Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Eur J Immunol ; 44(11): 3307-19, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25113564

RESUMEN

By restraining T-cell activation and promoting Treg-cell expansion, myeloid-derived suppressor cells (MDSCs) and tolerogenic DCs can control self-reactive and antigraft effector T cells in autoimmunity and transplantation. Their therapeutic use and characterization, however, is limited by their scarce availability in the peripheral blood of tumor-free donors. In the present study, we describe and characterize a novel population of human myeloid suppressor cells, named fibrocytic MDSC, which are differentiated from umbilical cord blood precursors by 4-day culture with FDA-approved cytokines (recombinant human-GM-CSF and recombinant human-G-CSF). This MDSC subset, characterized by the expression of MDSC-, DC-, and fibrocyte-associated markers, promotes Treg-cell expansion and induces normoglycemia in a xenogeneic mouse model of Type 1 diabetes. In order to exert their protolerogenic function, fibrocytic MDSCs require direct contact with activated T cells, which leads to the production and secretion of IDO. This new myeloid subset may have an important role in the in vitro and in vivo production of Treg cells for the treatment of autoimmune diseases, and in either the prevention or control of allograft rejection.


Asunto(s)
Indolamina-Pirrol 2,3,-Dioxigenasa/biosíntesis , Activación de Linfocitos/inmunología , Células Mieloides/inmunología , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología , Animales , Diferenciación Celular/inmunología , Línea Celular , Proliferación Celular , Diabetes Mellitus Tipo 1/inmunología , Femenino , Sangre Fetal/citología , Perfilación de la Expresión Génica , Rechazo de Injerto/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Células HEK293 , Humanos , Tolerancia Inmunológica , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Células Mieloides/citología , Proteínas Recombinantes/farmacología , Linfocitos T Reguladores/trasplante
2.
Biotechnol Bioeng ; 112(9): 1916-26, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25786390

RESUMEN

With a view toward reduction of graft loss, we explored pancreatic islet transplantation within fibrin matrices rendered pro-angiogenic by incorporation of minimal doses of vascular endothelial growth factor-A165 and platelet-derived growth factor-BB presented complexed to a fibrin-bound integrin-binding fibronectin domain. Engineered matrices allowed for extended release of pro-angiogenic factors and for their synergistic signaling with extracellular matrix-binding domains in the post-transplant period. Aprotinin addition delayed matrix degradation and prolonged pro-angiogenic factor availability within the graft. Both subcutaneous (SC) and epididymal fat pad (EFP) sites were evaluated. We show that in the SC site, diabetes reversal in mice transplanted with 1,000 IEQ of syngeneic islets was not observed for islets transplanted alone, while engineered matrices resulted in a diabetes median reversal time (MDRT) of 38 days. In the EFP site, the MDRT with 250 IEQ of syngeneic islets within the engineered matrices was 24 days versus 86 days for islets transplanted alone. Improved function of engineered grafts was associated with enhanced and earlier (by day 7) angiogenesis. Our findings show that by engineering the transplant site to promote prompt re-vascularization, engraftment and long-term function of islet grafts can be improved in relevant extrahepatic sites.


Asunto(s)
Fibrina/química , Trasplante de Islotes Pancreáticos , Islotes Pancreáticos/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/farmacología , Animales , Becaplermina , Proliferación Celular/efectos de los fármacos , Humanos , Hidrogeles/química , Ratones , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas c-sis/química , Proteínas Proto-Oncogénicas c-sis/deficiencia , Proteínas Proto-Oncogénicas c-sis/farmacología , Factor A de Crecimiento Endotelial Vascular/química
3.
Stem Cells ; 31(10): 2047-60, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23873634

RESUMEN

Regenerative medicine is transitioning into clinical programs using stem/progenitor cell therapies for repair of damaged organs. We summarize those for liver and pancreas, organs that share endodermal stem cell populations, biliary tree stem cells (hBTSCs), located in peribiliary glands. They are precursors to hepatic stem/progenitors in canals of Hering and to committed progenitors in pancreatic duct glands. They give rise to maturational lineages along a radial axis within bile duct walls and a proximal-to-distal axis starting at the duodenum and ending with mature cells in the liver or pancreas. Clinical trials have been ongoing for years assessing effects of determined stem cells (fetal-liver-derived hepatic stem/progenitors) transplanted into the hepatic artery of patients with various liver diseases. Immunosuppression was not required. Control subjects, those given standard of care for a given condition, all died within a year or deteriorated in their liver functions. Subjects transplanted with 100-150 million hepatic stem/progenitor cells had improved liver functions and survival extending for several years. Full evaluations of safety and efficacy of transplants are still in progress. Determined stem cell therapies for diabetes using hBTSCs remain to be explored but are likely to occur following ongoing preclinical studies. In addition, mesenchymal stem cells (MSCs) and hematopoietic stem cells (HSCs) are being used for patients with chronic liver conditions or with diabetes. MSCs have demonstrated significant effects through paracrine signaling of trophic and immunomodulatory factors, and there is limited evidence for inefficient lineage restriction into mature parenchymal or islet cells. HSCs' effects are primarily via modulation of immune mechanisms.


Asunto(s)
Hepatitis/terapia , Trasplante de Células Madre Mesenquimatosas , Pancreatitis/terapia , Diferenciación Celular , Linaje de la Célula , Hepatitis/inmunología , Humanos , Hígado/embriología , Hígado/inmunología , Hígado/patología , Células Madre Mesenquimatosas/fisiología , Páncreas/embriología , Páncreas/inmunología , Páncreas/patología , Pancreatitis/inmunología , Nicho de Células Madre
4.
Stem Cells ; 31(9): 1966-79, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23847135

RESUMEN

Peribiliary glands (PBGs) in bile duct walls, and pancreatic duct glands (PDGs) associated with pancreatic ducts, in humans of all ages, contain a continuous, ramifying network of cells in overlapping maturational lineages. We show that proximal (PBGs)-to-distal (PDGs) maturational lineages start near the duodenum with cells expressing markers of pluripotency (NANOG, OCT4, and SOX2), proliferation (Ki67), self-replication (SALL4), and early hepato-pancreatic commitment (SOX9, SOX17, PDX1, and LGR5), transitioning to PDG cells with no expression of pluripotency or self-replication markers, maintenance of pancreatic genes (PDX1), and expression of markers of pancreatic endocrine maturation (NGN3, MUC6, and insulin). Radial-axis lineages start in PBGs near the ducts' fibromuscular layers with stem cells and end at the ducts' lumens with cells devoid of stem cell traits and positive for pancreatic endocrine genes. Biliary tree-derived cells behaved as stem cells in culture under expansion conditions, culture plastic and serum-free Kubota's Medium, proliferating for months as undifferentiated cells, whereas pancreas-derived cells underwent only approximately 8-10 divisions, then partially differentiated towards an islet fate. Biliary tree-derived cells proved precursors of pancreas' committed progenitors. Both could be driven by three-dimensional conditions, islet-derived matrix components and a serum-free, hormonally defined medium for an islet fate (HDM-P), to form spheroids with ultrastructural, electrophysiological and functional characteristics of neoislets, including glucose regulatability. Implantation of these neoislets into epididymal fat pads of immunocompromised mice, chemically rendered diabetic, resulted in secretion of human C-peptide, regulatable by glucose, and able to alleviate hyperglycemia in hosts. The biliary tree-derived stem cells and their connections to pancreatic committed progenitors constitute a biological framework for life-long pancreatic organogenesis.


Asunto(s)
Sistema Biliar/citología , Linaje de la Célula , Organogénesis , Páncreas/citología , Páncreas/crecimiento & desarrollo , Células Madre/citología , Adulto , Animales , Antígenos de Neoplasias/metabolismo , Biomarcadores/metabolismo , Moléculas de Adhesión Celular/metabolismo , Diferenciación Celular/genética , Linaje de la Célula/genética , Diabetes Mellitus Experimental/fisiopatología , Diabetes Mellitus Experimental/terapia , Fenómenos Electrofisiológicos , Molécula de Adhesión Celular Epitelial , Regulación de la Expresión Génica , Humanos , Hiperglucemia/terapia , Islotes Pancreáticos/citología , Islotes Pancreáticos/fisiología , Islotes Pancreáticos/ultraestructura , Trasplante de Islotes Pancreáticos , Ratones , Organogénesis/genética , Conductos Pancreáticos/citología , Fenotipo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Esferoides Celulares/citología , Esferoides Celulares/metabolismo , Esferoides Celulares/ultraestructura , Nicho de Células Madre/genética , Células Madre/metabolismo
5.
Hepatology ; 54(6): 2159-72, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21809358

RESUMEN

UNLABELLED: Multipotent stem/progenitors are present in peribiliary glands of extrahepatic biliary trees from humans of all ages and in high numbers in hepato-pancreatic common duct, cystic duct, and hilum. They express endodermal transcription factors (e.g., Sox9, SOX17, FOXA2, PDX1, HES1, NGN3, PROX1) intranuclearly, stem/progenitor surface markers (EpCAM, NCAM, CD133, CXCR4), and sometimes weakly adult liver, bile duct, and pancreatic genes (albumin, cystic fibrosis transmembrane conductance regulator [CFTR], and insulin). They clonogenically expand on plastic and in serum-free medium, tailored for endodermal progenitors, remaining phenotypically stable as undifferentiated cells for months with a cell division initially every ≈36 hours and slowing to one every 2-3 days. Transfer into distinct culture conditions, each comprised of a specific mix of hormones and matrix components, yields either cords of hepatocytes (express albumin, CYP3A4, and transferrin), branching ducts of cholangiocytes (expressing anion exchanger-2-AE2 and CFTR), or regulatable C-peptide secreting neoislet-like clusters (expressing glucagon, insulin) and accompanied by changes in gene expression correlating with the adult fate. Transplantation into quiescent livers of immunocompromised mice results in functional human hepatocytes and cholangiocytes, whereas if into fat pads of streptozocin-induced diabetic mice, results in functional islets secreting glucose-regulatable human C-peptide. CONCLUSION: The phenotypes and availability from all age donors suggest that these stem/progenitors have considerable potential for regenerative therapies of liver, bile duct, and pancreatic diseases including diabetes.


Asunto(s)
Sistema Biliar/citología , Diferenciación Celular/fisiología , Hepatocitos/citología , Islotes Pancreáticos/citología , Células Madre Multipotentes/citología , Animales , Linaje de la Célula , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Humanos , Ratones , Ratones SCID , Regeneración/fisiología , Factores de Transcripción/biosíntesis , Trasplante Heterólogo
6.
Curr Opin Organ Transplant ; 16(1): 76-82, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21150613

RESUMEN

PURPOSE OF REVIEW: The promise of islet transplantation for type 1 diabetes has been hampered by the lack of a renewable source of insulin-producing cells. However, steadfast advances in the field have set the stage for stem cell-based approaches to take over in the near future. This review focuses on the most intriguing findings reported in recent years, which include not only progress in adult and embryonic stem cell differentiation, but also the direct reprogramming of nonendocrine tissues into insulin-producing beta cells. RECENT FINDINGS: In spite of their potential for tumorigenesis, human embryonic stem (hES) cells are poised to be in clinical trials within the next decade. This situation is mainly due to the preclinical success of a differentiation method that recapitulates beta cell development. In contrast, adult stem cells still need one such gold standard of differentiation, and progress is somewhat impeded by the lack of consensus on the best source. A concerted effort is necessary to bring their potential to clinical fruition. In the meantime, reported success in reprogramming might offer a 'third way' towards the rescue of pancreatic endocrine function. SUMMARY: Here we discuss the important strategic decisions that need to be made in order to maximize the therapeutic chances of each of the presented approaches.


Asunto(s)
Células Madre Adultas/trasplante , Células Madre Embrionarias/citología , Células Madre Embrionarias/trasplante , Trasplante de Islotes Pancreáticos/métodos , Islotes Pancreáticos/citología , Animales , Diferenciación Celular/fisiología , Diabetes Mellitus Tipo 1/cirugía , Humanos
7.
Am J Clin Dermatol ; 11 Suppl 1: 3-9, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20586498

RESUMEN

Tumor necrosis factor alpha (TNFalpha) plays a key pathophysiological role in psoriasis and psoriatic arthritis (PsA). Recent interest has thus focused on the clinical potential of TNFalpha antagonists (e.g. etanercept) in these settings. In psoriasis, several large pooled analyses and well-designed clinical trials documented the significant clinical efficacy and generally favorable tolerability of etanercept for up to 96 weeks. Similarly, in PsA, a large phase III trial showed that, etanercept significantly reduced arthritic symptoms and inhibited radiographic disease progression; sustained clinical benefit was again evident for up to 2 years. Etanercept is at the forefront of psoriatic disease management, and continued evolution and evaluation of the compound - for example, in detailed comparative studies and economic analyses - is likely to confirm a key role for etanercept in the treatment of psoriasis and PsA.


Asunto(s)
Artritis Psoriásica/tratamiento farmacológico , Inmunoglobulina G/uso terapéutico , Psoriasis/tratamiento farmacológico , Receptores del Factor de Necrosis Tumoral/uso terapéutico , Antirreumáticos/efectos adversos , Antirreumáticos/farmacología , Antirreumáticos/uso terapéutico , Artritis Psoriásica/fisiopatología , Ensayos Clínicos como Asunto , Progresión de la Enfermedad , Etanercept , Humanos , Inmunoglobulina G/efectos adversos , Inmunoglobulina G/farmacología , Psoriasis/fisiopatología , Receptores del Factor de Necrosis Tumoral/metabolismo
8.
Biol Cell ; 101(8): 431-40, 2009 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-19583566

RESUMEN

Beyond its role as an electron acceptor in aerobic respiration, oxygen is also a key effector of many developmental events. The oxygen-sensing machinery and the very fabric of cell identity and function have been shown to be deeply intertwined. Here we take a first look at how oxygen might lie at the crossroads of at least two of the major molecular pathways that shape pancreatic development. Based on recent evidence and a thorough review of the literature, we present a theoretical model whereby evolving oxygen tensions might choreograph to a large extent the sequence of molecular events resulting in the development of the organ. In particular, we propose that lower oxygenation prior to the expansion of the vasculature may favour HIF (hypoxia inducible factor)-mediated activation of Notch and repression of Wnt/beta-catenin signalling, limiting endocrine cell differentiation. With the development of vasculature and improved oxygen delivery to the developing organ, HIF-mediated support for Notch signalling may decline while the beta-catenin-directed Wnt signalling is favoured, which would support endocrine cell differentiation and perhaps exocrine cell proliferation/differentiation.


Asunto(s)
Oxígeno/metabolismo , Páncreas/crecimiento & desarrollo , Páncreas/metabolismo , Animales , Diferenciación Celular , Proliferación Celular , Humanos , Páncreas/citología , Transducción de Señal , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
9.
Methods Mol Biol ; 1871: 253-264, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30276744

RESUMEN

Diabetes mellitus is caused by either loss of pancreatic islets ß-cells (Type 1 Diabetes, T1D), insufficient insulin release in the islet ß-cells coupled with insulin resistance in target tissues (Type 2 Diabetes, T2D), or impaired insulin release (genetic forms of diabetes and, possibly, T1D subtypes). The investigation of the islet proteome could elucidate facets of the pathogenesis of diabetes. Enzymatically isolated and cultured (EIC) islets are frequently used to investigate biochemical signaling pathways that could trigger ß-cell changes and death in diabetes. However, they cannot fully reflect the natural protein composition and disease process of in vivo islets due to the stress from isolation procedures and in vitro culture. The laser capture microdissection method employs a high-energy laser source to separate the desired cells from the remaining tissue section in an environment which is well conserved and close to the natural condition. Here, we describe a label-free proteomic workflow of laser capture microdissected (LCM) human islets from fresh-frozen pancreas sections of cadaveric donors to obtain an accurate and unbiased profile of the pancreatic islet proteome. The workflow includes preparation of frozen tissue section, staining and dehydration, LCM islets collection, islet protein digestion, label-free Liquid Chromatography-Tandem Mass Spectrometry (LC-MS/MS), database search, and statistical analysis.


Asunto(s)
Cromatografía Liquida , Islotes Pancreáticos/metabolismo , Proteoma , Proteómica , Espectrometría de Masas en Tándem , Cromatografía Liquida/métodos , Biología Computacional/métodos , Análisis de Datos , Bases de Datos de Proteínas , Secciones por Congelación , Humanos , Inmunohistoquímica , Islotes Pancreáticos/citología , Captura por Microdisección con Láser , Proteoma/metabolismo , Proteómica/métodos , Espectrometría de Masas en Tándem/métodos
10.
Stem Cell Reports ; 12(3): 611-623, 2019 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-30773486

RESUMEN

The transplantation of human embryonic stem cell (hESC)-derived insulin-producing ß cells for the treatment of diabetes is finally approaching the clinical stage. However, even with state-of-the-art differentiation protocols, a significant percentage of undefined non-endocrine cell types are still generated. Most importantly, there is the potential for carry-over of non-differentiated cell types that may produce teratomas. We sought to modify hESCs so that their differentiated progeny could be selectively devoid of tumorigenic cells and enriched for cells of the desired phenotype (in this case, ß cells). Here we report the generation of a modified hESC line harboring two suicide gene cassettes, whose expression results in cell death in the presence of specific pro-drugs. We show the efficacy of this system at enriching for ß cells and eliminating tumorigenic ones both in vitro and in vivo. Our approach is innovative inasmuch as it allows for the preservation of the desired cells while eliminating those with the potential to develop teratomas.


Asunto(s)
Carcinogénesis/patología , Células Madre Embrionarias Humanas/patología , Células Secretoras de Insulina/patología , Animales , Carcinogénesis/genética , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Línea Celular , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Teratoma/genética , Teratoma/patología
11.
Lab Invest ; 88(11): 1167-77, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18779781

RESUMEN

Substantial amounts of nonendocrine cells are implanted as part of human islet grafts, and a possible influence of nonendocrine cells on clinical islet transplantation outcome has been postulated. There are currently no product release criteria specific for nonendocrine cells due to lack of available methods. The aims of this study were to develop a method for the evaluation of pancreatic ductal cells (PDCs) for clinical islet transplantation and to characterize them regarding phenotype, viability, and function. We assessed 161 human islet preparations using laser scanning cytometry (LSC/iCys) for phenotypic analysis of nonendocrine cells and flow cytometry (FACS) for PDC viability. PDC and beta-cells obtained from different density fractions during the islet cell purification were compared in terms of viability. Furthermore, we examined PDC ability to produce proinflammatory cytokines/chemokines, vascular endothelial growth factor (VEGF) and tissue factor (TF) relevant to islet graft outcome. Phenotypic analysis by LSC/iCys indicated that single staining for CK19 or CA19-9 was not enough for identifying PDCs, and that double staining for amylase and CK19 or CA19-9 allowed for quantitative evaluation of acinar cells and PDC content in human islet preparation. PDC showed a significantly higher viability than beta-cells (PDC vs beta-cell: 75.5+/-13.9 and 62.7+/-18.7%; P<0.0001). Although beta-cell viability was independent of its density, that of PDCs was higher as the density from which they were recovered increased. There was no correlation between PDCs and beta-cell viability (R(2)=0.0078). PDCs sorted from high-density fractions produced significantly higher amounts of proinflammatory mediators and VEGF, but not TF. We conclude that PDCs isolated from different fractions had different viability and functions. The precise characterization and assessment of these cells in addition to beta-cells in human islet cell products may be of assistance in understanding their contribution to islet engraftment and in developing strategies to enhance islet graft function.


Asunto(s)
Antígeno CA-19-9 , Células Secretoras de Insulina/citología , Trasplante de Islotes Pancreáticos , Queratina-19 , Conductos Pancreáticos/citología , Animales , Diabetes Mellitus Experimental , Humanos , Células Secretoras de Insulina/clasificación , Islotes Pancreáticos/citología , Citometría de Barrido por Láser , Ratones , Ratones Desnudos , Fenotipo
12.
Transplantation ; 86(1): 46-53, 2008 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-18622277

RESUMEN

BACKGROUND: Sirolimus plays a critical role in facilitating steroid-free immunosuppression, in conjunction with low dose tacrolimus, in current islet transplantation. Although several studies have investigated the effects of sirolimus on islet cells, conflicting results have been reported. In this study, we assessed the effects of sirolimus supplementation in culture media on human islet preparations, focusing on the anti-proinflammatory aspects. METHODS: Human islet preparations were divided into four groups: pure (purity >90%) sirolimus (30 ng/mL); pure control (0 ng/mL); impure (purity 40%-60%) sirolimus; and impure control. All groups were cultured for 3 days and assessed regarding glucose stimulated insulin release, fractional beta-cell viability, beta-cell, and macrophage content. Cytokine and chemokine production from islet preparations and sorted pancreatic ductal cells were also examined. RESULTS: Stimulated insulin release in the impure sirolimus group was significantly increased (P=0.024), as previously reported. Although fractional beta-cell viability showed no significant differences, beta-cell survival during culture significantly increased in impure sirolimus group when compared with the impure control group (P=0.015). Tumor necrosis factor-alpha, interleukin-1beta, monocyte chemotactic protein-1, and macrophage inflammatory protein-1beta production from the impure sirolimus group significantly decreased (P<0.05). Furthermore, tumor necrosis factor-alpha and macrophage inflammatory protein-1beta production from sorted ductal cells significantly decreased in the sirolimus group (P<0.05). The number of macrophages contained in islet preparations significantly decreased in the impure sirolimus group when compared with the impure control group (P<0.05). CONCLUSIONS: Sirolimus improved not only stimulated insulin release, but also beta-cell survival during culture. The antiinflammatory effects of sirolimus also appear beneficial to islet cells in culture and may be a useful strategy in improving islet transplantation outcomes.


Asunto(s)
Antiinflamatorios/farmacología , Inmunosupresores/farmacología , Mediadores de Inflamación/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Sirolimus/farmacología , Supervivencia Celular , Quimiocina CCL2/metabolismo , Quimiocina CCL4/metabolismo , Glucosa/metabolismo , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Interleucina-1beta/metabolismo , Islotes Pancreáticos/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Conductos Pancreáticos/efectos de los fármacos , Conductos Pancreáticos/metabolismo , Factores de Tiempo , Técnicas de Cultivo de Tejidos , Factor de Necrosis Tumoral alfa/metabolismo
13.
Stem Cells ; 25(12): 3155-64, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17761759

RESUMEN

Despite progress in our knowledge about pancreatic islet specification, most attempts at differentiating stem/progenitor cells into functional, transplantable beta cells have met only with moderate success thus far. A major challenge is the intrinsic simplicity of in vitro culture systems, which cannot approximate the physiological complexity of in vivo microenvironments. Oxygenation is a critical limitation of standard culture methods, and one of special relevance for the development of beta cells, known for their high O(2) requirements. Based on our understanding of islet physiology, we have tested the hypothesis that enhanced O(2) delivery (as provided by novel perfluorocarbon-based culture devices) may result in higher levels of beta-cell differentiation from progenitor cells in vitro. Using a mouse model of pancreatic development, we demonstrate that a physiological-like mode of O(2) delivery results in a very significant upregulation of endocrine differentiation markers (up to 30-fold for insulin one and 2), comparable to relevant in vivo controls. This effect was not observed by merely increasing environmental O(2) concentrations in conventional settings. Our findings indicate that O(2) plays an important role in the differentiation of beta cells from their progenitors and may open the door to more efficient islet differentiation protocols from embryonic and/or adult stem cells. Disclosure of potential conflicts of interest is found at the end of this article.


Asunto(s)
Diferenciación Celular/fisiología , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Consumo de Oxígeno/fisiología , Oxígeno/metabolismo , Animales , Células Cultivadas , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Oxígeno/fisiología , Embarazo
14.
Cell Transplant ; 17(5): 559-66, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18714675

RESUMEN

Riboflavin is a water-soluble vitamin that reduces the production of proinflammatory mediators and oxygen radicals. Because islet beta-cells are very sensitive to oxidative stress and to cytokines, we investigated the possible cytoprotective effects of riboflavin on insulinoma NIT-1 cells and on isolated rodent islets. NIT-1 cells and islets cultured in the presence or absence of 10 microM riboflavin were studied at baseline and after exposure to cytokines (TNF-alpha, IL-1beta, INF-gamma). Riboflavin treatment did not affect islet cell viability as assessed by flow cytometry for caspases activation. However, riboflavin prevented the cytokine-induced increase in IL-6 mRNA expression and p38 phosphorylation analyzed by real-time PCR and immunoassay, respectively. In summary, nontoxic doses of riboflavin prevent cytokines-induced p38 phosphorylation and IL-6 upregulation in islet cells. This observation, together with the safety profile of riboflavin in the clinical setting, makes it an appealing agent for islet cytoprotection in islet transplantation protocols.


Asunto(s)
Expresión Génica/efectos de los fármacos , Interleucina-6/biosíntesis , Islotes Pancreáticos/metabolismo , Riboflavina/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Células Cultivadas , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/terapia , Activación Enzimática/efectos de los fármacos , Interleucina-1beta/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Trasplante de Islotes Pancreáticos , Masculino , Ratones , Ratones Endogámicos NOD , Ratas , Riboflavina/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
15.
Cell Rep ; 22(9): 2408-2420, 2018 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-29490276

RESUMEN

Treatment of human pancreatic non-endocrine tissue with Bone Morphogenetic Protein 7 (BMP-7) leads to the formation of glucose-responsive ß-like cells. Here, we show that BMP-7 acts on extrainsular cells expressing PDX1 and the BMP receptor activin-like kinase 3 (ALK3/BMPR1A). In vitro lineage tracing indicates that ALK3+ cell populations are multipotent. PDX1+/ALK3+ cells are absent from islets but prominently represented in the major pancreatic ducts and pancreatic duct glands. We identified the purinergic receptor P2Y1 (P2RY1) as a surrogate surface marker for PDX1. Sorted P2RY1+/ALK3bright+ cells form BMP-7-expandable colonies characterized by NKX6.1 and PDX1 expression. Unlike the negative fraction controls, these colonies can be differentiated into multiple pancreatic lineages upon BMP-7 withdrawal. RNA-seq further corroborates the progenitor-like nature of P2RY1+/ALK3bright+ cells and their multilineage differentiation potential. Our studies confirm the existence of progenitor cells in the adult human pancreas and suggest a specific anatomical location within the ductal and glandular networks.


Asunto(s)
Proteína Morfogenética Ósea 7/farmacología , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/metabolismo , Páncreas Exocrino/metabolismo , Células Madre/citología , Células Madre/metabolismo , Adulto , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula/efectos de los fármacos , Linaje de la Célula/genética , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Replicación del ADN/efectos de los fármacos , Proteínas de Homeodominio/metabolismo , Humanos , Células Madre/efectos de los fármacos , Transactivadores/metabolismo
16.
Curr Opin Immunol ; 15(5): 507-11, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-14499257

RESUMEN

Clinical trials of islet transplantation are showing remarkable success, but they require administration of chronic immunosuppression, and are underscoring the large gap that exists between the number of human donors available and the number of patients that could benefit from the procedure. Recent progress has been made in the definition of key immunological mechanisms that are involved in determining islet transplant outcome. Clinical and preclinical studies, and studies in small animal model systems, will all eventually contribute to the definition of efficient and safe protocols for islet transplantation. If the use of xenografts is successful, it might represent a solution to the shortage of human organs.


Asunto(s)
Trasplante de Islotes Pancreáticos/inmunología , Animales , Rechazo de Injerto/inmunología , Humanos , Tolerancia Inmunológica , Terapia de Inmunosupresión , Ratones , Ratones Endogámicos NOD , Ratones Obesos , Modelos Animales , Trasplante Heterólogo/inmunología
17.
Transplantation ; 84(12): 1576-83, 2007 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-18165767

RESUMEN

BACKGROUND: Progressive graft dysfunction is commonly observed in recipients of islet allografts treated with high doses of rapamycin. This study aimed at evaluating the effect of rapamycin on pancreatic islet cell proliferation in vivo. METHODS: The murine pregnancy model was utilized, since a high rate of beta-cell proliferation occurs in a well-defined time frame. Rapamycin (0.2 mg/kg/day) was given to C57BL/6 mice for 5-7 days starting on day 7.5 of pregnancy. Cell proliferation was evaluated by detection of bromodeoxyuridine incorporation by immunohistochemistry. RESULTS: Pregnancy led to increased beta-cell proliferation and islet yield with skewing in islet size distribution as well as higher pancreatic insulin content, when compared to that of nonpregnant females. These effects of pregnancy on beta-cell proliferation and mass were significantly blunted by rapamycin treatment. Minimal effect of rapamycin was observed on islet function both in vivo and in vitro. Rapamycin treatment of islets in vitro resulted in reduced p70s6k phosphorylation, which was paralleled by increased ERK1/2 phosphorylation. CONCLUSIONS: Rapamycin treatment reduces the rate of beta-cell proliferation in vivo. This phenomenon may contribute to impair beta-cell renewal in transplanted patients and to the progressive dysfunction observed in islet graft recipients.


Asunto(s)
División Celular/efectos de los fármacos , Células Secretoras de Insulina/citología , Sirolimus/farmacología , Animales , Glucemia/metabolismo , Separación Celular/métodos , Inmunosupresores/farmacología , Insulina/análisis , Células Secretoras de Insulina/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Páncreas/citología , Páncreas/efectos de los fármacos , Proteínas Quinasas/metabolismo
18.
J Proteomics ; 150: 149-159, 2017 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-27620696

RESUMEN

The etiology of Type 1 Diabetes (T1D) remains elusive. Enzymatically isolated and cultured (EIC) islets cannot fully reflect the natural protein composition and disease process of in vivo islets, because of the stress from isolation procedures. In order to study islet protein composition in conditions close to the natural environment, we performed proteomic analysis of EIC islets, and laser capture microdissected (LCM) human islets and acinar tissue from fresh-frozen pancreas sections of three cadaveric donors. 1104 and 706 proteins were identified from 6 islets equivalents (IEQ) of LCM islets and acinar tissue, respectively. The proteomic profiles of LCM islets were reproducible within and among cadaveric donors. The endocrine hormones were only detected in LCM islets, whereas catalytic enzymes were significantly enriched in acinar tissue. Furthermore, high overlap (984 proteins) and similar function distribution were found between LCM and EIC islets proteomes, except that EIC islets had more acinar contaminants and stress-related signal transducer activity proteins. The comparison among LCM islets, LCM acinar tissue and EIC islets proteomes indicates that LCM combined with proteomic methods enables accurate and unbiased profiling of islet proteome from frozen pancreata. This paves the way for proteomic studies on human islets during the progression of T1D. SIGNIFICANCE: The etiological agent triggering autoimmunity against beta cells in Type 1 diabetes (T1D) remains obscure. The in vitro models available (enzymatically isolated and cultured islets, EIC islets) do not accurately reflect what happens in vivo due to lack of the natural environment where islets exist and the preparation-induced changes in cell physiology. The importance of this study is that we investigated the feasibility of laser capture microdissection (LCM) for the isolation of intact islets from frozen cadaveric pancreatic tissue sections. We compared the protein profile of LCM islets (9 replicates from 3 cadaveric donors) with that of both LCM acinar tissues (6 replicates from the same 3 cadaveric donor as LCM islets) and EIC islets (at least 4 replicates for each sample with the same islets equivalents) by using proteomics techniques with advanced instrumentation, nanoLC-Q Exactive HF Orbitrap mass spectrometry (nano LC-MS/MS). The results demonstrate that the LCM method is reliable in isolating islets with an intact environment. LCM-based islet proteomics is a feasible approach to obtain good proteome coverage for assessing the pathology of T1D using cadaveric pancreatic samples, even from very small sample amounts. Future applications of this LCM-based proteomic method may help us understand the pathogenesis of T1D and identify potential biomarkers for T1D diagnosis at an early stage.


Asunto(s)
Secciones por Congelación , Islotes Pancreáticos/metabolismo , Captura por Microdisección con Láser , Páncreas/metabolismo , Proteoma/análisis , Cadáver , Separación Celular/métodos , Células Cultivadas , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patología , Humanos , Islotes Pancreáticos/patología , Páncreas/patología , Cultivo Primario de Células , Proteoma/metabolismo , Proteómica/métodos , Manejo de Especímenes/métodos
19.
Diabetes ; 54(3): 720-6, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15734848

RESUMEN

Stem cell technologies hold great potential for the treatment of type 1 diabetes, provided that functional transplantable beta-cells can be selectively generated in an efficient manner. Such a process should recapitulate, at least to a certain extent, the embryonic development of beta-cells in vitro. However, progress at identifying the transcription factors involved in beta-cell development has not been accompanied by a parallel success at unraveling the pattern of their instructive extracellular signals. Here we present proof of principle of a novel approach to circumvent this problem, based on the use of the HIV/TAT protein transduction domain. Neurogenin 3 (ngn3), a factor whose expression is essential for pancreatic endocrine differentiation, was fused to the TAT domain. Administration of TAT/ngn3 to cultured pancreatic explants results in efficient uptake, nuclear translocation, and stimulation of downstream reporter and endogenous genes. Consistent with the predicted activity of the protein, e9.5 and e13.5 mouse pancreatic explants cultured in the presence of TAT/ngn3 show an increased level of endocrine differentiation compared with control samples. Our results raise the possibility of sequentially specifying stem/progenitor cells toward the beta-cell lineage, by using the appropriate sequence and combination of TAT-fused transcription factors.


Asunto(s)
Proteínas del Tejido Nervioso/biosíntesis , Páncreas/citología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Diferenciación Celular , Línea Celular , Expresión Génica , Productos del Gen tat/genética , Vectores Genéticos , Islotes Pancreáticos/citología , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/fisiología , Proteínas Recombinantes de Fusión/biosíntesis , Transducción Genética
20.
Transplantation ; 81(9): 1318-24, 2006 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-16699461

RESUMEN

BACKGROUND: Transplantation of pancreatic islets for the treatment of type 1 diabetes allows for physiologic glycemic control and insulin-independence when sufficient islets are implanted via the portal vein into the liver. Intrahepatic islet implantation requires specific infrastructure and expertise, and risks inherent to the procedure include bleeding, thrombosis, and elevation of portal pressure. Additionally, the relatively higher drug metabolite concentrations in the liver may contribute to the delayed loss of graft function of recent clinical trials. Identification of alternative implantation sites using biocompatible devices may be of assistance improving graft outcome. A desirable bioartificial pancreas should be easy to implant, biopsy, and retrieve, while allowing for sustained graft function. The subcutaneous (SC) site may require a minimally invasive procedure performed under local anesthesia, but its use has been hampered so far by lack of early vascularization, induction of local inflammation, and mechanical stress on the graft. METHODS: Chemically diabetic rats received syngeneic islets into the liver or SC into a novel biocompatible device consisting of a cylindrical stainless-steel mesh. The device was implanted 40 days prior to islet transplantation to allow embedding by connective tissue and neovascularization. Reversal of diabetes and glycemic control was monitored after islet transplantation. RESULTS: Syngeneic islets transplanted into a SC, neovascularized device restored euglycemia and sustained function long-term. Removal of graft-bearing devices resulted in hyperglycemia. Explanted grafts showed preserved islets and intense vascular networks. CONCLUSIONS: Ease of implantation, biocompatibility, and ability to maintain long-term graft function support the potential of our implantable device for cellular-based reparative therapies.


Asunto(s)
Diabetes Mellitus Experimental/cirugía , Trasplante de Islotes Pancreáticos/instrumentación , Trasplante de Islotes Pancreáticos/fisiología , Animales , Materiales Biocompatibles , Glucemia/metabolismo , Peso Corporal , Diabetes Mellitus Experimental/metabolismo , Modelos Animales de Enfermedad , Trasplante de Islotes Pancreáticos/métodos , Masculino , Politetrafluoroetileno , Ratas , Ratas Endogámicas Lew , Trasplante Isogénico , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA