Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
2.
Proc Natl Acad Sci U S A ; 107(9): 4305-10, 2010 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-20160085

RESUMEN

Islet transplantation is a promising treatment for diabetes but long-term success is limited by progressive graft loss. Aggregates of the beta cell peptide islet amyloid polypeptide (IAPP) promote beta cell apoptosis and rapid amyloid formation occurs in transplanted islets. Porcine islets are an attractive alternative islet source as they demonstrate long-term graft survival. We compared the capacity of transplanted human and porcine islets to form amyloid as an explanation for differences in graft survival. Human islets were transplanted into streptozotocin-diabetic immune-deficient mice. Amyloid deposition was detectable at 4 weeks posttransplantation and was associated with islet graft failure. More extensive amyloid deposition was observed after 8 weeks. By contrast, no amyloid was detected in transplanted neonatal or adult porcine islets that had maintained normoglycemia for up to 195 days. To determine whether differences in IAPP sequence between humans and pigs could explain differences in amyloid formation and transplant viability, we sequenced porcine IAPP. Porcine IAPP differs from the human sequence at 10 positions and includes substitutions predicted to reduce its amyloidogenicity. Synthetic porcine IAPP was considerably less amyloidogenic than human IAPP as determined by transmission electron microscopy, circular dichroism, and thioflavin T binding. Viability assays indicated that porcine IAPP is significantly less toxic to INS-1 beta cells than human IAPP. Our findings demonstrate that species differences in IAPP sequence can explain the lack of amyloid formation and improved survival of transplanted porcine islets. These data highlight the potential of porcine islet transplantation as a therapeutic approach for human diabetes.


Asunto(s)
Amiloide/metabolismo , Trasplante de Islotes Pancreáticos , Islotes Pancreáticos/metabolismo , Secuencia de Aminoácidos , Amiloide/química , Amiloide/fisiología , Animales , Dicroismo Circular , Rechazo de Injerto , Humanos , Polipéptido Amiloide de los Islotes Pancreáticos , Ratones , Microscopía Electrónica de Transmisión , Datos de Secuencia Molecular , Homología de Secuencia de Aminoácido , Especificidad de la Especie , Porcinos
3.
Cell Prolif ; 41(6): 843-858, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19040565

RESUMEN

OBJECTIVES: Islet-like clusters (ILCs), differentiated from human embryonic stem cells (hESCs), were characterized both before and after transplantation under the kidney capsule of streptozotocin-induced diabetic immuno-incompetent mice. MATERIALS AND METHODS: Multiple independent ILC preparations (n = 8) were characterized by immunohistochemistry, flow cytometry and cell insulin content, with six preparations transplanted into diabetic mice (n = 42), compared to controls, which were transplanted with either a human fibroblast cell line or undifferentiated hESCs (n = 28). RESULTS: Prior to transplantation, ILCs were immunoreactive for the islet hormones insulin, C-peptide and glucagon, and for the ductal epithelial marker cytokeratin-19. ILCs also had cellular insulin contents similar to or higher than human foetal islets. Expression of islet and pancreas-specific cell markers was maintained for 70 days post-transplantation. The mean survival of recipients was increased by transplanted ILCs as compared to transplanted human fibroblast cells (P < 0.0001), or undifferentiated hESCs (P < 0.042). Graft function was confirmed by secretion of human C-peptide in response to an oral bolus of glucose. CONCLUSIONS: hESC-derived ILC grafts continued to contain cells that were positive for islet endocrine hormones and were shown to be functional by their ability to secrete human C-peptide. Further enrichment and maturation of ILCs could lead to generation of a sufficient source of insulin-producing cells for transplantation into patients with type 1 diabetes.


Asunto(s)
Células Madre Embrionarias/citología , Células Endocrinas/citología , Trasplante de Islotes Pancreáticos , Islotes Pancreáticos/citología , Animales , Diferenciación Celular , Línea Celular , ADN/metabolismo , Células Madre Embrionarias/ultraestructura , Células Endocrinas/ultraestructura , Citometría de Flujo , Humanos , Insulina/metabolismo , Islotes Pancreáticos/ultraestructura , Estimación de Kaplan-Meier , Ratones , Ratones Endogámicos NOD
4.
Diabetologia ; 49(10): 2341-9, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16896936

RESUMEN

AIMS/HYPOTHESIS: The Edmonton Protocol for islet transplantation has provided hope for type 1 diabetic patients. However, this protocol requires lifelong immunosuppression, specifically sirolimus, a cellular antiproliferate. The effect of sirolimus on human pancreatic ductal cells (HDCs) is not known. This may be important since HDCs are believed to be islet precursors. Since neonatal porcine islets (NPIs), which contain many ductal precursor cells, could be a potential clinical source of islets, we also tested the effects of sirolimus on this tissue. METHODS: HDCs (n=4), NPIs (n=9) and human islets (n=5) were cultured with and without sirolimus (20 ng/ml) for 6 days. RESULTS: HDCs and NPIs cultured with sirolimus showed a 50 and 28% decrease, respectively, in cell number relative to control (p<0.05). Control cultures expanded 1.65- and 2.44-fold relative to time 0. Decreases in cell number of sirolimus-treated HDCs were not due to apoptosis as measured by TUNEL staining. No functional effects on human islets or NPIs were observed following static incubation with high glucose. Treatment of syngeneically transplanted and naïve BALC/c mice with sirolimus resulted in altered OGTT profiles with prolonged elevation of hyperglycaemia and weight gain. There was no difference in graft and organ insulin content between treatment groups. CONCLUSIONS/INTERPRETATION: Our results indicate that sirolimus decreases ductal cell numbers in culture and alters glucose-stimulated insulin secretion in vivo. The administration of sirolimus to islet transplant recipients is likely to impair graft function as a result of decreasing ductal neogenesis and induction of insulin resistance.


Asunto(s)
División Celular/efectos de los fármacos , Islotes Pancreáticos/citología , Islotes Pancreáticos/fisiología , Conductos Pancreáticos/citología , Conductos Pancreáticos/fisiología , Sirolimus/farmacología , Animales , Cadáver , Diabetes Mellitus Tipo 1/cirugía , Humanos , Insulina/metabolismo , Secreción de Insulina , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Trasplante de Islotes Pancreáticos , Ratones , Conductos Pancreáticos/efectos de los fármacos , Proteínas Quinasas/fisiología , Porcinos , Serina-Treonina Quinasas TOR
5.
Diabetologia ; 49(8): 1855-63, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16736130

RESUMEN

AIMS/HYPOTHESIS: Neurogenin 3 (NEUROG3), a basic helix-loop-helix transcription factor that is needed for endocrine cell development in the embryonic pancreas, has been shown to induce transdifferentiation of duct cells from adult pancreas towards a neuro-endocrine phenotype. Our study explored the endocrine transdifferentiation potential of NEUROG3 in neonatal pancreatic precursor cells. MATERIALS AND METHODS: A replication-deficient adenovirus expressing Neurog3 and green fluorescent protein (GFP) (Ad-NEUROG3) was used to infect neonatal pig pancreatic cell preparations enriched for endocrine islet and cytokeratin-positive precursor cells. GFP-positive cells were sorted using flow cytometry on days 3 and 8 after infection and characterised at the transcript and protein level. For in vivo experiments, the total population of Ad-NEUROG3-infected pancreatic cells was transplanted, then later removed for determination of graft hormone content and immunohistochemistry. RESULTS: Among the GFP-positive cells, the fraction of precursor cells decreased by more than 85% at day 8 after infection, while the fraction of glucagon-positive cells increased 2.5-fold and the beta cell number remained the same. Transplantation of the Ad-NEUROG3-infected pancreatic cell preparation failed to reverse streptozotocin-induced hyperglycaemia, while non-infected cells and a control cell preparation infected with replication-deficient adenovirus expressing only GFP were able to do so. At day 109 after transplantation, kidneys grafted with Ad-NEUROG3-infected pancreatic cells contained significantly decreased insulin and increased glucagon levels. Abundant glucagon-immunopositive cells were seen in Ad-NEUROG3-infected grafts, which were virtually devoid of proliferating insulin-positive cells. CONCLUSIONS/INTERPRETATION: In summary, adenoviral delivery of NEUROG3 to pancreatic precursor cells from neonatal pig pancreas promotes alpha cell differentiation in vitro and in vivo.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Células Secretoras de Glucagón/citología , Trasplante de Islotes Pancreáticos/fisiología , Proteínas del Tejido Nervioso/genética , Adenoviridae , Animales , Animales Modificados Genéticamente , Animales Recién Nacidos , Diferenciación Celular , Cartilla de ADN , Diabetes Mellitus Experimental/cirugía , Glucagón/análisis , Proteínas Fluorescentes Verdes/genética , Inmunohistoquímica , Insulina/análisis , Queratina-7 , Queratinas/análisis , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Porcinos , Sinaptofisina/análisis , Trasplante Heterólogo
6.
Diabetologia ; 47(10): 1810-8, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15517151

RESUMEN

AIMS/HYPOTHESIS: The aim of this study was to determine whether a simple alginate capsule can prolong islet survival and function during long-term tissue culture. We also wanted to observe the ability of these encapsulated islets to restore glucose responsiveness to diabetic recipients, along with the quantity of islets required to do so. METHODS: We compared the recovery and metabolic function of encapsulated canine islets with that of non-encapsulated canine islets following 1, 2 or 3 weeks of tissue culture. These culture preparations were also transplanted into diabetic nude mice and compared for their ability to reverse diabetes. Furthermore, short-term cultured encapsulated and non-encapsulated islets were transplanted in varying numbers to determine the minimum dose required to normalise blood glucose and prolong recipient survival. RESULTS: Islet recovery following 1, 2 and 3 weeks of tissue culture was significantly higher when islets were encapsulated. When these islets were recovered at 1, 2 and 3 weeks and transplanted into diabetic nude mice, survival at 100 days was 100% for all encapsulated groups, versus 66%, 33% and 33% respectively for the non-encapsulated islets. Additionally, substantially fewer short-term cultured islets were required to normalise blood glucose when the islets were encapsulated. Recipients of encapsulated islets also had significantly longer survival times than recipients of non-encapsulated preparations. CONCLUSIONS/INTERPRETATION: This study demonstrates that encapsulation of islets with purified alginate improves islet survival and function in vitro and in vivo.


Asunto(s)
Supervivencia de Injerto/fisiología , Trasplante de Islotes Pancreáticos/fisiología , Animales , Cápsulas , Técnicas de Cultivo de Célula/métodos , Perros , Insulina/metabolismo , Secreción de Insulina , Islotes Pancreáticos/citología , Islotes Pancreáticos/fisiología , Ratones , Modelos Animales , Trasplante Homólogo
7.
J Endocrinol ; 180(2): 213-25, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14765974

RESUMEN

The discovery of a pancreatic adult stem cell would have significant implications for cell-based replacement therapies for type 1 diabetes mellitus. Nestin, a marker for neural precursor cells, has been suggested as a possible marker for islet progenitor cells. We have characterized the expression and localization of nestin in both the intact human pancreas and clinical human pancreatic islet grafts. Nestin was found to be expressed at different levels in the acinar component of human pancreatic biopsies depending on donor, as well as in ductal structures and islets to some degree. In islets, insulin-producing beta-cells rarely co-expressed the protein, and in the ducts a small percentage (1-2%) of cells co-expressed nestin and cytokeratin 19 (CK19) while most expressed only CK19 (90%) or nestin (5-10%) alone. Assessment of nestin expression in neonatal pancreatic sections revealed an increased number of islet-associated positive cells as compared with adult islets. Nestin immunoreactivity was also found in cells of the pancreatic vasculature and mesenchyme as evidenced by co-localization with smooth muscle actin and vimentin. Samples from post-islet isolation clinical islet grafts revealed a pronounced heterogeneity in the proportion of nestin-positive cells (<1-72%). Co-localization studies in these grafts showed that nestin is not co-expressed in endocrine cells and rarely (<5%) with cytokeratin-positive ductal cells. However, relatively high levels of co-expression were found with acinar cells and cells expressing the mesenchymal marker vimentin. In conclusion we have shown a diffuse and variable expression of nestin in human pancreas that may be due to a number of different processes, including post-mortem tissue remodeling and cellular differentiation. For this reason nestin may not be a suitable marker solely for the identification of endocrine precursor cells in the pancreas.


Asunto(s)
Proteínas de Filamentos Intermediarios/genética , Proteínas del Tejido Nervioso , Páncreas/química , ARN Mensajero/análisis , Análisis de Varianza , Biomarcadores/análisis , Humanos , Inmunohistoquímica/métodos , Proteínas de Filamentos Intermediarios/análisis , Islotes Pancreáticos/química , Queratinas/análisis , Microscopía Fluorescente , Nestina , Conductos Pancreáticos/química , Reacción en Cadena de la Polimerasa/métodos , Vimentina/análisis
8.
Diabetologia ; 47(3): 499-508, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-14968299

RESUMEN

AIM/HYPOTHESIS: Embryonic stem (ES) cells have been proposed as a potential source of tissue for transplantation for the treatment of Type 1 diabetes. However, studies showing differentiation of beta cells from ES cells are controversial. The aim of this study was to characterise the insulin-expressing cells differentiated in vitro from ES cells and to assess their suitability for the treatment of diabetes. METHODS: ES cell-derived insulin-expressing cells were characterised by means of immunocytochemistry, RT-PCR and functional analyses. Activation of the Insulin I promoter during ES-cell differentiation was assessed in ES-cell lines transfected with a reporter gene. ES cell-derived cultures were transplanted into STZ-treated SCID-beige mice and blood glucose concentrations of diabetic mice were monitored for 3 weeks. RESULTS: Insulin-stained cells differentiated from ES cells were devoid of typical beta-cell granules, rarely showed immunoreactivity for C-peptide and were mostly apoptotic. The main producers of proinsulin/insulin in these cultures were neurons and neuronal precursors and a reporter gene under the control of the insulin I promoter was activated in cells with a neuronal phenotype. Insulin was released into the incubation medium but the secretion was not glucose-dependent. When the cultures were transplanted in diabetic mice they formed teratomas and did not reverse the hyperglycaemic state. CONCLUSIONS/INTERPRETATION: Our studies show that insulin-positive cells in vitro-differentiated from ES cells are not beta cells and suggest that alternative protocols, based on enrichment of ES cell-derived cultures with cells of the endodermal lineage, should be developed to generate true beta cells for the treatment of diabetes.


Asunto(s)
Insulina/genética , Células Madre/fisiología , Animales , Diferenciación Celular , Línea Celular , Genes Reporteros , Islotes Pancreáticos/citología , Islotes Pancreáticos/embriología , Islotes Pancreáticos/fisiología , Ratones , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/citología , Transfección
9.
Gene Ther ; 11(8): 694-700, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-14724669

RESUMEN

The immunoprotective nature of the testis has led to numerous investigations for its ability to protect cellular grafts. Sertoli cells (SCs) are at least partially responsible for this immunoprotective environment and survive allogeneic and xenogeneic transplantation. The ability of SCs to survive transplantation leads to the possibility that they could be engineered to deliver therapeutic proteins. As a model to test this hypothesis, we examined the ability of SCs that produce green fluorescent protein (GFP) to survive transplantation and continue expressing GFP. SCs were isolated from transgenic mice engineered to express GFP and transplanted as aggregates under the kidney capsule of severe combined immunodeficient (SCID) and Balb/c mice. Using this paradigm, it was possible to compare the survival of transgenic SCs directly in both immunodeficient and immunocompetent recipients. Fluorescence microscopy of the kidney capsule and immunohistochemistry of the grafts for GFP and GATA-4 revealed the presence of GFP-expressing SCs under the kidney capsule of SCID and Balb/c mice at both 30 and 60 days post-transplantation. In contrast, islets transplanted to Balb/c mice were rejected. Thus, SCs survive transplantation and continue to express GFP raising the possibility that SCs can be engineered using transgenic technology to produce proteins, such as insulin, factor VIII, or dopamine for the treatment of diabetes, hemophilia or Parkinson's disease, respectively.


Asunto(s)
Terapia Genética/métodos , Proteínas Luminiscentes/genética , Células de Sertoli/metabolismo , Células de Sertoli/trasplante , Animales , Expresión Génica , Ingeniería Genética , Proteínas Fluorescentes Verdes , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Ratones Transgénicos , Microscopía Fluorescente , Factores de Tiempo , Trasplante Homólogo
10.
J Endocrinol ; 177(1): 127-35, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12697044

RESUMEN

The expression of Galalpha-(1,3)Gal (alphaGal) on porcine islet cells remains controversial. Several groups have reported that porcine islet endocrine cells do not express alphaGal while we have shown in neonatal porcine islets (NPI) that beta cells do express this antigen. We hypothesize that endocrine cells expressing alphaGal on NPI are less mature cells that may have originated from ductal cells and that expression of this antigen disappears as they develop into fully mature beta cells. Thus, we further examined alphaGal expression on various porcine islet cell preparations and correlated this with the proportion of cytokeratin 7 (CK7)-positive ductal cells. In vitro and in vivo expression of alphaGal and CK7 was significantly (P<0.05) higher in less mature NPI cells compared with matured NPI and adult porcine islet cells while the reverse was observed in the proportion of beta cells. Moreover, a significantly higher proportion of CK7-positive cells was detected in the Gal-expressing population compared with non-expressing cells. In contrast, a higher proportion of beta cells was observed in the Gal-negative population compared with the Gal-positive population. These data showed a reduced expression of alphaGal and CK7 as porcine islet cells mature into beta cells suggesting a possible role for alphaGal in the maturation of pancreatic endocrine beta cells.


Asunto(s)
Antígenos Heterófilos/metabolismo , Senescencia Celular/fisiología , Disacáridos/metabolismo , Trasplante de Islotes Pancreáticos , Islotes Pancreáticos/metabolismo , Animales , Animales Recién Nacidos , Citometría de Flujo , Queratina-7 , Queratinas/metabolismo , Porcinos , Trasplante Heterólogo
11.
Lab Invest ; 81(12): 1683-92, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11742038

RESUMEN

The proinflammatory cytokines, interleukin-1beta (IL-1beta), tumor necrosis factor alpha (TNFalpha), and interferon gamma (IFNgamma), are cytotoxic to pancreatic islet beta cells, possibly by inducing nitric oxide and/or oxygen radical production in the beta cells. Peroxynitrite, the reaction product of nitric oxide and the superoxide radical, is a strong oxidant and cytotoxic mediator; therefore, we hypothesized that peroxynitrite might be a mediator of cytokine-induced islet beta-cell destruction. To test this hypothesis we incubated islets isolated from human pancreata with the cytokine combination of IL-1beta, TNFalpha, and IFNgamma. We found that these cytokines induced significant increases in nitrotyrosine, a marker of peroxynitrite, in islet beta cells, and the increase in nitrotyrosine preceded islet-cell destruction. Peroxynitrite mimicked the effects of cytokines on nitrotyrosine formation and islet beta-cell destruction. L-N(G)-monomethyl arginine, an inhibitor of nitric oxide synthase, prevented cytokine-induced nitric oxide production but not hydrogen peroxide production, nitrotyrosine formation, or islet beta-cell destruction. In contrast, guanidinoethyldisulphide, an inhibitor of inducible nitric oxide synthase and scavenger of peroxynitrite, prevented cytokine-induced nitric oxide and hydrogen peroxide production, nitrotyrosine formation, and islet beta-cell destruction. These results suggest that cytokine-induced peroxynitrite formation is dependent upon increased generation of superoxide (measured as hydrogen peroxide) and that peroxynitrite is a mediator of cytokine-induced destruction of human pancreatic islet beta cells.


Asunto(s)
Citocinas/farmacología , Islotes Pancreáticos/efectos de los fármacos , Ácido Peroxinitroso/fisiología , Tirosina/análogos & derivados , Muerte Celular , Combinación de Medicamentos , Guanidinas/farmacología , Humanos , Inmunohistoquímica/métodos , Técnicas In Vitro , Interferón gamma/farmacología , Interleucina-1/farmacología , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/patología , Islotes Pancreáticos/fisiopatología , Factor de Necrosis Tumoral alfa/farmacología , Tirosina/metabolismo
12.
Cell Transplant ; 10(7): 651-9, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11714201

RESUMEN

The development of effective protocols for the low-temperature banking of pancreatic islets is an important step in islet transplantation for the treatment of type I diabetes mellitus. We have been exploring the use of islets from the newborn pig as an alternative source of tissue for transplantation. Current cryopreservation protocols are empirically derived, but may be optimized by modeling osmotic responses during the cryopreservation process. This study determined the osmotic and cryoprotectant permeability parameters of cells isolated from the pancreas of newborn pigs. Key parameters are: the osmotically inactive fraction of cell volume, hydraulic conductivity, the permeability coefficients of dimethyl sulfoxide (DMSO) and ethylene glycol (EG) at varying temperatures, and the activation energies of these transport processes. Newborn pig islets were dispersed into single cells and kinetic and equilibrium cell volumes were recorded during osmotic excursions using an electronic particle counter interfaced to a computer. Data were fitted to theoretical descriptions of the osmotic responses of cells, based on the Kedem-Katchalsky approach. The hydraulic conductivity (Lp) in the absence of cryoprotectant was calculated as 0.050 +/- 0.005, 0.071 +/- 0.006, and 0.300 +/- 0.016 microm/min/atm at 4 degrees C, 10 degrees C, and 22 degrees C, respectively (mean +/- SEM, n = 7, 6, or 9). These values give an activation energy value of 16.69 kcal/mol when put into an Arrhenius plot. The solute permeability (Ps) values for 1 M DMSO were 0.89 +/- 0.12, 1.86 +/- 0.28, and 5.33 +/- 0.26 microm/min at 4 degrees C, 10 degrees C, and 22 degrees C, respectively (n = 11, 8, or 10) giving an activation energy of 15.98 kcal/mol. The Lp values for cells exposed to 1 M DMSO were 0.071 +/- 0.006, 0.084 +/- 0.008, and 0.185 +/- 0.014 microm/min/atm at 4 degrees C, 10 degrees C, and 22 degrees C, respectively. The activation energy for these values was 8.95 kcal/mol. The Ps values for 2 M DMSO were 1.11 +/- 0.13, 1.74 +/- 0.19, and 7.68 +/- 0.12 microm/min for the same temperatures, with a calculated activation energy of 17.89 kcal/mol. The Lp values in the presence of 2 M DMSO were 0.070 +/- 0.006, 0.085 +/- 0.008, and 0.192 +/- 0.009 microm/min/atm at 4 degrees C, 10 degrees C, and 22 degrees C, respectively, with an activation energy of 9.40 kcal/mol. Solutions of 1 M EG gave Ps values of 1.01 +/- 0.13, 1.45 +/- 0.25, and 4.90 +/- 0.48 microm/min at the three test temperatures. The resulting activation energy was 14.60 kcal/mol. The corresponding Lp values were 0.071 +/- 0.007, 0.068 +/- 0.006, and 0.219 +/- 0.012 microm/min/atm with an activation energy of 10.96 kcal/mol. The solute permeabilities in the presence of 2 M EG for newborn pig islet cells were 1.03 +/- 0.15, 1.42 +/- 0.23, and 5.56 +/- 0.22 microm/min; the activation energy was 15.70. The Lp values for cells in the presence of 2 M EG were 0.068 +/- 0.008, 0.071 +/- 0.006, and 0.225 +/- 0.010 microm/min/atm; the activation energy for these values was 11.49 kcal/mol. These key cryobiological parameters permit the mathematical modeling of osmotic responses of intact islets during the cryopreservation process, which may lead to further improvements in the low temperature storage of islets from newborn pigs.


Asunto(s)
Criopreservación/métodos , Trasplante de Islotes Pancreáticos/métodos , Animales , Animales Recién Nacidos , Membrana Celular/metabolismo , Células Cultivadas , Crioprotectores/farmacocinética , Diabetes Mellitus Tipo 1/terapia , Dimetilsulfóxido/farmacocinética , Glicol de Etileno/farmacocinética , Islotes Pancreáticos/citología , Soluciones Preservantes de Órganos/farmacología , Presión Osmótica , Porcinos , Trasplante Heterólogo
13.
Transplantation ; 72(4): 565-70, 2001 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-11544413

RESUMEN

BACKGROUND: Islet isolation from the pancreatic tissue matrix remains highly variable. Recent evidence suggests that intrinsic human pancreatic proteases, including trypsin, may inhibit effective collagenase enzymatic activity during islet isolation, thereby impairing the isolation success. In this study we have hypothesized that serine protease inhibition applied during pancreatic digestion, could improve yield and/or functional viability of islets isolated from human pancreases. METHODS: Twelve organ donor pancreases with 12.9+/-0.6 hr cold storage (mean+/-SEM) were perfused via their ducts with Liberase-HI enzyme in the presence (n=6) or absence (n=6) of 0.4 mM Pefabloc. All were then gently dissociated and their purified islets separated with Ficoll density gradient centrifugation. RESULTS: Donor-related factors (age, gender, cold storage time, body mass index, and pancreas weight) did not differ significantly between the two experimental groups. Pefabloc supplementation did not affect the digestion time, islets remaining trapped in exocrine tissue, or final islet purity. Islet recovery was increased in the Pefabloc-treated group (mean+/-SEM yield 323.8+/-80.8 x 10(3) islet equivalents vs. 130.8+/-13.6 x 10(3) islet equivalents, P<0.05). Cellular composition, DNA and insulin content, and insulin secretory activity of the isolated islets was similar. CONCLUSIONS: Inhibition of intrinsic protease activity within pancreases after prolonged cold storage improves isolation of viable islets.


Asunto(s)
Criopreservación , Islotes Pancreáticos , Páncreas , Inhibidores de Serina Proteinasa/uso terapéutico , Sulfonas/uso terapéutico , Recolección de Tejidos y Órganos/métodos , Recolección de Tejidos y Órganos/normas , Adolescente , Adulto , Cadáver , Humanos , Persona de Mediana Edad , Factores de Tiempo
14.
Diabetes ; 50(4): 710-9, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11289033

RESUMEN

Islet transplantation offers the prospect of good glycemic control without major surgical risks. After our initial report of successful islet transplantation, we now provide further data on 12 type 1 diabetic patients with brittle diabetes or problems with hypoglycemia previous to 1 November 2000. Details of metabolic control, acute complications associated with islet transplantation, and long-term complications related to immunosuppression therapy and diabetes were noted. Insulin secretion, both acute and over 30 min, was determined after intravenous glucose tolerance tests (IVGTTs). The median follow-up was 10.2 months (CI 6.5-17.4), and the longest was 20 months. Glucose control was stable, with pretransplant fasting and meal tolerance-stimulated glucose levels of 12.5+/-1.9 and 20.0+/-2.7 mmol/l, respectively, but decreased significantly, with posttransplant levels of 6.3+/-0.3 and 7.5+/-0.6 mmol/l, respectively (P < 0.006). All patients have sustained insulin production, as evidenced by the most current baseline C-peptide levels 0.66+/-0.06 nmol/l, increasing to 1.29+/-0.25 nmol/l 90 min after the meal-tolerance test. The mean HbA1c level decreased from 8.3+/-0.5% to the current level of 5.8+/-0.1% (P < 0.001). Presently, four patients have normal glucose tolerance, five have impaired glucose tolerance, and three have post-islet transplant diabetes (two of whom need oral hypoglycemic agents and low-dose insulin (<10 U/day). Three patients had a temporary increase in their liver-function tests. One patient had a thrombosis of a peripheral branch of the right portal vein, and two of the early patients had bleeding from the hepatic needle puncture site; but these technical problems were resolved. Two patients had transient vitreous hemorrhages. The two patients with elevated creatinine levels pretransplant had a significant increase in serum creatinine in the long term, although the mean serum creatinine of the group was unchanged. The cholesterol increased in five patients, and lipid-lowering therapy was required for three patients. No patient has developed cytomegalovirus infection or disease, posttransplant lymphoproliferative disorder, malignancies, or serious infection to date. None of the patients have been sensitized to donor antigen. In 11 of the 12 patients, insulin independence was achieved after 9,000 islet equivalents (IEs) per kilogram were transplanted. The acute insulin response and the insulin area under the curve (AUC) after IVGTT were consistently maintained over time. The insulin AUC from the IVGTT correlated to the number of islets transplanted, but more closely correlated when the cold ischemia time was taken into consideration (r = 0.83, P < 0.001). Islet transplantation has successfully corrected labile type 1 diabetes and problems with hypoglycemia, and our results show persistent insulin secretion. After a minimum of 9,000 IEs per kilogram are provided, insulin independence is usually attained. An elevation of creatinine appears to be a contraindication to this immunosuppressive regimen. For the subjects who had labile type 1 diabetes that was difficult to control, the risk-to-benefit ratio is in favor of islet transplantation.


Asunto(s)
Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/cirugía , Insulina/metabolismo , Trasplante de Islotes Pancreáticos/métodos , Adulto , Glucemia/análisis , Péptido C/sangre , Ensayos Clínicos como Asunto , Femenino , Estudios de Seguimiento , Humanos , Secreción de Insulina , Masculino , Complicaciones Posoperatorias , Periodo Posoperatorio , Resultado del Tratamiento
16.
Ann N Y Acad Sci ; 944: 47-61, 2001 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11797694

RESUMEN

The mechanisms involved in islet neogenesis have remained largely unexplored due to lack of an appropriate model. Furthermore, with the recent advances in islet transplantation, the need for alternative islet tissue sources is greater than ever. Therefore, the authors have refined a neonatal porcine islet (NPI) maturation model that offers an ideal tool to gain insight into islet growth as well as an alternative source of transplantable tissue. Recent knowledge in islet growth has resulted in endocrine tissue being derived from human pancreatic precursor tissue in vitro. The potential for large scale production of endocrine tissue in vitro has been indicated, however, more investigation must be done on the various signals and pathways involved in pancreatic development to optimize this technique. The authors believe that their NPI in vitro maturation model provides an ideal tool to study islet growth and maturation. Transduction of the NPI to overexpress genes of interest (i.e., PDX-1) or exposure of the NPI to various culture conditions will allow us to determine the effects on islet maturation. An understanding of NPI development gained will not only allow us to mature this unlimited tissue source for optimal xenotransplantation, but also elude to how human pancreatic endocrine precursor cells may be used to solve the current islet tissue supply problem.


Asunto(s)
Islotes Pancreáticos/crecimiento & desarrollo , Trasplante Heterólogo , Secuencia de Aminoácidos , Animales , Animales Recién Nacidos , Diferenciación Celular , Proteínas de Homeodominio/genética , Islotes Pancreáticos/citología , Trasplante de Islotes Pancreáticos , Modelos Biológicos , Datos de Secuencia Molecular , Porcinos , Transactivadores/genética
17.
Science ; 290(5498): 1959-62, 2000 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-11110661

RESUMEN

Genetic engineering of non-beta cells to release insulin upon feeding could be a therapeutic modality for patients with diabetes. A tumor-derived K-cell line was induced to produce human insulin by providing the cells with the human insulin gene linked to the 5'-regulatory region of the gene encoding glucose-dependent insulinotropic polypeptide (GIP). Mice expressing this transgene produced human insulin specifically in gut K cells. This insulin protected the mice from developing diabetes and maintained glucose tolerance after destruction of the native insulin-producing beta cells.


Asunto(s)
Diabetes Mellitus Experimental/terapia , Células Enteroendocrinas/citología , Células Enteroendocrinas/metabolismo , Terapia Genética , Glucosa/metabolismo , Insulina/metabolismo , Animales , Glucemia/metabolismo , Línea Celular , Clonación Molecular , Diabetes Mellitus Experimental/metabolismo , Polipéptido Inhibidor Gástrico/biosíntesis , Polipéptido Inhibidor Gástrico/genética , Expresión Génica , Ingeniería Genética , Glucosa/administración & dosificación , Prueba de Tolerancia a la Glucosa , Humanos , Insulina/biosíntesis , Insulina/genética , Ratones , Ratones Transgénicos , Proinsulina/genética , Regiones Promotoras Genéticas , Precursores de Proteínas/genética , Células Madre/citología , Células Madre/metabolismo , Estreptozocina , Transfección , Transgenes , Células Tumorales Cultivadas
18.
Diabetes ; 49(11): 1810-8, 2000 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11078447

RESUMEN

Testicular Sertoli cells protect pancreatic islet grafts from allo- and autoimmune destruction; however, the mechanism(s) of protection is unclear. The aim of this study was to determine whether Fas ligand (FasL) and/or transforming growth factor (TGF)-beta, immunoregulatory proteins produced by Sertoli cells, might mediate the protective effects of these cells against autoimmune destruction of islet beta-cells. Sertoli cells were purified from testes of NOD mice and implanted under the right renal capsule of diabetic NOD mice, whereas NOD islets were implanted under the left renal capsule. Of the mice that received islet and Sertoli cells grafts, 64% (9 of 14) remained normoglycemic at 60 days posttransplantation compared with 0% (0 of 6) of the mice that received islet grafts alone. Immunohistochemical examination of Sertoli cell grafts in normoglycemic mice revealed that TGF-beta1 expression by Sertoli cells remained high, whereas FasL expression by Sertoli cells decreased progressively posttransplantation. Also, plasma levels of TGF-beta1 were significantly elevated in mice that received Sertoli cells and islet grafts, and anti-TGF-beta1 antibody administration completely abrogated the protective effect of Sertoli cells on islet graft survival, whereas anti-FasL antibody did not. Islet graft destruction in anti-TGF-beta1-treated mice was associated with increases in interferon (IFN)-gamma-producing cells and decreases in interleukin (IL)-4-producing cells in the islet grafts. We conclude that 1) Sertoli cell production of TGF-beta1, not FasL, protects islet beta-cells from autoimmune destruction and 2) TGF-beta1 diverts islet-infiltrating cells from a beta-cell-destructive (IFN-gamma+) phenotype to a nondestructive (IL-4+) phenotype.


Asunto(s)
Autoinmunidad , Diabetes Mellitus Tipo 1/inmunología , Islotes Pancreáticos/inmunología , Células de Sertoli/fisiología , Factor de Crecimiento Transformador beta/farmacología , Animales , Anticuerpos Monoclonales/farmacología , Proteína Ligando Fas , Supervivencia de Injerto , Inmunohistoquímica , Trasplante de Islotes Pancreáticos , Riñón , Masculino , Glicoproteínas de Membrana/inmunología , Glicoproteínas de Membrana/fisiología , Ratones , Ratones Endogámicos NOD , Células de Sertoli/trasplante , Testículo/citología , Factor de Crecimiento Transformador beta/sangre , Factor de Crecimiento Transformador beta/inmunología
19.
Transplantation ; 70(6): 976-9, 2000 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-11014652

RESUMEN

BACKGROUND: Insulin has been implicated in the pathogenesis of type 1 diabetes and oral administration of insulin has been shown to delay the onset of diabetes in NOD mice. In this study we determined whether a single footpad injection of insulin will protect syngeneic islet grafts from autoimmune destruction when placed under the kidney capsule of diabetic NOD mice. METHODS: Five hundred islets were transplanted under the kidney capsule of diabetic female NOD mice in conjunction with a single footpad injection of either pork insulin in saline or mixed with incomplete Freund's adjuvant (IFA). Control groups received either IFA or saline alone. RESULTS: Seven of 11 animals (63.6%) given insulin in IFA exhibit long-term graft survival (>75 days; mean +/- SEM >85.4+/-16.1) whereas only 3 of 12 animals (25.0%) in the IFA group had graft survival longer than 75 days (mean +/- SEM >41.9+/-12.8 days). In contrast, none of the animals that received insulin in saline (17.3+/-2.5 days) and saline only (16.1+2.0 days) exhibit prolonged graft survival. CONCLUSION: These results suggest that a single footpad injection of insulin can protect the islet graft from immune attack in NOD mice.


Asunto(s)
Diabetes Mellitus/prevención & control , Insulina/administración & dosificación , Animales , Relación Dosis-Respuesta a Droga , Supervivencia de Injerto/efectos de los fármacos , Trasplante de Islotes Pancreáticos/inmunología , Trasplante de Islotes Pancreáticos/fisiología , Ratones , Ratones Endogámicos NOD , Prevención Secundaria , Factores de Tiempo , Trasplante Isogénico
20.
N Engl J Med ; 343(4): 230-8, 2000 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-10911004

RESUMEN

BACKGROUND: Registry data on patients with type 1 diabetes mellitus who undergo pancreatic islet transplantation indicate that only 8 percent are free of the need for insulin therapy at one year. METHODS: Seven consecutive patients with type 1 diabetes and a history of severe hypoglycemia and metabolic instability underwent islet transplantation in conjunction with a glucocorticoid-free immunosuppressive regimen consisting of sirolimus, tacrolimus, and daclizumab. Islets were isolated by ductal perfusion with cold, purified collagenase, digested and purified in xenoprotein-free medium, and transplanted immediately by means of a percutaneous transhepatic portal embolization. RESULTS: All seven patients quickly attained sustained insulin independence after transplantation of a mean (+/-SD) islet mass of 11,547+/-1604 islet equivalents per kilogram of body weight (median follow-up, 11.9 months; range, 4.4 to 14.9). All recipients required islets from two donor pancreases, and one required a third transplant from two donors to achieve sustained insulin independence. The mean glycosylated hemoglobin values were normal after transplantation in all recipients. The mean amplitude of glycemic excursions (a measure of fluctuations in blood glucose concentrations) was significantly decreased after the attainment of insulin independence (from 198+/-32 mg per deciliter [11.1+/-1.8 mmol per liter] before transplantation to 119+/-37 mg per deciliter [6.7+/-2.1 mmol per liter] after the first transplantation and 51+/-30 mg per deciliter [2.8+/-1.7 mmol per liter] after the attainment of insulin independence; P<0.001). There were no further episodes of hypoglycemic coma. Complications were minor, and there were no significant increases in lipid concentrations during follow-up. CONCLUSIONS: Our observations in patients with type 1 diabetes indicate that islet transplantation can result in insulin independence with excellent metabolic control when glucocorticoid-free immunosuppression is combined with the infusion of an adequate islet mass.


Asunto(s)
Diabetes Mellitus Tipo 1/cirugía , Inmunosupresores/uso terapéutico , Trasplante de Islotes Pancreáticos , Adulto , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales Humanizados , Glucemia/metabolismo , Péptido C/sangre , Daclizumab , Diabetes Mellitus Tipo 1/sangre , Quimioterapia Combinada , Prueba de Tolerancia a la Glucosa , Hemoglobina Glucada/metabolismo , Humanos , Inmunoglobulina G/uso terapéutico , Insulina/administración & dosificación , Trasplante de Islotes Pancreáticos/inmunología , Trasplante de Islotes Pancreáticos/métodos , Persona de Mediana Edad , Sirolimus/uso terapéutico , Tacrolimus/uso terapéutico , Acondicionamiento Pretrasplante
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA