Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
BMC Res Notes ; 12(1): 717, 2019 Oct 31.
Article in English | MEDLINE | ID: mdl-31672165

ABSTRACT

OBJECTIVE: CCAAT/Enhancer Binding proteins (C/EBPs) are transcription factors involved in the regulation of a variety of cellular processes. We used the Abcam Recombinant Anti-C/EBP beta antibody (E299) to detect C/EBPß expression during myogenesis. Though the antibody is monoclonal, and the immunogen used is highly specific to C/EBPß, we identified an intense band at 23 kDa on western blot that did not correspond to any of the known isoforms of C/EBPß, or family members predicted to cross-react. Absent in myoblast cells overexpressing C/EBPß, the band was present when C/EBPß was knocked down, confirming specificity for a protein other than C/EBPß. The objective of this work was to identify the contaminating reactivity. RESULTS: We performed immunoprecipitation followed by mass spectrometry to identified myosin light chain 4 (MYL4) as the unknown band, suggesting that the Abcam monoclonal antibody directed against C/EBPß is not pure, but contains a contaminating antibody against MYL4. Caution should be used when working in cells lines that express MYL4 to not confound the detection of MYL4 with that of C/EBPß isoforms.


Subject(s)
Antibodies, Monoclonal/immunology , CCAAT-Enhancer-Binding Protein-beta/immunology , Cell Differentiation/immunology , Myoblasts/immunology , Animals , Antibody Specificity/immunology , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cell Differentiation/genetics , Cell Line , Cross Reactions/immunology , Gene Expression Profiling , HEK293 Cells , Humans , Mice , Muscle Development/genetics , Muscle Development/immunology , Myoblasts/cytology , Myoblasts/metabolism , Myosin Light Chains/genetics , Myosin Light Chains/immunology , Myosin Light Chains/metabolism , Protein Isoforms/genetics , Protein Isoforms/immunology , Protein Isoforms/metabolism , Tandem Mass Spectrometry/methods
2.
Front Immunol ; 10: 1726, 2019.
Article in English | MEDLINE | ID: mdl-31396230

ABSTRACT

Background: IL-17A has effects on several cell types and is a therapeutic target in several inflammatory diseases. IL-17F shares 50% homology and biological activities with IL-17A. It is now of interest to target both cytokines. The objective was to compare the IL-17A and IL-17F effect on cytokine production by RA synoviocytes, and to extend to other cells. Methods: Cells (RA synoviocytes, psoriasis skin fibroblasts, endothelial cells, myoblasts, and hepatocytes) were cultured in the presence or not of: IL-17A, IL-17F, TNF, IL-1ß alone or their combinations, IL-17A/TNF, IL-17A/IL-1ß, IL-17A/TNF/IL-1ß, IL-17F/TNF, IL-17F/IL-1ß, and IL-17F/TNF/IL-1ß. All experiments were performed in parallel to reduce variability. After 48 h, supernatants were recovered and IL-6 and IL-8 levels were measured by ELISA. Results: IL-17A and IL-17F alone increased significantly IL-6 and IL-8 productions by synoviocytes, with a stronger effect for IL-17A. For IL-6 production, TNF or IL-1ß alone had the largest effect on myoblasts (5-fold increase), while for IL-8 production, it was on skin fibroblasts (5-fold increase). The IL-17A/TNF synergistic increase was observed on all cells for IL-6; and for IL-8, except for endothelial cells. For IL-17F/TNF, except with endothelial cells, a synergistic effect was also observed, but less powerful than with IL-17A/TNF. IL-17A/IL-1ß or IL-17F/IL-1ß effect was cell-type dependent, with an additive effect for synoviocytes (1.6 and 2-fold increase, respectively for IL-6, and 1.8 and 2-fold increase, respectively for IL-8) and a synergistic effect for hepatocytes (3.8 and 4.2-fold increase, respectively for IL-6, and 6 and 2-fold increase, respectively for IL-8). The three-cytokine combination induced an additive effect for synoviocytes and a synergistic effect for skin fibroblasts. Conclusion: IL-17A and IL-17F acted similarly by inducing pro-inflammatory cytokine secretion, with a stronger response intensity with IL-17A. Their activities were potentiated by the combination with TNF and IL-1ß, with an effect dependent on the cell type.


Subject(s)
Fibroblasts/immunology , Hepatocytes/immunology , Human Umbilical Vein Endothelial Cells/immunology , Interleukin-17/immunology , Myoblasts/immunology , Synoviocytes/immunology , Cells, Cultured , Fibroblasts/drug effects , Hepatocytes/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Interleukin-17/pharmacology , Interleukin-18/immunology , Myoblasts/drug effects , Synoviocytes/drug effects , Tumor Necrosis Factor-alpha/immunology
3.
Arthritis Rheumatol ; 71(8): 1371-1376, 2019 08.
Article in English | MEDLINE | ID: mdl-30861336

ABSTRACT

OBJECTIVE: Although more than a dozen myositis-specific autoantibodies (MSAs) have been identified, most patients with myositis are positive for a single MSA. The specific overexpression of a given myositis autoantigen in myositis muscle has been proposed as initiating and/or propagating autoimmunity against that particular autoantigen. The present study was undertaken to test this hypothesis. METHODS: In order to quantify autoantigen RNA expression, RNA sequencing was performed on muscle biopsy samples from control subjects, MSA-positive patients with myositis, regenerating mouse muscles, and cultured human muscle cells. RESULTS: Muscle biopsy samples were available from 20 control subjects and 106 patients with autoantibodies recognizing hydroxymethylglutaryl-coenzyme A reductase (n = 40), signal recognition particles (n = 9), Jo-1 (n = 18), nuclear matrix protein 2 (n = 12), Mi-2 (n = 11), transcription intermediary factor 1γ (n = 11), or melanoma differentiation-associated protein 5 (n = 5). The increased expression of a given autoantigen in myositis muscle was not associated with autoantibodies recognizing that autoantigen (all q > 0.05). In biopsy specimens from both myositis muscle and regenerating mouse muscles, autoantigen expression correlated directly with the expression of muscle regeneration markers and correlated inversely with the expression of genes encoding mature muscle proteins. Myositis autoantigens were also expressed at high levels in cultured human muscle cells. CONCLUSION: Most myositis autoantigens are highly expressed during muscle regeneration, which may relate to the propagation of autoimmunity. However, factors other than overexpression of specific autoantigens are likely to govern the development of unique autoantibodies in individual patients with myositis.


Subject(s)
Autoantibodies/immunology , Autoantigens/metabolism , Muscle, Skeletal/immunology , Myositis/immunology , Regeneration/immunology , Animals , Autoantigens/immunology , Biopsy , Cells, Cultured , Humans , Mice , Myoblasts/immunology , Myoblasts/metabolism , Myositis/physiopathology , RNA/immunology , RNA/metabolism
4.
J Biol Chem ; 293(40): 15594-15605, 2018 10 05.
Article in English | MEDLINE | ID: mdl-30139748

ABSTRACT

Recent studies have reported that the immune system significantly mediates skeletal muscle repair and regeneration. Additionally, biological scaffolds have been shown to play a role in polarizing the immune microenvironment toward pro-myogenic outcomes. Moreover, myostatin inhibitors are known to promote muscle regeneration and ameliorate fibrosis in animal models of Duchenne muscular dystrophy (DMD), a human disease characterized by chronic muscle degeneration. Biological scaffolds and myostatin inhibition can potentially influence immune-mediated regeneration in the dystrophic environment, but have not been evaluated together. Toward this end, here we created an injectable biological scaffold composed of hyaluronic acid and processed skeletal muscle extracellular matrix. This material formed a cytocompatible hydrogel at physiological temperatures in vitro When injected subfascially above the tibialis anterior muscles of both WT and dystrophic mdx-5Cv mice, a murine model of DMD, the hydrogel spreads across the entire muscle before completely degrading at 3 weeks in vivo We found that the hydrogel is associated with CD206+ pro-regenerative macrophage polarization and elevated anti-inflammatory cytokine expression in both WT and dystrophic mice. Co-injection of both hydrogel and myostatin inhibitor significantly increased FoxP3+ regulatory T cell modulation and Foxp3 gene expression in the scaffold immune microenvironment. Finally, delivery of myostatin inhibitor with the hydrogel increased its bioactivity in vivo, and transplantation of immortalized human myoblasts with the hydrogel promoted their survival in vivo This study identifies a key role for biological scaffolds and myostatin inhibitors in modulating a pro-regenerative immune microenvironment in dystrophic muscle.


Subject(s)
Antibodies, Monoclonal/pharmacology , Drug Delivery Systems/methods , Immunity, Innate/drug effects , Muscular Dystrophy, Animal/drug therapy , Myostatin/antagonists & inhibitors , Regeneration/drug effects , Absorbable Implants , Animals , Extracellular Matrix/chemistry , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Gene Expression Regulation , Humans , Hyaluronic Acid/chemistry , Hydrogels/chemistry , Immunity, Innate/genetics , Lectins, C-Type/genetics , Lectins, C-Type/immunology , Macrophages/cytology , Macrophages/drug effects , Macrophages/immunology , Mannose Receptor , Mannose-Binding Lectins/genetics , Mannose-Binding Lectins/immunology , Mice , Mice, Inbred mdx , Muscle Development/drug effects , Muscle Development/genetics , Muscle Development/immunology , Muscle, Skeletal/drug effects , Muscle, Skeletal/immunology , Muscle, Skeletal/pathology , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/immunology , Muscular Dystrophy, Animal/pathology , Myoblasts/cytology , Myoblasts/drug effects , Myoblasts/immunology , Myostatin/genetics , Myostatin/immunology , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology , Regeneration/genetics , Regeneration/immunology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Tissue Scaffolds
5.
J Gen Virol ; 98(12): 3008-3025, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29120300

ABSTRACT

Enterovirus 71 (EV71) has caused major outbreaks of hand, foot and mouth disease. EV71 infections increase the production of many host cytokines and pro-inflammatory factors, including interleukin (IL)-6, IL-10 and COX-2. Some of these molecules could stimulate the signal transducer and activator of transcription 3 (STAT3), which plays a key role in regulating host immune responses and several viral diseases. However, the role of STAT3 in EV71 infection remains unknown. This study found that the phosphorylation levels of STAT3 (pY705-STAT3) are closely related to EV71 infection. Further experiments revealed that STAT3 exerts an anti-EV71 activity. However, the antiviral activity of STAT3 is partially antagonized by EV71-induced miR-124, which directly targets STAT3 mRNA. Similarly, IL-6R, the α-subunit of the IL-6 receptor complex, exhibits anti-EV71 activity and is directly targeted by the virus-induced miR-124. These results indicate that EV71 can evade host IL-6R- and STAT3-mediated antiviral activities by EV71-induced miR-124. This suggests that controlling miR-124 and the downstream targets, IL-6R and STAT3, might benefit the antiviral treatment of EV71 infection.


Subject(s)
Enterovirus A, Human/genetics , Immune Evasion , MicroRNAs/genetics , RNA, Messenger/genetics , Receptors, Interleukin-6/genetics , STAT3 Transcription Factor/genetics , Base Sequence , Binding Sites , Cell Line, Tumor , Cyclooxygenase 2/genetics , Cyclooxygenase 2/immunology , Enterovirus A, Human/growth & development , Enterovirus A, Human/immunology , Gene Expression Regulation , HEK293 Cells , HeLa Cells , Humans , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-6/genetics , Interleukin-6/immunology , MicroRNAs/immunology , Myoblasts/immunology , Myoblasts/virology , Neurons/immunology , Neurons/virology , Phosphorylation , RNA, Messenger/immunology , Receptors, Interleukin-6/immunology , STAT3 Transcription Factor/immunology , Signal Transduction , Virus Replication
6.
Sci Rep ; 7(1): 7393, 2017 08 07.
Article in English | MEDLINE | ID: mdl-28785068

ABSTRACT

Inflammatory myopathy is a rare autoimmune muscle disorder. Treatment typically focuses on skeletal muscle weakness or inflammation within muscle, as well as complications of respiratory failure secondary to respiratory muscle weakness. Impaired respiratory muscle function contributes to increased dyspnea and reduced exercise capacity in pulmonary hypertension (PH), a debilitating condition that has few treatment options. The initiation and progression of PH is associated with inflammation and inflammatory cell recruitment and it is established that hypoxia-induced mitogenic factor (HIMF, also known as resistin-like molecule α), activates macrophages in PH. However, the relationship between HIMF and inflammatory myoblasts remains unclear. This study investigated the signaling pathway involved in interleukin-18 (IL-18) expression and its relationship with HIMF in cultured myoblasts. We found that HIMF increased IL-18 production in myoblasts and that secreted IL-18 promoted tube formation of the endothelial progenitor cells. We used the mouse xenograft model and the chick chorioallantoic membrane assay to further explore the role of HIMF in inflammatory myoblasts and angiogenesis in vivo. Thus, our study focused on the mechanism by which HIMF mediates IL-18 expression in myoblasts through angiogenesis in vitro and in vivo. Our findings provide an insight into HIMF functioning in inflammatory myoblasts.


Subject(s)
Intercellular Signaling Peptides and Proteins/metabolism , Interleukin-18/metabolism , Myoblasts/immunology , Neovascularization, Pathologic/metabolism , Up-Regulation , 3-Phosphoinositide-Dependent Protein Kinases/metabolism , Animals , Cell Line , Disease Models, Animal , Endothelial Progenitor Cells/cytology , Endothelial Progenitor Cells/immunology , Endothelial Progenitor Cells/metabolism , Humans , Mice , Myoblasts/cytology , Myoblasts/metabolism , Neovascularization, Pathologic/immunology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Transcription Factor AP-1/metabolism
7.
PLoS One ; 12(7): e0182040, 2017.
Article in English | MEDLINE | ID: mdl-28742154

ABSTRACT

BACKGROUND: Circulating lipopolysaccharide (LPS) concentrations are often elevated in patients with sepsis or with various endogenous diseases that are associated with metabolic endotoxemia. Involuntary loss of skeletal muscle, termed muscle wasting, is commonly observed in these conditions, suggesting that circulating LPS might play an essential role in its development. Although impairment of muscle regeneration is an important determinant of skeletal muscle wasting, it is unclear whether LPS affects this process and, if so, by what mechanism. Here, we used the C2C12 myoblast cell line to investigate the effects of LPS on myogenesis. METHODS: C2C12 myoblasts were grown to 80% confluence and induced to differentiate in the absence or presence of LPS (0.1 or 1 µg/mL); TAK-242 (1 µM), a specific inhibitor of Toll-like receptor 4 (TLR4) signaling; and a tumor necrosis factor (TNF)-α neutralizing antibody (5 µg/mL). Expression of a skeletal muscle differentiation marker (myosin heavy chain II), two essential myogenic regulatory factors (myogenin and MyoD), and a muscle negative regulatory factor (myostatin) was analyzed by western blotting. Nuclear factor-κB (NF-κB) DNA-binding activity was measured using an enzyme-linked immunosorbent assay. RESULTS: LPS dose-dependently and significantly decreased the formation of multinucleated myotubes and the expression of myosin heavy chain II, myogenin, and MyoD, and increased NF-κB DNA-binding activity and myostatin expression. The inhibitory effect of LPS on myogenic differentiation was reversible, suggesting that it was not caused by nonspecific toxicity. Both TAK-242 and anti-TNF-α reduced the LPS-induced increase in NF-κB DNA-binding activity, downregulation of myogenic regulatory factors, and upregulation of myostatin, thereby partially rescuing the impairment of myogenesis. CONCLUSIONS: Our data suggest that LPS inhibits myogenic differentiation via a TLR4-NF-κB-dependent pathway and an autocrine/paracrine TNF-α-induced pathway. These pathways may be involved in the development of muscle wasting caused by sepsis or metabolic endotoxemia.


Subject(s)
Lipopolysaccharides/immunology , Muscle Development , Myoblasts/cytology , NF-kappa B/immunology , Signal Transduction , Toll-Like Receptor 4/immunology , Animals , Cell Differentiation , Mice , Myoblasts/immunology , Myoblasts/metabolism , RNA, Messenger/genetics , Toll-Like Receptor 2/immunology , Toll-Like Receptor 4/genetics , Tumor Necrosis Factor-alpha/immunology
8.
Mol Immunol ; 82: 1-9, 2017 02.
Article in English | MEDLINE | ID: mdl-27992764

ABSTRACT

Chronic inflammatory diseases such as insulin resistance, Type 2 diabetes, neurodegenerative diseases etc., are shown to be caused due to imbalanced activation states of macrophages. MicroRNAs which are transcriptional/post-transcriptional regulators of gene expression drive several pathophysiological processes including macrophage polarization. However the functional role of microRNAs in regulating inflammation induced insulin resistance is ill defined. In our current study we observed that the expression of miR-712 was reduced in macrophages exposed to LPS and IFN-γ. Ectopic expression of miR-712 in RAW 264.7 mouse macrophages impaired the expression of iNOS protein and secretion of pro-inflammatory cytokines such as TNF-α, IL-6 and IFN-ß which in turn led to improved insulin stimulated glucose uptake in co-cultured L6 myoblasts. Mechanistically, we identified that miR-712 targets the 3'UTR of a potent inflammatory gene LRRK2 and dampens the phosphorylation of p38 and ERK1/2 kinases. Taken together, our data underscore the regulatory role of miR-712 in restoring insulin stimulated glucose uptake by myoblasts through down-regulating macrophage mediated inflammatory responses.


Subject(s)
Insulin Resistance/genetics , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/immunology , Macrophage Activation/genetics , MicroRNAs/immunology , Myoblasts/metabolism , Animals , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation/immunology , Glucose/metabolism , Immunoblotting , Inflammation/immunology , Macrophage Activation/immunology , Macrophages/immunology , Mice , Myoblasts/immunology , RAW 264.7 Cells , Real-Time Polymerase Chain Reaction
9.
J Autoimmun ; 75: 118-129, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27522114

ABSTRACT

Idiopathic inflammatory myopathies (IIMs) are diseases with muscle weakness, morphologically characterized by inflammatory infiltration and increased expression of MHC class I molecule on myofibers. Immunoproteasome, as a proteolytic complex that shapes the repertoire of antigenic peptides, has been previously demonstrated to be over-expressed in IIMs at mRNA level. In this study, we investigated the expression and the function of the immunoproteasome in IIMs in more detail. As shown by immunofluorescence staining, expression of relevant players of the immunoproteasome was detectable in the inflamed skeletal muscle tissue from IIM patients. In fact, two subunits of the immunoproteasome, ß1i or ß5i were upregulated in sporadic inclusion body myositis, immune-mediated necrotizing myopathies and dermatomyositis muscle biopsies and co-localized with the MHC class I expressing myofibers. Double immunofluorescence revealed that both myofibers and muscle infiltrating cells, including CD8+ T-cells and CD68 + macrophages in IIMs expressed ß1i or ß5i. In addition, we have also investigated the role of the immunoproteasome in myoblasts during in vitro inflammatory conditions. Using human primary myoblasts cultures we found that pro-inflammatory cytokines, TNF-α or IFN-γ upregulate ß1i or ß5i. Selective inhibition or depletion of ß5i amplified the TNF-α or IFN-γ mediated expression of cytokines/chemokines (myokines) in myoblasts. Furthermore, we demonstrated that specific inhibitors of ß1i or ß5i reduced the cell surface expression of MHC class I in myoblasts induced by IFN-γ. Taken together, our data suggest that the immunoproteasome is involved in pathologic MHC class I expression and maintenance of myokine production in IIMs. Thus, induction of the immunoproteasome was identified as a pathomechanism underlying inflammation in IIMs.


Subject(s)
Cytokines/immunology , Histocompatibility Antigens Class I/immunology , Muscle, Skeletal/immunology , Myositis/immunology , Proteasome Endopeptidase Complex/immunology , Adult , Aged , Aged, 80 and over , Blotting, Western , Cells, Cultured , Child, Preschool , Cytokines/genetics , Cytokines/metabolism , Dermatomyositis/genetics , Dermatomyositis/immunology , Dermatomyositis/metabolism , Female , Gene Expression/drug effects , Gene Expression/immunology , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism , Humans , Interferon-gamma/pharmacology , Male , Microscopy, Fluorescence , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Myoblasts/drug effects , Myoblasts/immunology , Myoblasts/metabolism , Myositis/genetics , Myositis/metabolism , Pancreatitis, Acute Necrotizing/genetics , Pancreatitis, Acute Necrotizing/immunology , Pancreatitis, Acute Necrotizing/metabolism , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Protein Subunits/genetics , Protein Subunits/immunology , Protein Subunits/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/pharmacology , Young Adult
10.
Int Immunopharmacol ; 34: 107-113, 2016 May.
Article in English | MEDLINE | ID: mdl-26943728

ABSTRACT

Emodin, a major component of Rheum palmatum, has been reported to significantly protect neural tissue against apoptosis and autophagy. However, the effects and underlying mechanisms of action of emodin in muscle atrophy are still poorly defined. In this study, we investigated the protective effects and the underlying mechanisms by which emodin acts on tumor necrosis factor alpha (TNF-α)-induced apoptosis and autophagy in mouse C2C12 myoblasts. Emodin, at various concentrations, decreased TNF-α-induced apoptosis in C2C12 myoblasts, which were analyzed by Hoechst 33342 staining and annexin V/PI analysis. Emodin also inhibited the collapse of the mitochondrial membrane potential and the generation of reactive oxygen species in TNF-α-stimulated C2C12 myoblasts. Consistent with these results, the expression of Bcl-2 was increased, whereas the expression of Bax, cleaved-caspase 3 and cleaved-PARP was decreased after emodin treatment. These data demonstrate that emodin attenuated apoptosis in TNF-α-stimulated C2C12 myoblasts through mitochondrial signaling pathways. In addition, emodin inhibited autophagy in TNF-α-stimulated C2C12 myoblasts by suppressing the expression of LC3-II, Beclin-1 and Atg7. Emodin also resulted in the upregulation of the phosphorylated forms of Akt. Taken together, these results suggest that emodin inhibited apoptosis and autophagy in TNF-α-induced C2C12 myoblasts, possibly through the activation of phosphorylated Akt. Our findings suggest that emodin could be a potential therapeutic agent in the treatment of muscle atrophy.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Emodin/pharmacology , Muscular Atrophy/drug therapy , Myoblasts/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Animals , Apoptosis/drug effects , Autophagy/drug effects , Cell Line , Membrane Potential, Mitochondrial/drug effects , Mice , Myoblasts/immunology , Phosphorylation , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Rheum/immunology , Tumor Necrosis Factor-alpha/immunology
11.
Biochimie ; 119: 16-26, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26453808

ABSTRACT

Uncontrolled inflammation leads to several diseases such as insulin resistance, T2D and several types of cancers. The functional role of microRNAs in inflammation induced insulin resistance is poorly studied. MicroRNAs are post-transcriptional regulatory molecules which mediate diverse biological processes. We here show that miR-16 expression levels are down-regulated in different inflammatory conditions such as LPS/IFNγ or palmitate treated macrophages, palmitate exposed myoblasts and insulin responsive tissues of high sucrose diet induced insulin resistant rats. Importantly, forced expression of miR-16 in macrophages impaired the production of TNF-α, IL-6 and IFN-ß leading to enhanced insulin stimulated glucose uptake in co-cultured skeletal myoblasts. Further, ectopic expression of miR-16 enhanced insulin stimulated glucose uptake in skeletal myoblasts via the up-regulation of GLUT4 and MEF2A, two key players involved in insulin stimulated glucose uptake. Collectively, our data highlight the important role of miR-16 in ameliorating inflammation induced insulin resistance.


Subject(s)
Down-Regulation , Insulin Resistance , Macrophage Activation , Macrophages/metabolism , MicroRNAs/metabolism , Myoblasts/metabolism , Animals , Cell Communication/drug effects , Cell Line , Cell Polarity/drug effects , Coculture Techniques , Dietary Sucrose/adverse effects , Down-Regulation/drug effects , Endotoxins/toxicity , Fatty Acids, Nonesterified/adverse effects , Interferon-gamma/genetics , Interferon-gamma/metabolism , Interferon-gamma/pharmacology , Macrophage Activation/drug effects , Macrophages/drug effects , Macrophages/immunology , Mice , MicroRNAs/antagonists & inhibitors , Muscle, Skeletal/metabolism , Myoblasts/drug effects , Myoblasts/immunology , RAW 264.7 Cells , Rats , Rats, Wistar , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology
12.
J Med Virol ; 87(5): 733-9, 2015 May.
Article in English | MEDLINE | ID: mdl-25656089

ABSTRACT

In addition to fever, rash, and arthralgia/arthritis, myalgia is another dominant symptom in Chikungunya virus (CHIKV) infection. How CHIKV induces myalgia is unclear. To better understand the viral factors involved in CHIKV-induced myalgia, CHIKVs were isolated from patients with and without myalgia designated myalgia-CHIKV and mild-CHIKV, respectively. The response of myoblasts to infection by the two groups of clinical isolates of CHIKV was investigated. Both groups of CHIKV replicated well in primary human myoblasts. However, the myalgia-CHIKVs replicated to a higher titer and caused the death of infected myoblast more rapidly than the mild-CHIKVs. CHIKV-infected myoblasts increased production of four out of five inflammatory cytokines examined (MCP-1, IP-10, MIP-1α, and IL-8) in comparison to mock-infected cells. Comparison between the myoblast inflammatory cytokine responses showed that myalgia-CHIKVs were stronger activators of cytokines than mild-CHIKVs. This means that recent epidemic strains of CHIKV exhibited different degrees of myoblast permissiveness as evidenced by differences in the ability to replicate and to stimulate inflammatory responses in myoblasts. This data suggest that the myopathic syndrome in recent epidemics is dependent upon the strain of CHIKV.


Subject(s)
Chikungunya Fever/virology , Chikungunya virus/physiology , Myalgia/virology , Myoblasts/immunology , Myoblasts/virology , Virus Replication , Adult , Cells, Cultured , Chikungunya Fever/epidemiology , Chikungunya Fever/pathology , Chikungunya virus/growth & development , Chikungunya virus/isolation & purification , Cytokines/metabolism , Humans , Viral Load
13.
Biochem Biophys Res Commun ; 458(4): 790-5, 2015 Mar 20.
Article in English | MEDLINE | ID: mdl-25686491

ABSTRACT

Cachexia or muscle wasting is a common condition that occurs in many chronic diseases. The wasting conditions are characterized by increased levels of TNF-α which was also known as cachectin in the past. But how TNF-α exerts its cachetic effects remains controversial. To clarify this issue, we investigated the impact of TNF-α on C2C12 cell myogenic differentiation. Our results demonstrate that myotube formation was completely inhibited by TNF-α when added to differentiating C2C12 myoblasts. The inhibitory effect of TNF-α on differentiation was accompanied by activation of NF-κB and down regulation of myogenin and Akt. Importantly, TNF-α's effect on differentiation was abolished when IGF-1 was added to the culture. IGF-1 treatment also inhibited NF-κB reporter activity and restored Akt levels. Our data suggest that TNF-α inhibits myogenic differentiation through NF-κB activation and impairment of IGF-1 signaling pathway. The reversal of TNF-α induced inhibition of myogenesis by IGF-1 may have significant therapeutic potential.


Subject(s)
Insulin-Like Growth Factor I/immunology , Muscle Fibers, Skeletal/cytology , Myoblasts/cytology , NF-kappa B/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , Cell Differentiation , Cell Line , Mice , Muscle Development , Muscle Fibers, Skeletal/immunology , Myoblasts/immunology , Signal Transduction
14.
Autoimmun Rev ; 13(11): 1160-6, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25172242

ABSTRACT

To our knowledge, no study has evaluated the involvement of T helper (Th)1- and Th2-chemokines in extra-ocular muscle (EOM) myopathy in "patients with thyroid-associated ophthalmopathy" (TAO-p). We tested the effects of interferon (IFN)γ and tumor necrosis factor (TNF)α stimulation, and of increasing concentrations of peroxisome proliferator-activated receptor (PPAR)γ agonists (pioglitazone or rosiglitazone; 0.1 µM-20 µM), on Th1-chemokine [C-X-C motif ligand (CXCL)10] and Th2-chemokine [C-C motif ligand (CCL)2] secretion in primary EOM cultures from TAO-p vs. control myoblasts. Moreover, we evaluated serum CXCL10 and CCL2 in active TAO-p with prevalent EOM involvement (EOM-p) vs. those with prevalent orbital fat expansion (OF-p). Serum CXCL10 was higher in OF-p and EOM-p vs. controls, while serum CCL2 was not significantly different in controls, or in OF-p and EOM-p. We showed the expression of PPARγ in EOM cells. In primary EOM cultures from TAO-p: a) CXCL10 was undetectable in the supernatant, IFNγ dose-dependently induced it, whereas TNFα did not; b) EOM produced basally low amounts of CCL2, TNFα dose-dependently induced it, whereas IFNγ did not; c) the combination of TNFα and IFNγ had a significant synergistic effect on CXCL10 and CCL2 secretion; and d) PPARγ agonists have an inhibitory role on the modulation of CXCL10, while they stimulate CCL2 secretion. EOM participates in the self-perpetuation of inflammation by releasing both Th1 (CXCL10) and Th2 (CCL2) chemokines under the influence of cytokines, in TAO. PPARγ agonist activation plays an inhibitory role on CXCL10, but stimulates the release of CCL2.


Subject(s)
Chemokine CCL2/metabolism , Chemokine CXCL10/metabolism , Graves Ophthalmopathy/immunology , Muscle, Skeletal/immunology , PPAR gamma/immunology , Humans , Muscle, Skeletal/metabolism , Myoblasts/immunology , Myoblasts/metabolism
15.
Mol Ther ; 22(5): 1008-17, 2014 May.
Article in English | MEDLINE | ID: mdl-24569833

ABSTRACT

Stem cell therapy is a promising strategy for treatment of muscular dystrophies. In addition to muscle fiber formation, reconstitution of functional stem cell pool by donor cells is vital for long-term treatment. We show here that some CD133(+) cells within human muscle are located underneath the basal lamina of muscle fibers, in the position of the muscle satellite cell. Cultured hCD133(+) cells are heterogeneous and multipotent, capable of forming myotubes and reserve satellite cells in vitro. They contribute to extensive muscle regeneration and satellite cell formation following intramuscular transplantation into irradiated and cryodamaged tibialis anterior muscles of immunodeficient Rag2-/γ chain-/C5-mice. Some donor-derived satellite cells expressed the myogenic regulatory factor MyoD, indicating that they were activated. In addition, when transplanted host muscles were reinjured, there was significantly more newly-regenerated muscle fibers of donor origin in treated than in control, nonreinjured muscles, indicating that hCD133(+) cells had given rise to functional muscle stem cells, which were able to activate in response to injury and contribute to a further round of muscle regeneration. Our findings provide new evidence for the location and characterization of hCD133(+) cells, and highlight that these cells are highly suitable for future clinical application.


Subject(s)
Antigens, CD/genetics , Cell- and Tissue-Based Therapy , Glycoproteins/genetics , Muscular Dystrophies/therapy , Peptides/genetics , Stem Cell Transplantation , AC133 Antigen , Animals , Antigens, CD/biosynthesis , Glycoproteins/biosynthesis , Humans , Mice , Muscular Dystrophies/genetics , MyoD Protein/biosynthesis , Myoblasts/cytology , Myoblasts/immunology , Myoblasts/metabolism , Regeneration/genetics , Regeneration/immunology , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/immunology , Satellite Cells, Skeletal Muscle/metabolism
16.
Protein Pept Lett ; 21(1): 45-51, 2014.
Article in English | MEDLINE | ID: mdl-23919379

ABSTRACT

Myostatin plays negative roles in muscle development. To block the inhibitory effects of myostatin on myogenesis, a 759 bp single chain variable fragment antibody (scFv) against myostatin was constructed and expressed in Escherichia coli. ELISA detection showed that the scFv could bind to myostatin, and change of the scFv N-terminal peptides decreased its binding affinity. MTT assay and cell morphology demonstrated that the cell number and viability of the C2C12 myoblast were enhanced by the scFv. Meanwhile, the scFv significantly inhibited the myostatin-induced expression of cyclin-dependent kinase inhibitor p21 and Smad binding element-luciferase activity. H2O2 increased the expression of Muscle RING Finger 1 (MuRF1) and Muscle Atrophy F-box (MAFbx) in myoblasts as well as myostatin and MuRF1 in myotubes, and the scFv significantly decreased the H2O2-elevated expression of these genes. Conclusively, the scFv we developed could antagonize the inhibitory effects of myostatin on myogenesis through Smad pathway and regulation of p21, MuRF1 and MAFbx gene expression. The scFv may have application in the therapy of muscular dystrophy and improvement of animal meat production.


Subject(s)
Muscle Development/physiology , Myoblasts/immunology , Myostatin/immunology , Single-Chain Antibodies/immunology , Antibody Affinity/immunology , Base Sequence , Cell Survival , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , Humans , Hydrogen Peroxide/pharmacology , Muscle Proteins/biosynthesis , Muscle, Skeletal/cytology , Muscle, Skeletal/growth & development , Muscular Dystrophies/therapy , Myoblasts/cytology , Protein Binding/physiology , SKP Cullin F-Box Protein Ligases/biosynthesis , Single-Chain Antibodies/biosynthesis , Smad Proteins/biosynthesis , Smad Proteins/immunology , Tripartite Motif Proteins , Ubiquitin-Protein Ligases/biosynthesis
17.
J Immunol Methods ; 393(1-2): 61-9, 2013 Jul 31.
Article in English | MEDLINE | ID: mdl-23603617

ABSTRACT

We investigated the effect of macrophage differentiation on the chemotactic activity to invade local damaged myoblasts using in vitro models of muscle injury. We found that: 1) the chemotactic activity of macrophages toward areas of damaged myoblasts might be induced more by live myoblasts than dead ones, 2) the chemotactic activity of macrophages is not due to velocity, but depends on the directionality toward damaged myoblast cells, and 3) macrophage differentiation strongly influence the chemotactic activity toward damaged myoblast cells through the expression of CCR2 and/or F-actin.


Subject(s)
Chemotaxis/immunology , Macrophages/immunology , Macrophages/pathology , Myoblasts/immunology , Myoblasts/pathology , Actins/immunology , Actins/metabolism , Animals , Cell Differentiation/immunology , Cell Line , Chemokine CCL2/immunology , Chemokine CCL2/metabolism , Macrophages/metabolism , Mice , Muscle, Skeletal/immunology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Diseases/immunology , Muscular Diseases/metabolism , Muscular Diseases/pathology , Myoblasts/metabolism , Receptors, CCR2/immunology , Receptors, CCR2/metabolism
18.
Biochem Biophys Res Commun ; 422(3): 522-6, 2012 Jun 08.
Article in English | MEDLINE | ID: mdl-22627135

ABSTRACT

CDA-II (cell differentiation agent II), isolated from healthy human urine, is a DNA methyltransferase inhibitor. Previous studies indicated that CDA-II played important roles in the regulation of cell growth and certain differentiation processes. However, it has not been determined whether CDA-II affects skeletal myogenesis. In this study, we investigated effects of CDA-II treatment on skeletal muscle progenitor cell differentiation, migration and proliferation. We found that CDA-II blocked differentiation of murine myoblasts C2C12 in a dose-dependent manner. CDA-II repressed expression of muscle transcription factors, such as Myogenin and Mef2c, and structural proteins, such as myosin heavy chain (Myh3), light chain (Mylpf) and MCK. Moreover, CDA-II inhibited C1C12 cell migration and proliferation. Thus, our data provide the first evidence that CDA-II inhibits growth and differentiation of muscle progenitor cells, suggesting that the use of CDA-II might affect skeletal muscle functions.


Subject(s)
Cell Differentiation/drug effects , DNA Modification Methylases/antagonists & inhibitors , Muscle Development/drug effects , Myoblasts, Skeletal/drug effects , Myoblasts/drug effects , Peptides/pharmacology , Phenylacetates/pharmacology , Animals , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Gene Expression/drug effects , MEF2 Transcription Factors , Mice , Muscle Development/genetics , Myoblasts/cytology , Myoblasts/immunology , Myoblasts, Skeletal/cytology , Myoblasts, Skeletal/enzymology , Myogenic Regulatory Factors/antagonists & inhibitors , Myogenin/antagonists & inhibitors , Myosin Heavy Chains/antagonists & inhibitors
19.
Cytokine Growth Factor Rev ; 22(3): 131-9, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21757394

ABSTRACT

Transforming Growth Factor-beta (TGF-ß) is a pro-sclerotic cytokine widely associated with the development of fibrosis in diabetic nephropathy. Central to the underlying pathology of tubulointerstitial fibrosis is epithelial-to-mesenchymal transition (EMT), or the trans-differentiation of tubular epithelial cells into myofibroblasts. This process is accompanied by a number of key morphological and phenotypic changes culminating in detachment of cells from the tubular basement membrane and migration into the interstitium. Ultimately these cells reside as activated myofibroblasts and further exacerbate the state of fibrosis. A large body of evidence supports a role for TGF-ß and downstream Smad signalling in the development and progression of renal fibrosis. Here we discuss a role for TGF-ß as the principle effector in the development of renal fibrosis in diabetic nephropathy, focusing on the role of the TGF-ß1 isoform and its downstream signalling intermediates, the Smad proteins. Specifically we review evidence for TGF-ß1 induced EMT in both the proximal and distal regions of the nephron and describe potential therapeutic strategies that may target TGF-ß1 activity.


Subject(s)
Diabetic Nephropathies/metabolism , Epithelial-Mesenchymal Transition , Fibroblasts/metabolism , Kidney Tubules/metabolism , Myoblasts/metabolism , Signal Transduction , Transforming Growth Factor beta1/metabolism , Animals , Cell Differentiation/immunology , Diabetic Nephropathies/immunology , Diabetic Nephropathies/pathology , Fibroblasts/immunology , Fibroblasts/pathology , Fibrosis/immunology , Fibrosis/metabolism , Fibrosis/pathology , Humans , Kidney Tubules/immunology , Kidney Tubules/pathology , Myoblasts/immunology , Myoblasts/pathology , Smad Proteins/immunology , Smad Proteins/metabolism , Transforming Growth Factor beta1/immunology
20.
J Muscle Res Cell Motil ; 32(1): 31-8, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21597958

ABSTRACT

We established a novel monoclonal antibody, Yaksa that is specific to a subpopulation of myogenic cells. The Yaksa antigen is not expressed on the surface of growing myoblasts but only on a subpopulation of myogenin-positive myocytes. When Yaksa antigen-positive mononucleated cells were freshly prepared from a murine myogenic cell by a cell sorter, they fused with each other and formed multinucleated myotubes shortly after replating while Yaksa antigen-negative cells scarcely generated myotubes. These results suggest that Yaksa could segregate fusion-competent, mononucleated cells from fusion-incompetent cells during muscle differentiation. The Yaksa antigen was also expressed in developing muscle and regenerating muscle in vivo and it was localized at sites of cell-cell contact between mono-nucleated muscle cells and between mono-nucleated muscle cells and myotubes. Thus, Yaksa that marks prefusion myocytes before myotube formation can be a useful tool to elucidate the cellular and molecular mechanisms of myogenic cell fusion.


Subject(s)
Antibodies, Monoclonal/isolation & purification , Muscle Cells/immunology , Muscle Development/immunology , Animals , Cell Differentiation/immunology , Cell Differentiation/physiology , Cell Line , Female , Mice , Mice, Inbred C57BL , Myoblasts/immunology , Myogenin/immunology , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...