Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 118(9)2021 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-33627403

RESUMO

Duchenne muscular dystrophy (DMD) is an X-linked recessive disorder characterized by progressive muscle degeneration and weakness due to mutations in the dystrophin gene. The symptoms of DMD share similarities with those of accelerated aging. Recently, hydrogen sulfide (H2S) supplementation has been suggested to modulate the effects of age-related decline in muscle function, and metabolic H2S deficiencies have been implicated in affecting muscle mass in conditions such as phenylketonuria. We therefore evaluated the use of sodium GYY4137 (NaGYY), a H2S-releasing molecule, as a possible approach for DMD treatment. Using the dys-1(eg33) Caenorhabditis elegans DMD model, we found that NaGYY treatment (100 µM) improved movement, strength, gait, and muscle mitochondrial structure, similar to the gold-standard therapeutic treatment, prednisone (370 µM). The health improvements of either treatment required the action of the kinase JNK-1, the transcription factor SKN-1, and the NAD-dependent deacetylase SIR-2.1. The transcription factor DAF-16 was required for the health benefits of NaGYY treatment, but not prednisone treatment. AP39 (100 pM), a mitochondria-targeted H2S compound, also improved movement and strength in the dys-1(eg33) model, further implying that these improvements are mitochondria-based. Additionally, we found a decline in total sulfide and H2S-producing enzymes in dystrophin/utrophin knockout mice. Overall, our results suggest that H2S deficit may contribute to DMD pathology, and rectifying/overcoming the deficit with H2S delivery compounds has potential as a therapeutic approach to DMD treatment.


Assuntos
Proteínas de Caenorhabditis elegans/genética , Distrofina/genética , Sulfeto de Hidrogênio/farmacologia , Mitocôndrias Musculares/efeitos dos fármacos , Morfolinas/farmacologia , Músculo Esquelético/efeitos dos fármacos , Distrofia Muscular Animal/tratamento farmacológico , Compostos Organofosforados/farmacologia , Compostos Organotiofosforados/farmacologia , Tionas/farmacologia , Animais , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Distrofina/deficiência , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica , Humanos , Sulfeto de Hidrogênio/metabolismo , Locomoção/efeitos dos fármacos , Locomoção/genética , Masculino , Camundongos , Camundongos Endogâmicos mdx , Mitocôndrias Musculares/metabolismo , Mitocôndrias Musculares/patologia , Proteínas Quinases Ativadas por Mitógeno/genética , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Morfolinas/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/metabolismo , Distrofia Muscular Animal/patologia , Distrofia Muscular de Duchenne/tratamento farmacológico , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/patologia , Compostos Organofosforados/metabolismo , Compostos Organotiofosforados/metabolismo , Prednisona/farmacologia , Sirtuínas/genética , Sirtuínas/metabolismo , Tionas/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Utrofina/deficiência , Utrofina/genética
2.
Biochem J ; 479(11): 1181-1204, 2022 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-35552369

RESUMO

The AMP-activated protein kinase (AMPK) αßγ heterotrimer is a primary cellular energy sensor and central regulator of energy homeostasis. Activating skeletal muscle AMPK with small molecule drugs improves glucose uptake and provides an opportunity for new strategies to treat type 2 diabetes and insulin resistance, with recent genetic and pharmacological studies indicating the α2ß2γ1 isoform combination as the heterotrimer complex primarily responsible. With the goal of developing α2ß2-specific activators, here we perform structure/function analysis of the 2-hydroxybiphenyl group of SC4, an activator with tendency for α2-selectivity that is also capable of potently activating ß2 complexes. Substitution of the LHS 2-hydroxyphenyl group with polar-substituted cyclohexene-based probes resulted in two AMPK agonists, MSG010 and MSG011, which did not display α2-selectivity when screened against a panel of AMPK complexes. By radiolabel kinase assay, MSG010 and MSG011 activated α2ß2γ1 AMPK with one order of magnitude greater potency than the pan AMPK activator MK-8722. A crystal structure of MSG011 complexed to AMPK α2ß1γ1 revealed a similar binding mode to SC4 and the potential importance of an interaction between the SC4 2-hydroxyl group and α2-Lys31 for directing α2-selectivity. MSG011 induced robust AMPK signalling in mouse primary hepatocytes and commonly used cell lines, and in most cases this occurred in the absence of changes in phosphorylation of the kinase activation loop residue α-Thr172, a classical marker of AMP-induced AMPK activity. These findings will guide future design of α2ß2-selective AMPK activators, that we hypothesise may avoid off-target complications associated with indiscriminate activation of AMPK throughout the body.


Assuntos
Proteínas Quinases Ativadas por AMP , Diabetes Mellitus Tipo 2 , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Linhagem Celular , Diabetes Mellitus Tipo 2/metabolismo , Camundongos , Músculo Esquelético/metabolismo , Fosforilação
3.
Am J Physiol Cell Physiol ; 323(2): C378-C384, 2022 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-35704693

RESUMO

Skeletal muscle atrophy and dysfunction contribute to morbidity and mortality in patients with cancer. Cachexia pathophysiology is highly complex, given that perturbations to the systemic cancer environment and the interaction with diverse tissues can contribute to wasting processes. Systemic interleukin 6 (IL-6) and glycoprotein 130 (gp130) receptors signaling have established roles in some types of cancer-induced muscle wasting through disruptions to protein turnover and oxidative capacity. Although exercise has documented benefits for cancer prevention and patient survival, there are significant gaps in our understanding of muscle adaptation and plasticity during severe cachexia. Preclinical models have provided valuable insight into the adaptive potential of muscle contraction within the cancer environment. We summarize the current understanding of how resistance-type exercise impacts mechanisms involved in cancer-induced muscle atrophy and dysfunction. Specifically, the role of IL-6 and gp130 receptors in the pathophysiology of muscle wasting and the adaptive response to exercise is explained. The discussion includes current knowledge gaps and future research directions needed to improve preclinical research and accelerate clinical translation in human patients with cancer.


Assuntos
Caquexia , Neoplasias , Caquexia/etiologia , Caquexia/prevenção & controle , Receptor gp130 de Citocina/metabolismo , Humanos , Interleucina-6/metabolismo , Contração Muscular , Músculo Esquelético/metabolismo , Atrofia Muscular/patologia , Neoplasias/metabolismo
4.
Am J Physiol Cell Physiol ; 321(2): C288-C296, 2021 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-34191629

RESUMO

Impaired oxidative capacity and mitochondrial function contribute to the dystrophic pathology in muscles of patients with Duchenne muscular dystrophy (DMD) and in relevant mouse models of the disease. Emerging evidence suggests an association between disrupted core clock expression and mitochondrial quality control, but this has not been established in muscles lacking dystrophin. We examined the diurnal regulation of muscle core clock and mitochondrial quality control expression in dystrophin-deficient C57BL/10ScSn-Dmdmdx (mdx) mice, an established model of DMD. Male C57BL/10 (BL/10; n = 18) and mdx mice (n = 18) were examined every 4 h beginning at the dark cycle. Throughout the entire light-dark cycle, extensor digitorum longus (EDL) muscles from mdx mice had decreased core clock mRNA expression (Arntl, Cry1, Cry2, Nr1d2; P < 0.05) and disrupted mitochondrial quality control mRNA expression related to biogenesis (decreased; Ppargc1a, Esrra; P < 0.05), fission (increased; Dnm1l; P < 0.01), fusion (decreased; Opa1, Mfn1; P < 0.05), and autophagy/mitophagy (decreased: Bnip3; P < 0.05; increased: Becn1; P < 0.05). Cosinor analysis revealed a decrease in the rhythmicity parameters mesor and amplitude for Arntl, Cry1, Cry2, Per2, and Nr1d1 (P < 0.001) in mdx mice. Diurnal oscillations in Esrra, Sirt1, Map1lc3b, and Sqstm1 were absent in mdx mice, along with decreased mesor and amplitude of Ppargc1a mRNA expression (P < 0.01). The expression of proteins involved in mitochondrial biogenesis (decreased: PPARGC1A, P < 0.05) and autophagy/mitophagy (increased: MAP1LC3BII, SQSTM1, BNIP3; P < 0.05) were also dysregulated in tibialis anterior muscles of mdx mice. These findings suggest that dystrophin deficiency in mdx mice impairs the regulation of the core clock and mitochondrial quality control, with relevance to DMD and related disorders.


Assuntos
Distrofina/deficiência , Mitocôndrias/metabolismo , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/fisiopatologia , Animais , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Distrofia Muscular de Duchenne/metabolismo , Utrofina/deficiência
5.
Semin Cell Dev Biol ; 54: 53-67, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26593326

RESUMO

While skeletal muscle mass is an established primary outcome related to understanding cancer cachexia mechanisms, considerable gaps exist in our understanding of muscle biochemical and functional properties that have recognized roles in systemic health. Skeletal muscle quality is a classification beyond mass, and is aligned with muscle's metabolic capacity and substrate utilization flexibility. This supplies an additional role for the mitochondria in cancer-induced muscle wasting. While the historical assessment of mitochondria content and function during cancer-induced muscle loss was closely aligned with energy flux and wasting susceptibility, this understanding has expanded to link mitochondria dysfunction to cellular processes regulating myofiber wasting. The primary objective of this article is to highlight muscle mitochondria and oxidative metabolism as a biological target of cancer cachexia and also as a cellular regulator of cancer-induced muscle wasting. Initially, we examine the role of muscle metabolic phenotype and mitochondria content in cancer-induced wasting susceptibility. We then assess the evidence for cancer-induced regulation of skeletal muscle mitochondrial biogenesis, dynamics, mitophagy, and oxidative stress. In addition, we discuss environments associated with cancer cachexia that can impact the regulation of skeletal muscle oxidative metabolism. The article also examines the role of cytokine-mediated regulation of mitochondria function, followed by the potential role of cancer-induced hypogonadism. Lastly, a role for decreased muscle use in cancer-induced mitochondrial dysfunction is reviewed.


Assuntos
Caquexia/etiologia , Caquexia/metabolismo , Músculo Esquelético/metabolismo , Neoplasias/complicações , Neoplasias/metabolismo , Animais , Humanos , Mitocôndrias/metabolismo , Modelos Biológicos , Oxirredução
6.
Am J Physiol Cell Physiol ; 315(1): C91-C103, 2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29641213

RESUMO

Systemic cytokines and contractile activity are established regulators of muscle protein turnover. Paradoxically, the IL-6 cytokine family, which shares the ubiquitously expressed membrane gp130 receptor, has been implicated in skeletal muscle's response to both contractions and cancer-induced wasting. Although we have reported that tumor-derived cachectic factors could suppress stretch-induced protein synthesis in cultured myotubes, the ability of systemic cytokines to disrupt in vivo eccentric contraction-induced protein synthesis has not been established. Therefore, we examined whether systemic IL-6 regulates basal and eccentric contraction-induced protein synthesis through muscle gp130 signaling. Systemic IL-6 overexpression was performed for 2 wk, and we then examined basal and eccentric contraction-induced protein synthesis and mammalian target of rapamycin complex 1 (mTORC1) signaling in tibialis anterior muscle of male wild-type, muscle-specific gp130 receptor knockout, and tumor-bearing ApcMin/+ mice. Systemic IL-6 overexpression suppressed basal protein synthesis and mTORC1 signaling independently of IL-6 level, which was rescued by muscle gp130 loss. Interestingly, only high systemic IL-6 levels suppressed eccentric contraction-induced protein synthesis. Systemic IL-6 overexpression in precachectic tumor-bearing ApcMin/+ mice accelerated cachexia development, which coincided with suppressed basal and eccentric contraction-induced muscle protein synthesis. The suppression of eccentric contraction-induced protein synthesis by IL-6 occurred independently of mTORC1 activation. Collectively, these findings demonstrate that basal protein synthesis suppression was more sensitive to circulating IL-6 compared with the induction of protein synthesis by eccentric contraction. However, systemic IL-6 can interact with the cancer environment to suppress eccentric contraction-induced protein synthesis independently of mTORC1 activation.


Assuntos
Interleucina-6/metabolismo , Contração Muscular/fisiologia , Proteínas Musculares/metabolismo , Biossíntese de Proteínas/fisiologia , Animais , Caquexia/metabolismo , Caquexia/fisiopatologia , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Células Musculares/metabolismo , Células Musculares/fisiologia , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/fisiologia , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiologia , Transdução de Sinais/fisiologia
7.
Exerc Sport Sci Rev ; 46(4): 247-253, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30001273

RESUMO

Skeletal muscle has the dynamic capability to modulate protein turnover in response to anabolic stimuli, such as feeding and contraction. We propose that anabolic resistance, the suppressed ability to induce protein synthesis, is central to cancer-induced muscle wasting. Furthermore, we propose that resistance exercise training has the potential to attenuate or treat cancer-induced anabolic resistance through improvements in oxidative metabolism.


Assuntos
Caquexia/terapia , Terapia por Exercício , Proteínas Musculares/biossíntese , Músculo Esquelético/fisiopatologia , Neoplasias/terapia , Treinamento Resistido , Humanos , Mitocôndrias Musculares/patologia
8.
Am J Physiol Endocrinol Metab ; 312(5): E447-E459, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28292759

RESUMO

Cachexia is a debilitating condition that occurs with chronic disease, including cancer; our research has shown that some regulation of cancer cachexia progression is affected by sex differences. The ApcMin/+ mouse is genetically predisposed to develop intestinal tumors; IL-6 signaling and hypogonadism are associated with cachexia severity in the male. This relationship in the female warrants further investigation, as we have shown that the ability of IL-6 to induce cachexia differs between the sexes. Since ovarian reproductive function relies on a complex system of endocrine signaling to affect whole body homeostasis, we examined the relationship between ovarian reproductive function and progression of cancer cachexia in the female ApcMin/+ mouse. Our study of ovarian reproductive function in female ApcMin/+ mice showed disease-related cessation of estrous cycling (acyclicity) in 38% of mice. Acyclicity, including morphological and functional losses and enhanced muscle inflammatory gene expression, was associated with severe cachexia. Interestingly, ovariectomy rescued body weight and muscle mass and function but increased muscle sensitivity to systemic IL-6 overexpression. In conclusion, our results provide evidence for a relationship between ovarian reproductive function and cachexia progression in female ApcMin/+ mice.


Assuntos
Polipose Adenomatosa do Colo/fisiopatologia , Caquexia/fisiopatologia , Ciclo Estral , Músculo Esquelético/fisiopatologia , Ovário/fisiopatologia , Polipose Adenomatosa do Colo/complicações , Animais , Caquexia/etiologia , Progressão da Doença , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
9.
Biochem Biophys Res Commun ; 493(3): 1236-1242, 2017 11 25.
Artigo em Inglês | MEDLINE | ID: mdl-28962861

RESUMO

Skeletal muscle atrophy is associated with a disruption in protein turnover involving increased protein degradation and suppressed protein synthesis. Although it has been well studied that the IGF-1/PI3K/Akt pathway plays an essential role in the regulation of the protein turnover, molecule(s) that triggers the change in protein turnover still remains to be elucidated. TRB3 has been shown to inhibit Akt through direct binding. In this study, we hypothesized that TRB3 in mouse skeletal muscle negatively regulates protein turnover via the disruption of Akt and its downstream molecules. Muscle-specific TRB3 transgenic (TRB3TG) mice had decreased muscle mass and fiber size, resulting in impaired muscle function. We also found that protein synthesis rate and signaling molecules, mTOR and S6K1, were significantly reduced in TRB3TG mice, whereas the protein breakdown pathway was significantly activated. In contrast, TRB3 knockout mice showed increased muscle mass and had an increase in protein synthesis rate, but decreases in FoxOs, atrogin-1, and MuRF-1. These findings indicate that TRB3 regulates protein synthesis and breakdown via the Akt/mTOR/FoxO pathways.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Músculo Esquelético/metabolismo , Animais , Proteínas de Ciclo Celular/genética , Feminino , Proteína Forkhead Box O1/metabolismo , Proteína Forkhead Box O3/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Proteínas Musculares/genética , Músculo Esquelético/fisiopatologia , Biossíntese de Proteínas/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Proteínas Ligases SKP Culina F-Box/genética , Serina-Treonina Quinases TOR/metabolismo
10.
Biochim Biophys Acta ; 1852(5): 816-25, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25555992

RESUMO

A devastating aspect of cancer cachexia is severe loss of muscle and fat mass. Though cachexia occurs in both sexes, it is not well-defined in the female. The Apc(Min/+) mouse is genetically predisposed to develop intestinal tumors; circulating IL-6 is a critical regulator of cancer cachexia in the male Apc(Min/+) mouse. The purpose of this study was to examine the relationship between IL-6 signaling and cachexia progression in the female Apc(Min/+) mouse. Male and female Apc(Min/+) mice were examined during the initiation and progression of cachexia. Another group of females had IL-6 overexpressed between 12 and 14 weeks or 15-18 weeks of age to determine whether IL-6 could induce cachexia. Cachectic female Apc(Min/+) mice lost body weight, muscle mass, and fat mass; increased muscle IL-6 mRNA expression was associated with these changes, but circulating IL-6 levels were not. Circulating IL-6 levels did not correlate with downstream signaling in muscle in the female. Muscle IL-6r mRNA expression and SOCS3 mRNA expression as well as muscle IL-6r protein and STAT3 phosphorylation increased with severe cachexia in both sexes. Muscle SOCS3 protein increased in cachectic females but decreased in cachectic males. IL-6 overexpression did not affect cachexia progression in female Apc(Min/+) mice. Our results indicate that female Apc(Min/+) mice undergo cachexia progression that is at least initially IL-6-independent. Future studies in the female will need to determine mechanisms underlying regulation of IL-6 response and cachexia induction.


Assuntos
Polipose Adenomatosa do Colo/genética , Caquexia/genética , Interleucina-6/genética , Transdução de Sinais , Polipose Adenomatosa do Colo/sangue , Polipose Adenomatosa do Colo/complicações , Proteína da Polipose Adenomatosa do Colo/genética , Animais , Western Blotting , Peso Corporal , Caquexia/etiologia , Caquexia/metabolismo , Progressão da Doença , Feminino , Expressão Gênica , Interleucina-6/sangue , Interleucina-6/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Tamanho do Órgão , Receptores de Interleucina-6/genética , Receptores de Interleucina-6/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores Sexuais , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina/genética , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Fatores de Tempo
11.
Exerc Sport Sci Rev ; 43(1): 34-40, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25390300

RESUMO

Severe burn trauma results in persistent skeletal muscle catabolism and prolonged immobilization. We hypothesize that structured rehabilitative exercise is a safe and efficacious strategy to restore lean body mass and physical function in burn victims. Here, we review the evidence for the utility of rehabilitative exercise training in restoring physiological function in burn survivors.


Assuntos
Queimaduras/reabilitação , Terapia por Exercício , Queimaduras/tratamento farmacológico , Queimaduras/fisiopatologia , Sistema Cardiovascular/fisiopatologia , Humanos , Pulmão/fisiopatologia , Músculo Esquelético/fisiopatologia
12.
Radiol Oncol ; 48(3): 247-56, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25177239

RESUMO

BACKGROUND: The purpose of this study was to determine the effect of two clinically relevant radiation doses on the susceptibility of mouse skeletal muscle to remodeling. MATERIALS AND METHODS: Alterations in muscle morphology and regulatory signaling were examined in tibialis anterior and gastrocnemius muscles after radiation doses that differed in total biological effective dose (BED). Female C57BL/6 (8-wk) mice were randomly assigned to non-irradiated control, four fractionated doses of 4 Gy (4x4 Gy; BED 37 Gy), or a single 16 Gy dose (16 Gy; BED 100 Gy). Mice were sacrificed 2 weeks after the initial radiation exposure. RESULTS: The 16 Gy, but not 4x4 Gy, decreased total muscle protein and RNA content. Related to muscle regeneration, both 16 Gy and 4x4 Gy increased the incidence of central nuclei containing myofibers, but only 16 Gy increased the extracellular matrix volume. However, only 4x4 Gy increased muscle 4-hydroxynonenal expression. While both 16 Gy and 4x4 Gy decreased IIB myofiber mean cross-sectional area (CSA), only 16 Gy decreased IIA myofiber CSA. 16 Gy increased the incidence of small diameter IIA and IIB myofibers, while 4x4 Gy only increased the incidence of small diameter IIB myofibers. Both treatments decreased the frequency and CSA of low succinate dehydrogenase activity (SDH) fibers. Only 16 Gy increased the incidence of small diameter myofibers having high SDH activity. Neither treatment altered muscle signaling related to protein turnover or oxidative metabolism. CONCLUSIONS: Collectively, these results demonstrate that radiation dose differentially affects muscle remodeling, and these effects appear to be related to fiber type and oxidative metabolism.

13.
JCI Insight ; 9(8)2024 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-38530354

RESUMO

Skeletal muscle wasting results from numerous pathological conditions affecting both the musculoskeletal and nervous systems. A unifying feature of these pathologies is the upregulation of members of the E3 ubiquitin ligase family, resulting in increased proteolytic degradation of target proteins. Despite the critical role of E3 ubiquitin ligases in regulating muscle mass, the specific proteins they target for degradation and the mechanisms by which they regulate skeletal muscle homeostasis remain ill-defined. Here, using zebrafish loss-of-function models combined with in vivo cell biology and proteomic approaches, we reveal a role of atrogin-1 in regulating the levels of the endoplasmic reticulum chaperone BiP. Loss of atrogin-1 resulted in an accumulation of BiP, leading to impaired mitochondrial dynamics and a subsequent loss in muscle fiber integrity. We further implicated a disruption in atrogin-1-mediated BiP regulation in the pathogenesis of Duchenne muscular dystrophy. We revealed that BiP was not only upregulated in Duchenne muscular dystrophy, but its inhibition using pharmacological strategies, or by upregulating atrogin-1, significantly ameliorated pathology in a zebrafish model of Duchenne muscular dystrophy. Collectively, our data implicate atrogin-1 and BiP in the pathogenesis of Duchenne muscular dystrophy and highlight atrogin-1's essential role in maintaining muscle homeostasis.


Assuntos
Modelos Animais de Doenças , Chaperona BiP do Retículo Endoplasmático , Homeostase , Proteínas Musculares , Músculo Esquelético , Distrofia Muscular de Duchenne , Proteínas Ligases SKP Culina F-Box , Peixe-Zebra , Animais , Proteínas Ligases SKP Culina F-Box/metabolismo , Proteínas Ligases SKP Culina F-Box/genética , Proteínas Musculares/metabolismo , Proteínas Musculares/genética , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/patologia , Distrofia Muscular de Duchenne/genética , Humanos , Chaperona BiP do Retículo Endoplasmático/metabolismo , Proteínas de Choque Térmico/metabolismo , Proteínas de Choque Térmico/genética , Proteínas de Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética , Retículo Endoplasmático/metabolismo , Dinâmica Mitocondrial
14.
Dis Model Mech ; 17(4)2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38602028

RESUMO

Duchenne muscular dystrophy (DMD) is a devastating monogenic skeletal muscle-wasting disorder. Although many pharmacological and genetic interventions have been reported in preclinical studies, few have progressed to clinical trials with meaningful benefit. Identifying therapeutic potential can be limited by availability of suitable preclinical mouse models. More rigorous testing across models with varied background strains and mutations can identify treatments for clinical success. Here, we report the generation of a DMD mouse model with a CRISPR-induced deletion within exon 62 of the dystrophin gene (Dmd) and the first generated in BALB/c mice. Analysis of mice at 3, 6 and 12 months of age confirmed loss of expression of the dystrophin protein isoform Dp427 and resultant dystrophic pathology in limb muscles and the diaphragm, with evidence of centrally nucleated fibers, increased inflammatory markers and fibrosis, progressive decline in muscle function, and compromised trabecular bone development. The BALB/c.mdx62 mouse is a novel model of DMD with associated variations in the immune response and muscle phenotype, compared with those of existing models. It represents an important addition to the preclinical model toolbox for developing therapeutic strategies.


Assuntos
Modelos Animais de Doenças , Distrofina , Camundongos Endogâmicos BALB C , Músculo Esquelético , Distrofia Muscular de Duchenne , Animais , Distrofia Muscular de Duchenne/patologia , Distrofia Muscular de Duchenne/genética , Distrofina/metabolismo , Distrofina/genética , Músculo Esquelético/patologia , Músculo Esquelético/metabolismo , Camundongos Endogâmicos mdx , Camundongos , Éxons/genética , Masculino , Fibrose , Fenótipo
15.
J Sports Sci ; 31(5): 488-96, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23121475

RESUMO

The purpose of this investigation was to examine the effects of cluster set configurations on power clean technique. Ten male, recreational weightlifters performed three sets of six repetitions at 80% of one repetition max with 0 (P0), 20 (P20) or 40 seconds (P40) inter-repetition rest. During the first and second set of P0, the catch and first pull were in a more forward position during repetition 6 as compared to repetition 1, respectively. During the second set of P40, differences in horizontal displacement were found between repetitions 1 and 6 for the second pull and the loop. During the third set of P40, differences in horizontal displacement were found between repetitions 1 and 6 for the first pull, transition, and beginning of the second pull. No differences in horizontal displacement were found between repetitions 1 and 6 during P20. During each set of P0, vertical displacement decreased between repetitions 1 and 6 (1.02 ± 0.07 m vs. 0.94 ± 0.06 m; Mean ± s). Cluster set configurations led to the maintenance of vertical displacement throughout all sets. The results demonstrate cluster set configurations with greater than 20 seconds inter-repetition rest maintain weightlifting technique to a greater extent than a traditional set configuration.


Assuntos
Desempenho Atlético/fisiologia , Movimento/fisiologia , Força Muscular , Músculo Esquelético/fisiologia , Treinamento Resistido , Análise e Desempenho de Tarefas , Levantamento de Peso/fisiologia , Adulto , Humanos , Masculino , Descanso , Adulto Jovem
16.
J Appl Physiol (1985) ; 135(1): 77-87, 2023 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-37262103

RESUMO

Duchenne muscular dystrophy (DMD) is a severe muscle wasting disease caused by mutations or deletions in the dystrophin gene, for which there remains no cure. As DMD patients also develop bone fragility because of muscle weakness and immobilization, better understanding of the pathophysiological mechanisms of dystrophin deficiency will help develop therapies to improve musculoskeletal health. Since alterations in muscle phenotype can influence bone structure, we investigated whether modifying muscle contractile activity through low-frequency stimulation (LFS) could alter bone architecture in mouse models of DMD. We tested the hypothesis that increasing muscle contractile activity could influence bone mass and structure in dystrophin-deficient (mdx) and dystrophin- and utrophin-deficient (dko) dystrophic mice. Tibial bone structure in dko mice was significantly different from that in mdx and wild-type (C57BL/10) control mice. Effects of LFS on bone architecture differed between dystrophic and healthy mice, with LFS thinning cortical bone in both dystrophic models. Bone mass was maintained in LFS-treated healthy mice, with a reduced proportion of high-density bone and concomitant increase in low-density bone. LFS-treated dko mice exhibited a net deficit in cortical thickness and reduced high-density bone but no equivalent increase in low-density bone. These alterations in bone structure and mineral density reduced mechanical strength in mdx and dko mice. The findings reveal that muscle activity can regulate bone mass, structure, mineral accrual, and strength, especially in the context of dystrophin and/or utrophin deficiency. The results provide unique insights into the development of bone fragility in DMD and for devising interventions to improve musculoskeletal health.NEW & NOTEWORTHY Patients with Duchenne muscular dystrophy (DMD) develop bone fragility because of muscle weakness and immobilization. We investigated whether increasing muscle contractile activity through low-frequency stimulation (LFS) could alter bone architecture in dystrophin-deficient (mdx) or dystrophin- and utrophin-deficient (dko) mouse models of DMD. Chronic LFS reduced tibial diaphysis cross sections in mdx and dko mice, without affecting bone shape in healthy mice. LFS affected the distribution of bone mineral density across all phenotypes, with the magnitude of effect being dependent on disease severity.


Assuntos
Distrofina , Distrofia Muscular de Duchenne , Animais , Camundongos , Camundongos Endogâmicos mdx , Utrofina/genética , Camundongos Endogâmicos C57BL , Músculo Esquelético , Debilidade Muscular , Modelos Animais de Doenças
17.
Eur J Appl Physiol ; 112(8): 3141-7, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22215288

RESUMO

The purpose of this study was to examine the effects of inter-repetition rest (IRR) on ratings of perceived exertion (RPE) in the power clean exercise in a multiple set protocol using peak power as an indication of fatigue. Ten resistance-trained males participated in four testing sessions which consisted of determination of a one repetition maximum (1RM) in the power clean exercise (session 1) and performance of three sets of six repetitions at 80% of 1RM with 0 (P0), 20 (P20), or 40 s (P40) IRR (sessions 2-4). Fatigue during all three conditions was indicated by a significant decrease in power of 9.0% (P0), 3.0% (P20) and 2.1% (P40), respectively. Significant difference in the rate of power decrease in P40 indicates less fatigue in comparison to P0 and P20. P40 resulted in a significantly lower RPE compared to P0 and P20 (7.43 ± 0.34, 6.46 ± 0.47, and 5.30 ± 0.55, respectively). RPE increased significantly (p ≤ 0.01) within each set (5.26 ± 0.37, 6.46 ± 0.44, and 7.46 ± 0.53; sets 1, 2, and 3, respectively). Significant difference in average RPE between the conditions indicates that RPE is not a determinant of intensity (% of 1RM) but the rate of fatigue (decreases in peak power). In addition, the fact that RPE increased between sets 1, 2 and 3 during all conditions support the same conclusion. The results demonstrate that increasing IRR in power clean training decreases the perception of effort and is inversely related to the rate of fatigue.


Assuntos
Contração Muscular , Fadiga Muscular , Força Muscular , Músculo Esquelético/fisiologia , Percepção , Treinamento Resistido , Levantamento de Peso , Adulto , Análise de Variância , Fenômenos Biomecânicos , Humanos , Masculino , Recuperação de Função Fisiológica , Fatores de Tempo , Adulto Jovem
18.
J Strength Cond Res ; 26(4): 883-9, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22228112

RESUMO

The effect of interrepetition rest (IRR) periods on power output during performance of multiple sets of power cleans is unknown. It is possible that IRR periods may attenuate the decrease in power output commonly observed within multiple sets. This may be of benefit for maximizing improvements in power with training. This investigation involved 10 college-aged men with proficiency in weightlifting. The subjects performed 3 sets of 6 repetitions of power cleans at 80% of their 1 repetition maximum with 0 (P0), 20 (P20), or 40 seconds (P40) of IRR. Each protocol (P0, P20, P40) was performed in a randomized order on different days each separated by at least 72 hours. The subjects performed the power cleans while standing on a force plate with 2 linear position transducers attached to the bar. Peak power, force, and velocity were obtained for each repetition and set. Peak power significantly decreased by 15.7% during P0 in comparison with a decrease of 5.5% (R1: 4,303 ± 567 W, R6: 4,055 ± 582 W) during P20 and a decrease of 3.3% (R1: 4,549 ± 659 W, R6: 4,363 ± 476 W) during P40. Peak force significantly decreased by 7.3% (R1: 2,861 ± 247 N, R6: 2,657 ± 225 N) during P0 in comparison with a decrease of 2.7% (R1: 2,811 ± 327 N, R6: 2,730 ± 285 N) during P20 and an increase of 0.4% (R1: 2,861 ± 323 N, R6: 2,862 ± 280 N) during P40. Peak velocity significantly decreased by 10.2% (R1: 1.97 ± 0.15 m·s(-1), R6: 1.79 ± 0.11 m·s(-1)) during P0 in comparison with a decrease of 3.8% (R1: 1.89 ± 0.13 m·s(-1), R6: 1.82 ± 0.12 m·s(-1)) during P20 and a decrease of 1.7% (R1: 1.93 ± 0.17 m·s(-1), R6: 1.89 ± 0.14 m·s(-1)) during P40. The results demonstrate that IRR periods allow for the maintenance of power in the power clean during a multiple set exercise protocol and that this may have implications for improved training adaptations.


Assuntos
Levantamento de Peso/fisiologia , Adulto , Humanos , Masculino , Força Muscular/fisiologia , Músculo Esquelético/fisiologia , Descanso/fisiologia , Adulto Jovem
19.
J Appl Physiol (1985) ; 133(1): 41-59, 2022 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-35511722

RESUMO

The strategy of gene delivery into skeletal muscles has provided exciting avenues in identifying new potential therapeutics toward muscular disorders and addressing basic research questions in muscle physiology through overexpression and knockdown studies. In vivo electroporation methodology offers a simple, rapidly effective technique for the delivery of plasmid DNA into postmitotic skeletal muscle fibers and the ability to easily explore the molecular mechanisms of skeletal muscle plasticity. The purpose of this review is to describe how to robustly electroporate plasmid DNA into different hindlimb muscles of rodent models. Furthermore, key parameters (e.g., voltage, hyaluronidase, and plasmid concentration) that contribute to the successful introduction of plasmid DNA into skeletal muscle fibers will be discussed. In addition, details on processing tissue for immunohistochemistry and fiber cross-sectional area (CSA) analysis will be outlined. The overall goal of this review is to provide the basic and necessary information needed for successful implementation of in vivo electroporation of plasmid DNA and thus open new avenues of discovery research in skeletal muscle physiology.


Assuntos
Eletroporação , Técnicas de Transferência de Genes , Animais , DNA , Eletroporação/métodos , Terapia Genética , Camundongos , Músculo Esquelético , Plasmídeos/genética
20.
J Cachexia Sarcopenia Muscle ; 13(3): 1541-1553, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35249268

RESUMO

BACKGROUND: Oxidative stress is implicated in the pathophysiology of Duchenne muscular dystrophy (DMD, caused by mutations in the dystrophin gene), which is the most common and severe of the muscular dystrophies. To our knowledge, the distribution of iron, an important modulator of oxidative stress, has not been assessed in DMD. We tested the hypotheses that iron accumulation occurs in mouse models of DMD and that modulation of iron through the diet or chelation could modify disease severity. METHODS: We assessed iron distribution and total elemental iron using LA-ICP-MS on skeletal muscle cross-sections of 8-week-old Bl10 control mice and dystrophic mdx mice (with moderate dystrophy) and dystrophin/utrophin-null mice (dko, with severe dystrophy). In addition, mdx mice (4 weeks) were treated with either an iron chelator (deferiprone 150 mg/kg/day) or iron-enriched feed (containing 1% added iron as carbonyl iron). Immunoblotting was used to determine the abundance of iron- and mitochondria-related proteins. (Immuno)histochemical and mRNA assessments of fibrosis and inflammation were also performed. RESULTS: We observed a significant increase in total elemental iron in hindlimb muscles of dko mice (+50%, P < 0.05) and in the diaphragm of mdx mice (+80%, P < 0.05), with both tissues exhibiting severe pathology. Iron dyshomeostasis was further evidenced by an increase in the storage protein ferritin (dko: +39%, P < 0.05) and ferroportin compared with Bl10 control mice (mdx: +152% and dko: +175%, P < 0.05). Despite having features of iron overload, dystrophic muscles had lower protein expression of ALAS-1, the rate-limiting enzyme for haem synthesis (dko -44%, P < 0.05), and the haem-containing protein myoglobin (dko -54%, P < 0.05). Deferiprone treatment tended to decrease muscle iron levels in mdx mice (-30%, P < 0.1), which was associated with lower oxidative stress and fibrosis, but suppressed haem-containing proteins and mitochondrial content. Increasing iron via dietary intervention elevated total muscle iron (+25%, P < 0.05) but did not aggravate the pathology. CONCLUSIONS: Muscles from dystrophic mice have increased iron levels and dysregulated iron-related proteins that are associated with dystrophic pathology. Muscle iron levels were manipulated by iron chelation and iron enriched feed. Iron chelation reduced fibrosis and reactive oxygen species (ROS) but also suppressed haem-containing proteins and mitochondrial activity. Conversely, iron supplementation increased ferritin and haem-containing proteins but did not alter ROS, fibrosis, or mitochondrial activity. Further studies are required to investigate the contribution of impaired ferritin breakdown in the dysregulation of iron homeostasis in DMD.


Assuntos
Sobrecarga de Ferro , Distrofia Muscular de Duchenne , Animais , Deferiprona , Distrofina/genética , Ferritinas , Fibrose , Heme/metabolismo , Ferro/metabolismo , Quelantes de Ferro , Sobrecarga de Ferro/etiologia , Camundongos , Camundongos Endogâmicos mdx , Distrofia Muscular de Duchenne/genética , Espécies Reativas de Oxigênio/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA