Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 181
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Methods ; 19(10): 1306-1319, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36064772

RESUMEN

Hematopoietic humanized (hu) mice are powerful tools for modeling the action of human immune system and are widely used for preclinical studies and drug discovery. However, generating a functional human T cell compartment in hu mice remains challenging, primarily due to the species-related differences between human and mouse thymus. While engrafting human fetal thymic tissues can support robust T cell development in hu mice, tissue scarcity and ethical concerns limit their wide use. Here, we describe the tissue engineering of human thymus organoids from inducible pluripotent stem cells (iPSC-thymus) that can support the de novo generation of a diverse population of functional human T cells. T cells of iPSC-thymus-engrafted hu mice could mediate both cellular and humoral immune responses, including mounting robust proinflammatory responses on T cell receptor engagement, inhibiting allogeneic tumor graft growth and facilitating efficient Ig class switching. Our findings indicate that hu mice engrafted with iPSC-thymus can serve as a new animal model to study human T cell-mediated immunity and accelerate the translation of findings from animal studies into the clinic.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Células Madre Pluripotentes Inducidas , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones SCID , Organoides , Linfocitos T , Timo
2.
Proc Natl Acad Sci U S A ; 119(52): e2214989119, 2022 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-36534805

RESUMEN

Historically, successful allotransplantation was only achieved by utilizing powerful immunosuppressive drugs that were exposing the patient to severe opportunistic infections. The thymus of the transplant recipient renders such therapy obligatory as it constitutively blocks self-reactive T cells while allowing alloreactive T cells to mature and populate the periphery. In 1992, a follow-up study revealed the presence of donor leukocytes in long-term transplant survivors. The stable persistence of recipient and donor leukocytes in the transplanted patient, referred to as "chimerism", was considered the reason why in some cases it was even possible to stop immunosuppressive treatment without damaging the transplanted organ. Unfortunately, it quickly became evident that stable, persistent allogeneic chimerism was not easily achievable by design. Recently, a novel approach has been identified to help address this clinical gap in knowledge: Cotransplantation of a donor graft with a thymic organoid populated with donor precursor cells generates stable, long-term chimerism in the recipient. In humanized mice, the implantation of thymic organoids, populated with human donor inducible pluripotent stem cell (iPSC)-derived thymic epithelial cells (TECs) and the same donor CD34+ bone marrow precursors, induces tolerance to human leukocyte antigen (HLA)-matched donor tissues/organs. This technology will allow successful allotransplantation of cells/organs even between Major Histocompatibility Complex (MHC)-noncompatible individuals and allow getting rid of immunosuppressive treatments reducing recipient morbidity.


Asunto(s)
Trasplante de Médula Ósea , Tolerancia Central , Humanos , Ratones , Animales , Estudios de Seguimiento , Tolerancia Inmunológica , Linfocitos T , Inmunosupresores , Antígenos de Histocompatibilidad
3.
Cardiovasc Diabetol ; 21(1): 130, 2022 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-35831885

RESUMEN

BACKGROUND: Liver pathology (LP) characteristic of non-alcoholic fatty acid disease (NAFLD)/non-alcoholic steatohepatitis (NASH) is a prevalent co-morbidity of type 2 diabetes (T2D). Accumulating evidence indicates that neutrophils driving insulin resistance (IR), including hepatic IR, precipitate T2D-associated NAFLD/NASH. We hypothesized that targeting neutrophil accumulation into insulin-sensitive tissues in mice using a CXCR2 antagonist under T2D-precipitating high fat diet (HFD) could improve insulin sensitivity and prevent the progression towards liver pathology reminiscent of NAFLD/NASH. METHODS: Mice were age-matched and on standard rodent chow prior to 1:1 randomization into control and HFD formulated with the CXCR2 antagonist AZD5069 or with biologically inactive substitute. They were monitored for metabolic changes including insulin sensitivity using the hyperinsulinemic-euglycemic clamp and hepatic histopathologic evaluation in H&E-stained sections as well as via immunofluorescence microscopy of liver sections for leukocyte markers, collagen 1A1 formation, α-smooth muscle actin (SMA), and galectin-3 expression, for 16 weeks. Statistical tests used to determine significant differences among study groups and outcomes include Student's t-test, one-way ANOVA, repeated measures two-way ANOVA, and Fisher's exact test, depending on the analytical question. RESULTS: Compared to mice on HFD, mice in the AZD5069-formulated HFD exhibited improved insulin sensitivity, a modest reduction in weight gain, and a significant improvement in LP and markers related to NAFLD/NASH. Mice in the AZD5069-formulated HFD also exhibited reduced neutrophil accumulation into the liver at the end of the 16 week study period. CONCLUSIONS: These results show, for the first time, the effectiveness of a selective CXCR2 antagonist to improve insulin sensitivity, concomitantly preventing the progression towards LP characteristic of NAFLD/NASH. This represents a novel approach to target IR and developing LP under T2D-susceptible conditions using a single agent. Furthermore, our data extend the growing evidence in support of neutrophils as a leukocyte population that imprints and maintains a chronic inflammatory state in the progression of dysregulated metabolism in liver-specific co-morbid conditions.


Asunto(s)
Diabetes Mellitus Tipo 2 , Resistencia a la Insulina , Enfermedad del Hígado Graso no Alcohólico , Animales , Diabetes Mellitus Tipo 2/metabolismo , Dieta Alta en Grasa , Humanos , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/prevención & control
4.
Xenotransplantation ; 24(1)2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-28058735

RESUMEN

Anti-CD154mAb is a powerful co-stimulation blockade agent that is efficacious in preventing rejection, even in xenogeneic settings. It has been used in the majority of successful long-term pig-to-non-human primate islet transplantation models. However, its clinical use was halted as a result of thromboembolic complications that were also observed in preclinical and clinical organ transplantation models. An anti-CD154mAb was administered to 14 streptozotocin-induced diabetic cynomolgus monkey recipients of porcine islets, some of which received the agent for many months. Monkeys were monitored for complications, and blood monitoring was carried out frequently. After euthanasia, multiple biopsies of all organs were examined for histological features of thromboembolism. Anti-CD154mAb prevented rejection of genetically engineered pig islets in all monkeys. No significant complications were attributable specifically to anti-CD154mAb. There was no evidence of thromboembolism in multiple histological sections from all major organs, including the brain. Our data suggest that in diabetic monkeys with pig islet grafts, anti-CD154mAb would appear to be an effective and safe therapy, and is not associated with thromboembolic complications.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Ligando de CD40/inmunología , Xenoinjertos/efectos de los fármacos , Trasplante de Islotes Pancreáticos/inmunología , Trasplante Heterólogo , Animales , Diabetes Mellitus Experimental/inmunología , Rechazo de Injerto/inmunología , Supervivencia de Injerto/inmunología , Xenoinjertos/inmunología , Inmunosupresores/farmacología , Macaca fascicularis , Porcinos , Trasplante Heterólogo/métodos
5.
J Autoimmun ; 72: 33-46, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27173406

RESUMEN

Type 1 diabetes (T1D) is an autoimmune disease characterized by pancreatic ß cell destruction induced by islet reactive T cells that have escaped central tolerance. Many physiological and environmental triggers associated with T1D result in ß cell endoplasmic reticulum (ER) stress and dysfunction, increasing the potential for abnormal post-translational modification (PTM) of proteins. We hypothesized that ß cell ER stress induced by environmental and physiological conditions generates abnormally-modified proteins for the T1D autoimmune response. To test this hypothesis we exposed the murine CD4(+) diabetogenic BDC2.5 T cell clone to murine islets in which ER stress had been induced chemically (Thapsigargin). The BDC2.5 T cell IFNγ response to these cells was significantly increased compared to non-treated islets. This ß cell ER stress increased activity of the calcium (Ca(2+))-dependent PTM enzyme tissue transglutaminase 2 (Tgase2), which was necessary for full stress-dependent immunogenicity. Indeed, BDC2.5 T cells responded more strongly to their antigen after its modification by Tgase2. Finally, exposure of non-antigenic murine insulinomas to chemical ER stress in vitro or physiological ER stress in vivo caused increased ER stress and Tgase2 activity, culminating in higher BDC2.5 responses. Thus, ß cell ER stress induced by chemical and physiological triggers leads to ß cell immunogenicity through Ca(2+)-dependent PTM. These findings elucidate a mechanism of how ß cell proteins are modified and become immunogenic, and reveal a novel opportunity for preventing ß cell recognition by autoreactive T cells.


Asunto(s)
Autoinmunidad/inmunología , Linfocitos T CD4-Positivos/inmunología , Diabetes Mellitus Tipo 1/inmunología , Estrés del Retículo Endoplásmico/inmunología , Células Secretoras de Insulina/inmunología , Secuencia de Aminoácidos , Animales , Autoantígenos/genética , Autoantígenos/inmunología , Autoinmunidad/genética , Western Blotting , Linfocitos T CD4-Positivos/metabolismo , Calcio/inmunología , Calcio/metabolismo , Línea Celular , Células Cultivadas , Cromogranina A/genética , Cromogranina A/inmunología , Cromogranina A/metabolismo , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/metabolismo , Estrés del Retículo Endoplásmico/genética , Proteínas de Unión al GTP/genética , Proteínas de Unión al GTP/inmunología , Proteínas de Unión al GTP/metabolismo , Humanos , Células Secretoras de Insulina/metabolismo , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Modelos Inmunológicos , Proteína Glutamina Gamma Glutamiltransferasa 2 , Procesamiento Proteico-Postraduccional/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Espectrometría de Masas en Tándem , Transglutaminasas/genética , Transglutaminasas/inmunología , Transglutaminasas/metabolismo
6.
Mol Ther ; 23(7): 1262-1277, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25903472

RESUMEN

One of the major obstacles in organ transplantation is to establish immune tolerance of allografts. Although immunosuppressive drugs can prevent graft rejection to a certain degree, their efficacies are limited, transient, and associated with severe side effects. Induction of thymic central tolerance to allografts remains challenging, largely because of the difficulty of maintaining donor thymic epithelial cells in vitro to allow successful bioengineering. Here, the authors show that three-dimensional scaffolds generated from decellularized mouse thymus can support thymic epithelial cell survival in culture and maintain their unique molecular properties. When transplanted into athymic nude mice, the bioengineered thymus organoids effectively promoted homing of lymphocyte progenitors and supported thymopoiesis. Nude mice transplanted with thymus organoids promptly rejected skin allografts and were able to mount antigen-specific humoral responses against ovalbumin on immunization. Notably, tolerance to skin allografts was achieved by transplanting thymus organoids constructed with either thymic epithelial cells coexpressing both syngeneic and allogenic major histocompatibility complexes, or mixtures of donor and recipient thymic epithelial cells. Our results demonstrate the technical feasibility of restoring thymic function with bioengineered thymus organoids and highlight the clinical implications of this thymus reconstruction technique in organ transplantation and regenerative medicine.


Asunto(s)
Células Epiteliales/inmunología , Tolerancia Inmunológica/inmunología , Timo/crecimiento & desarrollo , Trasplante Homólogo , Aloinjertos/inmunología , Animales , Bioingeniería , Células Epiteliales/citología , Ratones , Organoides/inmunología , Medicina Regenerativa , Timo/citología , Timo/inmunología
7.
Clin Immunol ; 160(1): 36-45, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25817545

RESUMEN

The pace at which nanotherapeutic technology for human disease is evolving has accelerated exponentially over the past five years. Most of the technology is centered on drug delivery which, in some instances, offers tunable control of drug release. Emerging technologies have resulted in improvements in tissue and cell targeting while others are at the initial stages of pairing drug release and drug release kinetics with microenvironmental stimuli or changes in homeostasis. Nanotherapeutics has only recently been adopted for consideration as a prophylaxis/treatment approach in autoimmunity. Herein, we summarize the current state-of-the art of nanotherapeutics specifically for type 1 diabetes mellitus and offer our view over the horizon of where we envisage this modality evolving towards.


Asunto(s)
Diabetes Mellitus Tipo 1/terapia , Sistemas de Liberación de Medicamentos/métodos , Inmunoterapia/métodos , Nanomedicina/métodos , Nanopartículas/uso terapéutico , Células Presentadoras de Antígenos/inmunología , Diabetes Mellitus Tipo 1/inmunología , Humanos , Linfocitos T/inmunología
8.
Clin Immunol ; 160(1): 103-23, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25773782

RESUMEN

We have developed novel antisense oligonucleotide-formulated microspheres that can reverse hyperglycemia in newly-onset diabetic mice. Dendritic cells taking up the microspheres adopt a restrained co-stimulation ability and migrate to the pancreatic lymph nodes when injected into an abdominal region that is drained by those lymph nodes. Furthermore, we demonstrate that the absolute numbers of antigen-specific Foxp3+ T regulatory cells are increased only in the lymph nodes draining the site of administration and that these T-cells proliferate independently of antigen supply in the microspheres. Taken together, our data add to the emerging model where antigen supply may not be a requirement in "vaccines" for autoimmune disease, but the site of administration - subserved by lymph nodes draining the target organ - is in fact critical to foster the generation of antigen-specific regulatory cells. The implications of these observations on "vaccine" design for autoimmunity are discussed and summarized.


Asunto(s)
Diabetes Mellitus Experimental/terapia , Diabetes Mellitus Tipo 1/terapia , Hiperglucemia/terapia , Microesferas , Linfocitos T Reguladores/citología , Animales , Antígeno B7-1/genética , Antígeno B7-2/genética , Glucemia/efectos de los fármacos , Antígeno CD11c/metabolismo , Antígenos CD40/genética , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Células Dendríticas/inmunología , Femenino , Factores de Transcripción Forkhead/análisis , Técnicas de Silenciamiento del Gen , Antígenos Comunes de Leucocito/metabolismo , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Activación de Linfocitos/inmunología , Macaca fascicularis , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Endogámicos NOD , Ratones Transgénicos , Oligonucleótidos Antisentido/genética , Páncreas/inmunología , Receptores de Antígenos de Linfocitos T/genética , Vacunas/administración & dosificación , Vacunas/inmunología
9.
Clin Immunol ; 160(1): 82-9, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25805654

RESUMEN

Herein, we highlight the technical feasibility of generating a functional mini thymus with a novel hydrogel system, based on a peptide-based self-assembly platform that can induce the formation of 3-D thymic epithelial cell (TEC) clusters. Amphiphilic peptide EAK16-II co-assembled with its histidinylated analogue EAKIIH6 into beta-sheet fibrils. When adaptor complexes (recombinant protein A/G molecules loaded with both anti-His and anti-EpCAM IgGs) were added to the mix, TECs were tethered to the hydrogel and formed 3-D mini clusters. TECs bound to the hydrogel composites retained their molecular properties; and when transplanted into athymic nude mice, they supported the development of functional T-cells. These mini thymic units of TECs can be useful in clinical applications to reconstitute T-cell adaptive immunity.


Asunto(s)
Bioingeniería/métodos , Hidrogeles/química , Oligopéptidos/química , Péptidos/química , Timo/citología , Andamios del Tejido , Animales , Proliferación Celular , Células Cultivadas , Células Epiteliales/citología , Linfopoyesis/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Linfocitos T/citología
10.
Xenotransplantation ; 22(1): 7-19, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25130196

RESUMEN

BACKGROUND: The need for pig islet xenotransplantation in patients with type 1 diabetes is compelling; however, the ideal age at which islets should be isolated from the donor pig remains uncertain. Pig islet transplantation in primates, as a valuable pre-clinical model, has been explored using adult, neonatal, fetal pig islets, and also pancreatic primordia from pig embryos as beta cell donors. Neonatal pig islets have some advantages over adult and fetal islets, but the optimal age within the first month of life at which neonatal islets should be isolated and transplanted is as yet unclear. METHODS: In an attempt to answer this question, we carried out a literature search, but limited the search primarily to evidence in the clinically-relevant pig-to-non-human primate model. RESULTS: We identified surprisingly few studies in this model directed to this topic. Even in pig-to-rodent models, there were few definitive data. CONCLUSION: From the few data available to us, we conclude that pancreatectomy and islet isolation from neonatal pigs may have advantages over adult pigs and that isolation during the first week of life may have minor advantages over later weeks.


Asunto(s)
Trasplante de Islotes Pancreáticos/métodos , Sus scrofa , Donantes de Tejidos , Trasplante Heterólogo/métodos , Factores de Edad , Animales , Animales Recién Nacidos , Células Cultivadas/trasplante , Diabetes Mellitus Experimental/cirugía , Diabetes Mellitus Tipo 1/cirugía , Trasplante de Tejido Fetal , Xenoinjertos , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Epiplón , Vena Porta , Primates , Roedores , Sus scrofa/embriología , Sus scrofa/crecimiento & desarrollo , Porcinos , Trasplante Heterotópico/métodos
11.
Xenotransplantation ; 22(4): 317-24, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26179209

RESUMEN

BACKGROUND: Pig islet grafts have been successful in treating diabetes in animal models. One remaining question is whether neonatal pig isletlike cell clusters (NICC) are resistant to the early loss of islets from the instant blood-mediated inflammatory reaction (IBMIR). METHODS: Neonatal isletlike cell clusters were harvested from three groups of piglets-(i) wild-type (genetically unmodified), (ii) α1,3-galactosyltransferase gene-knockout (GTKO)/CD46, and (iii) GTKO/CD46/CD39. NICC samples were mixed with human blood in vitro, and the following measurements were made-antibody binding; complement activation; speed of islet-induced coagulation; C-peptide; glutamic acid decarboxylase (GAD65) release; viability. RESULTS: Time to coagulation and viability were both reduced in all groups compared to freshly drawn non-anticoagulated human blood and autologous combinations, respectively. Antibody binding to the NICC occurred in all groups. CONCLUSIONS: Neonatal isletlike cell clusters were subject to humoral injury with no difference associated to their genetic characteristics.


Asunto(s)
Sangre/inmunología , Trasplante de Islotes Pancreáticos/inmunología , Trasplante Heterólogo/métodos , Animales , Animales Modificados Genéticamente , Animales Recién Nacidos , Antígenos CD/genética , Antígenos CD/inmunología , Apirasa/genética , Apirasa/inmunología , Coagulación Sanguínea , Activación de Complemento , Diabetes Mellitus/terapia , Galactosiltransferasas/deficiencia , Galactosiltransferasas/genética , Galactosiltransferasas/inmunología , Técnicas de Inactivación de Genes , Humanos , Técnicas In Vitro , Trasplante de Islotes Pancreáticos/efectos adversos , Trasplante de Islotes Pancreáticos/patología , Proteína Cofactora de Membrana/genética , Proteína Cofactora de Membrana/inmunología , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Sus scrofa , Trasplante Heterólogo/efectos adversos
12.
Xenotransplantation ; 22(1): 70-9, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25382150

RESUMEN

BACKGROUND: Xenotransplantation of porcine islets can reverse diabetes in non-human primates. The remaining hurdles for clinical application include safe and effective T-cell-directed immunosuppression, but protection against the innate immune system and coagulation dysfunction may be more difficult to achieve. Islet-targeted genetic manipulation of islet-source pigs represents a powerful tool to protect against graft loss. However, whether these genetic alterations would impair islet function is unknown. METHODS: On a background of α1,3-galactosyltransferase gene-knockout (GTKO)/human (h)CD46, additional genes (hCD39, human tissue factor pathway inhibitor, porcine CTLA4-Ig) were inserted in different combinations under an insulin promoter to promote expression in islets (confirmed by immunofluorescence). Seven pigs were tested for baseline and glucose/arginine-challenged levels of glucose, insulin, C-peptide, and glucagon. RESULTS: This preliminary study did not show definite evidence of ß-cell deficiencies, even when three transgenes were expressed under the insulin promoter. Of seven animals, all were normoglycemic at fasting, and five of seven had normal glucose disposal rates after challenge. All animals exhibited insulin, C-peptide, and glucagon responses to both glucose and arginine challenge; however, significant interindividual variation was observed. CONCLUSIONS: Multiple islet-targeted transgenic expression was not associated with an overtly detrimental effect on islet function, suggesting that complex genetic constructs designed for islet protection warrants further testing in islet xenotransplantation models.


Asunto(s)
Abatacept/genética , Antígenos CD/genética , Apirasa/genética , Glucosa/metabolismo , Insulina/genética , Islotes Pancreáticos/metabolismo , Lipoproteínas/genética , Regiones Promotoras Genéticas , Sus scrofa/metabolismo , Abatacept/biosíntesis , Animales , Animales Modificados Genéticamente , Antígenos CD/biosíntesis , Apirasa/biosíntesis , Arginina/farmacología , Glucemia/análisis , Péptido C/metabolismo , Línea Celular , Ayuno/sangre , Fibroblastos , Galactosiltransferasas/deficiencia , Galactosiltransferasas/genética , Regulación de la Expresión Génica , Técnicas de Inactivación de Genes , Genes Sintéticos , Glucagón/metabolismo , Glucosa/farmacología , Prueba de Tolerancia a la Glucosa , Humanos , Lipoproteínas/biosíntesis , Proteína Cofactora de Membrana/genética , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética , Porcinos , Transgenes
13.
Pediatr Diabetes ; 16(3): 151-63, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25652322

RESUMEN

Stem cell technology has recently gained a substantial amount of interest as one method to create a potentially limitless supply of transplantable insulin-producing cells to treat, and possibly cure diabetes mellitus. In this review, we summarize the state-of-the art of stem cell technology and list the potential sources of stem cells that have been shown to be useful as insulin-expressing surrogates. We also discuss the milestones that have been reached and those that remain to be addressed to generate bona fide beta cell-similar, insulin-producing surrogates. The caveats, limitations, and realistic expectations are also considered for current and future technology. In spite of the tremendous technical advances realized in the past decade, especially in the field of reprogramming adult somatic cells to become stem cells, the state-of-the art still relies on lengthy and cumbersome in vitro culture methods that yield cell populations that are not particularly glucose-responsive when transplanted into diabetic hosts. Despite the current impediments toward clinical translation, including the potential for immune rejection, the availability of technology to generate patient-specific reprogrammable stem cells has, and will be critical for, important insights into the genetics, epigenetics, biology, and physiology of insulin-producing cells in normal and pathologic states. This knowledge could accelerate the time to reach the desired breakthrough for safe and efficacious beta cell surrogates.


Asunto(s)
Diabetes Mellitus/terapia , Células Secretoras de Insulina/fisiología , Trasplante de Células Madre/tendencias , Diferenciación Celular , Humanos , Hígado/citología , Páncreas/citología
14.
Pediatr Diabetes ; 16(6): 393-401, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26084669

RESUMEN

Beta-cell replacement is the only physiologically relevant alternative to insulin injections in patients with type 1 diabetes (T1D). Pancreas and islet transplantation from deceased organ donors can provide a new beta-cell pool to produce insulin, help blood glucose management, and delay secondary diabetes complications. For children and adolescents with T1D, whole pancreas transplantation is not a viable option because of surgical complications, whereas islet transplantation, even if it is procedurally simpler, must still overcome the burden of immunosuppression to become a routine therapy for children in the future.


Asunto(s)
Diabetes Mellitus Tipo 1/cirugía , Terapia de Inmunosupresión/efectos adversos , Trasplante de Islotes Pancreáticos/efectos adversos , Factores de Edad , Animales , Complicaciones de la Diabetes/prevención & control , Diabetes Mellitus Tipo 1/complicaciones , Diabetes Mellitus Tipo 1/economía , Diabetes Mellitus Tipo 1/inmunología , Costos de la Atención en Salud , Humanos , Hiperglucemia/prevención & control , Hipoglucemia/prevención & control , Terapia de Inmunosupresión/economía , Trasplante de Islotes Pancreáticos/economía , Trasplante de Islotes Pancreáticos/inmunología , Trasplante de Islotes Pancreáticos/normas , Trasplante de Páncreas/efectos adversos , Guías de Práctica Clínica como Asunto , Donantes de Tejidos/provisión & distribución , Trasplante Autólogo/efectos adversos , Trasplante Autólogo/economía , Trasplante Autólogo/normas , Trasplante Heterólogo/efectos adversos , Trasplante Heterólogo/economía , Trasplante Heterólogo/normas , Trasplante Homólogo/efectos adversos , Trasplante Homólogo/economía , Trasplante Homólogo/normas , Estados Unidos , United States Food and Drug Administration
15.
J Autoimmun ; 53: 10-25, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25088457

RESUMEN

For reasons not fully understood, patients with an organ-specific autoimmune disease have increased risks of developing autoimmune responses against other organs/tissues. We identified ICA69, a known ß-cell autoantigen in Type 1 diabetes, as a potential common target in multi-organ autoimmunity. NOD mice immunized with ICA69 polypeptides exhibited exacerbated inflammation not only in the islets, but also in the salivary glands. To further investigate ICA69 autoimmunity, two genetically modified mouse lines were generated to modulate thymic ICA69 expression: the heterozygous ICA69(del/wt) line and the thymic medullary epithelial cell-specific deletion Aire-ΔICA69 line. Suboptimal central negative selection of ICA69-reactive T-cells was observed in both lines. Aire-ΔICA69 mice spontaneously developed coincident autoimmune responses to the pancreas, the salivary glands, the thyroid, and the stomach. Our findings establish a direct link between compromised thymic ICA69 expression and autoimmunity against multiple ICA69-expressing organs, and identify a potential novel mechanism for the development of multi-organ autoimmune diseases.


Asunto(s)
Autoantígenos/inmunología , Enfermedades Autoinmunes/inmunología , Tolerancia Inmunológica , Animales , Autoantígenos/genética , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/patología , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/inmunología , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Islotes Pancreáticos/inmunología , Islotes Pancreáticos/patología , Ratones , Ratones Endogámicos NOD , Ratones Transgénicos , Glándulas Salivales/inmunología , Glándulas Salivales/patología , Estómago/inmunología , Estómago/patología , Timo/inmunología , Timo/patología , Glándula Tiroides/inmunología , Glándula Tiroides/patología
16.
J Biol Chem ; 287(21): 17882-17893, 2012 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-22447927

RESUMEN

ICA69 (islet cell autoantigen 69 kDa) is a protein implicated in type 1 diabetes mellitus in both the non-obese diabetic (NOD) mouse model and humans. ICA69 is encoded by the Ica1 gene on mouse chromosome 6 A1-A2. We previously reported reduced ICA69 expression in the thymus of NOD mice compared with thymus of several non-diabetic mouse strains. We propose that reduced thymic ICA69 expression could result from variations in transcriptional regulation of the gene and that polymorphisms within the Ica1 core promoter may partially determine this transcriptional variability. We characterized the functional promoter of Ica1 in NOD mice and compared it with the corresponding portions of Ica1 in non-diabetic C57BL/6 mice. Luciferase reporter constructs demonstrated that the NOD Ica1 promoter region exhibited markedly reduced luciferase expression in transiently transfected medullary thymus epithelial (mTEC(+)) and B-cell (M12)-derived cell lines. However, in a non-diabetic strain, C57BL/6, the Ica1 promoter region was transcriptionally active when transiently transfected into the same cell lines. We concomitantly identified five single nucleotide polymorphisms within the NOD Ica1 promoter. One of these single nucleotide polymorphisms increases the binding affinity for the transcription factor AIRE (autoimmune regulator), which is highly expressed in thymic epithelial cells, where it is known to play a key role regulating self-antigen expression. We conclude that polymorphisms within the NOD Ica1 core promoter may determine AIRE-mediated down-regulation of ICA69 expression in medullary thymic epithelial cells, thus providing a novel mechanistic explanation for the loss of immunologic tolerance to this self-antigen in autoimmunity.


Asunto(s)
Autoantígenos/biosíntesis , Diabetes Mellitus Tipo 1/metabolismo , Regulación hacia Abajo , Polimorfismo de Nucleótido Simple , Factores de Transcripción/metabolismo , Transcripción Genética , Animales , Autoantígenos/genética , Línea Celular , Diabetes Mellitus Tipo 1/genética , Humanos , Ratones , Ratones Endogámicos NOD , Factores de Transcripción/genética , Proteína AIRE
17.
EMBO J ; 28(18): 2812-24, 2009 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-19680229

RESUMEN

Insulin expression in the thymus has been implicated in regulating the negative selection of autoreactive T cells and in mediating the central immune tolerance towards pancreatic beta-cells. To further explore the function of this ectopic insulin expression, we knocked out the mouse Ins2 gene specifically in the Aire-expressing medullary thymic epithelial cells (mTECs), without affecting its expression in the beta-cells. When further crossed to the Ins1 knockout background, both male and female pups (designated as ID-TEC mice for insulin-deleted mTEC) developed diabetes spontaneously around 3 weeks after birth. beta-cell-specific autoimmune destruction was observed, as well as islet-specific T cell infiltration. The presence of insulin-specific effector T cells was shown using ELISPOT assays and adoptive T cell transfer experiments. Results from thymus transplantation experiments proved further that depletion of Ins2 expression in mTECs was sufficient to break central tolerance and induce anti-insulin autoimmunity. Our observations may explain the rare cases of type 1 diabetes onset in very young children carrying diabetes-resistant HLA class II alleles. ID-TEC mice could serve as a new model for studying this pathology.


Asunto(s)
Diabetes Mellitus Experimental/genética , Diabetes Mellitus Tipo 1/genética , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Timo/patología , Alelos , Animales , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Tipo 1/inmunología , Modelos Animales de Enfermedad , Femenino , Tolerancia Inmunológica/genética , Masculino , Ratones , Ratones Noqueados , Modelos Genéticos , Linfocitos T/metabolismo , Timo/citología
18.
Lancet ; 379(9816): 672-83, 2012 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-22019026

RESUMEN

The shortage of organs and cells from deceased individuals continues to restrict allotransplantation. Pigs could provide an alternative source of tissue and cells but the immunological challenges and other barriers associated with xenotransplantation need to be overcome. Transplantation of organs from genetically modified pigs into non-human primates is now not substantially limited by hyperacute, acute antibody-mediated, or cellular rejection, but other issues have become more prominent, such as development of thrombotic microangiopathy in the graft or systemic consumptive coagulopathy in the recipient. To address these problems, pigs that express one or more human thromboregulatory or anti-inflammatory genes are being developed. The results of preclinical transplantation of pig cells--eg, islets, neuronal cells, hepatocytes, or corneas--are much more encouraging than they are for organ transplantation, with survival times greater than 1 year in all cases. Risk of transfer of an infectious microorganism to the recipient is small.


Asunto(s)
Animales Modificados Genéticamente , Rechazo de Injerto/prevención & control , Inmunosupresores/administración & dosificación , Primates , Porcinos , Trasplante Heterólogo , Animales , Trasplante de Córnea , Transfusión de Eritrocitos , Rechazo de Injerto/inmunología , Hepatocitos/trasplante , Humanos , Trasplante de Islotes Pancreáticos/inmunología , Neuronas/trasplante , Porcinos/genética , Linfocitos T/inmunología , Trasplante de Tejidos , Trasplante Heterólogo/inmunología
19.
PLoS Comput Biol ; 8(9): e1002700, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23028295

RESUMEN

The mitochondrial electron transport chain transforms energy satisfying cellular demand and generates reactive oxygen species (ROS) that act as metabolic signals or destructive factors. Therefore, knowledge of the possible modes and bifurcations of electron transport that affect ROS signaling provides insight into the interrelationship of mitochondrial respiration with cellular metabolism. Here, a bifurcation analysis of a sequence of the electron transport chain models of increasing complexity was used to analyze the contribution of individual components to the modes of respiratory chain behavior. Our algorithm constructed models as large systems of ordinary differential equations describing the time evolution of the distribution of redox states of the respiratory complexes. The most complete model of the respiratory chain and linked metabolic reactions predicted that condensed mitochondria produce more ROS at low succinate concentration and less ROS at high succinate levels than swelled mitochondria. This prediction was validated by measuring ROS production under various swelling conditions. A numerical bifurcation analysis revealed qualitatively different types of multistationary behavior and sustained oscillations in the parameter space near a region that was previously found to describe the behavior of isolated mitochondria. The oscillations in transmembrane potential and ROS generation, observed in living cells were reproduced in the model that includes interaction of respiratory complexes with the reactions of TCA cycle. Whereas multistationarity is an internal characteristic of the respiratory chain, the functional link of respiration with central metabolism creates oscillations, which can be understood as a means of auto-regulation of cell metabolism.


Asunto(s)
Proteínas del Complejo de Cadena de Transporte de Electrón/química , Mitocondrias/metabolismo , Proteínas Mitocondriales/química , Modelos Químicos , Modelos Moleculares , Especies Reactivas de Oxígeno/química , Sitios de Unión , Relojes Biológicos , Simulación por Computador , Transporte de Electrón , Radicales Libres , Oscilometría/métodos , Unión Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA