Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Commun Biol ; 3(1): 182, 2020 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-32313117

RESUMO

Understanding the signaling pathways that regulate proliferation and differentiation of muscle progenitors is essential for successful cell transplantation for treatment of Duchenne muscular dystrophy. Here, we report that a γ-secretase inhibitor, DAPT (N-[N-(3,5-difluorophenacetyl-L-alanyl)]-S-phenylglycine tertial butyl ester), which inhibits the release of NICD (Notch intercellular domain), promotes the fusion of human muscle progenitors in vitro and improves their engraftment in the tibialis anterior muscle of immune-deficient mice. Gene expression analysis revealed that DAPT severely down-regulates PTGER2, which encodes prostaglandin (PG) E2 receptor 2 (EP2), in human muscle progenitors in the differentiation condition. Functional analysis suggested that Notch signaling inhibits differentiation and promotes self-renewal of human muscle progenitors via PGE2/EP2 signaling in a cAMP/PKA-independent manner.


Assuntos
Diferenciação Celular , Proliferação de Células , Músculo Esquelético/metabolismo , Mioblastos Esqueléticos/metabolismo , Receptor Notch3/metabolismo , Receptores de Prostaglandina E Subtipo EP2/metabolismo , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Secretases da Proteína Precursora do Amiloide/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Fusão Celular , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Autorrenovação Celular , Sobrevivência Celular , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Dinoprostona/metabolismo , Dipeptídeos/farmacologia , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Humanos , Masculino , Camundongos Endogâmicos NOD , Camundongos Endogâmicos mdx , Camundongos SCID , Desenvolvimento Muscular , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patologia , Músculo Esquelético/lesões , Músculo Esquelético/patologia , Músculo Esquelético/cirurgia , Mioblastos Esqueléticos/efeitos dos fármacos , Mioblastos Esqueléticos/transplante , Receptor Notch3/genética , Receptores de Prostaglandina E Subtipo EP2/genética , Transdução de Sinais , Transplante de Células-Tronco
2.
J Cell Physiol ; 235(3): 2300-2309, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31489965

RESUMO

Niemann-Pick disease type C (NPC) is a genetic disorder in which patient cells have endosomal/lysosomal accumulation of cholesterol and sphingolipids. However, the relationship between sphingolipids and cholesterol accumulation in NPC cells has not been established. Here, we investigated the role of sphingomyelin (SM) on the accumulation of cholesterol in NPC cells. Reduction of SM by inhibition of the ceramide transfer protein CERT decreased the cholesterol accumulation in NPC cells. The accumulation of SM in NPC cells inhibited the transport of cholesterol to the endoplasmic reticulum. Overexpression of Rab9 in NPC cells reduced the cholesterol accumulation, which was recovered by treatment with SM. In NPC cells that overexpressed a Rab9 constitutively active mutant, SM treatment did not lead to the cholesterol accumulation. These results indicate that SM negatively regulates the Rab9-dependent vesicular trafficking of cholesterol, and a reduction in SM levels in NPC cells recovers the Rab9-dependent vesicular trafficking defect.


Assuntos
Transporte Biológico/fisiologia , Colesterol/metabolismo , Doença de Niemann-Pick Tipo C/metabolismo , Esfingomielinas/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo , Animais , Células CHO , Proteínas de Transporte/metabolismo , Linhagem Celular , Cricetulus , Retículo Endoplasmático/metabolismo , Glicoproteínas de Membrana/metabolismo , Transporte Proteico/fisiologia , Esfingolipídeos
3.
PLoS One ; 12(12): e0188821, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29194448

RESUMO

Duchenne muscular dystrophy (DMD) is a severe X-linked recessive muscle disorder caused by mutations in the dystrophin gene. Nonetheless, secondary processes involving perturbation of muscle regeneration probably exacerbate disease progression, resulting in the fatal loss of muscle in DMD patients. A dysfunction of undifferentiated myogenic cells is the most likely cause for the reduction of regenerative capacity of muscle. To clarify molecular mechanisms in perturbation of the regenerative capacity of DMD muscle, we have established several NCAM (CD56)-positive immortalized human dystrophic and non-dystrophic myogenic cell lines from DMD and healthy muscles. A pro-inflammatory cytokine, IL-1ß, promoted cell cycle progression of non-dystrophic myogenic cells but not DMD myogenic cells. In contrast, IL-1ß upregulated the Notch ligand Jagged1 gene in DMD myogenic cells but not in non-dystrophic myogenic cells. Knockdown of Jagged1 in DMD myogenic cells restored the IL-1ß-promoted cell cycle progression. Conversely, enforced expression of Jagged1-blocked IL-1ß promoted proliferation of non-dystrophic myogenic cells. In addition, IL-1ß prevented myogenic differentiation of DMD myogenic cells depending on Jagged1 but not of non-dystrophic myogenic cells. These results demonstrate that Jagged1 induced by IL-1ß in DMD myogenic cells modified the action of IL-1ß and reduced the ability to proliferate and differentiate. IL-1ß induced Jagged1 gene expression may be a feedback response to excess stimulation with this cytokine because high IL-1ß (200-1000 pg/ml) induced Jagged1 gene expression even in non-dystrophic myogenic cells. DMD myogenic cells are likely to acquire the susceptibility of the Jagged1 gene to IL-1ß under the microcircumstances in DMD muscles. The present results suggest that Jagged1 induced by IL-1ß plays a crucial role in the loss of muscle regeneration capacity of DMD muscles. The IL-1ß/Jagged1 pathway may be a new therapeutic target to ameliorate exacerbation of muscular dystrophy in a dystrophin-independent manner.


Assuntos
Interleucina-1beta/metabolismo , Proteína Jagged-1/metabolismo , Distrofia Muscular de Duchenne/metabolismo , Receptor Notch3/metabolismo , Diferenciação Celular , Células Cultivadas , Humanos , Desenvolvimento Muscular , Distrofia Muscular de Duchenne/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
4.
PLoS One ; 12(10): e0186584, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29049386

RESUMO

Previous studies have shown that phenotypic modulation of smooth muscle cells (SMCs) plays a pivotal role in human diseases. However, the molecular mechanisms underlying the reversible differentiation of SMCs remain elusive particularly because cultured SMCs that reproducibly exhibit bidirectional phenotypic modulation have not been established. Here we established an immortalized human bladder SMC line designated as hBS11. Under differentiation-inducing conditions, hBS11 cells underwent smooth muscle differentiation accompanied by the robust expression of smooth muscle differentiation markers and isoform-dependent reorganization of actin bundles. The cholinergic receptor agonist carbachol increased intracellular calcium in differentiated hBS11 cells in an acetylcholine muscarinic receptor-dependent manner. Differentiated hBS11 cells displayed contractile properties depending on the elevation in the levels of intracellular calcium. Depolarization of membrane potential triggered inward sodium current in differentiated hBS11 cells. However, differentiated hBS11 cells lost the differentiated phenotype and resumed mitosis when re-fed with growth medium. Our study provides direct evidence pertaining to the human bladder SMCs being able to retain the capacity of reversible differentiation and that the reorganization of actin bundles is involved in the reinstatement of contractility. Moreover, we have established a human SMC line retaining high proliferating potential without compromising differentiation potential.


Assuntos
Actinas/metabolismo , Diferenciação Celular , Músculo Liso/citologia , Bexiga Urinária/citologia , Linhagem Celular Transformada , Humanos , Músculo Liso/metabolismo , Bexiga Urinária/metabolismo
5.
Am J Pathol ; 187(12): 2674-2685, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28919112

RESUMO

Muscle dysfunction is the most important modifiable mediating factor in primary osteoarthritis (OA) because properly contracting muscles are a key absorber of forces acting on a joint. However, the pathological features of disuse muscle atrophy in OA patients have been rarely studied. Vastus medialis muscles of 14 female patients with OA (age range, 69 to 86 years), largely immobile for 1 or more years, were obtained during arthroplastic surgery and analyzed histologically. These were compared with female patients without arthritis, two with patellar fracture and two with patellar subluxation. Areas occupied by myofibers and adipose tissue were quantified. Large numbers of myofibers were lost in the vastus medialis of OA patients. The loss of myofibers was a possible cause of the reduction in muscle strength of the operated on knee. These changes were significantly correlated with an increase in intramuscular ectopic adipose tissue, and not observed in knees of nonarthritic patients. Resident platelet-derived growth factor receptor α-positive mesenchymal progenitor cells contributed to ectopic adipogenesis in vastus medialis muscles of OA patients. The present study suggests that significant loss of myofibers and ectopic adipogenesis in vastus medialis muscles are common pathological features of advanced knee OA patients with long-term loss of mobility. These changes may be related to the loss of joint function in patients with knee OA.


Assuntos
Tecido Adiposo , Coristoma/patologia , Transtornos Musculares Atróficos/patologia , Osteoartrite/complicações , Músculo Quadríceps/patologia , Adipogenia/fisiologia , Idoso , Idoso de 80 Anos ou mais , Feminino , Humanos , Transtornos Musculares Atróficos/etiologia
6.
PLoS One ; 12(7): e0181457, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28715470

RESUMO

Alendronate, a nitrogen-containing bisphosphonate, is well established as a treatment for osteoporosis through regulation of osteoclast activity. Previously, the pharmacological effects of bisphosphonates on cells outside the bone environment have been considered irrelevant because bisphosphonates target bone. Here we show that administration of alendronate impairs muscle regeneration in mice after bone fracture. A series of injections of alendronate alone or bone fracture alone did not affect muscle regeneration induced by cold injury. In contrast, alendronate treatment plus bone fracture severely impaired the regeneration of muscle that closely contacts the bone fracture site after cold injury. After cold injury, M-cadherin-positive myogenic cells disappeared in the damaged muscle areas of mice receiving the combination of alendronate treatment and bone fracture. The present results suggest that the muscle regeneration capacity is impaired by bone fracture in mice receiving alendronate treatment. The present research on the pharmacological effects of alendronate on muscle regeneration will aid in understanding of the in vivo action of alendronate on skeletal muscles.


Assuntos
Alendronato/farmacologia , Conservadores da Densidade Óssea/farmacologia , Lesão por Frio/fisiopatologia , Fraturas Ósseas/terapia , Músculo Esquelético/fisiopatologia , Regeneração/fisiologia , Alendronato/efeitos adversos , Animais , Conservadores da Densidade Óssea/efeitos adversos , Lesão por Frio/patologia , Lesão por Frio/terapia , Modelos Animais de Doenças , Fraturas Ósseas/patologia , Fraturas Ósseas/fisiopatologia , Masculino , Camundongos Endogâmicos ICR , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/patologia , Regeneração/efeitos dos fármacos
7.
Sci Rep ; 7: 46126, 2017 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-28555643

RESUMO

Duchenne muscular dystrophy (DMD) is a fatal progressive muscle-wasting disease. Various attempts are underway to convert severe DMD to a milder phenotype by modulating the splicing of the dystrophin gene and restoring its expression. In our previous study, we reported TG003, an inhibitor of CDC2-like kinase 1 (CLK1), as a splice-modifying compound for exon-skipping therapy; however, its metabolically unstable feature hinders clinical application. Here, we show an orally available inhibitor of CLK1, named TG693, which promoted the skipping of the endogenous mutated exon 31 in DMD patient-derived cells and increased the production of the functional exon 31-skipped dystrophin protein. Oral administration of TG693 to mice inhibited the phosphorylation of serine/arginine-rich proteins, which are the substrates of CLK1, and modulated pre-mRNA splicing in the skeletal muscle. Thus, TG693 is a splicing modulator for the mutated exon 31 of the dystrophin gene in vivo, possibly possessing therapeutic potential for DMD patients.


Assuntos
Distrofina/genética , Inibidores Enzimáticos/uso terapêutico , Éxons/genética , Distrofia Muscular de Duchenne/tratamento farmacológico , Distrofia Muscular de Duchenne/genética , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/antagonistas & inibidores , Tiazóis/uso terapêutico , Administração Oral , Animais , Inibidores Enzimáticos/administração & dosagem , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacocinética , Genes Reporter , Células HeLa , Humanos , Masculino , Camundongos , Músculo Esquelético/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Tirosina Quinases/metabolismo , Splicing de RNA/genética , Tiazóis/administração & dosagem , Tiazóis/química , Tiazóis/farmacocinética
8.
Int J Urol ; 24(6): 461-467, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28332237

RESUMO

OBJECTIVES: To examine the inhibitory effects of tumor necrosis factor-α on myogenic differentiation of human urethral rhabdosphincter cells. METHODS: A rhabdosphincter sample was obtained from a patient who underwent total cystectomy. To expand the lifespan of the primary cultured cells, rhabdosphincter myogenic cells were immortalized with mutated cyclin-dependent kinase 4, cyclin D1 and telomerase. The differential potential of the cells was investigated. The transfected human rhabdosphincter cells were induced for myogenic differentiation with recombinant human tumor necrosis factor-α and/or the tumor necrosis factor-α antagonist etanercept at different concentrations, and activation of signaling pathways was monitored. RESULTS: Human rhabdosphincter cells were selectively cultured for at least 40 passages. Molecular analysis confirmed the expression of myosin heavy chain, which is a specific marker of differentiated muscle cells, significantly increased after differentiation induction. Although tumor necrosis factor-α treatment reduced the myosin heavy chain expression in a concentration-dependent manner, etanercept inhibited this suppression. Tumor necrosis factor-α suppressed phosphorylation of protein kinase B and p38, whereas etanercept pretreatment promoted phosphorylation and myosin heavy chain expression in a concentration-dependent manner. CONCLUSIONS: Tumor necrosis factor-α inhibits differentiation of urethral rhabdosphincter cells in part through the p38 mitogen-activated protein kinase and phosphoinositide 3-kinase pathways. Inhibition of tumor necrosis factor-α might be a useful strategy to treat stress urinary incontinence.


Assuntos
Diferenciação Celular/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Mioblastos Esqueléticos/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Cistectomia , Etanercepte/farmacologia , Etanercepte/uso terapêutico , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Pessoa de Meia-Idade , Desenvolvimento Muscular/efeitos dos fármacos , Desenvolvimento Muscular/fisiologia , Músculo Esquelético/citologia , Mioblastos Esqueléticos/efeitos dos fármacos , Cadeias Pesadas de Miosina/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/efeitos dos fármacos , Cultura Primária de Células , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/isolamento & purificação , Uretra/citologia , Uretra/efeitos dos fármacos , Uretra/fisiologia , Neoplasias da Bexiga Urinária/cirurgia , Incontinência Urinária por Estresse/tratamento farmacológico , Incontinência Urinária por Estresse/fisiopatologia
9.
Int J Med Sci ; 13(9): 680-5, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27647997

RESUMO

Effects of myostatin (MSTN)-suppression on the regeneration of injured skeletal muscle under unloading condition were investigated by using transgenic mice expressing a dominant-negative form of MSTN (MSTN-DN). Both MSTN-DN and wild-type (WT) mice were subjected to continuous hindlimb suspension (HS) for 6 weeks. Cardiotoxin (CTX) was injected into left soleus muscle under anesthesia 2 weeks after the initiation of HS. Then, the soleus muscles were excised following 6-week HS (4 weeks after CTX-injection). CTX-injection caused to reduce the soleus fiber cross-sectional area (CSA) in WT mice under both unloading and weight-bearing conditions, but not in MSTN-DN mice. Under unloading condition, CTX-injected muscle weight and fiber CSA in MSTN-DN mice were significantly higher than those in WT mice. CTX-injected muscle had many damaged and regenerating fibers having central nuclei in both WT and MSTN-DN mice. Significant increase in the population of Pax7-positive nuclei in CTX-injected muscle was observed in MSTN-DN mice, but not in WT mice. Evidences indicate that the suppression of MSTN cause to increase the regenerative potential of injured soleus muscle via the increase in the population of muscle satellite cells regardless of unloading conditions.


Assuntos
Membro Posterior/crescimento & desenvolvimento , Músculo Esquelético/crescimento & desenvolvimento , Miostatina/biossíntese , Regeneração , Animais , Cardiotoxinas/administração & dosagem , Membro Posterior/efeitos dos fármacos , Membro Posterior/lesões , Membro Posterior/fisiopatologia , Humanos , Camundongos , Fibras Musculares Esqueléticas/efeitos dos fármacos , Fibras Musculares Esqueléticas/patologia , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/lesões , Músculo Esquelético/fisiopatologia , Miostatina/antagonistas & inibidores , Células Satélites de Músculo Esquelético/metabolismo , Células Satélites de Músculo Esquelético/patologia , Suporte de Carga
10.
Nucleic Acids Res ; 44(21): e158, 2016 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-27566152

RESUMO

Myoblasts are precursor skeletal muscle cells that differentiate into fused, multinucleated myotubes. Current single-cell microfluidic methods are not optimized for capturing very large, multinucleated cells such as myotubes. To circumvent the problem, we performed single-nucleus transcriptome analysis. Using immortalized human myoblasts, we performed RNA-seq analysis of single cells (scRNA-seq) and single nuclei (snRNA-seq) and found them comparable, with a distinct enrichment for long non-coding RNAs (lncRNAs) in snRNA-seq. We then compared snRNA-seq of myoblasts before and after differentiation. We observed the presence of mononucleated cells (MNCs) that remained unfused and analyzed separately from multi-nucleated myotubes. We found that while the transcriptome profiles of myoblast and myotube nuclei are relatively homogeneous, MNC nuclei exhibited significant heterogeneity, with the majority of them adopting a distinct mesenchymal state. Primary transcripts for microRNAs (miRNAs) that participate in skeletal muscle differentiation were among the most differentially expressed lncRNAs, which we validated using NanoString. Our study demonstrates that snRNA-seq provides reliable transcriptome quantification for cells that are otherwise not amenable to current single-cell platforms. Our results further indicate that snRNA-seq has unique advantage in capturing nucleus-enriched lncRNAs and miRNA precursors that are useful in mapping and monitoring differential miRNA expression during cellular differentiation.


Assuntos
Diferenciação Celular/genética , Mioblastos/citologia , Análise de Sequência de RNA/métodos , Linhagem Celular , Núcleo Celular/genética , Regulação da Expressão Gênica , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/fisiologia , MicroRNAs/genética , Fibras Musculares Esqueléticas/citologia , Mioblastos/fisiologia , Fator Regulador Miogênico 5/genética , RNA Longo não Codificante , Análise de Célula Única/métodos
11.
Neuropharmacology ; 110(Pt A): 458-469, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27539961

RESUMO

Sphingolipids such as sphingomyelin and glycosphingolipids (GSLs) derived from glucosylceramide (GlcCer), in addition to cholesterol, accumulate in cells/neurons in Niemann-Pick disease type C (NPC). The activities of acid sphingomyelinase and lysosomal glucocerebrosidase (GCase), which degrade sphingomyelin and GlcCer, respectively, are down-regulated in NPC cells, however, changes in GlcCer synthase activity have not yet been elucidated. We herein demonstrated for the first time that GlcCer synthase activity for the fluorescent ceramide, 4-nitrobenzo-2-oxa-1,3-diazole-labeled C6-ceramide (NBD-ceramide) increased in intact NPC1((-/-)) cells and cell lysates without affecting the protein levels. In NBD-ceramide-labeled NPC1((-/-)) cells, NBD-fluorescence preferentially accumulated in the Golgi complex and vesicular specks in the cytoplasm 40 and 150 min, respectively, after labeling, while a treatment for 48 h with the GlcCer synthase inhibitors, N-butyldeoxynojirimycin (NB-DNJ) and 1-phenyl-2-palmitoylamino-3-morpholino-1-propanol, accelerated the appearance of vesicular specks emitting NBD-fluorescence within 40 min. The treatment of NPC1((-/-)) cells with NB-DNJ for 48 h additionally increased the levels of cholesterol, but not those of sphingomyelin. Increases in the activity of GlcCer synthase and formation of vesicular specks emitting NBD-fluorescence in NPC1((-/-)) cells were dependent on cholesterol. LacCer taken up by endocytosis, which accumulated in the Golgi complex in normal cells, accumulated in vesicular specks after 10 and 40 min in NPC1((-/-)) cells, and this response was not accelerated by the NB-DNJ treatment, but was restored by the depletion of cholesterol. The cellular roles for enhanced GlcCer synthesis and increased levels of cholesterol in the trafficking of NBD-ceramide metabolites in NPC1((-/-)) cells have been discussed.


Assuntos
Transporte Biológico/fisiologia , Colesterol/metabolismo , Glucosilceramidas/metabolismo , Glucosiltransferases/metabolismo , Doença de Niemann-Pick Tipo C/metabolismo , Animais , Transporte Biológico/efeitos dos fármacos , Células CHO , Cricetulus , Endocitose/efeitos dos fármacos , Endocitose/fisiologia , Glucosiltransferases/antagonistas & inibidores , Complexo de Golgi/efeitos dos fármacos , Complexo de Golgi/metabolismo , Glicoproteínas de Membrana/deficiência , Glicoproteínas de Membrana/genética , Esfingomielinas/metabolismo
12.
PLoS Genet ; 12(7): e1006167, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27415617

RESUMO

Cell differentiation status is defined by the gene expression profile, which is coordinately controlled by epigenetic mechanisms. Cell type-specific DNA methylation patterns are established by chromatin modifiers including de novo DNA methyltransferases, such as Dnmt3a and Dnmt3b. Since the discovery of the myogenic master gene MyoD, myogenic differentiation has been utilized as a model system to study tissue differentiation. Although knowledge about myogenic gene networks is accumulating, there is only a limited understanding of how DNA methylation controls the myogenic gene program. With an aim to elucidate the role of DNA methylation in muscle development and regeneration, we investigate the consequences of mutating Dnmt3a in muscle precursor cells in mice. Pax3 promoter-driven Dnmt3a-conditional knockout (cKO) mice exhibit decreased organ mass in the skeletal muscles, and attenuated regeneration after cardiotoxin-induced muscle injury. In addition, Dnmt3a-null satellite cells (SCs) exhibit a striking loss of proliferation in culture. Transcriptome analysis reveals dysregulated expression of p57Kip2, a member of the Cip/Kip family of cyclin-dependent kinase inhibitors (CDKIs), in the Dnmt3a-KO SCs. Moreover, RNAi-mediated depletion of p57Kip2 replenishes the proliferation activity of the SCs, thus establishing a role for the Dnmt3a-p57Kip2 axis in the regulation of SC proliferation. Consistent with these findings, Dnmt3a-cKO muscles exhibit fewer Pax7+ SCs, which show increased expression of p57Kip2 protein. Thus, Dnmt3a is found to maintain muscle homeostasis by epigenetically regulating the proliferation of SCs through p57Kip2.


Assuntos
Inibidor de Quinase Dependente de Ciclina p57/metabolismo , DNA (Citosina-5-)-Metiltransferases/metabolismo , Desenvolvimento Muscular , Músculo Esquelético/metabolismo , Animais , Diferenciação Celular , Proliferação de Células , Metilação de DNA , DNA Metiltransferase 3A , Epigênese Genética , Feminino , Deleção de Genes , Perfilação da Expressão Gênica , Masculino , Camundongos , Camundongos Knockout , Proteína MyoD/metabolismo , Mioblastos/metabolismo , Fator de Transcrição PAX7/metabolismo , Regiões Promotoras Genéticas , Interferência de RNA , Regeneração , Tomografia Computadorizada por Raios X
13.
J Cachexia Sarcopenia Muscle ; 7(3): 366-76, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27239414

RESUMO

BACKGROUND: Muscle wasting during cancer cachexia contributes to patient morbidity. Cachexia-induced muscle damage may be understood by comparing its symptoms with those of other skeletal muscle diseases, but currently available data are limited. METHODS: We modelled cancer cachexia in mice bearing Lewis lung carcinoma/colon adenocarcinoma and compared the associated muscle damage with that in a murine muscular dystrophy model (mdx mice). We measured biochemical and immunochemical parameters: amounts/localization of cytoskeletal proteins and/or Ca(2+) signalling proteins related to muscle function and abnormality. We analysed intracellular Ca(2+) mobilization and compared results between the two models. Involvement of Ca(2+)-permeable channel transient receptor potential vanilloid 2 (TRPV2) was examined by inoculating Lewis lung carcinoma cells into transgenic mice expressing dominant-negative TRPV2. RESULTS: Tumourigenesis caused loss of body and skeletal muscle weight and reduced muscle force and locomotor activity. Similar to mdx mice, cachexia muscles exhibited myolysis, reduced sarcolemmal sialic acid content, and enhanced lysosomal exocytosis and sarcolemmal localization of phosphorylated Ca(2+)/CaMKII. Abnormal autophagy and degradation of dystrophin also occurred. Unlike mdx muscles, cachexia muscles did not exhibit regeneration markers (centrally nucleated fibres), and levels of autophagic proteolytic pathway markers increased. While a slight accumulation of TRPV2 was observed in cachexia muscles, Ca(2+) influx via TRPV2 was not elevated in cachexia-associated myotubes, and the course of cachexia pathology was not ameliorated by dominant-negative inhibition of TRPV2. CONCLUSIONS: Thus, cancer cachexia may induce muscle damage through TRPV2-independent mechanisms distinct from those in muscular dystrophy; this may help treat patients with tumour-induced muscle wasting.

14.
Brain Dev ; 38(8): 738-45, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27021413

RESUMO

BACKGROUND: Antisense oligonucleotides that induce exon skipping have been nominated as the most plausible treatment method for dystrophin expression in dystrophin-deficient Duchenne muscular dystrophy. Considering this therapeutic efficiency, small chemical compounds that can enable exon skipping have been highly awaited. In our previous report, a small chemical kinase inhibitor, TG003, was shown to enhance dystrophin expression by enhancing exon skipping. PURPOSE: Staurosporine (STS), a small chemical broad kinase inhibitor, was examined for enhanced skipping of a nonsense-encoding dystrophin exon. METHODS: STS was added to culture medium of HeLa cells transfected with minigenes expressing wild-type or mutated exon 31 with c.4303G>T (p.Glu1435X), and the resulting mRNAs were analyzed by RT-PCR amplification. Dystrophin mRNA and protein were analyzed in muscle cells treated with STS by RT-PCR and western blotting, respectively. RESULTS: STS did not alter splicing of the wild-type minigene. In the mutated minigene, STS increased the exon 31-skipped product. A combination of STS and TG003 did not significantly increase the exon 31-skipped product. STS enhanced skipping of exon 4 of the CDC-like kinase 1 gene, whereas TG003 suppressed it. Two STS analogs with selective kinase inhibitory activity did not enhance the mutated exon 31 skipping. When immortalized muscle cells with c.4303G>T in the dystrophin gene were treated with STS, skipping of the mutated exon 31 and dystrophin expression was enhanced. CONCLUSIONS: STS, a broad kinase inhibitor, was shown to enhance skipping of the mutated exon 31 and dystrophin expression, but selective kinase inhibitors did not.


Assuntos
Códon sem Sentido , Distrofina/metabolismo , Éxons , Estaurosporina/metabolismo , Desmina/metabolismo , Células HeLa , Humanos , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Tirosina Quinases/metabolismo , Transfecção
15.
Stem Cells ; 33(8): 2456-68, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25917344

RESUMO

Sarcopenia, age-related muscle weakness, increases the frequency of falls and fractures in elderly people, which can trigger severe muscle injury. Rapid and successful recovery from muscle injury is essential not to cause further frailty and loss of independence. In fact, we showed insufficient muscle regeneration in aged mice. Although the number of satellite cells, muscle stem cells, decreases with age, the remaining satellite cells maintain the myogenic capacity equivalent to young mice. Transplantation of young green fluorescent protein (GFP)-Tg mice-derived satellite cells into young and aged mice revealed that age-related deterioration of the muscle environment contributes to the decline in regenerative capacity of satellite cells. Thus, extrinsic changes rather than intrinsic changes in satellite cells appear to be a major determinant of inefficient muscle regeneration with age. Comprehensive protein expression analysis identified a decrease in insulin-like growth factor-II (IGF-II) level in regenerating muscle of aged mice. We found that pro- and big-IGF-II but not mature IGF-II specifically express during muscle regeneration and the expressions are not only delayed but also decreased in absolute quantity with age. Supplementation of pro-IGF-II in aged mice ameliorated the inefficient regenerative response by promoting proliferation of satellite cells, angiogenesis, and suppressing adipogenic differentiation of platelet derived growth factor receptor (PDGFR)α(+) mesenchymal progenitors. We further revealed that pro-IGF-II but not mature IGF-II specifically inhibits the pathological adipogenesis of PDGFRα(+) cells. Together, these results uncovered a distinctive pro-IGF-II-mediated self-reinforcement mechanism of muscle regeneration and suggest that supplementation of pro-IGF-II could be one of the most effective therapeutic approaches for muscle injury in elderly people.


Assuntos
Envelhecimento/metabolismo , Fator de Crescimento Insulin-Like II/metabolismo , Músculo Esquelético/fisiologia , Precursores de Proteínas/metabolismo , Regeneração/fisiologia , Células Satélites de Músculo Esquelético/metabolismo , Envelhecimento/genética , Animais , Fator de Crescimento Insulin-Like II/genética , Camundongos , Camundongos Knockout , Precursores de Proteínas/genética
16.
J Cell Biochem ; 116(9): 1898-907, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25716287

RESUMO

We examined the effect of the cellular sphingolipid level on the release of arachidonic acid (AA) and the activity of secretory phospholipase A2 (sPLA2 ) using two Chinese hamster ovary (CHO)-K1 cell mutants, LY-B and LY-A cells, deficient in sphingolipid synthesis. In LY-B cells, deficiency of sphingolipids enhanced the release of AA induced by bee venom sPLA2-III or human sPLA2-V. These alterations were reversed by replenishment of exogenous sphingomyelin (SM). In LY-A cells, deficiency of SM increased the release of AA induced by sPLA2. In CHO-K1 cells, decrease and increase of SM level in the plasma membrane by pharmacological methods increased and inhibited the release of AA, respectively. SM inhibited the activity of sPLA2 in vitro. Niemann-Pick disease type C (NPC) is a lysosomal storage disorder caused by mutation of either the NPC1 or NPC2 gene, and is characterized by accumulation of cholesterol and sphingolipids including SM in late endosomes/lysosomes. Increased levels of AA and sPLA2 activity are involved in various neurodegenerative diseases. In CHO cells lacking NPC1 (A101 cells), SM level was lower in the plasma membrane, while it was higher in late endosomes/lysosomes. The release of AA induced by sPLA2 was increased in A101 cells than that in parental cells (JP17 cells), which was attenuated by adding exogenous SM. In addition, sPLA2 -III-induced cytotoxicity in A101 cells was much higher than that in JP17 cells. These results suggest that SM in the plasma membrane plays important roles in regulating sPLA2 activity and the enzyme-induced cytotoxicity in A101 cells.


Assuntos
Ácido Araquidônico/biossíntese , Membrana Celular/metabolismo , Doença de Niemann-Pick Tipo C/enzimologia , Fosfolipases A2 Secretórias/metabolismo , Esfingomielinas/metabolismo , Animais , Células CHO , Cricetinae , Cricetulus , Fosfolipases A2 do Grupo III/metabolismo , Fosfolipases A2 do Grupo III/farmacologia , Fosfolipases A2 do Grupo V/metabolismo , Fosfolipases A2 do Grupo V/farmacologia , Humanos , Glicoproteínas de Membrana/deficiência , Modelos Biológicos , Fosfolipases A2 Secretórias/farmacologia , Esfingomielinas/deficiência
18.
Development ; 142(1): 51-61, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25480916

RESUMO

Muscle satellite cells are indispensable for muscle regeneration, but the functional diversity of their daughter cells is unknown. Here, we show that many Pax7(+)MyoD(-) cells locate both beneath and outside the basal lamina during myofiber maturation. A large majority of these Pax7(+)MyoD(-) cells are not self-renewed satellite cells, but have different potentials for both proliferation and differentiation from Pax7(+)MyoD(+) myoblasts (classical daughter cells), and are specifically marked by expression of the doublecortin (Dcx) gene. Transplantation and lineage-tracing experiments demonstrated that Dcx-expressing cells originate from quiescent satellite cells and that the microenvironment induces Dcx in myoblasts. Expression of Dcx seems to be necessary for myofiber maturation because Dcx-deficient mice exhibited impaired myofiber maturation resulting from a decrease in the number of myonuclei. Furthermore, in vitro and in vivo studies suggest that one function of Dcx in myogenic cells is acceleration of cell motility. These results indicate that Dcx is a new marker for the Pax7(+)MyoD(-) subpopulation, which contributes to myofiber maturation during muscle regeneration.


Assuntos
Diferenciação Celular , Proteínas Associadas aos Microtúbulos/metabolismo , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/fisiologia , Neuropeptídeos/metabolismo , Regeneração/fisiologia , Células-Tronco/citologia , Animais , Cardiotoxinas/administração & dosagem , Movimento Celular , Microambiente Celular , Proteínas do Domínio Duplacortina , Proteína Duplacortina , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Associadas aos Microtúbulos/deficiência , Proteína MyoD/metabolismo , Mioblastos/citologia , Mioblastos/metabolismo , Neuropeptídeos/deficiência , Fator de Transcrição PAX7/metabolismo , Células Satélites de Músculo Esquelético/citologia , Células-Tronco/metabolismo
20.
PLoS One ; 9(12): e114816, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25536520

RESUMO

The mechanisms responsible for active DNA demethylation remain elusive in Metazoa. A previous study that utilized zebrafish embryos provided a potent mechanism for active demethylation in which three proteins, AID, MBD4, and GADD45 are involved. We recently found age-dependent DNA hypomethylation in zebrafish, and it prompted us to examine if AID and MBD4 could be involved in the phenomenon. Unexpectedly, however, we found that most of the findings in the previous study were not reproducible. First, the injection of a methylated DNA fragment into zebrafish eggs did not affect either the methylation of genomic DNA, injected methylated DNA itself, or several loci tested or the expression level of aid, which has been shown to play a role in demethylation. Second, aberrant methylation was not observed at certain CpG islands following the injection of antisense morpholino oligonucleotides against aid and mbd4. Furthermore, we demonstrated that zebrafish MBD4 cDNA lacked a coding region for the methyl-CpG binding domain, which was assumed to be necessary for guidance to target regions. Taken together, we concluded that there is currently no evidence to support the proposed roles of AID and MBD4 in active demethylation in zebrafish embryos.


Assuntos
Citidina Desaminase/metabolismo , DNA Glicosilases/metabolismo , Metilação de DNA/genética , Embrião não Mamífero/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Processamento Alternativo/efeitos dos fármacos , Processamento Alternativo/genética , Animais , Ilhas de CpG/genética , Citidina Desaminase/genética , DNA Glicosilases/genética , Metilação de DNA/efeitos dos fármacos , Embrião não Mamífero/efeitos dos fármacos , Fertilização/genética , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Genoma , Injeções , Morfolinos/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Óvulo/efeitos dos fármacos , Óvulo/metabolismo , Estrutura Terciária de Proteína , Transcrição Gênica/efeitos dos fármacos , Proteínas de Peixe-Zebra/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...