Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
1.
Cells ; 13(11)2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38891104

RESUMEN

Mutations in the DMD gene cause fatal Duchenne Muscular Dystrophy (DMD). An attractive therapeutic approach is autologous cell transplantation utilizing myogenic progenitors derived from induced pluripotent stem cells (iPSCs). Given that a significant number of DMD mutations occur between exons 45 and 55, we developed a gene knock-in approach to correct any mutations downstream of exon 44. We applied this approach to two DMD patient-specific iPSC lines carrying mutations in exons 45 and 51 and confirmed mini-DYSTROPHIN (mini-DYS) protein expression in corrected myotubes by western blot and immunofluorescence staining. Transplantation of gene-edited DMD iPSC-derived myogenic progenitors into NSG/mdx4Cv mice produced donor-derived myofibers, as shown by the dual expression of human DYSTROPHIN and LAMIN A/C. These findings further provide proof-of-concept for the use of programmable nucleases for the development of autologous iPSC-based therapy for muscular dystrophies.


Asunto(s)
Sistemas CRISPR-Cas , Distrofina , Exones , Células Madre Pluripotentes Inducidas , Distrofia Muscular de Duchenne , Mutación , Células Madre Pluripotentes Inducidas/metabolismo , Distrofina/genética , Distrofina/metabolismo , Humanos , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , Distrofia Muscular de Duchenne/patología , Sistemas CRISPR-Cas/genética , Exones/genética , Mutación/genética , Animales , Ratones , Edición Génica/métodos , Fibras Musculares Esqueléticas/metabolismo
2.
NPJ Regen Med ; 9(1): 16, 2024 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-38575647

RESUMEN

Pluripotent stem cell (PSC)-based cell therapy is an attractive option for the treatment of multiple human disorders, including muscular dystrophies. While in vitro differentiating PSCs can generate large numbers of human lineage-specific tissue, multiple studies evidenced that these cell populations mostly display embryonic/fetal features. We previously demonstrated that transplantation of PSC-derived myogenic progenitors provides long-term engraftment and functional improvement in several dystrophic mouse models, but it remained unknown whether donor-derived myofibers mature to match adult tissue. Here, we transplanted iPAX7 myogenic progenitors into muscles of non-dystrophic and dystrophic mice and compared the transcriptional landscape of human grafts with respective in vitro-differentiated iPAX7 myotubes as well as human skeletal muscle biospecimens. Pairing bulk RNA sequencing with computational deconvolution of human reads, we were able to pinpoint key myogenic changes that occur during the in vitro-to-in vivo transition, confirm developmental maturity, and consequently evaluate their applicability for cell-based therapies.

3.
Int J Mol Sci ; 25(5)2024 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-38473751

RESUMEN

The diaphragm muscle is essential for breathing, and its dysfunctions can be fatal. Many disorders affect the diaphragm, including muscular dystrophies. Despite the clinical relevance of targeting the diaphragm, there have been few studies evaluating diaphragm function following a given experimental treatment, with most of these involving anti-inflammatory drugs or gene therapy. Cell-based therapeutic approaches have shown success promoting muscle regeneration in several mouse models of muscular dystrophy, but these have focused mainly on limb muscles. Here we show that transplantation of as few as 5000 satellite cells directly into the diaphragm results in consistent and robust myofiber engraftment in dystrophin- and fukutin-related protein-mutant dystrophic mice. Transplanted cells also seed the stem cell reservoir, as shown by the presence of donor-derived satellite cells. Force measurements showed enhanced diaphragm strength in engrafted muscles. These findings demonstrate the feasibility of cell transplantation to target the diseased diaphragm and improve its contractility.


Asunto(s)
Distrofia Muscular de Duchenne , Ratones , Animales , Distrofia Muscular de Duchenne/genética , Diafragma , Ratones Endogámicos mdx , Músculo Esquelético , Trasplante de Células
4.
Cells ; 12(8)2023 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-37190056

RESUMEN

Pluripotent stem (PS) cells enable the scalable production of tissue-specific derivatives with therapeutic potential for various clinical applications, including muscular dystrophies. Given the similarity to human counterparts, the non-human primate (NHP) is an ideal preclinical model to evaluate several questions, including delivery, biodistribution, and immune response. While the generation of human-induced PS (iPS)-cell-derived myogenic progenitors is well established, there have been no data for NHP counterparts, probably due to the lack of an efficient system to differentiate NHP iPS cells towards the skeletal muscle lineage. Here, we report the generation of three independent Macaca fascicularis iPS cell lines and their myogenic differentiation using PAX7 conditional expression. The whole-transcriptome analysis confirmed the successful sequential induction of mesoderm, paraxial mesoderm, and myogenic lineages. NHP myogenic progenitors efficiently gave rise to myotubes under appropriate in vitro differentiation conditions and engrafted in vivo into the TA muscles of NSG and FKRP-NSG mice. Lastly, we explored the preclinical potential of these NHP myogenic progenitors in a single wild-type NHP recipient, demonstrating engraftment and characterizing the interaction with the host immune response. These studies establish an NHP model system through which iPS-cell-derived myogenic progenitors can be studied.


Asunto(s)
Células Madre Pluripotentes Inducidas , Células Madre Pluripotentes , Animales , Ratones , Células Madre Pluripotentes Inducidas/metabolismo , Distribución Tisular , Células Madre Pluripotentes/metabolismo , Músculo Esquelético/metabolismo , Primates , Pentosiltransferasa/metabolismo
5.
Methods Mol Biol ; 2640: 129-142, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36995592

RESUMEN

Pluripotent stem cells have a multitude of potential applications in the areas of disease modeling, drug screening, and cell-based therapies for genetic diseases, including muscular dystrophies. The advent of induced pluripotent stem cell technology allows for the facile derivation of disease-specific pluripotent stem cells for any given patient. Targeted in vitro differentiation of pluripotent stem cells into the muscle lineage is a key step to enable all these applications. Transgene-based differentiation using conditional expression of the transcription factor PAX7 leads to the efficient derivation of an expandable and homogeneous population of myogenic progenitors suitable for both in vitro and in vivo applications. Here, we describe an optimized protocol for the derivation and expansion of myogenic progenitors from pluripotent stem cells using conditional expression of PAX7. Importantly, we further describe an optimized procedure for the terminal differentiation of myogenic progenitors into more mature myotubes, which are better suited for in vitro disease modeling and drug screening studies.


Asunto(s)
Células Madre Pluripotentes Inducidas , Distrofias Musculares , Células Madre Pluripotentes , Humanos , Fibras Musculares Esqueléticas , Distrofias Musculares/metabolismo , Diferenciación Celular , Desarrollo de Músculos/genética
6.
Circ Res ; 132(4): 519-540, 2023 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-36795845

RESUMEN

During cardiac development and morphogenesis, cardiac progenitor cells differentiate into cardiomyocytes that expand in number and size to generate the fully formed heart. Much is known about the factors that regulate initial differentiation of cardiomyocytes, and there is ongoing research to identify how these fetal and immature cardiomyocytes develop into fully functioning, mature cells. Accumulating evidence indicates that maturation limits proliferation and conversely proliferation occurs rarely in cardiomyocytes of the adult myocardium. We term this oppositional interplay the proliferation-maturation dichotomy. Here we review the factors that are involved in this interplay and discuss how a better understanding of the proliferation-maturation dichotomy could advance the utility of human induced pluripotent stem cell-derived cardiomyocytes for modeling in 3-dimensional engineered cardiac tissues to obtain truly adult-level function.


Asunto(s)
Células Madre Pluripotentes Inducidas , Ingeniería de Tejidos , Humanos , Ingeniería de Tejidos/métodos , Miocitos Cardíacos/fisiología , Miocardio , Diferenciación Celular/fisiología , Proliferación Celular
7.
EMBO Mol Med ; 15(2): e15315, 2023 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-36479608

RESUMEN

This commentary provides a brief overview of the steps necessary for the generation of an induced pluripotent stem (iPS) cell-derived clinical grade product. This process requires extensive, proper documentation as well as a thoughtful and systematic optimization of the manufacturing methods to ensure maintenance of the key biological features of the product, compliance with current good manufacturing practices (cGMP), and most importantly patient safety. The scale-up and optimization also ideally include the identification of efficient and cost-effective purification/isolation and expansion of the target cell population.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Células Madre Pluripotentes Inducidas , Humanos , Diferenciación Celular
8.
Cells ; 13(1)2023 12 29.
Artículo en Inglés | MEDLINE | ID: mdl-38201280

RESUMEN

In vitro-generated pluripotent stem cell (PSC)-derived Pax3-induced (iPax3) myogenic progenitors display an embryonic transcriptional signature, but upon engraftment, the profile of re-isolated iPax3 donor-derived satellite cells changes toward similarity with postnatal satellite cells, suggesting that engrafted PSC-derived myogenic cells remodel their transcriptional signature upon interaction within the adult muscle environment. Here, we show that engrafted myogenic progenitors also remodel their metabolic state. Assessment of oxygen consumption revealed that exposure to the adult muscle environment promotes overt changes in mitochondrial bioenergetics, as shown by the substantial suppression of energy requirements in re-isolated iPax3 donor-derived satellite cells compared to their in vitro-generated progenitors. Mass spectrometry-based metabolomic profiling further confirmed the relationship of engrafted iPax3 donor-derived cells to adult satellite cells. The fact that in vitro-generated myogenic progenitors remodel their bioenergetic signature upon in vivo exposure to the adult muscle environment may have important implications for therapeutic applications.


Asunto(s)
Células Madre Pluripotentes , Células Satélite del Músculo Esquelético , Adulto , Humanos , Metabolómica , Consumo de Oxígeno , Músculos
9.
NPJ Regen Med ; 7(1): 43, 2022 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-36056021

RESUMEN

Facioscapulohumeral muscular dystrophy (FSHD) is a genetically dominant progressive myopathy caused by improper silencing of the DUX4 gene, leading to fibrosis, muscle atrophy, and fatty replacement. Approaches focused on muscle regeneration through the delivery of stem cells represent an attractive therapeutic option for muscular dystrophies. To investigate the potential for cell transplantation in FSHD, we have used the doxycycline-regulated iDUX4pA-HSA mouse model in which low-level DUX4 can be induced in skeletal muscle. We find that mouse pluripotent stem cell (PSC)-derived myogenic progenitors engraft in muscle actively undergoing DUX4-mediated degeneration. Donor-derived muscle tissue displayed reduced fibrosis and importantly, engrafted muscles showed improved contractile specific force compared to non-transplanted controls. These data demonstrate the feasibility of replacement of diseased muscle with PSC-derived myogenic progenitors in a mouse model for FSHD, and highlight the potential for the clinical benefit of such a cell therapy approach.

10.
Cell Mol Life Sci ; 79(8): 406, 2022 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-35802202

RESUMEN

Muscular dystrophy encompasses a large number of heterogeneous genetic disorders characterized by progressive and devastating muscle wasting. Cell-based replacement strategies aimed at promoting skeletal muscle regeneration represent a candidate therapeutic approach to treat muscular dystrophies. Due to the difficulties of obtaining large numbers of stem cells from a muscle biopsy as well as expanding these in vitro, pluripotent stem cells (PSCs) represent an attractive cell source for the generation of myogenic progenitors, given that PSCs can repeatedly produce large amounts of lineage-specific tissue, representing an unlimited source of cells for therapy. In this review, we focus on the progress to date on different methods for the generation of human PSC-derived myogenic progenitor cells, their regenerative capabilities upon transplantation, their potential for allogeneic and autologous transplantation, as well as the specific challenges to be considered for future therapeutic applications.


Asunto(s)
Células Madre Pluripotentes Inducidas , Distrofias Musculares , Células Madre Pluripotentes , Diferenciación Celular , Humanos , Desarrollo de Músculos , Músculo Esquelético/fisiología , Distrofias Musculares/terapia , Trasplante de Células Madre
11.
Front Cell Dev Biol ; 10: 806564, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35663393

RESUMEN

The heart is one of the least regenerative organs. This is in large part due to the inability of adult mammalian cardiomyocytes to proliferate and divide. In recent years, a number of small molecules and molecular targets have been identified to stimulate cardiomyocyte proliferation, including p38 inhibition, YAP-Tead activation, fibroblast growth factor 1 and Neuregulin 1. Despite these exciting initial findings, a therapeutic approach to enhance cardiomyocyte proliferation in vivo is still lacking. We hypothesized that a more comprehensive in vitro validation using live-cell imaging and assessment of the proliferative effects on various cardiomyocyte sources might identify the most potent proliferative stimuli. Here, we used previously published stimuli to determine their proliferative effect on cardiomyocytes from different species and isolated from different developmental timepoints. Although all stimuli enhanced DNA synthesis and Histone H3 phosphorylation in neonatal rat ventricular cardiomyocytes to similar degrees, these effects varied substantially in mouse cardiomyocytes and human iPSC-derived cardiomyocytes. Our results highlight p21 inhibition and Yap-Tead activation as potent proliferative strategies to induce cultured cardiomyocyte cell cycle activity across mouse, rat and human cardiomyocytes.

12.
Stem Cell Reports ; 16(12): 2900-2912, 2021 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-34798067

RESUMEN

One major challenge in realizing cell-based therapy for treating muscle-wasting disorders is the difficulty in obtaining therapeutically meaningful amounts of engraftable cells. We have previously described a method to generate skeletal myogenic progenitors with exceptional engraftability from pluripotent stem cells via teratoma formation. Here, we show that these cells are functionally expandable in vitro while retaining their in vivo regenerative potential. Within 37 days in culture, teratoma-derived skeletal myogenic progenitors were expandable to a billion-fold. Similar to their freshly sorted counterparts, the expanded cells expressed PAX7 and were capable of forming multinucleated myotubes in vitro. Importantly, these cells remained highly regenerative in vivo. Upon transplantation, the expanded cells formed new DYSTROPHIN+ fibers that reconstituted up to 40% of tibialis anterior muscle volume and repopulated the muscle stem cell pool. Our study thereby demonstrates the possibility of producing large quantities of engraftable skeletal myogenic cells for transplantation.


Asunto(s)
Células Madre Pluripotentes Inducidas/patología , Desarrollo de Músculos , Músculo Esquelético/patología , Trasplante de Células Madre , Teratoma/patología , Animales , Compartimento Celular , Diferenciación Celular , Proliferación Celular , Ratones , Fibras Musculares Esqueléticas , RNA-Seq , Molécula 1 de Adhesión Celular Vascular/metabolismo
13.
Stem Cell Reports ; 16(11): 2752-2767, 2021 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-34653404

RESUMEN

Fukutin-related protein (FKRP) is a glycosyltransferase involved in glycosylation of alpha-dystroglycan (α-DG). Mutations in FKRP are associated with muscular dystrophies (MD) ranging from limb-girdle LGMDR9 to Walker-Warburg Syndrome (WWS), a severe type of congenital MD. Although hypoglycosylation of α-DG is the main hallmark of this group of diseases, a full understanding of the underlying pathophysiology is still missing. Here, we investigated molecular mechanisms impaired by FKRP mutations in pluripotent stem (PS) cell-derived myotubes. FKRP-deficient myotubes show transcriptome alterations in genes involved in extracellular matrix receptor interactions, calcium signaling, PI3K-Akt pathway, and lysosomal function. Accordingly, using a panel of patient-specific LGMDR9 and WWS induced PS cell-derived myotubes, we found a significant reduction in the autophagy-lysosome pathway for both disease phenotypes. In addition, we show that WWS myotubes display decreased ERK1/2 activity and increased apoptosis, which were restored in gene edited myotubes. Our results suggest the autophagy-lysosome pathway and apoptosis may contribute to the FKRP-associated MD pathogenesis.


Asunto(s)
Apoptosis/genética , Autofagia/genética , Predisposición Genética a la Enfermedad/genética , Distrofias Musculares/genética , Mutación , Pentosiltransferasa/genética , Línea Celular , Glicosilación , Humanos , Lisosomas/genética , Lisosomas/metabolismo , Fibras Musculares Esqueléticas/citología , Fibras Musculares Esqueléticas/metabolismo , Distrofias Musculares/metabolismo , Distrofias Musculares/patología , Distrofia Muscular de Cinturas/genética , Distrofia Muscular de Cinturas/metabolismo , Distrofia Muscular de Cinturas/patología , Pentosiltransferasa/metabolismo , Células Madre Pluripotentes/metabolismo , RNA-Seq/métodos , Transducción de Señal/genética , Transcriptoma/genética , Síndrome de Walker-Warburg/genética , Síndrome de Walker-Warburg/metabolismo , Síndrome de Walker-Warburg/patología
14.
Front Genet ; 12: 702547, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34408774

RESUMEN

This article will review myogenic cell transplantation for congenital and acquired diseases of skeletal muscle. There are already a number of excellent reviews on this topic, but they are mostly focused on a specific disease, muscular dystrophies and in particular Duchenne Muscular Dystrophy. There are also recent reviews on cell transplantation for inflammatory myopathies, volumetric muscle loss (VML) (this usually with biomaterials), sarcopenia and sphincter incontinence, mainly urinary but also fecal. We believe it would be useful at this stage, to compare the same strategy as adopted in all these different diseases, in order to outline similarities and differences in cell source, pre-clinical models, administration route, and outcome measures. This in turn may help to understand which common or disease-specific problems have so far limited clinical success of cell transplantation in this area, especially when compared to other fields, such as epithelial cell transplantation. We also hope that this may be useful to people outside the field to get a comprehensive view in a single review. As for any cell transplantation procedure, the choice between autologous and heterologous cells is dictated by a number of criteria, such as cell availability, possibility of in vitro expansion to reach the number required, need for genetic correction for many but not necessarily all muscular dystrophies, and immune reaction, mainly to a heterologous, even if HLA-matched cells and, to a minor extent, to the therapeutic gene product, a possible antigen for the patient. Finally, induced pluripotent stem cell derivatives, that have entered clinical experimentation for other diseases, may in the future offer a bank of immune-privileged cells, available for all patients and after a genetic correction for muscular dystrophies and other myopathies.

15.
Stem Cell Res ; 55: 102496, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34411972

RESUMEN

Satellite cells represent the main myogenic population accounting for skeletal muscle homeostasis and regeneration. While our knowledge of the signaling pathways controlling satellite cell regenerative capability is increasing, the underlying epigenetic mechanisms are still not clear, especially in the case of human satellite cells. Here, by performing chromatin accessibility profiling (ATAC-seq) in samples isolated from human and murine muscles, we investigated the changes in the epigenetic landscape occurring during the transition from activated satellite cells to myoblasts. Our analysis identifies a compendium of putative regulatory elements defining human activated satellite cells and myoblasts, respectively. A subset of these differentially accessible loci is shared by both murine and human satellite cells, includes elements associated with known self-renewal regulators, and is enriched for motifs bound by transcription factors participating in satellite cell regulation. Integration of transcriptional and epigenetic data reveals that known regulators of metabolic gene expression, such as PPARGC1A, represent potential PAX7 targets. Through characterization of genomic networks and the underlying effectors, our data represent an important starting point for decoding and manipulating the molecular mechanisms underlying human satellite cell muscle regenerative potential.


Asunto(s)
Cromatina , Células Satélite del Músculo Esquelético , Animales , Diferenciación Celular , Humanos , Ratones , Desarrollo de Músculos/genética , Músculo Esquelético , Factor de Transcripción PAX7/genética
16.
Cell Rep ; 36(2): 109360, 2021 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-34260922

RESUMEN

Mutations in the fukutin-related protein (FKRP) gene result in a broad spectrum of muscular dystrophy (MD) phenotypes, including the severe Walker-Warburg syndrome (WWS). Here, we develop a gene-editing approach that replaces the entire mutant open reading frame with the wild-type sequence to universally correct all FKRP mutations. We apply this approach to correct FKRP mutations in induced pluripotent stem (iPS) cells derived from patients displaying broad clinical severity. Our findings show rescue of functional α-dystroglycan (α-DG) glycosylation in gene-edited WWS iPS cell-derived myotubes. Transplantation of gene-corrected myogenic progenitors in the FKRPP448L-NSG mouse model gives rise to myofiber and satellite cell engraftment and, importantly, restoration of α-DG functional glycosylation in vivo. These findings suggest the potential feasibility of using CRISPR-Cas9 technology in combination with patient-specific iPS cells for the future development of autologous cell transplantation for FKRP-associated MDs.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos , Distroglicanos/genética , Terapia Genética , Distrofias Musculares/genética , Distrofias Musculares/terapia , Pentosiltransferasa/genética , Animales , Preescolar , Distroglicanos/metabolismo , Glicosilación , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Masculino , Ratones Mutantes , Fibras Musculares Esqueléticas/metabolismo , Mutación/genética , Fenotipo , Trasplante Autólogo , Síndrome de Walker-Warburg/genética
17.
Trends Cell Biol ; 31(3): 197-210, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33272829

RESUMEN

Fukutin-related protein (FKRP) is a glycosyltransferase involved in the functional glycosylation of α-dystroglycan (DG), a key component in the link between the cytoskeleton and the extracellular matrix (ECM). Mutations in FKRP lead to dystroglycanopathies with broad severity, including limb-girdle and congenital muscular dystrophy. Studies over the past 5 years have elucidated the function of FKRP, which has expanded the number of therapeutic opportunities for patients carrying FKRP mutations. These include small molecules, gene delivery, and cell therapy. Here we summarize recent findings on the function of FKRP and describe available models for studying diseases and testing therapeutics. Lastly, we highlight preclinical studies that hold potential for the treatment of FKRP-associated dystroglycanopathies.


Asunto(s)
Distrofias Musculares , Pentosiltransferasa , Distroglicanos/genética , Distroglicanos/metabolismo , Glicosilación , Glicosiltransferasas/metabolismo , Humanos , Distrofias Musculares/genética , Distrofias Musculares/terapia , Pentosiltransferasa/genética , Pentosiltransferasa/metabolismo
18.
Stem Cell Reports ; 16(1): 10-19, 2021 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-33275879

RESUMEN

Inducible expression of PAX7 in differentiating pluripotent stem cells (PSCs) allows massively scalable generation of human myogenic progenitors, which upon transplantation into dystrophic muscles give rise to donor-derived myofibers and satellite cells. Therefore, PSC-derived PAX7+ myogenic progenitors represent an attractive therapeutic approach to promote muscle regeneration. Work to date has used lentiviral vectors (LVs) that randomly integrate inducible PAX7 transgenes. Here, we investigated whether equivalent induction of the myogenic program could be achieved by targeting the PAX7 transgene into genomic safe harbor (GSH) sites. Across multiple PSC lines, we find that this approach consistently generates expandable myogenic progenitors in vitro, although scalability of expansion is moderately reduced compared with the LV approach. Importantly, transplantation of GSH-targeted myogenic progenitors produces robust engraftment, comparable with LV counterparts. These findings provide proof of concept for the use of GSH targeting as a potential alternative approach to generate therapeutic PSC-derived myogenic progenitors for clinical applications.


Asunto(s)
Factor de Transcripción PAX7/genética , Células Madre/metabolismo , Animales , Diferenciación Celular , Línea Celular , Modelos Animales de Enfermedad , Distrofina/genética , Cuerpos Embrioides/citología , Cuerpos Embrioides/metabolismo , Sitios Genéticos , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Humanos , Lentivirus/genética , Ratones , Desarrollo de Músculos , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/patología , Distrofia Muscular de Duchenne/terapia , Factor de Transcripción PAX7/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Células Madre Pluripotentes/trasplante , Trasplante de Células Madre , Células Madre/citología
19.
Nat Commun ; 11(1): 6222, 2020 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-33277476

RESUMEN

Using Hi-C, promoter-capture Hi-C (pCHi-C), and other genome-wide approaches in skeletal muscle progenitors that inducibly express a master transcription factor, Pax7, we systematically characterize at high-resolution the spatio-temporal re-organization of compartments and promoter-anchored interactions as a consequence of myogenic commitment and differentiation. We identify key promoter-enhancer interaction motifs, namely, cliques and networks, and interactions that are dependent on Pax7 binding. Remarkably, Pax7 binds to a majority of super-enhancers, and together with a cadre of interacting transcription factors, assembles feed-forward regulatory loops. During differentiation, epigenetic memory and persistent looping are maintained at a subset of Pax7 enhancers in the absence of Pax7. We also identify and functionally validate a previously uncharacterized Pax7-bound enhancer hub that regulates the essential myosin heavy chain cluster during skeletal muscle cell differentiation. Our studies lay the groundwork for understanding the role of Pax7 in orchestrating changes in the three-dimensional chromatin conformation in muscle progenitors.


Asunto(s)
Diferenciación Celular/genética , Cromatina/genética , Células Madre Embrionarias de Ratones/metabolismo , Desarrollo de Músculos/genética , Músculo Esquelético/metabolismo , Células 3T3-L1 , Animales , Células Cultivadas , Cromatina/metabolismo , Perfilación de la Expresión Génica/métodos , Ontología de Genes , Redes Reguladoras de Genes , Humanos , Ratones , Músculo Esquelético/citología , Factor de Transcripción PAX7/genética , Factor de Transcripción PAX7/metabolismo
20.
Skelet Muscle ; 10(1): 17, 2020 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-32493438

RESUMEN

BACKGROUND: Skeletal muscle function is essential for health, and it depends on the proper activity of myofibers and their innervating motor neurons. Each adult muscle is composed of different types of myofibers with distinct contractile and metabolic characteristics. The proper balance of myofiber types is disrupted in most muscle degenerative disorders, representing another factor compromising muscle function. One promising therapeutic approach for the treatment of these diseases is cell replacement based on the targeted differentiation of pluripotent stem cells (PSCs) towards the myogenic lineage. We have previously shown that transient induction of Pax3 or Pax7 in PSCs allows for the generation of skeletal myogenic progenitors endowed with myogenic regenerative potential, but whether they contribute to different fiber types remains unknown. RESULTS: Here, we investigate the fiber type composition of mouse PSC-derived myofibers upon their transplantation into dystrophic and non-dystrophic mice. Our data reveal that PSC-derived myofibers express slow and oxidative myosin heavy-chain isoforms, along with developmental myosins, regardless of the recipient background. Furthermore, transplantation of the mononuclear cell fraction re-isolated from primary grafts into secondary recipients results in myofibers that maintain preferential expression of slow and oxidative myosin heavy-chain isoforms but no longer express developmental myosins, thus indicating postnatal composition. CONCLUSIONS: Considering oxidative fibers are commonly spared in the context of dystrophic pathogenesis, this feature of PSC-derived myofibers could be advantageous for therapeutic applications.


Asunto(s)
Fibras Musculares Esqueléticas/metabolismo , Cadenas Pesadas de Miosina/genética , Células Madre Pluripotentes/citología , Animales , Diferenciación Celular , Línea Celular , Células Cultivadas , Regulación del Desarrollo de la Expresión Génica , Ratones , Desarrollo de Músculos , Fibras Musculares Esqueléticas/citología , Cadenas Pesadas de Miosina/metabolismo , Células Madre Pluripotentes/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA