Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
Mol Ther Nucleic Acids ; 33: 683-697, 2023 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-37650119

RESUMO

Autosomal recessive limb-girdle muscular dystrophy 21 (LGMDR21) is caused by pathogenic variants in protein O-glucosyltransferase 1 (POGLUT1), which is responsible for O-glucosylation of specific epidermal growth factor (EGF) repeats found in ∼50 mammalian proteins, including Notch receptors. Previous data from patient biopsies indicated that impaired Notch signaling, reduction of muscle stem cells, and accelerated differentiation are probably involved in disease etiopathology. Using patient induced pluripotent stem cells (iPSCs), their corrected isotypes, and control iPSCs, gene expression profiling indicated dysregulation of POGLUT1, NOTCH, muscle development, extracellular matrix (ECM), cell adhesion, and migration as involved pathways. They also exhibited reduced in vitro POGLUT1 enzymatic activity and NOTCH signaling as well as defective myogenesis, proliferation, migration and differentiation. Furthermore, in vivo studies demonstrated significant reductions in engraftment, muscle stem cell formation, PAX7 expression, and maintenance, along with an increased percentage of mislocalized PAX7+ cells in the interstitial space. Gene correction in patient iPSCs using CRISPR-Cas9 nickase led to the rescue of the main in vitro and in vivo phenotypes. These results demonstrate the efficacy of iPSCs and gene correction in disease modeling and rescue of the phenotypes and provide evidence of the involvement of muscle stem cell niche localization, PAX7 expression, and cell migration as possible mechanisms in LGMDR21.

2.
Sci Transl Med ; 14(669): eabo1981, 2022 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-36322628

RESUMO

Immune checkpoint inhibitors (ICIs) have been increasingly used in combination for cancer treatment but are associated with myocarditis. Here, we report that tumor-bearing mice exhibited response to treatment with combinatorial anti-programmed cell death 1 and anti-cytotoxic T lymphocyte antigen-4 antibodies but also presented with cardiovascular toxicities observed clinically with ICI therapy, including myocarditis and arrhythmia. Female mice were preferentially affected with myocarditis compared to male mice, consistent with a previously described genetic model of ICI myocarditis and emerging clinical data. Mechanistically, myocardial tissue from ICI-treated mice, the genetic mouse model, and human heart tissue from affected patients with ICI myocarditis all exhibited down-regulation of MANF (mesencephalic astrocyte-derived neurotrophic factor) and HSPA5 (heat shock 70-kDa protein 5) in the heart; this down-regulation was particularly notable in female mice. ICI myocarditis was amplified by heart-specific genetic deletion of mouse Manf and was attenuated by administration of recombinant MANF protein, suggesting a causal role. Ironically, both MANF and HSPA5 were transcriptionally induced by liganded estrogen receptor ß and inhibited by androgen receptor. However, ICI treatment reduced serum estradiol concentration to a greater extent in female compared to male mice. Treatment with an estrogen receptor ß-specific agonist and androgen depletion therapy attenuated ICI-associated cardiac effects. Together, our data suggest that ICI treatment inhibits estradiol-dependent expression of MANF/HSPA5 in the heart, curtailing the cardiomyocyte response to immune injury. This endocrine-cardiac-immune pathway offers new insights into the mechanisms of sex differences in cardiac disease and may offer treatment strategies for ICI myocarditis.


Assuntos
Miocardite , Humanos , Feminino , Masculino , Camundongos , Animais , Miocardite/complicações , Miocardite/tratamento farmacológico , Inibidores de Checkpoint Imunológico , Receptor beta de Estrogênio/metabolismo , Receptor beta de Estrogênio/uso terapêutico , Miócitos Cardíacos/metabolismo , Estradiol/efeitos adversos , Estradiol/metabolismo , Fatores de Crescimento Neural/efeitos adversos , Fatores de Crescimento Neural/metabolismo
3.
J Cell Biochem ; 123(2): 147-154, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34668236

RESUMO

Epigenetics play a fundamental role in induced pluripotent stem cell (iPSC) technology due to their effect on iPSC's reprogramming efficiency and their subsequent role in iPSC differentiation toward a specific lineage. Epigenetics can skew the differentiation course of iPSCs toward a specific lineage based on the epigenetic memory of the source cells, or even lead to acquisition of new cell phenotypes, due to its aberrations during reprogramming. This viewpoint discusses key features of the epigenetic process during iPSC reprogramming/differentiation and outlines important epigenetic factors that need to be considered for successful generation and differentiation of iPSCs for downstream applications.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos , Reprogramação Celular , Metilação de DNA , Epigênese Genética , Células-Tronco Pluripotentes Induzidas/metabolismo , Animais , Humanos
4.
Cells ; 10(10)2021 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-34685726

RESUMO

Advancements in reprogramming somatic cells into induced pluripotent stem cells (iPSCs) have provided a strong framework for in vitro disease modeling, gene correction and stem cell-based regenerative medicine. In cases of skeletal muscle disorders, iPSCs can be used for the generation of skeletal muscle progenitors to study disease mechanisms, or implementation for the treatment of muscle disorders. We have recently developed an improved directed differentiation method for the derivation of skeletal myogenic progenitors from hiPSCs. This method allows for a short-term (2 weeks) and efficient skeletal myogenic induction (45-65% of the cells) in human pluripotent stem cells (ESCs/iPSCs) using small molecules to induce mesoderm and subsequently myotomal progenitors, without the need for any gene integration or modification. After initial differentiation, skeletal myogenic progenitors can be purified from unwanted cells using surface markers (CD10+CD24-). These myogenic progenitors have been extensively characterized using in vitro gene expression/differentiation profiling as well as in vivo engraftment studies in dystrophic (mdx) and muscle injury (VML) rodent models and have been proven to be able to engraft and form mature myofibers as well as seeding muscle stem cells. The current protocol describes a detailed, step-by-step guide for this method and outlines important experimental details and troubleshooting points for its application in any human pluripotent stem cells.


Assuntos
Biomarcadores/metabolismo , Diferenciação Celular , Membrana Celular/metabolismo , Separação Celular/métodos , Desenvolvimento Muscular , Músculo Esquelético/citologia , Células-Tronco Pluripotentes/citologia , Proliferação de Células , Forma Celular , Sobrevivência Celular , Humanos , Fibras Musculares Esqueléticas/citologia
5.
Front Cell Dev Biol ; 9: 609984, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33585477

RESUMO

Cellular reprogramming is a fundamental topic in the research of stem cells and molecular biology. It is widely investigated and its understanding is crucial for learning about different aspects of development such as cell proliferation, determination of cell fate and stem cell renewal. Other factors involved during development include hypoxia and epigenetics, which play major roles in the development of tissues and organs. This review will discuss the involvement of hypoxia and epigenetics in the regulation of cellular reprogramming and how interplay between each factor can contribute to different cellular functions as well as tissue regeneration.

6.
Mol Ther ; 29(1): 121-131, 2021 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-32966776

RESUMO

Volumetric muscle loss injury is a common health problem with long-term disabilities. One common treatment is using muscle flaps from donor site, which has limited potentials due to donor site availability and morbidity. Although several stem cell therapies have been evaluated so far, most suffer from limited availability, immune incompatibility, or differentiation potential. Therefore, induced pluripotent stem cells (iPSCs) have a great promise for this purpose due to their unique differentiation, self-renewal, and immunocompatibility. Current study was designed to determine therapeutic potential of human iPSCs (hiPSCs) in a mouse model of volumetric muscle loss. Muscles were subjected to excision to generate 30%-40% muscle loss. Next, hiPSCs were differentiated toward skeletal myogenic progenitors and used with fibrin hydrogel to reconstruct the lost muscle. Histologic evaluation of the treated muscles indicated abundant engraftment of donor-derived mature fibers expressing human markers. Donor-derived fibers were also positive for the presence of neuromuscular junction (NMJ), indicating their proper innervation. Evaluation of the engrafted region indicated the presence of donor-derived satellite cells expressing human markers and Pax7. Finally, in situ muscle function analysis demonstrated significant improvement of the muscle contractility in muscles treated with hiPSCs. These results therefore provide key evidence for the therapeutic potential of human iPSCs in volumetric muscle loss injuries.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Doenças Musculares/patologia , Doenças Musculares/terapia , Transplante de Células-Tronco , Animais , Atrofia , Modelos Animais de Doenças , Sobrevivência de Enxerto , Camundongos , Músculo Esquelético/patologia , Doenças Musculares/etiologia , Transplante de Células-Tronco/efeitos adversos , Transplante de Células-Tronco/métodos , Resultado do Tratamento
7.
Nat Cell Biol ; 22(11): 1332-1345, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33106653

RESUMO

Dystrophin proteomic regulation in muscular dystrophies (MDs) remains unclear. We report that a long noncoding RNA (lncRNA), H19, associates with dystrophin and inhibits E3-ligase-dependent polyubiquitination at Lys 3584 (referred to as Ub-DMD) and its subsequent protein degradation. In-frame deletions in BMD and a DMD non-silent mutation (C3340Y) resulted in defects in the ability of the protein to interact with H19, which caused elevated Ub-DMD levels and dystrophin degradation. Dmd C3333Y mice exhibited progressive MD, elevated serum creatine kinase, heart dilation, blood vessel irregularity and respiratory failure with concurrently reduced dystrophin and increased Ub-DMD status. H19 RNA oligonucleotides conjugated with agrin (AGR-H19) and nifenazone competed with or inhibited TRIM63. Dmd C3333Y animals, induced-pluripotent-stem-cell-derived skeletal muscle cells from patients with Becker MD and mdx mice subjected to exon skipping exhibited inhibited dystrophin degradation, preserved skeletal and cardiac muscle histology, and improved strength and heart function following AGR-H19 or nifenazone treatment. Our study paves the way for meaningful targeted therapeutics for Becker MD and for certain patients with Duchenne MD.


Assuntos
Músculo Esquelético/metabolismo , Distrofias Musculares/prevenção & controle , Oligonucleotídeos/administração & dosagem , RNA Longo não Codificante/metabolismo , Animais , Antipirina/administração & dosagem , Antipirina/análogos & derivados , Cardiomiopatias/genética , Cardiomiopatias/metabolismo , Cardiomiopatias/patologia , Cardiomiopatias/prevenção & controle , Linhagem Celular , Modelos Animais de Doenças , Distrofina/genética , Distrofina/metabolismo , Inibidores Enzimáticos/administração & dosagem , Feminino , Meia-Vida , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Camundongos Mutantes , Proteínas Musculares/antagonistas & inibidores , Proteínas Musculares/metabolismo , Força Muscular , Músculo Esquelético/patologia , Músculo Esquelético/fisiopatologia , Distrofias Musculares/genética , Distrofias Musculares/metabolismo , Distrofias Musculares/patologia , Mutação , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Niacinamida/administração & dosagem , Niacinamida/análogos & derivados , Oligonucleotídeos/genética , Oligonucleotídeos/metabolismo , Estabilidade Proteica , Proteólise , RNA Longo não Codificante/genética , Proteínas com Motivo Tripartido/antagonistas & inibidores , Proteínas com Motivo Tripartido/metabolismo , Ubiquitina-Proteína Ligases/antagonistas & inibidores , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação
8.
J Cell Biochem ; 121(12): 4696-4698, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32692858

RESUMO

In this perspective, the potential application of stem cells for the treatment of COVID-19 related pneumonia and their potential mechanism of action have been overviewed.

9.
Stem Cells ; 37(12): 1615-1628, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31574188

RESUMO

Although the lack of dystrophin expression in muscle myofibers is the central cause of Duchenne muscular dystrophy (DMD), accumulating evidence suggests that DMD may also be a stem cell disease. Recent studies have revealed dystrophin expression in satellite cells and demonstrated that dystrophin deficiency is directly related to abnormalities in satellite cell polarity, asymmetric division, and epigenetic regulation, thus contributing to the manifestation of the DMD phenotype. Although metabolic and mitochondrial dysfunctions have also been associated with the DMD pathophysiology profile, interestingly, the role of dystrophin with respect to stem cells dysfunction has not been elucidated. In the past few years, editing of the gene that encodes dystrophin has emerged as a promising therapeutic approach for DMD, although the effects of dystrophin restoration in stem cells have not been addressed. Herein, we describe our use of a clustered regularly interspaced short palindromic repeats/Cas9-based system to correct the dystrophin mutation in dystrophic (mdx) muscle progenitor cells (MPCs) and show that the expression of dystrophin significantly improved cellular properties of the mdx MPCs in vitro. Our findings reveal that dystrophin-restored mdx MPCs demonstrated improvements in cell proliferation, differentiation, bioenergetics, and resistance to oxidative and endoplasmic reticulum stress. Furthermore, our in vivo studies demonstrated improved transplantation efficiency of the corrected MPCs in the muscles of mdx mice. Our results indicate that changes in cellular energetics and stress resistance via dystrophin restoration enhance muscle progenitor cell function, further validating that dystrophin plays a role in stem cell function and demonstrating the potential for new therapeutic approaches for DMD. Stem Cells 2019;37:1615-1628.


Assuntos
Distrofina/genética , Terapia Genética/métodos , Fibras Musculares Esqueléticas/patologia , Distrofia Muscular de Duchenne/terapia , Células Satélites de Músculo Esquelético/patologia , Animais , Sistemas CRISPR-Cas/genética , Diferenciação Celular/genética , Polaridade Celular/fisiologia , Proliferação de Células/genética , Modelos Animais de Doenças , Distrofina/metabolismo , Estresse do Retículo Endoplasmático/genética , Metabolismo Energético/genética , Epigênese Genética , Edição de Genes , Camundongos , Camundongos Endogâmicos mdx , Fibras Musculares Esqueléticas/metabolismo , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patologia , Estresse Oxidativo/genética , Células-Tronco/fisiologia
10.
Proc Natl Acad Sci U S A ; 116(10): 4346-4351, 2019 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-30760602

RESUMO

Optimal cell-based therapies for the treatment of muscle degenerative disorders should not only regenerate fibers but provide a quiescent satellite cell pool ensuring long-term maintenance and regeneration. Conditional expression of Pax3/Pax7 in differentiating pluripotent stem cells (PSCs) allows the generation of myogenic progenitors endowed with enhanced regenerative capacity. To identify the molecular determinants underlying their regenerative potential, we performed transcriptome analyses of these cells along with primary myogenic cells from several developmental stages. Here we show that in vitro-generated PSC-derived myogenic progenitors possess a molecular signature similar to embryonic/fetal myoblasts. However, compared with fetal myoblasts, following transplantation they show superior myofiber engraftment and ability to seed the satellite cell niche, respond to multiple reinjuries, and contribute to long-term regeneration. Upon engraftment, the transcriptome of reisolated Pax3/Pax7-induced PSC-derived myogenic progenitors changes toward a postnatal molecular signature, particularly in genes involved in extracellular matrix remodeling. These findings demonstrate that Pax3/Pax7-induced myogenic progenitors remodel their molecular signature and functionally mature upon in vivo exposure to the adult muscle environment.


Assuntos
Desenvolvimento Muscular/fisiologia , Fator de Transcrição PAX3/metabolismo , Fator de Transcrição PAX7/metabolismo , Células-Tronco Pluripotentes/metabolismo , Animais , Diferenciação Celular , Perfilação da Expressão Gênica , Camundongos , Desenvolvimento Muscular/genética , Músculo Esquelético , Mioblastos/metabolismo , Fator de Transcrição PAX3/genética , Fator de Transcrição PAX7/genética , Transcriptoma
11.
Cells ; 8(1)2019 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-30609814

RESUMO

Induced pluripotent stem cells (iPSCs) are the foundation of modern stem cell-based regenerative medicine, especially in the case of degenerative disorders, such as muscular dystrophies (MDs). Since their introduction in 2006, many studies have used iPSCs for disease modeling and identification of involved mechanisms, drug screening, as well as gene correction studies. In the case of muscular dystrophies, these studies commenced in 2008 and continue to address important issues, such as defining the main pathologic mechanisms in different types of MDs, drug screening to improve skeletal/cardiac muscle cell survival and to slow down disease progression, and evaluation of the efficiency of different gene correction approaches, such as exon skipping, Transcription activator-like effector nucleases (TALENs), Zinc finger nucleases (ZFNs) and RNA-guided endonuclease Cas9 (CRISPR/Cas9). In the current short review, we have summarized chronological progress of these studies and their key findings along with a perspective on the future road to successful iPSC-based cell therapy for MDs and the potential hurdles in this field.


Assuntos
Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/metabolismo , Distrofia Muscular de Duchenne/tratamento farmacológico , Medicina de Precisão , Animais , Sistemas CRISPR-Cas , Avaliação Pré-Clínica de Medicamentos , Marcação de Genes , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Modelos Biológicos , Nucleases dos Efetores Semelhantes a Ativadores de Transcrição/metabolismo , Nucleases de Dedos de Zinco/metabolismo
12.
Cell Rep ; 25(7): 1966-1981.e4, 2018 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-30428361

RESUMO

Myogenic differentiation of human pluripotent stem cells (hPSCs) has been done by gene overexpression or directed differentiation. However, viral integration, long-term culture, and the presence of unwanted cells are the main obstacles. By using CRISPR/Cas9n, a double-reporter human embryonic stem cell (hESC) line was generated for PAX7/MYF5, allowing prospective readout. This strategy allowed pathway screen to define efficient myogenic induction in hPSCs. Next, surface marker screen allowed identification of CD10 and CD24 for purification of myogenic progenitors and exclusion of non-myogenic cells. CD10 expression was also identified on human satellite cells and skeletal muscle progenitors. In vitro and in vivo studies using transgene and/or reporter-free hPSCs further validated myogenic potential of the cells by formation of new fibers expressing human dystrophin as well as donor-derived satellite cells in NSG-mdx4Cv mice. This study provides biological insights for myogenic differentiation of hPSCs using a double-reporter cell resource and defines an improved myogenic differentiation and purification strategy.


Assuntos
Separação Celular/métodos , Genes Reporter , Desenvolvimento Muscular , Músculo Esquelético/citologia , Células-Tronco Pluripotentes/metabolismo , Animais , Biomarcadores/metabolismo , Diferenciação Celular , Linhagem Celular , Autorrenovação Celular , Feminino , Células-Tronco Embrionárias Humanas/citologia , Células-Tronco Embrionárias Humanas/metabolismo , Humanos , Masculino , Mesoderma/citologia , Camundongos Endogâmicos mdx , Fator Regulador Miogênico 5/metabolismo , Fator de Transcrição PAX7/metabolismo , Células-Tronco Pluripotentes/citologia , Regeneração , Transdução de Sinais , Transplante de Células-Tronco , Fatores de Tempo , Transcriptoma/genética
13.
Stem Cell Res ; 27: 65-73, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29331939

RESUMO

Volumetric muscle defect, caused by trauma or combat injuries, is a major health concern leading to severe morbidity. It is characterized by partial or full thickness loss of muscle and its bio-scaffold, resulting in extensive fibrosis and scar formation. Therefore, the ideal therapeutic option is to use stem cells combined with bio-scaffolds to restore muscle. For this purpose, muscle-derived stem cells (MDSCs) are a great candidate due to their unique multi-lineage differentiation potential. In this study, we evaluated the regeneration potential of MDSCs for muscle loss repair using a novel in situ fibrin gel casting. Muscle defect was created by a partial thickness wedge resection in the tibialis anterior (TA) muscles of NSG mice which created an average of 25% mass loss. If untreated, this defect leads to severe muscle fibrosis. Next, MDSCs were delivered using a novel in situ fibrin gel casting method. Our results demonstrated MDSCs are able to engraft and form new myofibers in the defect when casted along with fibrin gel. LacZ labeled MDSCs were able to differentiate efficiently into new myofibers and significantly increase muscle mass. This was also accompanied by significant reduction of fibrotic tissue in the engrafted muscles. Furthermore, transplanted cells also contributed to new vessel formation and satellite cell seeding. These results confirmed the therapeutic potential of MDSCs and feasibility of direct in situ casting of fibrin/MDSC mixture to repair muscle mass defects.


Assuntos
Fibrina/uso terapêutico , Músculo Esquelético/patologia , Células-Tronco/fisiologia , Animais , Diferenciação Celular/fisiologia , Células Cultivadas , Fibrose/dietoterapia , Fibrose/terapia , Camundongos , Músculo Esquelético/lesões , Regeneração , Células-Tronco/citologia
14.
Stem Cell Res ; 24: 102-105, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-29034878

RESUMO

Recently, a new type of limb-girdle muscular dystrophy (LGMD type 2Z) has been identified due to a missense mutation in POGLUT1 (protein O-glucosyltransferase-Rumi), an enzyme capable of adding glucose to a distinct serine residue of epidermal growth factor-like repeats containing a C-X-S-X-(P/A)-C consensus sequence such as Notch receptors. Affected patients demonstrate reduced Notch signaling, decreased muscle stem cell pool and hypoglycosylation of α-dystroglycan, leading to LGMD phenotype. Here we report the generation and characterization of an iPSC line (CSCRMi001-A) from a LGMD-2Z patient with missense mutation in POGLUT1 which can be used for in vitro disease modeling.


Assuntos
Glucosiltransferases/genética , Células-Tronco Pluripotentes Induzidas/metabolismo , Distrofia Muscular do Cíngulo dos Membros/genética , Adulto , Linhagem Celular , Glucosiltransferases/metabolismo , Humanos , Masculino , Distrofia Muscular do Cíngulo dos Membros/metabolismo , Distrofia Muscular do Cíngulo dos Membros/patologia , Mutação , Mutação de Sentido Incorreto
15.
Cell Rep ; 19(13): 2867-2877, 2017 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-28658631

RESUMO

Pluripotent stem (PS)-cell-derived cell types hold promise for treating degenerative diseases. However, PS cell differentiation is intrinsically heterogeneous; therefore, clinical translation requires the development of practical methods for isolating progenitors from unwanted and potentially teratogenic cells. Muscle-regenerating progenitors can be derived through transient PAX7 expression. To better understand the biology, and to discover potential markers for these cells, here we investigate PAX7 genomic targets and transcriptional changes in human cells undergoing PAX7-mediated myogenic commitment. We identify CD54, integrin α9ß1, and Syndecan2 (SDC2) as surface markers on PAX7-induced myogenic progenitors. We show that these markers allow for the isolation of myogenic progenitors using both fluorescent- and CGMP-compatible magnetic-based sorting technologies and that CD54+α9ß1+SDC2+ cells contribute to long-term muscle regeneration in vivo. These findings represent a critical step toward enabling the translation of PS-cell-based therapies for muscle diseases.


Assuntos
Células-Tronco Pluripotentes Induzidas/metabolismo , Integrinas/metabolismo , Molécula 1 de Adesão Intercelular/metabolismo , Desenvolvimento Muscular/genética , Fator de Transcrição PAX7/genética , Sindecana-2/metabolismo , Animais , Diferenciação Celular , Expressão Gênica , Humanos , Masculino , Camundongos , Fator de Transcrição PAX7/metabolismo
16.
Stem Cell Reports ; 9(1): 12-22, 2017 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-28528701

RESUMO

Recent reports have documented the differentiation of human pluripotent stem cells toward the skeletal myogenic lineage using transgene- and cell purification-free approaches. Although these protocols generate myocytes, they have not demonstrated scalability, safety, and in vivo engraftment, which are key aspects for their future clinical application. Here we recapitulate one prominent protocol, and show that it gives rise to a heterogeneous cell population containing myocytes and other cell types. Upon transplantation, the majority of human donor cells could not contribute to myofiber formation. As a proof-of-principle, we incorporated the inducible PAX7 lentiviral system into this protocol, which then enabled scalable expansion of a homogeneous population of skeletal myogenic progenitors capable of forming myofibers in vivo. Our findings demonstrate the methods for scalable expansion of PAX7+ myogenic progenitors and their purification are critical for practical application to cell replacement treatment of muscle degenerative diseases.


Assuntos
Técnicas de Cultura de Células/métodos , Células Musculares/citologia , Células Musculares/transplante , Desenvolvimento Muscular , Fator de Transcrição PAX7/genética , Células-Tronco Pluripotentes/citologia , Animais , Diferenciação Celular , Linhagem Celular , Proliferação de Células , Células Cultivadas , Humanos , Lentivirus/genética , Masculino , Camundongos , Células Musculares/metabolismo , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/fisiologia , Células-Tronco Pluripotentes/metabolismo , Regeneração , Transgenes
17.
Sci Rep ; 7(1): 1177, 2017 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-28446779

RESUMO

Mammalian skeletal muscles contain a number of heterogeneous cell populations. Our previous study characterized a unique population of myogenic lineage stem cells that can be isolated from adult mammalian skeletal muscles upon injury. These injury-induced muscle-derived stem cell-like cells (iMuSCs) displayed a multipotent state with sensitiveness and strong migration abilities. Here, we report that these iMuSCs have the capability to form neurospheres that represent multiple neural phenotypes. The induced neuronal cells expressed various neuron-specific proteins, their mRNA expression during neuronal differentiation recapitulated embryonic neurogenesis, they generated action potentials, and they formed functional synapses in vitro. Furthermore, the transplantation of iMuSCs or their cell extracts into the muscles of mdx mice (i.e., a mouse model of Duchenne Muscular Dystrophy [DMD]) could restore the morphology of their previously damaged neuromuscular junctions (NMJs), suggesting that the beneficial effects of iMuSCs may not be restricted to cell restoration alone, but also due to their transient paracrine actions. The current study reveals the essential role of iMuSCs in the restoration of NMJs related to injuries and diseases.


Assuntos
Diferenciação Celular , Músculo Esquelético/citologia , Neurogênese , Neurônios/fisiologia , Células-Tronco/fisiologia , Animais , Modelos Animais de Doenças , Camundongos Endogâmicos mdx , Distrofia Muscular de Duchenne/terapia , Transplante de Células-Tronco/métodos
18.
PLoS One ; 12(4): e0176190, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28441415

RESUMO

Pluripotent stem cells (PSC) hold great promise for the treatment of human skeletal muscle diseases. However, it remains challenging to convert PSC to skeletal muscle cells, and the mechanisms by which the master regulatory transcription factor, Pax7, promotes muscle stem (satellite) cell identity are not yet understood. We have taken advantage of PSC-derived skeletal muscle precursor cells (iPax7), wherein the induced expression of Pax7 robustly initiates the muscle program and enables the in vitro generation of precursors that seed the satellite cell compartment upon transplantation. Remarkably, we found that chromatin accessibility in myogenic precursors pre-figures subsequent activation of myogenic differentiation genes. We also found that Pax7 binding is generally restricted to euchromatic regions and excluded from H3K27 tri-methylated regions in muscle cells, suggesting that recruitment of this factor is circumscribed by chromatin state. Further, we show that Pax7 binding induces dramatic, localized remodeling of chromatin characterized by the acquisition of histone marks associated with enhancer activity and induction of chromatin accessibility in both muscle precursors and lineage-committed myoblasts. Conversely, removal of Pax7 leads to rapid reversal of these features on a subset of enhancers. Interestingly, another cluster of Pax7 binding sites is associated with a durably accessible and remodeled chromatin state after removal of Pax7, and persistent enhancer accessibility is associated with subsequent, proximal binding by the muscle regulatory factors, MyoD1 and myogenin. Our studies provide new insights into the epigenetic landscape of skeletal muscle stem cells and precursors and the role of Pax7 in satellite cell specification.


Assuntos
Cromatina/metabolismo , Células Musculares/metabolismo , Desenvolvimento Muscular/fisiologia , Fator de Transcrição PAX7/metabolismo , Células-Tronco Pluripotentes/metabolismo , Animais , Linhagem Celular , Camundongos , Músculo Esquelético/metabolismo , Proteína MyoD/metabolismo , Miogenina/metabolismo , Fator de Transcrição PAX7/genética , Células Satélites de Músculo Esquelético/metabolismo
19.
Can J Physiol Pharmacol ; 95(7): 773-786, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28177678

RESUMO

Many obstacles compromise the efficacy of bone marrow mesenchymal stem cells (BM-MSCs) by inducing apoptosis in the grafted BM-MSCs. The current study investigates the effect of melatonin on important mediators involved in survival of BM-MSCs in hydrogen peroxide (H2O2) apoptosis model. In brief, BM-MSCs were isolated, treated with melatonin, and then exposed to H2O2. Their viability was assessed by MTT assay and apoptotic fractions were evaluated through Annexin V, Hoechst staining, and ADP/ATP ratio. Oxidative stress biomarkers including ROS, total antioxidant power (TAP), superoxide dismutase (SOD) and catalase (CAT) activity, glutathione (GSH), thiol molecules, and lipid peroxidation (LPO) levels were determined. Secretion of inflammatory cytokines (TNF-α and IL-6) were measured by ELISA assay. The protein expression of caspase-3, Bax, and Bcl-2, was also evaluated by Western blotting. Melatonin pretreatment significantly increased viability and decreased apoptotic fraction of H2O2-exposed BM-MSCs. Melatonin also decreased ROS generation, as well as increasing the activity of SOD and CAT enzymes and GSH content. Secretion of inflammatory cytokines in H2O2-exposed cells was also reduced by melatonin. Expression of caspase-3 and Bax proteins in H2O2-exposed cells was diminished by melatonin pretreatment. The findings suggest that melatonin may be an effective protective agent against H2O2-induced oxidative stress and apoptosis in MSC.


Assuntos
Citoproteção/efeitos dos fármacos , Peróxido de Hidrogênio/toxicidade , Melatonina/farmacologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Adulto , Caspase 3/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Células-Tronco Mesenquimais/citologia , Estresse Oxidativo/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Adulto Jovem , Proteína X Associada a bcl-2/genética
20.
J Pineal Res ; 61(4): 411-425, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27555371

RESUMO

Osteoarthritis (OA) is a degenerative joint disease characterized by progressive erosion of articular cartilage. As chondrocytes are the only cell type forming the articular cartilage, their gradual loss is the main cause of OA. There is a substantial body of published research that suggests reactive oxygen species (ROS) are major causative factors for chondrocyte damage and OA development. Oxidative stress elicited by ROS is capable of oxidizing and subsequently disrupting cartilage homeostasis, promoting catabolism via induction of cell death and damaging numerous components of the joint. IL-1ß and TNF-α are crucial inflammatory factors that play pivotal roles in the pathogenesis of OA. In this process, the mitochondria are the major source of ROS production in cells, suggesting a role of mitochondrial dysfunction in this type of arthritis. This may also be promoted by inflammatory cytokines such as IL-1ß and TNF-α which contribute to chondrocyte death. In patients with OA, the expression of endoplasmic reticulum (ER) stress-associated molecules is positively correlated with cartilage degeneration. Melatonin and its metabolites are broad-spectrum antioxidants and free radical scavengers which regulate a variety of molecular pathways such as inflammation, proliferation, apoptosis, and metastasis in different pathophysiological situations. Herein, we review the effects of melatonin on OA, focusing on its ability to regulate apoptotic processes and ER and mitochondrial activity. We also evaluate likely protective effects of melatonin on OA pathogenesis.


Assuntos
Apoptose , Condrócitos/metabolismo , Melatonina/metabolismo , Osteoartrite/metabolismo , Transdução de Sinais , Animais , Condrócitos/patologia , Estresse do Retículo Endoplasmático , Humanos , Interleucina-1beta/metabolismo , Osteoartrite/patologia , Espécies Reativas de Oxigênio/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...