Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Más filtros

Bases de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Adv Biosyst ; 3(9)2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31633004

RESUMEN

Pancreatic islet transplantation is a promising treatment for type I diabetes, which is a chronic autoimmune disease in which the host immune cells attack insulin-producing beta cells. The impact of this therapy is limited due to tissue availability and dependence on immunosuppressive drugs that prevent immune rejection of the transplanted cells. These issues can be solved by encapsulating stem cell-derived insulin-producing cells in an immunoprotective device. However, encapsulation exacerbates ischemia, and the lack of vasculature at the implantation site post-transplantation worsens graft survival. Here, an encapsulation device that supplements nutrients to the cells is developed to improve the survival of encapsulated stem cell-derived insulin-producing cells in the poorly vascularized subcutaneous space. An internal compartment in the device is fabricated to provide zero-order release of alanine and glutamine for several weeks. The amino acid reservoir sustains viability of insulin-producing cells in nutrient limiting conditions in vitro. Moreover, the reservoir also increases cell survival by 30% after transplanting the graft in the subcutaneous space.

2.
Stem Cell Reports ; 9(3): 807-819, 2017 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-28803916

RESUMEN

The advent of large-scale in vitro differentiation of human stem cell-derived insulin-producing cells (SCIPC) has brought us closer to treating diabetes using stem cell technology. However, decades of experiences from islet transplantation show that ischemia-induced islet cell death after transplant severely limits the efficacy of the therapy. It is unclear to what extent human SCIPC are susceptible to ischemia. In this study, we show that more than half of SCIPC die shortly after transplantation. Nutrient deprivation and hypoxia acted synergistically to kill SCIPC in vitro. Amino acid supplementation rescued SCIPC from nutrient deprivation, likely by providing cellular energy. Generating SCIPC under physiological oxygen tension of 5% conferred hypoxia resistance without affecting their differentiation or function. A two-pronged strategy of physiological oxygen acclimatization during differentiation and amino acid supplementation during transplantation significantly improved SCIPC survival after transplant.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Isquemia/terapia , Trasplante de Islotes Pancreáticos , Trasplante de Células Madre , Células Madre/metabolismo , Aminoácidos/farmacología , Animales , Muerte Celular/efectos de los fármacos , Hipoxia de la Célula/efectos de los fármacos , Citoprotección/efectos de los fármacos , Humanos , Células Secretoras de Insulina/efectos de los fármacos , Isquemia/patología , Ratones Endogámicos C57BL , Oxígeno/farmacología , Ácido Pirúvico/farmacología , Células Madre/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Supervivencia Tisular/efectos de los fármacos
3.
Diabetes ; 61(7): 1769-78, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22566533

RESUMEN

We evaluated the effects of hyperbaric oxygen therapy (HOT) on autoimmune diabetes development in nonobese diabetic (NOD) mice. Animals received no treatment or daily 60-min HOT 100% oxygen (HOT-100%) at 2.0 atmospheres absolute and were monitored for diabetes onset, insulitis, infiltrating cells, immune cell function, and ß-cell apoptosis and proliferation. Cyclophosphamide-induced diabetes onset was reduced from 85.3% in controls to 48% after HOT-100% (P < 0.005) and paralleled by lower insulitis. Spontaneous diabetes incidence reduced from 85% in controls to 65% in HOT-100% (P = 0.01). Prediabetic mice receiving HOT-100% showed lower insulitis scores, reduced T-cell proliferation upon stimulation in vitro (P < 0.03), increased CD62L expression in T cells (P < 0.04), reduced costimulation markers (CD40, DC80, and CD86), and reduced major histocompatibility complex class II expression in dendritic cells (DCs) (P < 0.025), compared with controls. After autoimmunity was established, HOT was less effective. HOT-100% yielded reduced apoptosis (transferase-mediated dUTP nick-end labeling-positive insulin-positive cells; P < 0.01) and increased proliferation (bromodeoxyuridine incorporation; P < 0.001) of insulin-positive cells compared with controls. HOT reduces autoimmune diabetes incidence in NOD mice via increased resting T cells and reduced activation of DCs with preservation of ß-cell mass resulting from decreased apoptosis and increased proliferation. The safety profile and noninvasiveness makes HOT an appealing adjuvant therapy for diabetes prevention and intervention trials.


Asunto(s)
Proliferación Celular , Diabetes Mellitus Tipo 1/prevención & control , Oxigenoterapia Hiperbárica , Células Secretoras de Insulina/fisiología , Animales , Apoptosis/inmunología , Antígeno B7-1/inmunología , Antígeno B7-2/biosíntesis , Antígeno B7-2/inmunología , Antígenos CD40/biosíntesis , Antígenos CD40/inmunología , Ciclofosfamida/efectos adversos , Células Dendríticas/inmunología , Diabetes Mellitus Tipo 1/inducido químicamente , Diabetes Mellitus Tipo 1/inmunología , Femenino , Genes MHC Clase II/inmunología , Inmunosupresores/efectos adversos , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/inmunología , Selectina L/biosíntesis , Selectina L/inmunología , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos NOD , Pancreatitis/inmunología , Pancreatitis/prevención & control , Linfocitos T/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA