Next Article in Journal
Green Preparation and Antibacterial Activity Evaluation of AgNPs-Blumea balsamifera Oil Nanoemulsion
Previous Article in Journal
Maillard Reaction-Derived S-Doped Carbon Dots Promotes Downregulation of PPARγ, C/EBPα, and SREBP-1 Genes In-Vitro
Previous Article in Special Issue
Flavonoids with Anti-Angiogenesis Function in Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

Systematic Review of Chemical Compounds with Immunomodulatory Action Isolated from African Medicinal Plants

by
Wendwaoga Arsène Nikiema
1,2,
Moussa Ouédraogo
1,2,3,*,
Windbedma Prisca Ouédraogo
1,2,3,
Souleymane Fofana
2,4,
Boris Honoré Amadou Ouédraogo
1,2,
Talwendpanga Edwige Delma
1,2,
Belem Amadé
1,2,
Gambo Moustapha Abdoulaye
1,2,
Aimé Serge Sawadogo
3,
Raogo Ouédraogo
2 and
Rasmané Semde
1,2,3
1
Laboratoire de Développement du Médicament, Ecole Doctorale Sciences et Santé, Université Joseph KI—ZERBO, 03 BP 7021 Ouagadougou 03, Burkina Faso
2
Centre d’Excellence Africain, Centre de Formation, de Recherche et d’Expertises en sciences du Médicament (CEA-CFOREM), Université Joseph KI—ZERBO, 03 BP 7021 Ouagadougou 03, Burkina Faso
3
Unité de Formation et de Recherche, Sciences de la Santé, Université Joseph KI—ZERBO, 03 BP 7021 Ouagadougou 03, Burkina Faso
4
Institut des Sciences de la Santé, Université NAZI Boni, 01 BP 1091 Bobo-Dioulasso 01, Burkina Faso
*
Author to whom correspondence should be addressed.
Submission received: 8 January 2024 / Revised: 25 March 2024 / Accepted: 29 March 2024 / Published: 26 April 2024
(This article belongs to the Special Issue Natural Bioactive Compounds and Human Health)

Abstract

:
A robust, well-functioning immune system is the cornerstone of good health. Various factors may influence the immune system’s effectiveness, potentially leading to immune system failure. This review aims to provide an overview of the structure and action of immunomodulators isolated from African medicinal plants. The research was conducted according to PRISMA guidelines. Full-text access research articles published in English up to December 2023, including plant characteristics, isolated phytochemicals, and immuno-modulatory activities, were screened. The chemical structures of the isolated compounds were generated using ChemDraw® (version 12.0.1076), and convergent and distinctive signaling pathways were highlighted. These phytochemicals with demonstrated immunostimulatory activity include alkaloids (berberine, piperine, magnoflorine), polysaccharides (pectin, glucan, acemannan, CALB-4, GMP90-1), glycosides (syringin, cordifolioside, tinocordiside, aucubin), phenolic compounds (ferulic acid, vanillic acid, eupalitin), flavonoids (curcumin, centaurein, kaempferin, luteolin, guajaverin, etc.), terpenoids (oleanolic acid, ursolic acid, betulinic acid, boswellic acids, corosolic acid, nimbidin, andrographolides). These discussed compounds exert their effects through various mechanisms, targeting the modulation of MAPKs, PI3K-Akt, and NF-kB. These mechanisms can support the traditional use of medicinal plants to treat immune-related diseases. The outcomes of this overview are to provoke structural action optimization, to orient research on particular natural chemicals for managing inflammatory, infectious diseases and cancers, or to boost vaccine immunogenicity.

1. Introduction

The immune system comprises a complex network of cells and biological mediators that safeguard the body against harm from foreign invaders like microbes and malignant cell infiltration while preventing excessive immune activation. It is distinguished by innate and adaptive immunities, working synergistically to protect the body [1].
Several endogenous and exogenous factors may influence the immune system’s effectiveness, potentially leading to malfunction. In conditions such as infectious diseases, e.g., COVID-19, dengue fever, and autoimmune diseases, e.g., celiac disease, type 1 diabetes, Addison’s disease, Graves’ disease, and Rheumatoid polyarthritis, there is an inappropriate immune response [2,3,4]. For instance, the interaction of the dengue virus with immune cells triggers a cytokine storm (involving IL1β, IL6, and tumor necrosis factor α), exacerbating the disease [5]. Also, in autoimmune diseases, self-reactive T cells and the exaggerated production of antibodies against the body’s tissues result in persistent inflammation [6]. In both categories of diseases, controlling the immune response is crucial.
Immunomodulation refers to any modification of the immune response and may involve the induction, expression, amplification, or inhibition of a part or phase of the immune response [7,8]. The concept of immunomodulation has gained significant attention, particularly with the resurgence of infectious diseases in recent years. Immunomodulators are categorized into immunostimulants, immunoadjuvants, and immunosuppressants.
Immunostimulants are pharmacological agents capable of strengthening the body’s resistance to infection. In healthy individuals, they serve as preventive measures and potentiators by enhancing the immune response. They can be used in immunotherapy for individuals with compromised immune systems. Notably, immunostimulants show promise in cancer treatment [9]. Immunosuppressants are critical in preventing organ transplant rejection and managing autoimmune diseases and immune-related disorders linked to infections. Immunoadjuvants stimulate the immune system by enhancing the antigenicity of vaccines without exerting a specific antigenic effect. They serve three main functions: aiding in antigen-targeting immune cells, acting as depots for the gradual release of the antigen, and modulating and reinforcing the type of immune response induced. They can influence cellular and humoral response choices, Th1 and Th2, immune protection versus immune destruction, and regeneration [10,11]. These constitute a new and promising application for immunoadjuvants.
Although synthetic immunomodulatory drugs offer many advantages, their undesirable side-effect profile and broad impact on the entire immune system are significant limitations to the extended use of these drugs, justifying the search for more effective and safer agents with targeted immunomodulatory activity. Using natural substances as immunoadjuvants during vaccine development to enhance immunogenicity is very promising [12]. Previous studies have shown that natural products with immunomodulatory activity have already been used to treat autoimmune diseases, inflammatory disorders, and cancer [13]. These substances constitute a valuable source of biologically active secondary metabolites, including alkaloids, polysaccharides, terpenoids, flavonoids, coumarins, glycosides, and proteins. This review aims to compile a comprehensive database of molecules from African medicinal plants capable of modulating immunity and improve our knowledge of the potential signaling pathways.

2. Results

2.1. Database Search Results

In the course of our database search, a total of 610 articles were initially identified (Figure 1). Upon removing duplicate entries (n = 587), 495 studies were subsequently excluded due to their classification as review articles or because the studies focused solely on crude extracts without evaluating isolated bioactive substances. Consequently, we scrutinized 92 full-text articles for eligibility, ultimately excluding 43. Our final dataset comprised 49 studies investigating the immunomodulatory activity of African medicinal plants.
The selected articles were published from 1994 to 2022. A total of 35 medicinal plant species belonging to 25 families were identified. The Liliaceae and Menispermaceae families were most represented with four and three plants species, respectively.
From medicinal plants, 86 molecules were isolated, 80 molecules were evaluated for their immunomodulatory proprieties (Table 1), and 56 had their structures represented (Figures 3–7).

2.2. African Medicinal Plants Used for Immunomodulation

Numerous medicinal plants used in traditional medicine systems have attracted the attention of scientists worldwide (Table 1). As discussed below, these medicinal plants exhibit immunomodulatory activity due to various chemical groups (Figure 2) and others medicinal properties, including antioxidant, anti-inflammatory, analgesic, and anti-arthritic activity.

2.3. Chemistry of Plant-Derived Immunomodulators

2.3.1. Alkaloids

Alkaloids isolated (Figure 3) from C. pareira, T. crispa, T. cordifolia, and P. longum exhibited immunomodulatory activities (Table 2).
Humoral immunity produces antigen-specific antibodies and is primarily driven by B cells. On the other hand, cell-mediated immunity does not depend on antibodies for its adaptive immune functions [75]. Mature T cells, macrophages, and the release of cytokines in response to an antigen primarily drive it. The alkaloid fraction of C. pareira tested on humoral and cell-mediated immunity by measuring the hemagglutination antibody titer and the delayed-type hypersensitivity (DTH) response demonstrated significant immunosuppressive effects at 25 to 100 mg/kg doses. It significantly (p < 0.01) reduced the humoral antibody titer and suppressed the DTH response (p < 0.01) at 75 mg/kg [76]. Berberine (compound 1), an isoquinoline alkaloid isolated from C. pareira, displayed no effect on splenocyte proliferation. However, it downregulated the Th1/Th2 cytokines’ expression (TNF-α, IL-2, IL-4, IL-10) in a mouse primary splenocytes model assay at a range of concentrations (0.8, 1.6, and 3.3 μM, or 0.5 mL/well) [14].
Phagocytosis is an essential cell-defense mechanism against foreign, non-self organisms, and has been used as a critical non-specific immunological parameter to evaluate immune functions. Phagocytes also kill microbes via an oxygen-independent mechanism, although not as effectively as oxygen-dependent mechanisms. Macrophages are essential for the phagocytosis mechanism.
Table 2. Isolated alkaloids with immunomodulatory activities.
Table 2. Isolated alkaloids with immunomodulatory activities.
Isolated Molecules (n°)ModelsPharmacodynamic Parameters
ED50/IC50
Biological EffectsCellular EffectReferences
1In vitro mouse primary splenocyte assay6.6 µMNo significant effect on cell viability at 0.8, 1.6, and 3.3 μMDownregulates splenocytes cytokines (IL2, 4, 10, TNFα) expression. [14]
2Mouse macrophage RAW 264.7 viability, chemotactic, phagocytic assay, ROS, NO, PGE2, and cytokine production, monocyte chemoattractant Protein-1 (MCP-1) production.ndIsolated compounds 2, 3, 4, and 5 at concentrations above 25 μg/mL showed toxic effects on macrophages’ viability (<90%).
Stimulation of cell migration.
Increase in macrophage migration.
Stimulation of cell migration, strong enhancement of macrophage phagocytic activity (81.01% compound 5).
Augmentation of ROS and NO generation.
Significantly stimulates PGE2 production,
enhances the MCP-1 level.
Significantly increases IL-1β, IL6, and TNFα production.
[17,18]
3nd
4nd
523.8 mM
6In vivo hematological assay,
in vitro Dalton’s lymphoma ascites (DLA), Ehrlich ascites carcinoma (EAC) cells assay, L929 cells
ndIncrease in white cell count (138.9%), stimulation of stem cell proliferation, enhancement of the number of plaque-forming cells (71.4%)
Cytotoxicity on DLA, EAC at 200 μg/mL, and L929 at 50 μg/mL.
Enhancement of the antibody production.[21]
nd = non-determined.
In a murine RAW macrophages in vitro assay, the crude extract of T. crispa (concentrations of 25–200 μg/mL) and the isolated alkaloids (compounds 2, 3, 4, and 5) at various concentrations (1.56, 3.12, 6.25, 12.5, and 25 μg/mL) increased chemotactic activity and enhanced macrophage phagocytic activity. Furthermore, they significantly increased cytokine levels (TNFα, IL1β, and IL6) [17]. Cytokines like TNFα contribute to antitumoral effects.
The treatment of Balb/c mice with the alcoholic extract (10 mg/dose/animal) of fruits of Piper longum and piperine (compound 6), a purified alkaloid, at 1.14 mg/dose/animal intraperitoneally for five consecutive days, yielded an increase in the white blood cell count by 142.8% and 138.9%, respectively. In addition, a cytotoxic effect was observed against L929 cancer cells at 100 µg/mL for the crude extract and 50 µg/mL for piperine [21].

2.3.2. Polysaccharides

Numerous studies have shown that plant polysaccharides can regulate the immune system in multiple ways and levels (Table 3). They not only activate immune cells, including T cells, B lymphocytes, macrophages, and dendritic cells, but they also activate and promote the production of cytokines (NO, TNFα, and IL6), thus showing regulatory effects on the immune system in various ways.
Dendritic cells (DCs) act as initiators of the initial immune response and play an essential part in regulating the immune system [77]. DCs recognize, capture, process, and present antigens to naive T cells, which stimulate the activation and proliferation of naive T cells for adaptive immune responses. Assessing immunomodulatory activity on DCs has been performed. Polysaccharides isolated from E. purpurea and Plantago asiatica could upregulate the maturation of DCs [22,23,64]. They act on cell maturation markers by enhancing the expression of surface molecules, including CD80, CD86, and MHC class II.
The thymus is an organ of the immune system, and is the site of production and maturation of T lymphocytes. The spleen is the body’s largest secondary lymphoid organ and, as such, hosts a wide range of immunological functions in addition to its hematopoietic function [78]. In addition to B and T cells, a small amount of macrophages and other cells, such as dendritic cells, are included in splenocytes. The activity of plant extracts has been studied on spleen and thymus cells.
The immunomodulatory effects of plant polysaccharides on macrophages are mainly achieved through the generation of reactive oxygen species (ROS), the secretion of cytokines, cell proliferation, and the phagocytic activity of macrophages. A water-soluble pectic extract from Allium cepa exhibited the capacity to enhance NO production in murine macrophages and stimulate the proliferation of splenocytes and thymocytes. An optimal concentration for proliferation was observed at 50 μg/mL [57]. In vitro, fructo-oligosaccharides (FOS) provoked a significant increase in the mitogenic activity of murine splenocytes and thymocytes after 24 h incubation at 5 and 50 μg/mL concentrations. Macrophage activation is involved in the first phase of the immune response, and interestingly, onion FOS significantly induced macrophage phagocytosis and NO release [58]. A water-soluble glucan isolated from M. oleifera exhibited significant macrophage activation and phagocytic activity along with the induction of monocyte NO release at 0.1 μg/mL [74]. The effect of GMP90-1 polysaccharides isolated from G. mangostana on the viability of RAW 264.7 macrophage was studied using an MTT assay. These studies showed that GMP90-1 polysaccharides inhibited cell growth at 400 µg/mL, whereas 50–200 µg/mL concentrations had no inhibitory effect. Additionally, GMP90-1 polysaccharides increased macrophage phagocytosis and induced NO production and cytokine expression (TNF-α, IL-6, IL-1β) at concentrations of 50, 100, and 200 µg/mL [26].
Table 3. Isolated polysaccharides with immunomodulator activities.
Table 3. Isolated polysaccharides with immunomodulator activities.
SourcesExtraction MethodMolecular Weight (kDa)Monosaccharide CompositionActive SubstanceBiological ActivityReferences
Allium cepaHot water1.8 × 102D-galactose: 6-O-Me-D-galactose: 3-O-acetyl-D-methyl galacturonate: D-methyl galacturonate1:1:1:1PectinEnhancement of NO production in macrophage,
stimulation of splenocyte and thymocyte proliferation.
[57]
Hot ethanol FOS: monosaccharide to hexasaccharideIncrease in splenocytes/thymocytes proliferation (~3-fold), macrophage phagocytic activity, NO production (~2.5-fold).[58]
Moringa oleiferaDistilled water70Gluc. (1→4)-α-D glucanIncrease in macrophage phagocytic activity, and in the number and percentage of globulin.[74]
Garcinia mangostana L. Water extraction5.3Ara., Gal., Rham.GMP90-1 = arabinofurane Enhancement of phagocytic activity (28.0%; 40.3% at 100 and 200 μg/mL, respectively), increase in NO secretion (2.2, 3.9, and 10.3 times at the concentrations of 50, 100, and 200, respectively), IL1β (38.42% at 200 μg/mL), IL6 (4.6, 5.1, and 8.5 times at 50, 100, and 200, respectively), TNFα (5.6, 41.7, and 200.1% at 50, 100, and 200 μg/mL, respectively).[26]
Aloe veraDistilled water-Man, Gluc, Gal.
62.9:13.1:0.6
Heteroglycan or acemannanIncrease in splenocyte proliferation (5.7 and 7.1% after 24 and 48 h, respectively).
Increase in IL-1 and TNFα secretion in irradiated mice (2.34 and 1.32~fold, respectively).
[52]
Echinaneae purpurea L. Water Diploid, tetraploidCPE2, CPE4Stimulation of lymphocyte proliferation and cytokine secretion.[24]
Gal, AraArabinogalactane [79,80]
Fructus aurantiiCold water, hot water3.14 × 102Man, Rha, GlcUA, GalUA, Gal, Ara 16.3:4.0:2.9:3.4:21.7:41.7Pectic polysaccharide CALB-4Promotion of PBMC proliferation.
Upregulation of NO production.
Affects TNFα, IL1β, IL6, and IL8 secretion.
Increases of proIL-1 expression.
[25]
Siraitia grosvenoriiHot water Gluc, Gal. Ara. Rham
5.8:0.77:0.38:0.12
Promotion of B and T lymphocyte proliferation.
Increase in thymus index. Increase in IL-2 and decrease in IL-1.
[27]
T. cordifoliaAcetone extract G1-4AUpregulation of TNFα, IL1β, IL6, IL10, IL12, and IFNγ expression. Enhancement of NO level.[20]
Tamarindus indicaFresh water Gal., Man., Gluc. Increase in phagocytic activity.
Inhibition of PHA-induced lymphocyte proliferation and leukocyte migration by 63–70.%
[72]
Salvia officinalis L.Ethanol-water10,000 < Mw > 50,000Rham. Ara., Xyl., Man., Gluc., Gal., UA.Arabinogalactans (A), Pectins (B), Glucurunoxylan polymers (D).Polysaccharides-induced thymocyte proliferation. [73]
Ara: arabinose; Gal: galactose; GalUA: galacturonic acid; Gluc: glucose; GlcUA: glucuronic acid; Man: manose; Rham: rhamnose; FOS: fructo-oligosaccharides; UA.: uronic acid.
Acemannan, a bioactive compound isolated from A. vera, displayed immunomodulatory activity. Studies in immunosuppressed mice indicated that acemannan treatment increased animal survivability and reduced mortality. It was established that acemannan upregulated cytokines’ (TNFα, IL1β, and IL6) production and improved peripheral lymphocyte counts, spleen cellularity, and the spleen index. Moreover, acemannan stimulated the macrophage nitric oxide release, surface molecule expression, and cell morphology in RAW 264.7 cells, a mouse macrophage cell line [52].
In vitro experiments demonstrated that tetraploid and diploid E. purpura enhanced the stimulation of mouse spleen lymphocytes by Concanavalin A. Tetraploid forms exhibited higher activity at lower concentrations and strongly promoted the release of IL-2 and IFNγ secretion [24].
Zampeng Shu et al. found that pectic polysaccharides (CALB-4) extracted from F. aurantii stimulated NO, TNF-α, IL-1β, IL-6, and IL-8 production depending on the concentration between 24 and 48 h. These polysaccharides also stimulated splenocyte proliferation and increased cyclophosphamide-induced carbon clearance [25]. Lymphocytes, which are essential contributors to the humoral immune response, were stimulated by polysaccharides extracted from S. grosvenonii [27]. These polysaccharides promoted splenocyte and thymocyte proliferation in an in vitro MTT model at 12.5 and 200 µg/mL concentrations. The effect on cytokine secretion was marked by a significant increase in IL-2 at 50 mg/kg and a notable decrease in IL-1β production at 400 mg/kg [27].
Polysaccharides isolated and purified from T. indica showed immunomodulatory activity by blocking mitotic activity induced by PHA on lymphocytes; they enhanced macrophages’ phagocytic activity, and inhibited leukocyte migration [72]. The immunomodulatory activity of water (A), ammonium oxalate (B), and potassium extractable (D) polysaccharide extracted from S. officinalis was evaluated via an in vitro co-mitogenic thymocyte test. Fraction A had an inhibitory effect at 300 μg/mL, and fractions B and D were at 1000 μg/mL. The inhibition was significant with fraction D. The optimum dose of this fraction was 100 μg/mL. Moreover, fraction D had a more marked SIcomit/SImito rate (3–4) than fractions A and B (≈2) [73].

2.3.3. Triterpenoids

Terpenoids, sometimes called isoprenoids, are a large and diverse class of naturally occurring organic chemicals that are similar to terpenes and are derived from assembled five-carbon isoprene units. Triterpenoids possess a rich chemistry and pharmacology with several pentacyclic motifs. They are used in inflammatory diseases and cancer therapeutics [81,82]. Numerous compounds (Figure 4) falling under the class of triterpenoids isolated from diverse medicinal plant species showed immunomodulatory properties.
Oleanolic acid (compound 7) and ursolic acid (compound 8): these pentacyclic terpenoids, extracted from various species of the Plantago genus, such as P. major, Ocimum sanctum, Psydium guajava, and Phyllantus amarus, have exhibited a range of pharmacological activities, including antioxidant and anti-inflammatory effects, functioning as immuno-inhibitors. Compounds 7 and 8 inhibited the peripheral blood proliferation of mononuclear cells (PMBCs) at 1.25 and 20 μg/mL, respectively. Ursolic acid displayed high activity at 40 μg/mL [63]. These compounds enhanced interferon-gamma (IFN-γ) secretion. In a model of human keratinocytes (HKLs), ursolic acid exhibited inhibitory effects on cell viability, while compounds 7 (15 μM) and 8 (30 μM) significantly increased respiratory burst levels. Both oleanolic acid (30 µM) and ursolic acid (30 µM) increased lysosomal enzyme activity, but ursolic acid (7.5 µM) inhibited lysosomal enzyme activity [46]. Corosolic acid (compound 9) isolated from P. guajava reduced HKL cell viability and significantly enhanced the cell respiratory burst level after 24 h of incubation. It also increased lysozyme activity without affecting NO production.
Andrographolides: in vivo and in vitro animal models were used to evaluate the immunomodulatory activity of these diterpenoids (compounds 1012) extracted from A. paniculata. Mice treated with different doses displayed a significant rise in the hemagglutination (HA) titer and in plaque-forming cells (PFC) in the spleen of sheep red blood cell (SRBC)-sensitized mice. Phagocytic activity assessed through carbon clearance exhibited a dose-dependent increase, and white blood cell counts were significantly increased [29].
The effect of compound 13 on the SRBC-induced delayed-type hypersensitivity (DTH) response indicated that oral administration inhibited the expression of the DTH response in mice. Splenocyte proliferation was inhibited at concentrations greater than 3.9 μg/mL, and the macrophage phagocytic activity function was enhanced [33].
Nimbidin (compound 14), isolated from A. indica, possesses immunomodulatory activity by inhibiting macrophage cell migration, phagocytosis, and phorbol myristate acetate (PMA)-stimulated respiratory bursts. Nimbidin also exhibited inhibitory effects on IL-1β release and NO and PGE2 production [31].
Astragalosides (AST IV compound 15; AST VII compound 16): these cycloartane triterpenes with saponin-like structures are mainly extracted from the Astragalus genus species. AST VII and Macrophyllosaponin B (compound 17) displayed low hemolytic activity at 500 µg/mL and increased splenocyte proliferation induced by Concanavalin A, lipopolysaccharide (LPS), and bovine serum albumin (BSA) in immunized mice. Immunoglobulin G1 and G2 antibody titers were increased by AST VII (120 µg) and Macrophyllosaponin B (90 µg), which stimulated IFNγ [83]. According to Nalbantsoy et al., Macrophyllosaponin B (156 µg/mL) exerts a suppressive effect on Th2 lymphocytes and a positive effect on Th1 lymphocytes by stimulating the release of specific cytokines (IL-2, IFNγ). It also inhibits the activity of inducible nitric oxide synthase (iNOS). In a murine model of a lymphoproliferation assay using MTT and a hemolysin spectrophotometry assay, AST IV increased T and B lymphocyte proliferation at 50–200 mg/kg. The activity of IL-1β at 1 nmol/L was increased. Additionally, TNF-α activity was inhibited with or without LPS stimulation [84].
β-aescin (compound 18), isolated from the roots of A. hippocastanum, has demonstrated important antiviral and virucidal activity against the dengue and VSH viruses by targeting their envelope. The crude extract increased the secretion of pro-inflammatory cytokines (TNF-α and IL-6), while the extracted compound β-aescin from A. hippocastanum displayed a synergistic effect with glucocorticoids, enhancing anti-inflammatory activity. β-aescin, in an in vitro model of RAW264.7 cells, decreased the concentration of TNF-α, IL-1β, and NO in a concentration-dependent manner [28,85].
Compound 19, a sesquiterpene tricyclic isolated from P. cablin, was studied for immunomodulatory activity in a mouse model. Oral administration significantly increased macrophage phagocytosis and boosted circulating immunoglobulin (IgM and IgG) while significantly decreasing the DTH response [34]. In the human peripheral blood mononuclear cells (PBMC) assay, compound 22 showed weak activity stimulating cell proliferation and a moderate stimulation of IFNγ secretion.

2.3.4. Polyphenols

The immune system plays a vital role in human well-being by increasing the immune response and providing protection. Polyphenols have well-demonstrated immunomodulatory effects, as they regulate the immune cells, macrophages, cytokines, and signaling pathways, and influence dendritic cells and lymphocytes (B and T), suppress T cell activation and natural killer cells, and suppress tumor-associated macrophages (Table 4).
Polyphenols are a heterogeneous group of phenolic compounds with two major classes: flavonoids and phenolic acids (Figure 5). They show immunomodulatory activity.
Table 4. Polyphenols isolated from African medicinal plants acting as immunomodulators.
Table 4. Polyphenols isolated from African medicinal plants acting as immunomodulators.
Isolated Molecules (n°)ModelsPharmacodynamic ParametersBiological EffectsCellular EffectReferences
ED50IC50
23Mouse macrophage,
lymphocytes (PBMCs) proliferation assay, natural killer cytotoxicity assay
ndndNo effect on cell viability.
Inhibition of lymphocyte proliferation.
Enhancement of NK cytotoxicity
Inhibition of PHA-induced IL2 release,
weak inhibition of TNFα production in PBMC
[38]
Splenocytes assay3.5 μg/mL Inhibition of splenocyte proliferationInhibition IL-2 synthesis[39]
18PBMC cell proliferation assay 1.5 mg/mLSuppression of lymphocyte proliferation [36]
24,
25
IFNγ promoter-driven luciferase reporter and T cells assay.75
0.9 mg/mL
Modulation of IFNγ transcription [35]
27,
28, 29
in vitro RAW 264.7 macrophage proliferation assayndndIncrease in macrophages’ proliferation (by 1.53-fold for compound 27 and 1.43-fold for compound 28). No significant increase was observed for compound 29. [37]
26Proliferation of murine splenocytes, macrophages, and human PBMCs.
NO production, lysosomal enzyme activity, and neutral red uptake assay
ndIncrease in cell viability in the absence of LPS (macrophages 23%, splenocytes 17%, human PBMCs 24%).
Increase in lysosome activity (57%) in a concentration-dependent manner.
Lack of effect on neutral red uptake.
Stimulation of NK cell activity (11%)
No effect on NO release.[41]
30, 31HKLsndndModulation affects the viability of HKLs.
No effect on cell viability
Increase in lysozyme activity
[46]
41, 43, 44In vitro mouse splenocyte proliferation assay, NK cell activity, cytotoxicity T lymphocyte activity, lysosomal enzyme activityndndInduction of splenocyte proliferation in the presence or absence of mitogen.
Enhancement of NK activity.
Inhibition of lysosomal function in a dose-dependent manner
Reduction of NO production (from 53.37 μM to 22.33 μM for compound 44; 20.66 μM for compound 43; and 28.64 μM compound 41)[50]
3238Human PBMCs assaynd
nd
nd
nd
Stimulation of PBMC proliferationStimulation of IFNγ secretion[46]
42Human PBMCs assayndndInhibition of cell proliferationInhibition of IL2 secretion.
Inhibition of NO release
[71]
39, 40Human PBMCs assay, RAW cells assayndndInhibition of cell proliferation (n°39).
Inhibition of lymphocyte proliferation.
No effect on NK cytotoxicity
nd = non-determined. The isolated polyphenol molecules with immunomodulatory activities reduced IL2 and NO secretion while increased IFNγ secretion.
Curcumin (compound 23) has demonstrated an in vitro immunomodulatory effect. On human PBMCs, compound 23 isolated from C. longa exhibited no effect on cell viability but significantly inhibited PHA-stimulated lymphocyte proliferation and enhanced natural killer cytotoxicity. Curcumin inhibited PHA-stimulated IL-2 production and had a weak effect on TNFα release [38]. Another model evaluating curcumin (2.5 μg/mL) activity on splenocytes revealed an inhibition of mitogen-induced splenocyte proliferation and IL-2 synthesis [39]. Centaurein flavonoids: centaurein (compound 24) and its aglycone centaureidin (compound 25), isolated from n-butanol fractions of B. pilosa, were studied for immunomodulatory activity through a cell transfection model with plasmids. They demonstrated increased IFNγ and induced the nuclear factor of activated T-cells (NFAT) and NFκB activity [35]. Another flavonoid, PA-1 (IC50 = 1.25–2.5 µg/mL), exhibited inhibitory activity on lymphocyte proliferation in an in vitro model of murine lymphocyte stimulation [36].
The ethanolic extract of B. diffusa roots exhibited antiproliferative activity on various human and murine cell lines and human PBMCs. Two flavonoids (compounds 39 and 40) were isolated from the ethanolic and chloroform extracts of B. diffusa roots. These compounds inhibited PBMC proliferation induced by PHA and the mixed lymphocyte reaction (MLR), natural killer cytotoxicity, and LPS-induced NO production. In RAW264.7 cells, compound 40 inhibited PHA-induced IL-2 release and LPS-induced TNFα [71,86].
Compound 26 at concentrations of 25 mM showed no toxic effect on murine splenocytes, macrophages, and human PBMCs. It induced cell proliferation, stimulated lysosomal activity, and increased the neutral red uptake in macrophages and the natural killer cell activity [41].
Flavonoids isolated from the ethanolic extract of C. viridiflorus leaves exhibited varying effects on RAW264.7 cells. Compounds 27 and 28 significantly increased cell proliferation, while compound 29 had no effect [87]. Certain flavonoids isolated from different extracts of P. guajava showed immunomodulatory activity in head-kidney leucocyte assays [46]. Hypophyllantin (compound 30) and guajaverin (compound 31) significantly affected cell viability, whereas avicularin (compound 32) did not. Compound 30 significantly increased the production of RBA, whereas compound 31 did not. Compound 30 also significantly increased the NOS production and lysozyme activity in HKL after 24 h of contact.
Compounds 32 and 33, isolated from P. major, exhibited significant (p < 0.05) human PBMC proliferation and IFNγ secretion stimulation. Compound 37 possesses intense activity stimulating human PBMC proliferation and IFNγ secretion (181 pg/mL). The stimulation index was 4.59. Compound 38 enhanced human PBMC proliferation at the range of 5 and 40 mg/mL concentrations. The activity of compound 36 was lesser than that of compound 37; however, this compound possesses higher activity than compounds 34 and 36 [63]. Compound 42 possesses inhibitory activity on human PBMC proliferation. The volatile oil extracted from Z. officinale (0.001–10 ng/mL) inhibited IL1α release in mice peritoneal macrophages. The DTH induced by DNFB was inhibited in a dose-dependent manner, and the inhibition rates were 31.6% (p < 0.01), 34.4% (p < 0.01), and 35.0 (p < 0.01), respectively. The thymus and spleen index decreased at 0.125, 0.25, and 0.5 g/kg bw doses. 6-gingerol (compound 45) is a main pharmacologic substance isolated from Z. officinale. The combination of LPS and 6-gingerol had no significant cytotoxicity on RAW264.7 cells. Nitrite production was also significantly (p < 0.05) inhibited dose-dependently: 6-gingerol significantly suppressed iNOS proteins as well as mRNA levels, TNFα, and IL10 release. The molecule had a protective effect by preventing the calcium overload induced by H2O2. It blocked PKC-α translocation and suppressed LPS-induced cytoplasmic I-κBα phosphorylation [68,70]. The effect of thymoquinone (compound 46), isolated from N. sativa, was evaluated on a rat Wistar cell proliferation model. A low concentration (1 μg/mL) did not significantly affect splenocyte viability and cell proliferation. At 5 μg/mL, a significant (p < 0.05) reduction of cell viability and proliferation was observed, whereas there was no effect on cytokines (IL4, IFNγ) production [67].

2.3.5. Coumarins

Coumarins isolated from various plant species (Figure 6) have demonstrated immunomodulatory effects (Table 5). Two coumarins isolated from A. vera, named compounds one and two were studied in an in vitro model. The compound one increased the macrophage phagocytic function in a concentration-dependent manner (50 to 200 µg/mL), with a maximum effect observed at 200 µg/mL, whereas compound two had no effect [51].
Esculetin (6,7-dihydrocoumarin, compound 47), another isolated coumarin, from A. vera enhanced the mitogenic effect of splenocytes stimulated with LPS and concanavalin A. It induced lymphokine-activated killer (LAK) activity in lymphocytes [53].
Two coumarin terpenoids isolated from F. szowitsiana, compounds 48 and 49, reduced PHA-induced splenocyte proliferation and preferentially induced IL-4 while suppressing IFNγ secretion [50].
Table 5. Isolated coumarins’ immunomodulator activities.
Table 5. Isolated coumarins’ immunomodulator activities.
Isolated Molecules (n°)ModelsPharmacodynamic ParametersBiological EffectsCellular EffectReferences
ED50IC50
47Murine macrophages and lymphocytes assayndndNo effect on macrophage viability.
Enhancement of endocytic activity induced by LPS on macrophages at concentrations of 80 and 120 mM.
Increase in mutagenic-induced cell proliferation.
Induction of LAK activity of splenic lymphocytes.
Enhances NO production and iNOS gene expression[53]
48, 49Murine splenocytes assayndndNo effect on cell viability for tested concentrations (0.5–15 μM). Compound 48 at concentration >0.5 μM decreased splenocytes stimulation index. Compound 49 decreased cell proliferation at lowest dose.
Suppression of PHA-induced cell proliferation.
Significantly augments IL4 secretion.
Inhibits IFNγ production.
Inhibits NO production by stimulated macrophages. Compound 48 increases PGE2 release; however, compound 49 inhibits it.
[50]
nd = non-determined.

2.3.6. Other Molecules: Glycosides

Syringin (compound 50), a phenolic glucoside isolated from T. crispa and T. cordifolia (Figure 7), was investigated in an in vitro study using RAW 264.7 mouse macrophage cultures. It significantly reduced macrophage phagocytic activity and cell chemotaxis. The impact on cytokine production included reducing TNFα, IL1β, and IL6 production [17,18].
Compounds 51, 52, 53, and 54 (Figure 7 and Table 6) increased cell phagocytic activity and NO production in cells [18]. Compound 55 tested on PBMC showed significant stimulation of cell proliferation and enhanced IFNγ secretion [63].
Mangiferin (compound 56), an isolated natural xanthone glucoside, demonstrated immunomodulatory activity. The 100 mg/kg dose significantly increased IgG and IgM levels, whereas IgA levels decreased in in vivo mice model [66].
Table 6. Isolated glycosides’ immunomodulator activity.
Table 6. Isolated glycosides’ immunomodulator activity.
Isolated Molecules (n°)ModelsPharmacodynamic ParametersBiological EffectsCellular EffectReferences
ED50IC50
50Murine RAW 264.7 cell viability assay
Chemotaxis assay
Phagocytosis assay
NO, ROS, PGE2 production
Monocyte chemoattractant Protein-1 production
Cytokine production
ndndToxicity effect above 25 μg/mL.
Reduction of cell chemotactic and phagocytosis activities.
Diminution of MCP-1 production (IC50 = 48.3)
Reduction of NO production.
Inhibition of PGE2 production (IC50 = 12.08 μM).
Decrease in IL1β, IL6, and TNFα production.
[17]
51, 52, 53, 54PMN cells viability assay
Phagocytosis assay
ROS, NO production assay
ndndIncrease in phagocytosis activityDose-dependent increase in NO and superoxide production [18]
55Human mononuclear cells assay
Lymphocytes transformations test
ndndStimulation of PBMC proliferationEnhancement of IFN-γ production[63]
nd = non-determined.

2.3.7. Proteins

The analysis of raw garlic extract revealed the presence of several proteins within the 10–75 kD range (Table 7). The mitogenic activity on peripheral blood lymphocytes (PBL) demonstrated a significant increase in cell proliferation at 10 mg/mL concentrations. Notably, protein QR-2 exhibited the highest mitogenic activity. Additionally, the modulatory effect on splenocytes and thymocytes displayed a stimulatory effect on cell proliferation, while QR-1 and -2 showed agglutination in rabbit erythrocytes [54].
The thymus is the primary immune organ that produces functional T cells. The effect of onion agglutinin (ACA) on thymocyte proliferation showed an 4- and 3.5-fold increase in cell proliferation at 0.01 μg/well and 0.1 μg/well, respectively, at 24 h. On the other hand, ACA showed a weak increase in LPS-induced B-lymphocyte proliferation (1.3-fold). It significantly elevated the expression of IL-2 and IFN-γ. The macrophages are the first line of defense of the body against infections. At 0.1 μg/well, ACA induces a significant increase (6–8-fold) in NO production by RAW264.7 at 24 h. The release of cytokines (TNFα and IL-12) was significantly stimulated [59].
Proteins isolated from A. membranaceus displayed immunomodulatory activities. These proteins significantly affected the proliferation of splenocytes, murine peritoneal macrophages, and bone marrow-derived dendritic cells (BMDCs) at 10–90 µg/mL, except at 10 µg/mL. The optimal activity was observed at a concentration of 50 μg/mL. These proteins promoted the phagocytosis effect of murine peritoneal macrophages, with the compound AMWPDG2 displaying the highest activity. Furthermore, these proteins significantly promoted the secretion of various cytokines and chemokines, including TNFα, IL-6, IL12p40, IL-1β, IL-1α, nitric oxide, hydrogen peroxide, and CXCL1 and CXCL3 secretion [61].
Table 7. Isolated proteins with immunomodulator activity.
Table 7. Isolated proteins with immunomodulator activity.
SourcesExtraction MethodIsolated ProteinsMolecular Weight (kDa)Biological Effects References
Allium sativum QR-1, QR-2, QR3 (7:28:1)13Mitogenic activity on human PBMC, murine splenocytes and thymocytes. QR-1 and QR-2 showed hemagglutination and mannose-binding activities. [54]
Allium cepaDialysis-D-mannose chromatographyACA:
Allium cepa Agglutinin
12ACA at 0.1 μg/well and 0.01 μg/well enhance thymocyte proliferation by ~4- and 3.5-fold, respectively, with a marginal effect on B cells proliferation (~1.3-fold at 0.01 μg/well), significantly increased cytokine production (TNFα, IL12), and IFN-γ and IL2 expression. ACA induced an ~8-fold increase in NO production by rat peritoneal cells at 12 and 24 h. ACA (0.01–10 μg/well) significantly enhanced IL12 (~3-fold) and TNFα (~2–3-fold) release. The phagocytosis activity is enhanced by 2-fold by ACA (0.1; 1; 10 μg).[59]
Tinospora cordifoliaChromatographyG1, G2, G310–80The proteins at a concentration range of 1–10 μg/mL showed mitogenic activity (3-fold) in murine splenocytes at 1–10 μg/mL and ~5–7-fold in thymocytes. They induced NO release by macrophages and enhanced macrophage phagocytosis activity. [19]
Astragalus membranaceus,Alkali extractionAMWP (16 aa)
AMWPDG2 (16 aa),
AMWPDG4 (15 aa),
AMWPDG6 (15 aa)
-
406.115
268.795
342.281
All proteins contain seven essential amino acids: Thr, Val., Met., Ile., Leu., Phe., and Lys.
Proteins at 50 μg/mL significantly promoted in murine peritoneal macrophage phagocytosis activity, secretion of immunomodulatory factors like NO (AMWPDG2 > AMWPDG4 = AMWPDG6) and H2O2 (AMWPDG2 > AMWPDG6 > AMWPDG4) and inflammatory cytokines (TNFa and IL6)
[61]
aa: amino acid; Thr.: threonine; Val.: valine; Ile: Isoleucine; Leu: leucine; Phe.: phenylalanine; Lys.: lysine; Met.: methionine.

2.4. Mechanism of Action of Plant-Derived Immunomodulators

Numerous studies have elucidated the molecular mechanism underlying the immunomodulatory effects of phytochemicals. These compounds activate macrophages and other cells, such as dendritic and lymphocyte cells, through Toll-like Receptors (TLR) [88]. Proinflammatory cytokines and other immune system mediators are closely associated with the induction of transcription factors, including NF-kB, the nuclear factor of activated T lymphocytes, signal transduction, and transcription activator (STAT).
Cell signaling is initiated by receptor stimulation, with most receptors belonging to the TLR family. TLRs are pivotal receptors for many natural substances, including lipopolysaccharides (LPS), natural polysaccharides, alkaloids, and terpenoids. The activation of TLRs triggers the recruitment of MyD88 and subsequently activates specific intracellular pathways. All TLR signaling pathways ultimately lead to the activation of the transcription factor NF-kappa B, which regulates the expression of numerous inflammatory cytokine genes. Three main cell-signaling pathways, including phosphoinositide (PI3K-Akt), Mitogen-Activated Protein Kinases (MAPKs), and nuclear factor kappa B (NF-kB), can activate and transcribe NF-kB.
The MAPK pathway consists of a three-tier kinase cascade in which MAP3K activates MAP2K, which activates MAPKs. MAPK signaling pathways include the activation of the extracellular-related kinase (ERK1/2), p38 isoforms (p38), and c-Jun NH2-terminal kinase (JNK1/2). Once activated, MAPKs can be phosphorylated and translocated into the nucleus, leading to the expression of related genes and cellular responses, such as the secretion of signaling molecules (NO and ROS) and cytokines (IL-1β, IL6, TNFα).
In resting cells, NF-κB in the cytoplasm tightly associates with the inhibitory protein IκB, forming the IκB kinase (IKK) complex. Activation of the NF-κB signaling pathway provokes the disintegration of the IKK complex, leading to the phosphorylation and degradation of IκB-α. Consequently, NF-κB is released, translocated into the nucleus, and bound to DNA, initiating the transcription of proinflammatory-related genes.
The isolated phytochemicals target many proteins in these signaling ways (Table 8). Oleanolic and ursolic acid inhibit TLR4. Oleanolic acid blocks TLR3 activation and inhibits mRNA expression while suppressing the activation of IKKα/β proteins [89]. Magnoflorine and G1-4A activate MAPKs while being inhibited by flavonoids, such as immunomodulator curcumin.
Ursolic acid and betulinic acid inhibit the degradation of Iκ-Bα, the phosphorylation of IkBα and p64 protein, the activation of Iκ-Bα kinase, and the translocation of p65 [90]. Astragaloside IV activates the phosphorylation of the p65, p38, ERK, and JNK proteins [91]. Magnoflorine, an alkaloid extracted from T. crispa, activates the phosphorylation of p65 and increases the phosphorylation and degradation of IκB. It also increases the phosphorylation of the JNK, ERK, and p38 proteins [92]. Curcumin inhibits LPS-induced NF-kB activation by suppressing the MAPK pathway [93]. The possible molecular mechanisms of isolated bioactive-induced immunomodulation are shown in Figure 8 and Table 8.
Table 8. Convergent mechanism of action of isolated compounds of African medicinal plants with immunomodulator activities.
Table 8. Convergent mechanism of action of isolated compounds of African medicinal plants with immunomodulator activities.
Compounds N°Phytochemical Group Cellular
Model
ReceptorTransduction Pathway Mechanism of ActionCellular Actions References
5Alkaloids Macrophages (U937) TLR4MAPKs,
PI3K-Akt
Augmentation of Akt phosphorylation,
induction of JNK, ERK, and p38 phosphorylation
Enhancement of upregulation of TNFα, IL1β, PGE2, COX-2 [92]
G1-4APolysaccharides Macrophages TLR4/MyD88MAPKs Activation of JNK, ERK, and p38 phosphorylationUpregulation of the expression of TNFα, IL6, IL12, IL10 [20]
8Triterpenoids Macrophages TLR4TLR4-
MyD88
Blocking TLR4/MyD88 Decrease in TNF-α,
IL-1β et IL-6 release
[94]
7TriterpenoidsTHP1 cellsTLR3MAPKs Inhibition of IκB phosphorylation and NF-κB translocation [89]
15Terpenoids saponinsMacrophages MAPKs/NFκB Increase in the phosphorylation of p65, p38, JNK, and ERK, and a decrease in their protein expression Increase in IL1β, IL6, TNFα, and inducible nitric oxide synthase[91]
23FlavonoidsDendritic cells MAPKs/NFκBSuppression of MAPKs and p65 activationReduction of inducible NO synthase and IL-12[93]
27FlavonoidsMacrophages TLR4MAPKs Suppression of phosphorylation of proteins p50/p65Increase in TNFα, IL1β, iNOS [95]
55Glycosides3T3-L1 adipocytes NF-κBSuppression of ERK phosphorylation and IκBα degradationInhibiting TNFα production[96]
56GlycosidesMouse primary hepatocytes MAPKsInhibiting the activation of c-JNK and ERK ½[97]
The proinflammatory mediators such TNFα, IL1β, IL6, IL10, COX-2, PGE2, and NO release are inversely modulated by inhibitors (7, 8, 23, 55, 56) and activators (5, 15, 27, G1-4A).

3. Discussion and Perspectives

This systematic review reported the immunomodulatory properties of 86 isolated phytochemicals or groups from 35 medicinal plant species belonging to 25 different families of African flora. Fifty-seven molecules had their structures identified and tested for immunomodulatory properties.
The review covered data from accessible full-text publications and did not take into account grey literature or others’ protected data. Nevertheless, based on the obtained data, an analysis of the chemical-based immunomodulatory properties of isolated molecules in various immune cell models, including macrophages, splenocytes, thymocytes, T and B lymphocytes, dendritic cells, and human PBMCs, was carried out.
The chemical structures of the isolated immunomodulators are organized according to phytochemical groups. These molecules offer a wide range of therapeutic options, such as treating immune-related inflammation, cancer, and oxidant stress-related diseases.

3.1. Alkaloids

Among the identified alkaloids, berberine, piperine, and magnoflorine showed relevant prospects.
Studies have shown that berberine, an iso-quinoline isolated from various medicinal plants, has a variety of pharmacological actions, including antiarthritic, antibacterial, and anticancer effects [98,99]. Berberine is active on several cancer cell lines and has remarkable antiviral activity on several viruses, justified by its immunomodulatory mechanism.
Berberine has been reported to attenuate the radio-resistance of colon cancer cells by repressing P-gp expression [100] and to sensitize breast cancer cells to different chemotherapeutic drugs [101]. Berberine also attenuates ovarian cancer cell resistance to cisplatin (DDP) by targeting the miRNA-21 to regulate the post-transcriptional expression of the tumor suppressor programmed cell death 4 [102]. Pretreatment with berberine promoted the antitumor effects of DDP on laryngeal cancer cells [103]. Pandey et al. showed the potential actions of berberine in attenuating resistance to 5-fluorouracil in gastric cancer cells [104].
The isoquinoline alkaloids have multi-target potential in multifactorial chronic diseases, including immune, metabolic, and neurological disorders, and present a high perspective in therapy [105].
The alkaloids also had antiviral activities on both RNA and DNA viruses. Their broad spectrum of activity is of interest for identifying new applications, like for berberine, an isoquinoline with clinical promise. Berberine effectively disrupts the replication process of various DNA and RNA viruses, including the human immunodeficiency virus and the new severe acute respiratory syndrome linked to coronavirus-2 (SARCOV-2) [106,107,108].
Berberine targets antiviral activity by inhibiting RTase [109] with an IC50 = 0.13 mM on HIV-1 NL4.3, activating protein kinase signaling pathways in several viruses (see the figure describing the mechanism of action: the kinase cascade) or the ERK and JNK signaling cascades to prevent the generation of virions. By activating the MAPK pathway, berberine reduces the virion titer during CHIKV infection [110] and suppresses RSV replication [111].
Piperine, another alkaloid isolated from P. longa, showed antitumoral, anti-inflammatory activities correlated with its immunomodulatory activity. It inhibited the translocation of NF-κB subunits like p50, p65, and c-Rel, as well as CREB, ATF-2, and c-Fos [112]. In an in vitro LPS-induced osteoarthritis model, piperine treatment showed anti-inflammatory activity by downregulating miR-127 and MyD88 expression. Piperine triggers apoptosis in ovarian cancer cells by increasing the JNK and p38 MAPK phosphorylation [113].
Based on these results, isolated alkaloids could be candidates for therapeutic agents for diseases-based NF-κB signaling pathways’ activation.

3.2. Polysaccharides and Proteins

Peritoneal macrophages possess important immunological functions, such as immune defense, surveillance, regulation, and antigen presentation [114]. Phagocytosis is the first step in the macrophage response to invading microorganisms, and the activation of phagocytosis elevates the innate immune response. Activated peritoneal macrophages induce NO production, and various immunostimulatory factors, such as IL-6 and TNF-α, play important roles in the phagocytosis, antigen presentation, and inflammatory regulation of macrophages [115]. It is important to measure the activity of immunostimulatory factors to highlight the immune-based mechanism of protection or pathophysiology genesis of immune-related diseases to guide medication. Numerous plant-isolated molecules act on macrophages, enhancing phagocytic activity and stimulating NO or TNFα release. The spleen is the largest immune organ reflecting the systemic immune status in the human body, and it plays an important role in anti-infection and anticancer activities [116]. The proliferation of spleen lymphocytes is key to activating cellular and humoral immunomodulatory responses. Some proteins (QR-1, QR-2, QR-3, AMWP, AMWPDG2, AMWPDG4, and AMWPDG6) and polysaccharides (Pectin, FOS, Glucan, CPE2, CPE4, and CALB-4) stimulate splenocytes’ and thymocytes’ proliferation. Polysaccharides help boost the immune system while also possessing anti-tumor properties.

3.3. Terpenoids

Among the terpenoids studied, oleanolic acid, ursolic acid, and betulinic acid have divergent effects depending on the concentration. Oleanolic acid stimulates T lymphocyte proliferation at 0.5 μg/mL, while ursolic and betulinic acid have an inhibitory effect with IC50 of 3.01 and 50 μg/mL, respectively [117]. Depending on their concentration, molecules belonging to the same phytochemical family can have opposite effects.
The chemical structure plays a significant role in the biological effect observed. Studying the structure–activity relationship will help elucidate the mechanism. Ursolic and oleanolic acids correlate with different biological activities, such as anti-inflammatory, anticancer, and antidiabetic [118]. Different mechanisms, including immunomodulation, could justify these proprieties.
Immunomodulators act by stimulating or suppressing the immune response. Therefore, they can be used in several therapeutic areas.
In vaccine development, new and improved adjuvants are needed to mitigate adverse effects and increase immunogenicity. AST VII and Mac B isolated from Astragalus have shown an immunostimulant effect by increasing IgG and IgG1 antibody titers with a smaller hemolytic effect. They also act by activating T and B lymphocytes during immunization with BSA [83]. The glucuronoxylan-D polymer isolated from S. officinalis, which has a significant comitogenic effect, has potential adjuvant properties [73].
Immunomodulators offer exciting prospects in the treatment of cancer. Those that strengthen the body’s ability to identify and eliminate cancer cells are revolutionizing treatment and can also help to strengthen the immune system. Immunomodulators have great potential in the treatment of autoimmune diseases. They, therefore, help to restore immunological balance.
They also offer excellent prospects for treating infectious diseases and combating bacterial resistance. B-aescin isolated from the seeds of A. hippocastanum has virucidal and antiviral activity in addition to its immunomodulatory effect [119]. Combining these effects would provide comprehensive treatment while strengthening the immune system.

3.4. Polyphenols

The negative impacts of synthetic drugs and the quest for natural alternatives for therapy have led to an increased demand for the multi-target action of phenols for enhancing immunity [120].
Combining quercetin with piperine, flavonoid, and no-flavonoid compounds presented an effective and potent anti-inflammatory strategy for treating acute colitis in mice. This anti-inflammatory effect was mediated by impaired DC immune responses [121]. Several flavonoids also display antitumor effects directly or indirectly. Luteolin enhanced the sensitivity of lapatinib in human breast cancer cells, and the combination of baicalein and cisplatin increased the apoptosis of gastric cancer and A549 lung adenocarcinoma cells in vitro [122,123].
The immunomodulatory effect is one of the most important mechanisms for the anti-tumor effect of flavonoids. They enhanced the cytotoxicity of NK and CTL cells to tumor cells via the upregulation of their activating receptor [124,125].
Flavonoids inhibit the production of various pro-inflammatory cytokines (IL6 and IL1β). Undeniably, the inflammatory tumor microenvironment is essential in the progression of malignant [126]. Baicalein (33) and baicalin (33′) are directly cytotoxic to some tumors. In addition to direct cytotoxicity, these two flavonoids stimulate the T cell-mediated immune response against tumors by reducing PD-L1 expression in cancer cells [127]. Kaempferol, curcumin, and quercetin inhibit the proliferation of numerous cell line [128].
NF-κB is essential in human cancer initiation, development, metastasis, and treatment resistance [129,130,131,132]. Many human cancers exhibit constitutive NF-κB activity due to the inflammatory microenvironment and various oncogenic mutations. NF-κB activity is associated with tumor cell proliferation [132]. In addition to suppressing apoptosis and promoting angiogenesis, it also induces an epithelial–mesenchymal transition, facilitating distant metastasis formation. NF-κB activation can also remodel the local metabolism and stimulate the immune system to promote tumor growth. The suppression of NF-κB in myeloid or tumor cells generally results in tumor regression, making the NF-κB pathway a promising therapeutic target. Howeve r, due to its vital role in various biological activities, selective targeting of components of the NF-κB pathway must be achieved for therapeutic purposes.
Since NF-κB plays an essential role in both tumor cells and the tumor microenvironment, targeting NF-κB as an anticancer therapy has been explored extensively over the last few decades. Hundreds of natural and synthetic compounds have been reported to inhibit NF-κB. However, their clinical application has shown little efficacy, except for certain types of lymphoma and leukemia [132].
Flavonoids such as curcumin, naringenin, and quercetin inhibit NF-κB through various signaling pathways, which could justify their anticancer activity.

3.5. Glycosides

The immunomodulatory properties of mangiferin have been demonstrated in several studies. It inhibits NF-KB by reducing the translocation of the p65 protein subunit. It also inhibits activation of the AGE-RAGE/mitogen-activated protein kinase (MAPK), the c-Jun N-terminal kinase (JNK), and the p38 pathways. The expression of extracellular regulated kinase 1/2 (ERK1/2) in the myocardium is also increased. Mangiferin could have beneficial effects in diabetic cardiomyopathy [133,134]. Mangiferin has antioxidant and anti-apoptotic properties via the MAPK/NF-κB/mitochondria-dependent pathways. It has an immunoprotective effect during cancer therapy [65,135]. Obesity is closely associated with a state of chronic inflammation, characterized by the abnormal production of cytokines and the activation of inflammatory signaling pathways in adipose tissue. The signaling pathways involve MAPKs.
Aucubin (55) suppressed the activation of extracellular signal-regulated kinase (ERK), the degradation of inhibitory kappa Ba (IκBα), and the subsequent activation of nuclear factor kappa B (NF-κB) [96]. Aucubin could improve obesity-induced atherosclerosis by attenuating TNF-α-induced inflammatory responses.

4. Methods

4.1. Search Strategy

The research was conducted according to PRISMA guidelines to identify studies about African medicinal plants used for their immunomodulatory properties. This exploration involved using the scholarly search engine Google Scholar and a comprehensive screening of prominent international databases, including PubMed, ScienceDirect, African Journal Online, and Embase. Our search queries incorporated specific keywords such as “immunomodulator” OR “immunity” AND “medicinal plant” OR “herbal plant’’ AND “phytochemicals”.
We exclusively considered scientific research articles published in English until December 2023, ensuring that they were accessible without restrictions. Article titles and abstracts were screened according to the research objectives, which focused on the immunomodulatory effects of African medicinal plant chemicals. This examination encompassed instances involving either full or partial isolation, coupled with the subsequent determination of the chemical structure of active compounds. Articles deemed relevant following independent evaluation were included.

4.2. Data Extraction

Eligible records were extracted into Microsoft Excel, 2013 by NWA and double-checked by OM, OWP, and RO. We systematically gathered data from each publication, including scientific names, botanical families, used parts of plants, extraction solvents, phytochemical groups, isolated compounds, and experimental models for objectifying immunomodulatory activities. These data were meticulously documented using a standardized Excel sheet form.
The risk of bias and the quality of each article were assessed by two independent reviewers.
Following this, we analyzed proposed mechanisms underpinning immunomodulatory activities to elucidate convergent and distinctive signaling pathways associated with the active compounds. The chemical structures of the isolated compounds were generated using ChemDraw® (version 12.0.1076). The specific scaffold (alkaloids, terpenoid, phenol, polyphenol, coumarin, or glycoside) was drawn in red, the sugar group and acid group were respectively drawn in pink and blue. In purpose to demonstrate stereochemistry, R and S chirality centers configuration were determined using the Cahn Ingold Prelog priority rules, and the E and Z absolute configurations for double bounds were indicated.
This systematic review protocol is registered on inplasy.com at number INPLASY202410116 (Accessed on 29 January 2024).

5. Conclusions

The present review is a contribution to highlighting African medicinal plants with isolated immunomodulatory molecules. The isolated chemicals, such oleanolic acid, ursolic acid, boswellic acid, betulinic acid, astragalosides, magnoflorine, luteolin, curcumin, andrographolides, centeurein, centaureidin, quercetin, guaverin, corosolic acid, naringenin, pectin, acemannan, nimbidin, syringin, esculetin, umbeliprenin, methyl galbanate, hypophyllantin, and baicalein, possess significant immunomodulatory properties. These molecules belong to various phytochemical groups: alkaloids, polyphenols, terpenoids, carbohydrates, glycosides, and proteins.
The chemical structures’ stereoisomers’ demonstration of the isolated compounds allows an analytical approach to the structure–activity relationship, as their main immunomodulatory transduction pathways are proposed. The pharmacological properties of the chemicals can be improved by chemical group optimizing to yield analogs with improved pharmacokinetic and pharmacodynamic properties.
The chemical actions comprise modulating the expression or phosphorylation status of various accessory proteins associated with the TLRs, STAT3, NF-κB, MAPKs, and PI3K/Akt pathways. The therapeutic perspectives of such immunomodulators are infectious, cancer, and chronic inflammatory diseases’ care, and the development of immunoadjuvants for vaccines.
The review data can also contribute to the registration of immunomodulatory plant-based traditional medicines, to orient researchers towards the screening of new immunomodulatory chemicals from the species or genera of other medicinal plants, or to allow the comparison with immunomodulatory medicinal plants and phytochemicals of other continents.

Author Contributions

Conceptualization, M.O. and W.A.N.; writing—original draft preparation, W.A.N.; writing—review and editing, M.O., W.P.O. and R.O.; Read and corrections: T.E.D., B.H.A.O., B.A., A.S.S., G.M.A., S.F. and R.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Any supplementary information can be obtained on request addressed to the corresponding author.

Acknowledgments

We would like to thank the ACE-CFOREM, a project of the Word Bank giving to N.W.A a scholarship and facilities to perform his studies.

Conflicts of Interest

The authors declare no conflicts of interest. The ACE-CFOREM, which gave the scholarship, had no role in the design of the study, in the collection, analyses, or interpretation of data, in the writing of the manuscript, or in the decision to publish the results.

References

  1. Nicholson, L.B. The Immune System. Essays Biochem. 2016, 60, 275–301. [Google Scholar] [CrossRef]
  2. Mohandas, S.; Jagannathan, P.; Henrich, T.J.; Sherif, Z.A.; Bime, C.; Quinlan, E.; Portman, M.A.; Gennaro, M.; Rehman, J. RECOVER Mechanistic Pathways Task Force Immune Mechanisms Underlying COVID-19 Pathology and Post-Acute Sequelae of SARS-CoV-2 Infection (PASC). Elife 2023, 12, e86014. [Google Scholar] [CrossRef]
  3. Nielsen, D.G. The Relationship of Interacting Immunological Components in Dengue Pathogenesis. Virol. J. 2009, 6, 211. [Google Scholar] [CrossRef]
  4. Pisetsky, D.S. Pathogenesis of Autoimmune Disease. Nat. Rev. Nephrol. 2023, 19, 509–524. [Google Scholar] [CrossRef]
  5. Khan, M.A.; Khan, Z.A.; Charles, M.; Pratap, P.; Naeem, A.; Siddiqui, Z.; Naqvi, N.; Srivastava, S. Cytokine Storm and Mucus Hypersecretion in COVID-19: Review of Mechanisms. J. Inflamm. Res. 2021, 14, 175–189. [Google Scholar] [CrossRef]
  6. Castro, C.; Gourley, M. Diagnostic Testing and Interpretation of Tests for Autoimmunity. J. Allergy Clin. Immunol. 2010, 125, S238–S247. [Google Scholar] [CrossRef]
  7. Mohamed, S.I.A.; Jantan, I.; Haque, M.A. Naturally Occurring Immunomodulators with Antitumor Activity: An Insight on Their Mechanisms of Action. Int. Immunopharmacol. 2017, 50, 291–304. [Google Scholar] [CrossRef]
  8. Shantilal, S.; Vaghela, J.S.; Sisodia, S.S. Review on Immunomodulation and Immunomodulatory Activity of Some Medicinal Plant. Eur. J. Biomed. 2018, 5, 163–174. [Google Scholar]
  9. Zhao, Z.; Zheng, L.; Chen, W.; Weng, W.; Song, J.; Ji, J. Delivery Strategies of Cancer Immunotherapy: Recent Advances and Future Perspectives. J. Hematol. Oncol. 2019, 12, 126. [Google Scholar] [CrossRef]
  10. Cox, E.; Verdonck, F.; Vanrompay, D.; Goddeeris, B. Adjuvants Modulating Mucosal Immune Responses or Directing Systemic Responses towards the Mucosa. Vet. Res. 2006, 37, 511–539. [Google Scholar] [CrossRef]
  11. Lutsiak, M.E.C.; Kwon, G.S.; Samuel, J. Biodegradable Nanoparticle Delivery of a Th2-Biased Peptide for Induction of Th1 Immune Responses. J. Pharm. Pharmacol. 2006, 58, 739–747. [Google Scholar] [CrossRef]
  12. Zhang, A.; Yang, X.; Li, Q.; Yang, Y.; Zhao, G.; Wang, B.; Wu, D. Immunostimulatory Activity of Water-Extractable Polysaccharides from Cistanche Deserticola as a Plant Adjuvant In Vitro and In Vivo. PLoS ONE 2018, 13, e0191356. [Google Scholar] [CrossRef]
  13. Bdallah, E.; Koko, W. Medicinal Plants of Antimicrobial and Immunomodulating Properties. Antimicrob. Res. Nov. Bioknowl. Educ. Programs 2017, 6, 127–139. [Google Scholar]
  14. Lin, W.-C.; Lin, J.-Y. Berberine Down-Regulates the Th1/Th2 Cytokine Gene Expression Ratio in Mouse Primary Splenocytes in the Absence or Presence of Lipopolysaccharide in a Preventive Manner. Int. Immunopharmacol. 2011, 11, 1984–1990. [Google Scholar] [CrossRef]
  15. Lee, C.-H.; Chen, J.-C.; Hsiang, C.-Y.; Wu, S.-L.; Wu, H.-C.; Ho, T.-Y. Berberine Suppresses Inflammatory Agents-Induced Interleukin-1β and Tumor Necrosis Factor-α Productions via the Inhibition of IκB Degradation in Human Lung Cells. Pharmacol. Res. 2007, 56, 193–201. [Google Scholar] [CrossRef]
  16. Okon, E.; Kukula-Koch, W.; Halasa, M.; Jarzab, A.; Baran, M.; Dmoszynska-Graniczka, M.; Angelis, A.; Kalpoutzakis, E.; Guz, M.; Stepulak, A.; et al. Magnoflorine—Isolation and the Anticancer Potential against NCI-H1299 Lung, MDA-MB-468 Breast, T98G Glioma, and TE671 Rhabdomyosarcoma Cancer Cells. Biomolecules 2020, 10, 1532. [Google Scholar] [CrossRef]
  17. Ahmad, W.; Jantan, I.; Kumolosasi, E.; Haque, M.A.; Bukhari, S.N.A. Immunomodulatory Effects of Tinospora crispa Extract and Its Major Compounds on the Immune Functions of RAW 264.7 Macrophages. Int. Immunopharmacol. 2018, 60, 141–151. [Google Scholar] [CrossRef]
  18. Sharma, U.; Bala, M.; Kumar, N.; Singh, B.; Munshi, R.K.; Bhalerao, S. Immunomodulatory Active Compounds from Tinospora cordifolia. J. Ethnopharmacol. 2012, 141, 918–926. [Google Scholar] [CrossRef]
  19. Aranha, I.; Clement, F.; Venkatesh, Y.P. Immunostimulatory Properties of the Major Protein from the Stem of the Ayurvedic Medicinal Herb, Guduchi (Tinospora cordifolia). J. Ethnopharmacol. 2012, 139, 366–372. [Google Scholar] [CrossRef]
  20. Gupta, P.K.; Rajan, M.G.R.; Kulkarni, S. Activation of Murine Macrophages by G1-4A, a Polysaccharide from Tinospora cordifolia, in TLR4/MyD88 Dependent Manner. Int. Immunopharmacol. 2017, 50, 168–177. [Google Scholar] [CrossRef]
  21. Sunila, E.S.; Kuttan, G. Immunomodulatory and Antitumor Activity of Piper longum Linn. and Piperine. J. Ethnopharmacol. 2004, 90, 339–346. [Google Scholar] [CrossRef] [PubMed]
  22. El-Ashmawy, N.E.; El-Zamarany, E.A.; Salem, M.L.; El-Bahrawy, H.A.; Al-Ashmawy, G.M. In Vitro and In Vivo Studies of the Immunomodulatory Effect of Echinacea Purpurea on Dendritic Cells. J. Genet. Eng. Biotechnol. 2015, 13, 185–192. [Google Scholar] [CrossRef] [PubMed]
  23. Benson, J.M.; Pokorny, A.J.; Rhule, A.; Wenner, C.A.; Kandhi, V.; Cech, N.B.; Shepherd, D.M. Echinacea Purpurea Extracts Modulate Murine Dendritic Cell Fate and Function. Food Chem. Toxicol. 2010, 48, 1170–1177. [Google Scholar] [CrossRef] [PubMed]
  24. Yang, G.; Li, K.; Liu, C.; Peng, P.; Bai, M.; Sun, J.; Li, Q.; Yang, Z.; Yang, Y.; Wu, H. A Comparison of the Immunostimulatory Effects of Polysaccharides from Tetraploid and Diploid Echinacea Purpurea. BioMed Res. Int. 2018, 2018, e8628531. [Google Scholar] [CrossRef] [PubMed]
  25. Shu, Z.; Yang, Y.; Xing, N.; Wang, Y.; Wang, Q.; Kuang, H. Structural Characterization and Immunomodulatory Activity of a Pectic Polysaccharide (CALB-4) from Fructus Aurantii. Int. J. Biol. Macromol. 2018, 116, 831–839. [Google Scholar] [CrossRef] [PubMed]
  26. Zhang, S.; Li, Z.; Wang, X.; An, L.; Bao, J.; Zhang, J.; Cui, J.; Li, Y.; Jin, D.-Q.; Tuerhong, M.; et al. Isolation, Structural Elucidation, and Immunoregulation Properties of an Arabinofuranan from the Rinds of Garcinia mangostana. Carbohydr. Polym. 2020, 246, 116567. [Google Scholar] [CrossRef] [PubMed]
  27. Ling, Z.; Ting Jun, H.; Chun Ni, L. Immunomodulatory and Antioxidant Activity of a Siraitia Grosvenorii Polysaccharide in Mice. Afr. J. Biotechnol. 2011, 10, 10045–10053. [Google Scholar] [CrossRef]
  28. Michelini, F.M.; Alché, L.E.; Bueno, C.A. Virucidal, Antiviral and Immunomodulatory Activities of β-Escin and Aesculus Hippocastanum Extract. J. Pharm. Pharmacol. 2018, 70, 1561–1571. [Google Scholar] [CrossRef]
  29. Low, M.; Khoo, C.S.; Münch, G.; Govindaraghavan, S.; Sucher, N.J. An In Vitro Study of Anti-Inflammatory Activity of Standardised Andrographis paniculata Extracts and Pure Andrographolide. BMC Complement. Altern. Med. 2015, 15, 18. [Google Scholar] [CrossRef]
  30. Naik, S.R.; Hule, A. Evaluation of Immunomodulatory Activity of an Extract of Andrographolides from Andographis paniculata. Planta Med. 2009, 75, 785–791. [Google Scholar] [CrossRef]
  31. Kaur, G.; Sarwar Alam, M.; Athar, M. Nimbidin Suppresses Functions of Macrophages and Neutrophils: Relevance to Its Antiinflammatory Mechanisms. Phytother. Res. 2004, 18, 419–424. [Google Scholar] [CrossRef]
  32. Vaghasiya, J.; Datani, M.; Nandkumar, K.; Malaviya, S.; Jivani, N. Comparative Evaluation of Alcoholic and Aqueous Extracts of Ocimum Sanctum for Immunomodulatory Activity. Int. J. Pharm. Biol. Res. 2010, 1, 25e9. [Google Scholar]
  33. Sharma, M.L.; Kaul, A.; Khajuria, A.; Singh, S.; Singh, G.B. Immunomodulatory Activity of Boswellic Acids (Pentacyclic Triterpene Acids) from Boswellia serrata. Phytother. Res. 1996, 10, 107–112. [Google Scholar] [CrossRef]
  34. Liao, J.; Wu, D.; Peng, S.; Xie, J.; Li, Y.; Su, J.; Chen, J.; Su, Z. Immunomodulatory Potential of Patchouli Alcohol Isolated from Pogostemon Cablin (Blanco) Benth (Lamiaceae) in Mice. Trop. J. Pharm. Res 2013, 12, 559–565. [Google Scholar] [CrossRef]
  35. Chang, S.-L.; Chiang, Y.-M.; Chang, C.L.-T.; Yeh, H.-H.; Shyur, L.-F.; Kuo, Y.-H.; Wu, T.-K.; Yang, W.-C. Flavonoids, Centaurein and Centaureidin, from Bidens pilosa, Stimulate IFN-γ Expression. J. Ethnopharmacol. 2007, 112, 232–236. [Google Scholar] [CrossRef] [PubMed]
  36. Pereira, R.L.; Ibrahim, T.; Lucchetti, L.; da Silva, A.J.; Gonçalves de Moraes, V.L. Immunosuppressive and Anti-Inflammatory Effects of Methanolic Extract and the Polyacetylene Isolated from Bidens pilosa L. Immunopharmacology 1999, 43, 31–37. [Google Scholar] [CrossRef] [PubMed]
  37. Abdelhady, M.I.S.; Kamal, A.M.; Rauf, A.; Mubarak, M.S.; Hadda, T.B. Bioassay-Guided Isolation and POM Analyses of a New Immunomodulatory Polyphenolic Constituent from Callistemon viridiflorus. Nat. Prod. Res. 2016, 30, 1131–1135. [Google Scholar] [CrossRef] [PubMed]
  38. Yadav, V.S.; Mishra, K.P.; Singh, D.P.; Mehrotra, S.; Singh, V.K. Immunomodulatory Effects of Curcumin. Immunopharmacol. Immunotoxicol. 2005, 27, 485–497. [Google Scholar] [CrossRef]
  39. Ranjan, D.; Chen, C.; Johnston, T.D.; Jeon, H.; Nagabhushan, M. Curcumin Inhibits Mitogen Stimulated Lymphocyte Proliferation, NFkappaB Activation, and IL-2 Signaling. J. Surg. Res. 2004, 121, 171–177. [Google Scholar] [CrossRef]
  40. Alonso-Castro, A.J.; Ortiz-Sánchez, E.; Domínguez, F.; Arana-Argáez, V.; Juárez-Vázquez, M.d.C.; Chávez, M.; Carranza-Álvarez, C.; Gaspar-Ramírez, O.; Espinosa-Reyes, G.; López-Toledo, G.; et al. Antitumor and Immunomodulatory Effects of Justicia spicigera Schltdl (Acanthaceae). J. Ethnopharmacol. 2012, 141, 888–894. [Google Scholar] [CrossRef]
  41. Del Carmen Juárez-Vázquez, M.; Josabad Alonso-Castro, A.; García-Carrancá, A. Kaempferitrin Induces Immunostimulatory Effects In Vitro. J. Ethnopharmacol. 2013, 148, 337–340. [Google Scholar] [CrossRef] [PubMed]
  42. Harikrishnan, H.; Jantan, I.; Haque, M.A.; Kumolosasi, E. Anti-Inflammatory Effects of Phyllanthus amarus Schum. & Thonn. through Inhibition of NF-κB, MAPK, and PI3K-Akt Signaling Pathways in LPS-Induced Human Macrophages. BMC Complement. Altern. Med. 2018, 18, 224. [Google Scholar] [CrossRef] [PubMed]
  43. Kassuya, C.A.L.; Leite, D.F.P.; de Melo, L.V.; Rehder, V.L.G.; Calixto, J.B. Anti-Inflammatory Properties of Extracts, Fractions and Lignans Isolated from Phyllanthus amarus. Planta Med. 2005, 71, 721–726. [Google Scholar] [CrossRef] [PubMed]
  44. Jantan, I.; Ilangkovan, M.; Yuandani; Mohamad, H.F. Correlation between the Major Components of Phyllanthus amarus and Phyllanthus urinaria and Their Inhibitory Effects on Phagocytic Activity of Human Neutrophils. BMC Complement. Altern. Med. 2014, 14, 429. [Google Scholar] [CrossRef]
  45. Shabbir, A.; Butt, H.; Shahzad, M.; Arshad, H.; Waheed, I. Immunostimulatory Effect of Methanolic Leaves Extract of Psidium guajava (Guava) on Humoral and Cell-Mediated Immunity in Mice. J. Anim. Plant Sci. 2016, 26, 1492–1500. [Google Scholar]
  46. Nhu, T.Q.; Dam, N.P.; Bich Hang, B.T.; Bach, L.T.; Thanh Huong, D.T.; Buu Hue, B.T.; Scippo, M.-L.; Phuong, N.T.; Quetin-Leclercq, J.; Kestemont, P. Immunomodulatory Potential of Extracts, Fractions and Pure Compounds from Phyllanthus amarus and Psidium guajava on Striped Catfish (Pangasianodon hypophthalmus) Head Kidney Leukocytes. Fish. Shellfish. Immunol. 2020, 104, 289–303. [Google Scholar] [CrossRef] [PubMed]
  47. Ben Sghaier, M.; Krifa, M.; Mensi, R.; Bhouri, W.; Ghedira, K.; Chekir-Ghedira, L. In Vitro and In Vivo Immunomodulatory and Anti-Ulcerogenic Activities of Teucrium ramosissimum Extracts. J. Immunotoxicol. 2011, 8, 288–297. [Google Scholar] [CrossRef]
  48. Nasr-Bouzaiene, N.; Sassi, A.; Bedoui, A.; Krifa, M.; Chekir-Ghedira, L.; Ghedira, K. Immunomodulatory and Cellular Antioxidant Activities of Pure Compounds from Teucrium ramosissimum Desf. Tumour Biol. 2016, 37, 7703–7712. [Google Scholar] [CrossRef] [PubMed]
  49. Askari, V.R.; Alavinezhad, A.; Rahimi, V.B.; Rezaee, S.A.; Boskabady, M.H. Immuno-Modulatory Effects of Methanolic Extract of Ferula szowitsiana on Isolated Human Th1/Th2/Treg Cytokines Levels, and Their Genes Expression and Nitric Oxide Production. Cytokine 2021, 138, 155387. [Google Scholar] [CrossRef]
  50. Zamani Taghizadeh Rabe, S.; Iranshahi, M.; Mahmoudi, M. In Vitro Anti-Inflammatory and Immunomodulatory Properties of Umbelliprenin and Methyl Galbanate. J. Immunotoxicol. 2016, 13, 209–216. [Google Scholar] [CrossRef]
  51. Zhang, X.; Wang, H.; Song, Y.; Nie, L.; Wang, L.; Liu, B.; Shen, P.; Liu, Y. Isolation, Structure Elucidation, Antioxidative and Immunomodulatory Properties of Two Novel Dihydrocoumarins from Aloe Vera. Bioorg. Med. Chem. Lett. 2006, 16, 949–953. [Google Scholar] [CrossRef] [PubMed]
  52. Kumar, S.; Tiku, A.B. Immunomodulatory Potential of Acemannan (Polysaccharide from Aloe Vera) against Radiation Induced Mortality in Swiss Albino Mice. Food Agric. Immunol. 2016, 27, 72–86. [Google Scholar] [CrossRef]
  53. Leung, K.; Leung, P.; Kong, L.; Leung, P. Immunomodulatory Effects of Esculetin (6,7-Dihydroxycoumarin) on Murine Lymphocytes and Peritoneal Macrophages. Cell Mol. Immunol. 2005, 2, 181–188. [Google Scholar] [PubMed]
  54. Clement, F.; Pramod, S.N.; Venkatesh, Y.P. Identity of the Immunomodulatory Proteins from Garlic (Allium sativum) with the Major Garlic Lectins or Agglutinins. Int. Immunopharmacol. 2010, 10, 316–324. [Google Scholar] [CrossRef] [PubMed]
  55. Chandrashekar, P.M.; Prashanth, K.V.H.; Venkatesh, Y.P. Isolation, Structural Elucidation and Immunomodulatory Activity of Fructans from Aged Garlic Extract. Phytochemistry 2011, 72, 255–264. [Google Scholar] [CrossRef] [PubMed]
  56. Chandrashekar, P.M.; Venkatesh, Y.P. Identification of the Protein Components Displaying Immunomodulatory Activity in Aged Garlic Extract. J. Ethnopharmacol. 2009, 124, 384–390. [Google Scholar] [CrossRef] [PubMed]
  57. Patra, P.; Sen, I.K.; Bhanja, S.K.; Nandi, A.K.; Samanta, S.; Das, D.; Devi, K.S.P.; Maiti, T.K.; Islam, S.S. Pectic Polysaccharide from Immature Onion Stick (Allium cepa): Structural and Immunological Investigation. Carbohydr. Polym. 2013, 92, 345–352. [Google Scholar] [CrossRef] [PubMed]
  58. Kumar, V.P.; Prashanth, K.V.H.; Venkatesh, Y.P. Structural Analyses and Immunomodulatory Properties of Fructo-Oligosaccharides from Onion (Allium cepa). Carbohydr. Polym. 2015, 117, 115–122. [Google Scholar] [CrossRef] [PubMed]
  59. Prasanna, V.K.; Venkatesh, Y.P. Characterization of Onion Lectin (Allium cepa Agglutinin) as an Immunomodulatory Protein Inducing Th1-Type Immune Response In Vitro. Int. Immunopharmacol. 2015, 26, 304–313. [Google Scholar] [CrossRef]
  60. Farag, M.R.; Alagawany, M. The Role of Astragalus membranaceus as Immunomodulator in Poultry. World’s Poult. Sci. J. 2019, 75, 43–54. [Google Scholar] [CrossRef]
  61. Huang, H.; Luo, S.-H.; Huang, D.-C.; Cheng, S.-J.; Cao, C.-J.; Chen, G.-T. Immunomodulatory Activities of Proteins from Astragalus membranaceus Waste. J. Sci. Food Agric. 2019, 99, 4174–4181. [Google Scholar] [CrossRef] [PubMed]
  62. Liang, Y.; Zhang, Q.; Zhang, L.; Wang, R.; Xu, X.; Hu, X. Astragalus membranaceus Treatment Protects Raw264.7 Cells from Influenza Virus by Regulating G1 Phase and the TLR3-Mediated Signaling Pathway. Evid.-Based Complement. Altern. Med. 2019, 2019, 2971604. [Google Scholar] [CrossRef] [PubMed]
  63. Chiang, L.-C.; Ng, L.T.; Chiang, W.; Chang, M.-Y.; Lin, C.-C. Immunomodulatory Activities of Flavonoids, Monoterpenoids, Triterpenoids, Iridoid Glycosides and Phenolic Compounds of Plantago Species. Planta Med. 2003, 69, 600–604. [Google Scholar] [CrossRef] [PubMed]
  64. Huang, D.; Nie, S.; Jiang, L.; Xie, M. A Novel Polysaccharide from the Seeds of Plantago Asiatica L. Induces Dendritic Cells Maturation through Toll-like Receptor 4. Int. Immunopharmacol. 2014, 18, 236–243. [Google Scholar] [CrossRef] [PubMed]
  65. García, D.; Leiro, J.; Delgado, R.; Sanmartín, M.L.; Ubeira, F.M. Mangifera Indica L. Extract (Vimang) and Mangiferin Modulate Mouse Humoral Immune Responses. Phytother. Res. 2003, 17, 1182–1187. [Google Scholar] [CrossRef] [PubMed]
  66. Rajendran, P.; Jayakumar, T.; Nishigaki, I.; Ekambaram, G.; Nishigaki, Y.; Vetriselvi, J.; Sakthisekaran, D. Immunomodulatory Effect of Mangiferin in Experimental Animals with Benzo(a)Pyrene-Induced Lung Carcinogenesis. Int. J. Biomed. Sci. 2013, 9, 68–74. [Google Scholar] [CrossRef] [PubMed]
  67. Gholamnezhad, Z.; Rafatpanah, H.; Sadeghnia, H.R.; Boskabady, M.H. Immunomodulatory and Cytotoxic Effects of Nigella sativa and Thymoquinone on Rat Splenocytes. Food Chem. Toxicol. 2015, 86, 72–80. [Google Scholar] [CrossRef] [PubMed]
  68. Zhou, H.; Deng, Y.; Xie, Q. The Modulatory Effects of the Volatile Oil of Ginger on the Cellular Immune Response In Vitro and In Vivo in Mice. J. Ethnopharmacol. 2006, 105, 301–305. [Google Scholar] [CrossRef] [PubMed]
  69. Carrasco, F.; Schmidt, G.; Lopes Romero, A.; Sartoretto, J.; Caparroz-Assef, S.; Bersani-Amado, C.; Cuman, R. Immunomodulatory Activity of Zingiber officinale Roscoe, Salvia officinalis L. and Syzygium aromaticum L. Essential Oils: Evidence for Humor- and Cell-Mediated Responses. J. Pharm. Pharmacol. 2009, 61, 961–967. [Google Scholar] [CrossRef]
  70. Lee, T.-Y.; Lee, K.-C.; Chen, S.-Y.; Chang, H.-H. 6-Gingerol Inhibits ROS and iNOS through the Suppression of PKC-α and NF-κB Pathways in Lipopolysaccharide-Stimulated Mouse Macrophages. Bioch. Biophys. Res. Commun. 2009, 382, 134–139. [Google Scholar] [CrossRef]
  71. Pandey, R.; Maurya, R.; Singh, G.; Sathiamoorthy, B.; Naik, S. Immunosuppressive Properties of Flavonoids Isolated from Boerhaavia diffusa Linn. Int. Immunopharmacol. 2005, 5, 541–553. [Google Scholar] [CrossRef]
  72. Sreelekha, T.T.; Vijayakumar, T.; Ankanthil, R.; Vijayan, K.K.; Nair, M.K. Immunomodulatory Effects of a Polysaccharide from Tamarindus Indica. Anti-Cancer Drugs 1993, 4, 209–212. [Google Scholar] [CrossRef] [PubMed]
  73. Capek, P.; Hrıbalová, V.; Švandová, E.; Ebringerová, A.; Sasinková, V.; Masarová, J. Characterization of Immunomodulatory Polysaccharides from Salvia officinalis L. Int. J. Biol. Macromol. 2003, 33, 113–119. [Google Scholar] [CrossRef] [PubMed]
  74. Mondal, S.; Chakraborty, I.; Pramanik, M.; Rout, D.; Islam, S. Structural Studies of an Immunoenhancing Polysaccharide Isolated from Mature Pods (Fruits) of Moringa oleifera (Sajina). Med. Chem. Res. 2004, 13, 390–400. [Google Scholar] [CrossRef]
  75. Sadeghalvad, M.; Mohammadi-Motlagh, H.-R.; Rezaei, N. Structure and Function of the Immune System. In Encyclopedia of Infection and Immunity; Rezaei, N., Ed.; Elsevier: Oxford, UK, 2022; pp. 24–38. ISBN 978-0-323-90303-5. [Google Scholar]
  76. Bafna, A.; Mishra, S. Antioxidant and Immunomodulatory Activity of the Alkaloidal Fraction of Cissampelos pareira Linn. Sci. Pharm. 2010, 78, 21–31. [Google Scholar] [CrossRef] [PubMed]
  77. Liu, K. Dendritic Cells. Encycl. Cell Biol. 2016, 741–749. [Google Scholar] [CrossRef]
  78. Parker, G.A.; Papenfuss, T.L. Immune System. In Atlas of Histology of the Juvenile Rat; Parker, G.A., Picut, C.A., Eds.; Academic Press: Boston, MA, USA, 2016; pp. 293–347. ISBN 978-0-12-802682-3. [Google Scholar]
  79. Luettig, B.; Steinmuller, C.; Gifford, G.E.; Wagner, H.; Lohmann-Matthes, M.-L. Macrophage Activation by the Polysaccharide Arabinogalactan Isolated From Plant Cell Cultures of Echinacea Purpurea. J. Natl. Cancer Inst. 1989, 81, 669–675. [Google Scholar] [CrossRef] [PubMed]
  80. Steinmüller, C.; Roesler, J.; Gröttrup, E.; Franke, G.; Wagner, H.; Lohmann-Matthes, M.L. Polysaccharides Isolated from Plant Cell Cultures of Echinacea Purpurea Enhance the Resistance of Immunosuppressed Mice against Systemic Infections with Candida Albicans and Listeria Monocytogenes. Int. J. Immunopharmacol. 1993, 15, 605–614. [Google Scholar] [CrossRef]
  81. Miranda, R.d.S.; Jesus, B.d.S.M.; Silva Luiz, S.R.; Viana, C.B.; Adão Malafaia, C.R.; Figueiredo, F.d.S.; Carvalho, T.d.S.C.; Silva, M.L.; Londero, V.S.; Costa-Silva, T.A.; et al. Antiinflammatory Activity of Natural Triterpenes—An Overview from 2006 to 2021. Phytother. Res. 2022, 36, 1459–1506. [Google Scholar] [CrossRef]
  82. Renda, G.; Gökkaya, İ.; Şöhretoğlu, D. Immunomodulatory Properties of Triterpenes. Phytochem. Rev. 2022, 21, 537–563. [Google Scholar] [CrossRef]
  83. Nalbantsoy, A.; Nesil, T.; Erden, S.; Calış, I.; Bedir, E. Adjuvant Effects of Astragalus saponins Macrophyllosaponin B and Astragaloside VII. J. Ethnopharmacol. 2011, 134, 897–903. [Google Scholar] [CrossRef] [PubMed]
  84. Wang, Y.-P.; Li, X.-Y.; Song, C.-Q.; Hu, Z.-B. Effect of Astragaloside IV on T, B Lymphocyte Proliferation and Peritoneal Macrophage Function in Mice. Acta Pharmacol. Sin. 2002, 23, 263–266. [Google Scholar] [PubMed]
  85. Xin, W.; Zhang, L.; Sun, F.; Jiang, N.; Fan, H.; Wang, T.; Li, Z.; He, J.; Fu, F. Escin Exerts Synergistic Anti-Inflammatory Effects with Low Doses of Glucocorticoids In Vivo and In Vitro. Phytomedicine 2011, 18, 272–277. [Google Scholar] [CrossRef] [PubMed]
  86. Mehrotra, S.; Singh, V.K.; Agarwal, S.S.; Maurya, R.; Srimal, R.C. Antilymphoproliferative Activity of Ethanolic Extract of Boerhaavia diffusa Roots. Exp. Mol. Pathol. 2002, 72, 236–242. [Google Scholar] [CrossRef] [PubMed]
  87. Kawasaki, T.; Kawai, T. Toll-like Receptor Signaling Pathways. Front. Immunol. 2014, 5, 461. [Google Scholar] [CrossRef] [PubMed]
  88. Lim, H.J.; Jang, H.-J.; Kim, M.H.; Lee, S.; Lee, S.W.; Lee, S.-J.; Rho, M.-C. Oleanolic Acid Acetate Exerts Anti-Inflammatory Activity via IKKα/β Suppression in TLR3-Mediated NF-κB Activation. Molecules 2019, 24, 4002. [Google Scholar] [CrossRef] [PubMed]
  89. Zhao, J.; Zheng, H.; Sui, Z.; Jing, F.; Quan, X.; Zhao, W.; Liu, G. Ursolic Acid Exhibits Anti-Inflammatory Effects through Blocking TLR4-MyD88 Pathway Mediated by Autophagy. Cytokine 2019, 123, 154726. [Google Scholar] [CrossRef] [PubMed]
  90. Kim, K.-S.; Lee, D.-S.; Kim, D.-C.; Yoon, C.-S.; Ko, W.; Oh, H.; Kim, Y.-C. Anti-Inflammatory Effects and Mechanisms of Action of Coussaric and Betulinic Acids Isolated from Diospyros Kaki in Lipopolysaccharide-Stimulated RAW 264.7 Macrophages. Molecules 2016, 21, 1206. [Google Scholar] [CrossRef] [PubMed]
  91. Li, Y.; Meng, T.; Hao, N.; Tao, H.; Zou, S.; Li, M.; Ming, P.; Ding, H.; Dong, J.; Feng, S.; et al. Immune Regulation Mechanism of Astragaloside IV on RAW264.7 Cells through Activating the NF-κB/MAPK Signaling Pathway. Int. Immunopharmacol. 2017, 49, 38–49. [Google Scholar] [CrossRef]
  92. Haque, M.A.; Jantan, I.; Harikrishnan, H.; Abdul Wahab, S.M. Magnoflorine Enhances LPS-Activated Pro-Inflammatory Responses via MyD88-Dependent Pathways in U937 Macrophages. Planta Med. 2018, 84, 1255–1264. [Google Scholar] [CrossRef]
  93. Kim, G.-Y.; Kim, K.-H.; Lee, S.-H.; Yoon, M.-S.; Lee, H.-J.; Moon, D.-O.; Lee, C.-M.; Ahn, S.-C.; Park, Y.C.; Park, Y.-M. Curcumin Inhibits Immunostimulatory Function of Dendritic Cells: MAPKs and Translocation of NF-Kappa B as Potential Targets. J. Immunol. 2005, 174, 8116–8124. [Google Scholar] [CrossRef] [PubMed]
  94. Hwang, Y.-J.; Song, J.; Kim, H.-R.; Hwang, K.-A. Oleanolic Acid Regulates NF-κB Signaling by Suppressing MafK Expression in RAW 264.7 Cells. BMB Rep 2014, 47, 524–529. [Google Scholar] [CrossRef] [PubMed]
  95. Zhong, R.; Miao, L.; Zhang, H.; Tan, L.; Zhao, Y.; Tu, Y.; Prieto, M.A.; Simal-Gandara, J.; Chen, L.; He, C.; et al. Anti-Inflammatory Activity of Flavonols via Inhibiting MAPK and NF-κB Signaling Pathways in RAW264.7 Macrophages. Curr. Res. Food Sci. 2022, 5, 1176–1184. [Google Scholar] [CrossRef] [PubMed]
  96. Park, K.S. Aucubin, a Naturally Occurring Iridoid Glycoside Inhibits TNF-α-Induced Inflammatory Responses through Suppression of NF-κB Activation in 3T3-L1 Adipocytes. Cytokine 2013, 62, 407–412. [Google Scholar] [CrossRef] [PubMed]
  97. Pal, P.B.; Sinha, K.; Sil, P.C. Mangiferin, a Natural Xanthone, Protects Murine Liver in Pb(II) Induced Hepatic Damage and Cell Death via MAP Kinase, NF-κB and Mitochondria Dependent Pathways. PLoS One 2013, 8, e56894. [Google Scholar] [CrossRef] [PubMed]
  98. Suchal, K.; Malik, S.; Khan, S.I.; Malhotra, R.K.; Goyal, S.N.; Bhatia, J.; Kumari, S.; Ojha, S.; Arya, D.S. Protective Effect of Mangiferin on Myocardial Ischemia-Reperfusion Injury in Streptozotocin-Induced Diabetic Rats: Role of AGE-RAGE/MAPK Pathways. Sci. Rep. 2017, 7, 42027. [Google Scholar] [CrossRef]
  99. Imenshahidi, M.; Hosseinzadeh, H. Berberis Vulgaris and Berberine: An Update Review. Phytother. Res. 2016, 30, 1745–1764. [Google Scholar] [CrossRef] [PubMed]
  100. Wang, K.; Feng, X.; Chai, L.; Cao, S.; Qiu, F. The Metabolism of Berberine and Its Contribution to the Pharmacological Effects. Drug Metab. Rev. 2017, 49, 139–157. [Google Scholar] [CrossRef] [PubMed]
  101. Guan, X.; Zheng, X.; Vong, C.T.; Zhao, J.; Xiao, J.; Wang, Y.; Zhong, Z. Combined Effects of Berberine and Evodiamine on Colorectal Cancer Cells and Cardiomyocytes In Vitro. Eur. J. Pharmacol. 2020, 875, 173031. [Google Scholar] [CrossRef]
  102. Gao, X.; Wang, J.; Li, M.; Wang, J.; Lv, J.; Zhang, L.; Sun, C.; Ji, J.; Yang, W.; Zhao, Z.; et al. Berberine Attenuates XRCC1-mediated Base Excision Repair and Sensitizes Breast Cancer Cells to the Chemotherapeutic Drugs. J. Cell. Mol. Med. 2019, 23, 6797–6804. [Google Scholar] [CrossRef]
  103. Liu, S.; Fang, Y.; Shen, H.; Xu, W.; Li, H. Berberine Sensitizes Ovarian Cancer Cells to Cisplatin through miR-21/PDCD4 Axis. Acta Biochim. Biophys. Sin. (Shanghai) 2013, 45, 756–762. [Google Scholar] [CrossRef] [PubMed]
  104. Palmieri, A.; Iapichino, A.; Cura, F.; Scapoli, L.; Carinci, F.; Mandrone, M.; Martinelli, M. Pre-Treatment with Berberine Enhances Effect of 5-Fluorouracil and Cisplatin in HEP2 Laryngeal Cancer Cell Line. J. Biol. Regul. Homeost. Agents 2018, 32, 167–177. [Google Scholar]
  105. Pandey, A.; Vishnoi, K.; Mahata, S.; Tripathi, S.C.; Misra, S.P.; Misra, V.; Mehrotra, R.; Dwivedi, M.; Bharti, A.C. Berberine and Curcumin Target Survivin and STAT3 in Gastric Cancer Cells and Synergize Actions of Standard Chemotherapeutic 5-Fluorouracil. Nutr. Cancer 2015, 67, 1293–1304. [Google Scholar] [CrossRef] [PubMed]
  106. Plazas, E.; 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]
  107. 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]
  108. Chin, L.W.; Cheng, Y.-W.; Lin, S.-S.; Lai, Y.-Y.; Lin, L.-Y.; Chou, M.-Y.; Chou, M.-C.; Yang, C.-C. Anti-Herpes Simplex Virus Effects of Berberine from Coptidis rhizoma, a Major Component of a Chinese Herbal Medicine, Ching-Wei-San. Arch. Virol. 2010, 155, 1933–1941. [Google Scholar] [CrossRef]
  109. 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.; et al. Berberine and Obatoclax Inhibit SARS-CoV-2 Replication in Primary Human Nasal Epithelial Cells In Vitro. Viruses 2021, 13, 282. [Google Scholar] [CrossRef]
  110. Bodiwala, H.S.; Sabde, S.; Mitra, D.; Bhutani, K.K.; Singh, I.P. Synthesis of 9-Substituted Derivatives of Berberine as Anti-HIV Agents. Eur. J. Med. Chem. 2011, 46, 1045–1049. [Google Scholar] [CrossRef]
  111. 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. Antiviral Res. 2019, 164, 52–60. [Google Scholar] [CrossRef]
  112. Shin, H.-B.; Choi, M.-S.; Yi, C.-M.; Lee, J.; Kim, N.-J.; Inn, K.-S. Inhibition of Respiratory Syncytial Virus Replication and Virus-Induced P38 Kinase Activity by Berberine. Int. Immunopharmacol. 2015, 27, 65–68. [Google Scholar] [CrossRef]
  113. Pradeep, C.R.; Kuttan, G. Piperine Is a Potent Inhibitor of Nuclear Factor-κB (NF-κB), c-Fos, CREB, ATF-2 and Proinflammatory Cytokine Gene Expression in B16F-10 Melanoma Cells. Int. Immunopharmacol. 2004, 4, 1795–1803. [Google Scholar] [CrossRef] [PubMed]
  114. Si, L.; Yang, R.; Lin, R.; Yang, S. Piperine Functions as a Tumor Suppressor for Human Ovarian Tumor Growth via Activation of JNK/P38 MAPK-Mediated Intrinsic Apoptotic Pathway. Biosci. Rep. 2018, 38, BSR20180503. [Google Scholar] [CrossRef] [PubMed]
  115. Hirayama, D.; Iida, T.; Nakase, H. The Phagocytic Function of Macrophage-Enforcing Innate Immunity and Tissue Homeostasis. Int. J. Mol. Sci. 2017, 19, 92. [Google Scholar] [CrossRef]
  116. Zhang, C.; Yang, M.; Ericsson, A.C. Function of Macrophages in Disease: Current Understanding on Molecular Mechanisms. Front. Immunol. 2021, 12, 620510. [Google Scholar] [CrossRef] [PubMed]
  117. Aliyu, M.; Zohora, F.; Saboor-Yaraghi, A.A.; Aliyu, M.; Zohora, F.; Saboor-Yaraghi, A.A. Spleen in Innate and Adaptive Immunity Regulation. AIMS Allergy Immunol. 2021, 5, 1–17. [Google Scholar] [CrossRef]
  118. Ayatollahi, A.M.; Ghanadian, M.; Afsharypour, S.; Abdella, O.M.; Mirzai, M.; Askari, G. Pentacyclic Triterpenes in Euphorbia microsciadia with Their T-Cell Proliferation Activity. Iran. J. Pharm. Res. 2011, 10, 287–294. [Google Scholar] [PubMed]
  119. Mioc, M.; Milan, A.; Malița, D.; Mioc, A.; Prodea, A.; Racoviceanu, R.; Ghiulai, R.; Cristea, A.; Căruntu, F.; Șoica, C. Recent Advances Regarding the Molecular Mechanisms of Triterpenic Acids: A Review (Part I). Int. J. Mol. Sci. 2022, 23, 7740. [Google Scholar] [CrossRef] [PubMed]
  120. Salinas, F.M.; Vázquez, L.; Gentilini, M.V.; O´Donohoe, A.; Regueira, E.; Nabaes Jodar, M.S.; Viegas, M.; Michelini, F.M.; Hermida, G.; Alché, L.E.; et al. Aesculus Hippocastanum L. Seed Extract Shows Virucidal and Antiviral Activities against Respiratory Syncytial Virus (RSV) and Reduces Lung Inflammation In Vivo. Antiviral Res. 2019, 164, 1–11. [Google Scholar] [CrossRef] [PubMed]
  121. Rathod, N.B.; Elabed, N.; Punia, S.; Ozogul, F.; Kim, S.-K.; Rocha, J.M. Recent Developments in Polyphenol Applications on Human Health: A Review with Current Knowledge. Plants 2023, 12, 1217. [Google Scholar] [CrossRef]
  122. Cavalcanti, E.; Vadrucci, E.; Delvecchio, F.R.; Addabbo, F.; Bettini, S.; Liou, R.; Monsurrò, V.; Huang, A.Y.-C.; Pizarro, T.T.; Santino, A.; et al. Administration of Reconstituted Polyphenol Oil Bodies Efficiently Suppresses Dendritic Cell Inflammatory Pathways and Acute Intestinal Inflammation. PLOS ONE 2014, 9, e88898. [Google Scholar] [CrossRef]
  123. Li, P.; Hu, J.; Shi, B.; Tie, J. Baicalein Enhanced Cisplatin Sensitivity of Gastric Cancer Cells by Inducing Cell Apoptosis and Autophagy via Akt/mTOR and Nrf2/Keap 1 Pathway. Biochem. Biophys. Res. Commun. 2020, 531, 320–327. [Google Scholar] [CrossRef]
  124. Zhang, L.; Liu, Q.; Huang, L.; Yang, F.; Liu, A.; Zhang, J. Combination of Lapatinib and Luteolin Enhances the Therapeutic Efficacy of Lapatinib on Human Breast Cancer through the FOXO3a/NQO1 Pathway. Biochem. Biophys. Res. Commun. 2020, 531, 364–371. [Google Scholar] [CrossRef]
  125. Bahrami, A.; Fereidouni, M.; Pirro, M.; Bianconi, V.; Sahebkar, A. Modulation of Regulatory T Cells by Natural Products in Cancer. Cancer Lett. 2019, 459, 72–85. [Google Scholar] [CrossRef] [PubMed]
  126. Chen, X.; Du, Y.; Lin, X.; Qian, Y.; Zhou, T.; Huang, Z. CD4+CD25+ Regulatory T Cells in Tumor Immunity. Int. Immunopharmacol. 2016, 34, 244–249. [Google Scholar] [CrossRef] [PubMed]
  127. Casey, S.C.; Amedei, A.; Aquilano, K.; Azmi, A.S.; Benencia, F.; Bhakta, D.; Bilsland, A.E.; Boosani, C.S.; Chen, S.; Ciriolo, M.R.; et al. Cancer Prevention and Therapy through the Modulation of the Tumor Microenvironment. Semin. Cancer Biol. 2015, 35, S199–S223. [Google Scholar] [CrossRef] [PubMed]
  128. Ke, M.; Zhang, Z.; Xu, B.; Zhao, S.; Ding, Y.; Wu, X.; Wu, R.; Lv, Y.; Dong, J. Baicalein and Baicalin Promote Antitumor Immunity by Suppressing PD-L1 Expression in Hepatocellular Carcinoma Cells. Int. Immunopharmacol. 2019, 75, 105824. [Google Scholar] [CrossRef] [PubMed]
  129. Bhosale, P.B.; Ha, S.E.; Vetrivel, P.; Kim, H.H.; Kim, S.M.; Kim, G.S. Functions of Polyphenols and Its Anticancer Properties in Biomedical Research: A Narrative Review. Transl. Cancer Res. 2020, 9, 7619–7631. [Google Scholar] [CrossRef] [PubMed]
  130. Meylan, E.; Dooley, A.L.; Feldser, D.M.; Shen, L.; Turk, E.; Ouyang, C.; Jacks, T. Requirement for NF-κB Signalling in a Mouse Model of Lung Adenocarcinoma. Nature 2009, 462, 104–107. [Google Scholar] [CrossRef]
  131. Xia, Y.; Yeddula, N.; Leblanc, M.; Ke, E.; Zhang, Y.; Oldfield, E.; Shaw, R.J.; Verma, I.M. Reduced Cell Proliferation by IKK2 Depletion in a Mouse Lung-Cancer Model. Nat. Cell Biol. 2012, 14, 257–265. [Google Scholar] [CrossRef]
  132. Bassères, D.S.; Ebbs, A.; Levantini, E.; Baldwin, A.S. Requirement of the NF-kappaB Subunit P65/RelA for K-Ras-Induced Lung Tumorigenesis. Cancer Res. 2010, 70, 3537–3546. [Google Scholar] [CrossRef]
  133. Keutgens, A.; Robert, I.; Viatour, P.; Chariot, A. Deregulated NF-kappaB Activity in Haematological Malignancies. Biochem. Pharmacol. 2006, 72, 1069–1080. [Google Scholar] [CrossRef] [PubMed]
  134. Hou, J.; Zheng, D.; Fung, G.; Deng, H.; Chen, L.; Liang, J.; Jiang, Y.; Hu, Y. Mangiferin Suppressed Advanced Glycation End Products (AGEs) through NF-κB Deactivation and Displayed Anti-Inflammatory Effects in Streptozotocin and High Fat Diet-Diabetic Cardiomyopathy Rats. Can. J. Physiol. Pharmacol. 2016, 94, 332–340. [Google Scholar] [CrossRef] [PubMed]
  135. Muruganandan, S.; Lal, J.; Gupta, P.K. Immunotherapeutic Effects of Mangiferin Mediated by the Inhibition of Oxidative Stress to Activated Lymphocytes, Neutrophils and Macrophages. Toxicology 2005, 215, 57–68. [Google Scholar] [CrossRef]
Figure 1. PRISMA flow diagram of the study selection.
Figure 1. PRISMA flow diagram of the study selection.
Molecules 29 02010 g001
Figure 2. Plant- derived immunomodulators. The isolated phytochemicals with immunomodulatory activities include polyphenols (carbohydrates, alkaloids, and proteins). * NB: Astragalus VII and Macrophyllosaponin B were isolated from the Astragalus genius in particular Astragalus trojanus Stev. and Astragalus oleifolius DC, which are not distributed in Africa.
Figure 2. Plant- derived immunomodulators. The isolated phytochemicals with immunomodulatory activities include polyphenols (carbohydrates, alkaloids, and proteins). * NB: Astragalus VII and Macrophyllosaponin B were isolated from the Astragalus genius in particular Astragalus trojanus Stev. and Astragalus oleifolius DC, which are not distributed in Africa.
Molecules 29 02010 g002
Figure 3. Structures of the identified alkaloids with immunomodulatory activities. The isolated alkaloids comprise a protoberberine isoquinoleic (1), aporphine isoquinoleine alkaloids (25), and a piperidine alkaloid (6). They have common scaffold highlighted in red. All have been tested and showed immunomodulatory activities.
Figure 3. Structures of the identified alkaloids with immunomodulatory activities. The isolated alkaloids comprise a protoberberine isoquinoleic (1), aporphine isoquinoleine alkaloids (25), and a piperidine alkaloid (6). They have common scaffold highlighted in red. All have been tested and showed immunomodulatory activities.
Molecules 29 02010 g003
Figure 4. Structures of identified terpenoids with immunomodulatory activities. These terpenoids are distinguished in triterpenoid pentacyclic saponin (7, 8, 9, 13, 20, 21), lactone sesquiterpenoid (10, 11, 12), tetracyclic saponin heterosid (14, 15, 16, 17), pentacyclic saponin heterosid (18), diterpenoid (19) and Acyclic monoterpenoid (22). They have in common adjacent 2–5 rings, except for linalool (22). It is an advantage to have simple and complex structures like in linalool and beta-aescin (18), respectively, exhibiting immunomodulatory activities, which will lengthen the list of analogues.
Figure 4. Structures of identified terpenoids with immunomodulatory activities. These terpenoids are distinguished in triterpenoid pentacyclic saponin (7, 8, 9, 13, 20, 21), lactone sesquiterpenoid (10, 11, 12), tetracyclic saponin heterosid (14, 15, 16, 17), pentacyclic saponin heterosid (18), diterpenoid (19) and Acyclic monoterpenoid (22). They have in common adjacent 2–5 rings, except for linalool (22). It is an advantage to have simple and complex structures like in linalool and beta-aescin (18), respectively, exhibiting immunomodulatory activities, which will lengthen the list of analogues.
Molecules 29 02010 g004
Figure 5. The structures of identified polyphenols acting as immunomodulators. The isolated polyphenols include flavonoids with the scaffold highlighted in red, polyphenolic acids with the acid group highlighted in blue, and ceto-phenolics. The thymoquinone is classed among polyphenols because in vivo metabolism gives phenolic compounds. The sugar group (highlighted in pink) is critical in the structure–activity relationship of flavonoids. Nevertheless, the active molecules have shown a similar way of modulating the immune system. They comprise phenol acid (38), cinnamic acid (23, 3437), lignan (30), flavonols (25, 28, 39), flavones (33, 42, 43), flavanone (41), heterosid flavonols (24, 26, 27, 29, 31, 32, 40), heterosid flavones (33′, 44), and other phenolic compounds (45, 46).
Figure 5. The structures of identified polyphenols acting as immunomodulators. The isolated polyphenols include flavonoids with the scaffold highlighted in red, polyphenolic acids with the acid group highlighted in blue, and ceto-phenolics. The thymoquinone is classed among polyphenols because in vivo metabolism gives phenolic compounds. The sugar group (highlighted in pink) is critical in the structure–activity relationship of flavonoids. Nevertheless, the active molecules have shown a similar way of modulating the immune system. They comprise phenol acid (38), cinnamic acid (23, 3437), lignan (30), flavonols (25, 28, 39), flavones (33, 42, 43), flavanone (41), heterosid flavonols (24, 26, 27, 29, 31, 32, 40), heterosid flavones (33′, 44), and other phenolic compounds (45, 46).
Molecules 29 02010 g005
Figure 6. Structures of African medicinal isolated coumarins acting as immunomodulators. Sesquiterpenyl coumarins (4849) with a long carbon chain show an identical mechanism of action while simple coumarin (47) has a different mechanism of action. The coumarin basic core is in red.
Figure 6. Structures of African medicinal isolated coumarins acting as immunomodulators. Sesquiterpenyl coumarins (4849) with a long carbon chain show an identical mechanism of action while simple coumarin (47) has a different mechanism of action. The coumarin basic core is in red.
Molecules 29 02010 g006
Figure 7. Structures of identified glycosides acting as immunomodulators. They comprise monosaccharides (50, 54, 55, 56), disaccharides (53) and aglycans (51, 52). The activity of glycosides is not linked only to the structure of the sugar unit (represented in red), but the aglycan part plays an important role. For example, the 11-hydroxymustakone molecule, without a sugar unit, has comparable activity to that of tinocordiside and cordifolioside.
Figure 7. Structures of identified glycosides acting as immunomodulators. They comprise monosaccharides (50, 54, 55, 56), disaccharides (53) and aglycans (51, 52). The activity of glycosides is not linked only to the structure of the sugar unit (represented in red), but the aglycan part plays an important role. For example, the 11-hydroxymustakone molecule, without a sugar unit, has comparable activity to that of tinocordiside and cordifolioside.
Molecules 29 02010 g007
Figure 8. Immunomodulatory signal-transduction pathways of some molecules. TLR: toll-like receptor; MyD88: myeloid differentiation primary response gene 88; MAPKs: mitogen-activated protein kinases; P13k-Akt: phosphatidylinositol-3-kinase-kinase B protein; Ik-Bk: inhibitory kappa B kinase; IKK: I-kB kinase; p38, p50, p65: proteins 38, 50, 65; P: phosphate; NF-kB: nuclear factor kappa B; ERK: extracellular signal-regulated kinase; JNK: c-Jun N-terminal kinase; Molecules 29 02010 i001: activation or stimulation by phytochemical; Molecules 29 02010 i002: inhibition or reduction by phytochemical; →: transduction way or activation.
Figure 8. Immunomodulatory signal-transduction pathways of some molecules. TLR: toll-like receptor; MyD88: myeloid differentiation primary response gene 88; MAPKs: mitogen-activated protein kinases; P13k-Akt: phosphatidylinositol-3-kinase-kinase B protein; Ik-Bk: inhibitory kappa B kinase; IKK: I-kB kinase; p38, p50, p65: proteins 38, 50, 65; P: phosphate; NF-kB: nuclear factor kappa B; ERK: extracellular signal-regulated kinase; JNK: c-Jun N-terminal kinase; Molecules 29 02010 i001: activation or stimulation by phytochemical; Molecules 29 02010 i002: inhibition or reduction by phytochemical; →: transduction way or activation.
Molecules 29 02010 g008
Table 1. African medicinal plants-based isolated immunomodulators.
Table 1. African medicinal plants-based isolated immunomodulators.
Species and Families of Plants Parts of Plant
Used
SolventChemical GroupsIsolated MoleculesOther Biological ActivityReference
Cissampelos pareira L.,
Menispermaceae
RootsMethanolAlkaloids Berberine (1),
Tetrandrine.
Antioxidant, antibacterial[14,15]
Tinospora crispa,
Menispermaceae
StemEthanolAlkaloids, glycosides, terpenoids N-formyllannonaine (2),
N-formylnornuceferine (3), Lysicamine (4) Magnoflorine (5) Syringin (50) 1-Octacosanol.
Anti-inflammatory, antioxidant[16,17]
Tinospora cordifolia (Wild) Hook. F. & Thomson, MenispermaceaeStemMethanol,
n-hexane, chloroform, ethyl acetate and
n-butanol
Alkaloids, glycosides, proteins11-hydroxymustakone (51),
N-methyl-2-pyrrolidone (52),
N-formylannonaine (2) Cordifolioside (53), Tinocordiside (54) Syringin (50)
[18,19,20]
Piper longum Linn. Piperaceae FruitsMethanolAlkaloidsPiperine (6)Anti-inflammatory, anti-infectious, antitumor, analgesic[21]
Echinacea purpura, Echinaceae Whole plant,
Root
Methanol,
ethanol,
aqueous
Polysaccharides, flavonoidsPolysaccharides, alkyl amides,
Arabinogalactans, Caffeic acid(34)
Antioxidant,
anti-inflammatory
[22,23,24]
Fructus aurantii,
Rutaceae
Fruit Polysaccharides Pectic polysaccharide: CALB-4 Anti-carcinogenic, antimicrobial[25]
Garcinia mangostana L., Guttiferae BarkMethanolPolysaccharidesArabinofuran (GMP90-1) Antioxidant, anti-inflammatory, antimicrobial[26]
Siraitia grosvenorii,
Cucurbitaceae
Whole plantAqueousPolysaccharides Polysaccharides Antioxidant, anti-inflammatory[27]
Aesculus hippocastanum, HippocastanaceaeSeedAlcoholicSaponins triterpenoides β-aescin (18)Antiviral[28]
Andrographis paniculata, AcanthaceaeWhole plantMethanol-waterTerpenoidsAndrographolide (10)
14-deoxyandrographolide (11); 14-deoxy-11,12-didehydroandrographolide (12),
Anticancer,
Anti-inflammatory
[29,30]
Azadirachta indica,
Meliaceae
Oil TerpenoidsNimbidin (14) Anti-inflammatory, anti-arthritic[31]
Ocimum sanctum
Lamiaceae
Whole plantAlcoholic, aqueousTerpenoids, essential oils, phenols, flavonoidsEugenol, Carvacrol, Oleanolic acid (7), Ursolic acid (8), Anti-inflammatory, antiallergic[32]
Boswellia serrata Roxb.
Burseraceae
Oleogum resin TerpenoidsBoswellic acids (13)Anti-inflammatory [33]
Pogostemon cablin Benth. LamiaceaeAerial partsEthanol aqueousTerpenoidsPA: Patchouli alcoholic (19)Antioxidant, Antimicrobial[34]
Biden Pilosa, Asteraceae Whole plantn-butanolFlavonoidsPolyacetylene 2-O-β-D-glucosyltrideca-11E-en-3,5,7,9-tetrayn-1,2-diol (PA-1), Centaurein (24), Centaureidin (25) Anti-inflammatory,
antihyperglycemic
[35,36]
Callistenom viridiflorus,
Myrtaceae
LeavesEthanolPhenols, flavonoidsApigenin 4′-O-β-d-glucopyranosyl- (1″’ → 4″)-O-β-d-glucopyranoside, Kaempferide (28), Isoquercetin (27), Hyperin (29)Anti-inflammatory, analgesic, antibacterial, antifungal.[37]
Curcuma longa,
Zingiberaceae
Rhizome FlavonoidsCurcumin (23)Anti-inflammatory, antimutagenic[38,39]
Justicia spicigera
Schltdl. Acanthaceae
LeavesEthanolFlavonoids Kaempferitrin (26)Antioxidant, antitumor[40,41]
Phyllantus amarus,
Euphorbiaceae
LeavesEthanol, fractions: ethyl acetate, dichloromethaneFlavonoids, lignanCorosolic acid (9),
Oleanolic acid (7), Phyllanthin, Hypophyllanthin (30)
Anti-inflammatory, antiviral, antimutagenic. [42,43,44]
Psidium guajava,
Myrtaceae
LeavesEthanolFlavonoids, glycosides, phenolic compounds, terpenoidsEllagic acid, Hyperin (29), Isoquercitin (27), Guajaverin (31), Avicularin (32), Asiatic acid (21), Maslinic acid (20), Corosolic acid (9), Oleanolic acid (7), Ursolic acid (8)Antiallergic, antitumoral, anti-inflammatory, analgesic, antimicrobial[45,46]
Teucrium ramosissimum Desf., LamiaceaeAerials partsChloroformFlavonoidsApigenin-7-glucoside (44), Genkwanin (43) Naringenin (41)Antioxidant, anti-inflammatory[47,48]
Ferula szowitsiana, ApiaceaeRootsMethanolCoumarins terpenoidsMethyl galbanate (49),
Umbelliprenin (48)
Anti-inflammatory, antioxidant[49,50]
Aloe vera,
Liliaceae
Whole rootsChloroformCoumarins, flavonoids, phenolics, carbohydrates, lignansEsculetin (6,7-dihydrocoumarin) (47) AcemannannAnti-inflammatory, antioxidant[51,52,53]
Allium sativum,
Alliaceae
BulbsPBSProteinsProteins (QR-1, QR-2, QR-3), Fructans, proteins (QA-1, QA-2, QA-3)Anti-inflammatory, antioxidant, antimicrobial, antitumor[54,55,56]
Allium cepa,
Alliaceae
BulbsEthanolProteins, polysaccharides, lectinsPectin, FOS (fructo-oligosaccharides), Agglutinin Antimicrobial[57,58,59]
Astragalus membranaceus,
Fabaceae
WasteAlkali solventProteins, saponins, alkaloids, polysaccharides, glucosides Proteins: AMWPDG2, AMWPDG4, AMWPDG6, Astragaloside IV (15), Astragaloside VII (16), Macrophyllosaponin B (17)Immunoadjuvants[60,61,62]
Plantago sp.
(P. major, P. asiatica)
Plantiginaceae.
LeavesAqueousFlavonoids, phenols, terpenoids, iridoids, Aucubin (55), Chlorogenic acid (35), Ferulic acid (36), p-Coumaric acid (37), Vanillic acid (38), Luteolin (42), Ursolic acid (8), Oleanolic acid (7), Baicalein (33), Baicalin (33′).Anticancer, antimicrobial, anti-inflammatory, antioxidant[63,64]
Mangifera indica L. AnacardiaceaeLeaves-Xanthone glucosideMangiferin (56)Antioxidant, antitumoral[65,66]
Nigella sativa L.
Ranunculaceae
SeedsEthanolic Volatile oilThymoquinone (46)Anti-inflammatory, antioxidant.[67]
Zingiber officinale
Zingiberaceae
Dried gingerDistilled waterVolatile oil, polyphenols6-Gingerol (45)Antibacterial, anti-inflammatory, antitumoral[68,69,70]
Boerhavia diffusa NyctaginaceaeLeavesHexane, chloroform, ethanolFlavonoidsEupalitin (BdI) (39), Eupalitin-3-O-β-D-galactopyranoside (BdII) (40)Anti-inflammatory[71]
Tamarindus indica
Leguminoseae
SeedsWaterPolysaccharidesPolysaccharidesAntitumoral[72]
Salvia officinalis L.
Lamiaceae
Arial partsMethanol-chloroformPolysaccharides, proteinsArabinogalactans (A), Pectins (B), Glucurunoxylan polymers (D).Anti-inflammatory[73]
Moringa oleifera,
Moringaceae
Mature podsAqueousPolysaccharides(1→4)-α-D glucanAnti-inflammatory[74]
The chemical compounds or groups (n = 86) were isolated from 35 medicinal plants species belonging to 25 different families. The numbers in parentheses are the appearance order numbers in the text of the 57 isolated chemicals whose structures were drawn (Figures 3–7).
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

Nikiema, W.A.; Ouédraogo, M.; Ouédraogo, W.P.; Fofana, S.; Ouédraogo, B.H.A.; Delma, T.E.; Amadé, B.; Abdoulaye, G.M.; Sawadogo, A.S.; Ouédraogo, R.; et al. Systematic Review of Chemical Compounds with Immunomodulatory Action Isolated from African Medicinal Plants. Molecules 2024, 29, 2010. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules29092010

AMA Style

Nikiema WA, Ouédraogo M, Ouédraogo WP, Fofana S, Ouédraogo BHA, Delma TE, Amadé B, Abdoulaye GM, Sawadogo AS, Ouédraogo R, et al. Systematic Review of Chemical Compounds with Immunomodulatory Action Isolated from African Medicinal Plants. Molecules. 2024; 29(9):2010. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules29092010

Chicago/Turabian Style

Nikiema, Wendwaoga Arsène, Moussa Ouédraogo, Windbedma Prisca Ouédraogo, Souleymane Fofana, Boris Honoré Amadou Ouédraogo, Talwendpanga Edwige Delma, Belem Amadé, Gambo Moustapha Abdoulaye, Aimé Serge Sawadogo, Raogo Ouédraogo, and et al. 2024. "Systematic Review of Chemical Compounds with Immunomodulatory Action Isolated from African Medicinal Plants" Molecules 29, no. 9: 2010. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules29092010

Article Metrics

Back to TopTop