Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Nat Med ; 28(4): 780-788, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35347281

RESUMO

Recessive dystrophic epidermolysis bullosa (RDEB) is a lifelong genodermatosis associated with blistering, wounding, and scarring caused by mutations in COL7A1, the gene encoding the anchoring fibril component, collagen VII (C7). Here, we evaluated beremagene geperpavec (B-VEC), an engineered, non-replicating COL7A1 containing herpes simplex virus type 1 (HSV-1) vector, to treat RDEB skin. B-VEC restored C7 expression in RDEB keratinocytes, fibroblasts, RDEB mice and human RDEB xenografts. Subsequently, a randomized, placebo-controlled, phase 1 and 2 clinical trial (NCT03536143) evaluated matched wounds from nine RDEB patients receiving topical B-VEC or placebo repeatedly over 12 weeks. No grade 2 or above B-VEC-related adverse events or vector shedding or tissue-bound skin immunoreactants were noted. HSV-1 and C7 antibodies sometimes presented at baseline or increased after B-VEC treatment without an apparent impact on safety or efficacy. Primary and secondary objectives of C7 expression, anchoring fibril assembly, wound surface area reduction, duration of wound closure, and time to wound closure following B-VEC treatment were met. A patient-reported pain-severity secondary outcome was not assessed given the small proportion of wounds treated. A global assessment secondary endpoint was not pursued due to redundancy with regard to other endpoints. These studies show that B-VEC is an easily administered, safely tolerated, topical molecular corrective therapy promoting wound healing in patients with RDEB.


Assuntos
Epidermólise Bolhosa Distrófica , Animais , Colágeno Tipo VII/genética , Colágeno Tipo VII/metabolismo , Epidermólise Bolhosa Distrófica/genética , Epidermólise Bolhosa Distrófica/metabolismo , Epidermólise Bolhosa Distrófica/terapia , Terapia Genética , Humanos , Queratinócitos/metabolismo , Camundongos , Pele/metabolismo
2.
Nat Commun ; 10(1): 5583, 2019 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-31811140

RESUMO

Mutations in fused in sarcoma (FUS) lead to amyotrophic lateral sclerosis (ALS) with varying ages of onset, progression and severity. This suggests that unknown genetic factors contribute to disease pathogenesis. Here we show the identification of muscleblind as a novel modifier of FUS-mediated neurodegeneration in vivo. Muscleblind regulates cytoplasmic mislocalization of mutant FUS and subsequent accumulation in stress granules, dendritic morphology and toxicity in mammalian neuronal and human iPSC-derived neurons. Interestingly, genetic modulation of endogenous muscleblind was sufficient to restore survival motor neuron (SMN) protein localization in neurons expressing pathogenic mutations in FUS, suggesting a potential mode of suppression of FUS toxicity. Upregulation of SMN suppressed FUS toxicity in Drosophila and primary cortical neurons, indicating a link between FUS and SMN. Our data provide in vivo evidence that muscleblind is a dominant modifier of FUS-mediated neurodegeneration by regulating FUS-mediated ALS pathogenesis.


Assuntos
Esclerose Lateral Amiotrófica/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas Nucleares/metabolismo , Proteína FUS de Ligação a RNA/metabolismo , Proteínas do Complexo SMN/metabolismo , Acetiltransferases/genética , Acetiltransferases/metabolismo , Esclerose Lateral Amiotrófica/genética , Animais , Citoplasma/metabolismo , Grânulos Citoplasmáticos/metabolismo , Drosophila/genética , Drosophila/metabolismo , Feminino , Células HEK293 , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Neurônios Motores/metabolismo , Mutação , Fenótipo , Proteína FUS de Ligação a RNA/genética , Proteína FUS de Ligação a RNA/toxicidade , Proteínas do Complexo SMN/genética , Fatores de Transcrição/metabolismo
3.
Cytoskeleton (Hoboken) ; 75(7): 323-335, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-30019417

RESUMO

The actin cortex that lines the plasma membrane of most eukaryotic cells resists external mechanical forces and plays critical roles in a variety of cellular processes including morphogenesis, cytokinesis, and cell migration. Despite its ubiquity and significance, we understand relatively little about the composition, dynamics, and structure of the actin cortex. Adenomatous polyposis coli (APC) proteins regulate the actin and microtubule cytoskeletons through a variety of mechanisms, and in some contexts, APC proteins are cortically enriched. Here we show that APC2 regulates cortical actin dynamics in the follicular epithelium and the nurse cells of the Drosophila ovary and in addition affects the distribution of cortical actin at the apical side of the follicular epithelium. To understand how APC2 influences these properties of the actin cortex, we investigated the mechanisms controlling the cortical localization of APC2 in S2 cultured cells. We previously showed that the N-terminal half of APC2 containing the Armadillo repeats and the C-terminal 30 amino acids (C30) are together necessary and sufficient for APC2's cortical localization. Our work presented here supports a model that cortical localization of APC2 is governed in part by self-association through the N-terminal APC Self-Association Domain (ASAD) and a highly conserved coiled-coil within the C30 domain.


Assuntos
Actinas/metabolismo , Proteínas do Domínio Armadillo/metabolismo , Proteínas de Drosophila/metabolismo , Ovário/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Sequência de Aminoácidos , Animais , Drosophila , Feminino , Ligação Proteica , Multimerização Proteica
4.
Cell Reprogram ; 12(3): 263-73, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20698768

RESUMO

Deciding to exit pluripotency and undergo differentiation is of singular importance for pluripotent cells, including embryonic stem cells (ESCs). The molecular mechanisms for these decisions to differentiate, as well as reversing those decisions during induced pluripotency (iPS), have focused largely on transcriptomic controls. Here, we explore the role of translational control for the maintenance of pluripotency and the decisions to differentiate. Global protein translation is significantly reduced in hESCs compared to their differentiated progeny. Furthermore, p70 S6K activation is restricted in hESCs compared to differentiated fibroblast-like cells. Disruption of p70 S6K-mediated translation by rapamycin or siRNA knockdown in undifferentiated hESCs does not alter cell viability or expression of the pluripotency markers Oct4 and Nanog. However, expression of constitutively active p70 S6K, but not wild-type p70 S6K, induces differentiation. Additionally, hESCs exhibit high levels of the mTORC1/p70 S6K inhibitory complex TSC1/TSC2 and preferentially express more rapamycin insensitive mTORC2 compared to differentiated cells. siRNA-mediated knockdown of both TSC2 and Rictor elevates p70 S6K activation and induces differentiation of hESCs. These results suggest that hESCs tightly regulate mTORC1/p70 S6K-mediated protein translation to maintain a pluripotent state as well as implicate a novel role for protein synthesis as a driving force behind hESC differentiation.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Embrionárias/citologia , Células-Tronco Pluripotentes/citologia , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Serina-Treonina Quinases TOR/fisiologia , Células-Tronco Embrionárias/efeitos dos fármacos , Ativação Enzimática , Humanos , Microscopia Eletrônica de Transmissão , Células-Tronco Pluripotentes/efeitos dos fármacos , Interferência de RNA , Sirolimo/farmacologia
5.
Stem Cell Res ; 4(1): 25-37, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19854689

RESUMO

While human embryonic stem cells (hESCs) are predisposed toward chromosomal aneploidities on 12, 17, 20, and X, rendering them susceptible to transformation, the specific genes expressed are not yet known. Here, by identifying the genes overexpressed in pluripotent rhesus ESCs (nhpESCs) and comparing them both to their genetically identical differentiated progeny (teratoma fibroblasts) and to genetically related differentiated parental cells (parental skin fibroblasts from whom gametes were used for ESC derivation), we find that some of those overexpressed genes in nhpESCs cluster preferentially on rhesus chromosomes 16, 19, 20, and X, homologues of human chromosomes 17, 19, 16, and X, respectively. Differentiated parental skin fibroblasts display gene expression profiles closer to nhpESC profiles than to teratoma cells, which are genetically identical to the pluripotent nhpESCs. Twenty over- and underexpressed pluripotency modulators, some implicated in neurogenesis, have been identified. The overexpression of some of these genes discovered using pedigreed nhpESCs derived from prime embryos generated by fertile primates, which is impossible to perform with the anonymously donated clinically discarded embryos from which hESCs are derived, independently confirms the importance of chromosome 17 and X regions in pluripotency and suggests specific candidates for targeting differentiation and transformation decisions.


Assuntos
Cromossomos Humanos , Células-Tronco Embrionárias/metabolismo , Expressão Gênica , Macaca mulatta/genética , Células-Tronco Pluripotentes/metabolismo , Animais , Diferenciação Celular , Linhagem Celular , Cromossomos Humanos Par 16 , Cromossomos Humanos Par 17 , Cromossomos Humanos Par 19 , Cromossomos Humanos X , Células-Tronco Embrionárias/citologia , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Masculino , Células-Tronco Pluripotentes/citologia , Teratoma/genética , Teratoma/patologia
6.
Stem Cell Res ; 5(3): 201-11, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20864427

RESUMO

Teratoma formation in xenografts is a sufficiently stringent pluripotency assay for stem cells. However, little is known about the composition and spatial relationships of tissues within teratomas that may provide clues about development and platforms for studying organ development. Additionally, teratoma formation and analysis lack standards for reporting as assays of pluripotency. Three of 27 total teratomas derived from pedigreed primate embryonic stem cells underwent quantitative three-dimensional high-resolution magnetic resonance microscopy (MRM). Teratomas were subsequently serially sectioned and tissue types identified, semiquantitated, and correlated with MRM images. All teratomas demonstrated tissue derivatives from the three germ layers and approximately 23 different tissue types were identified. Certain tissue groups attempted to form organs more frequently (e.g., trachea/bronchi, small intestine). MRM discriminated some tissues readily (e.g., bone, adipose, cartilage) while other tissue types with like MR intensities could not be distinguished. Semiquantitative histopathological analysis of teratomas demonstrates the ability to delineate multiple tissues as derived from ectoderm, mesoderm, or endoderm and to use this information for comparison to other teratomas. MRM provides rapid quantitative imaging of intact teratomas that complements histology and identifies sites of interest for additional biological studies.


Assuntos
Células-Tronco Embrionárias/patologia , Teratoma/patologia , Animais , Microscopia/instrumentação , Microscopia/métodos , Primatas
7.
Stem Cell Res ; 2(3): 178-87, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19393591

RESUMO

Here we have developed protocols using the baboon as a complementary alternative Old World Primate to rhesus and other macaques which have severe limitations in their availability. Baboons are not limited as research resources, they are evolutionarily closer to humans, and the multiple generations of pedigreed colonies which display complex human disease phenotypes all support their further optimization as an invaluable primate model. Since neither baboon-assisted reproductive technologies nor baboon embryonic stem cells (ESCs) have been reported, here we describe the first derivations and characterization of baboon ESC lines from IVF-generated blastocysts. Two ESCs lines (BabESC-4 and BabESC-15) display ESC morphology, express pluripotency markers (Oct-4, hTert, Nanog, Sox-2, Rex-1, TRA1-60, TRA1-81), and maintain stable euploid female karyotypes with parentage confirmed independently. They have been grown continuously for >430 and 290 days, respectively. Teratomas from both lines have all three germ layers. Availabilities of these BabESCs represent another important resource for stem cell biologists.


Assuntos
Linhagem Celular , Células-Tronco Embrionárias/citologia , Modelos Biológicos , Animais , Biomarcadores/metabolismo , Blastômeros/citologia , Diferenciação Celular , Células-Tronco Embrionárias/metabolismo , Células-Tronco Embrionárias/transplante , Feminino , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Imuno-Histoquímica , Cariotipagem , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Papio , Primatas , Medicina Regenerativa , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo , Telomerase/genética , Telomerase/metabolismo
8.
Curr Protoc Stem Cell Biol ; Chapter 1: Unit 1A.1, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18785157

RESUMO

Embryonic stem (ES) cells are a powerful research tool enabling the generation of mice with custom genetics, the study of the earliest stages of mammalian differentiation in vitro and, with the isolation of human ES cells, the potential of cell-based therapies for a number of diseases including Parkinson's and Type 1 diabetes. ES cells isolated from nonhuman primates (nhpES cells) offer the opportunity to ethically test the developmental potential of primate ES cells in chimeric offspring. If these cells have similar potency to mouse ES cells, this may open a new era of primate models of human disease. Nonhuman primates are the perfect model system for the preclinical testing of ES cell-derived therapies. In this unit, we describe methods for the derivation and characterization of nonhuman primate ES cells. With these protocols, the investigator will be able to isolate nhpES cells and perform the necessary tests to confirm the pluripotent phenotype.


Assuntos
Células-Tronco Embrionárias/citologia , Primatas/embriologia , Animais , Sequência de Bases , Técnicas de Cultura de Células , Separação Celular , Primers do DNA/genética , Células-Tronco Embrionárias/metabolismo , Células-Tronco Embrionárias/transplante , Expressão Gênica , Imuno-Histoquímica , Cariotipagem , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Fenótipo , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Células-Tronco Pluripotentes/transplante , Primatas/genética , Primatas/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Teratoma/etiologia , Transplante Heterólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA