Next Article in Journal
Evaluation of the Insect Resistance Efficacy of Transgenic Maize LD05 in China
Next Article in Special Issue
Ethnoveterinary Potential of Acacia (Vachellia and Senegalia) Species for Managing Livestock Health in Africa: From Traditional Uses to Therapeutic Applications
Previous Article in Journal
Advances in the Genus Ulva Research: From Structural Diversity to Applied Utility
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Therapeutic Potential of Erythrina Genus: Bioactive Phytoconstituents with Potent Antiviral and Antimicrobial Activities

by
Muchtaridi Muchtaridi
1,2,*,
Samuel Lestyawan
3,
Maitsa Alya Fakhirah
3,
Agus Rusdin
1,
Shela Salsabila
1,2,
Sandra Megantara
1,2,
Anas Subarnas
4 and
Nur Kusaira Khairul Ikram
5
1
Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, West Java, Indonesia
2
Research Collaboration Center for Radiopharmaceuticals Theranostic, Universitas Padjadjaran, Sumedang 45363, West Java, Indonesia
3
Apothecary Program, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, West Java, Indonesia
4
Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, West Java, Indonesia
5
Institute of Biological Sciences, Faculty of Science, Universiti Malaya, Kuala Lumpur 50603, Malaysia
*
Author to whom correspondence should be addressed.
Plants 2025, 14(19), 3053; https://doi.org/10.3390/plants14193053
Submission received: 23 August 2025 / Revised: 24 September 2025 / Accepted: 25 September 2025 / Published: 2 October 2025

Abstract

Infectious diseases present a significant global health challenge, further exacerbated by the rising prevalence of antimicrobial resistance and the limited availability of effective antiviral and antimicrobial agents. The Erythrina genus has garnered scientific interest due to its diverse array of bioactive phytoconstituents, with potential therapeutic relevance. This review aims to synthesize and critically assess the existing literature on the antiviral, antibacterial, antifungal, and antiplasmodial properties of Erythrina species. A comprehensive literature search was conducted using PubMed, Scopus, and Google Scholar databases. Relevant studies were identified through keyword searches combining pathogen-specific terms with “Erythrina”. The extracted data were categorized based on the pathogen type and its associated bioactive compounds. Several Erythrina species exhibited substantial antiviral activity against prominent viral pathogens, such as HIV and SARS-CoV-2. Notably, strong antibacterial efficacy was observed against Staphylococcus aureus, including multidrug-resistant strains. Antifungal activity was most pronounced against Candida albicans, while potent antiplasmodial effects were reported against both drug-sensitive and drug-resistant strains of Plasmodium falciparum. These pharmacological effects were predominantly attributed to prenylated flavonoids, isoflavones, pterocarpans, and erythrina-type alkaloids. Further mechanistic studies and in vivo evaluations are essential to fully assess their clinical efficacy and support the development of plant-derived antimicrobial agents.

Graphical Abstract

1. Introduction

Infectious diseases are caused by pathogens such as bacteria, viruses, fungi, and parasites, and continue to pose a major global health concern [1]. According to the World Health Organization (WHO), infectious diseases remain one of the leading causes of morbidity and mortality globally, exacerbated by the rise in antibiotic-resistant strains and the emergence of novel pathogens [2]. Current treatment modalities, including antibiotics and antiviral drugs, are becoming less effective due to pathogens’ resistance mechanisms and the limited development of new drugs [3]. Consequently, there is a growing interest in exploring alternative therapeutic sources, such as plant-derived compounds, which have historically been a rich source of novel bioactive agents with potential therapeutic benefits [4].
Erythrina plants, commonly known as “coral trees”, belong to the Fabaceae family and are renowned for their diverse medicinal properties. These plants have been traditionally utilized in diverse cultures for their purported antiviral and antimicrobial properties. Recent studies have identified several bioactive compounds from the Erythrina genus that demonstrate promising antiviral and antimicrobial activities [5,6]. Such findings suggest that Erythrina species could offer valuable alternatives or supplements to conventional treatments, addressing the pressing need for novel therapeutic agents against resistant pathogens and emerging infectious diseases.
This review aims to synthesize and critically evaluate the existing research on the antiviral and antimicrobial properties of Erythrina species. By summarizing identified bioactive compounds and their mechanisms of action, this review underscores the genus’s potential as a source of novel therapeutic agents. This synthesis of existing knowledge serves as a guide for future research directions and encourages further investigation into the clinical applications of Erythrina-derived compounds in the management of infectious diseases.
The persistent threat of infectious diseases continues to pose a significant global health challenge, resulting in substantial morbidity and mortality across both developing and industrialized nations. Despite substantial advancements in modern medicine, the emergence of antibiotic-resistant bacterial strains and novel viral pathogens, such as SARS-CoV-2, has undermined the effectiveness of current therapies [2,3]. The declining pipeline of novel antimicrobial agents further intensifies this crisis, underscoring the need to explore alternative and supplementary strategies to conventional pharmacotherapy.
The current frontline approach relies heavily on synthetic antimicrobials, including antibiotics, antifungals, antiprotozoals, and antiviral agents. While these therapies have improved global health outcomes, their long-term efficacy is limited by adaptive resistance mechanisms, narrow pathogen specificity, and adverse safety profiles. Resistance to β-lactam antibiotics, neuraminidase inhibitors, and even artemisinin-based compounds has been reported across diverse clinically relevant pathogens [4]. This has prompted the scientific community to reexamine the therapeutic value of natural products, particularly those derived from medicinal plants, which often exhibit multi-target activities and lower risk of resistance development.
Among the diverse botanical sources with ethnopharmacological relevance, Erythrina species are particularly notable for their longstanding ethnomedicinal use in Africa, Asia, and the Americas. These plants produce a wide range of secondary metabolites, including prenylated flavonoids, erythrina-type alkaloids, and pterocarpans, many of which exhibit potent antimicrobial, antiviral, antifungal, and antiplasmodial activities [6,7]. Several Erythrina species have demonstrated efficacy against priority pathogens such as S. aureus, C. albicans, Plasmodium falciparum, and even HIV and SARS-CoV-2, thereby positioning the genus as a promising reservoir of bioactive compounds for future drug development [8,9].
Despite numerous studies, with 81 published investigations collected from Scopus, PubMed, and Google Scholar databases reporting on the biological activities of isolated compounds and crude extracts from Erythrina species, including [10,11,12,13,14,15,16,17,18,19,20,21,22], antibacterial [23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41], antifungal [42,43,44,45,46,47], and antiplasmodial [48,49,50,51,52,53,54,55,56,57,58,59] properties, systematic and mechanistically oriented reviews remain limited. Existing reviews primarily concentrate on ethnobotany or individual pharmacological aspects, frequently disregarding integrative evaluations such as compound–pathogen interactions, comparative species analyses, and the potential role of Erythrina-derived compounds in combating antimicrobial resistance. Additionally, correlations between extract composition and in vitro potency, as well as cross-pathogen activity trends, have not been comprehensively examined. This review aims to address these gaps by providing a critical and integrative synthesis of the antiviral and antimicrobial properties of Erythrina species. By discussing phytoconstituent profiles, reported bioactivities, and proposed mechanisms of action, this review highlights the genus’s pharmacological importance and therapeutic potential. Ultimately, it seeks to inform and guide future translational research, supporting the advancement of Erythrina-derived compounds as promising candidates in combating infectious diseases and multidrug-resistant pathogens, as illustrated in Figure 1.
Figure 1 shows the broad spectrum of activities exhibited by Erythrina species, including antivirus, antibacterial, antifungal, and antiplasmodial effects. The most potent antiviral activity was observed in E. glauca against HIV, attributed to the active compound 3-O-Methylcalpocarpin, and in E. sigmoidea against SARS-CoV-2, linked to the compound gangetin. The most potent antibacterial activity was demonstrated by the methanol extract of E. caffra stem bark against S. aureus. In terms of antifungal activity, the ethanol stem bark extract of E. senegalensis showed the greatest potency against C. albicans. For antiplasmodial activity, the methanol root bark extract of E. sacleuxii exhibited the most significant effect against P. Falciparum.

2. Results

2.1. Antiviral Properties of Erythrina

Erythrina glauca has been reported to exhibit anti-HIV activity. Two compounds isolated from this species, sandwicensin and 3-O-methylcalopocarpin, demonstrated EC50 values of 2 µg/mL and 0.2 µg/mL, respectively [11]. These values indicate greater potency compared to compounds from Erythrina lysistemon, specifically 5-deoxyglyasperin F and 2′-hydroxyneobavaisoflavanone. These compounds exhibited significantly higher EC50 values, ranging from 11.5 µg/mL for 5-deoxyglyasperin F and 7.6 µg/mL for 2′-hydroxyneobavaisoflavanone [12]. A crude alkaloid fraction isolated from Erythrina abyssinica exhibited an EC50 value of 53 µg/mL, demonstrating lower potency compared to both E. glauca and E. lysistemon, despite the presence of bioactive compounds such as erythraline, erysodine, and related alkaloids [17]. In contrast, apigetrin from Erythrina variegata exhibited an EC50 of 100.59 µg/mL, demonstrating the least potential anti-HIV effect [10]. Notably, Erythrina senegalensis yielded diverse anti-HIV prenylated flavonoids, with 6.8-diprenylgenistein (EC50: 0.5 µM) exhibiting exceptional potency, surpassing all other compounds reported [12].
Phukhatmuen et al. [14] reported anti-SARS-CoV-2 activity in Erythrina subumbrans, isolating gangetinin as the active component, although specific EC50 values were not reported. Similarly, Nkengfack et al. [13] identified gangetin in Erythrina sigmoidea with comparable antiviral claims against SARS-CoV-2. Togola et al. [16] reported erybraedin D from Erythrina senegalensis, while Tanaka et al. [15] and Desta et al. [18], identified orientanol E from Erythrina variegata and erycaffra F from Erythrina caffra, respectively. However, in most cases, potency data (EC50 values) were not reported, limiting direct comparison.
Beyond HIV and SARS-CoV-2. other viruses have been investigated. Fahmy et al. [19] explored the anti-hepatitis viral activity of vitexin from Erythrina speciosa, which exhibited an EC50 of 125 µg/mL. In contrast, Hubert et al. [20] found significantly higher potency in compounds isolated from Erythrina senegalensis, such as 2.3-dihydro-2′-hydroxyosajin, osajin, and 6.8-diprenylgenistein, all with EC50 values around 67–72 µg/mL. These results indicate that E. senegalensis exhibits stronger anti-hepatitis viral activity than Erythrina speciosa. Another study analyzed Erythrina addisonae for anti-influenza potential and reported EC50 values ranging from 8.8 µg/mL to 26.44 µg/mL across several prenylated flavonoids, indicating considerably higher antiviral potency relative to both Erythrina speciosa and E. senegalensis [8].
Mollel et al. [21] investigated the anti-herpes simplex virus (HSV) activity of Erythrina abyssinica, noting an IC50 of 27 µg/mL from its stem bark aqueous extract. In a comparative context, Fahmy et al. [6] reported that vitexin from Erythrina speciosa showed an EC50 of 64 µg/mL, suggesting moderate activity. Pino et al. [60] evaluated Erythrina fusca and found its stem bark decoction had an EC50 of 243 ± 10.9 µg/mL, representing weaker activity among HSV-related studies. Similarly, González-Lavaut et al. [22] observed Erythrina poeppigiana to be slightly more potent than E. fusca, with an EC50 of 147.6 ± 4.3 µg/mL, yet still less active than E. abyssinica or Erythrina speciosa.
Finally, Rasool, et al. [61] reported the antiviral activity of Erythrina variegata against the Dengue virus, identifying multiple bioactive flavonoids including ericristagallin, osajin, and sigmoidins A–C. While no EC50 values were reported, the presence of these structurally diverse compounds aligns with the genus-wide trend of broad-spectrum antiviral potential observed in earlier studies. Table 1, presented below, provides a concise summary of several Erythrina species that have been tested for their antiviral activity.
The Erythrina genus is a potential inhibitor for the SARS-CoV-2 RNA-dependent RNA polymerase (RdRp), a critical enzyme for viral replication. Herlina et al. (2025) screened out 473 flavonoids from the genus Erythrina against the viral RNA-dependent RNA polymerase (RdRp) enzyme of the SARS-CoV-2 virus using computer-aided drug design [63]. The Erythrina genus, particularly butein, presents highly promising candidates for the development of SARS-CoV-2 RdRp inhibitors. Butein’s robust binding affinity, distinctive inhibitory mechanism, and favorable predicted safety profile establish a strong foundation for future experimental (in vitro) studies to substantiate its antiviral efficacy against COVID-19.
Similar to remdesivir, butein interacts with specific residues and nucleotides of the RNA primer and template of the SARS-CoV-2 virus. Figure 2 illustrates that butein is located between the primer and template RNA when interacting with amino acid residues in the palm subdomain, thereby presumably halting RNA chain elongation. As a result, this leads to inhibition of the SARS-CoV-2 RdRp activity. Nevertheless, our investigations employing pharmacophore modeling have revealed that butein necessitates modification, as it undergoes a loss of the chemical characteristics associated with hydrogen bond donor, aromatic hydrophobicity, and negative ionizability, as illustrated in Figure 2. The ionizable negative part plays a very important role in binding with magnesium ions that interact with the phosphate backbone of the diester and is part of the catalytic active site [64]. Butein lacks the necessary functional groups to compete effectively with remdesivir.

2.2. Antibacterial Activity of Erythrina

Table 2 summarizes the antibacterial activity of various Erythrina species. The methanol bark extract of E. caffra demonstrated remarkable efficacy against S. aureus, with a minimum inhibitory concentration (MIC) of 0.313 µg/mL [41]. In contrast, the ethanol bark extract of the same species showed only moderate inhibition at 39.1 µg/mL, indicating that the choice of extraction solvent significantly influences antibacterial potency [30]. Similarly, the methanol bark extract of E. abyssinica exhibited antibacterial activity with an MIC of 23 µg/mL. However, this effect was weaker compared to E. caffra, suggesting interspecies variations in phytochemical content of bioactive compounds [28].
Erybraedin, an isoflavonoid isolated from E. lysistemon, exhibited potent inhibitory activity against S. aureus (MIC: 2 µg/mL), underscoring the potential of Erythrina derivatives and isoflavonoids as antibacterial agents [35]. This finding is supported by the comparable activity of Eryvarin D from E. fusca, exhibiting comparable activity at 4 µg/mL against the same pathogen, suggesting consistency in antibacterial efficacy across species within the genus [27]. In contrast, Scandenone from E. addisonae showed weaker activity, with an MIC of 64 µg/mL. This highlights the variability in bioactivity depending on the compound class [32].
Ericristagallin, isolated from Erythrina subumbrans, exhibited remarkable activity against Methicillin-resistant S. aureus (MRSA) and Vancomycin-resistant S. aureus (VRSA), with minimum inhibitory concentrations (MICs) ranging between 0.39 and 1.56 µg/mL [65]. This inhibitory profile aligns with the findings from Erythrina zeyheri, where compounds, such as Erybraedin A and Eryzerin C, also exhibited potent activity against VRSA strains at MICs between 1.56 and 6.25 µg/mL, class-wide potency among prenylated isoflavonoids [36]. In contrast, the dichloromethane bark extract of Erythrina stricta exhibited diminished potency against MDRSA, with a minimum inhibitory concentration (MIC) of 31.25 µg/mL. This finding suggests a reduced efficacy of crude extracts in comparison to isolated compounds [23].
Furthermore, Erybraedin A from E. lysistemon exhibited consistent antibacterial activity against S. epidermidis, with a MIC of 2 µg/mL [35]. This is corroborated by the findings of reported comparable MIC values for Eryzerin C and Phaseollidin from the same species, suggesting these compounds contribute significantly to the antibacterial profile [32].
In contrast, within other bacterial groups, particularly Gram-negative pathogens, Neo-bavaisoflavone from E. sigmoidea exhibited inhibitory activity against E. coli, with minimum inhibitory concentrations (MICs) ranging from 8 to 32 µg/mL [25]. Similarly, flavanone derivatives from Erythrina livingstoniana exhibited minimum inhibitory concentrations (MICs) as low as 5 µg/mL [31]. In comparison, 6.8-diprenylgenistein and alpinumisoflavone from E. caffra with potent effects on E. coli and K. pneumoniae, showing MICs of 3.9–7.8 µg/mL [32]. These findings indicate consistent activity among prenylated flavonoids across Erythrina species against Enterobacteriaceae.
Lastly, Scandenone from E. addisonae demonstrated antibacterial activity against multiple strains, including E. faecalis, B. subtilis, and E. cloacae, with MIC values ranging from 16 to 64 µg/mL [8,32]. Although effective, these values indicate lower potency compared to compounds such as Erycristagallin or Erybraedin A. Collectively, these findings illustrate the broad-spectrum yet variable antibacterial potential of Erythrina, which is strongly influenced by the species differences, compound structure, and target pathogen.

2.3. Antifungal Activity of Erythrina

Table 3 summarizes the antifungal activity of various Erythrina species. A broad spectrum of activity has been documented, with particular efficacy against C. albicans, a common opportunistic fungal pathogen responsible for superficial infections, such as oral and vaginal candidiasis, and systemic infections in immunocompromised individuals. Among the tested extracts, the ethanol stem bark extract of E. senegalensis demonstrated the most potent inhibitory activity against C. albicans, with an MIC ranging between 4.00 and 15.63 µg/mL, indicating strong antifungal potential [42]. In contrast, other species such as E. stricta and E. lysistemon exhibited significantly higher MIC values (125 µg/mL and 25.000 µg/mL, respectively), highlighting the critical role of extraction method and plant part selection in modulating antifungal potency [23,37]. This variability in MIC underscores the influence of phytochemical diversity and compound abundance in different Erythrina taxa.
The antifungal spectrum of Erythrina extends beyond C. albicans, as several species have also exhibited inhibitory activity against other clinically relevant fungi, including C. glabrata, C. krusei, Cryptococcus neoformans, Aspergillus niger, Penicillium camemberti, Scopulariopsis brevicaulis, Pyricularia oryzae, and Rhizopus stolonifer. For example, E. crista-galli demonstrated activity against C. krusei with MIC values between 12.5 and 31.25 µg/mL, attributed to alkaloids such as erytharbine and erysotrine. Similarly, E. sacleuxii showed inhibition activity against P. oryzae through compounds such as prostratol C and orobol with a MIC of 20 µg/mL [31,44]. The identification of specific active constituents such as erypostyrene (from E. poeppigiana) and erysubin F (from E. sacleuxii) further reinforces the role of flavonoid and alkaloid subclasses within Erythrina as key contributors to its antifungal activity.
Collectively, the observed variation in antifungal efficacy, as evidenced by the wide MIC range, reflects the complexity of phytochemical interactions and highlights the need for further bioassay-guided fractionation to isolate and characterize specific bioactive compounds. These findings highlight the therapeutic potential of Erythrina as a valuable genus for the development of novel antifungal agents, particularly against resistant fungal pathogens.

2.4. Antiplasmodial Activity of Erythrina

Various Erythrina species have demonstrated activity against several Plasmodium species, including Plasmodium falciparum, the primary causative agent of malaria. Among them, the most potent compound reported is 5-hydroxysophoranone isolated from E. stricta and E. subumbrans with an IC50 of 2.5 µg/mL. The species also yielded other active compounds such as Erythrabbysin II (IC50 of 5.5 µg/mL), Soyaspongenol B (IC50; 4.6 µg/mL), and Erystagallin A (IC50; 3.8 µg/mL), which, although less potent than 5-hydroxysophoranone, still exhibited relatively low IC50 values [65]. In contrast, other Erythrina species demonstrated weaker antiplasmodial activity. For example, Lupeol isolated from E. caffra showed an IC50 of 41.7 µg/mL, while the aqueous stem bark extract of E. abbysicina had an IC50 of 47.74 µg/mL [49,78]. Beyond P. falciparum, Erythrina species have also shown activity against other Plasmodium species, as summarized in Table 4.

3. Discussion

Currently, effective treatments for infectious diseases are primarily based on well-established first-line therapies [84]. These include antiretroviral combinations for HIV, direct-acting antivirals for hepatitis, neuraminidase inhibitors for influenza, acyclovir derivatives for herpes simplex [85], β-lactams or glycopeptides for bacterial infections, azoles and polyenes for fungal diseases, and artemisinin-based combinations for malaria [86]. While these therapies have demonstrated efficacy, their utilization is frequently constrained by concerns related to resistance development, adverse effects, and accessibility. Consequently, there is an immediate need to investigate alternative sources, such as plant-derived compounds, which may present novel opportunities for complementary or replacement therapies.
The current findings reaffirm that multiple Erythrina species possess remarkable antiviral properties across a broad viral spectrum, including HIV, SARS-CoV-2, hepatitis, influenza, herpes simplex virus, and dengue virus. Among these, E. glauca displayed the most potent anti-HIV activity, with 3-O-methylcalopocarpin exhibiting an EC50 of 0.2 µg/mL, followed by sandwicensin with 2 µg/mL [11]. This result indicates a high intrinsic antiviral capacity of E. glauca constituents, especially in disrupting HIV replication and entry pathways [87]. The implication of this observation lies in the compound’s capacity to attenuate the cytopathic effects exerted by HIV on human T-lymphocytes, suggesting a possible role in host cell protection [88]. Mechanistically, this activity may be linked to the flavonoid core structure, which is known to interfere with viral enzymatic activity, particularly reverse transcriptase inhibition, or disrupt viral protein assembly by binding at critical viral-host interaction domains. This high degree of activity aligns with previously reported studies on prenylated flavonoids as antiviral agents, further confirming the structural suitability of this class in targeting retroviruses. The specificity and low EC50 values highlight the potential of Erythrina species, especially E. glauca, as a promising phytochemical source for antiretroviral drug discovery [11]. The chemical structure of 3-O-methylcalopocarpin and sandwicensin is provided in Figure 3.
A similarly strong antiviral potential is observed in E. senegalensis, particularly against hepatitis virus, where osajin demonstrated an EC50 of 67.54 µg/mL. Although less potent than anti-HIV candidates, its significance is magnified by its hepatoprotective effect via the inhibition of LDH leakage in CCl4-induced hepatotoxic models, pointing to a dual function of antiviral and organ-protective roles [89]. This duality suggests that osajin and related flavonoids may stabilize hepatocyte membranes or act as antioxidants, thereby indirectly mitigating virus-induced hepatic injury. Structurally, osajin contains prenyl side chains that may enhance its hepatic bioavailability and interaction with viral or host enzymes. These findings are consistent with literature reporting the cytoprotective action of prenylated isoflavones in models of oxidative stress and viral hepatitis, supporting its therapeutic relevance [20]. The chemical structure of osajin is provided in Figure 4a.
For the influenza virus, E. addisonae is the only species reported to exhibit activity with its compound erythradisson B, yielding a notably low EC50 of 8.8 µg/mL. The pharmacological implication is substantial, as this compound demonstrates a comparable inhibitory potential to early-generation neuraminidase inhibitors. The likely mechanism is direct inhibition of the neuraminidase enzyme, thereby preventing viral release and spread from infected epithelial cells [8]. Structurally, erythradisson B shares phenolic moieties that can engage the active site of neuraminidase via hydrogen bonding or hydrophobic interactions. This mirrors known inhibitory patterns of flavonoids in the sialidase enzyme pocket, consistent with earlier antiviral flavonoid research [90]. Chemical structure of erythradisson B is provided in Figure 4b.
The anti-herpes simplex virus activity of Erythrina abbysinica was particularly notable, with a stem bark aqueous extract achieving an IC50 of 27 µg/mL. Although not derived from a purified compound, the extract’s significant potency suggests the presence of bioactive alkaloids and coumarin-related constituents. The finding carries two key implications: first, it highlights the potential of crude plant materials in HSV inhibition; second, it emphasizes the need for compound-specific elucidation to validate the antiviral targets. Mechanistically, these constituents may inhibit HSV replication by disrupting viral DNA synthesis or glycoprotein-mediated entry, a mechanism commonly associated with erythrinan alkaloids. Compared to other plant-derived antivirals, the observed activity of E. abbysinica extract falls within a promising range for natural product leads [21].
In the antibacterial domain, Erythrina species, particularly E. caffra, demonstrated highly potent activity, with a methanol bark extract achieving a MIC of 0.313 µg/mL against S. aureus. This potency indicates a pharmacological profile competitive with first-line antibiotics, underscoring its potential relevance in combating Gram-positive bacterial infections. The significance of these findings is amplified by the persistent global threat of methicillin-resistant S. aureus (MRSA), where treatment options are limited. The underlying mechanism is likely driven by membrane disruption and enzyme inhibition, especially targeting bacterial topoisomerases, as suggested by the presence of prenylated flavonoids such as erybraedin A and phaseollidin. These molecules, due to their lipophilic side chains, preferentially integrate into the lipid bilayer of Gram-positive bacteria, causing membrane destabilization and leakage. This observation aligns with published SAR (structure–activity relationship) studies, which indicate that prenylation significantly boosts flavonoid antibacterial efficacy [28]. The chemical structure of erybraedin A and phaseollidin is provided in Figure 5.
Of particular significance is the consistent activity of Erythrina compounds against drug-resistant bacterial strains. For instance, ericristagallin from E. subumbrans exhibited minimum inhibitory concentrations (MICs) as low as 0.39 µg/mL against both MRSA and VRSA, suggesting its potential to circumvent resistance mechanisms. This activity suggests a non-classical mechanism, possibly bypassing the penicillin-binding proteins or efflux systems targeted by standard antibiotics [34]. The significance is further supported by the ability of E. stricta extracts to inhibit multidrug-resistant S. aureus at MICs of 31.25 µg/mL, reinforcing the hypothesis that these extracts contain multiple active constituents working synergistically. From a pharmacodynamic perspective, this multi-target approach is valuable as it reduces the evolutionary likelihood of resistance development, a major limitation of conventional monotherapeutic agents [36]. The structure of ericristagallin is provided in Figure 6.
In addition, the antifungal activity observed among various Erythrina species, notably E. senegalensis, underscores their clinical potential against fungal pathogens such as C. albicans, C. glabrata, and Cryptococcus neoformans. The MIC range of 4.00 to 15.63 µg/mL for E. senegalensis stem bark extract against C. albicans is within the threshold of therapeutic interest, particularly for resistant fungal strains. The pharmacological importance is elevated by the extract’s ability to disrupt biofilm formation, a known barrier to antifungal treatment. Mechanistically, bioactive compounds such as erypostyrene and erysubin F likely act through membrane destabilization or inhibition of ergosterol biosynthesis, impairing cell viability. The amphiphilic nature of these molecules facilitates their penetration into the fungal membrane, aligning with the literature on the antifungal role of prenylated pterocarpans [61,65]. The structure of erysubin F and erypostyrene is provided in Figure 7.
Finally, the antiplasmodial activity of Erythrina species provides further evidence of the genus’s broad-spectrum antimicrobial capacity. Notably, E. sacleuxii methanolic root bark extract exhibited an exceptionally low IC50 of 0.45 µg/mL against Plasmodium falciparum, and pure compounds such as phaseollidin and alpinumisoflavone demonstrated IC50 values below 2 µg/mL [69]. This indicates a high level of cytotoxic efficacy toward the parasite at low concentrations, comparable to established antimalarials. The likely mechanisms include interference with hemozoin formation, mitochondrial dysfunction, or oxidative damage induction [91]. The presence of multiple aromatic rings and phenolic hydroxyl groups in these compounds enhances their interaction with parasite biomolecules, particularly via redox cycling and mitochondrial disruption, as supported by analogous flavonoid-based antimalarial studies [92,93]. The structure of phaseollidin and alpinumisoflavone is provided in Figure 8.
Together, these findings affirm the pharmacological potential of Erythrina species as a reservoir of antiviral, antibacterial, antifungal, and antiplasmodial agents. The spectrum of activity observed across different pathogens is not only a testament to the genus’s chemical richness but also provides a strong rationale for further isolation, characterization, and preclinical evaluation of its constituents in the development of novel therapies.

4. Methodology

This review adopts a narrative-critical approach to comprehensively evaluate the antiviral and antimicrobial potential of Erythrina species, emphasizing phytochemical diversity and pharmacological relevance. A structured literature search was conducted across PubMed, Scopus, and Google Scholar up to June 2025 using targeted Boolean combinations, including “Erythrina AND antiviral,” “Erythrina AND antibacterial AND MIC,” “Erythrina AND antifungal AND MIC,” and “Erythrina AND antiplasmodial AND IC50,” alongside virus-specific queries (e.g., “HIV”, “SARS-CoV-2”). Titles, abstracts, and full texts were screened, with the top 50 entries per keyword set reviewed for eligibility. Peer-reviewed studies reporting quantitative in vitro or in vivo data on extracts or isolated compounds from Erythrina—with EC50. IC50. or MIC values were included. Non-pharmacological, non-English, or insufficiently detailed studies were excluded. Relevant data were extracted; categorized by pathogen type, species, and compound; and synthesized to identify mechanistic trends and comparative efficacy. The literature selection process is visually summarized in Figure 9, which provides a transparent overview of article identification, screening, and inclusion. While not adhering to PRISMA due to the narrative scope, the methodology reflects academic rigor, analytical coherence, and thematic precision in evaluating the phytotherapeutic promise of Erythrina in infectious disease contexts.

5. Conclusions

This comprehensive review systematically compiles and analyzes data across a wide spectrum of infectious targets to elucidate the antiviral, antibacterial, antifungal, and antiplasmodial potentials of the Erythrina genus. The literature demonstrates that multiple Erythrina species exhibit remarkable and often species-specific activities against clinically significant pathogens, including HIV, SARS-CoV-2, S. aureus, C. albicans, and P. falcipa-rum. These effects are primarily attributed to structurally diverse phytoconstituents, such as prenylated flavonoids, pterocarpans, and erythrina alkaloids, which act through mechanisms including enzymatic inhibition, membrane disruption, and modulation of microbial virulence. Notably, the genus consistently displays inhibitory profiles against multidrug-resistant bacteria and biofilm-forming fungi, positioning Erythrina-derived compounds as promising candidates for next-generation anti-infective development.

Author Contributions

Conceptualization, M.M., S.L., M.A.F. and A.R.; methodology, M.M., S.L., M.A.F. and A.R.; data curation, S.L. and M.A.F.; writing—original draft preparation, M.M., S.L., M.A.F., A.R. and S.S.; writing—review and editing, M.M., A.R., S.S., S.M., N.K.K.I. and A.S.; visualization, M.M., S.L., M.A.F. and A.R.; supervision, M.M., S.M., N.K.K.I. and A.S.; project administration, M.M.; funding acquisition, M.M. All authors have read and agreed to the published version of the manuscript.

Funding

The funding was secured through the Literature Review Grant from Research Grant Universitas Padjadjaran (HRU), designated as no. 2097/UN6.0/TU.00/2025.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

We extend our sincere gratitude to Universitas Padjadjaran for the financial support provided for this research and APC for publication. We also acknowledge the Fundamental Research Grant Scheme (FP034-2018A) and the SATU Joint Research Scheme (ST044-2023), which were awarded to N.K.K.I.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. WHO. Infectious Diseases. Available online: https://www.emro.who.int/health-topics/infectious-diseases/index.html (accessed on 11 September 2025).
  2. WHO. Antimicrobial Resistance. Available online: https://www.who.int/news-room/fact-sheets/detail/antimicrobial-resistance (accessed on 11 September 2025).
  3. Ventola, C.L. The antibiotic resistance crisis: Part 1: Causes and threats. Pharm. Ther. 2015, 40, 277–283. [Google Scholar]
  4. Newman, D.J.; Cragg, G.M. Natural Products as Sources of New Drugs over the Nearly Four Decades from 01/1981 to 09/2019. J. Nat. Prod. 2020, 83, 770–803. [Google Scholar] [CrossRef]
  5. Enoma, S.; Adewole, T.S.; Agunbiade, T.O.; Kuku, A. Antimicrobial Activities and Phylogenetic Study of Erythrina senegalensis DC (Fabaceae) Seed lectin. BioTechnologia 2023, 104, 21–32. [Google Scholar] [CrossRef] [PubMed]
  6. Fahmy, N.M.; Al-Sayed, E.; El-Shazly, M.; Singab, A.N. Comprehensive review on flavonoids biological activities of Erythrina plant species. Ind. Crops Prod. 2018, 123, 500–538. [Google Scholar] [CrossRef]
  7. Herlina, T.; Rizaldi Akili, A.W.; Nishinarizki, V.; Hardianto, A.; Latip, J.B. Review on antibacterial flavonoids from genus Erythrina: Structure-activity relationship and mode of action. Heliyon 2025, 11, e41395. [Google Scholar] [CrossRef]
  8. Nguyen, P.H.; Na, M.; Dao, T.T.; Ndinteh, D.T.; Mbafor, J.T.; Park, J.; Cheong, H.; Oh, W.K. New stilbenoid with inhibitory activity on viral neuraminidases from Erythrina addisoniae. Bioorganic Med. Chem. Lett. 2010, 20, 6430–6434. [Google Scholar] [CrossRef]
  9. Kaushal, A.; Sharma, M.; Navneet; Sharma, M. Ethnomedicinal, phytochemical, therapeutic and pharmacological review of the genus Erythrina. Int. J. Bot. Stud. 2020, 5, 642–648. [Google Scholar]
  10. Lee, J.; Oh, W.; Ahn, J.; Kim, Y.; Mbafor, J.; Wandji, J.; Fomum, Z. Prenylisoflavonoids from Erythrina senegalensis as Novel HIV-1 Protease Inhibitors. Planta Medica 2008, 75, 268–270. [Google Scholar] [CrossRef] [PubMed]
  11. McKee, T.C.; Bokesch, H.R.; McCormick, J.L.; Rashid, M.A.; Spielvogel, D.; Gustafson, K.R.; Alavanja, M.M.; Cardellina, J.H.; Boyd, M.R. Isolation and Characterization of New Anti-HIV and Cytotoxic Leads from Plants, Marine, and Microbial Organisms. J. Nat. Prod. 1997, 60, 431–438. [Google Scholar] [CrossRef]
  12. Mohammed, M.M.D.; Ibrahim, N.A.; Awad, N.E.; Matloub, A.A.; Mohamed-Ali, A.G.; Barakat, E.E.; Mohamed, A.E.; Colla, P.L. Anti-HIV-1 and cytotoxicity of the alkaloids of Erythrina abyssinica Lam. growing in Sudan. Nat. Prod. Res. 2012, 26, 1565–1575. [Google Scholar] [CrossRef]
  13. Nkengfack, A.E.; Azebaze, A.G.B.; Waffo, A.K.; Fomum, Z.T.; Meyer, M.; van Heerden, F.R. Cytotoxic isoflavones from Erythrina indica. Phytochemistry 2001, 58, 1113–1120. [Google Scholar] [CrossRef]
  14. Phukhatmuen, P.; Meesakul, P.; Suthiphasilp, V.; Charoensup, R.; Maneerat, T.; Cheenpracha, S.; Limtharakul, T.; Pyne, S.G.; Laphookhieo, S. Antidiabetic and antimicrobial flavonoids from the twigs and roots of Erythrina subumbrans (Hassk.) Merr. Heliyon 2021, 7, e06904. [Google Scholar] [CrossRef]
  15. Tanaka, H.; Sudo, M.; Kawamura, T.; Sato, M.; Yamaguchi, R.; Fukai, T.; Sakai, E.; Tanaka, N. Antibacterial Constituents from the Roots of Erythrina herbacea against Methicillin-resistant Staphylococcus aureus. Planta Medica 2010, 76, 916–919. [Google Scholar] [CrossRef]
  16. Togola, A.; Hedding, B.; Theis, A.; Wangensteen, H.; Rise, F.; Smestad Paulsen, B.; Diallo, D.; Egil Malterud, K. 15-Lipoxygenase Inhibitory Effects of Prenylated Flavonoids from Erythrina senegalensis. Planta Medica 2009, 75, 1168–1170. [Google Scholar] [CrossRef] [PubMed]
  17. Wardani, A.K.; Mun’im, A.; Yanuar, A. Inhibition of HIV-1 Reverse Transcriptase of Selected Indonesia Medicinal Plants and Isolation of the Inhibitor from Erythrina variegata L. Leaves. J. Young Pharm. 2018, 10, 169–172. [Google Scholar] [CrossRef]
  18. Desta, Z.Y.; Sewald, N.; Majinda, R.R.T. New flavonoids from the stem bark of Erythrina caffra Thunb. Nat. Prod. Res. 2014, 28, 667–673. [Google Scholar] [CrossRef] [PubMed]
  19. Fahmy, N.M.; Al-Sayed, E.; Moghannem, S.; Azam, F.; El-Shazly, M.; Singab, A.N. Breaking Down the Barriers to a Natural Antiviral Agent: Antiviral Activity and Molecular Docking of Erythrina speciosa Extract, Fractions, and the Major Compound. Chem. Biodivers. 2020, 17, e1900511. [Google Scholar] [CrossRef]
  20. Hubert, D.J.; Dawe, A.; Florence, N.T.; Gilbert, K.D.W.F.; Angele, T.N.; Buonocore, D.; Finzi, P.V.; Vidari, G.; Bonaventure, N.T.; Marzatico, F.; et al. In vitro hepatoprotective and antioxidant activities of crude extract and isolated compounds from Ficus gnaphalocarpa. Inflammopharmacology 2010, 19, 35–43. [Google Scholar] [CrossRef]
  21. Mollel, J.T.; Said, J.S.; Masalu, R.J.; Hannoun, C.; Mbunde, M.V.N.; Nondo, R.S.O.; Bergström, T.; Trybala, E. Anti-respiratory syncytial virus and anti-herpes simplex virus activity of six Tanzanian medicinal plants with extended studies of Erythrina abyssinica stem bark. J. Ethnopharmacol. 2022, 292, 115204. [Google Scholar] [CrossRef]
  22. González-Lavaut, J.A.; Prieto-González, S.; Garrido-Garrido, G.; García, M.; González-Guevara, J.L.; González-García, K.L. Antiviral activity of Cuban vegetable species. Pharmacologyonline 2006, 3, 527–530. [Google Scholar]
  23. Akter, K.; Barnes, E.C.; Loa-Kum-Cheung, W.L.; Yin, P.; Kichu, M.; Brophy, J.J.; Barrow, R.A.; Imchen, I.; Vemulpad, S.R.; Jamie, J.F. Antimicrobial and antioxidant activity and chemical characterisation of Erythrina stricta Roxb. (Fabaceae). J. Ethnopharmacol. 2016, 185, 171–181. [Google Scholar] [CrossRef] [PubMed]
  24. Chukwujekwu, J.C.; Van Heerden, F.R.; Van Staden, J. Antibacterial activity of flavonoids from the stem bark of Erythrina caffra thunb. Phytother. Res. 2010, 25, 46–48. [Google Scholar] [CrossRef]
  25. Djeussi, D.E.; Sandjo, L.P.; Noumedem, J.A.K.; Omosa, L.K.; T. Ngadjui, B.; Kuete, V. Antibacterial activities of the methanol extracts and compounds from Erythrina sigmoidea against Gram-negative multi-drug resistant phenotypes. BMC Complement. Altern. Med. 2015, 15, 453. [Google Scholar] [CrossRef] [PubMed]
  26. Ilodigwe, E. Wound Healing Activity of Ethanol Leaf Extract of Erythrina senegalensis. Br. J. Pharm. Res. 2014, 4, 531–540. [Google Scholar] [CrossRef]
  27. Innok, P.; Rukachaisirikul, T.; Phongpaichit, S.; Suksamrarn, A. Fuscacarpans A–C, new pterocarpans from the stems of Erythrina fusca. Fitoterapia 2010, 81, 518–523. [Google Scholar] [CrossRef]
  28. Jambwa, P.; Nkadimeng, S.M.; Mudimba, T.N.; Matope, G.; McGaw, L.J. Antibacterial and anti-inflammatory activity of plant species used in traditional poultry ethnomedicine in Zimbabwe: A first step to developing alternatives to antibiotic poultry feed additives. J. Ethnopharmacol. 2023, 300, 115687. [Google Scholar] [CrossRef]
  29. Kwesiga, G.; Kelling, A.; Kersting, S.; Sperlich, E.; von Nickisch-Rosenegk, M.; Schmidt, B. Total Syntheses of Prenylated Isoflavones from Erythrina sacleuxii and Their Antibacterial Activity: 5-Deoxy-3′-prenylbiochanin A and Erysubin F. J. Nat. Prod. 2020, 83, 3445–3453. [Google Scholar] [CrossRef]
  30. Olufunmiso, O.; Afolayan, A. In Vitro Antibacterial and Time-Kill Evaluation of the Erythrina caffra Thunb. Extract against Bacteria Associated with Diarrhoea. Sci. World J. 2012, 2012, 738314. [Google Scholar] [CrossRef]
  31. Ombito, J.O.; Bojase, G.; Majinda, R.; B. Masesane, I.; Schüffler, A.; Pusch, S.; Weber, C.; Opatz, T. Chemical constituents of the root wood of Erythrina sacleuxii and determination of the absolute configuration of suberectin. Bull. Chem. Soc. Ethiop. 2020, 34, 135–140. [Google Scholar] [CrossRef]
  32. Peleyeju, G.B.; Emmanuel, T.; Tata, C.M.; Djuidje Fotsing, M.C.; Niemann, N.; Rhyman, L.; Arderne, C.; Ndinteh, D.T.; Ramasami, P. Crystal structure and antibacterial activity of scandenone (warangalone) from Erythrina plants. J. Mol. Struct. 2019, 1191, 43–51. [Google Scholar] [CrossRef]
  33. Roumy, V.; Gutierrez-Choquevilca, A.-L.; Lopez Mesia, J.; Ruiz, L.; Ruiz Macedo, J.; Abedini, A.; Landoulsi, A.; Samaillie, J.; Hennebelle, T.; Rivière, C.; et al. In vitro antimicrobial activity of traditional plant used in mestizo shamanism from the Peruvian amazon in case of infectious diseases. Pharmacogn. Mag. 2015, 11, 625. [Google Scholar] [CrossRef]
  34. Rukachaisirikul, T.; Innok, P.; Suksamrarn, A. Erythrina Alkaloids and a Pterocarpan from the Bark of Erythrina subumbrans. J. Nat. Prod. 2008, 71, 156–158. [Google Scholar] [CrossRef]
  35. Sadgrove, N.J.; Oliveira, T.B.; Khumalo, G.P.; Vuuren, S.F.V.; van Wyk, B.E. Antimicrobial Isoflavones and Derivatives from Erythrina (Fabaceae): Structure Activity Perspective (Sar & Qsar) on Experimental and Mined Values Against Staphylococcus aureus. Antibiotics 2020, 9, 223. [Google Scholar] [CrossRef]
  36. Sato, M.; Tanaka, H.; Oh-Uchi, T.; Fukai, T.; Etoh, H.; Yamaguchi, R. Antibacterial activity of phytochemicals isolated from Erythrina zeyheri against vancomycin-resistant enterococci and their combinations with vancomycin. Phytother. Res. 2004, 18, 906–910. [Google Scholar] [CrossRef] [PubMed]
  37. Sato, M.; Tanaka, H.; Yamaguchi, R.; Oh-Uchi, T.; Etoh, H. Erythrina poeppigiana-derived phytochemical exhibiting antimicrobial activity against Candida albicans and methicillin-resistant Staphylococcus aureus. Lett. Appl. Microbiol. 2003, 37, 81–85. [Google Scholar] [CrossRef]
  38. Schultz, F.; Osuji, O.F.; Wack, B.; Anywar, G.; Garbe, L.-A. Antiinflammatory Medicinal Plants from the Ugandan Greater Mpigi Region Act as Potent Inhibitors in the COX-2/PGH2 Pathway. Plants 2021, 10, 351. [Google Scholar] [CrossRef] [PubMed]
  39. Tanaka, H.; Atsumi, I.; Shirota, O.; Sekita, S.; Sakai, E.; Sato, M.; Murata, J.; Murata, H.; Darnaedi, D.; Chen, I.S. Three New Constituents from the Roots of Erythrina variegata and Their Antibacterial Activity against Methicillin-Resistant Staphylococcus aureus. Chem. Biodivers. 2011, 8, 476–482. [Google Scholar] [CrossRef]
  40. Tanaka, H.; Sato, M.; Oh-Uchi, T.; Yamaguchi, R.; Etoh, H.; Shimizu, H.; Sako, M.; Takeuchi, H. Antibacterial properties of a new isoflavonoid from Erythrina poeppigiana against methicillin-resistant Staphylococcus aureus. Phytomedicine 2004, 11, 331–337. [Google Scholar] [CrossRef]
  41. Wintola, O.; Olufunmiso, O.; Afolayan, A. Brine Shrimps Toxicity and In vitro Antibacterial Potentials of the Crude Methanolic Stem Bark Extract of Erythrina caffra thunb. against Bacteria in Infections. 2015, 8, 4325–4334. [Google Scholar]
  42. Harley, B.K.; Quagraine, A.M.; Neglo, D.; Aggrey, M.O.; Orman, E.; Mireku-Gyimah, N.A.; Amengor, C.D.; Jato, J.; Saaka, Y.; Fleischer, T.C. Metabolite profiling, antifungal, biofilm formation prevention and disruption of mature biofilm activities of Erythrina senegalensis stem bark extract against Candida albicans and Candida glabrata. PLoS ONE 2022, 17, e0278096. [Google Scholar] [CrossRef]
  43. Sivalingam, A.M.; Pandian, A. Identification and characterization of silver nanoparticles from Erythrina indica and its antioxidant and Uropathogenic antimicrobial properties. Microb. Pathog. 2024, 190, 106635. [Google Scholar] [CrossRef]
  44. de Ávila, J.M.; Dalcol, I.I.; Pereira, A.O.; Santos, E.W.; Ferraz, A.; Santos, M.Z.; Mostardeiro, M.A.; Morel, A.F. Antimicrobial Evaluation of Erythrinan Alkaloids from Erythrina cristagalli L. Med. Chem. 2018, 14, 784–790. [Google Scholar] [CrossRef]
  45. Motsei, M.L.; Lindsey, K.L.; van Staden, J.; Jäger, A.K. Screening of traditionally used South African plants for antifungal activity against Candida albicans. J. Ethnopharmacol. 2003, 86, 235–241. [Google Scholar] [CrossRef] [PubMed]
  46. Ousmane, C.; Cyrille, G.K.R.; Lassina, S.P.; Karamoko, O. Antifungal Activities of Erythrina senegalensis Leaves Partitioned Extracts on the Germs Responsible for Opportunistic Cryptococcosis of HIV/AIDS. Sch. Acad. J. Biosci. 2023, 11, 455–460. [Google Scholar] [CrossRef]
  47. Surapuram, V.; Setzer, W.N.; McFeeters, R.L.; McFeeters, H. Antifungal Activity of Plant Extracts against Aspergillus niger and Rhizopus stolonifer. Nat. Product. Commun. 2014, 9, 1603–1605. [Google Scholar] [CrossRef]
  48. Begum, S.; Munissi, J.J.E.; Buriyo, A.S.; Makangara, J.J.; Lucantoni, L.; Avery, V.M.; Erdelyi, M.; Nyandoro, S.S. Antiplasmodial, Antimicrobial and Cytotoxic Activities of Extracts from Selected Medicinal Plants Growing in Tanzania. J. Biol. Act. Prod. Nat. 2020, 10, 165–176. [Google Scholar] [CrossRef]
  49. Waiganjo, B.; Moriasi, G.; Onyancha, J.; Elias, N.; Muregi, F. Antiplasmodial and Cytotoxic Activities of Extracts of Selected Medicinal Plants Used to Treat Malaria in Embu County, Kenya. J. Parasitol. Res. 2020, 2020, 1–12. [Google Scholar] [CrossRef]
  50. Zarev, Y.; Foubert, K.; Cos, P.; Maes, L.; Elgorashi, E.; Apers, S.; Ionkova, I.; Pieters, L. HPLC-DAD-SPE-NMR isolation of tetracyclic spiro-alkaloids with antiplasmodial activity from the seeds of Erythrina latissima. Nat. Prod. Res. 2019, 34, 1037–1040. [Google Scholar] [CrossRef]
  51. Andayi, A.W.; Yenesew, A.; Derese, S.; Midiwo, J.O.; Gitu, P.M.; Jondiko, O.J.; Akala, H.; Liyala, P.; Wangui, J.; Waters, N.C.; et al. Antiplasmodial Flavonoids from Erythrina sacleuxii. Planta Medica 2006, 72, 187–189. [Google Scholar] [CrossRef]
  52. Tjahjandarie, T.S.; Tanjung, M.; Rahmania, D.F.; Rhidoma, C.I.; Saputri, R.D. Calodioscurins A and B, two new isoprenylated xanthones from the stem bark of Calophyllum dioscurii P.F. Stevens. Nat. Prod. Res. 2019, 35, 1153–1158. [Google Scholar] [CrossRef]
  53. Jemimah Sandra, T.N.; Christelle Nadia, N.A.; Cedric, Y.; Guy-Armand, G.N.; Azizi, M.A.; Aboubakar Sidiki, N.N.; Alex Kevin, T.D.; Payne, V.K.; Hu, H. In vitro and in vivo antimalarial activities of the ethanol extract of Erythrina sigmoidea stem bark used for the treatment of malaria in the Western Region of Cameroon. Front. Parasitol. 2024, 3, 1359442. [Google Scholar] [CrossRef]
  54. Kamanzi Atindehou, K.; Schmid, C.; Brun, R.; Koné, M.W.; Traore, D. Antitrypanosomal and antiplasmodial activity of medicinal plants from Côte d’Ivoire. J. Ethnopharmacol. 2004, 90, 221–227. [Google Scholar] [CrossRef]
  55. Sazed, S.A.; Islam, O.; Bliese, S.L.; Hossainey, M.R.H.; Soma, M.A.; Rashid, M.A.; Rahman, M.S.; Alam, M.S. Phytochemical, Biological and Computational Investigations of Erythrina fusca Lour. to Assess Antimalarial Property against Plasmodium falciparum. Preprints 2020, 2020100576. [Google Scholar] [CrossRef]
  56. Yenesew, A.; Akala, H.M.; Twinomuhwezi, H.; Chepkirui, C.; Irungu, B.N.; Eyase, F.L.; Kamatenesi-Mugisha, M.; Kiremire, B.T.; Johnson, J.D.; Waters, N.C. The antiplasmodial and radical scavenging activities of flavonoids of Erythrina burttii. Acta Trop. 2012, 123, 123–127. [Google Scholar] [CrossRef] [PubMed]
  57. Aboelmagd, m.; Said, A.; Ross, S.; Haggag, E. A Novel Compound and Biological Evaluation of Phytoconstituents Isolated from Erythrina corallodendron L. Flowers. J. Adv. Pharm. Res. 2018, 2, 247–255. [Google Scholar] [CrossRef]
  58. Christopher, R.; Msonga, A.; Hoppe, H.C.; Boyom, F.F. Ethanol Extracts from Selected Tanzanian Medicinal Plants Selectively Inhibit Plasmodium falciparum Growth In Vitro. Tanzan. J. Sci. 2023, 49, 41–47. [Google Scholar] [CrossRef]
  59. Tati, H.; Euis, J.; Nurlelasari, N.; Dikdik, K.; Unang, S. Potential of Dadap Ayam (Erythrina variegata) Plant as Herbal Medicine. J. Med. Planta 2011, 1, 245902. [Google Scholar]
  60. Pino, S.; González, J.; González, J.; Garrido, G.; García, M.; Carballo, M.; Echemendía, O.; Urquiola, A.; Rastrelli, L.; Molina-Torres, J.; et al. Preliminary phytochemical screening and in vitro antiherpetic activity of Erythrina fusca Lour. Am. J. Pharm. 2004, 23, 453–458. [Google Scholar]
  61. Rasool, N.; Ashraf, A.; Waseem, M.; Hussain, W.; Mahmood, S. Computational exploration of antiviral activity of phytochemicals against NS2B/NS3 proteases from dengue virus. Turk. J. Biochem. 2018, 44, 261–277. [Google Scholar] [CrossRef]
  62. Tanaka, H.; Atsumi, I.; Hasegawa, M.; Hirata, M.; Sakai, T.; Sato, M.; Yamaguchi, R.; Tateishi, Y.; Tanaka, T.; Fukai, T. Two New Isoflavanones from the Roots of Erythrina variegata. Nat. Prod. Commun. 2015, 10, 499–501. [Google Scholar] [CrossRef]
  63. Herlina, T.; Nishinarizki, V.; Akili, A.W.R.; Hardianto, A.; Gaffar, S.; Muchtaridi, M.; Latip, J. Exploring Erythrina flavonoids as potential SARS-CoV-2 RdRp inhibitors through virtual screening, in silico ADMET evaluation, and molecular dynamics simulation studies. Sci. Rep. 2025, 15, 14259. [Google Scholar] [CrossRef] [PubMed]
  64. Yin, W.; Mao, C.; Luan, X.; Shen, D.D.; Shen, Q.; Su, H.; Wang, X.; Zhou, F.; Zhao, W.; Gao, M.; et al. Structural basis for inhibition of the RNA-dependent RNA polymerase from SARS-CoV-2 by remdesivir. Science 2020, 368, 1499–1504. [Google Scholar] [CrossRef] [PubMed]
  65. Rukachaisirikul, T.; Saekee, A.; Tharibun, C.; Watkuolham, S.; Suksamrarn, A. Biological Activities of the Chemical Constituents of Erythrina stricta and Erythrina subumbrans. Arch. Pharmacal Res. 2007, 30, 1398–1403. [Google Scholar] [CrossRef]
  66. Sato, M.; Tanaka, H.; Tani, N.; Nagayama, M.; Yamaguchi, R. Different antibacterial actions of isoflavones isolated from Erythrina poeppigiana against methicillin-resistant Staphylococcus aureus. Lett. Appl. Microbiol. 2006, 43, 243–248. [Google Scholar] [CrossRef]
  67. Sato, M.; Tanaka, H.; Yamaguchi, R.; Kato, K.; Etoh, H. Synergistic effects of mupirocin and an isoflavanone isolated from Erythrina variegata on growth and recovery of methicillin-resistant Staphylococcus aureus. Int. J. Antimicrob. Agents 2004, 24, 241–246. [Google Scholar] [CrossRef]
  68. Tanaka, H.; Hirata, M.; Etoh, H.; Sako, M.; Sato, M.; Murata, J.; Murata, H.; Darnaedi, D.; Fukai, T. Six New Constituents from the Roots of Erythrina variegata. Chem. Biodivers. 2004, 1, 1101–1108. [Google Scholar] [CrossRef]
  69. Khumalo, G.P.; Sadgrove, N.J.; Van Vuuren, S.F.; Van Wyk, B.-E. South Africa’s Best BARK Medicines Prescribed at the Johannesburg Muthi Markets for Skin, Gut, and Lung Infections: MIC’s and Brine Shrimp Lethality. Antibiotics 2021, 10, 681. [Google Scholar] [CrossRef]
  70. Sato, M.; Tanaka, H.; Fujiwara, S.; Hirata, M.; Yamaguchi, R.; Etoh, H.; Tokuda, C. Antibacterial property of isoflavonoids isolated from Erythrina variegata against cariogenic oral bacteria. Phytomedicine 2003, 10, 427–433. [Google Scholar] [CrossRef]
  71. Dzoyem, J.P.; McGaw, L.J.; Eloff, J.N. In vitro antibacterial, antioxidant and cytotoxic activity of acetone leaf extracts of nine under-investigated Fabaceae tree species leads to potentially useful extracts in animal health and productivity. BMC Complement. Altern. Med. 2014, 14, 147. [Google Scholar] [CrossRef]
  72. Redko, F.; Clavin, M.L.; Weber, D.; Ranea, F.; Anke, T.; Martino, V. Antimicrobial Isoflavonoids from Erythrina crista galli Infected with Phomopsis sp. Z. Für Naturforschung C 2007, 62, 164–168. [Google Scholar] [CrossRef]
  73. Bunalema, L.; Kirimuhuzya, C.; Tabuti, J.R.; Waako, P.; Magadula, J.J.; Otieno, N.; Orodho, J.A.; Okemo, P. The efficacy of the crude root bark extracts of Erythrina abyssinica on rifampicin resistant Mycobacterium tuberculosis. Afr. Health Sci. 2011, 11, 587–593. [Google Scholar]
  74. Mabona, U.; Viljoen, A.; Shikanga, E.; Marston, A.; Van Vuuren, S. Antimicrobial activity of southern African medicinal plants with dermatological relevance: From an ethnopharmacological screening approach, to combination studies and the isolation of a bioactive compound. J. Ethnopharmacol. 2013, 148, 45–55. [Google Scholar] [CrossRef]
  75. Bedane, K.G.; Kusari, S.; Eckelmann, D.; Masesane, I.B.; Spiteller, M.; Majinda, R.R.T. Erylivingstone A–C with antioxidant and antibacterial activities from Erythrina livingstoniana. Fitoterapia 2015, 105, 113–118. [Google Scholar] [CrossRef]
  76. Kgakatsi, N.A.; Majinda, R.R.T.; Masesane, I.B.; Nwamadi, M.S.; Demissie, T.B.; Ombito, J.O.; Gobe, I. New isoflavan from Erythrina livingstoniana. Nat. Prod. Res. 2022, 38, 493–502. [Google Scholar] [CrossRef] [PubMed]
  77. Tadjong Tcho, A.; Djouaka Bavoua, J.L.; Ngouonpe Wembe, A.; Gompe Bobda, E.G.; Majoumo, F.; Leuche, J.M.; Ndonfack Tiofack, E.; Mbah, J.A.; Toze, F.A.A. New flavanone and other constituents from Erythrina senegalensis A. DC. (fabaceae). Nat. Product. Res. 2024, 1–11. [Google Scholar] [CrossRef] [PubMed]
  78. Chukwujekwu, J.C.; de Kock, C.A.; Smith, P.J.; Van Heerden, F.R.; Van Staden, J. Antiplasmodial activity of compounds isolated from Erythrina caffra. South Afr. J. Bot. 2016, 106, 101–103. [Google Scholar] [CrossRef]
  79. Nondo, R.S.O.; Moshi, M.J.; Erasto, P.; Masimba, P.J.; Machumi, F.; Kidukuli, A.W.; Heydenreich, M.; Zofou, D. Anti-plasmodial activity of Norcaesalpin D and extracts of four medicinal plants used traditionally for treatment of malaria. BMC Complement. Altern. Med. 2017, 17, 167. [Google Scholar] [CrossRef]
  80. Prozesky, E.A.; Meyer, J.J.M.; Louw, A.I. In vitro antiplasmodial activity and cytotoxicity of ethnobotanically selected South African plants. J. Ethnopharmacol. 2001, 76, 239–245. [Google Scholar] [CrossRef]
  81. Ajaiyeoba, E.; Ashidi, J.; Abiodun, O.; Okpako, L.; Ogbole, O.; Akinboye, D.; Falade, C.; Bolaji, O.; Gbotosho, G.; Falade, M.; et al. Antimalarial Ethnobotany: In Vitro. Antiplasmodial Activity of Seven Plants Identified in the Nigerian Middle Belt. Pharm. Biol. 2005, 42, 588–591. [Google Scholar] [CrossRef]
  82. Ali, H.; König, G.M.; Khalid, S.A.; Wright, A.D.; Kaminsky, R. Evaluation of selected Sudanese medicinal plants for their in vitro activity against hemoflagellates, selected bacteria, HIV-1-RT and tyrosine kinase inhibitory, and for cytotoxicity. J. Ethnopharmacol. 2002, 83, 219–228. [Google Scholar] [CrossRef]
  83. Onyango, D.W.; Midiwo, J.O. In vivo Evaluation of Anti-malarial Activity of Stem and Root Extracts of Erythrina abyssinica. Eur. J. Med. Plants 2019, 27, 1–5. [Google Scholar] [CrossRef]
  84. Halloran, M.E.; Longini, I.M., Jr. Emerging, evolving, and established infectious diseases and interventions. Science 2014, 345, 1292–1294. [Google Scholar] [CrossRef]
  85. Shyr, Z.A.; Cheng, Y.S.; Lo, D.C.; Zheng, W. Drug combination therapy for emerging viral diseases. Drug Discov. Today 2021, 26, 2367–2376. [Google Scholar] [CrossRef] [PubMed]
  86. Vere Hodge, R.A.; Field, H.J. Antiviral agents for herpes simplex virus. Adv. Pharmacol. 2013, 67, 1–38. [Google Scholar] [CrossRef] [PubMed]
  87. Izquierdo-Pujol, J.; Puertas, M.C.; Martinez-Picado, J.; Morón-López, S. Targeting Viral Transcription for HIV Cure Strategies. Microorganisms 2024, 12, 752. [Google Scholar] [CrossRef] [PubMed]
  88. Vidya Vijayan, K.K.; Karthigeyan, K.P.; Tripathi, S.P.; Hanna, L.E. Pathophysiology of CD4+ T-Cell Depletion in HIV-1 and HIV-2 Infections. Front. Immunol. 2017, 8, 580. [Google Scholar] [CrossRef]
  89. Chen, P.; Zou, F.; Liu, W. Recent advancement in prevention against hepatotoxicity, molecular mechanisms, and bioavailability of gallic acid, a natural phenolic compound: Challenges and perspectives. Front. Pharmacol. 2025, 16, 1549526. [Google Scholar] [CrossRef]
  90. Muchtaridi, M.; Nuwarda, R.F.; Ikram, E.H.K.; Abdul Rahim, A.S.; Gazzali, A.M.; Wahab, H.A. Neuraminidase Inhibitor of Garcinia atroviridis L. Fruits and Leaves Using Partial Purification and Molecular Characterization. Molecules 2022, 27, 949. [Google Scholar] [CrossRef]
  91. Ariefta, N.R.; Narita, K.; Murata, T.; Nishikawa, Y. Evaluation of the antiplasmodial efficacy of synthetic 2,5-diphenyloxazole analogs of compounds naturally derived from Oxytropis lanata. Int. J. Parasitol. Drugs Drug Resist. 2024, 25, 100540. [Google Scholar] [CrossRef]
  92. Hodnick, W.F.; Milosavljević, E.B.; Nelson, J.H.; Pardini, R.S. Electrochemistry of flavonoids. Relationships between redox potentials, inhibition of mitochondrial respiration, and production of oxygen radicals by flavonoids. Biochem. Pharmacol. 1988, 37, 2607–2611. [Google Scholar] [CrossRef]
  93. Pal, C. Redox modulating small molecules having antimalarial efficacy. Biochem. Pharmacol. 2023, 218, 115927. [Google Scholar] [CrossRef]
Figure 1. Review for the most potent Erythrina sp. as an antivirus, antibacterial, antifungal, and antiplasmodial agent.
Figure 1. Review for the most potent Erythrina sp. as an antivirus, antibacterial, antifungal, and antiplasmodial agent.
Plants 14 03053 g001
Figure 2. Molecular docking simulation of butein (blue carbon) and remdesivir (gray carbon) by LigandScout 4.2 (under license acquired from Universitas Padjadjaran). Magenta highlights: negative ionizable; green highlights: hydrogen bond donor; red highlights: hydrogen bond acceptor; and blue highlights: ring aromatic hydrophobicity.
Figure 2. Molecular docking simulation of butein (blue carbon) and remdesivir (gray carbon) by LigandScout 4.2 (under license acquired from Universitas Padjadjaran). Magenta highlights: negative ionizable; green highlights: hydrogen bond donor; red highlights: hydrogen bond acceptor; and blue highlights: ring aromatic hydrophobicity.
Plants 14 03053 g002
Figure 3. Chemical structure of (a) 3-O-methylcalopocarpin and (b) sandwicensin.
Figure 3. Chemical structure of (a) 3-O-methylcalopocarpin and (b) sandwicensin.
Plants 14 03053 g003
Figure 4. Chemical structure of (a) Osajin and (b) Erythradisson B.
Figure 4. Chemical structure of (a) Osajin and (b) Erythradisson B.
Plants 14 03053 g004
Figure 5. Chemical structure of (a) erybraedin A and (b) phaseollidin.
Figure 5. Chemical structure of (a) erybraedin A and (b) phaseollidin.
Plants 14 03053 g005
Figure 6. Chemical structure of ericristagallin.
Figure 6. Chemical structure of ericristagallin.
Plants 14 03053 g006
Figure 7. Chemical structure of (a) erysubin F and (b) erypostyrene.
Figure 7. Chemical structure of (a) erysubin F and (b) erypostyrene.
Plants 14 03053 g007
Figure 8. Chemical structure of (a) phaseollidin and (b) alpinumisoflavone.
Figure 8. Chemical structure of (a) phaseollidin and (b) alpinumisoflavone.
Plants 14 03053 g008
Figure 9. Flow chart of methodology.
Figure 9. Flow chart of methodology.
Plants 14 03053 g009
Table 1. Antiviral activity of Erythrina.
Table 1. Antiviral activity of Erythrina.
NoSpeciesCompounds/ExtractsVirusRef.
1E. glaucaSandwicensin (EC50: 2 µg/mL)
3-O-methylcalopocarpin (EC50: 0.2 µg/mL)
HIV[11]
2E. lysistemon5-deoxyglyasperin F (EC50: 11.5 µg/mL)
2′-hydroxyneobavaisoflavanone (EC50: 7.6 µg/mL)
HIV[11]
3E. abbysinicaCrude alkaloid fraction (EC50: 53 µg/mL). The compounds identified were erythraline, erysodine, erysotrine, 8-oxoerythraline, and 11-methoxyerysodineHIV[12]
4E. variegataApigetrin (EC50: 100.59 µg/mL)HIV[17]
5E. senegalensisAuriculatin (EC50: 1.47 µg/mL)
Erysenegalensein O (EC50: 2.19 µg/mL)
Erysenegalensein D (EC50: 1.10 µg/mL)
Erysenegalensein N (EC50: 1.97 µg/mL)
6.8-diprenylgenistein (EC50: 0.203 µg/mL)
HIV[10]
6E. subumbransGangetininSARS-CoV-2[14]
7E. sigmoideaGangetinSARS-CoV-2[13]
8E. senagelensisErybraedin DSARS-CoV-2[16]
9E. variegataOrientanol ESARS-CoV-2[62]
10E. caffraErycaffra FSARS-CoV-2[18]
11E. speciosaVitexin (EC50: 125 µg/mL)Hepatitis Virus[19]
12E. senegalensis2. 3-dihydro-2′-hydroxyosajin (EC50: 71.8 ± 1.45 µg/mL)
osajin (67.54 ± 3.56 µg/mL)
6.8-diprenylgenistein (69.41 ± 2.56 µg/mL)
Hepatitis Virus[20]
13E. addisonaeErythradisson B (EC50: 8.8 µg/mL) Licoagrochalcone (EC50: 21.51 µg/mL)
Abyssinone VI (EC50: 26.44 µg/mL) 5-prenylbutein (EC50: 21.93 µg/mL)
Influenza[8]
14E. abbysinicaStem bark aqueous extract (IC50: 27 µg/mL)HSV[21]
15E. speciosaVitexin (EC50: 64 µg/mL)HSV[19]
16E. fuscaStem bark decoction (EC50: 243 ± 10.9 µg/mL)HSV[15]
17E. poeppigianaStem bark decoction (EC50: 147.6 ± 4.3 µg/mL)HSV[22]
18E. variegataEricristagallin, Osajin, Sigmoidin A, Sigmoidin B, Sigmoidin C, Robustone, Abyssinone, Eryvarin A, Eryvarin B, Euchrenone, Lupiwighteone, LaburnetineDengue Virus[61]
Table 2. Antibacterial activity of Erythrina.
Table 2. Antibacterial activity of Erythrina.
BacteriaErythrinaCompoundsMIC (µg/mL)Ref
Staphylococcus
S. aureusE. caffra6.8-diprenylgenistein7.8[24]
Ethanol bark extract (specific compounds are unknown)39.1[30]
Methanol bark extract (specific compounds are unknown)0.313[41]
E. lysistemonErybraedin A2[35]
Phaseollidin10
Abyssinone V-4′-methyl-ether59
Eryzerin C5
Alpumisoflavone31
Lysisteisoflavone62
E. senegalensisEthanol leaf extract (specific compounds are unknown)25[35]
E. strictaDichloromethane bark extract (specific compounds are unknown)7.81[23]
E. fuscaSandwicensin8[27]
Erythrabbysin A64
Erythrabbysin I64
Eryvarin D4
Scandeone8
E. abyssinicaMethanol bark extract (specific compounds are unknown)23[28]
Ethanol bark extract (specific compounds are unknown)62.5[38]
Ethyl acetate bark extract (specific compounds are unknown)83.3
E. amazoniaMethanol bark extract (specific compounds are unknown)75[33]
E. addisonaeScandenone64[32]
Methicillin-Resistant S. aureus (MRSA)E. subumbransEricristagallin0.39–1.56[65]
E. poeppigianaEryvarin D12.5[40]
3.9-dihydroxy-10-γ,γ- dimethylallyl- 6a,11adehydropterocarpan12.5
Isolupalpigenin1.56–3.13[66]
Erythrinin B6.25
Erypostyrene6.25[37]
Sandwicensin6.25–12.5
Erypoegin A25
Dimethylmedicarpin50
Angolensin50
E. sacleuxiiErysubin F15.4[29]
7.4′-dihydroxy-8.3′-diprenylflavone20.5
E. variegataEryvrain V12.5–25[39]
Eryvarin W1.56–3.13
Eryvarin X0.78–1.56
Bidwillon B3.13–6.25[67]
Eryvarin Q3.13–6.25[68]
E. strictaDichloromethane bark extract (specific compounds are unknown)31.25[23]
E. herbaceaErybacin A50[15]
Erybacin B12.5
Eryvariestyrene12.5
Glyasperin F50
Bidwillol A12.5
Phaseollinisoflavan50
Erythbidin50
Phaseollidin isoflavan12.5
Eryvarin L25
Glabrocoumarone A12.5
E. fuscaSandwicensin16[27]
Erythrabbysin A32
Erythrabbysin I64
Eryvarin D4
Vancomycin-Resistant S. aureus (VRSA)E. subumbransErycristagallin0.39–1.56[65]
E. zeyheriErybraedin A1.56–3.13[36]
Eryzerin A12.5–25
Eryzerin C6.25
Eryzerin D12.5
Eryzerin E12.5
Multidrug-Resistant S. aureus (MDRSA)E. strictaDichloromethane bark extract (specific compounds are unknown)31.25[23]
S. epidermidisE. lysistemonDichloromethane bark extract (specific compounds are unknown)40[69]
Erybraedin A2[35]
Phaseollidin5
Eryzerin C2
Lysisteisoflavone26
E. addisonaeScandenone32[32]
Actinomyces
A. viscosusE. variegata2-(γ,γ-dimethylallyl)-6a-hydroxyphaseollidin3.13[70]
Erystagallin A3.13
Erycristagallin1.56
Micrococcus
M. luteusE. caffraEthanol bark extract (specific compounds are unknown)39.1[30]
Methanol bark extract (specific compounds are unknown)0.156[41]
Enterococcus
E. faecalisE. caffraEthanol bark extract (specific compounds are unknown)39.1[30]
Methanol bark extract (specific compounds are unknown)0.156[41]
Acetone leaf extract (specific compounds are unknown)80[71]
E. addisonaeScandenone64[32]
Bacillus
B. cereusE. lysistemonErybraedin A1[35]
Phaseollidin10
Abyssinone V-4′-methyl-ether26
Eryzerin C10
Alpumisoflavone31
Lysisteisoflavone2
E. caffraMethanol bark extract (specific compounds are unknown)78[41]
E. addisonaeScandenone16[32]
B. pumilusE. caffraMethanol bark extract (specific compounds are unknown)39.1[41]
B. brevisE. cristagalliCoumestrol4.4[72]
Genistein13.5
Daidzein35
B subtilisE. addisonaeScandenone16[32]
Mycobacterium
M. tuberculosisE. strictaErythrabbysin II50[65]
Erystagallin A12.5
Erythrabbysin-150
5-hydroxysophoranone12.5
Sandwicensin50
E. subumbransErybraedin A25
Erythrabbysin II50
Erystagallin A12.5
Erythrabbysin-150
Erycristagallin12.5
5-hydroxysophoranone12.5
Erysubin F12.5
Rifampicin-Resistant M. tuberculosisE. abyssinicaMethanol bark extract (specific compounds are unknown)390[73]
M. smegmatisE. addisonaeScandenone100[32]
Propionibacterium
P. acnesE. lysistemonMethanol: Dichloromethane (1:1) leaf extract (specific compounds are unknown)80[74]
Escherichia
E. coliE. sigmoideaNeobavaisoflavone8–32[25]
E. livingstoniana5.7.3′-trihydroxy-4′-methoxy-5′-(3-methylbut2-enyl)flavanone5[75]
7.3′-dihydroxy-4′-methoxy-5′-(3-
methylbut-2-enyl)flavanone
5
(3S,3″R)-7-hydroxy-2′-methoxy-[3″-hydroxy-2″,2″-dimethylpyrano (3′,4′)] isoflavan63[76]
E. caffraAbyssione-V 4′-O-methyl ether3.9[24]
6.8-diprenylgenistein7.8
Alpinumisoflavone3.9
Methanol bark extract (specific compounds are unknown)0.02[41]
E. lysistemonErybraedin A2[35]
Phaseollidin20
Eryzerin C5
Lysisteisoflavone6
E. senegalensisEthanol leaf extract (specific compounds are unknown)25[26]
E. addisonaeScandenone64[32]
Enterobacter
E. cloacaeE. sigmoideaNeoisoflavone8[25]
E. aerogenesE. addisonaeScandenone64[32]
E. cloacaE. addisonaeScandenone64
Klebsiella
K. pneumoniaeE. sigmoideaNeoisoflavone8[25]
E. caffraAbyssione-V 4′-O-methyl ether3.9[24]
6.8-diprenylgenistein7.8
Alpinumisofl avone3.9
E. senegalensisEthanol leaf extract (specific compounds are unknown)6.25[26]
Pseudomonas
P. aeruginosaE. sigmoideaNeoisoflavone8[25]
E. lysistemonErybraedin A20[35]
Phaseollidin20
Cristacarpin78
Eryzerin C5
Alpumisoflavone20
Lysisteisoflavone31
Proteus
P. vulgarisE. caffraEthanol bark extract156.3[30]
Salmonella
S. typhiE. caffraMethanol bark extract (specific compounds are unknown)0.02[41]
S. enteretidisE. senegalensisCalopocarpin62.5[77]
E. abyssinicaMethanol bark extract (specific compounds are unknown)29[28]
Table 3. Antifungal activity of Erythrina.
Table 3. Antifungal activity of Erythrina.
FungiErythrina SpeciesCompoundsMIC (µg/mL)Ref
C. albicansE. indicaEthanol leaf extract (specific compounds are unknown)62.5[43]
E. senegalensisEthanol stem bark extract (specific compounds are unknown)4.00–15.63[42]
E. strictaDichloromethane bark extract (specific compounds are unknown)125[23]
E. poeppigianaErypostyrene50[37]
E. sacleuxiiErysubin F>32.021[29]
E. lysistemonAqueous extracts (specific compounds are unknown)25,000[45]
C. glabrataE. senegalensisEthanol stem bark extract (specific compounds are unknown)3.91–62.5[42]
C. kruseiE. crista-galliErytharbine, erysotrine, erythratidine N-oxide12.5–31.25[44]
C. neoformansE. senegalensisHydroalcoholic leaves extract (specific compounds are unknown)3120[46]
A. nigerE. senegalensisLectin seeds extract (specific compounds are unknown400[5]
E. lanceolataDichloromethane bark extract (specific compounds are unknown)1250[31]
P. camembretiE. senegalensisLectin seeds extract (specific compounds are unknown200[5]
S. brevicaulisE. senegalensisLectin seeds extract (specific compounds are unknown200[5]
P. oryzaeE. sacleuxiiProstratol C and orobol20[31]
R. stoloniferE. lanceolataDichloromethane bark extract (specific compounds are unknown)625[47]
Table 4. Antiplasmodial activity of Erythrina.
Table 4. Antiplasmodial activity of Erythrina.
PlasmodiumErythrina SpeciesCompoundsIC50
(µg/mL)
Ref
P. falciparumE. strictaErythrabbysin II5.5[65]
Erystagallin A3.8
5-hydroxysophoranone2.5
Soyaspongenol B4.6
E. subumbransErybraedin A3.4
Erythrabbysin II5.5
Erystagallin A3.8
5-hydroxysophoranone2.5
Erysubin F3.2
Soyaspongenol B4.6
E. latissimaErysodine6.5 ± 4.7[50]
Erysovine4.1 ± 0.6
Erysotrine20.6 ± 8.6
Erythraline7.3 ± 4.9
E. sacleuxiiMethanol stem bark extract (specific compounds are unknown)1.78 ± 0.93[48]
Methanol leaf extract (specific compounds are unknown)24.59 ± 10.54
Methanol root bark extract (specific compounds are unknown)0.45 ± 0.09
Acetone stem bark extract (specific compounds are unknown)3.8 ± 0.9[51]
Acetone root bark extract (specific compounds are unknown)2.2 ± 0.6
E. abbysinicaMethanol stem bark extract (specific compounds are unknown)37.37 ± 6.46[49]
Dichloromethane stem bark extract (specific compounds are unknown)5.37 ± 1.59
aqueous stem bark extract (specific compounds are unknown)47.74 ± 9.15
E. caffraErythrinasinate B24.4 ± 2.63[78]
Lupeol41.7 ± 9.74
E. ovalifoliaErythrisenegalone1.69[52]
Alpinumisoflavone1.98
Phaseollidin1.66
Sandwicensin1.83
E. burttiiButtinol A7.6 ± 0.3[56]
Buttinol B19.1 ± 0.6
Buttinol C9.3 ± 0.9
Buttinol H13.3 ± 2.5
Buttinol D4.9 ± 0.3
4-O-Methylsigmoidin B12.4 ± 1.7
Abbysinone V5.7 ± 0.5
Abbysinone V methyl ether10.7 ± 2.4
Calocarpin19.4 ± 1.8
E. senegalensisEthanol stem bark extract (specific compounds are unknown)1.82[54]
E. fuscaMethanol stem bark extract (specific compounds are unknown)13[55]
N-hexane stem bark extract (specific compounds are unknown)21
Chloroform stem bark extract (specific compounds are unknown)22
Aqueous stem bark extract (specific compounds are unknown)5
E. sigmoideaEthanol stem bark extract (specific compounds are unknown)6.44 ± 0.08[53]
Aqueous stem bark extract (specific compounds are unknown)29.51 ± 3.63
E. haerdiiEthanol leaf extract (specific compounds are unknown)25.6 ± 2.5 [58]
Ethanol root bark extract (specific compounds are unknown)11.0 ± 0.7
Ethanol stem bark extract (specific compounds are unknown)8.6 ± 0.8
E. variegataEthyl acetate leaf extract (specific compounds are unknown)16.7[59]
N-butanol leaf extract (specific compounds are unknown)13.2
10.11-dioxoerythratidine9.3
terpenoid pentacyclic glycoside1.8
E. corallodendronIsoschaftoside1.702[57]
Vicenin II1.744
Chloroquine-Resistant P. falciparumE. abbysinicaMethanol stem bark extract (specific compounds are unknown)34.13 ± 9.79[49]
Dichloromethane stem bark extract (specific compounds are unknown)6.99 ± 0.76
Aqueous stem bark extract (specific compounds are unknown)50.11 ± 10.23
E. schliebeniiEthanol root extract (specific compounds are unknown)1.87 ± 0.44[79]
Aqueous stem bark extract (specific compounds are unknown)7.04 ± 0.72
E. burttiiButtinol A8.5 ± 0.6 [56]
Buttinol B21.1 ± 0.8
Buttinol C9.1 ± 1.2
Buttinol H20.3 ± 4.1
Buttinol D6.1 ± 1.5
4-O-Methylsigmoidin B12.7 ± 2.3
Abbysinone V6.6 ± 1.3
Abbysinone V methyl ether11.9 ± 2.1
Calocarpin17 ± 1.5
E. lysistemonaqueous stem bark extract (specific compounds are unknown)8.27[80]
E. fuscaMethanol stem bark extract (specific compounds are unknown)8[55]
N-hexane stem bark extract (specific compounds are unknown)5
Chloroform stem bark extract (specific compounds are unknown)13
Aqueous stem bark extract (specific compounds are unknown)18
E. sacleuxiiAcetone stem bark extract (specific compounds are unknown)6.3 ± 1.4[51]
Acetone root bark extract (specific compounds are unknown)1.34 ± 0.3
E. haerdiiEthanol leaf extract (specific compounds are unknown)36.4 ± 1.9 [58]
Ethanol root bark extract (specific compounds are unknown)15.2 ± 0.9
Ethanol stem bark extract (specific compounds are unknown)7.6 ± 0.7
E. variegataMethanol leaf extract (specific compounds are unknown)6.8 [59]
Ethyl acetate leaf extract (specific compounds are unknown)26.5
N-butanol leaf extract (specific compounds are unknown)5.1
10.11-dioxoerythratidine3.3
terpenoid pentacyclic glycoside3.2
E. corallodendronIsoschaftoside1.1049[57]
Vicenin II1.4099
Multidrug-Resistant P. falciparumE. senegalensisMethanol stem bark extract (specific compounds are unknown)99.6 ± 1.25[81]
E. abbysinicaMethanol stem bark extract (specific compounds are unknown)7.81[82]
E. subumbransVogelin C2.8[34]
Lespedezaflavanone B3.7
Abbysinone V7.0
E. sigmoideaEthanol stem bark extract (specific compounds are unknown)7.53 ± 0.22[53]
Aqueous stem bark extract (specific compounds are unknown)35.23 ± 3.17
E. fuscaLonchocarpol A9.18[83]
Phaseollidin9.09
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

Muchtaridi, M.; Lestyawan, S.; Fakhirah, M.A.; Rusdin, A.; Salsabila, S.; Megantara, S.; Subarnas, A.; Khairul Ikram, N.K. Therapeutic Potential of Erythrina Genus: Bioactive Phytoconstituents with Potent Antiviral and Antimicrobial Activities. Plants 2025, 14, 3053. https://doi.org/10.3390/plants14193053

AMA Style

Muchtaridi M, Lestyawan S, Fakhirah MA, Rusdin A, Salsabila S, Megantara S, Subarnas A, Khairul Ikram NK. Therapeutic Potential of Erythrina Genus: Bioactive Phytoconstituents with Potent Antiviral and Antimicrobial Activities. Plants. 2025; 14(19):3053. https://doi.org/10.3390/plants14193053

Chicago/Turabian Style

Muchtaridi, Muchtaridi, Samuel Lestyawan, Maitsa Alya Fakhirah, Agus Rusdin, Shela Salsabila, Sandra Megantara, Anas Subarnas, and Nur Kusaira Khairul Ikram. 2025. "Therapeutic Potential of Erythrina Genus: Bioactive Phytoconstituents with Potent Antiviral and Antimicrobial Activities" Plants 14, no. 19: 3053. https://doi.org/10.3390/plants14193053

APA Style

Muchtaridi, M., Lestyawan, S., Fakhirah, M. A., Rusdin, A., Salsabila, S., Megantara, S., Subarnas, A., & Khairul Ikram, N. K. (2025). Therapeutic Potential of Erythrina Genus: Bioactive Phytoconstituents with Potent Antiviral and Antimicrobial Activities. Plants, 14(19), 3053. https://doi.org/10.3390/plants14193053

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