Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Med ; 4(1): 31-50.e8, 2023 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-36417917

RESUMEN

BACKGROUND: Adeno-associated virus (AAV) vectors are a promising vehicle for noninvasive gene delivery to the central nervous system via intravenous infusion. However, naturally occurring serotypes have a limited ability to transduce the brain, and translating engineered capsids from mice to nonhuman primates has proved challenging. METHODS: In this study, we use an mRNA-based directed-evolution strategy in multiple strains of mice as well as a de novo selection in cynomolgus macaques to identify families of engineered vectors with increased potency in the brain and decreased tropism for the liver. FINDINGS: We compare the transgene expression capabilities of several engineered vectors and show that while some of our novel macaque-derived variants significantly outperform AAV9 in transducing the macaque brain following systemic administration, mouse-derived variants-both those identified in this study and those reported by other groups-universally do not. CONCLUSIONS: Together, the results of this work introduce a class of primate-derived engineered AAV capsids with increased therapeutic potential and highlight the critical need for using appropriate animal models to both identify and evaluate novel AAVs intended for delivery to the human central nervous system. FUNDING: This work was funded primarily through an anonymous philanthropic gift to the P.C.S. lab at the Broad Institute of MIT and Harvard and by a grant from the Howard Hughes Medical Institute to P.C.S.


Asunto(s)
Cápside , Macaca , Humanos , Animales , Ratones , Cápside/metabolismo , Macaca/genética , Vectores Genéticos/genética , Sistema Nervioso Central/metabolismo , Transgenes , Primates/genética , Dependovirus/genética , Dependovirus/metabolismo
2.
Cell ; 184(19): 4919-4938.e22, 2021 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-34506722

RESUMEN

Replacing or editing disease-causing mutations holds great promise for treating many human diseases. Yet, delivering therapeutic genetic modifiers to specific cells in vivo has been challenging, particularly in large, anatomically distributed tissues such as skeletal muscle. Here, we establish an in vivo strategy to evolve and stringently select capsid variants of adeno-associated viruses (AAVs) that enable potent delivery to desired tissues. Using this method, we identify a class of RGD motif-containing capsids that transduces muscle with superior efficiency and selectivity after intravenous injection in mice and non-human primates. We demonstrate substantially enhanced potency and therapeutic efficacy of these engineered vectors compared to naturally occurring AAV capsids in two mouse models of genetic muscle disease. The top capsid variants from our selection approach show conserved potency for delivery across a variety of inbred mouse strains, and in cynomolgus macaques and human primary myotubes, with transduction dependent on target cell expressed integrin heterodimers.


Asunto(s)
Cápside/metabolismo , Dependovirus/metabolismo , Evolución Molecular Dirigida , Técnicas de Transferencia de Gen , Músculo Esquelético/metabolismo , Secuencia de Aminoácidos , Animales , Cápside/química , Células Cultivadas , Modelos Animales de Enfermedad , Células HEK293 , Humanos , Integrinas/metabolismo , Macaca fascicularis , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Fibras Musculares Esqueléticas/metabolismo , Distrofia Muscular de Duchenne/patología , Distrofia Muscular de Duchenne/terapia , Miopatías Estructurales Congénitas/patología , Miopatías Estructurales Congénitas/terapia , Multimerización de Proteína , Proteínas Tirosina Fosfatasas no Receptoras/genética , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo , Proteínas Tirosina Fosfatasas no Receptoras/uso terapéutico , ARN Guía de Kinetoplastida/metabolismo , Recombinación Genética/genética , Especificidad de la Especie , Transgenes
3.
Skelet Muscle ; 10(1): 28, 2020 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-33036659

RESUMEN

Satellite cells are the canonical muscle stem cells that regenerate damaged skeletal muscle. Loss of function of these cells has been linked to reduced muscle repair capacity and compromised muscle health in acute muscle injury and congenital neuromuscular diseases. To identify new pathways that can prevent loss of skeletal muscle function or enhance regenerative potential, we established an imaging-based screen capable of identifying small molecules that promote the expansion of freshly isolated satellite cells. We found several classes of receptor tyrosine kinase (RTK) inhibitors that increased freshly isolated satellite cell numbers in vitro. Further exploration of one of these compounds, the RTK inhibitor CEP-701 (also known as lestaurtinib), revealed potent activity on mouse satellite cells both in vitro and in vivo. This expansion potential was not seen upon exposure of proliferating committed myoblasts or non-myogenic fibroblasts to CEP-701. When delivered subcutaneously to acutely injured animals, CEP-701 increased both the total number of satellite cells and the rate of muscle repair, as revealed by an increased cross-sectional area of regenerating fibers. Moreover, freshly isolated satellite cells expanded ex vivo in the presence of CEP-701 displayed enhanced muscle engraftment potential upon in vivo transplantation. We provide compelling evidence that certain RTKs, and in particular RET, regulate satellite cell expansion during muscle regeneration. This study demonstrates the power of small molecule screens of even rare adult stem cell populations for identifying stem cell-targeting compounds with therapeutic potential.


Asunto(s)
Células Madre Embrionarias Humanas/efectos de los fármacos , Desarrollo de Músculos , Inhibidores de Proteínas Quinasas/farmacología , Células Satélite del Músculo Esquelético/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Carbazoles/farmacología , Proliferación Celular , Células Cultivadas , Furanos/farmacología , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/fisiología , Humanos , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/citología , Músculo Esquelético/fisiología , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Regeneración , Células Satélite del Músculo Esquelético/citología , Células Satélite del Músculo Esquelético/fisiología
4.
Cell Rep ; 27(4): 1254-1264.e7, 2019 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-31018138

RESUMEN

In vivo delivery of genome-modifying enzymes holds significant promise for therapeutic applications and functional genetic screening. Delivery to endogenous tissue stem cells, which provide an enduring source of cell replacement during homeostasis and regeneration, is of particular interest. Here, we use a sensitive Cre/lox fluorescent reporter system to test the efficiency of genome modification following in vivo transduction by adeno-associated viruses (AAVs) in tissue stem and progenitor cells. We combine immunophenotypic analyses with in vitro and in vivo assays of stem cell function to reveal effective targeting of skeletal muscle satellite cells, mesenchymal progenitors, hematopoietic stem cells, and dermal cell subsets using multiple AAV serotypes. Genome modification rates achieved through this system reached >60%, and modified cells retained key functional properties. This study establishes a powerful platform to genetically alter tissue progenitors within their physiological niche while preserving their native stem cell properties and regulatory interactions.


Asunto(s)
Diferenciación Celular , Dependovirus/genética , Genoma , Células Madre Hematopoyéticas/citología , Células Satélite del Músculo Esquelético/citología , Piel/citología , Animales , Movimiento Celular , Dependovirus/clasificación , Femenino , Técnicas de Transferencia de Gen , Terapia Genética , Células Madre Hematopoyéticas/metabolismo , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Células Satélite del Músculo Esquelético/metabolismo , Piel/metabolismo
5.
Nat Methods ; 13(10): 868-74, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27595405

RESUMEN

CRISPR-Cas9 delivery by adeno-associated virus (AAV) holds promise for gene therapy but faces critical barriers on account of its potential immunogenicity and limited payload capacity. Here, we demonstrate genome engineering in postnatal mice using AAV-split-Cas9, a multifunctional platform customizable for genome editing, transcriptional regulation, and other previously impracticable applications of AAV-CRISPR-Cas9. We identify crucial parameters that impact efficacy and clinical translation of our platform, including viral biodistribution, editing efficiencies in various organs, antigenicity, immunological reactions, and physiological outcomes. These results reveal that AAV-CRISPR-Cas9 evokes host responses with distinct cellular and molecular signatures, but unlike alternative delivery methods, does not induce extensive cellular damage in vivo. Our study provides a foundation for developing effective genome therapeutics.


Asunto(s)
Sistemas CRISPR-Cas/genética , Dependovirus/genética , Técnicas de Transferencia de Gen , Ingeniería Genética/métodos , Vectores Genéticos/genética , Animales , Ensayo de Inmunoadsorción Enzimática , Edición Génica , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
6.
Science ; 351(6271): 407-411, 2016 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-26721686

RESUMEN

Frame-disrupting mutations in the DMD gene, encoding dystrophin, compromise myofiber integrity and drive muscle deterioration in Duchenne muscular dystrophy (DMD). Removing one or more exons from the mutated transcript can produce an in-frame mRNA and a truncated, but still functional, protein. In this study, we developed and tested a direct gene-editing approach to induce exon deletion and recover dystrophin expression in the mdx mouse model of DMD. Delivery by adeno-associated virus (AAV) of clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9 endonucleases coupled with paired guide RNAs flanking the mutated Dmd exon23 resulted in excision of intervening DNA and restored the Dmd reading frame in myofibers, cardiomyocytes, and muscle stem cells after local or systemic delivery. AAV-Dmd CRISPR treatment partially recovered muscle functional deficiencies and generated a pool of endogenously corrected myogenic precursors in mdx mouse muscle.


Asunto(s)
Terapia Genética/métodos , Distrofia Muscular de Duchenne/terapia , Células Satélite del Músculo Esquelético/metabolismo , Transducción Genética/métodos , Animales , Sistemas CRISPR-Cas , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Dependovirus , Modelos Animales de Enfermedad , Exones , Mutación del Sistema de Lectura , Ratones , Ratones Endogámicos mdx , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/genética , Miocardio/metabolismo , ARN Mensajero/genética , Eliminación de Secuencia
7.
Cell ; 155(4): 909-921, 2013 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-24209627

RESUMEN

Ex vivo expansion of satellite cells and directed differentiation of pluripotent cells to mature skeletal muscle have proved difficult challenges for regenerative biology. Using a zebrafish embryo culture system with reporters of early and late skeletal muscle differentiation, we examined the influence of 2,400 chemicals on myogenesis and identified six that expanded muscle progenitors, including three GSK3ß inhibitors, two calpain inhibitors, and one adenylyl cyclase activator, forskolin. Forskolin also enhanced proliferation of mouse satellite cells in culture and maintained their ability to engraft muscle in vivo. A combination of bFGF, forskolin, and the GSK3ß inhibitor BIO induced skeletal muscle differentiation in human induced pluripotent stem cells (iPSCs) and produced engraftable myogenic progenitors that contributed to muscle repair in vivo. In summary, these studies reveal functionally conserved pathways regulating myogenesis across species and identify chemical compounds that expand mouse satellite cells and differentiate human iPSCs into engraftable muscle.


Asunto(s)
Evaluación Preclínica de Medicamentos , Desarrollo de Músculos/efectos de los fármacos , Animales , Colforsina/farmacología , Técnicas de Cultivo , AMP Cíclico/metabolismo , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Músculo Esquelético/citología , Músculo Esquelético/fisiología , Distrofias Musculares/terapia , Células Satélite del Músculo Esquelético/metabolismo , Trasplante de Células Madre , Pez Cebra/embriología , Pez Cebra/metabolismo
8.
Nat Biotechnol ; 31(10): 898-907, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24013197

RESUMEN

In a cell-free approach to regenerative therapeutics, transient application of paracrine factors in vivo could be used to alter the behavior and fate of progenitor cells to achieve sustained clinical benefits. Here we show that intramyocardial injection of synthetic modified RNA (modRNA) encoding human vascular endothelial growth factor-A (VEGF-A) results in the expansion and directed differentiation of endogenous heart progenitors in a mouse myocardial infarction model. VEGF-A modRNA markedly improved heart function and enhanced long-term survival of recipients. This improvement was in part due to mobilization of epicardial progenitor cells and redirection of their differentiation toward cardiovascular cell types. Direct in vivo comparison with DNA vectors and temporal control with VEGF inhibitors revealed the greatly increased efficacy of pulse-like delivery of VEGF-A. Our results suggest that modRNA is a versatile approach for expressing paracrine factors as cell fate switches to control progenitor cell fate and thereby enhance long-term organ repair.


Asunto(s)
Linaje de la Célula , Infarto del Miocardio/terapia , Miocardio/patología , ARN Mensajero/metabolismo , Regeneración , Células Madre/citología , Células Madre/metabolismo , Animales , Apoptosis , Biomarcadores/metabolismo , Diferenciación Celular , Proliferación Celular , Modelos Animales de Enfermedad , Células Endoteliales/patología , Técnicas de Transferencia de Gen , Humanos , Cinética , Luciferasas/metabolismo , Ratones , Modelos Biológicos , Músculo Esquelético/metabolismo , Infarto del Miocardio/fisiopatología , Miocardio/metabolismo , ARN Mensajero/genética , Trasplante de Células Madre , Análisis de Supervivencia , Resultado del Tratamiento , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
9.
Cell ; 152(3): 570-83, 2013 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-23352431

RESUMEN

Long noncoding RNAs (lncRNAs) are often expressed in a development-specific manner, yet little is known about their roles in lineage commitment. Here, we identified Braveheart (Bvht), a heart-associated lncRNA in mouse. Using multiple embryonic stem cell (ESC) differentiation strategies, we show that Bvht is required for progression of nascent mesoderm toward a cardiac fate. We find that Bvht is necessary for activation of a core cardiovascular gene network and functions upstream of mesoderm posterior 1 (MesP1), a master regulator of a common multipotent cardiovascular progenitor. We also show that Bvht interacts with SUZ12, a component of polycomb-repressive complex 2 (PRC2), during cardiomyocyte differentiation, suggesting that Bvht mediates epigenetic regulation of cardiac commitment. Finally, we demonstrate a role for Bvht in maintaining cardiac fate in neonatal cardiomyocytes. Together, our work provides evidence for a long noncoding RNA with critical roles in the establishment of the cardiovascular lineage during mammalian development.


Asunto(s)
Diferenciación Celular , Células Madre Embrionarias/metabolismo , Miocitos Cardíacos/citología , ARN Largo no Codificante , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Redes Reguladoras de Genes , Humanos , Mesodermo/citología , Mesodermo/metabolismo , Ratones , Miocitos Cardíacos/metabolismo , Complejo Represivo Polycomb 2/metabolismo , Ratas
10.
Annu Rev Pathol ; 8: 441-75, 2013 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-23121053

RESUMEN

Skeletal muscle is a highly specialized, postmitotic tissue that must withstand chronic mechanical and physiological stress throughout life to maintain proper contractile function. Muscle damage or disease leads to progressive weakness and disability, and manifests in more than 100 different human disorders. Current therapies to treat muscle degenerative diseases are limited mostly to the amelioration of symptoms, although promising new therapeutic directions are emerging. In this review, we discuss the pathological basis for the most common muscle degenerative diseases and highlight new and encouraging experimental and clinical opportunities to prevent or reverse these afflictions.


Asunto(s)
Enfermedades Musculares/patología , Enfermedades Musculares/terapia , Animales , Humanos , Músculo Esquelético/lesiones , Músculo Esquelético/patología , Distrofias Musculares/patología , Distrofias Musculares/terapia
11.
Stem Cell Rev Rep ; 6(2): 297-306, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20180049

RESUMEN

The recent generation of induced pluripotent stem cells (iPSCs) from somatic cells provides an invaluable resource for drug or toxicology screening, medical research, and patient-specific cell therapy. However, there are currently a number of obstacles including virus integration and the genetic alteration of iPSCs that will need to be overcome before these cells may be considered safe for clinical applications. Here, we highlight the potential and challenges of iPSC research and review advances in reprogramming methods that have rapidly moved the field closer to realizing the goal of generating safe iPSCs for transplantation.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Células Madre Pluripotentes Inducidas/citología , Humanos
12.
Int J Dev Biol ; 54(5): 877-86, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-19876814

RESUMEN

Although human induced pluripotent stem cells (hiPSCs) hold great promise as a source of differentiated cells for vast therapeutic implications, many obstacles still need to be surmounted before this can become a reality. One obstacle, a robust feeder- and serum-free system to generate and expand hiPSCs in culture is still unavailable. Here, for the first time, we describe a novel establishment and maintenance culture technique that uses human dermal fibroblasts to generate hiPSCs by introducing four factors, Klf4, Oct4, Sox2, and c-Myc under serum- and feeder-independent conditions. We have used a serum replacement product, conditioned medium (CM), or feeder-free medium (FFM) supplemented with high elevated basic-fibroblast growth factor in the absence or presence of Matrigel. Our FFM system in the presence of Matrigel enhanced the efficiency of alkaline phosphatase-positive colonies at a frequency at least 10-fold greater than the conventional method on feeder cells. The established hiPSCs are similar to human embryonic stem cells in many aspects including morphology, passaging, surface and pluripotency markers, normal karyotype, gene expression, ultrastructure, and in vitro differentiation. Such hiPSCs could be useful particularly in the context of in vitro disease modeling, pharmaceutical screening and in cellular replacement therapies once the safety issues have been overcome.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Diferenciación Celular/genética , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Adulto , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Medio de Cultivo Libre de Suero/farmacología , Técnica del Anticuerpo Fluorescente , Expresión Génica/efectos de los fármacos , Humanos , Células Madre Pluripotentes Inducidas/ultraestructura , Cariotipificación , Factor 4 Similar a Kruppel , Microscopía Electrónica de Rastreo , Microscopía Electrónica de Transmisión , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
13.
J Proteome Res ; 8(6): 3098-108, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19334765

RESUMEN

We isolated a moderately halophilic bacterium with high level of tolerance to two toxic oxyanions, selenite and tellurite, from hypersaline soil in Garmsar, Iran. 16s rRNA sequence analysis revealed that the isolate, strain MAM, had 98% similarity with Halomonas elongate, and is closely related to other species of the genus Halomonas. We observed that the tolerance to tellurite and its removal increased significantly when both selenite and tellurite were added to the culture media, suggesting a positive synergism of selenite on tellurite tolerance and removal. We applied a proteomic approach to study the proteome response of Halomonas sp. strain MAM to selenite, tellurite, and selenite + tellurite. Out of approximately 800 protein spots detected on 2-DE gels, 208 spots were differentially expressed in response to at least one of treatments. Of them, 70 CBB stained spots were analyzed by MALDI TOF/TOF mass spectrometry, leading to identification of 36 proteins. Our results revealed that several mechanisms including fatty acid synthesis, energy production, cell transport, oxidative stress detoxification, DNA replication, transcription and translation contributed in bacterial response and/or adaptation. These results provided new insights into the general mechanisms on the tolerance of halophilic bacteria to these two toxic oxyanions and the use of them for bioremediation of contaminated saline soils and wastes discharge sites.


Asunto(s)
Proteínas Bacterianas/metabolismo , Halomonas/efectos de los fármacos , Halomonas/fisiología , Proteoma/metabolismo , Selenito de Sodio/farmacología , Telurio/farmacología , Acetil-CoA Carboxilasa/metabolismo , Biodegradación Ambiental , Electroforesis en Gel Bidimensional , Halomonas/genética , Halomonas/ultraestructura , Filogenia , Proteómica/métodos , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Estrés Fisiológico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...