Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 15(1): 2093, 2024 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-38453931

RESUMO

Adhesions are critical for anchoring cells in their environment, as signaling platforms and for cell migration. In line with these diverse functions different types of cell-matrix adhesions have been described. Best-studied are the canonical integrin-based focal adhesions. In addition, non-canonical integrin adhesions lacking focal adhesion proteins have been discovered. These include reticular adhesions also known as clathrin plaques or flat clathrin lattices, that are enriched in clathrin and other endocytic proteins, as well as extensive adhesion networks and retraction fibers. How these different adhesion types that share a common integrin backbone are related and whether they can interconvert is unknown. Here, we identify the protein stonin1 as a marker for non-canonical αVß5 integrin-based adhesions and demonstrate by live cell imaging that canonical and non-canonical adhesions can reciprocally interconvert by the selective exchange of components on a stable αVß5 integrin scaffold. Hence, non-canonical adhesions can serve as points of origin for the generation of canonical focal adhesions.


Assuntos
Adesões Focais , Integrinas , Integrinas/metabolismo , Adesões Focais/metabolismo , Junções Célula-Matriz/metabolismo , Movimento Celular , Clatrina/metabolismo , Adesão Celular
2.
eNeuro ; 2024 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-38388443

RESUMO

The type I transmembrane protein BT-IgSF is predominantly localized in the brain and testes. It belongs to the CAR subgroup of Ig cell adhesion proteins, that are hypothesized to regulate connexin expression or localization. Here, we studied the putative link between BT-IgSF and connexins in astrocytes, ependymal cells and neurons of the mouse. Global knockout of BT-IgSF caused an increase in the clustering of connexin43 (Gja1), but not of connexin30 (Gjb6), on astrocytes and ependymal cells. Additionally, knockout animals displayed reduced expression levels of connexin43 protein in the cortex and hippocampus. Importantly, analysis of biocytin spread in hippocampal or cortical slices from mature mice of either sex revealed a decrease in astrocytic cell-cell coupling in the absence of BT-IgSF. Blocking either protein biosynthesis or proteolysis showed that the lysosomal pathway increased connexin43 degradation in astrocytes. Localization of connexin43 in subcellular compartments was not impaired in astrocytes of BT-IgSF mutants. In contrast to connexin43 the localization and expression of connexin36 (Gjd2) on neurons was not affected by the absence of BT-IgSF. Overall, our data indicate that the IgCAM BT-IgSF is essential for correct gap junction-mediated astrocyte-to-astrocyte cell communication.Significance Statement Astrocytes regulate a variety of physiological processes in the developing and adult brain that are essential for proper brain function. Astrocytes form extensive networks in the brain and communicate via gap junctions. Disruptions of gap junction coupling are found in several diseases such as neurodegeneration or epilepsy. Here, we demonstrate that the cell adhesion protein BT-IgSF is essential for gap junction mediated coupling between astrocytes in the cortex and hippocampus.

3.
Nat Commun ; 14(1): 8043, 2023 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-38114482

RESUMO

The complex neuromuscular network that controls body movements is the target of severe diseases that result in paralysis and death. Here, we report the development of a robust and efficient self-organizing neuromuscular junction (soNMJ) model from human pluripotent stem cells that can be maintained long-term in simple adherent conditions. The timely application of specific patterning signals instructs the simultaneous development and differentiation of position-specific brachial spinal neurons, skeletal muscles, and terminal Schwann cells. High-content imaging reveals self-organized bundles of aligned muscle fibers surrounded by innervating motor neurons that form functional neuromuscular junctions. Optogenetic activation and pharmacological interventions show that the spinal neurons actively instruct the synchronous skeletal muscle contraction. The generation of a soNMJ model from spinal muscular atrophy patient-specific iPSCs reveals that the number of NMJs and muscle contraction is severely affected, resembling the patient's pathology. In the future, the soNMJ model could be used for high-throughput studies in disease modeling and drug development. Thus, this model will allow us to address unmet needs in the neuromuscular disease field.


Assuntos
Células-Tronco Pluripotentes Induzidas , Atrofia Muscular Espinal , Humanos , Junção Neuromuscular/patologia , Neurônios Motores/fisiologia , Fibras Musculares Esqueléticas/patologia , Atrofia Muscular Espinal/patologia , Músculo Esquelético/patologia , Células-Tronco Pluripotentes Induzidas/patologia
4.
Methods Mol Biol ; 2640: 259-276, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36995601

RESUMO

The bHLH transcription factor MyoD is a master regulator of myogenic differentiation, and its sustained expression in fibroblasts suffices to differentiate them into muscle cells. MyoD expression oscillates in activated muscle stem cells of developing, postnatal and adult muscle under various conditions: when the stem cells are dispersed in culture, when they remain associated with single muscle fibers, or when they reside in muscle biopsies. The oscillatory period is around 3 h and thus much shorter than the cell cycle or circadian rhythm. Unstable MyoD oscillations and long periods of sustained MyoD expression are observed when stem cells undergo myogenic differentiation. The oscillatory expression of MyoD is driven by the oscillatory expression of the bHLH transcription factor Hes1 that periodically represses MyoD. Ablation of the Hes1 oscillator interferes with stable MyoD oscillations and leads to prolonged periods of sustained MyoD expression. This interferes with the maintenance of activated muscle stem cells and impairs muscle growth and repair. Thus, oscillations of MyoD and Hes1 control the balance between the proliferation and differentiation of muscle stem cells. Here, we describe time-lapse imaging methods using luciferase reporters, which can monitor dynamic MyoD gene expression in myogenic cells.


Assuntos
Fibras Musculares Esqueléticas , Proteína MyoD , Proteína MyoD/genética , Proteína MyoD/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Células-Tronco , Diferenciação Celular , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Músculo Esquelético/metabolismo
5.
J Cachexia Sarcopenia Muscle ; 13(1): 713-727, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34821076

RESUMO

BACKGROUND: Sepsis and inflammation can cause intensive care unit-acquired weakness (ICUAW). Increased interleukin-6 (IL-6) plasma levels are a risk factor for ICUAW. IL-6 signalling involves the glycoprotein 130 (gp130) receptor and the JAK/STAT-pathway, but its role in sepsis-induced muscle wasting is uncertain. In a clinical observational study, we found that the IL-6 target gene, SOCS3, was increased in skeletal muscle of ICUAW patients indicative for JAK/STAT-pathway activation. We tested the hypothesis that the IL-6/gp130-pathway mediates ICUAW muscle atrophy. METHODS: We sequenced RNA (RNAseq) from tibialis anterior (TA) muscle of cecal ligation and puncture-operated (CLP) and sham-operated wildtype (WT) mice. The effects of the IL-6/gp130/JAK2/STAT3-pathway were investigated by analysing the atrophy phenotype, gene expression, and protein contents of C2C12 myotubes. Mice lacking Il6st, encoding gp130, in myocytes (cKO) and WT controls, as well as mice treated with the JAK2 inhibitor AG490 or vehicle were exposed to CLP or sham surgery for 24 or 96 h. RESULTS: Analyses of differentially expressed genes in RNAseq (≥2-log2-fold change, P < 0.01) revealed an activation of IL-6-signalling and JAK/STAT-signalling pathways in muscle of septic mice, which occurred after 24 h and lasted at least for 96 h during sepsis. IL-6 treatment of C2C12 myotubes induced STAT3 phosphorylation (three-fold, P < 0.01) and Socs3 mRNA expression (3.1-fold, P < 0.01) and caused myotube atrophy. Knockdown of Il6st diminished IL-6-induced STAT3 phosphorylation (-30.0%; P < 0.01), Socs3 mRNA expression, and myotube atrophy. JAK2 (- 29.0%; P < 0.01) or STAT3 inhibition (-38.7%; P < 0.05) decreased IL-6-induced Socs3 mRNA expression. Treatment with either inhibitor attenuated myotube atrophy in response to IL-6. CLP-operated septic mice showed an increased STAT3 phosphorylation and Socs3 mRNA expression in TA muscle, which was reduced in septic Il6st-cKO mice by 67.8% (P < 0.05) and 85.6% (P < 0.001), respectively. CLP caused a loss of TA muscle weight, which was attenuated in Il6st-cKO mice (WT: -22.3%, P < 0.001, cKO: -13.5%, P < 0.001; WT vs. cKO P < 0.001). While loss of Il6st resulted in a reduction of MuRF1 protein contents, Atrogin-1 remained unchanged between septic WT and cKO mice. mRNA expression of Trim63/MuRF1 and Fbxo32/Atrogin-1 were unaltered between CLP-treated WT and cKO mice. AG490 treatment reduced STAT3 phosphorylation (-22.2%, P < 0.05) and attenuated TA muscle atrophy in septic mice (29.6% relative reduction of muscle weight loss, P < 0.05). The reduction in muscle atrophy was accompanied by a reduction in Fbxo32/Atrogin-1-mRNA (-81.3%, P < 0.05) and Trim63/MuRF1-mRNA expression (-77.6%, P < 0.05) and protein content. CONCLUSIONS: IL-6 via the gp130/JAK2/STAT3-pathway mediates sepsis-induced muscle atrophy possibly contributing to ICUAW.


Assuntos
Receptor gp130 de Citocina , Interleucina-6 , Janus Quinase 2 , Atrofia Muscular , Fator de Transcrição STAT3 , Sepse , Animais , Receptor gp130 de Citocina/genética , Receptor gp130 de Citocina/metabolismo , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , Janus Quinase 2/genética , Janus Quinase 2/metabolismo , Camundongos , Músculo Esquelético/fisiopatologia , Atrofia Muscular/etiologia , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Sepse/complicações , Sepse/metabolismo
6.
Exp Cell Res ; 409(2): 112933, 2021 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-34793773

RESUMO

The balance between proliferation and differentiation of muscle stem cells is tightly controlled, ensuring the maintenance of a cellular pool needed for muscle growth and repair. Muscle stem cells can proliferate, they can generate differentiating cells, or they self-renew to produce new stem cells. Notch signaling plays a crucial role in this process. Recent studies revealed that expression of the Notch effector HES1 oscillates in activated muscle stem cells. The oscillatory expression of HES1 periodically represses transcription from the genes encoding the myogenic transcription factor MYOD and the Notch ligand DLL1, thereby driving MYOD and DLL1 oscillations. This oscillatory network allows muscle progenitor cells and activated muscle stem cells to remain in a proliferative and 'undecided' state, in which they can either differentiate or self-renew. When HES1 is downregulated, MYOD oscillations become unstable and are replaced by sustained expression, which drives the cells into terminal differentiation. During development and regeneration, proliferating stem cells contact each other and the stability of the oscillatory expression depends on regular DLL1 inputs provided by neighboring cells. In such communities of cells that receive and provide Notch signals, the appropriate timing of DLL1 inputs is important, as sustained DLL1 cannot replace oscillatory DLL1. Thus, in cell communities, DLL1 oscillations ensure the appropriate balance between self-renewal and differentiation. In summary, oscillations in myogenic cells are an important example of dynamic gene expression determining cell fate.


Assuntos
Diferenciação Celular , Músculo Esquelético/citologia , Periodicidade , Receptores Notch/metabolismo , Células-Tronco/citologia , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Humanos , Ligantes , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Músculo Esquelético/metabolismo , Receptores Notch/genética , Células-Tronco/metabolismo , Fatores de Transcrição HES-1/genética , Fatores de Transcrição HES-1/metabolismo
7.
Elife ; 102021 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-34350830

RESUMO

Acute skeletal muscle injury is followed by an inflammatory response, removal of damaged tissue, and the generation of new muscle fibers by resident muscle stem cells, a process well characterized in murine injury models. Inflammatory cells are needed to remove the debris at the site of injury and provide signals that are beneficial for repair. However, they also release chemokines, reactive oxygen species, as well as enzymes for clearance of damaged cells and fibers, which muscle stem cells have to withstand in order to regenerate the muscle. We show here that MET and CXCR4 cooperate to protect muscle stem cells against the adverse environment encountered during muscle repair. This powerful cyto-protective role was revealed by the genetic ablation of Met and Cxcr4 in muscle stem cells of mice, which resulted in severe apoptosis during early stages of regeneration. TNFα neutralizing antibodies rescued the apoptosis, indicating that TNFα provides crucial cell-death signals during muscle repair that are counteracted by MET and CXCR4. We conclude that muscle stem cells require MET and CXCR4 to protect them against the harsh inflammatory environment encountered in an acute muscle injury.


Assuntos
Fator de Crescimento de Hepatócito/genética , Inflamação/fisiopatologia , Fibras Musculares Esqueléticas/fisiologia , Receptores CXCR4/genética , Regeneração , Células-Tronco/fisiologia , Animais , Fator de Crescimento de Hepatócito/metabolismo , Camundongos , Músculo Esquelético/fisiologia , Receptores CXCR4/metabolismo
9.
Nat Commun ; 12(1): 1318, 2021 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-33637744

RESUMO

Cell-cell interactions mediated by Notch are critical for the maintenance of skeletal muscle stem cells. However, dynamics, cellular source and identity of functional Notch ligands during expansion of the stem cell pool in muscle growth and regeneration remain poorly characterized. Here we demonstrate that oscillating Delta-like 1 (Dll1) produced by myogenic cells is an indispensable Notch ligand for self-renewal of muscle stem cells in mice. Dll1 expression is controlled by the Notch target Hes1 and the muscle regulatory factor MyoD. Consistent with our mathematical model, our experimental analyses show that Hes1 acts as the oscillatory pacemaker, whereas MyoD regulates robust Dll1 expression. Interfering with Dll1 oscillations without changing its overall expression level impairs self-renewal, resulting in premature differentiation of muscle stem cells during muscle growth and regeneration. We conclude that the oscillatory Dll1 input into Notch signaling ensures the equilibrium between self-renewal and differentiation in myogenic cell communities.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Diferenciação Celular/fisiologia , Desenvolvimento Muscular/fisiologia , Músculos/metabolismo , Células-Tronco/fisiologia , Animais , Proteínas de Ligação ao Cálcio/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Células HEK293 , Humanos , Camundongos , Camundongos Knockout , Desenvolvimento Muscular/genética , Mutação , Proteína MyoD/genética , Proteína MyoD/metabolismo , Transdução de Sinais/fisiologia , Fatores de Transcrição HES-1/metabolismo , Transcriptoma
10.
Front Cell Dev Biol ; 9: 634708, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33598464

RESUMO

The Arp2/3 complex generates branched actin filament networks operating in cell edge protrusion and vesicle trafficking. Here we employ a conditional knockout mouse model permitting tissue- or cell-type specific deletion of the murine Actr3 gene (encoding Arp3). A functional Actr3 gene appeared essential for fibroblast viability and growth. Thus, we developed cell lines for exploring the consequences of acute, tamoxifen-induced Actr3 deletion causing near-complete loss of functional Arp2/3 complex expression as well as abolished lamellipodia formation and membrane ruffling, as expected. Interestingly, Arp3-depleted cells displayed enhanced rather than reduced cell spreading, employing numerous filopodia, and showed little defects in the rates of random cell migration. However, both exploration of new space by individual cells and collective migration were clearly compromised by the incapability to efficiently maintain directionality of migration, while the principal ability to chemotax was only moderately affected. Examination of actin remodeling at the cell periphery revealed reduced actin turnover rates in Arp2/3-deficient cells, clearly deviating from previous sequestration approaches. Most surprisingly, induced removal of Arp2/3 complexes reproducibly increased FMNL formin expression, which correlated with the explosive induction of filopodia formation. Our results thus highlight both direct and indirect effects of acute Arp2/3 complex removal on actin cytoskeleton regulation.

11.
Development ; 147(22)2020 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-33028610

RESUMO

The Arp2/3 complex is essential for the assembly of branched filamentous actin, but its role in physiology and development is surprisingly little understood. Melanoblasts deriving from the neural crest migrate along the developing embryo and traverse the dermis to reach the epidermis, colonising the skin and eventually homing within the hair follicles. We have previously established that Rac1 and Cdc42 direct melanoblast migration in vivo We hypothesised that the Arp2/3 complex might be the main downstream effector of these small GTPases. Arp3 depletion in the melanocyte lineage results in severe pigmentation defects in dorsal and ventral regions of the mouse skin. Arp3 null melanoblasts demonstrate proliferation and migration defects and fail to elongate as their wild-type counterparts. Conditional deletion of Arp3 in primary melanocytes causes improper proliferation, spreading, migration and adhesion to extracellular matrix. Collectively, our results suggest that the Arp2/3 complex is absolutely indispensable in the melanocyte lineage in mouse development, and indicate a significant role in developmental processes that require tight regulation of actin-mediated motility.


Assuntos
Complexo 2-3 de Proteínas Relacionadas à Actina/metabolismo , Adesão Celular , Proliferação de Células , Melanócitos/metabolismo , Pigmentação da Pele , Pele/metabolismo , Complexo 2-3 de Proteínas Relacionadas à Actina/genética , Animais , Linhagem Celular , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Melanócitos/citologia , Camundongos , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Pele/citologia , Proteína cdc42 de Ligação ao GTP/genética , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteínas rac1 de Ligação ao GTP/genética , Proteínas rac1 de Ligação ao GTP/metabolismo
12.
Proc Natl Acad Sci U S A ; 117(13): 7471-7481, 2020 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-32170013

RESUMO

Eps15-homology domain containing protein 2 (EHD2) is a dynamin-related ATPase located at the neck of caveolae, but its physiological function has remained unclear. Here, we found that global genetic ablation of EHD2 in mice leads to increased lipid droplet size in fat tissue. This organismic phenotype was paralleled at the cellular level by increased fatty acid uptake via a caveolae- and CD36-dependent pathway that also involves dynamin. Concomitantly, elevated numbers of detached caveolae were found in brown and white adipose tissue lacking EHD2, and increased caveolar mobility in mouse embryonic fibroblasts. EHD2 expression itself was down-regulated in the visceral fat of two obese mouse models and obese patients. Our data suggest that EHD2 controls a cell-autonomous, caveolae-dependent fatty acid uptake pathway and imply that low EHD2 expression levels are linked to obesity.


Assuntos
Proteínas de Transporte/metabolismo , Cavéolas/metabolismo , Ácidos Graxos/metabolismo , Animais , Transporte Biológico , Membrana Celular/metabolismo , Células HeLa , Humanos , Gotículas Lipídicas/metabolismo , Metabolismo dos Lipídeos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
13.
Genes Dev ; 33(23-24): 1673-1687, 2019 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-31699777

RESUMO

Knockout of the ubiquitously expressed miRNA-17∼92 cluster in mice produces a lethal developmental lung defect, skeletal abnormalities, and blocked B lymphopoiesis. A shared target of miR-17∼92 miRNAs is the pro-apoptotic protein BIM, central to life-death decisions in mammalian cells. To clarify the contribution of miR-17∼92:Bim interactions to the complex miR-17∼92 knockout phenotype, we used a system of conditional mutagenesis of the nine Bim 3' UTR miR-17∼92 seed matches. Blocking miR-17∼92:Bim interactions early in development phenocopied the lethal lung phenotype of miR-17∼92 ablation and generated a skeletal kinky tail. In the hematopoietic system, instead of causing the predicted B cell developmental block, it produced a selective inability of B cells to resist cellular stress; and prevented B and T cell hyperplasia caused by Bim haploinsufficiency. Thus, the interaction of miR-17∼92 with a single target is essential for life, and BIM regulation by miRNAs serves as a rheostat controlling cell survival in specific physiological contexts.


Assuntos
Linfócitos B/citologia , Proteína 11 Semelhante a Bcl-2/metabolismo , Sobrevivência Celular/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Hematopoese/genética , MicroRNAs/metabolismo , Regiões 3' não Traduzidas/genética , Animais , Linfócitos B/patologia , Proteína 11 Semelhante a Bcl-2/genética , Técnicas de Inativação de Genes , Pulmão/embriologia , Camundongos , MicroRNAs/genética , Mutação , Estresse Fisiológico
14.
PLoS One ; 14(10): e0223620, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31600286

RESUMO

Endothelial nitric oxide synthase (eNOS)-related vessel relaxation is a highly coordinated process that regulates blood flow and pressure and is dependent on caveolae. Here, we investigated the role of caveolar plasma membrane stabilization by the dynamin-related ATPase EHD2 on eNOS-nitric oxide (NO)-dependent vessel relaxation. Loss of EHD2 in small arteries led to increased numbers of caveolae that were detached from the plasma membrane. Concomitantly, impaired relaxation of mesenteric arteries and reduced running wheel activity were observed in EHD2 knockout mice. EHD2 deletion or knockdown led to decreased production of nitric oxide (NO) although eNOS expression levels were not changed. Super-resolution imaging revealed that eNOS was redistributed from the plasma membrane to internalized detached caveolae in EHD2-lacking tissue or cells. Following an ATP stimulus, reduced cytosolic Ca2+ peaks were recorded in human umbilical vein endothelial cells (HUVECs) lacking EHD2. Our data suggest that EHD2-controlled caveolar dynamics orchestrates the activity and regulation of eNOS/NO and Ca2+ channel localization at the plasma membrane.


Assuntos
Vasos Sanguíneos/fisiologia , Proteínas de Transporte/metabolismo , Cavéolas/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Óxido Nítrico/metabolismo , Vasodilatação/fisiologia , Animais , Cálcio/metabolismo , Cavéolas/ultraestrutura , Membrana Celular/metabolismo , Citosol/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Artérias Mesentéricas/diagnóstico por imagem , Artérias Mesentéricas/metabolismo , Camundongos Endogâmicos C57BL , Condicionamento Físico Animal
15.
Genes Dev ; 33(9-10): 524-535, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-30862660

RESUMO

The balance between proliferation and differentiation of muscle stem cells is tightly controlled, ensuring the maintenance of a cellular pool needed for muscle growth and repair. We demonstrate here that the transcriptional regulator Hes1 controls the balance between proliferation and differentiation of activated muscle stem cells in both developing and regenerating muscle. We observed that Hes1 is expressed in an oscillatory manner in activated stem cells where it drives the oscillatory expression of MyoD. MyoD expression oscillates in activated muscle stem cells from postnatal and adult muscle under various conditions: when the stem cells are dispersed in culture, when they remain associated with single muscle fibers, or when they reside in muscle biopsies. Unstable MyoD oscillations and long periods of sustained MyoD expression are observed in differentiating cells. Ablation of the Hes1 oscillator in stem cells interfered with stable MyoD oscillations and led to prolonged periods of sustained MyoD expression, resulting in increased differentiation propensity. This interfered with the maintenance of activated muscle stem cells, and impaired muscle growth and repair. We conclude that oscillatory MyoD expression allows the cells to remain in an undifferentiated and proliferative state and is required for amplification of the activated stem cell pool.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/genética , Proteína MyoD/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Fatores de Transcrição HES-1/metabolismo , Animais , Células Cultivadas , Camundongos , Proteína MyoD/genética , Receptores Notch/metabolismo , Transdução de Sinais , Fatores de Transcrição HES-1/genética
16.
Elife ; 62017 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-28463680

RESUMO

The equilibrium between proliferation and quiescence of myogenic progenitor and stem cells is tightly regulated to ensure appropriate skeletal muscle growth and repair. The non-receptor tyrosine phosphatase Ptpn11 (Shp2) is an important transducer of growth factor and cytokine signals. Here we combined complex genetic analyses, biochemical studies and pharmacological interference to demonstrate a central role of Ptpn11 in postnatal myogenesis of mice. Loss of Ptpn11 drove muscle stem cells out of the proliferative and into a resting state during muscle growth. This Ptpn11 function was observed in postnatal but not fetal myogenic stem cells. Furthermore, muscle repair was severely perturbed when Ptpn11 was ablated in stem cells due to a deficit in stem cell proliferation and survival. Our data demonstrate a molecular difference in the control of cell cycle withdrawal in fetal and postnatal myogenic stem cells, and assign to Ptpn11 signaling a key function in satellite cell activity.


Assuntos
Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Células Satélites de Músculo Esquelético/citologia , Células Satélites de Músculo Esquelético/metabolismo , Animais , Citocinas/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Camundongos Endogâmicos C57BL , Proteína Tirosina Fosfatase não Receptora Tipo 11/genética , Transdução de Sinais
17.
Exp Cell Res ; 314(5): 1048-60, 2008 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-18061163

RESUMO

Raver1 is an hnRNP protein that interacts with the ubiquitous splicing regulator PTB and binds to cytoskeletal components like alpha-actinin and vinculin/metavinculin. Cell culture experiments suggested that raver1 functions as corepressor in PTB-regulated splicing reactions and may thereby increase proteome complexity. To determine the role of raver1 in vivo, we inactivated the gene by targeted disruption in the mouse. Here we report that raver1-deficient mice develop regularly to adulthood and show no obvious anatomical or behavioral defects. In keeping with this notion, cells from raver1-null mice were indistinguishable from wild type cells and displayed normal growth, motility, and cytoskeletal architecture in culture. Moreover, alternative splicing of exons, including the model exon 3 of alpha-tropomyosin, was not markedly changed in mutant mice, suggesting that the role of raver1 for PTB-mediated exon repression is not absolutely required to generate splice variants during mouse development. Interestingly however, loss of raver1 caused significantly reduced plasticity of synapses on acute hippocampal slices, as elicited by electrophysiological measurements of markedly lower LTP and LTD in mutant neurons. Our results provide evidence that raver1 may play an important role for the regulation of neuronal synaptic plasticity, possibly by controlling especially the late LTP via posttranscriptional mechanisms.


Assuntos
Proteínas de Transporte/fisiologia , Ribonucleoproteínas Nucleares Heterogêneas/fisiologia , Plasticidade Neuronal , Proteínas Nucleares/fisiologia , Sinapses/fisiologia , Animais , Células Cultivadas , Proteínas do Citoesqueleto/fisiologia , Eletrofisiologia , Embrião de Mamíferos , Fibroblastos , Hipocampo , Potenciação de Longa Duração , Camundongos , Camundongos Knockout , Proteínas Nucleares/deficiência , Fenótipo , Proteínas de Ligação a RNA , Ribonucleoproteínas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...