Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Hum Mol Genet ; 31(2): 189-206, 2021 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-34392367

RESUMEN

Leukemia inhibitory factor (LIF) can influence development by increasing cell proliferation and inhibiting differentiation. Because of its potency for expanding stem cell populations, delivery of exogenous LIF to diseased tissue could have therapeutic value. However, systemic elevations of LIF can have negative, off-target effects. We tested whether inflammatory cells expressing a LIF transgene under control of a leukocyte-specific, CD11b promoter provide a strategy to target LIF to sites of damage in the mdx mouse model of Duchenne muscular dystrophy, leading to increased numbers of muscle stem cells and improved muscle regeneration. However, transgene expression in inflammatory cells did not increase muscle growth or increase numbers of stem cells required for regeneration. Instead, transgene expression disrupted the normal dispersion of macrophages in dystrophic muscles, leading to transient increases in muscle damage in foci where macrophages were highly concentrated during early stages of pathology. The defect in inflammatory cell dispersion reflected impaired chemotaxis of macrophages to C-C motif chemokine ligand-2 and local increases of LIF production that produced large aggregations of cytolytic macrophages. Transgene expression also induced a shift in macrophage phenotype away from a CD206+, M2-biased phenotype that supports regeneration. However, at later stages of the disease when macrophage numbers declined, they dispersed in the muscle, leading to reductions in muscle fiber damage, compared to non-transgenic mdx mice. Together, the findings show that macrophage-mediated delivery of transgenic LIF exerts differential effects on macrophage dispersion and muscle damage depending on the stage of dystrophic pathology.


Asunto(s)
Distrofia Muscular Animal , Distrofia Muscular de Duchenne , Animales , Humanos , Factor Inhibidor de Leucemia/genética , Factor Inhibidor de Leucemia/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Endogámicos mdx , Ratones Transgénicos , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Distrofia Muscular Animal/genética , Distrofia Muscular de Duchenne/metabolismo
2.
FASEB J ; 36(3): e22192, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35174906

RESUMEN

Modulating the number of muscle stems cells, called satellite cells, during early postnatal development produces long-term effects on muscle growth. We tested the hypothesis that high expression levels of the anti-aging protein Klotho in early postnatal myogenesis increase satellite cell numbers by influencing the epigenetic regulation of genes that regulate myogenesis. Our findings show that elevated klotho expression caused a transient increase in satellite cell numbers and slowed muscle fiber growth, followed by a period of accelerated muscle growth that leads to larger fibers. Klotho also transcriptionally downregulated the H3K27 demethylase Jmjd3, leading to increased H3K27 methylation and decreased expression of genes in the canonical Wnt pathway, which was associated with a delay in muscle differentiation. In addition, Klotho stimulation and Jmjd3 downregulation produced similar but not additive reductions in the expression of Wnt4, Wnt9a, and Wnt10a in myogenic cells, indicating that inhibition occurred through a common pathway. Together, our results identify a novel pathway through which Klotho influences myogenesis by reducing the expression of Jmjd3, leading to reductions in the expression of Wnt genes and inhibition of canonical Wnt signaling.


Asunto(s)
Histona Demetilasas con Dominio de Jumonji/genética , Proteínas Klotho/metabolismo , Desarrollo de Músculos , Mioblastos/metabolismo , Animales , Línea Celular , Regulación hacia Abajo , Regulación del Desarrollo de la Expresión Génica , Histona Demetilasas con Dominio de Jumonji/metabolismo , Proteínas Klotho/genética , Ratones , Ratones Endogámicos C57BL , Mioblastos/citología , Vía de Señalización Wnt
3.
Exp Physiol ; 108(12): 1531-1547, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37864311

RESUMEN

NEW FINDINGS: What is the central question of this study? Does the hormone Klotho affect the myogenic response of muscle cells to mechanical loading or exercise? What is the main finding and its importance? Klotho prevents direct, mechanical activation of genes that regulate muscle differentiation, including genes that encode the myogenic regulatory factor myogenin and proteins in the canonical Wnt signalling pathway. Similarly, elevated levels of klotho expression in vivo prevent the exercise-induced increase in myogenin-expressing cells and reduce exercise-induced activation of the Wnt pathway. These findings demonstrate a new mechanism through which the responses of muscle to the mechanical environment are regulated. ABSTRACT: Muscle growth is influenced by changes in the mechanical environment that affect the expression of genes that regulate myogenesis. We tested whether the hormone Klotho could influence the response of muscle to mechanical loading. Applying mechanical loads to myoblasts in vitro increased RNA encoding transcription factors that are expressed in activated myoblasts (Myod) and in myogenic cells that have initiated terminal differentiation (Myog). However, application of Klotho to myoblasts prevented the loading-induced activation of Myog without affecting loading-induced activation of Myod. This indicates that elevated Klotho inhibits mechanically-induced differentiation of myogenic cells. Elevated Klotho also reduced the transcription of genes encoding proteins involved in the canonical Wnt pathway or their target genes (Wnt9a, Wnt10a, Ccnd1). Because the canonical Wnt pathway promotes differentiation of myogenic cells, these findings indicate that Klotho inhibits the differentiation of myogenic cells experiencing mechanical loading. We then tested whether these effects of Klotho occurred in muscles of mice experiencing high-intensity interval training (HIIT) by comparing wild-type mice and klotho transgenic mice. The expression of a klotho transgene combined with HIIT synergized to tremendously elevate numbers of Pax7+ satellite cells and activated MyoD+ cells. However, transgene expression prevented the increase in myogenin+ cells caused by HIIT in wild-type mice. Furthermore, transgene expression diminished the HIIT-induced activation of the canonical Wnt pathway in Pax7+ satellite cells. Collectively, these findings show that Klotho inhibits loading- or exercise-induced activation of muscle differentiation and indicate a new mechanism through which the responses of muscle to the mechanical environment are regulated.


Asunto(s)
Músculos , Células Satélite del Músculo Esquelético , Animales , Ratones , Diferenciación Celular , Hormonas/metabolismo , Desarrollo de Músculos/genética , Músculo Esquelético/metabolismo , Músculos/metabolismo , Proteína MioD/metabolismo , Miogenina/metabolismo , Células Satélite del Músculo Esquelético/metabolismo
4.
J Immunol ; 205(6): 1664-1677, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32817369

RESUMEN

Changes in macrophage phenotype in injured muscle profoundly influence regeneration. In particular, the shift of macrophages from a proinflammatory (M1 biased) phenotype to a proregenerative (M2 biased) phenotype characterized by expression of CD206 and CD163 is essential for normal repair. According to the current canonical mechanism regulating for M1/M2 phenotype transition, signaling through PPARδ is necessary for obtaining the M2-biased phenotype. Our findings confirm that the murine myeloid cell-targeted deletion of Ppard reduces expression in vitro of genes that are activated in M2-biased macrophages; however, the mutation in mice in vivo increased numbers of CD206+ M2-biased macrophages and did not reduce the expression of phenotypic markers of M2-biased macrophages in regenerating muscle. Nevertheless, the mutation impaired CCL2-mediated chemotaxis of macrophages and slowed revascularization of injured muscle. In contrast, null mutation of IL-10 diminished M2-biased macrophages but produced no defects in muscle revascularization. Our results provide two significant findings. First, they illustrate that mechanisms that regulate macrophage phenotype transitions in vitro are not always predictive of mechanisms that are most important in vivo. Second, they show that mechanisms that regulate macrophage phenotype transitions differ in different in vivo environments.


Asunto(s)
Interleucina-10/metabolismo , Macrófagos/fisiología , Músculo Esquelético/fisiología , Enfermedades Musculares/metabolismo , Células Mieloides/fisiología , PPAR delta/metabolismo , Enfermedad Aguda , Animales , Diferenciación Celular , Movimiento Celular , Células Cultivadas , Citocinas/metabolismo , Humanos , Interleucina-10/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Enfermedades Musculares/genética , Enfermedades Musculares/inmunología , PPAR delta/genética , Fenotipo , Regeneración , Células TH1/inmunología , Células Th2/inmunología
5.
Hum Mol Genet ; 27(1): 14-29, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29040534

RESUMEN

Duchenne muscular dystrophy (DMD) is a muscle wasting disease in which inflammation influences the severity of pathology. We found that the onset of muscle inflammation in the mdx mouse model of DMD coincides with large increases in expression of pro-inflammatory cytokines [tumor necrosis factor-α (TNFα); interferon gamma (IFNγ)] and dramatic reductions of the pro-myogenic protein Klotho in muscle cells and large increases of Klotho in pro-regenerative, CD206+ macrophages. Furthermore, TNFα and IFNγ treatments reduced Klotho in muscle cells and increased Klotho in macrophages. Because CD206+/Klotho+ macrophages were concentrated at sites of muscle regeneration, we tested whether macrophage-derived Klotho promotes myogenesis. Klotho transgenic macrophages had a pro-proliferative influence on muscle cells that was ablated by neutralizing antibodies to Klotho and conditioned media from Klotho mutant macrophages did not increase muscle cell proliferation in vitro. In addition, transplantation of bone marrow cells from Klotho transgenic mice into mdx recipients increased numbers of myogenic cells and increased the size of muscle fibers. Klotho also acted directly on macrophages, stimulating their secretion of TNFα. Because TNFα is a muscle mitogen, we tested whether the pro-proliferative effects of Klotho on muscle cells were mediated by TNFα and found that increased proliferation caused by Klotho was reduced by anti-TNFα. Collectively, these data show that pro-inflammatory cytokines contribute to silencing of Klotho in dystrophic muscle, but increase Klotho expression by macrophages. Our findings also show that macrophage-derived Klotho can promote muscle regeneration by expanding populations of muscle stem cells and increasing muscle fiber growth in dystrophic muscle.


Asunto(s)
Glucuronidasa/fisiología , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/metabolismo , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Silenciador del Gen , Humanos , Inflamación/genética , Proteínas Klotho , Macrófagos/metabolismo , Ratones , Ratones Endogámicos mdx , Músculo Esquelético/metabolismo , Distrofia Muscular Animal/genética , Distrofia Muscular de Duchenne/fisiopatología , Mioblastos/metabolismo
6.
FASEB J ; 33(1): 1415-1427, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30130434

RESUMEN

Aging is associated with diminished muscle mass, reductions in muscle stem cell functions, and increased muscle fibrosis. The immune system, especially macrophages, can have important roles in modulating muscle growth and regeneration, suggesting that the immune system may also have significant influences on muscle aging. Moreover, the immune system experiences changes in function during senescence, suggesting that regulatory interaction between muscle cells and the immune system may also change during aging. In this study, we performed bone marrow transplantations between age-mismatched donor and recipient mice to test the influence of the age of the immune system on muscle aging. Transplantation of young bone marrow cells into old recipients prevented sarcopenia and prevented age-related change in muscle fiber phenotype. Transplantation of old bone marrow cells into young animals reduced satellite cell numbers and promoted satellite cells to switch toward a fibrogenic phenotype. We also demonstrated that conditioned media from young, but not old, bone marrow cells promoted myoblast proliferation in vitro, and we found that factors released by young bone marrow cells were more supportive of myotube differentiation in vitro. Together, our results demonstrate that aging of bone marrow cells promotes the age-related reduction of satellite cell number and function and contributes to sarcopenia.-Wang, Y., Wehling-Henricks, M., Welc, S. S., Fisher, A. L., Zuo, Q., Tidball, J. G. Aging of the immune system causes reductions in muscle stem cell populations, promotes their shift to a fibrogenic phenotype, and modulates sarcopenia.


Asunto(s)
Envejecimiento/fisiología , Sistema Inmunológico/fisiología , Sarcopenia/patología , Células Satélite del Músculo Esquelético/patología , Animales , Biomarcadores/metabolismo , Células de la Médula Ósea/patología , Diferenciación Celular , Proliferación Celular , Senescencia Celular/fisiología , Femenino , Fibrosis , Trasplante de Células Madre Hematopoyéticas , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Fibras Musculares Esqueléticas/fisiología
7.
Exp Physiol ; 105(1): 132-147, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31724771

RESUMEN

NEW FINDINGS: What is the central question of this study? Does modulating the expression of Klotho affect myogenesis following acute injury of healthy, non-senescent muscle? What is the main finding and its importance? Klotho can accelerate muscle growth following acute injury of healthy, adult mice, which supports the possibility that increased delivery of Klotho could have therapeutic value for improving repair of damaged muscle. ABSTRACT: Skeletal muscle injuries activate a complex programme of myogenesis that can restore normal muscle structure. We tested whether modulating the expression of klotho influenced the response of mouse muscles to acute injury. Our findings show that klotho expression in muscle declines at 3 days post-injury. That reduction in klotho expression coincided with elevated expression of targets of Wnt signalling (Ccnd1; Myc) and increased MyoD+ muscle cell numbers, reflecting the onset of myogenic cell differentiation. klotho expression subsequently increased at 7 days post-injury with elevated expression occurring primarily in inflammatory lesions, which was accompanied by reduced expression of Wnt target genes (Ccnd1: 91%; Myc: 96%). Introduction of a klotho transgene maintained high levels of klotho expression over the course of muscle repair and attenuated the increases in Ccnd1 and Myc expression that occurred at 3 days post-injury. Correspondingly, transgene expression reduced Wnt signalling in Pax7+ cells, reflected by reductions in Pax7+ cells expressing active ß-catenin, and reduced the numbers of MyoD+ cells at 3 days post-injury. At 21 days post-injury, muscles in klotho transgenic mice showed increased Pax7+ and decreased myogenin+ cell densities and large increases in myofibre size. Likewise, treating myogenic cells in vitro with Klotho reduced Myod expression but did not affect Pax7 expression. Muscle inflammation was only slightly modulated by increased klotho expression, initially reducing the expression of M2-biased macrophage markers Cd163 and Cd206 at 3 days post-injury and later increasing the expression of pan-macrophage marker F480 and Cd68 at 21 days post-injury. Collectively, our study shows that Klotho modulates myogenesis and that increased expression accelerates muscle growth after injury.


Asunto(s)
Glucuronidasa/metabolismo , Desarrollo de Músculos , Músculo Esquelético/lesiones , Músculo Esquelético/metabolismo , Vía de Señalización Wnt , Animales , Células Cultivadas , Ciclina D1/metabolismo , Proteínas Klotho , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fibras Musculares Esqueléticas/citología , Mioblastos/citología , Proteínas Proto-Oncogénicas c-myc/metabolismo
8.
Hum Mol Genet ; 25(12): 2465-2482, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27154199

RESUMEN

Duchenne muscular dystrophy (DMD) is a lethal muscle disease involving progressive loss of muscle regenerative capacity and increased fibrosis. We tested whether epigenetic silencing of the klotho gene occurs in the mdx mouse model of DMD and whether klotho silencing is an important feature of the disease. Our findings show that klotho undergoes muscle-specific silencing at the acute onset of mdx pathology. Klotho experiences increased methylation of CpG sites in its promoter region, which is associated with gene silencing, and increases in a repressive histone mark, H3K9me2. Expression of a klotho transgene in mdx mice restored their longevity, reduced muscle wasting, improved function and greatly increased the pool of muscle-resident stem cells required for regeneration. Reductions of fibrosis in late, progressive stages of the mdx pathology achieved by transgene expression were paralleled by reduced expression of Wnt target genes (axin-2), transforming growth factor-beta (TGF-ß1) and collagens types 1 and 3, indicating that Klotho inhibition of the profibrotic Wnt/TGFß axis underlies its anti-fibrotic effect in aging, dystrophic muscle. Thus, epigenetic silencing of klotho during muscular dystrophy contributes substantially to lost regenerative capacity and increased fibrosis of dystrophic muscle during late progressive stages of the disease.


Asunto(s)
Fibrosis/genética , Glucuronidasa/genética , Distrofia Muscular Animal/genética , Distrofia Muscular de Duchenne/genética , Animales , Proteína Axina/biosíntesis , Colágeno Tipo I/biosíntesis , Colágeno Tipo III/biosíntesis , Modelos Animales de Enfermedad , Fibrosis/patología , Regulación de la Expresión Génica , Silenciador del Gen , Glucuronidasa/antagonistas & inhibidores , Humanos , Proteínas Klotho , Ratones , Ratones Endogámicos mdx , Músculo Esquelético/patología , Distrofia Muscular Animal/patología , Distrofia Muscular de Duchenne/patología , Regeneración/genética , Factor de Crecimiento Transformador beta1/biosíntesis
9.
Hum Mol Genet ; 25(23): 5167-5177, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27798095

RESUMEN

FDA-approved mineralocorticoid receptor (MR) antagonists are used to treat heart failure. We have recently demonstrated efficacy of MR antagonists for skeletal muscles in addition to heart in Duchenne muscular dystrophy mouse models and that mineralocorticoid receptors are present and functional in skeletal muscles. The goal of this study was to elucidate the underlying mechanisms of MR antagonist efficacy on dystrophic skeletal muscles. We demonstrate for the first time that infiltrating myeloid cells clustered in damaged areas of dystrophic skeletal muscles have the capacity to produce the natural ligand of MR, aldosterone, which in excess is known to exacerbate tissue damage. Aldosterone synthase protein levels are increased in leukocytes isolated from dystrophic muscles compared with controls and local aldosterone levels in dystrophic skeletal muscles are increased, despite normal circulating levels. All genes encoding enzymes in the pathway for aldosterone synthesis are expressed in muscle-derived leukocytes. 11ß-HSD2, the enzyme that inactivates glucocorticoids to increase MR selectivity for aldosterone, is also increased in dystrophic muscle tissues. These results, together with the demonstrated preclinical efficacy of antagonists, suggest MR activation is in excess of physiological need and likely contributes to the pathology of muscular dystrophy. This study provides new mechanistic insight into the known contribution of myeloid cells to muscular dystrophy pathology. This first report of myeloid cells having the capacity to produce aldosterone may have implications for a wide variety of acute injuries and chronic diseases with inflammation where MR antagonists may be therapeutic.


Asunto(s)
Insuficiencia Cardíaca/tratamiento farmacológico , Antagonistas de Receptores de Mineralocorticoides/administración & dosificación , Músculo Esquelético/efectos de los fármacos , Distrofia Muscular de Duchenne/tratamiento farmacológico , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/biosíntesis , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/genética , Aldosterona/metabolismo , Animales , Citocromo P-450 CYP11B2/biosíntesis , Citocromo P-450 CYP11B2/genética , Modelos Animales de Enfermedad , Corazón/efectos de los fármacos , Corazón/fisiopatología , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/patología , Humanos , Ratones , Músculo Esquelético/patología , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patología , Células Mieloides/efectos de los fármacos , Células Mieloides/patología
10.
Development ; 141(6): 1184-96, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24595286

RESUMEN

Much of the focus in muscle regeneration has been placed on the identification and delivery of stem cells to promote regenerative capacity. As those efforts have advanced, we have learned that complex features of the microenvironment in which regeneration occurs can determine success or failure. The immune system is an important contributor to that complexity and can determine the extent to which muscle regeneration succeeds. Immune cells of the myeloid lineage play major regulatory roles in tissue regeneration through two general, inductive mechanisms: instructive mechanisms that act directly on muscle cells; and permissive mechanisms that act indirectly to influence regeneration by modulating angiogenesis and fibrosis. In this article, recent discoveries that identify inductive actions of specific populations of myeloid cells on muscle regeneration are presented, with an emphasis on how processes in muscle and myeloid cells are co-regulated.


Asunto(s)
Músculo Esquelético/fisiología , Células Mieloides/fisiología , Regeneración/fisiología , Animales , Linaje de la Célula , Citocinas/fisiología , Fibrosis , Humanos , Inflamación/patología , Inflamación/fisiopatología , Macrófagos/clasificación , Macrófagos/fisiología , Modelos Biológicos , Desarrollo de Músculos/fisiología , Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/lesiones , Células Mieloides/clasificación , Transducción de Señal
11.
J Physiol ; 592(21): 4627-38, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-25194047

RESUMEN

The secondary loss of neuronal nitric oxide synthase (nNOS) that occurs in dystrophic muscle is the basis of numerous, complex and interacting features of the dystrophic pathology that affect not only muscle itself, but also influence the interaction of muscle with other tissues. Many mechanisms through which nNOS deficiency contributes to misregulation of muscle development, blood flow, fatigue, inflammation and fibrosis in dystrophic muscle have been identified, suggesting that normalization in NO production could greatly attenuate diverse aspects of the pathology of muscular dystrophy through multiple regulatory pathways. However, the relative importance of the loss of nNOS from the sarcolemma versus the importance of loss of total nNOS from dystrophic muscle remains unknown. Although most current evidence indicates that nNOS localization at the sarcolemma is not required to achieve NO-mediated reductions of pathology in muscular dystrophy, the question remains open concerning whether membrane localization would provide a more efficient rescue from features of the dystrophic phenotype.


Asunto(s)
Distrofia Muscular de Duchenne/enzimología , Distrofia Muscular de Duchenne/fisiopatología , Óxido Nítrico Sintasa de Tipo I/deficiencia , Animales , Distrofina/deficiencia , Distrofina/genética , Distrofina/metabolismo , Humanos , Óxido Nítrico/metabolismo , Sarcolema/enzimología
12.
J Immunol ; 189(7): 3669-80, 2012 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-22933625

RESUMEN

We examined the function of IL-10 in regulating changes in macrophage phenotype during muscle growth and regeneration following injury. Our findings showed that the Th1 cytokine response in inflamed muscle is characterized by high levels of expression of CD68, CCL-2, TNF-α, and IL-6 at 1 d postinjury. During transition to the Th2 cytokine response, expression of those transcripts declined, whereas CD163, IL-10, IL-10R1, and arginase-1 increased. Ablation of IL-10 amplified the Th1 response at 1 d postinjury, causing increases in IL-6 and CCL2, while preventing a subsequent increase in CD163 and arginase-1. Reductions in muscle fiber damage that normally occurred between 1 and 4 d postinjury did not occur in IL-10 mutants. In addition, muscle regeneration and growth were greatly slowed by loss of IL-10. Furthermore, myogenin expression increased in IL-10 mutant muscle at 1 d postinjury, suggesting that the mutation amplified the transition from the proliferative to the early differentiation stages of myogenesis. In vitro assays showed that stimulation of muscle cells with IL-10 had no effect on cell proliferation or expression of MyoD or myogenin. However, coculturing muscle cells with macrophages activated with IL-10 to the M2 phenotype increased myoblast proliferation without affecting MyoD or myogenin expression, showing that M2 macrophages promote the early, proliferative stage of myogenesis. Collectively, these data show that IL-10 plays a central role in regulating the switch of muscle macrophages from a M1 to M2 phenotype in injured muscle in vivo, and this transition is necessary for normal growth and regeneration of muscle.


Asunto(s)
Inmunofenotipificación , Interleucina-10/fisiología , Macrófagos/clasificación , Macrófagos/inmunología , Músculo Esquelético/inmunología , Regeneración/inmunología , Animales , Aumento de la Célula , Proliferación Celular , Citocinas/biosíntesis , Femenino , Interleucina-10/deficiencia , Interleucina-10/genética , Macrófagos/patología , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Esquelético/crecimiento & desarrollo , Músculo Esquelético/lesiones , Regeneración/genética , Células TH1/inmunología , Células TH1/metabolismo , Células Th2/inmunología , Células Th2/metabolismo , Regulación hacia Arriba/inmunología
13.
Hum Mol Genet ; 20(4): 790-805, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21118895

RESUMEN

M1 macrophages play a major role in worsening muscle injury in the mdx mouse model of Duchenne muscular dystrophy. However, mdx muscle also contains M2c macrophages that can promote tissue repair, indicating that factors regulating the balance between M1 and M2c phenotypes could influence the severity of the disease. Because interleukin-10 (IL-10) modulates macrophage activation in vitro and its expression is elevated in mdx muscles, we tested whether IL-10 influenced the macrophage phenotype in mdx muscle and whether changes in IL-10 expression affected the pathology of muscular dystrophy. Ablation of IL-10 expression in mdx mice increased muscle damage in vivo and reduced mouse strength. Treating mdx muscle macrophages with IL-10 reduced activation of the M1 phenotype, assessed by iNOS expression, and macrophages from IL-10 null mutant mice were more cytolytic than macrophages isolated from wild-type mice. Our data also showed that muscle cells in mdx muscle expressed the IL-10 receptor, suggesting that IL-10 could have direct effects on muscle cells. We assayed whether ablation of IL-10 in mdx mice affected satellite cell numbers, using Pax7 expression as an index, but found no effect. However, IL-10 mutation significantly increased myogenin expression in vivo during the acute and the regenerative phase of mdx pathology. Together, the results show that IL-10 plays a significant regulatory role in muscular dystrophy that may be caused by reducing M1 macrophage activation and cytotoxicity, increasing M2c macrophage activation and modulating muscle differentiation.


Asunto(s)
Interleucina-10/genética , Interleucina-10/metabolismo , Activación de Macrófagos/fisiología , Macrófagos/inmunología , Macrófagos/metabolismo , Distrofia Muscular de Duchenne , Fenotipo , Animales , Diferenciación Celular , Línea Celular , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Músculo Esquelético/citología , Músculo Esquelético/patología , Músculo Esquelético/fisiología , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/patología , Fagocitosis/fisiología , Receptores de Interleucina-10/genética , Receptores de Interleucina-10/metabolismo , Regeneración
14.
J Immunol ; 187(10): 5419-28, 2011 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-22013114

RESUMEN

Duchenne muscular dystrophy is a degenerative disorder that leads to death by the third decade of life. Previous investigations have shown that macrophages that invade dystrophic muscle are a heterogeneous population consisting of M1 and M2 macrophages that promote injury and repair, respectively. In the present investigation, we tested whether IFN-γ worsens the severity of mdx dystrophy by activating macrophages to a cytolytic M1 phenotype and by suppressing the activation of proregenerative macrophages to an M2 phenotype. IFN-γ is a strong inducer of the M1 phenotype and is elevated in mdx dystrophy. Contrary to our expectations, null mutation of IFN-γ caused no reduction of cytotoxicity of macrophages isolated from mdx muscle and did not reduce muscle fiber damage in vivo or improve gross motor function of mdx mice at the early, acute peak of pathology. In contrast, ablation of IFN-γ reduced muscle damage in vivo during the regenerative stage of the disease and increased activation of the M2 phenotype and improved motor function of mdx mice at that later stage of the disease. IFN-γ also inhibited muscle cell proliferation and differentiation in vitro, and IFN-γ mutation increased MyoD expression in mdx muscle in vivo, showing that IFN-γ can have direct effects on muscle cells that could impair repair. Taken together, the findings show that suppression of IFN-γ signaling in muscular dystrophy reduces muscle damage and improves motor performance by promoting the M2 macrophage phenotype and by direct actions on muscle cells.


Asunto(s)
Inhibidores de Crecimiento/efectos adversos , Inmunosupresores/efectos adversos , Interferón gamma/fisiología , Activación de Macrófagos/inmunología , Músculo Esquelético/inmunología , Músculo Esquelético/patología , Distrofia Muscular de Duchenne/inmunología , Distrofia Muscular de Duchenne/patología , Animales , Células Cultivadas , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Inhibidores de Crecimiento/deficiencia , Inhibidores de Crecimiento/fisiología , Inmunofenotipificación , Inmunosupresores/farmacología , Interferón gamma/efectos adversos , Interferón gamma/deficiencia , Activación de Macrófagos/genética , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Ratones Noqueados , Ratones Transgénicos , Músculo Esquelético/crecimiento & desarrollo , Distrofia Muscular de Duchenne/etiología , Regeneración/genética , Regeneración/inmunología
15.
Muscle Nerve ; 45(2): 266-73, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22246884

RESUMEN

INTRODUCTION: p38γ kinase is highly enriched in skeletal muscle and is implicated in myotube formation. However, the activation status of p38γ in muscle is unclear. METHODS: p38γ activity in slow and fast adult mouse skeletal muscle tissue was examined, as was the impact of p38γ deficiency on muscle development and gene expression. RESULTS: p38γ is preferentially activated in slow muscle, but it is inactive in fast muscle types. Furthermore, the loss of p38γ in mice led to decreased muscle mass associated with a smaller myofiber diameter in slow muscle, but there was no impact on fast muscle in either mass or myofiber diameter. Finally, p38γ-deficient muscle showed selective changes in genes related to muscle growth in slow muscle fibers. CONCLUSION: This study provides evidence that p38γ is selectively activated in slow skeletal muscle and is involved in the normal growth and development of a subset of skeletal muscle.


Asunto(s)
Regulación de la Expresión Génica/genética , Fibras Musculares de Contracción Lenta/enzimología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Calcineurina/genética , Calcineurina/metabolismo , Transportador de Glucosa de Tipo 4/genética , Transportador de Glucosa de Tipo 4/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Cadenas Pesadas de Miosina/genética , Cadenas Pesadas de Miosina/metabolismo , Tamaño de los Órganos/genética , PPAR gamma/genética , PPAR gamma/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , ARN Mensajero , Transactivadores/genética , Transactivadores/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/deficiencia
16.
Aging Cell ; 21(10): e13690, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36098370

RESUMEN

Intramuscular macrophages play key regulatory roles in determining the response of skeletal muscle to injury and disease. Recent investigations showed that the numbers and phenotype of intramuscular macrophages change during aging, suggesting that those changes could influence the aging process. We tested that hypothesis by generating a mouse model that harbors a myeloid cell-specific mutation of Spi1, which is a transcription factor that is essential for myeloid cell development. The mutation reduced the numbers of macrophages biased to the CD163+/CD206+ M2 phenotype in muscles of aging mice without affecting the numbers of CD68-expressing macrophages and reduced the expression of transcripts associated with the M2-biased phenotype. The mutation did not affect the colony-forming ability or the frequency of specific subpopulations of bone marrow hematopoietic cells and did not affect myeloid/lymphoid cell ratios in peripheral blood leukocyte populations. Cellularity of most myeloid lineage cells was not influenced by the mutation. The Spi1 mutation in bone marrow-derived macrophages in vitro also did not affect expression of transcripts that indicate the M2-biased phenotype. Thus, myeloid cell-targeted mutation of Spi1 influences macrophage phenotype in muscle but did not affect earlier stages of differentiation of cells in the macrophage lineage. The mutation reduced age-related muscle fibrosis, which is consistent with the reduction of M2-biased macrophages, and reduced expression of the pro-fibrotic enzyme arginase. Most importantly, the mutation prevented sarcopenia. Together, our observations indicate that intramuscular, M2-biased macrophages play significant roles in promoting detrimental, age-related changes in muscle.


Asunto(s)
Sarcopenia , Animales , Ratones , Arginasa/metabolismo , Fibrosis , Macrófagos/metabolismo , Músculo Esquelético/metabolismo , Mutación/genética , Células Mieloides , Sarcopenia/genética , Sarcopenia/metabolismo , Sarcopenia/prevención & control , Factores de Transcripción/metabolismo
17.
Hum Mol Genet ; 18(3): 482-96, 2009 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-18996917

RESUMEN

Duchenne muscular dystrophy (DMD) is the most common, lethal, muscle-wasting disease of childhood. Previous investigations have shown that muscle macrophages may play an important role in promoting the pathology in the mdx mouse model of DMD. In the present study, we investigate the mechanism through which macrophages promote mdx dystrophy and assess whether the phenotype of the macrophages changes between the stage of peak muscle necrosis (4 weeks of age) and muscle regeneration (12 weeks). We find that 4-week-old mdx muscles contain a population of pro-inflammatory, classically activated M1 macrophages that lyse muscle in vitro by NO-mediated mechanisms. Genetic ablation of the iNOS gene in mdx mice also significantly reduces muscle membrane lysis in 4-week-old mdx mice in vivo. However, 4-week mdx muscles also contain a population of alternatively activated, M2a macrophages that express arginase. In vitro assays show that M2a macrophages reduce lysis of muscle cells by M1 macrophages through the competition of arginase in M2a cells with iNOS in M1 cells for their common, enzymatic substrate, arginine. During the transition from the acute peak of mdx pathology to the regenerative stage, expression of IL-4 and IL-10 increases, either of which can deactivate the M1 phenotype and promote activation of a CD163+, M2c phenotype that can increase tissue repair. Our findings further show that IL-10 stimulation of macrophages activates their ability to promote satellite cell proliferation. Deactivation of the M1 phenotype is also associated with a reduced expression of iNOS, IL-6, MCP-1 and IP-10. Thus, these results show that distinct subpopulations of macrophages can promote muscle injury or repair in muscular dystrophy, and that therapeutic interventions that affect the balance between M1 and M2 macrophage populations may influence the course of muscular dystrophy.


Asunto(s)
Arginina/metabolismo , Macrófagos/inmunología , Músculo Esquelético/patología , Distrofia Muscular de Duchenne/inmunología , Animales , Proliferación Celular , Células Cultivadas , Citocinas/inmunología , Humanos , Activación de Macrófagos , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Músculo Esquelético/inmunología , Músculo Esquelético/metabolismo , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/inmunología , Distrofia Muscular Animal/metabolismo , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Fenotipo , Índice de Severidad de la Enfermedad
18.
Hum Mol Genet ; 18(18): 3439-51, 2009 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-19542095

RESUMEN

Duchenne muscular dystrophy (DMD) involves a complex pathophysiology that is not easily explained by the loss of the protein dystrophin, the primary defect in DMD. Instead, many features of the pathology are attributable to the secondary loss of neuronal nitric oxide synthase (nNOS) from dystrophin-deficient muscle. In this investigation, we tested whether the loss of nNOS contributes to the increased fatigability of mdx mice, a model of DMD. Our findings show that the expression of a muscle-specific, nNOS transgene increases the endurance of mdx mice and enhances glycogen metabolism during treadmill-running, but did not affect vascular perfusion of muscles. We also find that the specific activity of phosphofructokinase (PFK; the rate limiting enzyme in glycolysis) is positively affected by nNOS in muscle; PFK-specific activity is significantly reduced in mdx muscles and the muscles of nNOS null mutants, but significantly increased in nNOS transgenic muscles and muscles from mdx mice that express the nNOS transgene. PFK activity measured under allosteric conditions was significantly increased by nNOS, but unaffected by endothelial NOS or inducible NOS. The specific domain of nNOS that positively regulates PFK activity was assayed by cloning and expressing different domains of nNOS and assaying their effects on PFK activity. This approach yielded a polypeptide that included the flavin adenine dinucleotide (FAD)-binding domain of nNOS as the region of the molecule that promotes PFK activity. Smaller peptides in this domain were then synthesized and used in activity assays that showed a 36-amino acid peptide in the FAD-binding domain in which most of the positive allosteric activity of nNOS for PFK resides. Mapping this peptide onto the structure of nNOS shows that the peptide is exposed on the surface, readily available for binding. Collectively, these findings indicate that defects in glycolytic metabolism and increased fatigability in dystrophic muscle may be caused in part by the loss of positive allosteric interactions between nNOS and PFK.


Asunto(s)
Distrofias Musculares/enzimología , Distrofias Musculares/fisiopatología , Óxido Nítrico Sintasa de Tipo I/metabolismo , Fosfofructoquinasas/metabolismo , Regulación Alostérica , Animales , Flavina-Adenina Dinucleótido/metabolismo , Glucógeno/metabolismo , Glucólisis , Ratones , Ratones Endogámicos mdx , Ratones Transgénicos , Modelos Moleculares , Distrofias Musculares/genética , Óxido Nítrico Sintasa de Tipo I/química , Óxido Nítrico Sintasa de Tipo I/genética , Condicionamiento Físico Animal , Unión Proteica , Estructura Terciaria de Proteína , Especificidad por Sustrato
20.
Exp Gerontol ; 145: 111200, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33359378

RESUMEN

Skeletal muscle regeneration that follows acute injury is strongly influenced by interactions with immune cells that invade and proliferate in the damaged tissue. Discoveries over the past 20 years have identified many of the key mechanisms through which myeloid cells, especially macrophages, regulate muscle regeneration. In addition, lymphoid cells that include CD8+ T-cells and regulatory T-cells also significantly affect the course of muscle regeneration. During aging, the regenerative capacity of skeletal muscle declines, which can contribute to progressive loss of muscle mass and function. Those age-related reductions in muscle regeneration are accompanied by systemic, age-related changes in the immune system, that affect many of the myeloid and lymphoid cell populations that can influence muscle regeneration. In this review, we present recent discoveries that indicate that aging of the immune system contributes to the diminished regenerative capacity of aging muscle. Intrinsic, age-related changes in immune cells modify their expression of factors that affect the function of a population of muscle stem cells, called satellite cells, that are necessary for normal muscle regeneration. For example, age-related reductions in the expression of growth differentiation factor-3 (GDF3) or CXCL10 by macrophages negatively affect adult myogenesis, by disrupting regulatory interactions between macrophages and satellite cells. Those changes contribute to a reduction in the numbers and myogenic capacity of satellite cells in old muscle, which reduces their ability to restore damaged muscle. In addition, aging produces changes in the expression of molecules that regulate the inflammatory response to injured muscle, which also contributes to age-related defects in muscle regeneration. For example, age-related increases in the production of osteopontin by macrophages disrupts the normal inflammatory response to muscle injury, resulting in regenerative defects. These nascent findings represent the beginning of a newly-developing field of investigation into mechanisms through which aging of the immune system affects muscle regeneration.


Asunto(s)
Regeneración , Células Satélite del Músculo Esquelético , Inmunomodulación , Desarrollo de Músculos , Músculo Esquelético
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA