Next Article in Journal
Synthesis, Antimicrobial and Antibiofilm Activities, and Molecular Docking Investigations of 2-(1H-Indol-3-yl)-1H-benzo[d]imidazole Derivatives
Previous Article in Journal
New 1,2,3-Triazole/1,2,4-triazole Hybrids as Aromatase Inhibitors: Design, Synthesis, and Apoptotic Antiproliferative Activity
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

A Bidens pilosa L. Non-Polar Extract Modulates the Polarization of Human Macrophages and Dendritic Cells into an Anti-Inflammatory Phenotype

by
Xandy Melissa Rodríguez Mesa
1,
Leonardo Andres Contreras Bolaños
1,
Geison Modesti Costa
2,
Antonio Luis Mejia
1 and
Sandra Paola Santander González
1,*
1
Phytoimmunomodulation Research Group, Juan N. Corpas University Foundation, Bogotá 111161, Colombia
2
Phytochemistry Research Group (GIFUJ), Pontificia Universidad Javeriana, Bogotá 110231, Colombia
*
Author to whom correspondence should be addressed.
Molecules 2023, 28(20), 7094; https://doi.org/10.3390/molecules28207094
Submission received: 28 August 2023 / Revised: 3 October 2023 / Accepted: 10 October 2023 / Published: 14 October 2023
(This article belongs to the Section Natural Products Chemistry)

Abstract

:
Different communities around the world traditionally use Bidens pilosa L. for medicinal purposes, mainly for its anti-inflammatory, antinociceptive, and antioxidant properties; it is used as an ingredient in teas or herbal medicines for the treatment of pain, inflammation, and immunological disorders. Several studies have been conducted that prove the immunomodulatory properties of this plant; however, it is not known whether the immunomodulatory properties of B. pilosa are mediated by its ability to modulate antigen-presenting cells (APCs) such as macrophages (MØs) and dendritic cells (DCs) (through polarization or the maturation state, respectively). Different polar and non-polar extracts and fractions were prepared from the aerial part of B. pilosa. Their cytotoxic and immunomodulatory effects were first tested on human peripheral blood mononuclear cells (PBMCs) and phytohemagglutinin (PHA)-stimulated PBMCs, respectively, via an MTT assay. Then, the non-cytotoxic plant extracts and fractions that showed the highest immunomodulatory activity were selected to evaluate their effects on human MØ polarization and DC maturation (cell surface phenotype and cytokine secretion) through multiparametric flow cytometry. Finally, the chemical compounds of the B. pilosa extract that showed the most significant immunomodulatory effects on human APCs were identified using gas chromatography coupled with mass spectrometry. The petroleum ether extract and the ethyl acetate and hydroalcoholic fractions obtained from B. pilosa showed low cytotoxicity and modulated the PHA-stimulated proliferation of PBMCs. Furthermore, the B. pilosa petroleum ether extract induced M2 polarization or a hybrid M1/M2 phenotype in MØs and a semi-mature status in DCs, regardless of exposure to a maturation stimulus. The immunomodulatory activity of the non-polar (petroleum ether) extract of B. pilosa on human PBMC proliferation, M2 polarization of MØs, and semi-mature status in DCs might be attributed to the low–medium polarity components in the extract, such as phytosterol terpenes and fatty acid esters.

Graphical Abstract

1. Introduction

Immunomodulation is a strategy that encompasses different therapeutic approaches to modify the immune response [1]. This therapeutic strategy has been used in conditions that require the activation of the immune response, such as infectious diseases, immunodeficiencies, and cancer, as well as in those that demand the control or reduction of the inflammatory response to treat allergies and autoimmune diseases or to prevent transplant rejection.
Different immunomodulatory therapies have focused on regulating the activity of cells of the innate (granulocytes and natural killer (NK) cells) or adaptive (T cells and B cells) immune system [2]. However, in the last 20 years, new therapies have begun to focus on the modulation of antigen-presenting cells (APCs) (macrophages (MØs) and dendritic cells (DCs)) [3,4]. Antigen-presenting cells modulate the activation and inactivation of innate and adaptive immune responses, as well as the balance between them [5,6]. It is well known that alterations in the phenotype and functionality of APCs favor the development of cancer and inflammatory and autoimmune diseases [6,7,8].
Macrophages are a functionally heterogeneous group of cells that can rapidly change their function in response to surrounding signals [6]. Cytokines and Toll-like receptor (TLR) agonists can induce macrophage activation and polarization (M1 or M2) of phenotypes, characterized by the expression of different gene and protein profiles [9]. M1 macrophages, essential for the development of effective antimicrobial and antitumor immune responses, display a proinflammatory phenotype distinguished by the secretion of tumor necrosis factor (TNF) alpha, nitric oxide (NO), inducible nitric oxide synthase (iNOS) interleukin (IL) 12, and IL-23, which favor Th1 and Th17 responses [6,10,11]. In contrast, M2 macrophages exhibit potent anti-inflammatory activity and antagonize the MØ M1 response [12,13]. M2 MØs play a crucial role in tissue homeostasis, wound healing, and fibrosis [14,15,16]. M2 macrophages also participate in the development of the Th2 responses against some fungi and helminths [6,17,18].
Dendritic cells are essential in triggering innate and adaptive immune responses and enforcing a protective tolerance to self-antigens [19]. The activation of immature DCs in tissues depends largely on the maturation status (immature, semi-mature, or mature) they acquire in response to a stimulus [20]. When DCs recognize an extracellular or intracellular stimulus that is insufficient to induce their activation, they remain immature. Immature DCs can efficiently capture antigens, although their processing and the resulting intracellular signaling do not increase the expression of molecules associated with naïve T cell activation for generating effector T cells. Therefore, immature DCs contribute to the induction of tolerance and control of immune responses [21]. On the other hand, immature DCs can mature upon recognizing pathogen- and damage-associated molecular patterns (PAMPs and DAMPs) through TLRs, collectins, or scavenger receptors. Mature DCs overexpress major histocompatibility complex (MHC) class I and II molecules, co-stimulatory molecules (CD80, CD83, CD86), release cytokines (IL-12 and IL-1ꞵ), and can induce clonal expansion of antigen-specific naïve T cells and their differentiation into effector T cells [19,22].
Several investigations have shown that some plant extracts and chemical compounds can activate or inhibit APC responses. For example, curcuminoids obtained from Curcuma longa L. or polysaccharides isolated from Echinacea purpurea (L.) Moench induce M2 and M1 polarization of MØs [23,24], respectively. Likewise, terpenes and flavonoids decrease DC activation, while polysaccharides such as acemannan obtained from Aloe vera (L.) Burm.f., angelane from Angelica gigas Nakai, and galactomannan from Caesalpinia spinosa induce DC maturation [25,26,27].
Bidens pilosa is a plant traditionally used in tropical and subtropical areas around the world to reduce inflammation and pain. These uses have been recorded in America, Africa, Asia, and Oceania [28]. Currently, several scientific works have demonstrated and confirmed that different chemical compounds of this plant modulate the synthesis of inflammatory mediators (IL-1β, IL-6, TNF-α) and reactive oxygen species (ROS) in vitro [29,30,31]. In addition, in vivo studies in animal models have shown that the anti-inflammatory and antinociceptive properties that B. pilosa are associated with include polyacetylenes (PCs) and phenolic compounds [32,33,34].
Although different extracts and chemical compounds of B. pilosa have been shown to modulate the innate immune response, their ability to regulate the activation or inhibition of APCs is unknown. Therefore, this study aimed to evaluate the effect of different extracts and fractions of B. pilosa on MØ polarization and DC maturation.

2. Results and Discussion

2.1. Bidens pilosa Extracts and Fractions Showed No Cytotoxic Activity towards Human Peripheral Blood Mononuclear Cells and Modulated Their Proliferation and Cytokine Secretion

None of the tested extracts and fractions of B. pilosa were cytotoxic to human PBMCs in either of the three evaluations, and their IC50 values were above the theoretical figure considered cytotoxic (IC50 < 100 µg/mL) (Figure 1A). The activity of all B. pilosa extracts and fractions was tested on PHA-stimulated human PBMC cultures for 12, 24, and 48 h; however, only the treatments that had the most significant effects are shown at 48 h.
In PBMCs stimulated and treated at the same time, the PI decreased by 23 to 65% in cultures exposed to the petroleum ether extract (2) (1.56 to 200 µg/mL), by 10 to 45% in those treated with the ethyl acetate fraction (3.3) (1.56 to 200 µg/mL), and by 2 to 14% in cultures exposed to the hydroalcoholic fraction (25 to 200 µg/mL), respectively (Figure 1B). Additionally, cells exposed with this hydroalcoholic fraction at lower concentrations (1.56 to 12.5 µg/mL) showed an increase in the IP by 9% to 3%, respectively (Figure 1B).
Furthermore, the production of proinflammatory cytokines (IL-1β, IL-6, IL-8, and TNF) in PHA-stimulated PBMC cultures was modulated by some of the treatments derived from B. pilosa. In particular, IL-6 was significantly decreased in the presence of the petroleum ether extract (2) and the hydroalcoholic fraction (3.4) (* p < 0.05); the latter also induced a decreased production of IL-1β (* p < 0.05). IL-10 and IL-12p70 were not detected in the supernatants of any culture or control.
The addition of the petroleum ether extract (2) (1.56 to 200 µg/mL) to already proliferating PBMCs decreased the PI by 4% to 14% and the ethyl acetate (3.3) and hydroalcoholic (3.4) fractions by 9% to 19% and 8% to 18%, respectively (Figure 1C). These treatments significantly decreased TNF levels (** p < 0.01 and *** p < 0.001, respectively) but did not alter the concentration of IL-1β, IL-6, or IL-8 in culture supernatants.
B. pilosa has previously been reported to have low cytotoxic activity, even in tumor cell lines. Kviecinski et al. demonstrated that high concentrations of B. pilosa extracts of low and intermediate polarity—obtained through a supercritical fluid process—decreased the viability of human breast cancer MCF7 cells. Furthermore, they calculated the IC50 values of such extracts to be between 437 μg/mL and 291 μg/mL after 24 h and 48 h incubation [35]. Similarly, an aqueous (polar) extract of B. pilosa var. minor Sheriff was found to be slightly cytotoxic to human leukemic cell lines (L1210, U937, K562, Raji, and P3HR1), showing IC50 values between 145 μg/mL and 586 μg/mL [36].
Some authors have shown that a crude methanolic extract of B. pilosa containing medium- and high-polarity compounds inhibited the proliferation of human PBMC stimulated with PHA. In contrast, other authors demonstrated that the aqueous infusion of B. pilosa increases cytokine production (TNF and IL-1β) by human whole blood, both in the presence or not in the presence of lipopolysaccharide (LPS) [37].
This last result could be associated with the activity of PCs which, as observed in models of Listeria monocytogenes, Candida parapsilosis, and Eimeria tenella infection, enhance the phagocytic activity of macrophages and increase the production of IFN-γ by T cells [34,38,39,40]. On the other hand, in in vivo models, the anti-inflammatory potential of B. pilosa has been determined. In a colitis animal model, aqueous extract decreased the leukocytic infiltration in the intestine and the production of inflammatory cytokines such as TNF-α [33,41].
These assays reporting anti-inflammatory or proinflammatory responses may be associated with the activity of different secondary or primary metabolites present in the extracts, which may generate one response or the other. In our assays, extract 2, which is enriched with secondary metabolites, exhibits anti-inflammatory activity that is also reported in the literature (Table 1 and Table 2).
Furthermore, the pharmaceutical product FITOPROT (B. pilosa extract combined with curcuminoids) can reduce the synthesis of proinflammatory cytokines (IL-1β, IL-6, IL-8, and TNF-α) by human HaCaT keratinocytes exposed to 5-fluorouracil (5FU) [29].

2.2. The Non-Polar Extract and the More Polar Fractions Obtained from B. pilosa Promoted an Anti-Inflammatory Profile of Human Macrophages and Dendritic Cells

Petroleum ether extract (2), ethyl acetate (3.3), and hydroalcoholic (3.4) fractions obtained from B. pilosa were selected for further studies on their activity on human MØs and DCs.
Notably, the B. pilosa petroleum ether extract (2) polarized M1-preconditioned MØs into the M2 phenotype (CD80Low/CD86Low/CD163High/CD206Med/CD209High/iNOSLow) and increased the IL-10 levels in culture supernatants (Figure 2).
Moreover, the B. pilosa ethyl acetate (3.3) and hydroalcoholic (3.4) fractions polarized M1-preconditioned MØ towards an “incomplete” M2 phenotype (CD80Low/CD86Low/CD163Med/Low/CD206Med/CD209Med/Low/iNOSLow) and induced a simultaneous increase in IL-10 production. Importantly, none of the extracts or fractions of B. pilosa L. analyzed could polarize M1-preconditioned MØ into the proinflammatory phenotype (CD80High/CD86High/CD163Low/CD206Low/CD209Low /iNOSHigh) or increase the levels of IL-1β, IL-12p70, and TNF in culture supernatants (Figure 2).
Furthermore, after being added to the M2-preconditioned MØ cultures, the petroleum ether extract (2) of B. pilosa induced a significant increase in IL-8 and IL-10 (Figure 3), cytokines associated with an anti-inflammatory profile M2 [42,43,44,45]. This finding agrees with the results described above, according to which this non-polar extract of B. pilosa showed an M2-polarizing effect on M1-preconditioned MØs (Figure 3A,C).
The ethyl acetate (3.3) and hydroalcoholic (3.4) fractions of B. pilosa also polarized the M2-preconditioned MØs to an “incomplete” M2 phenotype (CD80Low/CD86Low/CD163Mid/Low/CD206Mid/CD209Mid/Low/iNOSLow). In addition, fraction 3.3 induced the production of IL-6 and IL-10, whereas fraction 3.4 favored the synthesis of IL-6, IL-8, and IL-10. It should be noted that IL-6 has also been associated with the development of anti-inflammatory MØs in both in vitro and in vivo models of neuroinflammation [46,47] (Figure 3). These results indicate that the petroleum ether extract (2) of B. pilosa enhances the complete switching of MØs into the M2 anti-inflammatory phenotype, regardless of the growth stimuli present during their differentiation.
Moreover, immature DCs exposed to the petroleum ether extract (2) of B. pilosa expressed a phenotypic profile identified as CD206High/CD209High/CD83High/CD86High/HLA-DRHigh. A phenotypic change (CD206High/CD209High/CD83Med/CD86Med/HLA-DRMed/Low) was also observed in cultures exposed to the plant-derived fractions 3.3 and 3.4.
None of the B. pilosa extracts or fractions tested were found to induce the synthesis of proinflammatory cytokines (IL-1β, IL-12p70, and TNF) when added to immature DC cultures (Figure 4D).
Altogether, these data indicated that the petroleum ether extract of B. pilosa is the only treatment that favored the progression of immature DCs into a “semi-mature” status, characterized by the increased expression of major histocompatibility complex class II (MHC-II) and co-stimulatory (CD83 and CD86) molecules, and no increase in the synthesis of the proinflammatory cytokines that were evaluated [48] (Figure 4).
It is well known that immature and semi-mature DCs induce T cell anergy and, thus, tolerance to peripheral self-antigens. These observations have opened new perspectives for developing therapies against allergies and autoimmune diseases and allergies [48,49].
Therefore, the finding that the petroleum ether extract of B. pilosa induced immature DCs to progress to a semi-mature status makes this phytochemical a promising therapeutic compound that should be tested in contexts of chronic inflammation where regulation of the immune response is highly required.
Given the anti-inflammatory properties of the petroleum ether extract (2) of B. pilosa on the polarization into M2 MØs, additional assays were performed to confirm its activity in the polarization of M1-inflammatory MØs (stimulated with IFN-γ) towards M2. The results showed that this extract favored the co-expression of CD209 and CD206 associated with an M2 profile but did not significantly decrease the expression of HLA-DR, CD86, or iNOS, with a production of IL12p70, TNF, IL-6, and IL-8 (Figure 5A). Therefore, macrophages treated with petroleum ether show a hybrid M1/M2 phenotype (CD206High/CD209High/CD86Med/iNOSMed/). The functionality of the M1/M2 phenotype of macrophages has not yet been well described; however, some authors describe that this profile would be associated with the regulation of the assembly and structure of the extracellular matrix, which is essential during the repair process and homeostasis of the tissues [50].
In the same way, and taking into account that the petroleum ether extract (2) on DCs induces the semi-maturation of these cells, (Figure 4) it was established if this extract can or cannot favor their maturation in co-treatment with a maturation stimulus such as LPS. The results obtained showed that the petroleum ether extract of B. pilosa does, indeed, change the mature phenotype induced for LPS (CD206Med/CD209Med/CD83High/CD86High/HLA-DRHigh) toward a phenotype status CD206High/CD209High/CD83High/CD86High/HLA-DRMed; additionally, treatment only modulated a reduced TNF production, without affecting production of other cytokines when compared with the vehicle control. Since this profile is similar to a semi-mature profile, the extract seems to be inhibiting the complete maturation of dendritic cells.
The observed results show that Bidens apolar extract has anti-inflammatory potential that could be used in future therapeutic approaches for the treatment of chronic inflammatory conditions, such as autoimmune diseases, in which APCs play an important role in the chronicity and maintenance of these conditions [51,52,53] due to the overproduction of proinflammatory cytokines (IL-1β, IL-6, and TNF-α) and overexpression of some transcription factors, such as the nuclear factor kappa B (NF-κB) and the signal transducer and activator of transcription 3 (STAT3) [54].
The development and chronicity of several autoimmune diseases (systemic lupus erythematosus (SEL), primary Sjögren’s syndrome, thyroid autoimmunity, type I diabetes (T1D), and rheumatoid arthritis (RA)) are associated with a constant synthesis of IL-1β [55,56]. This cytokine affects cell survival by decreasing the DNA content and reducing protein synthesis, metabolism, energy production, and the cell cycle, thus favoring the development of autoimmune disorders [57]. For example, elevated serum levels of IL-1β along with those of TNF-α have been associated with increased pancreatic β-cell destruction in patients with T1D, at all stages of the disease [58,59,60,61,62].
High levels of IL-1β are also observed in the serum and cerebrospinal fluid (CSF) of patients with multiple sclerosis. IL-1β alters the excitation–inhibition balance of neurons, and its elevated levels in CSF are associated with brain lesions [63,64]. These observations corroborate the relationship between chronic IL-1β production and disease severity. Furthermore, in patients with RA, a synergistic interaction between TNF-α, IL-1β, and IL-6 is related to disease progression, as it promotes chronic joint inflammation, constant activation of proinflammatory MØs, and erosive changes in cartilages and bones [65,66,67]. Similarly, TNF-α, IL-1β, and IL-6 secreted by MØs also play an essential role in the pathophysiology of autoimmune inflammatory bowel diseases such as colitis. These cytokines accumulate in intestinal tissue and induce extracellular matrix degradation, epithelial damage, endothelial activation, and blood vessel disruption [68,69,70].
Altogether, the above evidence supports the critical role of proinflammatory cytokines in the progression of autoimmune diseases. Some clinical trials with antibodies antagonizing the biological activity of cytokine, such as tocilizumab (anti-IL-6R mAb), have yielded effective therapeutic results for inflammatory autoimmune disorders such as RA and systemic and polyarticular juvenile idiopathic arthritis [71].
Therefore, the search for new natural immunomodulators capable of controlling or decreasing the proliferation and differentiation of APC cells and, thus, the synthesis of cytokines responsible for maintaining chronic inflammation has become an important field of research for managing autoimmune diseases.
This is the first study that shows the anti-inflammatory properties of B. pilosa extracts on MØ polarization and the DCs’ maturation status. Currently, our research group is evaluating the cell signaling pathways involved in Bidens activity on these cells, which will complement the characterization of this activity and allow us to progress in the pre-clinical models associated with the control of inflammation in autoimmune diseases.

2.3. The Medium–Low Polarity Compounds of the Petroleum Ether Extract of B. pilosa Would Be Associated with the Induction of Anti-Inflammatory Human Macrophages and Dendritic Cells

The petroleum ether extract of B. pilosa showed the highest immunomodulatory activity among all the extracts and fractions of B. pilosa tested here. Therefore, this extract was chemically analyzed via GC-MS and was shown to contain 17 compounds. These were mainly terpenes (48%), phytosterols (27%), fatty acids (14%), and aliphatic compounds (9%).
Some of the molecules identified in the petroleum ether extract of B. pilosa, such as germacrene D, (+)-Spathulenol, α-Humulene epoxide II, Ethyl palmitate, Methyl linolenate, Ethyl linoleate, Ethyl linolenate, Stigmasterol, Beta-Sitosterol, and Friedelan-3-one, have been previously reported by other authors (Table 1) [28,72].
Table 1. Compounds of the petroleum ether extract of B. pilosa previously reported to have immunomodulatory activity. Structures taken from: pubchem.ncbi.nim.nih.gov (accessed on 7 February 2023).
Table 1. Compounds of the petroleum ether extract of B. pilosa previously reported to have immunomodulatory activity. Structures taken from: pubchem.ncbi.nim.nih.gov (accessed on 7 February 2023).
RT
(min)
Abundance
(%)
Similarity
(%)
CompoundStructureBiological ActivityReference
8.6771.1692Germacrene DMolecules 28 07094 i001Anti-inflammatory. Inhibits calcium mobilization, chemotaxis, and ROS production in neutrophils.[73]
9.9932.6089(+)-SpathulenolMolecules 28 07094 i002Antinociceptive, antiproliferative activity against lymphocytes by inducing apoptosis.[74]
10.0634.2988Caryophyllene epoxideMolecules 28 07094 i003Anti-tumor, anti-inflammatory, analgesic.[75,76]
10.2731.7491α-Humulene epoxide IIMolecules 28 07094 i004Antioxidant, anti-inflammatory. [77,78]
13.3373.2590Ethyl palmitateMolecules 28 07094 i005Anti-inflammatory, anti-arthritic, and immunomodulatory.
In vivo animal model: decreases PGE2 levels, plasma levels of TNF-α, and IL-6. Reduces NF-κB expression in liver and lung tissues and neutrophil tissue infiltration.
[79,80]
14.7971.1885Methyl linolenateMolecules 28 07094 i006Antioxidant and anti-inflammatory, decreases NO, iNOS, COX-2, and IL-1β synthesis in LPS-stimulated MØs.[81,82]
15.5773.6083Ethyl linoleateMolecules 28 07094 i007Anti-inflammatory. Down-regulates iNOS and COX-2 expression, reduces NO and PGE2 production in LPS-activated RAW 264.7 cells.[83]
15.6735.3487Ethyl linolenateMolecules 28 07094 i008--
27.62016.0685StigmasterolMolecules 28 07094 i009Antimicrobial, antioxidant.
In vitro anti-inflammatory activity against newborn mouse chondrocytes and primary cultures of IL-1β-stimulated patient-derived chondrocytes. Inhibits the NF-kB pathway and several proinflammatory mediators and mediators of matrix degradation.
Anti-inflammatory activity for microglia.
[84,85]
28.44710.3286Beta-SitosterolMolecules 28 07094 i010Anti-inflammatory.
Alleviates inflammatory response by inhibiting activation of ERK/p38 and NF-κB pathways in LPS-exposed BV2 cells.
Antiangiogenic activity in in vitro and in vivo models of rheumatoid arthritis.
[86,87]
32.4429.1085Friedelan-3-oneMolecules 28 07094 i011Antioxidant, anti-inflammatory, and immunomodulatory activities.
In vivo model: decreases DSS-induced colitis and regulates autophagy.
It is associated with the inhibition of ROS production (intracellular/extracellular), TNF-α, IL-1β, and IL-6.
It also inhibits cell proliferation.
[88,89]
Abbreviations: COX-2: Cyclooxygenase-2; DSS: Dextran sulfate sodium; IL: Interleukin; HO-1: Heme oxygenase-1; iNOS: Inducible nitric oxide synthase; LPS: Lipopolysaccharide; NF-κB: nuclear factor kappa B; NO: Nitric oxide; PGE2: Prostaglandin E2; ROS: Reactive oxygen species; RT: Retention time; TNF-α: Tumor necrosis factor alpha.
Nevertheless, novel compounds were also identified in the petroleum ether extract of B. pilosa, namely dihydroactinidiolide, ent-germacra-4(15),5,10(14)-trien-1.beta-ol, neophytadiene, 1,1,6-trimethyl-3-methylene-2-(3,6,9,13-tetramethyl-6-ethene-10,14-dimethylene-pentadec-4-enyl) cyclohexane, tetrapentacontane, and methylcomate C (Table 2).
Table 2. Novel compounds in the petroleum ether extract of B. pilosa with immunomodulatory activity. Structures taken from: pubchem.ncbi.nim.nih.gov (accessed on 7 February 2023).
Table 2. Novel compounds in the petroleum ether extract of B. pilosa with immunomodulatory activity. Structures taken from: pubchem.ncbi.nim.nih.gov (accessed on 7 February 2023).
RT
(min)
Abundance
(%)
Similarity
(%)
CompoundStructureBiological ActivityReference
9.6100.6493DihydroactinidiolideMolecules 28 07094 i012Antioxidant activity.[90]
10.8472.2382ent-Germacra-4(15),5,10(14)-trien-1.beta.-olMolecules 28 07094 i013--
11.8331.2988NeophytadieneMolecules 28 07094 i014Anti-inflammatory activity.
Inhibits the production of NO and the inflammatory cytokines TNF-α, IL-6, and IL-10 in in vitro (RAW 264.7 cells) and in vivo (Sprague Dawley rats) models.
[91]
22.8971.95851,1,6-trimethyl-3-methylene-2-(3,6,9,13-tetramethyl-6-ethenye-10,14-dimethylene-pentadec-4-enyl) cyclohexaneMolecules 28 07094 i015Antiproliferative activity against tumor cells.[92]
23.4578.8383TetrapentacontaneMolecules 28 07094 i016Antioxidant and anti-inflammatory activities.[93]
29.1235.1589Methyl commate CMolecules 28 07094 i017Antioxidant, anti-inflammatory, antidiabetic, and antihyperlipidemic activities.[94,95]
Abbreviations: IL: Interleukin; NO: Nitric oxide; ROS: Reactive oxygen species; RT: Retention time; TNF-α: Tumor necrosis factor alpha.
Some of the terpenes of B. pilosa identified in this study have anti-inflammatory activity. Friedelan-3-one, a pentacyclic triterpene, abounds in the petroleum ether extract of B. pilosa and has also been isolated from other plants, such as Azima tetracantha Lam. It exhibits anti-inflammatory, analgesic, and antipyretic effects in animal models of inflammation [96]. For example, it counteracts dextran sodium sulfate (DSS)-induced murine colitis by reducing IL-1β and IL-6 synthesis and potentiating IL-10 synthesis. Furthermore, according to network pharmacology integrated with bioinformatics, friedelan-3-one had several potential targets, such as the C-C chemokine receptor type 2 (CCR2) [89]. This receptor is essential for promoting macrophage M1/M2 polarization and inducing DC maturation, as it up-regulates the expression of costimulatory molecules [97,98]. Thus, friedelan-3-one could exert its immunomodulatory activity on MØs and DCs by binding to the CCR2 receptor.
Essential oils enriched in terpenes derived from Cannabis sativa L. (β-pinene, myrcene, cis-α-bergamotene, α-humulene, trans-caryophyllene) show anti-inflammatory activity on the murine macrophage RAW 264.7 cell line by inhibiting nitric oxide radical production [99]. Similarly, another terpene, neophytadiene, isolated from the marine algae Turbinaria ornata (50 and 100 μM/mL) inhibits NO production and induces down-regulation of iNOS and NF-κB in murine macrophage RAW 264.7 stimulated with LPS and also in an in vivo model of inflammation induced by LPS in Sprague Dawley rats [91].
Beta-caryophyllene modulates the cannabinoid CB2 receptor (CB2R) involved in M2 polarization of MØs and DC maturation [52,100,101]. However, the activity of oxidized caryophyllene on these APCs remains unclear. Although oxidized caryophyllene has no affinity for CB2R, it has shown anti-inflammatory and analgesic properties [75,76] and the ability to modulate the STAT3-mediated signaling pathway in tumor cells. Considering that this pathway is also important for MØ polarization and DC maturation [102,103,104], it is possible to hypothesize that oxidized caryophyllene could act on MØs and DCs by modulating STAT3-mediated pathways; however, studies are needed to confirm this mechanism of action.
Stigmasterol was another abundant compound identified in the petroleum ether extract of B. pilosa. It is an anti-inflammatory phytosterol that decreases neuroinflammation in in vivo and in vitro models of Alzheimer’s disease. Specifically, it attenuates the inflammatory response of microglia via NF-κB signaling and nucleotide-binding domains, leucine-rich–containing family pyrin domain–containing-3 (NLRP3) via adenine monophosphate activated protein kinase (AMPK) activation [105]. Furthermore, stigmasterol inhibits phagocytosis, NO, and TNF-α production, Cyclooxygenase (COX-2) expression, iNOS, and phosphorylated extracellular signal-regulated protein kinase (p-ERK) in LPS-activated RAW264.7 cells [106]. These findings indicate that stigmasterol could strongly inhibit M1 polarization of MØs. However, M2 polarization was not assessed in any of these studies. No data on the activity of stigmasterol on human DCs were found in the literature reviewed.
Beta-sitosterol, a sterol commonly found in plants, exhibits anti-inflammatory activity. It alleviates the inflammatory response by inhibiting the activation of ERK/p38- and NF-κB-mediated pathways in LPS-exposed microglia cells [86]. Beta-sitosterol also attenuates inflammation in an animal model of RA by modulating M2 polarization of MØs [107]. The mechanism of action responsible for this immunomodulatory activity could be associated with the affinity of beta-sitosterol to the glucocorticoid receptor (GR). The structural similarity of sitosterol to steroids and corticosteroids would explain its ability to reduce airway inflammation in in vivo models of asthma [108]. Although beta-sitosterol is not an exclusive compound of medicinal plants with immunomodulatory activity, it could act in synergy with other anti-inflammatory molecules identified in the petroleum ether extract of B. pilosa and potentiate their effects.
Macrophages express the GR; glucocorticoids such as dexamethasone act on LPS-stimulated MØs by inhibiting the p38 MAPK pathway mainly activated in M1 MØs [109,110]. A GR-mediated anti-inflammatory effect has also been evidenced in DCs that acquire tolerogenic properties when exposed to glucocorticoids [83].
Fatty acids are another group of compounds in the petroleum ether extract of B. pilosa with potential immunomodulatory activity on APCs. The literature on metabolic networks and signaling pathways involved in MØ polarization indicates that both the availability and type of substrates in the surrounding media are critical for the functionality of these cells [111]. Saturated fatty acids are known to be proinflammatory and induce M1 MØs through TLR2 and TLR4 activation. In contrast, unsaturated fatty acids inhibit these TLR-dependent signaling pathways and act as ligands of PPAR-γ, thus favoring M2 polarization of MØs [112,113,114,115]. Ethyl linoleate was a fatty acid identified in the petroleum ether extract of B. pilosa. It is also isolated from Allium sativum and down-regulates iNOS and COX-2 expression in LPS-activated RAW 264.7 cells [83]. Another fatty acid, ethyl palmitate, is reported to ameliorate carrageenan-induced rat paw edema, and reduce PGE2 levels, plasma TNF-α and IL-6 concentrations, and NF-κB expression in liver and lung cells, as well as neutrophil tissue infiltration in a rat model of LPS-induced endotoxemia [80].
Given the modulatory activity of the phytochemicals described above, it is possible to postulate that the terpenes, phytosterols, and fatty acids identified in the petroleum ether extract of B. pilosa have anti-inflammatory effects that could also modulate human MØ polarization and DC semi-mature differentiation. However, there are no specific studies on the mechanisms (receptors involved, pathway activation/inactivation, metabolic activity, protein expression, and gene activation/inactivation) by which these natural compounds could exert such immunomodulatory effects on the APC.

3. Materials and Methods

3.1. Obtaining Plant Material

The cultivation and collection of the plant material of Bidens pilosa L. was carried out in the Jorge Piñeros Corpas Medicinal Garden of the Juan N. Corpas University Foundation, in Bogotá D.C, Colombia. The taxonomic identification of the plant was carried out in the national herbarium of the Faculty of Sciences of the National University of Colombia (registration number, 609178). The use of this plant for research purposes was approved by the Ministry of Environment and Development sustainable in Colombia, under the resolution of access to genetic resources number 315 of 2021, framework Permit to Collect Specimens of Wild Species of Biological Diversity for Non-Commercial Scientific Research Purposes Resolution No. 00596 of 2018. The aerial part of the plant (leaves, stems, and flowers) were dried at 24–26 °C under aseptic conditions, then pulverized in a mill, packed, and stored in the dark at an average temperature of 19 °C.

3.2. Obtaining Extracts and Fractions

The pulverized dry aerial parts from B. pilosa sample (500 g) were mixed with petroleum ether (4 L) for fifteen days to separate the petroleum-ether-soluble and petroleum-ether-insoluble phases. The benzine-insoluble phase (more polar) was then subjected to ethanolic extraction (4 L) using a Soxhlet unit. Then, this ethanolic extract was fractionated using continuous liquid–liquid fractionation with solvents of increasing polarity. As a result, B. pilosa fractions in n-hexane, chloroform, ethyl acetate, and the hydroalcoholic residue (ethanol-water 1:1) were produced. All extracts and fractions were dissolved in dimethyl sulfoxide (DMSO).

3.3. Screening of the Immunomodulatory Activity of Bidens pilosa L. in Human PBMCs

3.3.1. Isolation and Culture of Human Peripheral Blood Mononuclear Cells

Human PBMCs were isolated from buffy coats of blood units donated to the blood bank of the District Institute of Science, Biotechnology, and Innovation in Health, Bogotá, Colombia. Buffy coats were diluted in phosphate-buffered saline 1X (PBS 1X) and centrifuged in Ficoll-Paque™ Premium (Cytiva, Uppsala, Sweden) density gradients (1.084 g/mL) at 800 gravities for 40 min at 20 °C to isolate PBMCs. Cells were washed and resuspended in Dulbecco’s modified Eagle’s medium (DMEM) (Lonza, Visp, Switzerland) supplemented with 10% fetal bovine serum (FBS) (Eurobio, Les Ulis, France), 100 U/mL penicillin, 100 µg/mL streptomycin (Corning Mediatech, Corning, NY, USA, EE. UU.), and 1 mM sodium pyruvate (Sigma Aldrich, St. Louis, MO, USA, EE. UU.). Cells were counted with trypan blue in the Neubauer chamber and the cells with a greater viability (90%) were used for cytotoxicity and cell proliferation assays.

3.3.2. Cytotoxicity Assays on PBMCs

PBMCs were cultured in 96-well plates (100,000 cells per well) and treated with different doses (1.56 µg/mL to 200 µg/mL) of extracts and fractions of B. pilosa at 37 °C, 5% CO2, for 12, 24, and 48 h. PBMCs exposed only to DMSO (vehicle control), untreated cells, or PBMCs stimulated with PHA at 0.04 μg/mL (PHA, MICROGEN LABG&M, Bogotá, Colombia) or blank reagent (well without cells), or cells treated with CUR (Sigma Aldrich, EE. UU.) or Δ9-THC (Restek, Bellefonte, PA, USA, EE. UU.) at different doses (0.015, 0.0045, and 0.135 and 3, 9, and 27 µg/mL, respectively) were run in parallel as controls. The latter were used as immunomodulatory (antiproliferative and anti-inflammatory) controls in the assays. Two independent experiments were carried out, and each concentration was tested in triplicate.
After incubation, culture media were replaced with 100 μL medium without phenol red. Then, 50 μL of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazole bromide (MTT; Sigma, St. Louis, MO, USA, EE. UU.) dissolved in PBS 1X was added, and cells were incubated at 37 °C, 5% CO2 for 4 h. The resulting formazan crystals were dissolved in DMSO before reading absorbance (Abs) at 550 nanometers (nm) on an AccuReader microplate reader, model M965 (Metertech, Taiwan). The percentages of viable cells were calculated as
(Abs of the cells that receive the treatments − Abs reagent blank) × 100)/(Abs control with DMSO − Abs reagent blank)
With these values, the inhibitory concentration 50 (IC50) for each of the plant extracts and fractions was calculated.
Although there is no agreement on IC50 values specifically indicating the exact cytotoxic figure [116], different authors consider that an IC50 < 100 μg/mL could be considered a cytotoxic concentration [117,118]. A two-way ANOVA test was used to compare the mean values for each treatment and the vehicle control, using the GraphPad Prism software version 9.0 (GraphPad Prism Inc., San Diego, CA, USA).

3.3.3. Cell Proliferation Assays on PBMCs

The synergistic or antagonistic effect of B. pilosa on cell proliferation was evaluated on human PBMC cultures stimulated with PHA at 0.04 μg/mL (PHA, LABG&M, Colombia), according to the standardization of this methodology in our laboratory, and treated with the plant extracts or fractions. Two types of experiments were performed: In the first, PBMCs were treated simultaneously with PHA and extracts or fractions of B. pilosa at different doses (1.56 to 200 µg/mL) for 12, 24, and 48 h. In the second type of experiment, the PBMCs were stimulated with PHA (0.04 μg/mL) and allowed to proliferate for 24 h before the addition of plant extracts or fractions, and then, they were incubated with these treatments for 12, 24, and 48 h. For each type of experiment, proliferation (PHA), vehicle (PHA and DMSO), cells treated with CUR or THC at different doses (0.015, 0.0045 and 0.135 and 3, 9, and 27 µg/mL, respectively), and reagent blank control cultures were run in parallel. Two independent experiments were carried out, and each concentration was tested in triplicate.
Cell proliferation was assessed via the MTT colorimetric assay as described above. The proliferation index (PI) was calculated using the following equation:
PI = (Abs of treated cells − Abs of the reagent blank)/Abs of the vehicle control.

3.4. Measurement of Cytokines in Culture Supernatants

The levels (pg/mL) of IL-1β, IL-6, IL-8, IL-10, IL-12p70, and TNF released in the supernatants of cultures were quantified using the commercial BD Cytometric Bead Array (CBA) Human Inflammatory Cytokine kit (Becton, Dickinson and Company BD Life Sciences-Biosciences, Franklin Lakes, NJ, USA, EE. UU.) in FACSAria II (Becton Dickinson, Franklin Lakes, NJ, USA, EE. UU.) citometer and FCAP array software v 3.0 (BD, Bioscience, Franklin Lakes, NJ, USA, EE. UU.) for analysis. A one-way ANOVA test was used to evaluate differences between levels of cytokines using GraphPad Prism software version 9.0.

3.5. Analysis of the Immunomodulatory Activity of B. pilosa L. on Human MØs and CDs

3.5.1. Culture and Differentiation of Human Monocyte-Derived MØs

Human CD14+ monocytes were isolated from PBMCs using the Human CD14 Beads, QuadroMACS Starting Kit (LS) (No. 130-091-051) (Miltenyi Biotec GmbH, Bergisch, Gladbach, Germany) according to the manufacturer’s instructions. The isolated monocytes were then cultured to be differentiated into MØs or DCs.Briefly, monocytes (700,000 cells per well) were cultured in 12-well plates with RPMI medium and 20 ng/mL of granulocyte-monocyte colony-stimulating factor (GM-CSF, R&D system, Minneapolis, MN, USA, EE. UU.) that favors M1 polarization (M1-preconditioned MØs), or 20 ng/mL of macrophage colony-stimulating factor (M-CSF, R&D system, EE. UU.) that favors M2 polarization (M2-preconditioned MØs) at 37 °C, 5% CO2 for six days, replacing the culture media on the third day [119,120].
The expression of common macrophage markers was evaluated on the sixth day with antibodies: CD16 APC/CY7 (Biolegend, San Diego, CA, USA, EE. UU.), CD163 PERCP (Biolegend, EE. UU.), and CD86 PE (BD Pharmingen™, Franklin Lakes, NJ, USA, EE. UU.), using FACSAria II cytometer (Becton Dickinson, EE. UU.).
At the sixth day, M1- and M2-preconditioned MØs were then exposed to B. pilosa extracts or selected fractions (40 µg/mL according to the activity observed in PBMCs) in duplicate at 37 °C, 5% CO2 for 48 h.
M1- and M2-preconditioned MØ (non-polarized or untreated), polarized M1 (addition of interferon gamma (IFN-γ at 40 ng/mL) to the M1-preconditioned MØ culture on the sixth day) (R&D system, EE. UU.), polarized M2 (addition of IL-4 at 40 ng/mL to the M2-preconditioned MØ culture on the sixth day) (R&D system, EE. UU.), vehicle (addition of DMSO on the sixth day), and CUR at 15 ng/mL controls were run in parallel. Two independent experiments were carried out in duplicate.
The ability of the petroleum ether extract of B. pilosa to modulate or reprogram M1 polarizing toward M2 was also evaluated. To this purpose, M1-preconditioned MØs were treated with IFN-γ at the sixth day for 48 h and were then treated with the petroleum ether extract of B. pilosa at 40 µg/mL or DMSO (vehicle control) or CUR at 15 ng/mL in duplicate for an additional 48 h at 37 °C, 5% CO2.
The concentration tested in duplicate was selected according to the activity observed in M1- or M2-preconditioned MØs. Staining conditions of these cells were created and analysis was carried out in the same way as previously described.

3.5.2. Culture and Differentiation of Human Monocyte-Derived DCs

CD14+ human monocytes (600,000 cells per well) were cultured in 12-well plates with RPMI medium (Lonza, Switzerland) and 100 ng/mL GM-CSF and 20 ng/mL IL-4 (R&D system, EE. UU.) at 37 °C, 5% CO2 for five days, replacing the culture media on the third day.
The immature phenotype of these DCs (CD206high, CD209high, CD86low, and HLA-DRlow) was determined via flow cytometry using CD206 PE/CY7 (Biolegend, EE. UU.), CD209 PERCP/CY5 (BD Pharmingen™, EE. UU.), and HLA-DR FITC (Biolegend, EE. UU.) antibodies using a FACSAria II cytometer (Becton Dickinson, EE. UU.).
Immature DCs were then treated with the extracts and fractions of B. pilosa, selected in duplicate, at 37 °C, 5% CO2 for 48 h. Immature DC (untreated), mature DC (addition of lipopolysaccharide [LPS] 1 μg/mL on the fifth day) (Sigma-Aldrich, EE. UU.), immature cells treated with CUR at 15 ng/mL, or vehicle (addition of DMSO on the fifth day) controls were run in parallel in two independent experiments.
The capability of the petroleum ether extract of B. pilosa to modulate the maturation process of DCs in an inflammatory context was also evaluated. To this end, immature DCs (derived from monocytes cultured with GM-CSF and IL-4 for five days) were treated on the fifth day with LPS (1 μg/mL) (maturating stimulus), LPS and either petroleum ether extract of B. pilosa or DMSO (vehicle control) at the same time, or CUR at 15 ng/mL in duplicate and further incubated at 37 °C, 5% CO2 for 48 h.

3.5.3. Characterization of the Phenotype of MØs and DCs

After incubation, MØs and DCs were phenotypically characterized by assessing cytokine synthesis and surface marker expressions. For this purpose, culture supernatants were collected for cytokine quantitation using the CBA Human Inflammatory Cytokine kit (Becton, Dickinson and Company BD Life Sciences-Biosciences, EE. UU.) and analyzed via flow cytometry using FACSAria II (Becton Dickinson, EE. UU.) and FCAP array software v 3.0 (BD, Bioscience, EE. UU.). A one-way ANOVA test was used to evaluate differences between levels of cytokines of treated cells and DMSO-treated cells using GraphPad Prism software version 9.0.
Additionally, MØs and DCs were re-collected and stained with fluorochrome-conjugated antibodies and analyzed via flow cytometry. First, Fc receptors were blocked using Human TruStain FcX™ (Biolegend, EE. UU.) according to the manufacturer’s instructions. Then, MØs were stained with anti-CD16 Alexa Fluor 700 (BD Pharmingen™, EE. UU.), anti-CD68 FITC (Biolegend, EE. UU.), anti-CD86 PE (BD Pharmingen™, EE. UU.), anti-iNOS Alexa fluor 594 (Novus Biologicals™, Centennial, CO, USA, EE. UU.), anti-CD206 PE/CY7 (Biolegend, EE. UU.), anti-CD209 PERCP (BD Pharmingen™, EE. UU.), and anti-CD163 PE (Biolegend, EE. UU.) antibodies to assess the M1 profile (CD16High, CD68High, CD86High, iNOSHigh) and M2 profile (CD16High, CD206High, CD209High, CD163High).
Similarly, DCs were stained with anti-CD86 PE (BD Pharmingen™, EE. UU.), anti-HLA-DR FITC (BD Pharmingen™, EE. UU.), anti-CD83 Alexa Fluor 700 (Invitrogen, Waltham, MA, USA, EE. UU.), anti-CD206 PE/CY7, and anti-CD209 Percp-Cy5 (BD Pharmingen™, EE. UU.) antibodies to assess their immature (CD206High, CD209High, CD86Low, CD83Low, and HLA-DRLow), semi-mature (CD206high, CD209high, CD86high, CD83high, and HLA-DRhigh), and mature (CD206low, CD209low, CD86high, CD83high, and HLA-DRhigh) profile.
Stained cells were acquired on a FACSAria II flow cytometer (Becton Dickinson), and data were analyzed using the FlowJo™ Software version 10.0 (FlowJo, Ashland, OR, USA: Becton, Dikinson and Company, Franklin Lakes, NJ, USA). Results were expressed as mean ± SD. Mean values of cultures exposed to B. pilosa and of vehicle control cultures were compared by a two-way ANOVA test using the GraphPad Prism software, v. 9.0.

3.6. Chemical Characterization through Gas Chromatography Coupled with Mass Spectrometry (GC/MS)

Chromatographic profiling and mass spectra analyses of B. pilosa of petroleum ether extract were carried out using the Shimadzu single quadrupole GCMS-QP2020 NX gas chromatograph–mass spectrometer (Shimadzu Scientific Instruments, Kyoto, Japan, MD, EE. UU.), equipped with an HP5-MS capillary column (30 m in length × 0.25 mm in diameter × 0.25 µm in thickness) (Agilent J&W Scientific, Palo Alto, CA, USA, EE. UU.). Helium gas was used as the carrier gas at a constant 1 mL/min flow rate. For GC-MS spectral detection, the spectrometer was set up in positive ionization mode with electron impact (EI) at 70 eV, in full scan mode from 100 to 800 m/z. The injection quantity of 1 μL was used (10:1 split ratio), and the injector temperature was 280 °C.
The program used was set initially at 100 °C for 3 min, gradually raised at 15 °C/min up to 220 °C, which was maintained for 3 min, and then raised at 8 °C/min up to 300 °C for 10 min. The phytochemical constituents present in the extract were tentatively identified by their retention time (RT) (min), peak area, and peak height, and by comparing mass spectral patterns with the spectral data of authentic compounds stored in the National Institute of Standards and Technology (NIST) 08 mass spectral library with a similarity percentage greater than 80%.

4. Conclusions

Extracts and fractions of different polarities obtained from B. pilosa were found to be slightly cytotoxic or non-cytotoxic to primary human cells (PBMCs and APCs). This characteristic, together with their anti-inflammatory properties, makes them a promising source of compounds to be considered in developing new immunomodulatory therapies to regulate the response of immune system cells under pathological conditions.
The low–medium polarity products of B. pilosa (petroleum ether extract and ethyl acetate fraction) showed better antiproliferative activity on human PBMCs when added concomitantly with PHA than when added to already proliferating PBMCs.
The most hydrophobic extract (petroleum ether) of B. pilosa exerted the most significant antiproliferative effect on PHA-stimulated PBMCs and was the most efficient at promiting MØ polarization into the M2 phenotype and at driving DCs to acquire a semi-mature status. These in vitro results indicate that the petroleum ether extract of B. pilosa, enriched in immunomodulatory compounds such as terpenes and fatty acids, could down-regulate the immune response by acting on different cells involved in innate and adaptive immunity.
These results were supported by the finding that this extract induced M1-preconditioned MØs to express an M1/M2 hybrid phenotype and semi-mature DCs to remain as such despite being exposed to LPS.
All the findings described above allow us to consider future research focused on the therapeutic application of the anti-inflammatory properties of B. pilosa-derived products in inflammatory settings.

Author Contributions

X.M.R.M.: Methodology, Analysis, Writing—Original Draft, and Writing—Review and Editing; L.A.C.B.: Methodology and Validation; G.M.C.: Writing—Original Draft and Writing—Review and Editing; A.L.M.: Writing—Original Draft; S.P.S.G.: Methodology Design, Supervision, Writing—Original Draft, and Writing—Review and Editing. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by the Pontificia Universidad Javeriana; the Ministry of Science, Technology and Innovation; the Ministry of National Education; the Ministry of Industry, Commerce and Tourism; and ICETEX. Second Call of the Scientific Ecosystem, Colombia Científica, 792-2017. Program “Generation of alternatives therapeutics in cancer from plants through translational research and development processes, articulated in environmentally and economically sustainable valuable systems” (Contract No. FP44842-221-2018) and the Juan N. Corpas University Foundation.

Acknowledgments

The authors thank the Juan N. Corpas University Foundation and everyone participating in this ongoing project. They also thank Martha Mesa for revising and editing the English language.

Conflicts of Interest

The authors declare no conflict of interest.

Sample Availability

Not applicable.

Abbreviations

(5FU)—5-fluorouracil, (Abs)—absorbance, (AMPK)—adenine monophosphate activated protein kinase, (APCs)—antigen-presenting cells, (CB2R)—cannabinoid CB2 receptor, (CBA)—cytometric bead array, (CCR2)—C-C chemokine receptor type 2, (COX-2)—Cyclooxygenase, (CSF)—cerebrospinal fluid, (CUR)—curcumin, (DCs)—dendritic cells, (DMSO)—dimethyl sulfoxide, (DSS)—dextran sodium sulfate, (eV)—electron Volts, (MHC)—major histocompatibility complex, (DMEM)—Dulbecco’s modified Eagle’s medium, (Min)—minutes, (FBS)—fetal bovine serum, (GR)—glucocorticoid receptor, (GM-CSF)—granulocyte-monocyte colony-stimulating factor, (h)—hours, (HO-1)—Heme oxygenase-1, (IC50)—inhibitory concentration 50, (IFN-γ)—interferon gamma, (IL)—interleukin, (iNOS)—inducible nitric oxide synthase, (LPS)—lipopolysaccharide, (M-CSF)—macrophage colony-stimulating factor, (MØs)—macrophages, (MTT)—3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazole bromide, (nm)—nanometers, (NK)—natural killer, (NO)—nitric oxide, (NLRP3)—nucleotide-binding domain, leucine-rich–containing family pyrin domain–containing-3, (NF-κB)—nuclear factor kappa B, (PAMPs)—pathogen-associated molecular patterns, (PBMCs)—peripheral blood mononuclear cells, (PBS 1X)—phosphate-buffered saline 1X, (p-ERK)—phosphorylated extracellular signal-regulated protein kinase, (PGE2)—Prostaglandin E2, (PHA)—phytohemagglutinin, (PCs)—polyacetylenes, (PI)—proliferation index, (RA)—rheumatoid arthritis, (ROS)—reactive oxygen species, (RT)—retention time, (SEL)—systemic lupus erythematosus, (STAT3)—activator of transcription 3, (T1D)—type I diabetes, (THC)—delta-9-tetrahydrocannabinol, (TLR)—toll-like receptor, (TNF)—tumor necrosis factor.

References

  1. Gea-Banacloche, J.C. Immunomodulation. In Principles of Molecular Medicine; Runge, M.S., Patterson, C., Eds.; Humana Press: Totowa, NJ, USA, 2006; pp. 893–904. [Google Scholar] [CrossRef]
  2. Masihi, K.N. Fighting infection using immunomodulatory agents. Expert. Opin. Biol. Ther. 2001, 1, 641–653. [Google Scholar] [CrossRef] [PubMed]
  3. Martin, K.; Schreiner, J.; Zippelius, A. Modulation of APC Function and Anti-Tumor Immunity by Anti-Cancer Drugs. Front. Immunol. 2015, 6, 501. [Google Scholar] [CrossRef] [PubMed]
  4. Bashaw, A.A.; Leggatt, G.R.; Chandra, J.; Tuong, Z.K.; Frazer, I.H. Modulation of antigen presenting cell functions during chronic HPV infection. Papillomavirus Res. 2017, 4, 58–65. [Google Scholar] [CrossRef] [PubMed]
  5. Banchereau, J.; Briere, F.; Caux, C.; Davoust, J.; Lebecque, S.; Liu, Y.J.; Pulendran, B.; Palucka, K. Immunobiology of dendritic cells. Annu. Rev. Immunol. 2000, 18, 767–811. [Google Scholar] [CrossRef]
  6. Murray, P.J.; Wynn, T.A. Protective and pathogenic functions of macrophage subsets. Nat. Rev. Immunol. 2011, 11, 723–737. [Google Scholar] [CrossRef] [PubMed]
  7. Moradali, M.F.; Mostafavi, H.; Ghods, S.; Hedjaroude, G.A. Immunomodulating and anticancer agents in the realm of macromycetes fungi (macrofungi). Int Immunopharmacol. 2007, 7, 701–724. [Google Scholar] [CrossRef]
  8. Coutant, F.; Miossec, P. Altered dendritic cell functions in autoimmune diseases: Distinct and overlapping profiles. Nat. Rev. Rheumatol. 2016, 12, 703–715. [Google Scholar] [CrossRef] [PubMed]
  9. Murray, P.J.; Allen, J.E.; Biswas, S.K.; Fisher, E.A.; Gilroy, D.W.; Goerdt, S.; Gordon, S.; Hamilton, J.A.; Ivashkiv, L.B.; Lawrence, T.; et al. Macrophage activation and polarization: Nomenclature and experimental guidelines. Immunity 2014, 41, 14–20. [Google Scholar] [CrossRef] [PubMed]
  10. Hashimoto, D.; Chow, A.; Greter, M.; Saenger, Y.; Kwan, W.H.; Leboeuf, M.; Ginhoux, F.; Ochando, J.C.; Kunisaki, Y.; van Rooijen, N.; et al. Pretransplant CSF-1 therapy expands recipient macrophages and ameliorates GVHD after allogeneic hematopoietic cell transplantation. J. Exp. Med. 2011, 208, 1069–1082. [Google Scholar] [CrossRef]
  11. Serbina, N.V.; Salazar-Mather, T.P.; Biron, C.A.; Kuziel, W.A.; Pamer, E.G. TNF/iNOS-producing dendritic cells mediate innate immune defense against bacterial infection. Immunity 2003, 19, 59–70. [Google Scholar] [CrossRef]
  12. de Waal Malefyt, R.; Figdor, C.G.; Huijbens, R.; Mohan-Peterson, S.; Bennett, B.; Culpepper, J.; Dang, W.; Zurawski, G.; E de Vries, J. Effects of IL-13 on phenotype, cytokine production, and cytotoxic function of human monocytes. Comparison with IL-4 and modulation by IFN-gamma or IL-10. J. Immunol. 1993, 151, 6370–6381. [Google Scholar] [CrossRef]
  13. Gordon, S. Alternative activation of macrophages. Nat. Rev. Immunol. 2003, 3, 23–35. [Google Scholar] [CrossRef] [PubMed]
  14. Sindrilaru, A.; Peters, T.; Wieschalka, S.; Baican, C.; Baican, A.; Peter, H.; Hainzl, A.; Schatz, S.; Qi, Y.; Schlecht, A.; et al. An unrestrained proinflammatory M1 macrophage population induced by iron impairs wound healing in humans and mice. J. Clin. Investig. 2011, 121, 985–997. [Google Scholar] [CrossRef] [PubMed]
  15. Wynn, T.A. Fibrotic disease and the TH1/TH2 paradigm. Nat. Rev. Immunol. 2004, 4, 583–594. [Google Scholar] [CrossRef] [PubMed]
  16. Xiao, W.; Hong, H.; Kawakami, Y.; Lowell, C.A.; Kawakami, T. Regulation of myeloproliferation and M2 macrophage programming in mice by Lyn/Hck, SHIP, and Stat5. J. Clin. Investig. 2008, 118, 924–934. [Google Scholar] [CrossRef] [PubMed]
  17. Anthony, R.M.; Urban, J.F.; Alem, F.; Hamed, H.A.; Rozo, C.T.; Boucher, J.L.; Van Rooijen, N.; Gause, W.C. Memory TH2 cells induce alternatively activated macrophages to mediate protection against nematode parasites. Nat. Med. 2006, 12, 955–960. [Google Scholar] [CrossRef]
  18. Bhatia, S.; Fei, M.; Yarlagadda, M.; Qi, Z.; Akira, S.; Saijo, S.; Iwakura, Y.; van Rooijen, N.; Gibson, G.A.; Croix, C.M.S.; et al. Rapid Host Defense against Aspergillus fumigatus Involves Alveolar Macrophages with a Predominance of Alternatively Activated Phenotype. PLoS ONE 2011, 6, e15943. [Google Scholar] [CrossRef] [PubMed]
  19. Dalod, M.; Chelbi, R.; Malissen, B.; Lawrence, T. Dendritic cell maturation: Functional specialization through signaling specificity and transcriptional programming. EMBO J. 2014, 33, 1104–1116. [Google Scholar] [CrossRef]
  20. Lutz, M.B.; Schuler, G. Immature, semi-mature and fully mature dendritic cells: Which signals induce tolerance or immunity? Trends Immunol. 2002, 23, 445–449. [Google Scholar] [CrossRef] [PubMed]
  21. Williams, C.A.; Harry, R.A.; McLeod, J.D. Apoptotic cells induce dendritic cell-mediated suppression via interferon-γ-induced IDO. Immunology 2008, 124, 89–101. [Google Scholar] [CrossRef]
  22. Mellman, I.; Steinman, R.M. Dendritic cells: Specialized and regulated antigen processing machines. Cell 2001, 106, 255–258. [Google Scholar] [CrossRef]
  23. Catanzaro, M.; Corsini, E.; Rosini, M.; Racchi, M.; Lanni, C. Immunomodulators Inspired by Nature: A Review on Curcumin and Echinacea. Molecules 2018, 23, 2778. [Google Scholar] [CrossRef]
  24. Gao, S.; Zhou, J.; Liu, N.; Wang, L.; Gao, Q.; Wu, Y.; Zhao, Q.; Liu, P.; Wang, S.; Liu, Y.; et al. Curcumin induces M2 macrophage polarization by secretion IL-4 and/or IL-13. J. Mol. Cell Cardiol. 2015, 85, 131–139. [Google Scholar] [CrossRef] [PubMed]
  25. Kim, J.Y.; Yoon, Y.D.; Ahn, J.M.; Kang, J.S.; Park, S.K.; Lee, K.; Bin Song, K.; Kim, H.M.; Han, S.-B. Angelan isolated from Angelica gigas Nakai induces dendritic cell maturation through toll-like receptor 4. Int. Immunopharmacol. 2007, 7, 78–87. [Google Scholar] [CrossRef] [PubMed]
  26. Lee, J.K.; Lee, M.K.; Yun, Y.P.; Kim, Y.; Kim, J.S.; Kim, Y.S.; Kim, K.; Han, S.S.; Lee, C.-K. Acemannan purified from Aloe vera induces phenotypic and functional maturation of immature dendritic cells. Int. Immunopharmacol. 2001, 1, 1275–1284. [Google Scholar] [CrossRef]
  27. Santander, S.P.; Aoki, M.; Hernandez, J.F.; Pombo, M.; Moins-Teisserenc, H.; Mooney, N.; Fiorentino, S. Galactomannan from Caesalpinia spinosa induces phenotypic and functional maturation of human dendritic cells. Int. Immunopharmacol. 2011, 11, 652–660. [Google Scholar] [CrossRef]
  28. Bartolome, A.P.; Villaseñor, I.M.; Yang, W.-C. Bidens pilosa L. (Asteraceae): Botanical Properties, Traditional Uses, Phytochemistry, and Pharmacology. Evid. Based Complement. Altern. Med. ECAM 2013, 2013, 340215. [Google Scholar]
  29. Dos Santos Filho, E.X.; Da Silva, A.C.G.; De Ávila, R.I.; Batista, A.C.; Marreto, R.N.; Lima, E.M.; de Oliveira, C.M.A.; Mendonça, E.F.; Valadares, M.C. Chemopreventive effects of FITOPROT against 5-fluorouracil-induced toxicity in HaCaT cells. Life Sci. 2018, 193, 300–308. [Google Scholar] [CrossRef] [PubMed]
  30. Quaglio, A.E.V.; Cruz, V.M.; Almeida-Junior, L.D.; Costa, C.A.R.A.; Di Stasi, L.C. Bidens pilosa (Black Jack) Standardized Extract Ameliorates Acute TNBS-induced Intestinal Inflammation in Rats. Planta Med. 2020, 86, 319–330. [Google Scholar] [CrossRef] [PubMed]
  31. Zhang, C.; Li, K.; Yang, Z.; Wang, Y.; Si, H. The Effect of the Aqueous Extract of Bidens Pilosa L. on Androgen Deficiency Dry. Eye in Rats. Cell Physiol. Biochem. Int. J. Exp. Cell Physiol. Biochem. Pharmacol. 2016, 39, 266–277. [Google Scholar] [CrossRef]
  32. Fotso, A.; Longo, F.; Djomeni, P.; Kouam, S.; Spiteller, M.; Dongmo, A.; Savineau, J.P. Analgesic and antiinflammatory activities of the ethyl acetate fraction of Bidens pilosa (Asteraceae). Inflammopharmacology 2014, 22, 105–114. [Google Scholar] [CrossRef]
  33. 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]
  34. Rodríguez-Mesa, X.M.; Contreras Bolaños, L.A.; Mejía, A.; Pombo, L.M.; Modesti Costa, G.; Santander González, S.P. Immunomodulatory Properties of Natural Extracts and Compounds Derived from Bidens pilosa L.: Literature Review. Pharmaceutics 2023, 15, 1491. [Google Scholar] [CrossRef] [PubMed]
  35. Kviecinski, M.R.; Benelli, P.; Felipe, K.B.; Correia JF, G.; Pich, C.T.; Ferreira, S.R.S.; Pedrosa, R. SFE from Bidens pilosa Linné to obtain extracts rich in cytotoxic polyacetylenes with antitumor activity. J. Supercrit. Fluids. 2011, 56, 243–248. [Google Scholar] [CrossRef]
  36. Chang, J.S.; Chiang, L.C.; Chen, C.C.; Liu, L.T.; Wang, K.C.; Lin, C.C. Antileukemic activity of Bidens pilosa L. var. minor (Blume) Sherff and Houttuynia cordata Thunb. Am. J. Chin. Med. 2001, 29, 303–312. [Google Scholar] [CrossRef]
  37. Abajo, C.; Ángeles Boffill, M.; del Campo, J.; Alexandra Méndez, M.; González, Y.; Mitjans, M.; Vinardell, M.P. In vitro study of the antioxidant and immunomodulatory activity of aqueous infusion of Bidens pilosa. J. Ethnopharmacol. 2004, 93, 319–323. [Google Scholar] [CrossRef] [PubMed]
  38. Chung, C.Y.; Yang, W.C.; Liang, C.L.; Liu, H.Y.; Lai, S.K.; Chang, C.L.T. Data on the effect of Cytopiloyne against Listeria monocytogenes infection in mice. Data Brief 2016, 7, 995–998. [Google Scholar] [CrossRef] [PubMed]
  39. Chung, C.Y.; Yang, W.C.; Liang, C.L.; Liu, H.Y.; Lai, S.K.; Chang, C.L.T. Cytopiloyne, a polyacetylenic glucoside from Bidens pilosa, acts as a novel anticandidal agent via regulation of macrophages. J. Ethnopharmacol. 2016, 184, 72–80. [Google Scholar] [CrossRef] [PubMed]
  40. Yang, W.C.; Yang, C.Y.; Liang, Y.C.; Yang, C.W.; Li, W.Q.; Chung, C.Y.; Yang, M.-T.; Kuo, T.-F.; Lin, C.-F.; Liang, C.-L.; et al. Anti-coccidial properties and mechanisms of an edible herb, Bidens pilosa, and its active compounds for coccidiosis. Sci. Rep. 2019, 9, 2896. [Google Scholar] [CrossRef] [PubMed]
  41. Abiodun, O.O.; Sosanya, A.S.; Nwadike, N.; Oshinloye, A.O. Beneficial effect of Bidens pilosa L. (Asteraceae) in a rat model of colitis. J. Basic Clin. Physiol. Pharmacol. 2020, 31. [Google Scholar] [CrossRef] [PubMed]
  42. Corre, I.; Pineau, D.; Hermouet, S. Interleukin-8: An autocrine/paracrine growth factor for human hematopoietic progenitors acting in synergy with colony stimulating factor-1 to promote monocyte-macrophage growth and differentiation. Exp. Hematol. 1999, 27, 28–36. [Google Scholar] [CrossRef] [PubMed]
  43. Li, A.; Dubey, S.; Varney, M.L.; Dave, B.J.; Singh, R.K. IL-8 Directly Enhanced Endothelial Cell Survival, Proliferation, and Matrix Metalloproteinases Production and Regulated Angiogenesis1. J. Immunol. 2003, 170, 3369–3376. [Google Scholar] [CrossRef] [PubMed]
  44. Xu, H.; Lai, W.; Zhang, Y.; Liu, L.; Luo, X.; Zeng, Y.; Wu, H.; Lan, Q.; Chu, Z. Tumor-associated macrophage-derived IL-6 and IL-8 enhance invasive activity of LoVo cells induced by PRL-3 in a KCNN4 channel-dependent manner. BMC Cancer 2014, 14, 330. [Google Scholar] [CrossRef]
  45. Zheng, T.; Ma, G.; Tang, M.; Li, Z.; Xu, R. IL-8 Secreted from M2 Macrophages Promoted Prostate Tumorigenesis via STAT3/MALAT1 Pathway. Int. J. Mol. Sci. 2018, 20, 98. [Google Scholar] [CrossRef] [PubMed]
  46. Casella, G.; Garzetti, L.; Gatta, A.T.; Finardi, A.; Maiorino, C.; Ruffini, F.; Martino, G.; Muzio, L.; Furlan, R. IL4 induces IL6-producing M2 macrophages associated to inhibition of neuroinflammation in vitro and in vivo. J. Neuroinflamm. 2016, 13, 139. [Google Scholar] [CrossRef] [PubMed]
  47. Scheller, J.; Chalaris, A.; Schmidt-Arras, D.; Rose-John, S. The pro- and anti-inflammatory properties of the cytokine interleukin-6. Biochim. Biophys. Acta BBA Mol. Cell Res. 2011, 1813, 878–888. [Google Scholar] [CrossRef] [PubMed]
  48. Lutz, M. Therapeutic Potential of Semi-Mature Dendritic Cells for Tolerance Induction. Front. Immunol. 2012, 3, 123. [Google Scholar] [CrossRef] [PubMed]
  49. Dudek, A.M.; Martin, S.; Garg, A.D.; Agostinis, P. Immature, Semi-Mature, and Fully Mature Dendritic Cells: Toward a DC-Cancer Cells Interface That Augments Anticancer Immunity. Front. Immunol. 2013, 4, 438. [Google Scholar] [CrossRef] [PubMed]
  50. Witherel, C.E.; Sao, K.; Brisson, B.K.; Han, B.; Volk, S.W.; Petrie, R.J.; Han, L.; Spiller, K.L. Regulation of extracellular matrix assembly and structure by hybrid M1/M2 macrophages. Biomaterials 2021, 269, 120667. [Google Scholar] [CrossRef] [PubMed]
  51. Henriquez, J.E.; Crawford, R.B.; Kaminski, N.E. Suppression of CpG-ODN-mediated IFNα and TNFα response in human plasmacytoid dendritic cells (pDC) by cannabinoid receptor 2 (CB2)-specific agonists. Toxicol. Appl. Pharmacol. 2019, 369, 82–89. [Google Scholar] [CrossRef] [PubMed]
  52. Rodríguez Mesa, X.M.; Moreno Vergara, A.F.; Contreras Bolaños, L.A.; Guevara Moriones, N.; Mejía Piñeros, A.L.; Santander González, S.P. Therapeutic Prospects of Cannabinoids in the Immunomodulation of Prevalent Autoimmune Diseases. Cannabis Cannabinoid Res. 2021, 6, 196–210. [Google Scholar] [CrossRef] [PubMed]
  53. Tsokos, G.C.; Lo, M.S.; Costa Reis, P.; Sullivan, K.E. New insights into the immunopathogenesis of systemic lupus erythematosus. Nat. Rev. Rheumatol. 2016, 12, 716–730. [Google Scholar] [CrossRef] [PubMed]
  54. Hirano, T. IL-6 in inflammation, autoimmunity and cancer. Int. Immunol. 2021, 33, 127–148. [Google Scholar] [CrossRef] [PubMed]
  55. Wewers, M.D.; Dare, H.A.; Winnard, A.V.; Parker, J.M.; Miller, D.K. IL-1 beta-converting enzyme (ICE) is present and functional in human alveolar macrophages: Macrophage IL-1 beta release limitation is ICE independent. J. Immunol. 1997, 159, 5964–5972. [Google Scholar] [CrossRef] [PubMed]
  56. Zhao, R.; Zhou, H.; Su, S.B. A critical role for interleukin-1β in the progression of autoimmune diseases. Int. Immunopharmacol. 2013, 17, 658–669. [Google Scholar] [CrossRef]
  57. Sparre, T.; Bjerre Christensen, U.; Gotfredsen, C.F.; Mose Larsen, P.; Fey, S.J.; Hjernø, K. Changes in expression of IL-1β influenced proteins in transplanted islets during development of diabetes in diabetes-prone BB rats. Diabetologia 2004, 47, 892–908. [Google Scholar]
  58. Corbett, J.A.; Kwon, G.; Turk, J.; McDaniel, M.L. IL-1.beta. induces the coexpression of both nitric oxide synthase and cyclooxygenase by Islets of Langerhans: Activation of cyclooxygenase by nitric oxide. Biochemistry 1993, 32, 13767–13770. [Google Scholar] [CrossRef] [PubMed]
  59. Dogan, Y.; Akarsu, S.; Ustundag, B.; Yilmaz, E.; Gurgoze, M.K. Serum IL-1β, IL-2, and IL-6 in Insulin-Dependent Diabetic Children. Mediat. Inflamm. 2006, 2006, e59206. [Google Scholar] [CrossRef] [PubMed]
  60. Oberholzer, A.; Oberholzer, C.; Moldawer, L.L. Cytokine signaling-regulation of the immune response in normal and critically ill states. Crit. Care Med. 2000, 28, N3. [Google Scholar] [CrossRef] [PubMed]
  61. Rabinovitch, A. An update on cytokines in the pathogenesis of insulin-dependent diabetes mellitus. Diabetes Metab. Rev. 1998, 14, 129–151. [Google Scholar] [CrossRef]
  62. Reddy, S.; Young, M. IL-1β Expression in Islet Cells of the NOD Mouse and Its Spatial Relationship to Beta Cells and Inducible Nitric Oxide Synthase. Ann. NY Acad. Sci. 2002, 958, 190–193. [Google Scholar] [CrossRef] [PubMed]
  63. Cannella, B.; Raine, C.S. The adhesion molecule and cytokine profile of multiple sclerosis lesions. Ann. Neurol. 1995, 37, 424–435. [Google Scholar] [CrossRef] [PubMed]
  64. Olmos, G.; Lladó, J. Tumor Necrosis Factor Alpha: A Link between Neuroinflammation and Excitotoxicity. Mediat. Inflamm. 2014, 2014, 861231. [Google Scholar] [CrossRef] [PubMed]
  65. Bansard, C.; Lequerré, T.; Derambure, C.; Vittecoq, O.; Hiron, M.; Daragon, A.; Pouplin, S.; Daveau, M.; Boyer, O.; Tron, F.; et al. Gene profiling predicts rheumatoid arthritis responsiveness to IL-1Ra (anakinra). Rheumatology 2011, 50, 283–292. [Google Scholar] [CrossRef]
  66. Olsen, N.J.; Stein, C.M. New Drugs for Rheumatoid Arthritis. N. Engl. J. Med. 2004, 350, 2167–2179. [Google Scholar] [CrossRef] [PubMed]
  67. Smolen, J.S.; Aletaha, D.; Koeller, M.; Weisman, M.H.; Emery, P. New therapies for treatment of rheumatoid arthritis. Lancet 2007, 370, 1861–1874. [Google Scholar] [CrossRef]
  68. Adegbola, S.O.; Sahnan, K.; Warusavitarne, J.; Hart, A.; Tozer, P. Anti-TNF Therapy in Crohn’s Disease. Int. J. Mol. Sci. 2018, 19, 2244. [Google Scholar] [CrossRef] [PubMed]
  69. Levin, A.D.; Wildenberg, M.E.; van den Brink, G.R. Mechanism of Action of Anti-TNF Therapy in Inflammatory Bowel Disease. J. Crohns Colitis. 2016, 10, 989–997. [Google Scholar] [CrossRef]
  70. Sands, B.E.; Kaplan, G.G. The Role of TNFα in Ulcerative Colitis. J. Clin. Pharmacol. 2007, 47, 930–941. [Google Scholar] [CrossRef]
  71. Kishimoto, T.; Kang, S.; Tanaka, T. IL-6: A New Era for the Treatment of Autoimmune Inflammatory Diseases. In Innovative Medicine; Nakao, K., Minato, N., Uemoto, S., Eds.; Springer: Tokyo, Japan, 2015; pp. 131–147. [Google Scholar]
  72. Geissberger, P.; Séquin, U. Constituents of Bidens pilosa L.: Do the components found so far explain the use of this plant in traditional medicine? Acta Trop. 1991, 48, 251–261. [Google Scholar] [CrossRef] [PubMed]
  73. Schepetkin, I.A.; Özek, G.; Özek, T.; Kirpotina, L.N.; Khlebnikov, A.I.; Quinn, M.T. Chemical Composition and Immunomodulatory Activity of Hypericum perforatum Essential Oils. Biomolecules 2020, 10, 916. [Google Scholar] [CrossRef] [PubMed]
  74. Ziaei, A.; Ramezani, M.; Wright, L.; Paetz, C.; Schneider, B.; Amirghofran, Z. Identification of spathulenol in Salvia mirzayanii and the immunomodulatory effects. Phytother. Res. PTR. 2011, 25, 557–562. [Google Scholar] [CrossRef] [PubMed]
  75. Fidyt, K.; Fiedorowicz, A.; Strządała, L.; Szumny, A. β-caryophyllene and β-caryophyllene oxide—Natural compounds of anticancer and analgesic properties. Cancer Med. 2016, 5, 3007–3017. [Google Scholar] [CrossRef] [PubMed]
  76. Moghrovyan, A.; Parseghyan, L.; Sevoyan, G.; Darbinyan, A.; Sahakyan, N.; Gaboyan, M.; Karabekian, Z.; Voskanyan, A. Antinociceptive, anti-inflammatory, and cytotoxic properties of Origanum vulgare essential oil, rich with β-caryophyllene and β-caryophyllene oxide. Korean J. Pain 2022, 35, 140–151. [Google Scholar] [CrossRef] [PubMed]
  77. Sharma, P.; Shah, G.C. Composition and antioxidant activity of Senecio nudicaulis Wall. ex DC. (Asteraceae): A medicinal plant growing wild in Himachal Pradesh, India. Nat. Prod. Res. 2015, 29, 883–886. [Google Scholar] [CrossRef] [PubMed]
  78. Shebaby, W.; Saliba, J.; Faour, W.H.; Ismail, J.; El Hage, M.; Daher, C.F.; Taleb, R.I.; Nehmeh, B.; Dagher, C.; Chrabieh, E.; et al. In vivo and in vitro anti-inflammatory activity evaluation of Lebanese Cannabis sativa L. ssp. indica (Lam.). J. Ethnopharmacol. 2021, 270, 113743. [Google Scholar] [CrossRef]
  79. Jiménez-Ferrer, E.; Vargas-Villa, G.; Martínez-Hernández, G.B.; González-Cortazar, M.; Zamilpa, A.; García-Aguilar, M.P.; Arenas-Ocampo, M.L.; Herrera-Ruiz, M. Fatty-Acid-Rich Agave angustifolia Fraction Shows Antiarthritic and Immunomodulatory Effect. Molecules 2022, 27, 7204. [Google Scholar] [CrossRef]
  80. Saeed, N.M.; El-Demerdash, E.; Abdel-Rahman, H.M.; Algandaby, M.M.; Al-Abbasi, F.A.; Abdel-Naim, A.B. Anti-inflammatory activity of methyl palmitate and ethyl palmitate in different experimental rat models. Toxicol. Appl. Pharmacol. 2012, 264, 84–93. [Google Scholar] [CrossRef] [PubMed]
  81. Fu, Y.P.; Yuan, H.; Xu, Y.; Liu, R.M.; Luo, Y.; Xiao, J.H. Protective effects of Ligularia fischeri root extracts against ulcerative colitis in mice through activation of Bcl-2/Bax signalings. Phytomedicine Int. J. Phytother. Phytopharm. 2022, 99, 154006. [Google Scholar] [CrossRef] [PubMed]
  82. Kim, T.H.; Truong, V.L.; Jeong, W.S. Phytochemical Composition and Antioxidant and Anti-Inflammatory Activities of Ligularia fischeri Turcz: A Comparison between Leaf and Root Extracts. Plants 2022, 11, 3005. [Google Scholar] [CrossRef] [PubMed]
  83. Park, S.Y.; Seetharaman, R.; Ko, M.J.; Kim, D.Y.; Kim, T.H.; Yoon, M.K.; Kwak, J.H.; Lee, S.J.; Bae, Y.S.; Choi, Y.W. Ethyl linoleate from garlic attenuates lipopolysaccharide-induced pro-inflammatory cytokine production by inducing heme oxygenase-1 in RAW264.7 cells. Int. Immunopharmacol. 2014, 19, 253–261. [Google Scholar] [CrossRef] [PubMed]
  84. Gabay, O.; Sanchez, C.; Salvat, C.; Chevy, F.; Breton, M.; Nourissat, G.; Wolf, C.; Jacques, C.; Berenbaum, F. Stigmasterol: A phytosterol with potential anti-osteoarthritic properties. Osteoarthr. Cartil. 2010, 18, 106–116. [Google Scholar] [CrossRef] [PubMed]
  85. Swamy, M.K.; Arumugam, G.; Kaur, R.; Ghasemzadeh, A.; Yusoff, M.M.; Sinniah, U.R. GC-MS Based Metabolite Profiling, Antioxidant and Antimicrobial Properties of Different Solvent Extracts of Malaysian Plectranthus amboinicus Leaves. Evid. Based Complement. Alternat Med. 2017, 2017, e1517683. [Google Scholar] [CrossRef]
  86. Sun, Y.; Gao, L.; Hou, W.; Wu, J. β-Sitosterol Alleviates Inflammatory Response via Inhibiting the Activation of ERK/p38 and NF-κB Pathways in LPS-Exposed BV2 Cells. BioMed Res. Int. 2020, 2020, 7532306. [Google Scholar] [CrossRef] [PubMed]
  87. Qian, K.; Zheng, X.X.; Wang, C.; Huang, W.G.; Liu, X.B.; Xu, S.D.; Liu, D.-K.; Liu, M.-Y.; Lin, C.-S. β-Sitosterol Inhibits Rheumatoid Synovial Angiogenesis Through Suppressing VEGF Signaling Pathway. Front. Pharmacol. 2022, 12, 816477. [Google Scholar] [CrossRef] [PubMed]
  88. Feigni, O.Y.M.; Mbiantcha, M.; Yousseu, W.N.; Tsafack, G.E.; Djuichou, F.N.S.; Noungoua, C.M.; Atsafack, C.M.L.; Ateufack, G. Immunomodulatory, anti-infammatory and antioxidant activities of aqueous and ethanolic extracts of Cissus quadrangularis Linn. (Vitaceae) in chronic pai. Eur. PMC 2022. [Google Scholar] [CrossRef]
  89. Shi, B.; Liu, S.; Huang, A.; Zhou, M.; Sun, B.; Cao, H.; Shan, J.; Sun, B.; Lin, J. Revealing the Mechanism of Friedelin in the Treatment of Ulcerative Colitis Based on Network Pharmacology and Experimental Verification. Evid. Based Complement. Altern. Med. ECAM. 2021, 2021, 4451779. [Google Scholar] [CrossRef] [PubMed]
  90. Das, M.; Devi, K.P. Dihydroactinidiolide regulates Nrf2/HO-1 expression and inhibits caspase-3/Bax pathway to protect SH-SY5Y human neuroblastoma cells from oxidative stress induced neuronal apoptosis. Neurotoxicology 2021, 84, 53–63. [Google Scholar] [CrossRef] [PubMed]
  91. Bhardwaj, M.; Sali, V.K.; Mani, S.; Vasanthi, H.R. Neophytadiene from Turbinaria ornata Suppresses LPS-Induced Inflammatory Response in RAW 264.7 Macrophages and Sprague Dawley Rats. Inflammation 2020, 43, 937–950. [Google Scholar] [CrossRef] [PubMed]
  92. Mishra, S.; Verma, S.S.; Rai, V.; Awasthee, N.; Arya, J.S.; Maiti, K.K.; Gupta, S.C. Curcuma raktakanda Induces Apoptosis and Suppresses Migration in Cancer Cells: Role of Reactive Oxygen Species. Biomolecules 2019, 9, 159. [Google Scholar] [CrossRef] [PubMed]
  93. Tanod, W.A.; Yanuhar, U.; Maftuch Putra, M.Y.; Risjani, Y. Screening of NO Inhibitor Release Activity from Soft Coral Extracts Origin Palu Bay, Central Sulawesi, Indonesia. Anti-Inflamm. Anti-Allergy Agents Med. Chem. 2019, 18, 126–141. [Google Scholar] [CrossRef] [PubMed]
  94. Gairola, K.; Gururani, S.; Kumar, R.; Prakash, O.; Agarwal, S.; Dubey, S. Phytochemical composition antioxidant and anti-inflammatory activities of essential oil of Acmella uliginosa (Sw.) Cass. grown in North India Terai region of Uttarakhand. Trends Phytochem Res. 2021, 5, 44–52. [Google Scholar] [CrossRef]
  95. Vats, S.; Gupta, T. Evaluation of bioactive compounds and antioxidant potential of hydroethanolic extract of Moringa oleifera Lam. from Rajasthan, India. Physiol. Mol. Biol. Plants 2017, 23, 239–248. [Google Scholar] [CrossRef]
  96. Antonisamy, P.; Duraipandiyan, V.; Ignacimuthu, S. Anti-inflammatory, analgesic and antipyretic effects of friedelin isolated from Azima tetracantha Lam. in mouse and rat models. J. Pharm. Pharmacol. 2011, 63, 1070–1077. [Google Scholar] [CrossRef] [PubMed]
  97. Deci, M.B.; Ferguson, S.W.; Scatigno, S.L.; Nguyen, J. Modulating Macrophage Polarization through CCR2 Inhibition and Multivalent Engagement. Mol. Pharm. 2018, 15, 2721–2731. [Google Scholar] [CrossRef]
  98. Jimenez, F.; Quinones, M.P.; Martinez, H.G.; Estrada, C.A.; Clark, K.; Garavito, E.; Ibarra, J.; Melby, P.C.; Ahuja, S.S. CCR2 Plays a Critical Role in Dendritic Cell Maturation: Possible Role of, C.C.L.2.; NF-κB. J. Immunol. 2010, 184, 5571–5581. [Google Scholar] [CrossRef] [PubMed]
  99. Gallily, R.; Yekhtin, Z.; Hanuš, L.O. The Anti-Inflammatory Properties of Terpenoids from Cannabis. Cannabis Cannabinoid Res. 2018, 3, 282–290. [Google Scholar] [CrossRef] [PubMed]
  100. Gaffal, E.; Kemter, A.M.; Scheu, S.; Leite Dantas, R.; Vogt, J.; Baune, B.; Tüting, T.; Zimmer, A.; Alferink, J. Cannabinoid Receptor 2 Modulates Maturation of Dendritic Cells and Their Capacity to Induce Hapten-Induced Contact Hypersensitivity. Int. J. Mol. Sci. 2020, 21, 475. [Google Scholar] [CrossRef] [PubMed]
  101. Tortora, C.; Di Paola, A.; Argenziano, M.; Creoli, M.; Marrapodi, M.M.; Cenni, S.; Tolone, C.; Rossi, F.; Strisciuglio, C. Effects of CB2 Receptor Modulation on Macrophage Polarization in Pediatric Celiac Disease. Biomedicines 2022, 10, 874. [Google Scholar] [CrossRef] [PubMed]
  102. Balic, J.J.; Albargy, H.; Luu, K.; Kirby, F.J.; Jayasekara, W.S.N.; Mansell, F.; Garama, D.J.; De Nardo, D.; Baschuk, N.; Louis, C.; et al. STAT3 serine phosphorylation is required for TLR4 metabolic reprogramming and IL-1β expression. Nat. Commun. 2020, 11, 3816. [Google Scholar] [CrossRef]
  103. Sulczewski, F.B.; Martino, L.A.; Salles, D.; Yamamoto, M.M.; Rosa, D.S.; Boscardin, S.B. STAT3 signaling modulates the immune response induced after antigen targeting to conventional type 1 dendritic cells through the DEC205 receptor. Front. Immunol. 2022, 13, 1006996. [Google Scholar] [CrossRef] [PubMed]
  104. Yuan, F.; Fu, X.; Shi, H.; Chen, G.; Dong, P.; Zhang, W. Induction of Murine Macrophage M2 Polarization by Cigarette Smoke Extract via the JAK2/STAT3 Pathway. PLoS ONE 2014, 9, e107063. [Google Scholar] [CrossRef] [PubMed]
  105. Jie, F.; Yang, X.; Yang, B.; Liu, Y.; Wu, L.; Lu, B. Stigmasterol attenuates inflammatory response of microglia via NF-κB and NLRP3 signaling by AMPK activation. Biomed. Pharmacother. 2022, 153, 113317. [Google Scholar] [CrossRef] [PubMed]
  106. Yuan, L.; Zhang, F.; Shen, M.; Jia, S.; Xie, J. Phytosterols Suppress Phagocytosis and Inhibit Inflammatory Mediators via ERK Pathway on LPS-Triggered Inflammatory Responses in RAW264.7 Macrophages and the Correlation with Their Structure. Foods 2019, 8, 582. [Google Scholar] [CrossRef]
  107. Liu, R.; Hao, D.; Xu, W.; Li, J.; Li, X.; Shen, D.; Sheng, K.; Zhao, L.; Xu, W.; Gao, Z.; et al. β-Sitosterol modulates macrophage polarization and attenuates rheumatoid inflammation in mice. Pharm. Biol. 2019, 57, 161–168. [Google Scholar] [CrossRef] [PubMed]
  108. Xu, J.; Yang, L.; Lin, T. β-sitosterol targets glucocorticoid receptor to reduce airway inflammation and remodeling in allergic asthma. Pulm. Pharmacol. Ther. 2023, 78, 102183. [Google Scholar] [CrossRef] [PubMed]
  109. Baumann, D.; Drebant, J.; Hägele, T.; Burger, L.; Serger, C.; Lauenstein, C.; Dudys, P.; Erdmann, G.; Offringa, R. p38 MAPK signaling in M1 macrophages results in selective elimination of M2 macrophages by MEK inhibition. J. Immunother. Cancer 2021, 9, e002319. [Google Scholar] [CrossRef]
  110. Bhattacharyya, S.; Brown, D.E.; Brewer, J.A.; Vogt, S.K.; Muglia, L.J. Macrophage glucocorticoid receptors regulate Toll-like receptor 4–mediated inflammatory responses by selective inhibition of p38 MAP kinase. Blood 2007, 109, 4313–4319. [Google Scholar] [CrossRef]
  111. Castoldi, A.; Naffah de Souza, C.; Câmara, N.O.S.; Moraes-Vieira, P.M. The Macrophage Switch in Obesity Development. Front. Immunol. 2016, 6, 637. [Google Scholar] [CrossRef] [PubMed]
  112. Hwang, D.H.; Kim, J.A.; Lee, J.Y. Mechanisms for the activation of Toll-like receptor 2/4 by saturated fatty acids and inhibition by docosahexaenoic acid. Eur. J. Pharmacol. 2016, 785, 24–35. [Google Scholar] [CrossRef] [PubMed]
  113. Marion-Letellier, R.; Savoye, G.; Ghosh, S. Fatty acids, eicosanoids and PPAR gamma. Eur. J. Pharmacol. 2016, 785, 44–49. [Google Scholar] [CrossRef]
  114. Varga, T.; Czimmerer, Z.; Nagy, L. PPARs are a unique set of fatty acid regulated transcription factors controlling both lipid metabolism and inflammation. Biochim. Biophys. Acta 2011, 1812, 1007–1022. [Google Scholar] [CrossRef] [PubMed]
  115. Wu, H.M.; Ni, X.X.; Xu, Q.Y.; Wang, Q.; Li, X.Y.; Hua, J. Regulation of lipid-induced macrophage polarization through modulating peroxisome proliferator-activated receptor-gamma activity affects hepatic lipid metabolism via a Toll-like receptor 4/NF-κB signaling pathway. J. Gastroenterol. Hepatol. 2020, 35, 1998–2008. [Google Scholar] [CrossRef]
  116. Velandia, S.A.; Quintero, E.; Stashenko, E.E.; Ocazionez, R.E. Actividad antiproliferativa de aceites esenciales de plantas cultivadas en Colombia. Acta Biológica Colomb. 2018, 23, 189–198. [Google Scholar] [CrossRef]
  117. Ferraz, R.P.C.; Bomfim, D.S.; Carvalho, N.C.; Soares, M.B.P.; da Silva, T.B.; Machado, W.J.; Prata, A.P.N.; Costa, E.V.; Moraes, V.R.S.; Nogueira, P.C.L.; et al. Cytotoxic effect of leaf essential oil of Lippia gracilis Schauer (Verbenaceae). Phytomed. Int. J. Phytother. Phytopharm. 2013, 20, 615–621. [Google Scholar] [CrossRef]
  118. Mazzio, E.A.; Soliman, K.F.A. In vitro screening for the tumoricidal properties of international medicinal herbs. Phytother. Res. PTR 2009, 23, 385–398. [Google Scholar] [CrossRef] [PubMed]
  119. Zarif, J.C.; Hernandez, J.R.; Verdone, J.E.; Campbell, S.P.; Drake, C.G.; Pienta, K.J. A phased strategy to differentiate human CD14+monocytes into classically and alternatively activated macrophages and dendritic cells. BioTechniques 2016, 61, 33–41. [Google Scholar] [CrossRef] [PubMed]
  120. Mia, S.; Warnecke, A.; Zhang, X.M.; Malmström, V.; Harris, R.A. An optimized protocol for human M2 macrophages using M-CSF and IL-4/IL-10/TGF-β yields a dominant immunosuppressive phenotype. Scand. J. Immunol. 2014, 79, 305–314. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Effects of B. pilosa L. on human PBMCs. (A). Heat maps of percentage (%) of viable PBMCs treated at different concentrations of petroleum ether extract (2), ethyl acetate fraction (3.3), or hydroalcoholic fraction (3.4) of B. pilosa and cultured at 37 °C, 5% CO2 for 48 h. Control cultures were prepared with untreated PBMCs, PBMCs treated with curcumin (CUR) or delta-9-tetrahydrocannabinol (Δ9-THC), or DMSO (vehicle control). The percentage of viable cells relative to the vehicle control was determined via the colorimetric MTT assay. (B). Heat maps of proliferation index (PI) of PBMCs were stimulated with PHA (0.04 μg/mL) and simultaneously exposed to different concentrations of CUR, Δ9-THC or petroleum ether extract (2), or ethyl acetate fraction (3) or hydroalcoholic fraction (4) of B. pilosa and cultured at 37 °C, 5% CO2 for 48 h. (C). Heat maps of PI of PBMCs that were stimulated with PHA (0.04 μg/mL) at 37 °C, 5% CO2 for 24 h, and then exposed to the extracts of B. pilosa or controls, as described in (B). Then, the PI was determined via the MTT colorimetric assay, relative to the respective vehicle control. Statistical significance for all experiments was obtained using two-way ANOVA and Dunnet’s post hoc multiple comparison test (* p < 0.05, ** p < 0.01, *** p < 0.001).
Figure 1. Effects of B. pilosa L. on human PBMCs. (A). Heat maps of percentage (%) of viable PBMCs treated at different concentrations of petroleum ether extract (2), ethyl acetate fraction (3.3), or hydroalcoholic fraction (3.4) of B. pilosa and cultured at 37 °C, 5% CO2 for 48 h. Control cultures were prepared with untreated PBMCs, PBMCs treated with curcumin (CUR) or delta-9-tetrahydrocannabinol (Δ9-THC), or DMSO (vehicle control). The percentage of viable cells relative to the vehicle control was determined via the colorimetric MTT assay. (B). Heat maps of proliferation index (PI) of PBMCs were stimulated with PHA (0.04 μg/mL) and simultaneously exposed to different concentrations of CUR, Δ9-THC or petroleum ether extract (2), or ethyl acetate fraction (3) or hydroalcoholic fraction (4) of B. pilosa and cultured at 37 °C, 5% CO2 for 48 h. (C). Heat maps of PI of PBMCs that were stimulated with PHA (0.04 μg/mL) at 37 °C, 5% CO2 for 24 h, and then exposed to the extracts of B. pilosa or controls, as described in (B). Then, the PI was determined via the MTT colorimetric assay, relative to the respective vehicle control. Statistical significance for all experiments was obtained using two-way ANOVA and Dunnet’s post hoc multiple comparison test (* p < 0.05, ** p < 0.01, *** p < 0.001).
Molecules 28 07094 g001
Figure 2. Effects of the petroleum ether extract and the ethyl acetate and hydroalcoholic fractions of B. pilosa on the polarization of M1-preconditioned MØs. Human M1-preconditioned MØs (derived from monocytes cultured with 20 ng/mL of GM-CSF for six days) were exposed to petroleum ether extract (2), the ethyl acetate fraction (3.3), or the hydroalcoholic fraction (3.4) of B. pilosa and cultured at 37 °C, 5% CO2 for 48 h. M1-preconditioned MØ (non-polarized or untreated), polarized M1 (addition of IFN-γ at 40 ng/mL on the sixth day), M1-preconditioned treated with curcumin (CUR) at 15 ng/mL (immunomodulator control), and vehicle (addition of DMSO on the sixth day) MØ culture controls were run in parallel. Macrophage phenotype was assessed by staining with fluorescent antibodies and flow cytometry. Culture supernatants were collected for cytokine quantification using CBA kit and flow cytometry. (A). Representative histograms comparing MØ phenotypic markers. (B). Representative contour plots showing co-expression of MØ phenotypic markers. (C). Column graph shows the percentages (mean ± SD) of MØ (CD16+ cells) expressing M1 or M2 phenotypic markers. (D). Column graph shows the cytokine levels in culture supernatants (mean ± SD). * p < 0.05, *** p < 0.001. One-way or two-way ANOVA tests and Dunnet’s post hoc multiple comparison test. n = 2.
Figure 2. Effects of the petroleum ether extract and the ethyl acetate and hydroalcoholic fractions of B. pilosa on the polarization of M1-preconditioned MØs. Human M1-preconditioned MØs (derived from monocytes cultured with 20 ng/mL of GM-CSF for six days) were exposed to petroleum ether extract (2), the ethyl acetate fraction (3.3), or the hydroalcoholic fraction (3.4) of B. pilosa and cultured at 37 °C, 5% CO2 for 48 h. M1-preconditioned MØ (non-polarized or untreated), polarized M1 (addition of IFN-γ at 40 ng/mL on the sixth day), M1-preconditioned treated with curcumin (CUR) at 15 ng/mL (immunomodulator control), and vehicle (addition of DMSO on the sixth day) MØ culture controls were run in parallel. Macrophage phenotype was assessed by staining with fluorescent antibodies and flow cytometry. Culture supernatants were collected for cytokine quantification using CBA kit and flow cytometry. (A). Representative histograms comparing MØ phenotypic markers. (B). Representative contour plots showing co-expression of MØ phenotypic markers. (C). Column graph shows the percentages (mean ± SD) of MØ (CD16+ cells) expressing M1 or M2 phenotypic markers. (D). Column graph shows the cytokine levels in culture supernatants (mean ± SD). * p < 0.05, *** p < 0.001. One-way or two-way ANOVA tests and Dunnet’s post hoc multiple comparison test. n = 2.
Molecules 28 07094 g002
Figure 3. Effects of the petroleum ether extract and the ethyl acetate and hydroalcoholic fractions of B. pilosa on the polarization of M2-preconditioned MØs. Human M2-preconditioned MØs (derived from monocytes cultured with 20 ng/mL of M-CSF for six days) were exposed to petroleum ether extract (2), the ethyl acetate fraction (3.3), or the hydroalcoholic fraction (3.4) of B. pilosa and cultured at 37 °C, 5% CO2 for 48 h. M2-preconditioned (non-polarized or untreated), polarized M2 (addition of IL-4 at 40 ng/mL on the sixth day), M2 treated with curcumin (CUR) at 15 ng/mL (immunomodulator control), and vehicle (addition of DMSO on the sixth day) MØ culture controls were run in parallel. Macrophage phenotype was assessed by staining with fluorescent antibodies and using flow cytometry. Culture supernatants were collected for cytokine quantification using CBA kit and flow cytometry. (A). Representative histograms comparing MØ phenotypic markers. (B). Representative contour plots showing co-expression of MØ phenotypic markers. (C). Column graph shows the percentages (mean ± SD) of MØ (CD16+ cells) expressing M1 or M2 phenotypic markers. (D). Column graph shows the cytokine levels in culture supernatants (mean ± SD). * p < 0.05, ** p < 0.01, *** p < 0.001. One-way or two-way ANOVA tests and Dunnet’s post hoc multiple comparison test.
Figure 3. Effects of the petroleum ether extract and the ethyl acetate and hydroalcoholic fractions of B. pilosa on the polarization of M2-preconditioned MØs. Human M2-preconditioned MØs (derived from monocytes cultured with 20 ng/mL of M-CSF for six days) were exposed to petroleum ether extract (2), the ethyl acetate fraction (3.3), or the hydroalcoholic fraction (3.4) of B. pilosa and cultured at 37 °C, 5% CO2 for 48 h. M2-preconditioned (non-polarized or untreated), polarized M2 (addition of IL-4 at 40 ng/mL on the sixth day), M2 treated with curcumin (CUR) at 15 ng/mL (immunomodulator control), and vehicle (addition of DMSO on the sixth day) MØ culture controls were run in parallel. Macrophage phenotype was assessed by staining with fluorescent antibodies and using flow cytometry. Culture supernatants were collected for cytokine quantification using CBA kit and flow cytometry. (A). Representative histograms comparing MØ phenotypic markers. (B). Representative contour plots showing co-expression of MØ phenotypic markers. (C). Column graph shows the percentages (mean ± SD) of MØ (CD16+ cells) expressing M1 or M2 phenotypic markers. (D). Column graph shows the cytokine levels in culture supernatants (mean ± SD). * p < 0.05, ** p < 0.01, *** p < 0.001. One-way or two-way ANOVA tests and Dunnet’s post hoc multiple comparison test.
Molecules 28 07094 g003
Figure 4. Effects of the petroleum ether extract and the ethyl acetate and hydroalcoholic fractions of B. pilosa on the maturation status of human DCs. Human immature DCs were exposed to petroleum ether extract (2), the ethyl acetate fraction (3.3), or the hydroalcoholic fraction (3.4) of B. pilosa at 37 °C, 5% CO2 for 48 h. Immature (untreated), mature (addition of LPS at 1 μg/mL on the fifth day), treated with curcumin (CUR) at 15 ng/mL (immunomodulator control), and vehicle (addition of DMSO on the fifth day) DC control cultures were run in parallel. Dendritic cell phenotype was assessed by staining with fluorescent antibodies and using flow cytometry. Culture supernatants were collected for cytokine quantification using CBA and flow cytometry. (A). Representative histograms comparing DC phenotypic markers. (B). Representative contour plots showing co-expression of DC phenotypic markers. (C). Column graphs show the percentages (mean ± SD) of total CD206+CD209+ DCs and CD206+CD209+ subsets in the CD83+, CD86+, or HLA-DR+ DCs. (D). Column graphs show cytokine levels (mean ± SD) in culture supernatants. * p < 0.05, ** p < 0.01, *** p < 0.001. One-way or two-way ANOVA tests and Dunnet’s post hoc multiple comparison test.
Figure 4. Effects of the petroleum ether extract and the ethyl acetate and hydroalcoholic fractions of B. pilosa on the maturation status of human DCs. Human immature DCs were exposed to petroleum ether extract (2), the ethyl acetate fraction (3.3), or the hydroalcoholic fraction (3.4) of B. pilosa at 37 °C, 5% CO2 for 48 h. Immature (untreated), mature (addition of LPS at 1 μg/mL on the fifth day), treated with curcumin (CUR) at 15 ng/mL (immunomodulator control), and vehicle (addition of DMSO on the fifth day) DC control cultures were run in parallel. Dendritic cell phenotype was assessed by staining with fluorescent antibodies and using flow cytometry. Culture supernatants were collected for cytokine quantification using CBA and flow cytometry. (A). Representative histograms comparing DC phenotypic markers. (B). Representative contour plots showing co-expression of DC phenotypic markers. (C). Column graphs show the percentages (mean ± SD) of total CD206+CD209+ DCs and CD206+CD209+ subsets in the CD83+, CD86+, or HLA-DR+ DCs. (D). Column graphs show cytokine levels (mean ± SD) in culture supernatants. * p < 0.05, ** p < 0.01, *** p < 0.001. One-way or two-way ANOVA tests and Dunnet’s post hoc multiple comparison test.
Molecules 28 07094 g004
Figure 5. Effects of the petroleum ether extract of B. pilosa on M1 polarization of macrophages and maturation of LPS-stimulated dendritic cells. (A). Human M1-preconditioned MØs were exposed to IFN-γ at 40 ng/mL (M1-polarizing stimulus) for 48 h and then treated with curcumin at 15 ng/mL (CUR) (immunomodulator control), petroleum ether extract (2) of B. pilosa, or DMSO (vehicle control) for an additional 48 h, incubated at 37 °C, 5% CO2. Column graphs show the percentages (mean ± SD) of MØs (CD16+ cells) expressing M1 or M2 phenotypic markers. Also, representative contour plots of MØs co-expressing M1 or M2 markers are shown. (B). Column graphs show cytokine levels (mean ± SD) in MØ culture supernatants. (C). Human immature DCs were exposed to LPS at 1 μg/mL (maturating stimulus) and the CUR at 15 ng/mL or petroleum ether extract (2) of B. pilosa at the same time, or DMSO (vehicle control) on fifth day and further incubated at 37 °C, 5% CO2 for 48 h. DCs were evaluated by staining with fluorescent antibodies and using flow cytometry. Column graphs show the percentages (mean ± SD) of total CD206+CD209+ DCs and CD206+CD209+ subsets in the CD83+, CD86+, or HLA-DR+ DCs. Also, representative contour plots of DCs co-expressing phenotypic markers are shown. (D). Column graphs show cytokine levels (mean ± SD) in DC culture supernatants (* p < 0.05, ** p < 0.01, *** p < 0.001). One-way or two-way ANOVA tests and Dunnet’s post hoc multiple comparison test.
Figure 5. Effects of the petroleum ether extract of B. pilosa on M1 polarization of macrophages and maturation of LPS-stimulated dendritic cells. (A). Human M1-preconditioned MØs were exposed to IFN-γ at 40 ng/mL (M1-polarizing stimulus) for 48 h and then treated with curcumin at 15 ng/mL (CUR) (immunomodulator control), petroleum ether extract (2) of B. pilosa, or DMSO (vehicle control) for an additional 48 h, incubated at 37 °C, 5% CO2. Column graphs show the percentages (mean ± SD) of MØs (CD16+ cells) expressing M1 or M2 phenotypic markers. Also, representative contour plots of MØs co-expressing M1 or M2 markers are shown. (B). Column graphs show cytokine levels (mean ± SD) in MØ culture supernatants. (C). Human immature DCs were exposed to LPS at 1 μg/mL (maturating stimulus) and the CUR at 15 ng/mL or petroleum ether extract (2) of B. pilosa at the same time, or DMSO (vehicle control) on fifth day and further incubated at 37 °C, 5% CO2 for 48 h. DCs were evaluated by staining with fluorescent antibodies and using flow cytometry. Column graphs show the percentages (mean ± SD) of total CD206+CD209+ DCs and CD206+CD209+ subsets in the CD83+, CD86+, or HLA-DR+ DCs. Also, representative contour plots of DCs co-expressing phenotypic markers are shown. (D). Column graphs show cytokine levels (mean ± SD) in DC culture supernatants (* p < 0.05, ** p < 0.01, *** p < 0.001). One-way or two-way ANOVA tests and Dunnet’s post hoc multiple comparison test.
Molecules 28 07094 g005
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

Rodríguez Mesa, X.M.; Contreras Bolaños, L.A.; Modesti Costa, G.; Mejia, A.L.; Santander González, S.P. A Bidens pilosa L. Non-Polar Extract Modulates the Polarization of Human Macrophages and Dendritic Cells into an Anti-Inflammatory Phenotype. Molecules 2023, 28, 7094. https://doi.org/10.3390/molecules28207094

AMA Style

Rodríguez Mesa XM, Contreras Bolaños LA, Modesti Costa G, Mejia AL, Santander González SP. A Bidens pilosa L. Non-Polar Extract Modulates the Polarization of Human Macrophages and Dendritic Cells into an Anti-Inflammatory Phenotype. Molecules. 2023; 28(20):7094. https://doi.org/10.3390/molecules28207094

Chicago/Turabian Style

Rodríguez Mesa, Xandy Melissa, Leonardo Andres Contreras Bolaños, Geison Modesti Costa, Antonio Luis Mejia, and Sandra Paola Santander González. 2023. "A Bidens pilosa L. Non-Polar Extract Modulates the Polarization of Human Macrophages and Dendritic Cells into an Anti-Inflammatory Phenotype" Molecules 28, no. 20: 7094. https://doi.org/10.3390/molecules28207094

Article Metrics

Back to TopTop