36 research outputs found

    Modulation of Klotho expression in injured muscle perturbs Wnt signalling and influences the rate of muscle growth

    Get PDF
    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 β‐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

    Differential Effects of Myeloid Cell PPARδ and IL-10 in Regulating Macrophage Recruitment, Phenotype, and Regeneration following Acute Muscle Injury

    Get PDF
    Changes in macrophage phenotype in injured muscle profoundly influence regeneration. In particular, the shift of macrophages from a pro-inflammatory (M1-biased) phenotype to a pro-regenerative (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 IL10 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

    Neuronal Nitric Oxide Synthase-Rescue of Dystrophin/Utrophin Double Knockout Mice does not Require nNOS Localization to the Cell Membrane

    Get PDF
    Survival of dystrophin/utrophin double-knockout (dko) mice was increased by muscle-specific expression of a neuronal nitric oxide synthase (nNOS) transgene. Dko mice expressing the transgene (nNOS TG+/dko) experienced delayed onset of mortality and increased life-span. The nNOS TG+/dko mice demonstrated a significant decrease in the concentration of CD163+, M2c macrophages that can express arginase and promote fibrosis. The decrease in M2c macrophages was associated with a significant reduction in fibrosis of heart, diaphragm and hindlimb muscles of nNOS TG+/dko mice. The nNOS transgene had no effect on the concentration of cytolytic, CD68+, M1 macrophages. Accordingly, we did not observe any change in the extent of muscle fiber lysis in the nNOS TG+/dko mice. These findings show that nNOS/NO (nitric oxide)-mediated decreases in M2c macrophages lead to a reduction in the muscle fibrosis that is associated with increased mortality in mice lacking dystrophin and utrophin. Interestingly, the dramatic and beneficial effects of the nNOS transgene were not attributable to localization of nNOS protein at the cell membrane. We did not detect any nNOS protein at the sarcolemma in nNOS TG+/dko muscles. This important observation shows that sarcolemmal localization is not necessary for nNOS to have beneficial effects in dystrophic tissue and the presence of nNOS in the cytosol of dystrophic muscle fibers can ameliorate the pathology and most importantly, significantly increase life-span

    Arginine Metabolism by Macrophages Promotes Cardiac and Muscle Fibrosis in mdx Muscular Dystrophy

    Get PDF
    Duchenne muscular dystrophy (DMD) is the most common, lethal disease of childhood. One of 3500 new-born males suffers from this universally-lethal disease. Other than the use of corticosteroids, little is available to affect the relentless progress of the disease, leading many families to use dietary supplements in hopes of reducing the progression or severity of muscle wasting. Arginine is commonly used as a dietary supplement and its use has been reported to have beneficial effects following short-term administration to mdx mice, a genetic model of DMD. However, the long-term effects of arginine supplementation are unknown. This lack of knowledge about the long-term effects of increased arginine metabolism is important because elevated arginine metabolism can increase tissue fibrosis, and increased fibrosis of skeletal muscles and the heart is an important and potentially life-threatening feature of DMD.We use both genetic and nutritional manipulations to test whether changes in arginase metabolism promote fibrosis and increase pathology in mdx mice. Our findings show that fibrotic lesions in mdx muscle are enriched with arginase-2-expressing macrophages and that muscle macrophages stimulated with cytokines that activate the M2 phenotype show elevated arginase activity and expression. We generated a line of arginase-2-null mutant mdx mice and found that the mutation reduced fibrosis in muscles of 18-month-old mdx mice, and reduced kyphosis that is attributable to muscle fibrosis. We also observed that dietary supplementation with arginine for 17-months increased mdx muscle fibrosis. In contrast, arginine-2 mutation did not reduce cardiac fibrosis or affect cardiac function assessed by echocardiography, although 17-months of dietary supplementation with arginine increased cardiac fibrosis. Long-term arginine treatments did not decrease matrix metalloproteinase-2 or -9 or increase the expression of utrophin, which have been reported as beneficial effects of short-term treatments.Our findings demonstrate that arginine metabolism by arginase promotes fibrosis of muscle in muscular dystrophy and contributes to kyphosis. Our findings also show that long-term, dietary supplementation with arginine exacerbates fibrosis of dystrophic heart and muscles. Thus, commonly-practiced dietary supplementation with arginine by DMD patients has potential risk for increasing pathology when performed for long periods, despite reports of benefits acquired with short-term supplementation

    nNOS protein and nitric oxide are increased in dko mice expressing a nNOS transgene.

    No full text
    <p>A. Overexpression of the nNOS transgene was confirmed by western blots for all mice assayed in the investigation. Upper panel shows a blot for nNOS in a representative nNOS TG+/dko muscle extract (Tg+) and a representative nNOS TG-/dko extract (TG-). Lower panel shows the same blot stained with Ponceau red prior to antibody labeling to confirm uniform loading of samples. B. Percent cytosolic nitric oxide (NO) in wild-type (white bar), nNOS TG-/dko (black bar) and nNOS TG+/dko (grey bar) muscle. n = 5 mice per group. Error bars represent standard error of the mean. *  =  significant difference as compared to wild-type. #  =  significant difference as compared to nNOS TG-/dko. p<0.05. C. Wild-type muscle showing NO present in the cytosol and highly concentrated at the sarcolemma. Bar = 50 µm. D. nNOS TG-/dko muscle exhibits low levels of NO in muscle fibers, but strong NO production by infiltrating mononuclear cells (arrow). E. Increased levels of NO are evident in the cytosol of muscle fibers from nNOS TG+/dko mice.</p

    Fibrosis is inversely correlated with nNOS expression in dko muscle.

    No full text
    <p>Linear regression was used to quantify the relationship between nNOS expression (measured densitometrically following western blotting) and hydroxyproline content in quadriceps muscles from nNOS TG+/dko and nNOS TG-/dko mice. n = 5 mice per group.</p

    nNOS transgene expression does not reduce muscle fiber damage in dko mice.

    No full text
    <p>A. The percentage of damaged fibers in soleus muscles of nNOS TG-/dko mice (black bar) and nNOS TG+/dko mice (grey bar) does not differ. n = 5 mice per group. B. The aggregate distribution of intracellular fluorescence in all soleus fibers from each group. A rightward shift on the abscissa would indicate an increase in the number of fibers with lesions. The black and white, striped peaks represent fibers of nNOS TG-/dko mice, the red peaks represent fibers from nNOS TG+/dko mice and the striped, red areas indicate overlap between the groups. n = 5 mice per group.</p
    corecore