Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.225
Filtrar
Más filtros

Intervalo de año de publicación
1.
Annu Rev Immunol ; 34: 609-33, 2016 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-27168246

RESUMEN

The immune system is responsible for defending an organism against the myriad of microbial invaders it constantly confronts. It has become increasingly clear that the immune system has a second major function: the maintenance of organismal homeostasis. Foxp3(+)CD4(+) regulatory T cells (Tregs) are important contributors to both of these critical activities, defense being the primary purview of Tregs circulating through lymphoid organs, and homeostasis ensured mainly by their counterparts residing in parenchymal tissues. This review focuses on so-called tissue Tregs. We first survey existing information on the phenotype, function, sustaining factors, and human equivalents of the three best-characterized tissue-Treg populations-those operating in visceral adipose tissue, skeletal muscle, and the colonic lamina propria. We then attempt to distill general principles from this body of work-as concerns the provenance, local adaptation, molecular sustenance, and targets of action of tissue Tregs, in particular.


Asunto(s)
Tejido Adiposo/inmunología , Colon/inmunología , Membrana Mucosa/inmunología , Músculo Esquelético/inmunología , Linfocitos T Reguladores/inmunología , Animales , Factores de Transcripción Forkhead/metabolismo , Homeostasis , Humanos , Especificidad de Órganos
2.
Nat Immunol ; 20(5): 626-636, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30936495

RESUMEN

Muscle damage elicits a sterile immune response that facilitates complete regeneration. Here, we used mass spectrometry-based lipidomics to map the mediator lipidome during the transition from inflammation to resolution and regeneration in skeletal muscle injury. We observed temporal regulation of glycerophospholipids and production of pro-inflammatory lipid mediators (for example, leukotrienes and prostaglandins) and specialized pro-resolving lipid mediators (for example, resolvins and lipoxins) that were modulated by ibuprofen. These time-dependent profiles were recapitulated in sorted neutrophils and Ly6Chi and Ly6Clo muscle-infiltrating macrophages, with a distinct pro-resolving signature observed in Ly6Clo macrophages. RNA sequencing of macrophages stimulated with resolvin D2 showed similarities to transcriptional changes found during the temporal transition from Ly6Chi macrophage to Ly6Clo macrophage. In vivo, resolvin D2 increased Ly6Clo macrophages and functional improvement of the regenerating muscle. These results reveal dynamic lipid mediator signatures of innate immune cells and provide a proof of concept for their exploitable effector roles in muscle regeneration.


Asunto(s)
Mediadores de Inflamación/inmunología , Lípidos/inmunología , Macrófagos/inmunología , Músculo Esquelético/inmunología , Regeneración/inmunología , Animales , Ácidos Docosahexaenoicos/inmunología , Ácidos Docosahexaenoicos/farmacología , Expresión Génica/efectos de los fármacos , Expresión Génica/inmunología , Perfilación de la Expresión Génica , Ontología de Genes , Secuenciación de Nucleótidos de Alto Rendimiento , Metabolismo de los Lípidos/inmunología , Lípidos/análisis , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Ratones Endogámicos C57BL , Músculo Esquelético/lesiones , Músculo Esquelético/fisiopatología , Regeneración/genética
3.
Cell ; 155(6): 1282-95, 2013 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-24315098

RESUMEN

Long recognized to be potent suppressors of immune responses, Foxp3(+)CD4(+) regulatory T (Treg) cells are being rediscovered as regulators of nonimmunological processes. We describe a phenotypically and functionally distinct population of Treg cells that rapidly accumulated in the acutely injured skeletal muscle of mice, just as invading myeloid-lineage cells switched from a proinflammatory to a proregenerative state. A Treg population of similar phenotype accumulated in muscles of genetically dystrophic mice. Punctual depletion of Treg cells during the repair process prolonged the proinflammatory infiltrate and impaired muscle repair, while treatments that increased or decreased Treg activities diminished or enhanced (respectively) muscle damage in a dystrophy model. Muscle Treg cells expressed the growth factor Amphiregulin, which acted directly on muscle satellite cells in vitro and improved muscle repair in vivo. Thus, Treg cells and their products may provide new therapeutic opportunities for wound repair and muscular dystrophies.


Asunto(s)
Músculo Esquelético/citología , Músculo Esquelético/fisiología , Regeneración , Linfocitos T Reguladores/fisiología , Anfirregulina , Animales , Familia de Proteínas EGF , Glicoproteínas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Tejido Linfoide/citología , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/inmunología , Músculo Esquelético/lesiones , Distrofias Musculares/patología , Distrofias Musculares/fisiopatología , Distrofias Musculares/terapia , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología , Transcriptoma
4.
Proc Natl Acad Sci U S A ; 119(15): e2111445119, 2022 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-35377804

RESUMEN

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.


Asunto(s)
Células Asesinas Naturales , Músculo Esquelético , Enfermedades Musculares , Neutrófilos , Regeneración , Células Satélite del Músculo Esquelético , Animales , Fibrosis , Células Asesinas Naturales/inmunología , Ratones , Músculo Esquelético/inmunología , Músculo Esquelético/patología , Enfermedades Musculares/inmunología , Enfermedades Musculares/patología , Infiltración Neutrófila , Neutrófilos/inmunología , Regeneración/inmunología , Células Satélite del Músculo Esquelético/inmunología , Factor de Crecimiento Transformador beta/metabolismo
5.
J Autoimmun ; 148: 103286, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39033686

RESUMEN

Immune-mediated necrotizing myopathy (IMNM) is a rare and newly recognized autoimmune disease within the spectrum of idiopathic inflammatory myopathies. It is characterized by myositis-specific autoantibodies, elevated serum creatine kinase levels, inflammatory infiltrate, and weakness. IMNM can be classified into three subtypes based on the presence or absence of specific autoantibodies: anti-signal recognition particle myositis, anti-3-hydroxy-3-methylglutaryl-coenzyme A reductase myositis, and seronegative IMNM. In recent years, IMNM has gained increasing attention and emerged as a research hotspot. Recent studies have suggested that the pathogenesis of IMNM is linked to aberrant activation of immune system, including immune responses mediated by antibodies, complement, and immune cells, particularly macrophages, as well as abnormal release of inflammatory factors. Non-immune mechanisms such as autophagy and endoplasmic reticulum stress also participate in this process. Additionally, genetic variations associated with IMNM have been identified, providing new insights into the genetic mechanisms of the disease. Progress has also been made in IMNM treatment research, including the use of immunosuppressants and the development of biologics. Despite the challenges in understanding the etiology and treatment of IMNM, the latest research findings offer important guidance and insights for delving deeper into the disease's pathogenic mechanisms and identifying new therapeutic strategies.


Asunto(s)
Autoanticuerpos , Miositis , Humanos , Miositis/inmunología , Miositis/terapia , Miositis/patología , Miositis/diagnóstico , Miositis/etiología , Autoanticuerpos/inmunología , Necrosis/inmunología , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/terapia , Enfermedades Autoinmunes/etiología , Enfermedades Autoinmunes/diagnóstico , Animales , Inmunosupresores/uso terapéutico , Músculo Esquelético/inmunología , Músculo Esquelético/patología , Músculo Esquelético/metabolismo
6.
J Autoimmun ; 146: 103232, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38692172

RESUMEN

The link between type I IFN and adaptive immunity, especially T-cell immunity, in JDM still remained largely unclear. This study aimed to understand the effect of elevated type I IFN signaling on CD8+ T cell-associated muscle damage in juvenile dermatomyositis (JDM). This study used flow cytometry (FC) and RT‒PCR were used to examine the circulating cell ratio and type I IFN response. And scRNA-seq was used to examine peripheral immunity in 6 active JDM patients, 3 stable JDM patients, 3 juvenile IMNM patients and 3 age-matched healthy children. In vivo validation experiments were conducted using a mouse model induced by STING agonists and an experimental autoimmune myositis model (EAM). In vitro experiments were conducted using isolated CD8+ T-cells from JDM patients and mice. We found that active JDM patients showed an extensive type I IFN response and a decreased CD8+ T-cell ratio in the periphery (P < 0.05), which was correlated with muscle involvement (P < 0.05). Both new active JDM patients and all active JDM patients showed decreased CD8+ TCM cell ratios compared with age and gender matched stable JDM patients (P < 0.05). Compared with new pediatirc systemic lupus erythematosus (SLE) patients, new active JDM patients displayed decreased CD8+ T-cell and CD8+ TCM cell ratios (P < 0.05). Active JDM patient skeletal muscle biopsies displayed an elevated type I IFN response, upregulated MHC-I expression and CD8+ T-cell infiltration, which was validated in EAM mice. sc-RNAseq demonstrated that type I IFN signalling is the kinetic factor of abnormal differentiation and enhances the cytotoxicity of peripheral CD8+ T cells in active JDM patients, which was confirmed by in vivo and in vitro validation experiments. In summary, the elevated type I IFN signalling affected the differentiation and function of CD8+ T cells in active JDM patients. Skeletal muscle-infiltrating CD8+ T cells might migrate from the periphery under the drive of type I IFN and increased MHC I signals. Therapies targeting autoantigen-specific CD8+ T cells may represent a potential new treatment direction.


Asunto(s)
Autoantígenos , Linfocitos T CD8-positivos , Dermatomiositis , Interferón Tipo I , Músculo Esquelético , Transducción de Señal , Humanos , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Interferón Tipo I/metabolismo , Animales , Músculo Esquelético/inmunología , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Ratones , Transducción de Señal/inmunología , Autoantígenos/inmunología , Femenino , Dermatomiositis/inmunología , Dermatomiositis/patología , Dermatomiositis/metabolismo , Masculino , Niño , Modelos Animales de Enfermedad , Adolescente , Preescolar
7.
J Med Virol ; 96(7): e29811, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39011825

RESUMEN

The recent outbreak of monkeypox virus (MPXV) was unprecedented in its size and distribution. Those living with uncontrolled HIV and low CD4 T cell counts might develop a fulminant clinical mpox course with increased mortality, secondary infections, and necrotizing lesions. Fatal cases display a high and widespread MPXV tissue burden. The underlying pathomechanisms are not fully understood. We report here the pathological findings of an MPXV-driven abscess in gastrocnemius muscle requiring surgery in an immunocompromised patient with severe mpox. Presence of virus particles and infectivity were confirmed by electron microscopy, expansion microscopy, and virus culture, respectively. MPXV tissue distribution by immunohistochemistry (IHC) showed a necrotic core with infection of different cell types. In contrast, at the lesion rim fibroblasts were mainly infected. Immune cells were almost absent in the necrotic core, but were abundant at the infection rim and predominantly macrophages. Further, we detected high amounts of alternatively activated GPNMB+-macrophages at the lesion border. Of note, macrophages only rarely colocalized with virus-infected cells. Insufficient clearance of infected cells and infection of lesion-associated fibroblasts sustained by the abundance of profibrotic macrophages might lead to the coalescing of lesions and the severe and persistent clinical mpox course observed in immunocompromised patients.


Asunto(s)
Huésped Inmunocomprometido , Monkeypox virus , Mpox , Músculo Esquelético , Humanos , Músculo Esquelético/virología , Músculo Esquelético/patología , Músculo Esquelético/inmunología , Mpox/virología , Mpox/inmunología , Monkeypox virus/inmunología , Masculino , Macrófagos/inmunología , Macrófagos/virología , Fibroblastos/virología , Fibroblastos/inmunología , Inmunohistoquímica , Absceso/inmunología , Absceso/virología , Absceso/patología , Persona de Mediana Edad
8.
Blood ; 138(4): 304-317, 2021 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-33786586

RESUMEN

Hematopoiesis is extrinsically controlled by cells of the bone marrow microenvironment, including skeletal lineage cells. The identification and subsequent studies of distinct subpopulations of maturing skeletal cells is currently limited because of a lack of methods to isolate these cells. We found that murine Lin-CD31-Sca-1-CD51+ cells can be divided into 4 subpopulations by using flow cytometry based on their expression of the platelet-derived growth factor receptors ⍺ and ß (PDGFR⍺ and PDGFRß). The use of different skeletal lineage reporters confirmed the skeletal origin of the 4 populations. Multiplex immunohistochemistry studies revealed that all 4 populations were localized near the growth plate and trabecular bone and were rarely found near cortical bone regions or in central bone marrow. Functional studies revealed differences in their abundance, colony-forming unit-fibroblast capacity, and potential to differentiate into mineralized osteoblasts or adipocytes in vitro. Furthermore, the 4 populations had distinct gene expression profiles and differential cell surface expression of leptin receptor (LEPR) and vascular cell adhesion molecule 1 (VCAM-1). Interestingly, we discovered that 1 of these 4 different skeletal populations showed the highest expression of genes involved in the extrinsic regulation of B lymphopoiesis. This cell population varied in abundance between distinct hematopoietically active skeletal sites, and significant differences in the proportions of B-lymphocyte precursors were also observed in these distinct skeletal sites. This cell population also supported pre-B lymphopoiesis in culture. Our method of isolating 4 distinct maturing skeletal populations will help elucidate the roles of distinct skeletal niche cells in regulating hematopoiesis and bone.


Asunto(s)
Linfocitos B/inmunología , Diferenciación Celular/inmunología , Linfopoyesis/inmunología , Músculo Esquelético/inmunología , Animales , Diferenciación Celular/genética , Linfopoyesis/genética , Ratones , Ratones Transgénicos
9.
Immunity ; 41(1): 36-48, 2014 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-25035952

RESUMEN

Chronic, low-grade adipose tissue inflammation is a key etiological mechanism linking the increasing incidence of type 2 diabetes (T2D) and obesity. It is well recognized that the immune system and metabolism are highly integrated, and macrophages, in particular, have been identified as critical effector cells in the initiation of inflammation and insulin resistance. Recent advances have been made in the understanding of macrophage recruitment and retention to adipose tissue and the participation of other immune cell populations in the regulation of this inflammatory process. Here we discuss the pathophysiological link between macrophages, obesity, and insulin resistance, highlighting the dynamic immune cell regulation of adipose tissue inflammation. We also describe the mechanisms by which inflammation causes insulin resistance and the new therapeutic targets that have emerged.


Asunto(s)
Tejido Adiposo/inmunología , Diabetes Mellitus Tipo 2/inmunología , Resistencia a la Insulina/inmunología , Macrófagos/inmunología , Obesidad/inmunología , Tejido Adiposo/citología , Animales , Citocinas/inmunología , Citocinas/metabolismo , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/fisiopatología , Humanos , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Hígado/citología , Hígado/inmunología , Activación de Macrófagos/inmunología , Ratones , Músculo Esquelético/citología , Músculo Esquelético/inmunología , Obesidad/tratamiento farmacológico , Obesidad/fisiopatología , Transducción de Señal/inmunología
10.
Proc Natl Acad Sci U S A ; 117(34): 20729-20740, 2020 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-32796104

RESUMEN

Tissue-resident macrophages can originate from embryonic or adult hematopoiesis. They play important roles in a wide range of biological processes including tissue remodeling during organogenesis, organ homeostasis, repair following injury, and immune response to pathogens. Although the origins and tissue-specific functions of resident macrophages have been extensively studied in many other tissues, they are not well characterized in skeletal muscle. In the present study, we have characterized the ontogeny of skeletal muscle-resident macrophages by lineage tracing and bone marrow transplant experiments. We demonstrate that skeletal muscle-resident macrophages originate from both embryonic hematopoietic progenitors located within the yolk sac and fetal liver as well as definitive hematopoietic stem cells located within the bone marrow of adult mice. Single-cell-based transcriptome analyses revealed that skeletal muscle-resident macrophages are distinctive from resident macrophages in other tissues as they express a distinct complement of transcription factors and are composed of functionally diverse subsets correlating to their origins. Functionally, skeletal muscle-resident macrophages appear to maintain tissue homeostasis and promote muscle growth and regeneration.


Asunto(s)
Macrófagos/inmunología , Músculo Esquelético/inmunología , Animales , Médula Ósea/metabolismo , Trasplante de Médula Ósea/métodos , Diferenciación Celular/genética , Linaje de la Célula/genética , Desarrollo Embrionario , Femenino , Heterogeneidad Genética , Hematopoyesis/genética , Células Madre Hematopoyéticas/metabolismo , Homeostasis , Macrófagos/metabolismo , Macrófagos/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/inmunología , Monocitos/metabolismo , Organogénesis/genética
11.
Proc Natl Acad Sci U S A ; 117(25): 14354-14364, 2020 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-32513697

RESUMEN

Ischemic injury initiates a sterile inflammatory response that ultimately participates in the repair and recovery of tissue perfusion. Macrophages are required for perfusion recovery during ischemia, in part because they produce growth factors that aid in vascular remodeling. The input signals governing this pro-revascularization phenotype remain of interest. Here we found that hindlimb ischemia increases levels of resolvin D1 (RvD1), an inflammation-resolving lipid mediator that targets macrophages via its receptor, ALX/FPR2. Exogenous RvD1 enhances perfusion recovery during ischemia, and mice deficient in Alx/Fpr2 have an endogenous defect in this process. Mechanistically, RNA sequencing revealed that RvD1 induces a transcriptional program in macrophages characteristic of a pro-revascularization phenotype. Vascularization of ischemic skeletal muscle, as well as cutaneous wounds, is impaired in mice with myeloid-specific deficiency of Alx/Fpr2, and this is associated with altered expression of pro-revascularization genes in skeletal muscle and macrophages isolated from skeletal muscle. Collectively, these results uncover a role of ALX/FPR2 in revascularization that may be amenable to therapeutic targeting in diseases associated with altered tissue perfusion and repair.


Asunto(s)
Ácidos Docosahexaenoicos/metabolismo , Isquemia/inmunología , Neovascularización Fisiológica/inmunología , Receptores de Formil Péptido/metabolismo , Receptores de Lipoxina/metabolismo , Cicatrización de Heridas/inmunología , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Técnicas de Inactivación de Genes , Humanos , Isquemia/patología , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Noqueados , Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/inmunología , Músculo Esquelético/patología , Cultivo Primario de Células , RNA-Seq , Receptores de Formil Péptido/genética , Receptores de Lipoxina/genética , Transducción de Señal/inmunología , Piel/irrigación sanguínea , Piel/inmunología , Piel/lesiones , Piel/patología , Transcripción Genética/inmunología
12.
Hum Mol Genet ; 29(3): 353-368, 2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31696230

RESUMEN

Duchenne muscular dystrophy (DMD) is a lethal muscle wasting disorder caused by mutations in the DMD gene that leads to the absence or severe reduction of dystrophin protein in muscle. The mdx mouse, also dystrophin deficient, is the model most widely used to study the pathology and test potential therapies, but the phenotype is milder than human DMD. This limits the magnitude and range of histological damage parameters and molecular changes that can be measured in pre-clinical drug testing. We used 3 weeks of voluntary wheel running to exacerbate the mdx phenotype. In mdx mice, voluntary exercise increased the amount of damaged necrotic tissue and macrophage infiltration. Global gene expression profiling revealed that exercise induced additional and larger gene expression changes in mdx mice and the pathways most impacted by exercise were all related to immune function or cell-extracellular matrix (ECM) interactions. When we compared the matrisome and inflammation genes that were dysregulated in mdx with those commonly differentially expressed in DMD, we found the exercised mdx molecular signature more closely resembled that of DMD. These gene expression changes in the exercised mdx model thus provide more scope to assess the effects of pre-clinical treatments. Our gene profiling comparisons also highlighted upregulation of ECM proteins involved in innate immunity pathways, proteases that can release them, downstream receptors and signaling molecules in exercised mdx and DMD, suggesting that the ECM could be a major source of pro-inflammatory molecules that trigger and maintain the immune response in dystrophic muscle.


Asunto(s)
Proteínas de la Matriz Extracelular/metabolismo , Regulación de la Expresión Génica , Inmunidad/inmunología , Inflamación/patología , Actividad Motora , Músculo Esquelético/patología , Distrofia Muscular de Duchenne/patología , Animales , Proteínas de la Matriz Extracelular/genética , Perfilación de la Expresión Génica , Humanos , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Masculino , Ratones , Ratones Endogámicos mdx , Músculo Esquelético/inmunología , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/inmunología , Distrofia Muscular de Duchenne/metabolismo
13.
EMBO J ; 37(10)2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29632021

RESUMEN

Opa1 participates in inner mitochondrial membrane fusion and cristae morphogenesis. Here, we show that muscle-specific Opa1 ablation causes reduced muscle fiber size, dysfunctional mitochondria, enhanced Fgf21, and muscle inflammation characterized by NF-κB activation, and enhanced expression of pro-inflammatory genes. Chronic sodium salicylate treatment ameliorated muscle alterations and reduced the muscle expression of Fgf21. Muscle inflammation was an early event during the progression of the disease and occurred before macrophage infiltration, indicating that it is a primary response to Opa1 deficiency. Moreover, Opa1 repression in muscle cells also resulted in NF-κB activation and inflammation in the absence of necrosis and/or apoptosis, thereby revealing that the activation is a cell-autonomous process and independent of cell death. The effects of Opa1 deficiency on the expression NF-κB target genes and inflammation were absent upon mitochondrial DNA depletion. Under Opa1 deficiency, blockage or repression of TLR9 prevented NF-κB activation and inflammation. Taken together, our results reveal that Opa1 deficiency in muscle causes initial mitochondrial alterations that lead to TLR9 activation, and inflammation, which contributes to enhanced Fgf21 expression and to growth impairment.


Asunto(s)
ADN Mitocondrial/genética , GTP Fosfohidrolasas/fisiología , Inflamación/etiología , Músculo Esquelético/patología , Enfermedades Musculares/etiología , Receptor Toll-Like 9/metabolismo , Animales , Apoptosis , Células Cultivadas , Citocinas/metabolismo , Femenino , Inflamación/metabolismo , Inflamación/patología , Masculino , Ratones Noqueados , Músculo Esquelético/inmunología , Enfermedades Musculares/metabolismo , Enfermedades Musculares/patología , Necrosis , Regeneración , Receptor Toll-Like 9/genética
14.
Am J Physiol Regul Integr Comp Physiol ; 322(1): R41-R54, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34786967

RESUMEN

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.


Asunto(s)
Proliferación Celular , Ejercicio Físico , Hipertrofia/metabolismo , Mediadores de Inflamación/metabolismo , Inflamación/metabolismo , Macrófagos/metabolismo , Músculo Esquelético/metabolismo , Regeneración , Crecimiento del Músculo Esquelético , Animales , Humanos , Hipertrofia/inmunología , Hipertrofia/patología , Hipertrofia/fisiopatología , Inflamación/inmunología , Inflamación/patología , Inflamación/fisiopatología , Macrófagos/inmunología , Músculo Esquelético/inmunología , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Fenotipo , Transducción de Señal
15.
J Immunol ; 204(8): 2203-2215, 2020 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-32161098

RESUMEN

Myeloid cells are critical to the development of fibrosis following muscle injury; however, the mechanism of their role in fibrosis formation remains unclear. In this study, we demonstrate that myeloid cell-derived TGF-ß1 signaling is increased in a profibrotic ischemia reperfusion and cardiotoxin muscle injury model. We found that myeloid-specific deletion of Tgfb1 abrogates the fibrotic response in this injury model and reduces fibro/adipogenic progenitor cell proliferation while simultaneously enhancing muscle regeneration, which is abrogated by adaptive transfer of normal macrophages. Similarly, a murine TGFBRII-Fc ligand trap administered after injury significantly reduced muscle fibrosis and improved muscle regeneration. This study ultimately demonstrates that infiltrating myeloid cell TGF-ß1 is responsible for the development of traumatic muscle fibrosis, and its blockade offers a promising therapeutic target for preventing muscle fibrosis after ischemic injury.


Asunto(s)
Fibrosis/inmunología , Fibrosis/patología , Macrófagos/inmunología , Músculo Esquelético/inmunología , Músculo Esquelético/patología , Células Mieloides/inmunología , Factor de Crecimiento Transformador beta1/inmunología , Animales , Cardiotoxinas , Fibrosis/complicaciones , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Mieloides/patología , Fenotipo , Daño por Reperfusión/inducido químicamente , Daño por Reperfusión/complicaciones , Daño por Reperfusión/inmunología
16.
Mar Drugs ; 20(5)2022 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-35621964

RESUMEN

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.


Asunto(s)
Dipéptidos , Macrófagos , Músculo Esquelético , Fagocitosis , Animales , Citocinas/metabolismo , Dipéptidos/metabolismo , Dipéptidos/farmacología , Imidazoles/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/fisiología , Ratones , Músculo Esquelético/inmunología , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiología , Fagocitosis/efectos de los fármacos
17.
Int J Mol Sci ; 23(3)2022 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-35163799

RESUMEN

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.


Asunto(s)
Entrenamiento Aeróbico/veterinaria , Inflamación/genética , Lipopolisacáridos/efectos adversos , Músculo Esquelético/inmunología , Receptor Toll-Like 4/genética , Adaptación Fisiológica , Animales , Citocinas/metabolismo , Transportador de Glucosa de Tipo 4/metabolismo , Inflamación/inducido químicamente , Inflamación/metabolismo , Masculino , Ratones , Mitocondrias Musculares/metabolismo , Músculo Esquelético/efectos de los fármacos , Mutación , Biogénesis de Organelos , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Condicionamiento Físico Animal , Regulación hacia Arriba
18.
Lab Invest ; 101(6): 719-732, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33674785

RESUMEN

Immune cells are involved in skeletal muscle regeneration. The mechanism by which Treg cells are involved in the regeneration of injured skeletal muscle is still unclear. The purpose of this study was to explore the role of programmed death-1 in contused skeletal muscle regeneration, and to clarify the regulation of programmed death-1 on Treg cell generation and macrophage polarization, in order to deepen our understanding of the relationship between the immune system and injured skeletal muscle regeneration. The results show that programmed death-1 knockdown reduced the number of Treg cells and impaired contused skeletal muscle regeneration compared with those of wild-type mice. The number of pro-inflammatory macrophages in the contused skeletal muscle of programmed death-1 knockout mice increased, and the expression of pro-inflammatory factors and oxidative stress factors increased, while the number of anti-inflammatory macrophages and the expression of anti-inflammatory factors, antioxidant stress factors, and muscle regeneration-related factors decreased. These results suggest that programmed death-1 can promote contused skeletal muscle regeneration by regulating Treg cell generation and macrophage polarization.


Asunto(s)
Contusiones/inmunología , Músculo Esquelético/inmunología , Receptor de Muerte Celular Programada 1/metabolismo , Regeneración , Animales , Macrófagos/fisiología , Masculino , Ratones Noqueados , Estrés Oxidativo , Linfocitos T Reguladores/fisiología
19.
Hum Mol Genet ; 28(12): 2030-2045, 2019 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-30759207

RESUMEN

Mineralocorticoid receptor (MR) drugs have been used clinically for decades to treat cardiovascular diseases. MR antagonists not only show preclinical efficacy for heart in Duchenne muscular dystrophy (DMD) models but also improve skeletal muscle force and muscle membrane integrity. The mechanisms of action of MR antagonists in skeletal muscles are entirely unknown. Since MR are present in many cell types in the muscle microenvironment, it is critical to define cell-intrinsic functions in each cell type to ultimately optimize antagonist efficacy for use in the widest variety of diseases. We generated a new conditional knockout of MR in myofibers and quantified cell-intrinsic mechanistic effects on functional and histological parameters in a DMD mouse model. Skeletal muscle MR deficiency led to improved respiratory muscle force generation and less deleterious fibrosis but did not reproduce MR antagonist efficacy on membrane susceptibility to induced damage. Surprisingly, acute application of MR antagonist to muscles led to improvements in membrane integrity after injury independent of myofiber MR. These data demonstrate that MR antagonists are efficacious to dystrophic skeletal muscles through both myofiber intrinsic effects on muscle force and downstream fibrosis and extrinsic functions on membrane stability. MR antagonists may therefore be applicable for treating more general muscle weakness and possibly other conditions that result from cell injuries.


Asunto(s)
Antagonistas de Receptores de Mineralocorticoides/uso terapéutico , Músculo Esquelético/patología , Distrofia Muscular de Duchenne/patología , Animales , Membrana Celular/efectos de los fármacos , Membrana Celular/fisiología , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Fuerza Muscular/efectos de los fármacos , Músculo Esquelético/inmunología , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiopatología , Distrofia Muscular de Duchenne/tratamiento farmacológico , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/fisiopatología , Receptores de Mineralocorticoides/genética , Receptores de Mineralocorticoides/metabolismo , Espironolactona/uso terapéutico
20.
Am J Pathol ; 190(1): 190-205, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31726040

RESUMEN

Duchenne muscular dystrophy (DMD) causes severe disability and death of young men because of progressive muscle degeneration aggravated by sterile inflammation. DMD is also associated with cognitive and bone-function impairments. This complex phenotype results from the cumulative loss of a spectrum of dystrophin isoforms expressed from the largest human gene. Although there is evidence for the loss of shorter isoforms having impact in the central nervous system, their role in muscle is unclear. We found that at 8 weeks, the active phase of pathology in dystrophic mice, dystrophin-null mice (mdxßgeo) presented with a mildly exacerbated phenotype but without an earlier onset, increased serum creatine kinase levels, or decreased muscle strength. However, at 12 months, mdxßgeo diaphragm strength was lower, whereas fibrosis increased, compared with mdx. The most striking features of the dystrophin-null phenotype were increased ectopic myofiber calcification and altered macrophage infiltration patterns, particularly the close association of macrophages with calcified fibers. Ectopic calcification had the same temporal pattern of presentation and resolution in mdxßgeo and mdx muscles, despite significant intensity differences across muscle groups. Comparison of the rare dystrophin-null patients against those with mutations affecting full-length dystrophins may provide mechanistic insights for developing more effective treatments for DMD.


Asunto(s)
Calcinosis/patología , Distrofina/metabolismo , Fibrosis/patología , Macrófagos/inmunología , Distrofia Muscular Animal/patología , Distrofia Muscular de Duchenne/patología , Calcificación Vascular/patología , Animales , Calcinosis/inmunología , Calcinosis/metabolismo , Distrofina/genética , Fibrosis/inmunología , Fibrosis/metabolismo , Inflamación , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos mdx , Músculo Esquelético/inmunología , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Distrofia Muscular Animal/inmunología , Distrofia Muscular Animal/metabolismo , Distrofia Muscular de Duchenne/inmunología , Distrofia Muscular de Duchenne/metabolismo , Calcificación Vascular/inmunología , Calcificación Vascular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA