Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
NPJ Regen Med ; 9(1): 10, 2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38424446

RESUMO

Skeletal muscle function crucially depends on innervation while repair of skeletal muscle relies on resident muscle stem cells (MuSCs). However, it is poorly understood how innervation affects MuSC properties and thereby regeneration of skeletal muscle. Here, we report that loss of innervation causes precocious activation of MuSCs concomitant with the expression of markers of myogenic differentiation. This aberrant activation of MuSCs after loss of innervation is accompanied by profound alterations on the mRNA and protein level. Combination of muscle injury with loss of innervation results in impaired regeneration of skeletal muscle including shifts in myogenic populations concomitant with delayed maturation of regenerating myofibers. We further demonstrate that loss of innervation leads to alterations in myofibers and their secretome, which then affect MuSC behavior. In particular, we identify an increased secretion of Osteopontin and transforming growth factor beta 1 (Tgfb1) by myofibers isolated from mice which had undergone sciatic nerve transection. The altered secretome results in the upregulation of early activating transcription factors, such as Junb, and their target genes in MuSCs. However, the combination of different secreted factors from myofibers after loss of innervation is required to cause the alterations observed in MuSCs after loss of innervation. These data demonstrate that loss of innervation first affects myofibers causing alterations in their secretome which then affect MuSCs underscoring the importance of proper innervation for MuSC functionality and regeneration of skeletal muscle.

2.
Front Mol Neurosci ; 17: 1356326, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38419795

RESUMO

Guanosine diphosphate-mannose pyrophosphorylase B (GMPPB) catalyzes the conversion of mannose-1-phosphate and GTP to GDP-mannose, which is required as a mannose donor for the biosynthesis of glycan structures necessary for proper cellular functions. Mutations in GMPPB have been associated with various neuromuscular disorders such as muscular dystrophy and myasthenic syndromes. Here, we report that GMPPB protein abundance increases during brain and skeletal muscle development, which is accompanied by an increase in overall protein mannosylation. To model the human disorder in mice, we generated heterozygous GMPPB KO mice using CIRSPR/Cas9. While we were able to obtain homozygous KO mice from heterozygous matings at the blastocyst stage, homozygous KO embryos were absent beyond embryonic day E8.5, suggesting that the homozygous loss of GMPPB results in early embryonic lethality. Since patients with GMPPB loss-of-function manifest with neuromuscular disorders, we investigated the role of GMPPB in vitro. Thereby, we found that the siRNA-mediated knockdown of Gmppb in either primary myoblasts or the myoblast cell line C2C12 impaired myoblast differentiation and resulted in myotube degeneration. siRNA-mediated knockdown of Gmppb also impaired the neuron-like differentiation of N2A cells. Taken together, our data highlight the essential role of GMPPB during development and differentiation, especially in myogenic and neuronal cell types.

3.
Mol Ther ; 31(9): 2612-2632, 2023 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-37452493

RESUMO

Rhabdomyosarcoma is the most common pediatric soft tissue tumor, comprising two major subtypes: the PAX3/7-FOXO1 fusion-negative embryonal and the PAX3/7-FOXO1 fusion-positive alveolar subtype. Here, we demonstrate that the expression levels of the transcriptional repressor TRPS1 are specifically enhanced in the embryonal subtype, resulting in impaired terminal myogenic differentiation and tumor growth. During normal myogenesis, expression levels of TRPS1 have to decrease to allow myogenic progression, as demonstrated by overexpression of TRPS1 in myoblasts impairing myotube formation. Consequentially, myogenic differentiation in embryonal rhabdomyosarcoma in vitro as well as in vivo can be achieved by reducing TRPS1 levels. Furthermore, we show that TRPS1 levels in RD cells, the bona fide model cell line for embryonal rhabdomyosarcoma, are regulated by miR-1 and that TRPS1 and MYOD1 share common genomic binding sites. The myogenin (MYOG) promoter is one of the critical targets of TRPS1 and MYOD1; we demonstrate that TRPS1 restricts MYOG expression and thereby inhibits terminal myogenic differentiation. Therefore, reduction of TRPS1 levels in embryonal rhabdomyosarcoma might be a therapeutic approach to drive embryonal rhabdomyosarcoma cells into myogenic differentiation, thereby generating postmitotic myotubes.


Assuntos
MicroRNAs , Rabdomiossarcoma Embrionário , Humanos , Criança , Rabdomiossarcoma Embrionário/genética , Rabdomiossarcoma Embrionário/metabolismo , Rabdomiossarcoma Embrionário/patologia , Miogenina/genética , Miogenina/metabolismo , Diferenciação Celular/genética , MicroRNAs/genética , Desenvolvimento Muscular/genética , Linhagem Celular Tumoral , Proteínas Repressoras
4.
Comput Struct Biotechnol J ; 20: 6348-6359, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36420144

RESUMO

Wnt signaling is essential for embryonic development and tissue homeostasis. So far, little is known about the importance and functional relevance of the different regions in WNT proteins including regions in their C-terminus identified as hairpin and linker. However, it was shown that the C-terminus of WNT7A comprising the linker and the hairpin region is sufficient to elicit signaling. Here, we demonstrate that actually the hairpin region of WNT7A in its C-terminus is fully sufficient to induce non-canonical signaling in myogenic cells while the linker region alone did not show biological activity. Of note, all known non-canonical signaling branches of WNT7A signaling in skeletal muscle were activated by the hairpin region of WNT7A thereby inducing hypertrophy in myotubes, symmetric expansion of satellite stem cells and migration of myoblasts. Furthermore, we demonstrate that the linker region in the C-terminus of WNT7A binds to the FZD7 receptor while it does not activate non-canonical Wnt signaling. However, the hairpin and the linker region of WNT7A can activate canonical Wnt signaling independent of each other suggesting that specificity of downstream signaling might be depending on those specific regions in the C-terminus.

5.
Front Cell Dev Biol ; 9: 721543, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34676210

RESUMO

Myogenic differentiation, muscle stem cell functionality, and regeneration of skeletal muscle are cellular processes under tight control of various signaling pathways. Here, we investigated the role of non-canonical NF-κB signaling in myogenic differentiation, muscle stem cell functionality, and regeneration of skeletal muscle. We stimulated non-canonical NF-κB signaling with an agonistically acting antibody of the lymphotoxin beta receptor (LTßR). Interestingly, we found that stimulation of non-canonical NF-κB signaling through the LTßR agonist impairs myogenic differentiation, muscle stem cell function, and regeneration of skeletal muscle. Furthermore, we show that stimulation of non-canonical NF-κB signaling by the LTßR agonist coincides with activation of canonical NF-κB signaling. We suggest a direct crosstalk between canonical and non-canonical NF-κB signaling during myogenic differentiation which is required for proper myogenic differentiation and thereby regeneration of skeletal muscle.

6.
Cell Rep ; 35(10): 109223, 2021 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-34107247

RESUMO

During aging, the regenerative capacity of skeletal muscle decreases due to intrinsic changes in muscle stem cells (MuSCs) and alterations in their niche. Here, we use quantitative mass spectrometry to characterize intrinsic changes in the MuSC proteome and remodeling of the MuSC niche during aging. We generate a network connecting age-affected ligands located in the niche and cell surface receptors on MuSCs. Thereby, we reveal signaling by integrins, Lrp1, Egfr, and Cd44 as the major cell communication axes perturbed through aging. We investigate the effect of Smoc2, a secreted protein that accumulates with aging, primarily originating from fibro-adipogenic progenitors. Increased levels of Smoc2 contribute to the aberrant Integrin beta-1 (Itgb1)/mitogen-activated protein kinase (MAPK) signaling observed during aging, thereby causing impaired MuSC functionality and muscle regeneration. By connecting changes in the proteome of MuSCs to alterations of their niche, our work will enable a better understanding of how MuSCs are affected during aging.


Assuntos
Matriz Extracelular/metabolismo , Integrinas/metabolismo , Músculo Esquelético/metabolismo , Células-Tronco/metabolismo , Diferenciação Celular , Humanos
7.
J Clin Invest ; 131(9)2021 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-33755596

RESUMO

GDP-mannose-pyrophosphorylase-B (GMPPB) facilitates the generation of GDP-mannose, a sugar donor required for glycosylation. GMPPB defects cause muscle disease due to hypoglycosylation of α-dystroglycan (α-DG). Alpha-DG is part of a protein complex, which links the extracellular matrix with the cytoskeleton, thus stabilizing myofibers. Mutations of the catalytically inactive homolog GMPPA cause alacrima, achalasia, and mental retardation syndrome (AAMR syndrome), which also involves muscle weakness. Here, we showed that Gmppa-KO mice recapitulated cognitive and motor deficits. As structural correlates, we found cortical layering defects, progressive neuron loss, and myopathic alterations. Increased GDP-mannose levels in skeletal muscle and in vitro assays identified GMPPA as an allosteric feedback inhibitor of GMPPB. Thus, its disruption enhanced mannose incorporation into glycoproteins, including α-DG in mice and humans. This increased α-DG turnover and thereby lowered α-DG abundance. In mice, dietary mannose restriction beginning after weaning corrected α-DG hyperglycosylation and abundance, normalized skeletal muscle morphology, and prevented neuron degeneration and the development of motor deficits. Cortical layering and cognitive performance, however, were not improved. We thus identified GMPPA defects as the first congenital disorder of glycosylation characterized by α-DG hyperglycosylation, to our knowledge, and we have unraveled underlying disease mechanisms and identified potential dietary treatment options.


Assuntos
Distroglicanas , Guanosina Difosfato Manose , Músculo Esquelético/metabolismo , Doenças Neuromusculares , Nucleotidiltransferases/deficiência , Animais , Distroglicanas/genética , Distroglicanas/metabolismo , Glicosilação , Guanosina Difosfato Manose/genética , Guanosina Difosfato Manose/metabolismo , Humanos , Camundongos , Camundongos Knockout , Doenças Neuromusculares/dietoterapia , Doenças Neuromusculares/genética , Doenças Neuromusculares/metabolismo , Nucleotidiltransferases/metabolismo
8.
Vitam Horm ; 116: 295-311, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33752822

RESUMO

Regeneration of skeletal muscle is a finely tuned process which is depending on muscle stem cells, a population of stem cells in skeletal muscle which is also termed satellite cells. Muscle stem cells are a prerequisite for regeneration of skeletal muscle. Of note, the muscle stem cell population is heterogeneous and subpopulations can be identified depending on gene expression or phenotypic traits. However, all muscle stem cells express the transcription factor Pax7 and their functionality is tightly controlled by intrinsic signaling pathways and extrinsic signals. The latter ones include signals form the stem cell niche as well as circulating factors such as growth factors and hormones. Among them are Wnt proteins, growth factors like IGF-1 or FGF-2 and hormones such as thyroid hormones and the anti-aging hormone Klotho. A highly orchestrated interplay between those factors and muscle stem cells is important for their full functionality and ultimately regeneration of skeletal muscle as outlined here.


Assuntos
Células Satélites de Músculo Esquelético , Diferenciação Celular/fisiologia , Desenvolvimento Muscular/genética , Músculo Esquelético/metabolismo , Regeneração/fisiologia , Células Satélites de Músculo Esquelético/metabolismo , Células-Tronco
9.
J Vis Exp ; (168)2021 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-33645580

RESUMO

Adult skeletal muscle tissue harbors a stem cell population that is indispensable for its ability to regenerate. Upon muscle damage, muscle stem cells leave their quiescent state and activate the myogenic program ultimately leading to the repair of damaged tissue concomitant with the replenishment of the muscle stem cell pool. Various factors influence muscle stem cell activity, among them intrinsic stimuli but also signals from the direct muscle stem cell environment, the stem cell niche. The isolation and culture of single myofibers with their associated muscle stem cells preserves most of the interaction of the stem cell with its niche and is, therefore, the closest possibility to study muscle stem cell functionality ex vivo. Here, a protocol for the isolation, culture, siRNA transfection and immunostaining of muscle stem cells on their respective myofibers from mouse EDL (extensor digitorum longus) muscles is provided. The experimental conditions outlined here allow the study and manipulation of muscle stem cells ex vivo including investigation of myogenic activity without the inherent need for in vivo animal experiments.


Assuntos
Células-Tronco Adultas/citologia , Técnicas de Cultura de Células/métodos , Fibras Musculares Esqueléticas/citologia , Células-Tronco/citologia , Animais , Células Cultivadas , Colagenases/metabolismo , Camundongos Endogâmicos C57BL , Desenvolvimento Muscular , RNA Interferente Pequeno/metabolismo , Regeneração , Fixação de Tecidos , Transfecção
10.
Sci Transl Med ; 13(580)2021 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-33568522

RESUMO

Heart failure with preserved ejection fraction (HFpEF) is a highly prevalent and intractable form of cardiac decompensation commonly associated with diastolic dysfunction. Here, we show that diastolic dysfunction in patients with HFpEF is associated with a cardiac deficit in nicotinamide adenine dinucleotide (NAD+). Elevating NAD+ by oral supplementation of its precursor, nicotinamide, improved diastolic dysfunction induced by aging (in 2-year-old C57BL/6J mice), hypertension (in Dahl salt-sensitive rats), or cardiometabolic syndrome (in ZSF1 obese rats). This effect was mediated partly through alleviated systemic comorbidities and enhanced myocardial bioenergetics. Simultaneously, nicotinamide directly improved cardiomyocyte passive stiffness and calcium-dependent active relaxation through increased deacetylation of titin and the sarcoplasmic reticulum calcium adenosine triphosphatase 2a, respectively. In a long-term human cohort study, high dietary intake of naturally occurring NAD+ precursors was associated with lower blood pressure and reduced risk of cardiac mortality. Collectively, these results suggest NAD+ precursors, and especially nicotinamide, as potential therapeutic agents to treat diastolic dysfunction and HFpEF in humans.


Assuntos
Insuficiência Cardíaca , Animais , Estudos de Coortes , Insuficiência Cardíaca/tratamento farmacológico , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Niacinamida/farmacologia , Niacinamida/uso terapêutico , Ratos , Ratos Endogâmicos Dahl , Volume Sistólico
11.
In Vitro Cell Dev Biol Anim ; 56(8): 585-592, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32964376

RESUMO

The isolation and cultivation of intact, single myofibers presents a superior approach for studying myogenic cells in their native position. The cells' characteristics remain more similar to muscle tissue than in cell culture. Nevertheless, no routinely used method in higher vertebrates exists. Therefore, we aimed at establishing the isolation and cultivation of single myofibers from porcine muscle. For the first time, we implemented the isolation of intact myofibers from porcine fibularis tertius muscle by enzymatic digestion and their subsequent cultivation under floating conditions. Confocal microscopy showed intact myofibrill structures in isolated myofibers. Myogenic cells were able to proliferate at their parent myofiber as shown by the increase of myonuclear number during culture. Additionally, the described method can be used to investigate myogenic cells migrated from isolated myofibers. These cells expressed myogenic markers and were able to differentiate. In the future, our method can be used for genetic manipulation of cells at myofibers, investigation of growth factors or pharmacological substances, and determination of interactions between myofibers and associated cells. Working with isolated myofibers has the potential to bridge conventional cell culture and animal experiments. Adapting the method to porcine muscle allows for application possibilities in veterinary medicine as well as in biomedical research, which cannot be addressed in rodent model systems.


Assuntos
Técnicas de Cultura de Células/métodos , Separação Celular , Fibras Musculares Esqueléticas/citologia , Animais , Núcleo Celular/metabolismo , Proliferação de Células , Dissecação , Membro Posterior/fisiologia , Suínos
12.
Front Cell Dev Biol ; 8: 639, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32793592

RESUMO

Increased life expectancy in modern society comes at the cost of age-associated disabilities and diseases. Aged brains not only show reduced excitability and plasticity, but also a decline in inhibition. Age-associated defects in inhibitory circuits likely contribute to cognitive decline and age-related disorders. Molecular mechanisms that exert epigenetic control of gene expression contribute to age-associated neuronal impairments. Both DNA methylation, mediated by DNA methyltransferases (DNMTs), and histone modifications maintain neuronal function throughout lifespan. Here we provide evidence that DNMT1 function is implicated in the age-related loss of cortical inhibitory interneurons. Dnmt1 deletion in parvalbumin-positive interneurons attenuates their age-related decline in the cerebral cortex. Moreover, conditional Dnmt1-deficient mice show improved somatomotor performance and reduced aging-associated transcriptional changes. A decline in the proteostasis network, responsible for the proper degradation and removal of defective proteins, is implicated in age- and disease-related neurodegeneration. Our data suggest that DNMT1 acts indirectly on interneuron survival in aged mice by modulating the proteostasis network during life-time.

13.
EBioMedicine ; 58: 102898, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32711251

RESUMO

BACKGROUND: One-third of all deaths in hospitals are caused by sepsis. Despite its demonstrated prevalence and high case fatality rate, antibiotics remain the only target-oriented treatment option currently available. Starting from results showing that low-dose anthracyclines protect against sepsis in mice, we sought to find new causative treatment options to improve sepsis outcomes. METHODS: Sepsis was induced in mice, and different treatment options were evaluated regarding cytokine and biomarker expression, lung epithelial cell permeability, autophagy induction, and survival benefit. Results were validated in cell culture experiments and correlated with patient samples. FINDINGS: Effective low-dose epirubicin treatment resulted in substantial downregulation of the sphingosine 1-phosphate (S1P) degrading enzyme S1P lyase (SPL). Consequent accumulation and secretion of S1P in lung parenchyma cells stimulated the S1P-receptor type 3 (S1PR3) and mitogen-activated protein kinases p38 and ERK, reducing tissue damage via increased disease tolerance. The protective effects of SPL inhibition were absent in S1PR3 deficient mice. Sepsis patients showed increased expression of SPL, stable expression of S1PR3, and increased levels of mucin-1 and surfactant protein D as indicators of lung damage. INTERPRETATION: Our work highlights a tissue-protective effect of SPL inhibition in sepsis due to activation of the S1P/S1PR3 axis and implies that SPL inhibitors and S1PR3 agonists might be potential therapeutics to protect against sepsis by increasing disease tolerance against infections. FUNDING: This study was supported by the Center for Sepsis Control and Care (CSCC), the German Research Foundation (DFG), RTG 1715 (to M. H. G. and I. R.) and the National Institutes of Health, Grant R01GM043880 (to S. S.).


Assuntos
Aldeído Liases/metabolismo , Epirubicina/administração & dosagem , Sepse/tratamento farmacológico , Receptores de Esfingosina-1-Fosfato/metabolismo , Animais , Autofagia , Permeabilidade da Membrana Celular , Células Cultivadas , Modelos Animais de Doenças , Regulação para Baixo , Epirubicina/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Camundongos , Mucina-1/metabolismo , Estudos Prospectivos , Proteína D Associada a Surfactante Pulmonar/metabolismo , Distribuição Aleatória , Sepse/etiologia , Sepse/metabolismo , Receptores de Esfingosina-1-Fosfato/genética , Resultado do Tratamento , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
14.
Mech Ageing Dev ; 189: 111283, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32544406

RESUMO

Aging is characterized by a progressive decline in tissue and organ function often linked to a reduced stem cell functionality, a cell population important for regeneration. Skeletal muscle mass and regenerative capacity decrease with advancing age. Muscle stem cells, also termed satellite cells, are a prerequisite for regeneration of skeletal muscle. Their functionality declines with increasing age, driven by intrinsic changes and changes in the stem cell niche. Here, we discuss the current understanding how muscle stem cells are affected during aging. The aging associated alterations include among others upregulation of developmental pathways in aged muscle stem cells and changes in the extracellular matrix.


Assuntos
Envelhecimento/metabolismo , Matriz Extracelular/metabolismo , Músculo Esquelético/metabolismo , Células Satélites de Músculo Esquelético/metabolismo , Animais , Humanos
15.
Mol Ther Oncolytics ; 16: 134-146, 2020 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-32055677

RESUMO

Cancer cachexia is a complex metabolic disease so far lacking effective therapy, and it accounts for approximately one third of all cancer-related deaths worldwide. The extracellular ligand Wnt7a has a dual function in skeletal muscle, inducing the anabolic AKT/mammalian target of rapamycin (mTOR) pathway in myofibers and driving muscle stem cell expansion in skeletal muscle, making it a promising candidate for treatment of muscle wasting diseases. In murine and human myotubes, Wnt7a activates the anabolic AKT/mTOR pathway, thereby preventing cachexia-induced atrophy with a single application being sufficient to prevent atrophy independently of the tumor cell type causing cachexia. Addition of Wnt7a also improved activation and differentiation of muscle stem cells in cancer cachexia, a condition under which skeletal muscle regeneration is severely impaired due to stalled muscle stem cell differentiation. Finally, we show that Wnt7a prevents cancer cachexia in an in vivo mouse model based on C26 colon carcinoma cells. Wnt7a has a dual role in cachectic skeletal muscle; that is, it effectively counteracts muscle wasting through activation of the anabolic AKT/mTOR pathway and, furthermore, reverts the loss of muscle stem cell functionality due to cancer cachexia, making Wnt7a a promising candidate for an ameliorative treatment of cancer cachexia.

16.
J Vis Exp ; (151)2019 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-31609335

RESUMO

Skeletal muscle possesses an enormous capacity to regenerate after injury. This process is mainly driven by muscle stem cells, also termed satellite cells. Satellite cells are characterized by the expression of the transcription factor Pax7 and their location underneath the basal lamina in the resting skeletal muscle. Upon injury, satellite cells get activated, undergo self-renewal or differentiation to either form new myofibers or to fuse with damaged ones. The functionality of satellite cells in vivo can be investigated using a cardiotoxin based injury model of skeletal muscle. To study the function of one gene during the regeneration of skeletal muscle, transgenic mouse models are mostly used. Here, we present an alternative method to transgenic mice, to investigate the gene function in satellite cells during regeneration, e.g., in cases where transgenic mice are not available. We combine the cardiotoxin mediated injury of a specific skeletal muscle with the injection of a self-delivering siRNA into the regenerating muscle which is then taken up by satellite cells among other cells. Thereby, we provide a method to analyze gene function in satellite cells during regeneration under physiological conditions without the need for transgenic mice.


Assuntos
Cardiotoxinas/farmacologia , Músculo Esquelético/fisiologia , RNA Interferente Pequeno , Regeneração/fisiologia , Cicatrização , Animais , Diferenciação Celular , Separação Celular , Modelos Animais de Doenças , Feminino , Masculino , Camundongos , Camundongos Transgênicos , Músculo Esquelético/efeitos dos fármacos , Fator de Transcrição PAX7 , RNA Interferente Pequeno/metabolismo , Células Satélites de Músculo Esquelético/fisiologia , Venenos de Serpentes/farmacologia
18.
Cell Mol Life Sci ; 76(13): 2559-2570, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-30976839

RESUMO

Skeletal muscle regeneration is a finely tuned process involving the activation of various cellular and molecular processes. Satellite cells, the stem cells of skeletal muscle, are indispensable for skeletal muscle regeneration. Their functionality is critically modulated by intrinsic signaling pathways as well as by interactions with the stem cell niche. Here, we discuss the properties of satellite cells, including heterogeneity regarding gene expression and/or their phenotypic traits and the contribution of satellite cells to skeletal muscle regeneration. We also summarize the process of regeneration with a specific emphasis on signaling pathways, cytoskeletal rearrangements, the importance of miRNAs, and the contribution of non-satellite cells such as immune cells, fibro-adipogenic progenitor cells, and PW1-positive/Pax7-negative interstitial cells.


Assuntos
Células-Tronco Adultas/citologia , Desenvolvimento Muscular , Músculo Esquelético/citologia , Músculo Esquelético/fisiologia , Regeneração , Adulto , Diferenciação Celular , Humanos
19.
Methods Mol Biol ; 2045: 25-36, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30838602

RESUMO

The isolation and culture of single floating myofibers with their adjacent muscle stem cells allow the analysis and comparison of muscle stem cells from aged and young mice. This method has the advantage that muscle stem cells are cultured on the myofiber, thereby culturing them in conditions as close to their endogenous niche as possible. Here we describe the isolation, culture, transfection with siRNA, and subsequent immunostaining for muscle stem cells on their adjacent myofibers from aged and young mice.


Assuntos
Células-Tronco Adultas/citologia , Técnicas de Cultura de Células/métodos , Fibras Musculares Esqueléticas/citologia , Músculo Esquelético/citologia , Células-Tronco Adultas/metabolismo , Envelhecimento , Animais , Anticorpos , Diferenciação Celular , Colagenases , Imunofenotipagem , Camundongos , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Proteína MyoD/imunologia , Proteína MyoD/metabolismo , Fator de Transcrição PAX7/imunologia , Fator de Transcrição PAX7/metabolismo , RNA Interferente Pequeno , Células Satélites de Músculo Esquelético/citologia , Células Satélites de Músculo Esquelético/metabolismo , Transfecção , Fluxo de Trabalho
20.
Skelet Muscle ; 8(1): 20, 2018 07 04.
Artigo em Inglês | MEDLINE | ID: mdl-29973273

RESUMO

BACKGROUND: Klotho is a well-known anti-aging hormone, which serves as a suppressor of aging through a variety of mechanisms. Aging of skeletal muscle is concomitant with a decrease in muscle stem cell function resulting in impaired regeneration. METHODS: Here we investigate the functional role of the anti-aging hormone Klotho for muscle stem cell function after cardiotoxin-induced injury of skeletal muscle using a klotho hypomorphic mouse line, which is characterized by a premature aging phenotype. Furthermore, we perform floating single myofiber cultures with their adjacent muscle stem cells to investigate the interplay between canonical Wnt signaling and Klotho function. RESULTS: We demonstrate that muscle stem cell numbers are significantly decreased in klotho hypomorphic mice. Furthermore, we show that muscle stem cell function is also severely impaired upon loss of klotho expression, in culture and during regeneration in vivo. Moreover, we demonstrate that addition of recombinant Klotho protein inhibits aberrant excessive Wnt signaling in aged muscle stem cells thereby restoring their functionality. CONCLUSIONS: The anti-aging hormone Klotho counteracts aberrant canonical Wnt signaling in muscle stem cells and might be one of the naturally occurring inhibitors of canonical Wnt signaling in skeletal muscle.


Assuntos
Glucuronidase/fisiologia , Músculo Esquelético/fisiologia , Mioblastos Esqueléticos/fisiologia , Regeneração/fisiologia , Envelhecimento/patologia , Envelhecimento/fisiologia , Senilidade Prematura/patologia , Senilidade Prematura/fisiopatologia , Animais , Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Células Cultivadas , Glucuronidase/antagonistas & inibidores , Glucuronidase/deficiência , Glucuronidase/genética , Proteínas Klotho , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Desenvolvimento Muscular/fisiologia , Mioblastos Esqueléticos/efeitos dos fármacos , Mioblastos Esqueléticos/patologia , RNA Mensageiro/genética , Via de Sinalização Wnt/fisiologia , Proteína Wnt3A/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...