Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Stem Cell ; 31(7): 974-988.e5, 2024 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-38843830

RESUMEN

Cellular therapies with cardiomyocytes produced from induced pluripotent stem cells (iPSC-CMs) offer a potential route to cardiac regeneration as a treatment for chronic ischemic heart disease. Here, we report successful long-term engraftment and in vivo maturation of autologous iPSC-CMs in two rhesus macaques with small, subclinical chronic myocardial infarctions, all without immunosuppression. Longitudinal positron emission tomography imaging using the sodium/iodide symporter (NIS) reporter gene revealed stable grafts for over 6 and 12 months, with no teratoma formation. Histological analyses suggested capability of the transplanted iPSC-CMs to mature and integrate with endogenous myocardium, with no sign of immune cell infiltration or rejection. By contrast, allogeneic iPSC-CMs were rejected within 8 weeks of transplantation. This study provides the longest-term safety and maturation data to date in any large animal model, addresses concerns regarding neoantigen immunoreactivity of autologous iPSC therapies, and suggests that autologous iPSC-CMs would similarly engraft and mature in human hearts.


Asunto(s)
Células Madre Pluripotentes Inducidas , Macaca mulatta , Miocitos Cardíacos , Animales , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/citología , Diferenciación Celular , Humanos , Trasplante Autólogo , Tomografía de Emisión de Positrones , Factores de Tiempo , Infarto del Miocardio/terapia , Infarto del Miocardio/patología
2.
FASEB J ; 37(7): e23029, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37310585

RESUMEN

The increasing incidence of cardiovascular disease (CVD) has led to a significant ongoing need to address this surgically through coronary artery bypass grafting (CABG) and percutaneous coronary interventions (PCI). From this, there continues to be a substantial burden of mortality and morbidity due to complications arising from endothelial damage, resulting in restenosis. Whilst mast cells (MC) have been shown to have a causative role in atherosclerosis and other vascular diseases, including restenosis due to vein engraftment; here, we demonstrate their rapid response to arterial wire injury, recapitulating the endothelial damage seen in PCI procedures. Using wild-type mice, we demonstrate accumulation of MC in the femoral artery post-acute wire injury, with rapid activation and degranulation, resulting in neointimal hyperplasia, which was not observed in MC-deficient KitW-sh/W-sh mice. Furthermore, neutrophils, macrophages, and T cells were abundant in the wild-type mice area of injury but reduced in the KitW-sh/W-sh mice. Following bone-marrow-derived MC (BMMC) transplantation into KitW-sh/W-sh mice, not only was the neointimal hyperplasia induced, but the neutrophil, macrophage, and T-cell populations were also present in these transplanted mice. To demonstrate the utility of MC as a target for therapy, we administered the MC stabilizing drug, disodium cromoglycate (DSCG) immediately following arterial injury and were able to show a reduction in neointimal hyperplasia in wild-type mice. These studies suggest a critical role for MC in inducing the conditions and coordinating the detrimental inflammatory response seen post-endothelial injury in arteries undergoing revascularization procedures, and by targeting the rapid MC degranulation immediately post-surgery with DSCG, this restenosis may become a preventable clinical complication.


Asunto(s)
Aterosclerosis , Intervención Coronaria Percutánea , Lesiones del Sistema Vascular , Animales , Ratones , Hiperplasia , Mastocitos , Arterias , Constricción Patológica
3.
J Clin Invest ; 130(8): 4167-4181, 2020 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-32369445

RESUMEN

There are more than 7000 described rare diseases, most lacking specific treatment. Autosomal-dominant hyper-IgE syndrome (AD-HIES, also known as Job's syndrome) is caused by mutations in STAT3. These patients present with immunodeficiency accompanied by severe nonimmunological features, including skeletal, connective tissue, and vascular abnormalities, poor postinfection lung healing, and subsequent pulmonary failure. No specific therapies are available for these abnormalities. Here, we investigated underlying mechanisms in order to identify therapeutic targets. Histological analysis of skin wounds demonstrated delayed granulation tissue formation and vascularization during skin-wound healing in AD-HIES patients. Global gene expression analysis in AD-HIES patient skin fibroblasts identified deficiencies in a STAT3-controlled transcriptional network regulating extracellular matrix (ECM) remodeling and angiogenesis, with hypoxia-inducible factor 1α (HIF-1α) being a major contributor. Consistent with this, histological analysis of skin wounds and coronary arteries from AD-HIES patients showed decreased HIF-1α expression and revealed abnormal organization of the ECM and altered formation of the coronary vasa vasorum. Disease modeling using cell culture and mouse models of angiogenesis and wound healing confirmed these predicted deficiencies and demonstrated therapeutic benefit of HIF-1α-stabilizing drugs. The study provides mechanistic insights into AD-HIES pathophysiology and suggests potential treatment options for this rare disease.


Asunto(s)
Matriz Extracelular/metabolismo , Síndrome de Job/metabolismo , Neovascularización Fisiológica , Piel/metabolismo , Cicatrización de Heridas , Heridas y Lesiones/metabolismo , Animales , Matriz Extracelular/genética , Matriz Extracelular/patología , Femenino , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Síndrome de Job/genética , Síndrome de Job/patología , Masculino , Ratones , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Piel/irrigación sanguínea , Piel/patología , Heridas y Lesiones/genética , Heridas y Lesiones/patología
4.
Stem Cell Res ; 45: 101821, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32344328

RESUMEN

We have successfully generated induced pluripotent stem cell (iPSC) lines derived from peripheral blood mononuclear cells of five patients with Cerebral autosomal-dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL). These cells carry the genetic NOTCH3 mutation present in their parental cells. These iPSC cells exhibited normal karyotype and phenotype, which were sustained through propagation. Furthermore, these iPSCs displayed the capacity of differentiating toward the three germ layers in vitro.


Asunto(s)
CADASIL , Células Madre Pluripotentes Inducidas , CADASIL/genética , Humanos , Leucocitos Mononucleares , Imagen por Resonancia Magnética , Mutación , Fenotipo , Receptor Notch3/genética , Receptores Notch/genética
5.
Stem Cell Res ; 41: 101586, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31707214

RESUMEN

Autosomal dominant Hyper IgE syndrome (AD-HIES), a rare immune deficiency affecting fewer than one per million people, is caused by heterozygous deleterious mutations in STAT3. STAT3 signaling plays crucial roles in basic cellular functions affecting broad aspects of cellular homeostasis. Accordingly, in addition to immunological deficits, patients experience severe multisystem non-immunological features. Human induced pluripotent stem cells (hiPSC) are well established as in vivo disease models for various human pathologies. We describe the generation of iPSC from three AD-HIES patients. These iPSCs express pluripotency markers, differentiate into three germ layers, have normal karyotype and similar genome identity to parental cells.


Asunto(s)
Diferenciación Celular , Fibroblastos/patología , Células Madre Pluripotentes Inducidas/patología , Síndrome de Job/genética , Síndrome de Job/patología , Mutación , Factor de Transcripción STAT3/genética , Células Cultivadas , Fibroblastos/metabolismo , Genes Dominantes , Humanos , Células Madre Pluripotentes Inducidas/metabolismo
6.
Stem Cell Res ; 38: 101481, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31234109

RESUMEN

Chemokine receptor 5 (CCR5) is the primary coreceptor for HIV entry into macrophages. Individuals with a homozygous deletion of 32 bp in the CCR5 gene (CCR5Δ32) are highly resistant to HIV infection (Samson et al., 1996). Allogeneic stem cell transplantation from a healthy donor with the homozygous CCR5Δ32 variant to an HIV positive individual has demonstrated efficient long-term control of HIV. We identified three individuals with this homozygous CCR5Δ32 variant, and successfully generated induced pluripotent stem cell (iPSC) lines from their dermal fibroblasts. The iPSCs lines carrying homozygous CCR5Δ32 variant displayed phenotypically normal and the potential to differentiation toward the three germ layers.


Asunto(s)
Secuencia de Bases , Homocigoto , Células Madre Pluripotentes Inducidas/metabolismo , Receptores CCR5 , Eliminación de Secuencia , Adulto , Línea Celular , Femenino , Humanos , Masculino , Persona de Mediana Edad , Receptores CCR5/genética , Receptores CCR5/metabolismo
7.
Sci Rep ; 8(1): 5907, 2018 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-29651156

RESUMEN

Nonhuman primate (NHP) models are more predictive than rodent models for developing induced pluripotent stem cell (iPSC)-based cell therapy, but robust and reproducible NHP iPSC-cardiomyocyte differentiation protocols are lacking for cardiomyopathies research. We developed a method to differentiate integration-free rhesus macaque iPSCs (RhiPSCs) into cardiomyocytes with >85% purity in 10 days, using fully chemically defined conditions. To enable visualization of intracellular calcium flux in beating cardiomyocytes, we used CRISPR/Cas9 to stably knock-in genetically encoded calcium indicators at the rhesus AAVS1 safe harbor locus. Rhesus cardiomyocytes derived by our stepwise differentiation method express signature cardiac markers and show normal electrochemical coupling. They are responsive to cardiorelevant drugs and can be successfully engrafted in a mouse myocardial infarction model. Our approach provides a powerful tool for generation of NHP iPSC-derived cardiomyocytes amenable to utilization in basic research and preclinical studies, including in vivo tissue regeneration models and drug screening.


Asunto(s)
Calcio/metabolismo , Efecto Fundador , Células Madre Pluripotentes Inducidas/metabolismo , Infarto del Miocardio/terapia , Miocitos Cardíacos/metabolismo , Animales , Biomarcadores/metabolismo , Sistemas CRISPR-Cas , Calcio/análisis , Fármacos Cardiovasculares/farmacología , Diferenciación Celular , Línea Celular , Dependovirus/genética , Dependovirus/metabolismo , Modelos Animales de Enfermedad , Fluorescencia , Expresión Génica , Técnicas de Sustitución del Gen , Genes Reporteros , Sitios Genéticos , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Macaca mulatta , Ratones , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/trasplante , Proteína Homeótica Nanog/genética , Proteína Homeótica Nanog/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Antígenos Embrionarios Específico de Estadio/genética , Antígenos Embrionarios Específico de Estadio/metabolismo , Trasplante Heterólogo
8.
Sci Signal ; 9(458): ra121, 2016 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-27965423

RESUMEN

ACDC (arterial calcification due to deficiency of CD73) is an autosomal recessive disease resulting from loss-of-function mutations in NT5E, which encodes CD73, a 5'-ectonucleotidase that converts extracellular adenosine monophosphate to adenosine. ACDC patients display progressive calcification of lower extremity arteries, causing limb ischemia. Tissue-nonspecific alkaline phosphatase (TNAP), which converts pyrophosphate (PPi) to inorganic phosphate (Pi), and extracellular purine metabolism play important roles in other inherited forms of vascular calcification. Compared to cells from healthy subjects, induced pluripotent stem cell-derived mesenchymal stromal cells (iMSCs) from ACDC patients displayed accelerated calcification and increased TNAP activity when cultured under conditions that promote osteogenesis. TNAP activity generated adenosine in iMSCs derived from ACDC patients but not in iMSCs from control subjects, which have CD73. In response to osteogenic stimulation, ACDC patient-derived iMSCs had decreased amounts of the TNAP substrate PPi, an inhibitor of extracellular matrix calcification, and exhibited increased activation of AKT, mechanistic target of rapamycin (mTOR), and the 70-kDa ribosomal protein S6 kinase (p70S6K), a pathway that promotes calcification. In vivo, teratomas derived from ACDC patient cells showed extensive calcification and increased TNAP activity. Treating mice bearing these teratomas with an A2b adenosine receptor agonist, the mTOR inhibitor rapamycin, or the bisphosphonate etidronate reduced calcification. These results show that an increase of TNAP activity in ACDC contributes to ectopic calcification by disrupting the extracellular balance of PPi and Pi and identify potential therapeutic targets for ACDC.


Asunto(s)
Fosfatasa Alcalina/metabolismo , Enfermedades Genéticas Congénitas/enzimología , Células Madre Pluripotentes Inducidas/enzimología , Células Madre Mesenquimatosas/enzimología , Transducción de Señal , Calcificación Vascular/enzimología , 5'-Nucleotidasa/deficiencia , Adenosina/genética , Adenosina/metabolismo , Fosfatasa Alcalina/genética , Animales , Proteínas Ligadas a GPI/deficiencia , Enfermedades Genéticas Congénitas/genética , Enfermedades Genéticas Congénitas/patología , Humanos , Células Madre Pluripotentes Inducidas/patología , Células Madre Mesenquimatosas/patología , Ratones , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/genética , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Calcificación Vascular/genética , Calcificación Vascular/patología
9.
Sci Transl Med ; 6(227): 227ra34, 2014 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-24622514

RESUMEN

Veins grafted into an arterial environment undergo a complex vascular remodeling process. Pathologic vascular remodeling often results in stenosed or occluded conduit grafts. Understanding this complex process is important for improving the outcome of patients with coronary and peripheral artery disease undergoing surgical revascularization. Using in vivo murine cell lineage-tracing models, we show that endothelial-derived cells contribute to neointimal formation through endothelial-to-mesenchymal transition (EndMT), which is dependent on early activation of the Smad2/3-Slug signaling pathway. Antagonism of transforming growth factor-ß (TGF-ß) signaling by TGF-ß neutralizing antibody, short hairpin RNA-mediated Smad3 or Smad2 knockdown, Smad3 haploinsufficiency, or endothelial cell-specific Smad2 deletion resulted in decreased EndMT and less neointimal formation compared to controls. Histological examination of postmortem human vein graft tissue corroborated the changes observed in our mouse vein graft model, suggesting that EndMT is operative during human vein graft remodeling. These data establish that EndMT is an important mechanism underlying neointimal formation in interpositional vein grafts, and identifies the TGF-ß-Smad2/3-Slug signaling pathway as a potential therapeutic target to prevent clinical vein graft stenosis.


Asunto(s)
Transdiferenciación Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Mesodermo/efectos de los fármacos , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Venas/crecimiento & desarrollo , Venas/trasplante , Animales , Anticuerpos Neutralizantes/farmacología , Linaje de la Célula/efectos de los fármacos , Células Endoteliales/citología , Células Endoteliales/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Mesodermo/citología , Mesodermo/metabolismo , Ratones , Neointima/metabolismo , Transducción de Señal/efectos de los fármacos , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Factores de Transcripción de la Familia Snail , Factores de Transcripción/metabolismo , Venas/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA