Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Dev Cell ; 58(24): 2896-2913.e6, 2023 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-38056454

RESUMO

Skeletal muscle repair relies on heterogeneous populations of satellite cells (SCs). The mechanisms that regulate SC homeostasis and state transition during activation are currently unknown. Here, we investigated the emerging role of non-genetic micro-heterogeneity, i.e., intrinsic cell-to-cell variability of a population, in this process. We demonstrate that micro-heterogeneity of the membrane protein CRIPTO in mouse-activated SCs (ASCs) identifies metastable cell states that allow a rapid response of the population to environmental changes. Mechanistically, CRIPTO micro-heterogeneity is generated and maintained through a process of intracellular trafficking coupled with active shedding of CRIPTO from the plasma membrane. Irreversible perturbation of CRIPTO micro-heterogeneity affects the balance of proliferation, self-renewal, and myogenic commitment in ASCs, resulting in increased self-renewal in vivo. Our findings demonstrate that CRIPTO micro-heterogeneity regulates the adaptative response of ASCs to microenvironmental changes, providing insights into the role of intrinsic heterogeneity in preserving stem cell population diversity during tissue repair.


Assuntos
Células Satélites de Músculo Esquelético , Animais , Camundongos , Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Músculo Esquelético/metabolismo , Células Satélites de Músculo Esquelético/metabolismo , Células-Tronco
2.
Front Psychol ; 14: 1233176, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37901077

RESUMO

The speech of individuals with schizophrenia exhibits atypical prosody and pragmatic dysfunctions, producing monotony. The paper presents the outcomes of corpus-based research on the prosodic features of the pathology as they manifest in real-life spontaneous interactions. The research relies on a corpus of schizophrenic speech recorded during psychiatric interviews (CIPPS) compared to a sampling of non-pathological speech derived from the LABLITA corpus of spoken Italian, which has been selected according to comparability requirements. Corpora has been intensively analyzed in the Language into Act Theory (L-AcT) frame, which links prosodic cues and pragmatic values. A cluster of linguistic parameters marked by prosody has been considered: utterance boundaries, information structure, speech disfluency, and prosodic prominence. The speech flow of patients turns out to be organized into small chunks of information that are shorter and scarcely structured, with an atypical proportion of post-nuclear information units (Appendix). It is pervasively scattered with silences, especially with long pauses between utterances and long silences at turn-taking. Fluency is hindered by retracing phenomena that characterize complex information structures. The acoustic parameters that give rise to prosodic prominence (f0 mean, f0 standard deviation, spectral emphasis, and intensity variation) have been measured considering the pragmatic roles of the prosodic units, distinguishing prominences within the illocutionary units (Comment) from those characterizing Topic units. Patients show a flattening of the Comment-prominence, reflecting impairments in performing the illocutionary activity. Reduced values of spectral emphasis and intensity variation also suggest a lack of engagement in communication. Conversely, Topic-prominence shows higher values for f0 standard deviation and spectral emphasis, suggesting effort when defining the domain of relevance of the illocutionary force. When comparing Topic and Comment-prominences of patients, the former consistently exhibit higher values across all parameters. In contrast, the non-pathological group displays the opposite pattern.

3.
Int J Mol Sci ; 24(15)2023 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-37569832

RESUMO

Duchenne muscular dystrophy (DMD) is a muscle disease caused by mutations in the dystrophin gene characterized by myofiber fragility and progressive muscle degeneration. The genetic defect results in a reduced number of self-renewing muscle stem cells (MuSCs) and an impairment of their activation and differentiation, which lead to the exhaustion of skeletal muscle regeneration potential and muscle replacement by fibrotic and fatty tissue. In this study, we focused on an unexplored strategy to improve MuSC function and to preserve their niche based on the regenerative properties of mesenchymal stromal cells from the amniotic membrane (hAMSCs), that are multipotent cells recognized to have a role in tissue repair in different disease models. We demonstrate that the hAMSC secretome (CM hAMSC) and extracellular vesicles (EVs) isolated thereof directly stimulate the in vitro proliferation and differentiation of human myoblasts and mouse MuSC from dystrophic muscles. Furthermore, we demonstrate that hAMSC secreted factors modulate the muscle stem cell niche in dystrophic-mdx-mice. Interestingly, local injection of EV hAMSC in mdx muscles correlated with an increase in the number of activated Pax7+/Ki67+ MuSCs and in new fiber formation. EV hAMSCs also significantly reduced muscle collagen deposition, thus counteracting fibrosis and MuSCs exhaustion, two hallmarks of DMD. Herein for the first time we demonstrate that CM hAMSC and EVs derived thereof promote muscle regeneration by supporting proliferation and differentiation of resident muscle stem cells. These results pave the way for the development of a novel treatment to counteract DMD progression by reducing fibrosis and enhancing myogenesis in dystrophic muscles.


Assuntos
Vesículas Extracelulares , Células-Tronco Mesenquimais , Distrofia Muscular de Duchenne , Células Satélites de Músculo Esquelético , Humanos , Animais , Camundongos , Camundongos Endogâmicos mdx , Âmnio , Músculo Esquelético , Distrofina/genética , Distrofia Muscular de Duchenne/genética , Modelos Animais de Doenças
4.
Clin Linguist Phon ; : 1-15, 2023 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-37253140

RESUMO

Sociolinguistic factors such as status and prestige can significantly impact the persistence of an accent in a patient with Foreign Accent Syndrome (FAS). FAS is a rare acquired syndrome that affects a speaker's accent typically caused by a stroke or trauma. In this presented FAS case study, we explore two distinct perspectives on a shift from a Sicilian to a North-East variety of Italian accent, caused by an accident trauma. Data have been collected with an ethnographic approach to explore the patient's narrative towards his 'foreign accent'. Firstly, the study analyzes the perception of native listeners through a speech sample perception test of different varieties of Italian. The listeners' responses revealed a diversified classification of the accent, highlighting the listener's crucial role in assigning the status of 'foreignness' to a particular variety. Additionally, an analysis with Praat software showed that the FAS speaker used a variety with some Sicilian and North-East traits. Secondly, the study investigated the patient's perception of their new accent through an ethnographic approach and participant observer technique. The results revealed a typology of FAS speakers that correlated with sociolinguistic factors not previously identified by research. In conclusion, this study sheds light on the complex interplay between sociolinguistic factors and FAS, demonstrating the importance of exploring FAS under various perspectives of research.

5.
Int J Mol Sci ; 24(5)2023 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-36901738

RESUMO

Histone deacetylases (HDACs) are enzymes that regulate the deacetylation of numerous histone and non-histone proteins, thereby affecting a wide range of cellular processes. Deregulation of HDAC expression or activity is often associated with several pathologies, suggesting potential for targeting these enzymes for therapeutic purposes. For example, HDAC expression and activity are higher in dystrophic skeletal muscles. General pharmacological blockade of HDACs, by means of pan-HDAC inhibitors (HDACi), ameliorates both muscle histological abnormalities and function in preclinical studies. A phase II clinical trial of the pan-HDACi givinostat revealed partial histological improvement and functional recovery of Duchenne Muscular Dystrophy (DMD) muscles; results of an ongoing phase III clinical trial that is assessing the long-term safety and efficacy of givinostat in DMD patients are pending. Here we review the current knowledge about the HDAC functions in distinct cell types in skeletal muscle, identified by genetic and -omic approaches. We describe the signaling events that are affected by HDACs and contribute to muscular dystrophy pathogenesis by altering muscle regeneration and/or repair processes. Reviewing recent insights into HDAC cellular functions in dystrophic muscles provides new perspectives for the development of more effective therapeutic approaches based on drugs that target these critical enzymes.


Assuntos
Histona Desacetilases , Distrofia Muscular de Duchenne , Humanos , Histona Desacetilases/metabolismo , Distrofia Muscular de Duchenne/genética , Carbamatos/farmacologia , Músculo Esquelético/metabolismo , Inibidores de Histona Desacetilases/farmacologia
6.
Cell Death Dis ; 13(9): 793, 2022 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-36114172

RESUMO

Muscle-resident non-myogenic mesenchymal cells play key roles that drive successful tissue regeneration within the skeletal muscle stem cell niche. These cells have recently emerged as remarkable therapeutic targets for neuromuscular disorders, although to date they have been poorly investigated in facioscapulohumeral muscular dystrophy (FSHD). In this study, we characterised the non-myogenic mesenchymal stromal cell population in FSHD patients' muscles with signs of disease activity, identified by muscle magnetic resonance imaging (MRI), and compared them with those obtained from apparently normal muscles of FSHD patients and from muscles of healthy, age-matched controls. Our results showed that patient-derived cells displayed a distinctive expression pattern of mesenchymal markers, along with an impaired capacity to differentiate towards mature adipocytes in vitro, compared with control cells. We also demonstrated a significant expansion of non-myogenic mesenchymal cells (identified as CD201- or PDGFRA-expressing cells) in FSHD muscles with signs of disease activity, which correlated with the extent of intramuscular fibrosis. In addition, the accumulation of non-myogenic mesenchymal cells was higher in FSHD muscles that deteriorate more rapidly. Our results prompt a direct association between an accumulation, as well as an altered differentiation, of non-myogenic mesenchymal cells with muscle degeneration in FSHD patients. Elucidating the mechanisms and cellular interactions that are altered in the affected muscles of FSHD patients could be instrumental to clarify disease pathogenesis and identifying reliable novel therapeutic targets.


Assuntos
Células-Tronco Mesenquimais , Distrofia Muscular Facioescapuloumeral , Diferenciação Celular/fisiologia , Humanos , Imageamento por Ressonância Magnética/métodos , Células-Tronco Mesenquimais/patologia , Músculo Esquelético/metabolismo , Distrofia Muscular Facioescapuloumeral/genética , Distrofia Muscular Facioescapuloumeral/metabolismo , Distrofia Muscular Facioescapuloumeral/patologia
7.
Adv Exp Med Biol ; 1382: 71-84, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36029404

RESUMO

Muscular dystrophies are a complex group of inherited neuromuscular disorders that progressively lead to a loss of muscle fibers and mobility and muscle weakness; over time, they evolve to an increasing level of disability. Muscular dystrophies are mostly caused by genetic mutations in proteins responsible for maintaining sarcolemma structures, such as an absence or reductions of dystrophin expression, conditions which are strictly related to muscular disorders that affect most people with this disease. Along the years, with the recent advances in the understanding of muscular dystrophies, it has been shown that many changes in Post-Translational Modifications (PTMs) of muscle proteins are associated with muscular dystrophies, wherein pathogenic alterations in the modulation of these muscle proteins are directly related to the incidence of this disease. An increase in the identification of the genetic bases and molecular mechanisms involved in the most common form of muscular dystrophies, including PTMs changes, holds potential to develop new therapeutic approaches. In this chapter we will describe the most common muscular dystrophies and changes in PTM processes such as phosphorylation and glycosylation that are very important in the evolution of the disease, highlighting the lack of mass spectrometry-based (MS-based) studies of these PTMs, suggesting that the application of this technique could reveal important informations about the molecular mechanisms of muscular dystrophies.


Assuntos
Distrofias Musculares , Humanos , Proteínas Musculares , Mutação , Processamento de Proteína Pós-Traducional
8.
Front Bioeng Biotechnol ; 9: 652970, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34095095

RESUMO

Mesenchymal stromal cells (MSCs) are multipotent cells found in different tissues: bone marrow, peripheral blood, adipose tissues, skeletal muscle, perinatal tissues, and dental pulp. MSCs are able to self-renew and to differentiate into multiple lineages, and they have been extensively used for cell therapy mostly owing to their anti-fibrotic and immunoregulatory properties that have been suggested to be at the basis for their regenerative capability. MSCs exert their effects by releasing a variety of biologically active molecules such as growth factors, chemokines, and cytokines, either as soluble proteins or enclosed in extracellular vesicles (EVs). Analyses of MSC-derived secretome and in particular studies on EVs are attracting great attention from a medical point of view due to their ability to mimic all the therapeutic effects produced by the MSCs (i.e., endogenous tissue repair and regulation of the immune system). MSC-EVs could be advantageous compared with the parental cells because of their specific cargo containing mRNAs, miRNAs, and proteins that can be biologically transferred to recipient cells. MSC-EV storage, transfer, and production are easier; and their administration is also safer than MSC therapy. The skeletal muscle is a very adaptive tissue, but its regenerative potential is altered during acute and chronic conditions. Recent works demonstrate that both MSCs and their secretome are able to help myofiber regeneration enhancing myogenesis and, interestingly, can be manipulated as a novel strategy for therapeutic interventions in muscular diseases like muscular dystrophies or atrophy. In particular, MSC-EVs represent promising candidates for cell free-based muscle regeneration. In this review, we aim to give a complete picture of the therapeutic properties and advantages of MSCs and their products (MSC-derived EVs and secreted factors) relevant for skeletal muscle regeneration in main muscular diseases.

9.
EMBO Rep ; 21(9): e50863, 2020 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-32754983

RESUMO

We show that extracellular vesicles (EVs) released by mesenchymal cells (i.e., fibro-adipogenic progenitors-FAPs) mediate microRNA (miR) transfer to muscle stem cells (MuSCs) and that exposure of dystrophic FAPs to HDAC inhibitors (HDACis) increases the intra-EV levels of a subset of miRs, which cooperatively target biological processes of therapeutic interest, including regeneration, fibrosis, and inflammation. Increased levels of miR-206 in EVs released by FAPs of muscles from Duchenne muscular dystrophy (DMD) patients or mdx mice exposed to HDACi are associated with enhanced regeneration and decreased fibrosis. Consistently, EVs from HDACi-treated dystrophic FAPs can stimulate MuSC activation and expansion ex vivo, and promote regeneration, while inhibiting fibrosis and inflammation of dystrophic muscles, upon intramuscular transplantation in mdx mice, in vivo. AntagomiR-mediated blockade of individual miRs reveals a specific requirement of miR-206 for EV-induced expansion of MuSCs and regeneration of dystrophic muscles, and indicates that cooperative activity of HDACi-induced miRs accounts for the net biological effect of these EVs. These data point to pharmacological modulation of EV content as novel strategy for therapeutic interventions in muscular dystrophies.


Assuntos
Vesículas Extracelulares , MicroRNAs , Animais , Inibidores de Histona Desacetilases/farmacologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , MicroRNAs/genética , Músculo Esquelético
10.
Int J Mol Sci ; 21(3)2020 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-32023816

RESUMO

Duchenne muscular dystrophy (DMD) is a genetic disease characterized by muscle wasting and chronic inflammation, leading to impaired satellite cells (SCs) function and exhaustion of their regenerative capacity. We previously showed that lack of PKCθ in mdx mice, a mouse model of DMD, reduces muscle wasting and inflammation, and improves muscle regeneration and performance at early stages of the disease. In this study, we show that muscle regeneration is boosted, and fibrosis reduced in mdxθ-/- mice, even at advanced stages of the disease. This phenotype was associated with a higher number of Pax7 positive cells in mdxθ-/- muscle compared with mdx muscle, during the progression of the disease. Moreover, the expression level of Pax7 and Notch1, the pivotal regulators of SCs self-renewal, were upregulated in SCs isolated from mdxθ-/- muscle compared with mdx derived SCs. Likewise, the expression of the Notch ligands Delta1 and Jagged1 was higher in mdxθ-/- muscle compared with mdx. The expression level of Delta1 and Jagged1 was also higher in PKCθ-/- muscle compared with WT muscle following acute injury. In addition, lack of PKCθ prolonged the survival and sustained the differentiation of transplanted myogenic progenitors. Overall, our results suggest that lack of PKCθ promotes muscle repair in dystrophic mice, supporting stem cells survival and maintenance through increased Delta-Notch signaling.


Assuntos
Cardiotoxinas/efeitos adversos , Músculo Esquelético/lesões , Distrofia Muscular de Duchenne/genética , Proteína Quinase C-theta/genética , Células-Tronco/citologia , Animais , Diferenciação Celular , Células Cultivadas , Masculino , Camundongos , Camundongos Endogâmicos mdx , Músculo Esquelético/citologia , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/fisiologia , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/terapia , Fator de Transcrição PAX7/metabolismo , Receptor Notch1/metabolismo , Regeneração , Transdução de Sinais , Transplante de Células-Tronco , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo
11.
Nat Commun ; 9(1): 3950, 2018 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-30262909

RESUMO

The endocannabinoid system refers to a widespread signaling system and its alteration is implicated in a growing number of human diseases. However, the potential role of endocannabinoids in skeletal muscle disorders remains unknown. Here we report the role of the endocannabinoid CB1 receptors in Duchenne's muscular dystrophy. In murine and human models, CB1 transcripts show the highest degree of expression at disease onset, and then decline overtime. Similar changes are observed for PAX7, a key regulator of muscle stem cells. Bioinformatics and biochemical analysis reveal that PAX7 binds and upregulates the CB1 gene in dystrophic more than in healthy muscles. Rimonabant, an antagonist of CB1, promotes human satellite cell differentiation in vitro, increases the number of regenerated myofibers, and prevents locomotor impairment in dystrophic mice. In conclusion, our study uncovers a PAX7-CB1 cross talk potentially exacerbating DMD and highlights the role of CB1 receptors as target for potential therapies.


Assuntos
Distrofia Muscular de Duchenne/genética , Receptor CB1 de Canabinoide/genética , Animais , Ácidos Araquidônicos/metabolismo , Sequência de Bases , Biomarcadores/metabolismo , Diglicerídeos/metabolismo , Endocanabinoides/metabolismo , Glicerídeos/metabolismo , Células HEK293 , Humanos , Luciferases/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Atividade Motora/efeitos dos fármacos , Células Musculares/efeitos dos fármacos , Células Musculares/metabolismo , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Distrofia Muscular de Duchenne/patologia , Distrofia Muscular de Duchenne/fisiopatologia , Fator de Transcrição PAX7/genética , Fator de Transcrição PAX7/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptor CB1 de Canabinoide/metabolismo , Regeneração/efeitos dos fármacos , Rimonabanto/farmacologia , Transcrição Gênica/efeitos dos fármacos
12.
Sci Rep ; 8(1): 3448, 2018 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-29472596

RESUMO

Skeletal muscle exhibits a high regenerative capacity, mainly due to the ability of satellite cells to replicate and differentiate in response to appropriate stimuli. Epigenetic control is effective at different stages of this process. It has been shown that the chromatin-remodeling factor HDAC4 is able to regulate satellite cell proliferation and commitment. However, its molecular targets are still uncovered. To explain the signaling pathways regulated by HDAC4 in satellite cells, we generated tamoxifen-inducible mice with conditional inactivation of HDAC4 in Pax7+ cells (HDAC4 KO mice). We found that the proliferation and differentiation of HDAC4 KO satellite cells were compromised, although similar amounts of satellite cells were found in mice. Moreover, we found that the inhibition of HDAC4 in satellite cells was sufficient to block the differentiation process. By RNA-sequencing analysis we identified P21 and Sharp1 as HDAC4 target genes. Reducing the expression of these target genes in HDAC4 KO satellite cells, we also defined the molecular pathways regulated by HDAC4 in the epigenetic control of satellite cell expansion and fusion.


Assuntos
Inibidor de Quinase Dependente de Ciclina p21/genética , Epigênese Genética , Histona Desacetilases/fisiologia , Células Satélites de Músculo Esquelético/fisiologia , Fatores de Transcrição/genética , Animais , Diferenciação Celular/genética , Proliferação de Células/genética , Células Cultivadas , Histona Desacetilases/genética , Camundongos , Camundongos Knockout , Fator de Transcrição PAX7/genética , Células Satélites de Músculo Esquelético/citologia , Transdução de Sinais , Tamoxifeno/farmacologia
13.
Methods Mol Biol ; 1687: 231-256, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29067668

RESUMO

Functional interactions between muscle (satellite) stem cells-MuSCs-and other cellular components of their niche (the fibro-adipogenic progenitors-FAPs) coordinate regeneration of injured as well as diseased skeletal muscles. These interactions are largely mediated by secretory networks, whose integrity is critical to determine whether repair occurs by compensatory regeneration leading to formation of new contractile fibers, or by maladaptive formation of fibrotic scars and fat infiltration. Here we provide the description of methods for isolation of FAPs and MuSCs from muscles of wild type and dystrophic mice, and protocols of cocultures as well as MuSC's exposure to FAP- derived exosomes. These methods and protocols can be exploited in murine models of acute muscle injury to investigate salient features of physiological repair, and in models of muscular diseases to identify dysregulated networks that compromise functional interactions between cellular components of the regeneration environment during disease progression. We predict that exporting these procedures to patient-derived muscle samples will contribute to advance our understanding of human skeletal myogenesis and related disorders.


Assuntos
Tecido Adiposo/citologia , Separação Celular/métodos , Células Satélites de Músculo Esquelético/citologia , Células-Tronco/citologia , Adipogenia/genética , Tecido Adiposo/metabolismo , Tecido Adiposo/patologia , Animais , Diferenciação Celular/genética , Fibrose/genética , Fibrose/patologia , Humanos , Camundongos , Desenvolvimento Muscular/genética , Mioblastos/citologia , Mioblastos/metabolismo , Mioblastos/patologia , Células Satélites de Músculo Esquelético/metabolismo , Células Satélites de Músculo Esquelético/patologia
14.
Stem Cells Int ; 2016: 6093601, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26839565

RESUMO

In the context of regenerative medicine, based on the potential of stem cells to restore diseased tissues, epigenetics is becoming a pivotal area of interest. Therapeutic interventions that promote tissue and organ regeneration have as primary objective the selective control of gene expression in adult stem cells. This requires a deep understanding of the epigenetic mechanisms controlling transcriptional programs in tissue progenitors. This review attempts to elucidate the principle epigenetic regulations responsible of stem cells differentiation. In particular we focus on the current understanding of the epigenetic networks that regulate differentiation of muscle progenitors by the concerted action of chromatin-modifying enzymes and noncoding RNAs. The novel exciting role of exosome-bound microRNA in mediating epigenetic information transfer is also discussed. Finally we show an overview of the epigenetic strategies and therapies that aim to potentiate muscle regeneration and counteract the progression of Duchenne Muscular Dystrophy (DMD).

15.
Epigenomics ; 6(5): 547-60, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25431946

RESUMO

Duchenne muscular dystrophy (DMD) is a life-threatening genetic disease that currently has no available cure. A number of pharmacological strategies that aim to target events downstream of the genetic defect are currently under clinical investigation, and some of these are outlined in this report. In particular, we focus on the ability of histone deacetylase inhibitors to promote muscle regeneration and prevent the fibro-adipogenic degeneration of dystrophic mice. We describe the rationale behind the translation of histone deacetylase inhibitors into a clinical approach, which inspired the first clinical trial with an epigenetic drug as a potential therapeutic option for DMD patients.


Assuntos
Epigênese Genética , Inibidores de Histona Desacetilases/uso terapêutico , Distrofia Muscular de Duchenne/tratamento farmacológico , Distrofia Muscular de Duchenne/genética , Acetilação , Animais , Carbamatos/farmacologia , Carbamatos/uso terapêutico , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Ensaios Clínicos Fase I como Assunto , Ensaios Clínicos Fase II como Assunto , Epigênese Genética/efeitos dos fármacos , Regulação da Expressão Gênica , Inibidores de Histona Desacetilases/farmacologia , Histonas/metabolismo , Humanos , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/metabolismo , Mutação , Regeneração/efeitos dos fármacos , Regeneração/genética , Células Satélites de Músculo Esquelético/citologia , Células Satélites de Músculo Esquelético/efeitos dos fármacos , Células Satélites de Músculo Esquelético/metabolismo
16.
Genes Dev ; 28(8): 841-57, 2014 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-24682306

RESUMO

Fibro-adipogenic progenitors (FAPs) are important components of the skeletal muscle regenerative environment. Whether FAPs support muscle regeneration or promote fibro-adipogenic degeneration is emerging as a key determinant in the pathogenesis of muscular diseases, including Duchenne muscular dystrophy (DMD). However, the molecular mechanism that controls FAP lineage commitment and activity is currently unknown. We show here that an HDAC-myomiR-BAF60 variant network regulates the fate of FAPs in dystrophic muscles of mdx mice. Combinatorial analysis of gene expression microarray, genome-wide chromatin remodeling by nuclease accessibility (NA) combined with next-generation sequencing (NA-seq), small RNA sequencing (RNA-seq), and microRNA (miR) high-throughput screening (HTS) against SWI/SNF BAF60 variants revealed that HDAC inhibitors (HDACis) derepress a "latent" myogenic program in FAPs from dystrophic muscles at early stages of disease. Specifically, HDAC inhibition induces two core components of the myogenic transcriptional machinery, MYOD and BAF60C, and up-regulates the myogenic miRs (myomiRs) (miR-1.2, miR-133, and miR-206), which target the alternative BAF60 variants BAF60A and BAF60B, ultimately directing promyogenic differentiation while suppressing the fibro-adipogenic phenotype. In contrast, FAPs from late stage dystrophic muscles are resistant to HDACi-induced chromatin remodeling at myogenic loci and fail to activate the promyogenic phenotype. These results reveal a previously unappreciated disease stage-specific bipotency of mesenchimal cells within the regenerative environment of dystrophic muscles. Resolution of such bipotency by epigenetic intervention with HDACis provides a molecular rationale for the in situ reprogramming of target cells to promote therapeutic regeneration of dystrophic muscles.


Assuntos
Histona Desacetilases/metabolismo , MicroRNAs/metabolismo , Músculo Esquelético/fisiologia , Distrofias Musculares/genética , Distrofias Musculares/fisiopatologia , Células-Tronco/metabolismo , Animais , Reprogramação Celular/genética , Cromatina/genética , Montagem e Desmontagem da Cromatina/fisiologia , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Inibidores de Histona Desacetilases/farmacologia , Histona Desacetilases/genética , Ácidos Hidroxâmicos/farmacologia , Camundongos , Camundongos Endogâmicos mdx , Proteínas Musculares/genética , Proteínas Musculares/metabolismo
17.
Mol Med ; 19: 79-87, 2013 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-23552722

RESUMO

Previous work has established the existence of dystrophin-nitric oxide (NO) signaling to histone deacetylases (HDACs) that is deregulated in dystrophic muscles. As such, pharmacological interventions that target HDACs (that is, HDAC inhibitors) are of potential therapeutic interest for the treatment of muscular dystrophies. In this study, we explored the effectiveness of long-term treatment with different doses of the HDAC inhibitor givinostat in mdx mice--the mouse model of Duchenne muscular dystrophy (DMD). This study identified an efficacy for recovering functional and histological parameters within a window between 5 and 10 mg/kg/d of givinostat, with evident reduction of the beneficial effects with 1 mg/kg/d dosage. The long-term (3.5 months) exposure of 1.5-month-old mdx mice to optimal concentrations of givinostat promoted the formation of muscles with increased cross-sectional area and reduced fibrotic scars and fatty infiltration, leading to an overall improvement of endurance performance in treadmill tests and increased membrane stability. Interestingly, a reduced inflammatory infiltrate was observed in muscles of mdx mice exposed to 5 and 10 mg/kg/d of givinostat. A parallel pharmacokinetic/pharmacodynamic analysis confirmed the relationship between the effective doses of givinostat and the drug distribution in muscles and blood of treated mice. These findings provide the preclinical basis for an immediate translation of givinostat into clinical studies with DMD patients.


Assuntos
Carbamatos/uso terapêutico , Inibidores de Histona Desacetilases/uso terapêutico , Distrofia Muscular de Duchenne/tratamento farmacológico , Animais , Carbamatos/farmacologia , Células Cultivadas , Teste de Esforço , Fibrose/tratamento farmacológico , Fibrose/patologia , Inibidores de Histona Desacetilases/farmacologia , Humanos , Camundongos , Camundongos Endogâmicos mdx , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/patologia , Músculo Esquelético/fisiopatologia , Distrofia Muscular de Duchenne/patologia , Distrofia Muscular de Duchenne/fisiopatologia , Mioblastos/citologia , Mioblastos/efeitos dos fármacos , Corrida
18.
EMBO Mol Med ; 5(4): 626-39, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23505062

RESUMO

HDAC inhibitors (HDACi) exert beneficial effects in mdx mice, by promoting endogenous regeneration; however, the cellular determinants of HDACi activity on dystrophic muscles have not been determined. We show that fibroadipogenic progenitors (FAP) influence the regeneration potential of satellite cells during disease progression in mdx mice and mediate HDACi ability to selectively promote regeneration at early stages of disease. FAPs from young mdx mice promote, while FAPs from old mdx mice repress, satellite cell-mediated formation of myotubes. In young mdx mice HDACi inhibited FAP adipogenic potential, while enhancing their ability to promote differentiation of adjacent satellite cells, through upregulation of the soluble factor follistatin. By contrast, FAPs from old mdx mice were resistant to HDACi-mediated inhibition of adipogenesis and constitutively repressed satellite cell-mediated formation of myotubes. We show that transplantation of FAPs from regenerating young muscles restored HDACi ability to increase myofibre size in old mdx mice. These results reveal that FAPs are key cellular determinants of disease progression in mdx mice and mediate a previously unappreciated stage-specific beneficial effect of HDACi in dystrophic muscles.


Assuntos
Adipogenia/efeitos dos fármacos , Inibidores de Histona Desacetilases/administração & dosagem , Músculos/fisiopatologia , Distrofias Musculares/tratamento farmacológico , Células Satélites de Músculo Esquelético/citologia , Células-Tronco/citologia , Fatores Etários , Animais , Células Cultivadas , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Camundongos Knockout , Camundongos SCID , Músculos/efeitos dos fármacos , Distrofias Musculares/fisiopatologia , Células Satélites de Músculo Esquelético/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos
19.
EMBO J ; 31(2): 301-16, 2012 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-22068056

RESUMO

Tissue-specific transcriptional activators initiate differentiation towards specialized cell types by inducing chromatin modifications permissive for transcription at target loci, through the recruitment of SWItch/Sucrose NonFermentable (SWI/SNF) chromatin-remodelling complex. However, the molecular mechanism that regulates SWI/SNF nuclear distribution in response to differentiation signals is unknown. We show that the muscle determination factor MyoD and the SWI/SNF subunit BAF60c interact on the regulatory elements of MyoD-target genes in myoblasts, prior to activation of transcription. BAF60c facilitates MyoD binding to target genes and marks the chromatin for signal-dependent recruitment of the SWI/SNF core to muscle genes. BAF60c phosphorylation on a conserved threonine by differentiation-activated p38α kinase is the signal that promotes incorporation of MyoD-BAF60c into a Brg1-based SWI/SNF complex, which remodels the chromatin and activates transcription of MyoD-target genes. Our data support an unprecedented two-step model by which pre-assembled BAF60c-MyoD complex directs recruitment of SWI/SNF to muscle loci in response to differentiation cues.


Assuntos
Proteínas Cromossômicas não Histona/fisiologia , Sistema de Sinalização das MAP Quinases , Desenvolvimento Muscular/fisiologia , Proteínas Musculares/fisiologia , Proteína MyoD/fisiologia , Fatores de Transcrição/fisiologia , Animais , Linhagem Celular , Cromatina/genética , Proteínas Cromossômicas não Histona/antagonistas & inibidores , Proteínas Cromossômicas não Histona/química , Proteínas Cromossômicas não Histona/genética , DNA Helicases/fisiologia , Fibroblastos/metabolismo , Regulação da Expressão Gênica/genética , Células HeLa/metabolismo , Humanos , Camundongos , Complexos Multiproteicos , Proteínas Musculares/antagonistas & inibidores , Proteínas Musculares/química , Proteínas Musculares/genética , Mioblastos/metabolismo , Proteínas Nucleares/fisiologia , Fosforilação , Fosfotreonina/análise , Processamento de Proteína Pós-Traducional , Interferência de RNA , RNA Interferente Pequeno/farmacologia , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/química , Fatores de Transcrição/genética , Técnicas do Sistema de Duplo-Híbrido , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia
20.
Mol Med ; 17(5-6): 457-65, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21308150

RESUMO

Histone deacetylases inhibitors (HDACi) include a growing number of drugs that share the ability to inhibit the enzymatic activity of some or all the HDACs. Experimental and preclinical evidence indicates that these epigenetic drugs not only can be effective in the treatment of malignancies, inflammatory diseases and degenerative disorders, but also in the treatment of genetic diseases, such as muscular dystrophies. The ability of HDACi to counter the progression of muscular dystrophies points to HDACs as a crucial link between specific genetic mutations and downstream determinants of disease progression. It also suggests the contribution of epigenetic events to the pathogenesis of muscular dystrophies. Here we describe the experimental evidence supporting the key role of HDACs in the control of the transcriptional networks underlying the potential of dystrophic muscles either to activate compensatory regeneration or to undergo fibroadipogenic degeneration. Studies performed in mouse models of Duchenne muscular dystrophy (DMD) indicate that dystrophin deficiency leads to deregulated HDAC activity, which perturbs downstream networks and can be restored directly, by HDAC blockade, or indirectly, by reexpression of dystrophin. This evidence supports the current view that HDACi are emerging candidate drugs for pharmacological interventions in muscular dystrophies, and reveals unexpected common beneficial outcomes of pharmacological treatment or gene therapy.


Assuntos
Inibidores de Histona Desacetilases/uso terapêutico , Distrofias Musculares/tratamento farmacológico , Distrofias Musculares/genética , Animais , Distrofina/metabolismo , Humanos , Camundongos , Distrofias Musculares/metabolismo , Distrofia Muscular de Duchenne/tratamento farmacológico , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...