Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.221
Filter
1.
Brain Nerve ; 76(5): 646-654, 2024 May.
Article in Japanese | MEDLINE | ID: mdl-38741508

ABSTRACT

Immune-mediated necrotizing myopathy (IMNM) is a form of autoimmune myositis characterized by the presence of necrotic and regenerating process as a major finding in the muscle. Anti-SRP and anti-HMGCR have been identified as IMNM-specific autoantibodies. Patients with this disease often present with severe muscle weakness and markedly elevated serum creatine kinase (CK) levels. Differentiation from muscular dystrophy is challenging in certain cases. When patients meet the condition "subacute onset", "hyperCKemia over 1000 IU/L", and "clinical diagnosis of muscular dystrophy lacking molecular diagnosis", the possibility of IMNM should be considered. Autoantibody measurement, including of anti-SRP and HMGCR antibodies, is recommended. Treatment with corticosteroid in combination with immunosuppressants, intravenous immunoglobulin, and rituximab can be performed.


Subject(s)
Autoantibodies , Necrosis , Humans , Autoantibodies/immunology , Autoimmune Diseases/immunology , Autoimmune Diseases/diagnosis , Myositis/immunology , Myositis/diagnosis , Hydroxymethylglutaryl CoA Reductases/immunology , Immunoglobulins, Intravenous/administration & dosage , Muscle, Skeletal/pathology , Muscle, Skeletal/immunology , Signal Recognition Particle/immunology
2.
Curr Top Dev Biol ; 158: 239-251, 2024.
Article in English | MEDLINE | ID: mdl-38670708

ABSTRACT

Muscle regeneration is a complex process orchestrated by multiple steps. Recent findings indicate that inflammatory responses could play central roles in bridging initial muscle injury responses and timely muscle injury reparation. The various types of immune cells and cytokines have crucial roles in muscle regeneration process. In this review, we provide an overview of the functions of acute inflammation in muscle regeneration.


Subject(s)
Immune System , Muscle, Skeletal , Regeneration , Regeneration/immunology , Regeneration/physiology , Animals , Humans , Muscle, Skeletal/physiology , Muscle, Skeletal/immunology , Inflammation/immunology , Cytokines/metabolism
3.
Mar Drugs ; 20(5)2022 May 05.
Article in English | MEDLINE | ID: mdl-35621964

ABSTRACT

Balenine is one of the endogenous imidazole dipeptides derived from marine products. It is composed of beta-alanine and 3-methyl-L-histidine, which exist mainly in the muscles of marine organisms. The physiological functions of dietary balenine are not well-known. In this study, we investigated whether the supplementation of dietary balenine was associated with muscle function in a cardiotoxin-indued muscle degeneration/regeneration model. Through morphological observation, we found that the supplementation of balenine-enriched extract promoted the regeneration stage. In addition, the expression of regeneration-related myogenic marker genes, such as paired box protein 7, MyoD1, myogenin, and Myh3, in a group of mice fed a balenine-enriched extract diet was higher than that in a group fed a normal diet. Moreover, the supplementation of balenine-enriched extract promoted the expression of anti-inflammatory cytokines as well as pro-inflammatory cytokines at the degeneration stage. Interestingly, phagocytic activity in the balenine group was significantly higher than that in the control group in vitro. These results suggest that balenine may promote the progress of muscle regeneration by increasing the phagocytic activity of macrophages.


Subject(s)
Dipeptides , Macrophages , Muscle, Skeletal , Phagocytosis , Animals , Cytokines/metabolism , Dipeptides/metabolism , Dipeptides/pharmacology , Imidazoles/metabolism , Macrophages/drug effects , Macrophages/physiology , Mice , Muscle, Skeletal/immunology , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiology , Phagocytosis/drug effects
4.
BMJ Case Rep ; 15(5)2022 May 23.
Article in English | MEDLINE | ID: mdl-35606030

ABSTRACT

Hydroxyl-methyl-glutaryl-Co-A reductase (HMGCR) immune mediated necrotising myopathy (IMNM) is a rare autoimmune myositis that is thought to be triggered by statins and responds to immunomodulation. We report a case of a woman in her 30s with HMGCR IMNM without a history of statin exposure who had a clear flare of her myositis after beginning mushroom supplements. Mushrooms are natural HMGCR inhibitors, and this is the first case to demonstrate a flare triggered by mushrooms in a patient with known HMGCR IMNM. This case highlights the importance of reviewing diet and supplements in patients with IMNM. It also emphasises the importance of strict statin avoidance for patients with IMNM even when the myositis is under good control.


Subject(s)
Agaricales , Autoimmune Diseases , Dietary Supplements , Muscular Diseases , Adult , Autoantibodies/immunology , Autoimmune Diseases/chemically induced , Autoimmune Diseases/diagnosis , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Dietary Supplements/adverse effects , Female , Humans , Hydroxymethylglutaryl CoA Reductases/adverse effects , Hydroxymethylglutaryl CoA Reductases/immunology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Muscle, Skeletal/immunology , Muscle, Skeletal/pathology , Muscular Diseases/chemically induced , Muscular Diseases/diagnosis , Muscular Diseases/immunology , Muscular Diseases/pathology , Myositis/chemically induced , Myositis/diagnosis , Myositis/immunology , Myositis/pathology , Necrosis/chemically induced , Necrosis/immunology , Phytotherapy/adverse effects , Symptom Flare Up
5.
Proc Natl Acad Sci U S A ; 119(15): e2111445119, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35377804

ABSTRACT

Volumetric muscle loss (VML) overwhelms the innate regenerative capacity of mammalian skeletal muscle (SkM), leading to numerous disabilities and reduced quality of life. Immune cells are critical responders to muscle injury and guide tissue resident stem cell­ and progenitor-mediated myogenic repair. However, how immune cell infiltration and intercellular communication networks with muscle stem cells are altered following VML and drive pathological outcomes remains underexplored. Herein, we contrast the cellular and molecular mechanisms of VML injuries that result in the fibrotic degeneration or regeneration of SkM. Following degenerative VML injuries, we observed the heightened infiltration of natural killer (NK) cells as well as the persistence of neutrophils beyond 2 wk postinjury. Functional validation of NK cells revealed an antagonistic role in neutrophil accumulation in part via inducing apoptosis and CCR1-mediated chemotaxis. The persistent infiltration of neutrophils in degenerative VML injuries was found to contribute to impairments in muscle stem cell regenerative function, which was also attenuated by transforming growth factor beta 1 (TGFß1). Blocking TGFß signaling reduced neutrophil accumulation and fibrosis and improved muscle-specific force. Collectively, these results enhance our understanding of immune cell­stem cell cross talk that drives regenerative dysfunction and provide further insight into possible avenues for fibrotic therapy exploration.


Subject(s)
Killer Cells, Natural , Muscle, Skeletal , Muscular Diseases , Neutrophils , Regeneration , Satellite Cells, Skeletal Muscle , Animals , Fibrosis , Killer Cells, Natural/immunology , Mice , Muscle, Skeletal/immunology , Muscle, Skeletal/pathology , Muscular Diseases/immunology , Muscular Diseases/pathology , Neutrophil Infiltration , Neutrophils/immunology , Regeneration/immunology , Satellite Cells, Skeletal Muscle/immunology , Transforming Growth Factor beta/metabolism
6.
Adipocyte ; 11(1): 190-201, 2022 12.
Article in English | MEDLINE | ID: mdl-35412419

ABSTRACT

Bariatric surgery (BS) is an effective treatment for obesity. Adipose tissue, liver tissue and skeletal muscle are important metabolic tissues. This study investigated hub genes and their association with immune infiltration in these metabolic tissues of obese patients after BS by bioinformatic analysis with Gene Expression Omnibus datasets. Differentially expressed genes (DEGs) were identified, and a protein-protein interaction network was constructed to identify hub genes. As a result, 121 common DEGs were identified and mainly enriched in cytokine-cytokine receptor interactions, chemokine signaling pathway, neutrophil activation and immune responses. Immune cell infiltration analysis showed that the abundance of M1 macrophages was significantly lower in adipose and liver tissue after BS (p<0.05). Ten hub genes (TYROBP, TLR8, FGR, NCF2, HCK, CCL2, LAPTM5, MNDA and S100A9) that were all downregulated after BS were also associated with immune cells. Consistently, results in the validated dataset showed that the expression levels of these hub genes were increased in obese patients and mice, and decreased after BS. In conclusion, this study analysed the potential immune and inflammatory mechanisms of BS in three key metabolic tissues of obese patients, and revealed hub genes associated with immune cell infiltration, thus providing potential targets for obesity treatment.


Subject(s)
Bariatric Surgery , Immune System Phenomena , Obesity , Adipose Tissue/immunology , Animals , Chemokines , Cytokines , Gene Expression Profiling , Humans , Immune System Phenomena/genetics , Liver/immunology , Mice , Muscle, Skeletal/immunology , Neutrophils , Obesity/complications , Obesity/genetics , Obesity/metabolism , Receptors, Cytokine
7.
Nat Commun ; 13(1): 879, 2022 02 15.
Article in English | MEDLINE | ID: mdl-35169163

ABSTRACT

Dysregulation of the balance between pro-inflammatory and anti-inflammatory macrophages has a key function in the pathogenesis of Duchenne muscular dystrophy (DMD), a fatal genetic disease. We postulate that an evolutionarily ancient protective mechanism against infection, known as trained immunity, drives pathological inflammation in DMD. Here we show that bone marrow-derived macrophages from a murine model of DMD (mdx) exhibit cardinal features of trained immunity, consisting of transcriptional hyperresponsiveness associated with metabolic and epigenetic remodeling. The hyperresponsive phenotype is transmissible by bone marrow transplantation to previously healthy mice and persists for up to 11 weeks post-transplant. Mechanistically, training is induced by muscle extract in vitro. The functional and epigenetic changes in bone marrow-derived macrophages from dystrophic mice are TLR4-dependent. Adoptive transfer experiments further support the TLR4-dependence of trained macrophages homing to damaged muscles from the bone marrow. Collectively, this suggests that a TLR4-regulated, memory-like capacity of innate immunity induced at the level of the bone marrow promotes dysregulated inflammation in DMD.


Subject(s)
Bone Marrow Transplantation , Immunity, Innate/immunology , Macrophages/immunology , Muscle, Skeletal/pathology , Muscular Dystrophy, Duchenne/pathology , Toll-Like Receptor 4/immunology , Animals , Bone Marrow Cells/immunology , Cell Line , Disease Models, Animal , Inflammation/immunology , L Cells , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Mice, Knockout , Muscle, Skeletal/immunology , Muscular Dystrophy, Duchenne/immunology , Tissue Extracts/pharmacology , Transcription, Genetic/genetics
9.
Int J Mol Sci ; 23(3)2022 Feb 07.
Article in English | MEDLINE | ID: mdl-35163799

ABSTRACT

Endurance exercise induces various adaptations that yield health benefits; however, the underlying molecular mechanism has not been fully elucidated. Given that it has recently been accepted that inflammatory responses are required for a specific muscle adaptation after exercise, this study investigated whether toll-like receptor (TLR) 4, a pattern recognition receptor that induces proinflammatory cytokines, is responsible for exercise-induced adaptations in mouse skeletal muscle. The TLR4 mutant (TLR4m) and intact TLR4 control mice were each divided into 2 groups (sedentary and voluntary wheel running) and were housed for six weeks. Next, we removed the plantaris muscle and evaluated the expression of cytokines and muscle regulators. Exercise increased cytokine expression in the controls, whereas a smaller increase was observed in the TLR4m mice. Mitochondrial markers and mitochondrial biogenesis inducers, including peroxisome proliferator-activated receptor beta and heat shock protein 72, were increased in the exercised controls, whereas this upregulation was attenuated in the TLR4m mice. In contrast, exercise increased the expression of molecules such as peroxisome proliferator-activated receptor-gamma coactivator 1-alpha and glucose transporter 4 in both the controls and TLR4m mice. Our findings indicate that exercise adaptations such as mitochondrial biogenesis are mediated via TLR4, and that TLR4-mediated inflammatory responses could be involved in the mechanism of adaptation.


Subject(s)
Endurance Training/veterinary , Inflammation/genetics , Lipopolysaccharides/adverse effects , Muscle, Skeletal/immunology , Toll-Like Receptor 4/genetics , Adaptation, Physiological , Animals , Cytokines/metabolism , Glucose Transporter Type 4/metabolism , Inflammation/chemically induced , Inflammation/metabolism , Male , Mice , Mitochondria, Muscle/metabolism , Muscle, Skeletal/drug effects , Mutation , Organelle Biogenesis , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Physical Conditioning, Animal , Up-Regulation
10.
Nat Commun ; 13(1): 166, 2022 01 10.
Article in English | MEDLINE | ID: mdl-35013338

ABSTRACT

Muscle cell death in polymyositis is induced by CD8+ cytotoxic T lymphocytes. We hypothesized that the injured muscle fibers release pro-inflammatory molecules, which would further accelerate CD8+ cytotoxic T lymphocytes-induced muscle injury, and inhibition of the cell death of muscle fibers could be a novel therapeutic strategy to suppress both muscle injury and inflammation in polymyositis. Here, we show that the pattern of cell death of muscle fibers in polymyositis is FAS ligand-dependent necroptosis, while that of satellite cells and myoblasts is perforin 1/granzyme B-dependent apoptosis, using human muscle biopsy specimens of polymyositis patients and models of polymyositis in vitro and in vivo. Inhibition of necroptosis suppresses not only CD8+ cytotoxic T lymphocytes-induced cell death of myotubes but also the release of inflammatory molecules including HMGB1. Treatment with a necroptosis inhibitor or anti-HMGB1 antibodies ameliorates myositis-induced muscle weakness as well as muscle cell death and inflammation in the muscles. Thus, targeting necroptosis in muscle cells is a promising strategy for treating polymyositis providing an alternative to current therapies directed at leukocytes.


Subject(s)
HMGB1 Protein/antagonists & inhibitors , Imidazoles/pharmacology , Indoles/pharmacology , Muscle Fibers, Skeletal/drug effects , Myositis/prevention & control , Necroptosis/drug effects , Polymyositis/genetics , Animals , Antibodies, Neutralizing/pharmacology , C-Reactive Protein/administration & dosage , Fas Ligand Protein/genetics , Fas Ligand Protein/immunology , Female , Gene Expression Regulation , Granzymes/genetics , Granzymes/immunology , HMGB1 Protein/genetics , HMGB1 Protein/immunology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle Fibers, Skeletal/immunology , Muscle Fibers, Skeletal/pathology , Muscle Strength/drug effects , Muscle Strength/immunology , Muscle, Skeletal/drug effects , Muscle, Skeletal/immunology , Muscle, Skeletal/pathology , Myositis/chemically induced , Myositis/genetics , Myositis/immunology , Necroptosis/genetics , Necroptosis/immunology , Perforin/genetics , Perforin/immunology , Polymyositis/immunology , Polymyositis/pathology , Signal Transduction , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/pathology
11.
J Nutr Biochem ; 99: 108855, 2022 01.
Article in English | MEDLINE | ID: mdl-34517096

ABSTRACT

Patients with inflammatory bowel disease (IBD) have higher incidence of extraintestinal manifestations (EIM), including liver disorders, sarcopenia, and neuroinflammation. Fermented rice bran (FRB), generated from rice bran (RB), is rich in bioactive compounds, and exhibits anti-colitis activity. However, its role in EIM prevention is still unclear. Here, for the first time, we investigated whether EIM in female C57Bl/6N mice is attenuated by FRB supplementation. EIM was induced by repeated administration of 1.5% dextran sulfate sodium (DSS) in drinking water (4 d) followed by drinking water (12 d). Mice were divided into 3 groups-control (AIN93M), 10% RB, and 10% FRB. FRB ameliorated relapsing colitis and inflammation in muscle by significantly lowering proinflammatory cytokines Tnf-α and Il-6 in serum and advanced glycation end product-specific receptor (Ager) in serum and muscle when compared with the RB and control groups. As FRB reduced aspartate aminotransferase levels and oxidative stress, it might prevent liver disorders. FRB downregulated proinflammatory cytokine and chemokine transcripts responsible for neuroinflammation in the hippocampus and upregulated mRNA expression of G protein coupled receptors (GPRs), Gpr41 and Gpr43, in small and large intestines, which may explain the FRB-mediated protective mechanism. Hence, FRB can be used as a supplement to prevent IBD-associated EIM.


Subject(s)
Colitis/drug therapy , Colitis/immunology , Dietary Fiber/administration & dosage , Oryza/chemistry , Plant Preparations/administration & dosage , Animals , Chemokines/genetics , Chemokines/immunology , Chronic Disease/therapy , Colitis/chemically induced , Colitis/genetics , Dextran Sulfate/adverse effects , Dietary Fiber/analysis , Dietary Supplements/analysis , Disease Models, Animal , Female , Hippocampus/immunology , Humans , Interleukin-6/genetics , Interleukin-6/immunology , Intestines/immunology , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/immunology , Oxidative Stress , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
12.
Am J Physiol Regul Integr Comp Physiol ; 322(1): R41-R54, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34786967

ABSTRACT

Macrophages are one of the top players when considering immune cells involved with tissue homeostasis. Recently, increasing evidence has demonstrated that macrophages could also present two major subsets during tissue healing: proliferative macrophages (M1-like), which are responsible for increasing myogenic cell proliferation, and restorative macrophages (M2-like), which are involved in the end of the mature muscle myogenesis. The participation and characterization of these macrophage subsets are critical during myogenesis to understand the inflammatory role of macrophages during muscle recovery and to create supportive strategies that can improve mass muscle maintenance. Indeed, most of our knowledge about macrophage subsets comes from skeletal muscle damage protocols, and we still do not know how these subsets can contribute to skeletal muscle adaptation. Thus, this narrative review aims to collect and discuss studies demonstrating the involvement of different macrophage subsets during the skeletal muscle damage/regeneration process, showcasing an essential role of these macrophage subsets during muscle adaptation induced by acute and chronic exercise programs.


Subject(s)
Cell Proliferation , Exercise , Hypertrophy/metabolism , Inflammation Mediators/metabolism , Inflammation/metabolism , Macrophages/metabolism , Muscle, Skeletal/metabolism , Regeneration , Skeletal Muscle Enlargement , Animals , Humans , Hypertrophy/immunology , Hypertrophy/pathology , Hypertrophy/physiopathology , Inflammation/immunology , Inflammation/pathology , Inflammation/physiopathology , Macrophages/immunology , Muscle, Skeletal/immunology , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Phenotype , Signal Transduction
13.
Front Immunol ; 12: 783616, 2021.
Article in English | MEDLINE | ID: mdl-34899749

ABSTRACT

Idiopathic inflammatory myopathy (IIM) is a heterogeneous group of acquired, autoimmune muscle diseases characterized by muscle inflammation and extramuscular involvements. Present literatures have revealed that dysregulated cell death in combination with impaired elimination of dead cells contribute to the release of autoantigens, damage-associated molecular patterns (DAMPs) and inflammatory cytokines, and result in immune responses and tissue damages in autoimmune diseases, including IIMs. This review summarizes the roles of various forms of programmed cell death pathways in the pathogenesis of IIMs and provides evidence for potential therapeutic targets.


Subject(s)
Muscle, Skeletal/pathology , Myositis/pathology , Regulated Cell Death , Alarmins/immunology , Alarmins/metabolism , Animals , Autoantigens/immunology , Autoantigens/metabolism , Cytokines/immunology , Cytokines/metabolism , Humans , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Muscle, Skeletal/immunology , Muscle, Skeletal/metabolism , Myositis/immunology , Myositis/metabolism , Signal Transduction
14.
Front Immunol ; 12: 777502, 2021.
Article in English | MEDLINE | ID: mdl-34925350

ABSTRACT

Background: Immune-mediated necrotizing myopathy (IMNM) is characterized by markedly elevated creatinine kinase and histologically scattered necrotic muscle fibers and generally associated with autoantibodies against signal recognition particle (SRP) or 3-hydroxy-3-methylglutaryl-coA-reductase (HMGCR). Poor clinical response to conventional therapies and relapses commonly occur in severe cases. Anti-B-cell therapies have been used in refractory/relapsing cases. Methods: The characteristics of a patient with IMNM associated with anti-SRP antibodies including physical examination, laboratory tests, and disease activity assessment were evaluated. Conventional therapy, belimumab treatment schedule, and follow-up data were recorded. Medical records of IMNM patients treated in our department from September 2014 to June 2021 were reviewed to evaluate the efficacy and safety of anti-B-cell therapy for anti-SRP IMNM. A literature review of patients with anti-SRP IMNM treated with anti-B-cell therapies was performed. Results: We describe a case of a 47-year-old woman with IMNM associated with anti-SRP antibodies who relapsed twice after conventional therapy but showed good response and tolerance to belimumab at 28 weeks follow-up. In this review, three patients from our department were treated with rituximab. Two of the three patients rapidly improved after treatment. Twenty patients and five retrospective studies were included in the literature review. All patients were administered rituximab as an anti-B-cell drug. Conclusion: Despite a lack of rigorous clinical trials, considerable experience demonstrated that anti-B-cell therapy might be effective for patients with IMNM associated with anti-SRP antibodies. Belimumab in association with steroids might be an encouraging option for refractory/relapsing cases.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Autoantibodies/blood , Immunosuppressive Agents/therapeutic use , Muscular Diseases/drug therapy , Signal Recognition Particle/immunology , Antibodies, Monoclonal, Humanized/pharmacology , Autoantibodies/immunology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Biopsy , Female , Glucocorticoids/pharmacology , Glucocorticoids/therapeutic use , Humans , Immunosuppressive Agents/pharmacology , Middle Aged , Muscle, Skeletal/immunology , Muscle, Skeletal/pathology , Muscular Diseases/blood , Muscular Diseases/diagnosis , Muscular Diseases/immunology , Necrosis/blood , Necrosis/diagnosis , Necrosis/drug therapy , Necrosis/immunology , Retrospective Studies , Treatment Outcome
15.
Life Sci ; 287: 120117, 2021 Dec 15.
Article in English | MEDLINE | ID: mdl-34740577

ABSTRACT

Denervated skeletal muscular atrophy is primarily characterized by loss of muscle strength and mass and an unideal functional recovery of the muscle after extended denervation. This review emphasizes the interaction between the immune system and the denervated skeletal muscle. Immune cells such as neutrophils, macrophages and T-cells are activated and migrate to denervated muscle, where they release a high concentration of cytokines and chemokines. The migration of these immune cells, the transformation of different functional immune cell subtypes, and the cytokine network in the immune microenvironment may be involved in the regulatory process of muscle atrophy or repair. However, the exact mechanisms of the interaction between these immune cells and immune molecules in skeletal muscles are unclear. In this paper, the immune microenvironment regulation of muscle atrophy induced by peripheral nerve injury is reviewed.


Subject(s)
Biomedical Research/trends , Cellular Microenvironment/physiology , Immunity, Cellular/physiology , Muscle, Skeletal/immunology , Muscular Atrophy/immunology , Peripheral Nerve Injuries/immunology , Animals , Humans , Macrophages/immunology , Macrophages/metabolism , Muscle Denervation/methods , Muscle, Skeletal/innervation , Muscle, Skeletal/metabolism , Muscular Atrophy/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Peripheral Nerve Injuries/metabolism
16.
Nat Commun ; 12(1): 6769, 2021 11 24.
Article in English | MEDLINE | ID: mdl-34819506

ABSTRACT

Adeno-associated virus (AAV)-mediated CRISPR-Cas9 editing holds promise to treat many diseases. The immune response to bacterial-derived Cas9 has been speculated as a hurdle for AAV-CRISPR therapy. However, immunological consequences of AAV-mediated Cas9 expression have thus far not been thoroughly investigated in large mammals. We evaluate Cas9-specific immune responses in canine models of Duchenne muscular dystrophy (DMD) following intramuscular and intravenous AAV-CRISPR therapy. Treatment results initially in robust dystrophin restoration in affected dogs but also induces muscle inflammation, and Cas9-specific humoral and cytotoxic T-lymphocyte (CTL) responses that are not prevented by the muscle-specific promoter and transient prednisolone immune suppression. In normal dogs, AAV-mediated Cas9 expression induces similar, though milder, immune responses. In contrast, other therapeutic (micro-dystrophin and SERCA2a) and reporter (alkaline phosphatase, AP) vectors result in persistent expression without inducing muscle inflammation. Our results suggest Cas9 immunity may represent a critical barrier for AAV-CRISPR therapy in large mammals.


Subject(s)
CRISPR-Cas Systems/immunology , Genetic Therapy/adverse effects , Genetic Vectors/immunology , Muscle, Skeletal/immunology , Muscular Dystrophy, Duchenne/therapy , Animals , CRISPR-Cas Systems/genetics , Dependovirus/genetics , Disease Models, Animal , Dogs , Dystrophin/genetics , Dystrophin/immunology , Gene Editing/methods , Genes, Reporter/genetics , Genes, Reporter/immunology , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Humans , Muscle, Skeletal/pathology , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/immunology , Muscular Dystrophy, Duchenne/pathology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/immunology
17.
Front Immunol ; 12: 729017, 2021.
Article in English | MEDLINE | ID: mdl-34603301

ABSTRACT

Piscine orthoreovirus (PRV-1) infection causes heart and skeletal muscle inflammation (HSMI) in farmed Atlantic salmon (Salmo salar). The virus is also associated with focal melanized changes in white skeletal muscle where PRV-1 infection of macrophages appears to be important. In this study, we studied the macrophage polarization into M1 (pro-inflammatory) and M2 (anti-inflammatory) phenotypes during experimentally induced HSMI. The immune response in heart with HSMI lesions was characterized by CD8+ and MHC-I expressing cells and not by polarized macrophages. Fluorescent in situ hybridization (FISH) assays revealed localization of PRV-1 in a few M1 macrophages in both heart and skeletal muscle. M2 type macrophages were widely scattered in the heart and were more abundant in heart compared to the skeletal muscle. However, the M2 macrophages did not co-stain for PRV-1. There was a strong cellular immune response to the infection in the heart compared to that of the skeletal muscle, seen as increased MHC-I expression, partly in cells also containing PRV-1 RNA, and a high number of cytotoxic CD8+ granzyme producing cells that targeted PRV-1. In skeletal muscle, MHC-I expressing cells and CD8+ cells were dispersed between myocytes, but these cells did not stain for PRV-1. Gene expression analysis by RT-qPCR complied with the FISH results and confirmed a drop in level of PRV-1 following the cell mediated immune response. Overall, the results indicated that M1 macrophages do not contribute to the initial development of HSMI. However, large numbers of M2 macrophages reside in the heart and may contribute to the subsequent fast recovery following clearance of PRV-1 infection.


Subject(s)
CD8-Positive T-Lymphocytes/virology , Fish Diseases/virology , Heart/virology , Macrophages/virology , Orthoreovirus/pathogenicity , Retroviridae Infections/virology , Salmo salar/virology , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Fish Diseases/immunology , Fish Diseases/metabolism , Host-Pathogen Interactions , Immunity, Cellular , Macrophages/immunology , Macrophages/metabolism , Muscle, Skeletal/immunology , Muscle, Skeletal/metabolism , Muscle, Skeletal/virology , Myocardium/immunology , Myocardium/metabolism , Orthoreovirus/immunology , Phenotype , Retroviridae Infections/immunology , Retroviridae Infections/metabolism , Salmo salar/immunology , Salmo salar/metabolism , Time Factors , Viral Load
18.
Front Endocrinol (Lausanne) ; 12: 697204, 2021.
Article in English | MEDLINE | ID: mdl-34594301

ABSTRACT

Skeletal muscles secrete various factors, such as proteins/peptides, nucleotides, and metabolites, which are referred to as myokines. Many of these factors are transported into extracellular bodily fluids in a free or protein-bound form. Furthermore, several secretory factors have been shown to be wrapped up by small vesicles, particularly exosomes, secreted into circulation, and subsequently regulate recipient cells. Thus, exosome contents can be recognized as myokines. In recipient cells, proteins, microRNAs, and metabolites in exosomes can regulate the expression and activity of target proteins associated with nutrient metabolism and immune function. The levels of circulating exosomes and their contents are altered in muscle disorders and metabolic-related states, such as metabolic dysfunction, sarcopenia, and physical fitness. Therefore, such circulating factors could mediate various interactions between skeletal muscle and other organs and may be useful as biomarkers reflecting physiological and pathological states associated with muscular function. Here, this review summarizes secretory regulation of muscle-derived exosomes. Their metabolic and immunological roles and the significance of their circulating levels are also discussed.


Subject(s)
Exosomes/physiology , Muscle, Skeletal/immunology , Muscle, Skeletal/metabolism , Animals , Biomarkers/metabolism , Cell Communication/genetics , Cell Communication/immunology , Energy Metabolism/genetics , Energy Metabolism/physiology , Humans , Immune System/metabolism , Immune System/physiology , MicroRNAs/genetics , MicroRNAs/metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/ultrastructure
19.
Aging (Albany NY) ; 13(18): 21962-21974, 2021 09 19.
Article in English | MEDLINE | ID: mdl-34537761

ABSTRACT

Dulaglutide, a glucagon-like peptide-1 receptor (GLP-1R) agonist, is widely used to treat diabetes. However, its effects on muscle wasting due to aging are poorly understood. In the current study, we investigated the therapeutic potential and underlying mechanism of dulaglutide in muscle wasting in aged mice. Dulaglutide improved muscle mass and strength in aged mice. Histological analysis revealed that the cross-sectional area of the tibialis anterior (TA) in the dulaglutide-treated group was thicker than that in the vehicle group. Moreover, dulaglutide increased the shift toward middle and large-sized fibers in both young and aged mice compared to the vehicle. Dulaglutide increased myofiber type I and type IIa in young (18.5% and 8.2%) and aged (1.8% and 19.7%) mice, respectively, compared to the vehicle group. Peroxisome proliferator-activated receptor-gamma coactivator-1α (PGC-1α), a master regulator of mitochondrial biogenesis, decreased but increased by dulaglutide in aged mice. The expression of atrophic factors such as myostatin, atrogin-1, and muscle RING-finger protein-1 was decreased in aged mice, whereas that of the myogenic factor, MyoD, was increased in both young and aged mice following dulaglutide treatment. In aged mice, optic atrophy-1 (OPA-1) protein was decreased, whereas Toll-like receptor-9 (TLR-9) and its targeting inflammatory cytokines (interleukin-6 [IL-6] and tumor necrosis factor-α [TNF-α]) were elevated in the TA and quadriceps (QD) muscles. In contrast, dulaglutide administration reversed this expression pattern, thereby significantly attenuating the expression of inflammatory cytokines in aged mice. These data suggest that dulaglutide may exert beneficial effects in the treatment of muscle wasting due to aging.


Subject(s)
Aging/metabolism , Glucagon-Like Peptides/analogs & derivatives , Immunoglobulin Fc Fragments/administration & dosage , Muscle, Skeletal/physiopathology , Recombinant Fusion Proteins/administration & dosage , Sarcopenia/drug therapy , Sarcopenia/immunology , Toll-Like Receptor 9/immunology , Aging/drug effects , Aging/genetics , Animals , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/immunology , Glucagon-Like Peptides/administration & dosage , Humans , Hypoglycemic Agents/administration & dosage , Interleukin-6/genetics , Interleukin-6/immunology , Male , Mice , Muscle Proteins/genetics , Muscle Proteins/immunology , Muscle, Skeletal/drug effects , Muscle, Skeletal/immunology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/immunology , SKP Cullin F-Box Protein Ligases/genetics , SKP Cullin F-Box Protein Ligases/immunology , Sarcopenia/etiology , Sarcopenia/genetics , Signal Transduction/drug effects , Toll-Like Receptor 9/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
20.
Sci Rep ; 11(1): 19160, 2021 09 27.
Article in English | MEDLINE | ID: mdl-34580412

ABSTRACT

This study investigated whether oxidative and glycolytic rat skeletal muscles respond differently to a high-fat (HF) sucrose-enriched diet with respect to diacylglycerol (DAG) and ceramides accumulation, protein kinase C (PKC) activation, glucose metabolism, and the expression of inflammatory genes. HF diet (8 weeks) suppressed insulin-stimulated glycogen synthesis and glucose oxidation in soleus (Sol), extensor digitorum longus (EDL) and epitrochlearis (Epit) muscles. However, DAG and ceramides levels increased in Sol and EDL, but not in Epit muscles of HF-fed rats. Additionally, membrane-bound PKC-delta and PKC-theta increased in Sol and EDL, whereas in Epit muscles both PKC isoforms were reduced by HF diet. In Epit muscles, HF diet also increased the expression of tumor necrosis factor-α (TNF-α) receptors (CD40 and FAS), toll-like receptor 4 (TLR4), and nuclear factor kappa light polypeptide gene enhancer in B cells (NF-kB), whereas in Sol and EDL muscles the expression of these inflammatory genes remained unchanged upon HF feeding. In conclusion, HF diet caused DAG and ceramides accumulation, PKC activation, and the induction of inflammatory pathways in a fiber type-specific manner. These findings help explain why oxidative and glycolytic muscles similarly develop insulin resistance, despite major differences in their metabolic characteristics and responsiveness to dietary lipid abundance.


Subject(s)
Glycolysis/immunology , Insulin Resistance/immunology , Muscle, Skeletal/metabolism , Obesity/metabolism , Animals , Ceramides/analysis , Ceramides/metabolism , Diet, High-Fat/adverse effects , Dietary Sucrose/adverse effects , Diglycerides/analysis , Diglycerides/metabolism , Disease Models, Animal , Humans , Inflammation/diagnosis , Inflammation/immunology , Inflammation/metabolism , Insulin/metabolism , Male , Muscle, Skeletal/immunology , Obesity/etiology , Obesity/immunology , Oxidative Stress/immunology , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...