sharma PC, Thakur VK, Rahman MM, Dhobi M. Bioactivity and In Silico Studies of Isoquinoline and Related Alkaloids as Promising Antiviral Agents: An Insight. Biomolecules. 2023; 13(1):17. https://doi.org/10.3390/biom13010017

Chicago/Turabian Style

Sharma, Divya, Neetika Sharma, Namish Manchanda, Satyendra K. Prasad, Prabodh Chander Sharma, Vijay Kumar Thakur, M. Mukhlesur Rahman, and Mahaveer Dhobi. 2023. "Bioactivity and In Silico Studies of Isoquinoline and Related Alkaloids as Promising Antiviral Agents: An Insight" Biomolecules 13, no. 1: 17. https://doi.org/10.3390/biom13010017

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