Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Adv Exp Med Biol ; 1147: 319-344, 2019.
Article in English | MEDLINE | ID: mdl-31147885

ABSTRACT

The muscular dystrophies are an heterogeneous group of inherited myopathies characterised by the progressive wasting of skeletal muscle tissue. Pericytes have been shown to make muscle in vitro and to contribute to skeletal muscle regeneration in several animal models, although recent data has shown this to be controversial. In fact, some pericyte subpopulations have been shown to contribute to fibrosis and adipose deposition in muscle. In this chapter, we explore the identity and the multifaceted role of pericytes in dystrophic muscle, potential therapeutic applications and the current need to overcome the hurdles of characterisation (both to identify pericyte subpopulations and track cell fate), to prevent deleterious differentiation towards myogenic-inhibiting subpopulations, and to improve cell proliferation and engraftment efficacy.


Subject(s)
Muscular Dystrophies , Pericytes , Animals , Cell Differentiation , Muscle Development , Muscle, Skeletal , Regeneration
2.
Stem Cell Reports ; 12(3): 461-473, 2019 03 05.
Article in English | MEDLINE | ID: mdl-30745033

ABSTRACT

Satellite cells are responsible for skeletal muscle regeneration. Upon activation, they proliferate as transient amplifying myoblasts, most of which fuse into regenerating myofibers. Despite their remarkable differentiation potential, these cells have limited migration capacity, which curtails clinical use for widespread forms of muscular dystrophy. Conversely, skeletal muscle perivascular cells have less myogenic potential but better migration capacity than satellite cells. Here we show that modulation of Notch and PDGF pathways, involved in developmental specification of pericytes, induces perivascular cell features in adult mouse and human satellite cell-derived myoblasts. DLL4 and PDGF-BB-treated cells express markers of perivascular cells and associate with endothelial networks while also upregulating markers of satellite cell self-renewal. Moreover, treated cells acquire trans-endothelial migration ability while remaining capable of engrafting skeletal muscle upon intramuscular transplantation. These results extend our understanding of muscle stem cell fate plasticity and provide a druggable pathway with clinical relevance for muscle cell therapy.


Subject(s)
Biomarkers/metabolism , Cell Movement/physiology , Receptors, Notch/metabolism , Receptors, Platelet-Derived Growth Factor/metabolism , Satellite Cells, Skeletal Muscle/metabolism , Signal Transduction/physiology , Stem Cells/metabolism , Animals , Endothelial Cells/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Muscle Development/physiology , Muscle, Skeletal/metabolism , Myoblasts/metabolism , Pericytes/metabolism , Regeneration/physiology , Up-Regulation/physiology
3.
Cell Rep ; 23(3): 899-908, 2018 Apr 17.
Article in English | MEDLINE | ID: mdl-29669293

ABSTRACT

Generating human skeletal muscle models is instrumental for investigating muscle pathology and therapy. Here, we report the generation of three-dimensional (3D) artificial skeletal muscle tissue from human pluripotent stem cells, including induced pluripotent stem cells (iPSCs) from patients with Duchenne, limb-girdle, and congenital muscular dystrophies. 3D skeletal myogenic differentiation of pluripotent cells was induced within hydrogels under tension to provide myofiber alignment. Artificial muscles recapitulated characteristics of human skeletal muscle tissue and could be implanted into immunodeficient mice. Pathological cellular hallmarks of incurable forms of severe muscular dystrophy could be modeled with high fidelity using this 3D platform. Finally, we show generation of fully human iPSC-derived, complex, multilineage muscle models containing key isogenic cellular constituents of skeletal muscle, including vascular endothelial cells, pericytes, and motor neurons. These results lay the foundation for a human skeletal muscle organoid-like platform for disease modeling, regenerative medicine, and therapy development.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Models, Biological , Tissue Engineering , Cell Differentiation , Cell Lineage , Humans , Hydrogels/chemistry , Muscle Development , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophies/metabolism , Muscular Dystrophies/pathology , Tissue Scaffolds/chemistry
4.
EMBO Mol Med ; 10(2): 254-275, 2018 02.
Article in English | MEDLINE | ID: mdl-29242210

ABSTRACT

Transferring large or multiple genes into primary human stem/progenitor cells is challenged by restrictions in vector capacity, and this hurdle limits the success of gene therapy. A paradigm is Duchenne muscular dystrophy (DMD), an incurable disorder caused by mutations in the largest human gene: dystrophin. The combination of large-capacity vectors, such as human artificial chromosomes (HACs), with stem/progenitor cells may overcome this limitation. We previously reported amelioration of the dystrophic phenotype in mice transplanted with murine muscle progenitors containing a HAC with the entire dystrophin locus (DYS-HAC). However, translation of this strategy to human muscle progenitors requires extension of their proliferative potential to withstand clonal cell expansion after HAC transfer. Here, we show that reversible cell immortalisation mediated by lentivirally delivered excisable hTERT and Bmi1 transgenes extended cell proliferation, enabling transfer of a novel DYS-HAC into DMD satellite cell-derived myoblasts and perivascular cell-derived mesoangioblasts. Genetically corrected cells maintained a stable karyotype, did not undergo tumorigenic transformation and retained their migration ability. Cells remained myogenic in vitro (spontaneously or upon MyoD induction) and engrafted murine skeletal muscle upon transplantation. Finally, we combined the aforementioned functions into a next-generation HAC capable of delivering reversible immortalisation, complete genetic correction, additional dystrophin expression, inducible differentiation and controllable cell death. This work establishes a novel platform for complex gene transfer into clinically relevant human muscle progenitors for DMD gene therapy.


Subject(s)
Chromosomes, Artificial, Human , Dystrophin/genetics , Genetic Therapy/methods , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/therapy , Animals , Cells, Cultured , Genetic Vectors , Humans , Mice , Models, Animal , Mutation
SELECTION OF CITATIONS
SEARCH DETAIL
...