Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Moringa isothiocyanate-1 inhibits LPS-induced inflammation in mouse myoblasts and skeletal muscle

  • Badi Sri Sailaja,

    Roles Data curation, Formal analysis, Investigation, Methodology, Visualization, Writing – review & editing

    Affiliation Department of Plant Biology, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, United States of America

  • Sohaib Hassan,

    Roles Data curation, Formal analysis, Investigation, Software, Visualization

    Affiliation Department of Genetics and the Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway Township, New Jersey, United States of America

  • Evan Cohen,

    Roles Data curation, Formal analysis, Investigation, Software

    Affiliation Department of Genetics and the Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway Township, New Jersey, United States of America

  • Irina Tmenova,

    Roles Data curation, Formal analysis, Investigation

    Affiliation Department of Plant Biology, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, United States of America

  • Renalison Farias-Pereira,

    Roles Visualization, Writing – original draft, Writing – review & editing

    Affiliation Department of Plant Biology, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, United States of America

  • Michael P. Verzi,

    Roles Methodology, Resources, Supervision, Validation, Writing – review & editing

    Affiliation Department of Genetics and the Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway Township, New Jersey, United States of America

  • Ilya Raskin

    Roles Conceptualization, Funding acquisition, Project administration, Resources, Supervision, Writing – review & editing

    raskin@rutgers.edu

    Affiliation Department of Plant Biology, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, United States of America

Abstract

This study aims to investigate the anti-inflammatory effects of moringa isothiocyanate-1 (MIC-1) extracted from seeds of Moringa oleifera Lam. in lipopolysaccharide (LPS)-induced inflammation models. MIC-1 decreased nitric oxide production and reduced the expression of pro-inflammatory markers (TNF-α, Ifn-α, IL-1β, IL-6) in C2C12 myoblasts. The daily oral treatment of MIC-1 (80 mg/kg) for three days significantly reduced the expression of pro-inflammatory markers in gastrocnemius muscle tissue of LPS-treated C57BL/6 male mice. Transcriptomic analysis provided further insights into the inhibitory effects of MIC-1 on the LPS-induced inflammation, which suggested that MIC-1 affects inflammation and immunity-related genes in myoblasts and skeletal muscle tissue. MIC-1 inhibited the nuclear accumulation of the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) in the LPS-treated myoblasts. Our data support the hypothesis that the MIC-1’s effects in the muscle cells are mediated through the inhibition of the NF-κB translocation in the nucleus, which, in turn, results in immunomodulating and anti-inflammatory responses at the gene expression levels.

Introduction

Moringa isothiocyanate-1 (MIC-1, also known as moringin) is an isothiocyanate that has similar biological activities as other natural isothiocyanates, such as sulforaphane from cruciferous vegetables. Isothiocyanates are the class of plant secondary metabolites originating from the sulfur-containing compounds glucosinolates. MIC-1 is found in Moringa oleifera Lam., a plant used as food and medicine, with concentrations up to 40% in the hydro-alcoholic moringa seed extract [1]. In contrast to liquid sulforaphane, MIC-1 contains a sugar group that makes it a stable solid at room temperatures [2]. MIC-1’s chemical structure also improves its in vivo stability and bioavailability [2, 3]. Sixty percent of MIC-1, applied as pure compound or as a component of moringa seed extract, was not degraded in the in vitro gastrointestinal digestion model [3]. Orally administered MIC-1, up to 100 mg/kg bw/d, did not cause toxic effects in rats [4]. These properties make MIC-1 a potentially useful addition to the human diet.

Previous reports showed that MIC-1 has anti-inflammatory, antioxidant, and antimicrobial activities [1, 2, 58]. The biological activities of moringa isothiocyanates, including MIC-1, have been recently reviewed by Lopez-Rodriguez et. al. [9]. In vitro studies showed that MIC-1 inhibits pro-inflammatory pathways [1, 7, 8, 1014], and reduces lipopolysaccharide (LPS)-induced inflammation by inhibiting nuclear factor kappa B (NF-κB) pathway in macrophages [8]. NF-κB is a transcription factor that induces inflammation by upregulating cytokines, such as the tumor necrosis factor alpha (TNF-α), interleukins (IL), and interferons (Ifn). Consistently, previous studies have shown that MIC-1 decreases pro-inflammatory cytokines (TNF-α, IL-6, IL-1β, and Ifn-α) in macrophages [2, 7, 8, 1012]. MIC-1 reduced the production of nitric oxide (NO), a pro-inflammatory marker, by downregulating the inducible nitric oxide synthase (iNOS) gene in LPS-treated macrophages [1]. MIC-1 inhibited inflammatory pathways, including LPS, NF- κB, IL-1, NO signaling pathways in skin carcinogenesis and nephropathy models in vitro as well [13, 14].

The anti-inflammatory mechanisms of MIC-1 have also been investigated by using in vivo models. We previously showed that the daily oral supplementation with MIC-1 (80 mg/kg bw) for 3 days protects from systemic inflammation induced by LPS in mice [8]. We used transcriptomics analyses and targeted molecular biology to show that MIC-1 downregulated inflammatory pathways and pro-inflammatory cytokines reversing the LPS-induced inflammatory response in spleen, liver, kidney and colon [8]. Others have shown that MIC-1 (10 mg/kg bw; ip) reduced pro-inflammatory cytokines in brain in a murine model for Parkinson’s disease [10], and the transdermal application of 2% MIC-1, as a cream, had anti-inflammatory effects in spinal cord tissue in a murine model for multiple sclerosis [15]. Other studies have suggested that MIC-1, administered in moringa extracts, reduces inflammatory markers in blood, adipose tissue, liver, and intestine in mice with diet-induced metabolic dysfunction [6, 16]. Although there is evidence that MIC-1 acts as anti-inflammatory compound in several models, there is no report that investigated if MIC-1’s anti-inflammatory effects extend to skeletal muscle.

Since MIC-1 and other isothiocyanates have anti-inflammatory properties, MIC-1 may be used to promote skeletal muscle health. Pre-clinical and clinical studies have suggested that sulforaphane ameliorates muscle damage caused by inflammation [1720]. Sulforaphane inhibited NF- κB pathway, including the levels of IL-1β and IL-6, in a genetic model for muscle dystrophy [17]. Also, MIC-1 may improve skeletal muscle health due to its antioxidant effects, as seen in moringa leaf extracts-treated C2C12 cells [2124]. For instance, moringa leaf extract reduced oxidative stress induced by hydrogen peroxide in C2C12 myotubes [24]. Skeletal muscle plays a major role in a host response to sepsis contributing to signaling pathways of inflammation and metabolic dysfunctions [25, 26]. Since others have shown that both C2C12 myoblasts in vitro and gastrocnemius muscle in vivo are responsive to LPS-induced inflammation by increasing the pro-inflammatory cytokines, such as TNF-α and IL-6 [27], we decided to evaluate the effects of MIC-1 on LPS-induced inflammation in mouse myoblasts and gastrocnemius muscle tissue. Our current study confirms that MIC-1 reduces LPS-induced muscle inflammation by downregulating pro-inflammatory cytokines via NF-κB pathway, and MIC-1 likely inhibits other inflammation-associated muscle disorders as well.

Materials and methods

Materials

MIC-1 (purity >98%) was isolated from Moringa oleifera seed extract as previously reported [1]. All other materials were purchased from commercial suppliers. C2C12 cells were obtained from American Type Culture Collection (ATCC, Manassas, VA, USA). C57BL/6 male mice were acquired from Charles River Laboratories (Malvern, PA, USA). Cell culture materials: Dulbecco’s modified Eagle’s medium (DMEM), penicillin G, streptomycin, and fetal bovine serum (FBS) were obtained from Gibco Inc. (Grand Island, NY). 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) was obtained from Cayman Chemical (Ann Arbor, MI, USA) and Griess reagent system was acquired from Promega Corporation (Madison, WI, USA). Lipopolysaccharide (LPS, Escherichia coli 0111: B4), dimethyl sulfoxide (DMSO), and other chemicals were purchased from Sigma (St. Louis, MO, USA). Gene probes were obtained from Integrated DNA Technologies (Coralville, IA, USA), and the probe sequences are the same as our previous study [8]. RNeasy Mini Kit was obtained from Qiagen (Germantown, MD, USA). Anti-NF-κB p65 antibody (ab16502) was obtained from Abcam (Cambridge, MA, USA), and 4′,6-diamidino-2-phenylindole (DAPI) and Alexa Fluor™ 680 phalloidin were purchased from Invitrogen (Carlsbad, CA, USA).

In vitro assays

C2C12 myoblasts were cultured in a growth medium (90% DMEM, 10% FBS, 100 units/mL penicillin, and 100 μg/mL streptomycin) at 37°C in a humidified atmosphere with 5% carbon dioxide. MIC-1 solutions were made in ethanol. Myoblasts were treated with 0.475% ethanol (vehicle), or 1–10 μM MIC-1, with or without 1 μg/mL LPS for 24 h before the in vitro assays.

Cell viability was measured by using MTT, which was added to the growth medium to create purple formazan crystals [1]. Absorbance values were obtained by spectrophotometric measurements at 570 nm, and values were reported as % of control (vehicle).

Nitric oxide (NO) production was evaluated by using Griess method [1]. The concentrations of NO in the cell media were calculated by using nitrite as standard. Absorbance was read at 550 nm, and values were reported as % of LPS-treated group.

Immunofluorescence staining was used for NF-κB nuclear translocation assessment as previously described [8]. Fixed cells were stained with anti-NF-κB p65 antibody with fluorescent counterstains for F-actin and DAPI. The percentage of nucleic positive cells to NF-kB (p65) was quantified by counting 100 cells per field in three separate experiments [28].

Animals and tissue collection

Eighteen C57BL/6 male mice were randomly separated into vehicle control, LPS-treated, or LPS-MIC-1-treated group as previously described [8]. MIC-1 (80 mg/kg bw) was orally administered by gavage daily for 3 days before LPS treatment (10 mg/kg bw; ip). Gastrocnemius muscles were collected from euthanized mice after 16 h of LPS treatment. Fat and connective tissues were trimmed from muscles before freezing in liquid nitrogen for further analyses. The handling and care of animals used in this study followed the standard procedures recommended by the Guide for the Care and Use of Laboratory Animals of the National Institutes of Health. All efforts were made to minimize suffering and distress. No animal showed behavioral and physiological signs of pain and distress including changes in feeding pattern, body temperature, and social interaction after 16 h of LPS treatment. Animals were sacrificed by carbon dioxide asphyxiation. The protocol was approved by the Rutgers University Institutional Animal Care and Use Committee (ID999900474).

RNA sequencing and analysis

RNA-sequencing data (Gene Expression Omnibus accession #GSE209706) were analyzed by using the transcript abundances from pseudoalignments in Kallisto (v0.46.11), an RNA-seq quantification program [8, 29]. The differentially expressed genes were indicated by the normalized fragments per kilobase reads per million (FPKM) values. FPKM values (> 1) were analyzed by using tximport package (v 1.18.02) in R (v 4.0.3), and DESeq2 (v 1.30.13) to determine the differences in gene expression (P-value < 0.05, or < 0.001; absolute log2-fold change > 1; FPKM > 1) [30, 31]. Morpheus heat mapping software (Broad Institute) was used to illustrate the identified clusters of differentially expressed genes by using k-means clustering analysis. Gene Ontology (GO) analysis by the Database for Annotation, Visualization and Integrated Discovery (DAVID, v6.84) was used to identify the biological processes of genes, expressed as negative log10 (P-values). Reproducibility of RNA-seq data is shown in principal component analysis (PCA) plots (S1 Fig); DESeq2 analysis of triplicates for C2C12 samples and muscle tissue samples was conducted to create PCA plots. The data were first turned into a DESeq2 object, then ’regularized log’ transformed and plotted in a PCA plot.

Gene expression

Reverse transcription quantitative real-time PCR was performed to measure the expression of pro-inflammatory genes as previously described [8]. Power SYBR Green PCR Master Mix was used to detect the amplification of the target genes by using the QuantStudio™ 3 Real-Time PCR System (Applied Biosystems, Waltham, MA, USA). GAPDH gene expression was used as housekeeping gene to normalize the expression of target genes.

Statistical analysis

Data are expressed as mean ± standard deviation (SD) of three experiments in triplicate. One-way analysis variance or Student’s t-test were used to determine statistical differences (P < 0.05) by using GraphPad (GraphPad, La Jolla, CA).

Results

Effect of MIC-1 on LPS-induced inflammation in C2C12 murine muscle cells

To assess the cytotoxicity of MIC-1, we performed a cell viability assay in C2C12 murine cells treated with MIC-1 (1, 5 or 10 μM). MIC-1 did not significantly affect the cellular viability at the tested concentrations (Fig 1A). Next, we evaluated the effects of MIC-1 on NO production with LPS (1 μg/mL) and found a significant dose dependent reduction in NO production (Fig 1B). Furthermore, we found that 1 μM MIC-1 downregulated significantly the pro-inflammatory genes TNF-α, Ifn-α, IL-1β, and IL-6 (Fig 1C). These results suggested that MIC-1 inhibits LPS-induced inflammation without major effects on the viability of muscle cells.

thumbnail
Fig 1. Effect of moringa isothiocyanate-1 (MIC-1) in C2C12 myoblasts.

Cells were treated with MIC-1 (1, 5 or 10 μM), or 0.475% ethanol (vehicle), with or without 1 μg/mL lipopolysaccharide (LPS) for 24 h. (A) Cell viability. (B) Nitric oxide (NO) production. (C) Gene expression of pro-inflammatory markers (TNF-α, Ifn-α, IL-1β and IL-6). Data are represented as mean ± S.D. (n = 3). * P < 0.05, obtained by t-test when compared to LPS-treated group.

https://doi.org/10.1371/journal.pone.0279370.g001

Transcriptome analysis of MIC-1 in LPS-induced inflammation muscle cells

Transcriptomic analysis of 1 μM MIC-1 and LPS-treated myoblasts was performed to evaluate the effects of MIC-1 on inflammation in C2C12 skeletal muscle cells. Control samples were compared to LPS-treated samples to assess the effects of LPS, and LPS-treated samples were compared to LPS-MIC-1-treated samples to assess the effects of MIC-1 on the LPS-induced inflammation. As shown in Fig 2A, 1397 genes were upregulated, and 1336 genes were downregulated due to LPS treatment (control vs LPS-treated samples). A total of 2509 genes were upregulated, and 2510 genes were downregulated due to MIC-1 treatment (LPS-treated samples vs LPS-MIC-1-treated samples) (Fig 2A). The differentially expressed genes (adjusted P-value < 0.001 and FPKM > 1), identified by DESeq2 analysis using the pairwise comparisons (control vs LPS-treated samples; LPS-treated samples vs LPS-MIC-1-treated samples), were further clustered by using k-means to identify clusters of genes showing common responses to these treatments. This list of 3,567 genes were clustered in five clusters (Fig 2B). Cluster V was particularly interesting, as 1,104 genes were induced by LPS and subsequently repressed by the MIC-1 treatment (Fig 2B). GO analysis of these genes revealed that functions such as innate immune response, inflammatory response, and immune system processes were induced by LPS, and subsequently repressed by MIC-1 (Fig 2C; S1 File). These results suggest that the anti-inflammatory properties of MIC-1 in the LPS-treated myoblasts result from a direct effect of MIC-1 on the inflammation and immunity pathways.

thumbnail
Fig 2. Transcriptome analysis of moringa isothiocyanate-1 (MIC-1) in C2C12 myoblasts.

(A) Venn diagrams depict genes significantly (absolute log2-fold change > 1; Adj. P < 0.001) upregulated/downregulated due to LPS (control vs. lipopolysaccharide (LPS)-treated samples) or MIC-1 (LPS-treated samples vs. LPS-MIC-1-treated samples) in C2C12 murine muscle cells. (B) Heat map of differentially expressed genes in five k-means clusters. Column, row, and color represent a sample, gene, and expression level, respectively. (C) Gene ontology (GO) analysis of upregulated genes due to LPS (control vs. LPS-treated samples) and downregulated genes due to MIC-1 (LPS-treated samples vs. LPS-MIC-1-treated samples), represented as negative log10 (P-values).

https://doi.org/10.1371/journal.pone.0279370.g002

Transcriptome analysis of MIC-1 effect in the LPS-induced muscle tissue

MIC-1 (80 mg/kg bw) was orally administered to C57BL/6 male mice for three days before LPS treatment (10 mg/kg bw; ip). Thereafter, the gastrocnemius muscle tissues of animals were collected 16 h after the LPS injection for transcriptomic analyses by RNA-seq (Fig 3A). We compared untreated control vs LPS-treated samples, and LPS-treated samples vs LPS-MIC-1-treated samples to understand the response to LPS and its attenuation by MIC-1 in the skeletal muscle tissue. Our results showed that 116 genes were upregulated due to LPS (control vs LPS-treated samples) (Fig 3B). A total of 1374 genes were upregulated, and 952 genes were downregulated due to MIC-1 (LPS-treated samples vs LPS-MIC-1-treated samples) (Fig 3B). The differentially expressed genes (adjusted P-value < 0.05), identified by DESeq2 analysis using the pairwise comparisons (control vs LPS-treated samples; LPS-treated samples vs LPS-MIC-1-treated samples), were further clustered by using k-means to identify clusters of genes showing common responses to these treatments. Five k-means clusters were obtained from this list of 1,049 genes (Fig 3C). GO analysis of the upregulated genes by LPS showed that the biological functions (GO terms) were related to lipid, protein, and redox metabolism (Fig 3D). GO analysis showed that biological pathways suppressed by MIC-1 were associated with immune system and inflammatory responses in the skeletal muscle tissue (Fig 3D; S2 File). These results support the hypothesis that MIC-1 attenuates the LPS-induced inflammation in the skeletal muscle tissue.

thumbnail
Fig 3. Transcriptome analysis of moringa isothiocyanate-1 (MIC-1) in muscle tissue.

(A) Schematic diagram of the experimental protocol. MIC-1 was given orally daily for 3 days to C57BL/6 male mice before lipopolysaccharide (LPS) treatment. Mice were randomly divided into three groups: control (10% DMSO; p.o.); LPS (10 mg/kg bw; ip); LPS + MIC-1 (80 mg/kg bw; p.o.). Muscle tissues were collected 16 h after LPS treatment (n = 3). (B) Venn diagrams depict genes significantly (absolute log2-fold change > 1; P < 0.05) upregulated/downregulated due to LPS (control vs. LPS-treated samples) or MIC-1 (LPS-treated samples vs. LPS-MIC-1-treated samples) in murine gastrocnemius tissues. (C) Heat map of differentially expressed genes in five k-means clusters. Column, row, and color represent a sample, gene, and expression level, respectively. (D) Gene ontology (GO) analysis of upregulated genes due to LPS (control vs. LPS-treated samples), and downregulated genes due to MIC-1 (LPS-treated samples vs. LPS-MIC-1-treated samples), represented as negative log10 (P-values).

https://doi.org/10.1371/journal.pone.0279370.g003

MIC-1 inhibits LPS-induced acute inflammation in mice

To validate the transcriptomic analyses, we evaluated the effects of MIC-1 treatment on the pro-inflammatory cytokines in murine muscle tissues by RT-qPCR. As expected, our results showed that MIC-1 decreased LPS-induced gene expressions of TNF-α, Ifn-α, IL-1β, and IL-6 in muscle tissue (Fig 4). These results corroborated with our transcriptomic analysis by RNA-Seq data showing the suppression of inflammatory cascades by MIC-1.

thumbnail
Fig 4. Moringa isothiocyanate-1 (MIC-1) reduced lipopolysaccharide (LPS)-induced pro-inflammatory markers in muscle tissue.

TNF-α, Ifn-α, IL-1β, and IL-6 mRNA levels in the gastrocnemius muscle tissues treated with LPS and/or MIC-1 were measured by RT-qPCR. Data are represented as mean ± S.D. (n = 3). * P < 0.05, obtained by t-test when compared to LPS-treated group.

https://doi.org/10.1371/journal.pone.0279370.g004

MIC-1 inhibited the NF-κB nuclear translocation in skeletal muscle cells

NF-κB is transcription factor that regulates pro-inflammatory cytokines, and our previous study showed that MIC-1 inhibited the nuclear translocation of NF-κB in macrophages [8]. To test whether MIC-1 inhibits nuclear translocation of NF-κB, we performed immunofluorescence staining of NF-κB in C2C12 myoblasts. Confocal microscope imaging showed that LPS increased the nuclear translocation of NF-κB, which was inhibited by 1 μM MIC-1 in an hour (Fig 5). These results further confirm that the inhibition of NF-κB pathway is part of the mechanisms of action of MIC-1 on inflammation in skeletal muscle.

thumbnail
Fig 5. Moringa isothiocyanate-1 (MIC-1) inhibited the NF-κB nuclear translocation in skeletal muscle cells.

The effects of MIC-1 on lipopolysaccharide (LPS)-induced nuclear translocation of NF-κB in C2C12 muscle cells. Treatments: untreated control, LPS (10 μg/mL), and LPS + 1 μM MIC-1 for 1 h. The NF-κB (p65) location was observed using FITC-anti-NF-κB p65 antibody, phalloidin-actin (F-actin; cytoskeleton), and DAPI (nuclei). (A) Representative pictures taken by using confocal microscopy. Scale bar: 20 μm. (B) The percentage of nucleic positive cells to NF-κB (p65). Data are represented as mean ± SD (n = 6, from three independent experiments). * P < 0.05, obtained by t-test when compared to LPS-treated group.

https://doi.org/10.1371/journal.pone.0279370.g005

Discussion

MIC-1 is a chemically stable anti-inflammatory isothiocyanate from M. oleifera seeds. Our previous studies documented that MIC-1 activates anti-inflammatory processes in macrophages by inhibiting the NF-κB pathway [2, 7, 8]. However, the mechanisms of the anti-inflammatory action of MIC-1 in skeletal muscles have not been investigated. Since inflammation is an underlying cause of multiple muscle disorders, this study uses global transcriptomic analyses, selective gene expression analyses, and immunofluorescence staining to elucidate MIC-1’s anti-inflammatory mechanisms using the LPS-induced inflammation models in murine C2C12 myoblasts and skeletal muscle tissue. Our results showed that MIC-1 suppressed the LPS-induced inflammation by decreasing the nuclear translocation of NF-κB in the skeletal muscle cells. MIC-1’s anti-inflammatory properties were confirmed in skeletal muscle of mice, suggesting that the oral administration of MIC-1 prevents systemic inflammation.

LPS, a membrane component of Gram-negative bacteria, is used to induce inflammation and sepsis in pre-clinical studies [1, 2, 7, 8, 1012, 25]. The induced innate immune response is initiated by recognizing LPS via Toll-like receptors (TLR), which activate NF-κB pathway inducing the expression of cytokines (e.g., TNF-α, IL-6 and IL-1β) to protect against infections [26, 32, 33]. Our data confirmed that LPS increased pro-inflammatory markers, such as cytokines and NO production in murine muscle. Our data also confirmed that the proliferating myoblasts are responsive to LPS-induced inflammation in a manner similar to differentiated myotube cells as previously reported by others [27]. It is also known that LPS can change protein and lipid metabolism and these metabolic changes occur before inducing an immune response in skeletal muscle [26, 32]. Others suggest that the LPS-induced metabolic changes, including immune response, are dependent on skeletal muscle type and exposure time [3234]. Although in the present study we collected only one type of murine skeletal muscle (gastrocnemius, a fast-twitch glycolytic skeletal muscle) at one time point, we consistently showed that LPS treatment induces the upregulation of inflammatory markers in the skeletal muscle.

NF-κB is a transcription factor that regulates innate and adaptive immune responses mediated by inflammation. As a result, NF-κB activation contributes to the etiology of various inflammatory diseases. Our results showed that MIC-1 reduced the LPS-induced inflammation, at least partially, via NF-κB pathway in skeletal muscle. MIC-1 decreased the NF-κB’s nuclear translocation, as visualized by immunofluorescence staining. This observation may be validated further with cell fractionation experiments [35]. The observed MIC-1’s anti-inflammatory effects in skeletal muscle cells were similar to its effects in macrophages, which suggest that the oral administration of MIC-1 affects multiple inflammation-mediated processes [8]. Sulforaphane from Brassicaceae plants is the best studied natural isothiocyanate, however, in contrast to MIC-1 it is a chemically unstable liquid at room temperature (2). Similarly to MIC-1, sulforaphane also reduced inflammatory pathways in skeletal muscle [1720]. Sulforaphane inhibited the NF-κB pathway followed by the downregulation of the inflammatory cytokines (TNF-α, IL-1β, IL-6) in skeletal muscle of mdx mice, a genetic model for muscle dystrophy [17]. Moreover, sulforaphane regulated the NF-κB’s upstream regulators by increasing the inhibitor of κB (IκB) and decreasing IκB kinase (IKK) at protein levels in skeletal muscle of mdx mice [17]. IKK is responsible for the phosphorylation of IκB releasing NF-κB into the nucleus, which suggests that sulforaphane inhibited NF-κB’s nuclear translocation by regulating its cytoplasmic content [17]. MIC-1 exerts its anti-inflammatory properties by inhibiting NF-κB pathway, however whether MIC-1 inhibits NF-κB pathway post-translationally via IKK/IκB is not yet known. Further studies may be needed to evaluate the effects of MIC-1 on specific NF-κB isoforms and related molecules, as well as to confirm NF-κB involvement using inhibitors or loss of function mutations.

MIC-1 may also regulate inflammation via other pathways, such as interferons (Ifn) signaling pathway. MIC-1 downregulated the expression of a type I interferon (Ifn-α), which attracts immune response cells to the infected areas. Interferons can induce immune response independently of NF-κB pathway, for example via activation of the Janus kinase (JAK)/signal transducer and activator of transcription (STAT) signaling pathway [36, 37]. Although sulforaphane reduced the phosphorylation of JAK2 and STAT3 in liver and adipose tissues of high fat diet-fed rats [36], sulforaphane and MIC-1 inhibited interferon-stimulated genes without regulating the phosphorylation of STATs in Ba/F3 lymphocytes and HeLa epithelial cells [37]. MIC-1 and sulforaphane may inhibit JAK/STAT pathway by downstream inhibitory events or other posttranslational modifications [37]. In addition, previous reports suggest that sulforaphane reduces interferon response by downregulating the stimulator of interferon genes (STING) via the nuclear factor erythroid 2–related factor 2 (Nrf2) pathway [38, 39]. We previously reported that MIC-1 increases the nuclear translocation of Nrf2, followed by a reduction of oxidative stress and inflammation markers induced by LPS in macrophages [8]. Consistently, moringa leaf extract increased the activities of Nrf2 target enzymes (i.e., superoxide dismutase and catalase) in C2C12 myotubes [22, 24]. Therefore, MIC-1’s anti-inflammatory properties may also include JAK/STAT, and STING/Nrf2 pathways. Other pathways identified by our GO analyses (S1 and S2 Files) may also be involved in the MIC-1’s anti-inflammatory effects in skeletal muscle, however how these pathways interact due to MIC-1 needs further investigation. We did not evaluate the effects of MIC-1 alone (without LPS induction) in muscle cells. However, we previously reported that MIC-1 showed similar anti-inflammatory effects in murine and human macrophages with and without LPS induction [1, 8].

The effective and safe dosages of MIC-1, given orally, used in rodent models can be translated to 6.5–16 mg/kg bw (390–960 mg/60 kg bw) in human equivalent doses [4, 8, 40]. However, clinical trials are yet to show MIC-1’s effectiveness and safety in humans. The clinical trials that investigated the effects of sulforaphane on inflammatory markers are still not definitive [18, 41]. While sulforaphane (30 mg/d for 4 weeks) reduced exercise-induced IL-6 levels in young healthy men [18], sulforaphane (up to 26.6 mg/d for 4 weeks) did not change inflammatory markers, including IL-6 levels, in patients with chronic obstructive pulmonary disease [41]. Pre-clinical studies showed that MIC-1 and sulforaphane have similar biological properties, but the anti-inflammatory effects of isothiocyanates in humans still need to be elucidated. In summary, MIC-1 suppresses the inflammatory pathways and pro-inflammatory cytokines induced by LPS in skeletal muscle. Overall, our study suggests that MIC-1 is a potential intervention to reduce systemic inflammation, inflammation-associated metabolic, and muscular dysfunctions.

Supporting information

S1 Fig. Principal component analysis (PCA) plots of RNA-seq data.

DESeq2 analysis of the 3 replicates for C2C12 samples (A) and muscle tissue samples (B) was conducted to create PCA plots. The data were first turned into a DESeq2 object, then ’regularized log’ transformed and plotted in a PCA plot.

https://doi.org/10.1371/journal.pone.0279370.s001

(TIF)

S1 File. Gene Ontology (GO) analysis of C2C12 myoblasts.

Gene counts, gene list, and GO terms of comparisons between control (Ctrl) vs lipopolysaccharide (LPS)-treated samples, and LPS-treated samples vs LPS-moringa isothiocyanate 1 (MIC-1)-treated samples.

https://doi.org/10.1371/journal.pone.0279370.s002

(XLSX)

S2 File. Gene Ontology (GO) analysis of gastrocnemius muscle of C57BL/6 male mice.

Gene counts, gene list, and GO terms of comparisons between control (Ctrl) vs lipopolysaccharide (LPS)-treated samples, and LPS-treated samples vs LPS-moringa isothiocyanate 1 (MIC-1)-treated samples.

https://doi.org/10.1371/journal.pone.0279370.s003

(XLSX)

References

  1. 1. Jaja-Chimedza A, Graf BL, Simmler C, Kim Y, Kuhn P, Pauli GF, et al. Biochemical characterization and anti-inflammatory properties of an isothiocyanate-enriched moringa (Moringa oleifera) seed extract. PLoS One. 2017;12(8):e0182658. pmid:28792522
  2. 2. Waterman C, Cheng DM, Rojas-Silva P, Poulev A, Dreifus J, Lila MA, et al. Stable, water extractable isothiocyanates from Moringa oleifera leaves attenuate inflammation in vitro. Phytochemistry. 2014;103:114–22. pmid:24731259
  3. 3. Wolff K, Jaja-Chimedza A, Kim Y, Waterman C, Poulev A, Raskin I, et al. Moringa isothiocyanate-1 is bioaccessible and bioavailable as a stable unmodified compound. Phytochem Lett. 2020;38:33–8.
  4. 4. Kim Y, Jaja-Chimedza A, Merrill D, Mendes O, Raskin I. A 14-day repeated-dose oral toxicological evaluation of an isothiocyanate-enriched hydro-alcoholic extract from Moringa oleifera Lam. seeds in rats. Toxicol reports. 2018;5:418–26. pmid:29854612
  5. 5. Padla EP, Solis LT, Levida RM, Shen CC, Ragasa CY. Antimicrobial isothiocyanates from the seeds of Moringa oleifera Lam. Z Naturforsch C. 2012;67(11–12):557–64. pmid:23413749
  6. 6. Waterman C, Rojas-Silva P, Tumer TB, Kuhn P, Richard AJ, Wicks S, et al. Isothiocyanate-rich Moringa oleifera extract reduces weight gain, insulin resistance, and hepatic gluconeogenesis in mice. Mol Nutr Food Res. 2015;59(6):1013–24. pmid:25620073
  7. 7. Cheng D, Gao L, Su S, Sargsyan D, Wu R, Raskin I, et al. Moringa isothiocyanate activates Nrf2: potential role in diabetic nephropathy. AAPS J. 2019;21(2):31. pmid:30783799
  8. 8. Sailaja BS, Aita R, Maledatu S, Ribnicky D, Verzi MP, Raskin I. Moringa isothiocyanate-1 regulates Nrf2 and NF-κB pathway in response to LPS-driven sepsis and inflammation. PLoS One. 2021;16(4):e0248691. pmid:33793581
  9. 9. Lopez-Rodriguez NA, Gaytán-Martínez M, de la Luz Reyes-Vega M, Loarca-Piña G. Glucosinolates and isothiocyanates from Moringa oleifera: chemical and biological approaches. Plant Foods Hum Nutr. 2020 Dec;75(4):447–57. pmid:32909179
  10. 10. Giacoppo S, Rajan TS, De Nicola GR, Iori R, Rollin P, Bramanti P, et al. The isothiocyanate isolated from Moringa oleifera shows potent anti-inflammatory activity in the treatment of murine subacute Parkinson’s disease. Rejuvenation Res. 2017;20(1):50–63. pmid:27245199
  11. 11. Giacoppo S, Rajan TS, Iori R, Rollin P, Bramanti P, Mazzon E. The α-cyclodextrin complex of the Moringa isothiocyanate suppresses lipopolysaccharide-induced inflammation in RAW 264.7 macrophage cells through Akt and p38 inhibition. Inflamm Res. 2017;66(6):487–503. pmid:28289752
  12. 12. Rajan TS, Giacoppo S, Iori R, De Nicola GR, Grassi G, Pollastro F, et al. Anti-inflammatory and antioxidant effects of a combination of cannabidiol and moringin in LPS-stimulated macrophages. Fitoterapia. 2016;112:104–15. pmid:27215129
  13. 13. Wang C, Wu R, Sargsyan D, Zheng M, Li S, Yin R, et al. CpG methyl-seq and RNA-seq epigenomic and transcriptomic studies on the preventive effects of Moringa isothiocyanate in mouse epidermal JB6 cells induced by the tumor promoter TPA. J Nutr Biochem. 2019;68:69–78. pmid:31030169
  14. 14. Li S, Li W, Wu R, Yin R, Sargsyan D, Raskin I, et al. Epigenome and transcriptome study of moringa isothiocyanate in mouse kidney mesangial cells induced by high glucose, a potential model for diabetic-induced nephropathy. AAPS J. 2019;22(1):8. pmid:31807911
  15. 15. Giacoppo S, Iori R, Bramanti P, Mazzon E. Topical moringin-cream relieves neuropathic pain by suppression of inflammatory pathway and voltage-gated ion channels in murine model of multiple sclerosis. Mol Pain. 2017;13:1744806917724318. pmid:28741431
  16. 16. Jaja-Chimedza A, Zhang L, Wolff K, Graf BL, Kuhn P, Moskal K, et al. A dietary isothiocyanate-enriched moringa (Moringa oleifera) seed extract improves glucose tolerance in a high-fat-diet mouse model and modulates the gut microbiome. J Funct Foods. 2018;47:376–85. pmid:30930963
  17. 17. Sun CC, Li SJ, Yang CL, Xue RL, Xi YY, Wang L, et al. Sulforaphane attenuates muscle inflammation in dystrophin-deficient mdx mice via NF-E2-related Factor 2 (Nrf2)-mediated inhibition of NF-κB signaling pathway. J Biol Chem. 2015;290(29):17784–95. pmid:26013831
  18. 18. Sato K, Kihara H, Kumazawa Y, Tatara K. Oral chronic sulforaphane effects on heavy resistance exercise: implications for inflammatory and muscle damage parameters in young practitioners. Nutrition. 2021;90:111266. pmid:34004418
  19. 19. Faridvand Y, Haddadi P, Nejabati HR, Ghaffari S, Zamani-Gharehchamani E, Nozari S, et al. Sulforaphane modulates CX3CL1/CX3CR1 axis and inflammation in palmitic acid-induced cell injury in C2C12 skeletal muscle cells. Mol Biol Rep. 2020;47(10):7971–7. pmid:33034881
  20. 20. Wang M, Pu D, Zhao Y, Chen J, Zhu S, Lu A, et al. Sulforaphane protects against skeletal muscle dysfunction in spontaneous type 2 diabetic db/db mice. Life Sci. 2020;255:117823. pmid:32445760
  21. 21. Duranti G, Maldini M, Crognale D, Sabatini S, Corana F, Horner K, et al. Moringa oleifera leaf extract influences oxidative metabolism in C2C12 myotubes through SIRT1-PPARα pathway. Phytomedicine Plus. 2021;1(1):100014.
  22. 22. Duranti G, Maldini M, Crognale D, Horner K, Dimauro I, Sabatini S, et al. Moringa oleifera leaf extract upregulates Nrf2/HO-1 expression and ameliorates redox status in C2C12 skeletal muscle cells. Molecules. 2021 Aug;26(16):5041. pmid:34443628
  23. 23. Yun YR, Oh SJ, Lee MJ, Choi YJ, Park SJ, Lee MA, et al. Antioxidant activity and calcium bioaccessibility of Moringa oleifera leaf hydrolysate, as a potential calcium supplement in food. Food Sci Biotechnol. 2020 Nov;29(11):1563–71. pmid:33088605
  24. 24. Ceci R, Maldini M, Olson ME, Crognale D, Horner K, Dimauro I, et al. Moringa oleifera leaf extract protects C2C12 myotubes against H(2)O(2)-induced oxidative stress. Antioxidants. 2022;11(8):1435. pmid:35892637
  25. 25. Cao YY, Wang Z, Yu T, Zhang Y, Wang ZH, Lu ZM, et al. Sepsis induces muscle atrophy by inhibiting proliferation and promoting apoptosis via PLK1-AKT signalling. J Cell Mol Med. 2021;25(20):9724–39. pmid:34514712
  26. 26. Moreno-Rupérez Á, Priego T, González-Nicolás MÁ, López-Calderón A, Lázaro A, Martín AI. Role of glucocorticoid signaling and HDAC4 activation in diaphragm and gastrocnemius proteolytic activity in septic rats. Int J Mol Sci. 2022;23(7):3641. pmid:35408999
  27. 27. Frost RA, Nystrom GJ, Lang CH. Lipopolysaccharide regulates proinflammatory cytokine expression in mouse myoblasts and skeletal muscle. Am J Physiol Regul Integr Comp Physiol. 2002 Sep;283(3):R698–709. pmid:12185005
  28. 28. Rocha-Ramírez LM, Hernández-Chiñas U, Baños-Rojas D, Xicohtencatl-Cortés J, Chávez-Berrocal ME, Rico-Rosillo G, et al. Pet serine protease from enteroaggregative Escherichia coli stimulates the inflammatory response activating human macrophages. BMC Microbiol. 2016;16(1):158. pmid:27439312
  29. 29. Bray NL, Pimentel H, Melsted P, Pachter L. Near-optimal probabilistic RNA-seq quantification. Nat Biotechnol. 2016;34(5):525–7. pmid:27043002
  30. 30. Soneson C, Love MI, Robinson MD. Differential analyses for RNA-seq: transcript-level estimates improve gene-level inferences. F1000Research. 2015;4:1521. pmid:26925227
  31. 31. Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014;15(12):550. pmid:25516281
  32. 32. Magnoni LJ, Roher N, Crespo D, Krasnov A, Planas J V. In vivo molecular responses of fast and slow muscle fibers to lipopolysaccharide in a Teleost fish, the rainbow trout (Oncorhynchus mykiss). Biology (Basel). 2015;4(1):67–87. pmid:25658438
  33. 33. Lang CH, Silvis C, Deshpande N, Nystrom G, Frost RA. Endotoxin stimulates in vivo expression of inflammatory cytokines tumor necrosis factor alpha, interleukin-1 beta, -6, and high-mobility-group protein-1 in skeletal muscle. Shock. 2003;19(6):538–46. pmid:12785009
  34. 34. Mattingly AJ, Laitano O, Garcia CK, Robinson GP, Clanton TL. Lipopolysaccharide-induced cytokine secretion from in vitro mouse slow and fast limb muscle. Shock. 2022;57(4):600–7. pmid:34798635
  35. 35. Vecchiotti D, Verzella D, Capece D, Nolfi MDV, Di Francesco B, Cornice J, et al. Biochemical methods to analyze the subcellular localization of NF-κB proteins using cell fractionation. Methods Mol Biol. 2021;2366:19–25.
  36. 36. Ashmawy AI, El-Abhar HS, Abdallah DM, Ali MA. Chloroquine modulates the sulforaphane anti-obesity mechanisms in a high-fat diet model: role of JAK-2/ STAT-3/ SOCS-3 pathway. Eur J Pharmacol. 2022;927:175066. pmid:35643302
  37. 37. Michl C, Vivarelli F, Weigl J, De Nicola GR, Canistro D, Paolini M, et al. The chemopreventive phytochemical moringin isolated from Moringa oleifera seeds inhibits JAK/STAT signaling. PLoS One. 2016;11(6):e0157430. pmid:27304884
  38. 38. Genova E, Apollonio M, Decorti G, Tesser A, Tommasini A, Stocco G. In vitro effects of sulforaphane on interferon-driven inflammation and exploratory evaluation in two healthy volunteers. Molecules. 2021;26(12):3602. pmid:34204601
  39. 39. Olagnier D, Brandtoft AM, Gunderstofte C, Villadsen NL, Krapp C, Thielke AL, et al. Nrf2 negatively regulates STING indicating a link between antiviral sensing and metabolic reprogramming. Nat Commun. 2018;9(1):3506. pmid:30158636
  40. 40. Nair AB, Jacob S. A simple practice guide for dose conversion between animals and human. J basic Clin Pharm. 2016;7(2):27–31. pmid:27057123
  41. 41. Wise RA, Holbrook JT, Criner G, Sethi S, Rayapudi S, Sudini KR, et al. Lack of effect of oral sulforaphane administration on Nrf2 expression in COPD: a randomized, double-Blind, placebo controlled trial. PLoS One. 2016;11(11):e0163716. pmid:27832073