Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Transplant Direct ; 8(10): e1367, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36204182

RESUMEN

Islet and ß-cell function is intrinsic to glucose homeostasis. Pancreatectomy and islet autotransplantation (PIAT) for chronic pancreatitis (CP) treatment is a useful model for assessing islet function in the absence of immune-suppression and to perform extensive presurgical metabolic evaluations not possible from deceased donors. We recently showed that in CP-PIAT patients, preoperative islet identity loss presented with postoperative glycemic loss. Here, we examine presurgical islet function using Homeostatic Model Assessment-Beta Cell Function (%) (HOMA-ß) and glycemic variables and compared them with postsurgical insulin independence and their predicted alignment with Secretory Unit of Islet Transplant Objects (SUITO) and beta cell score after transplantation (BETA-2) scores. Methods: Seven CP-PIAT patients were assessed for ß-cell function metrics, including pretransplant and 6-mo posttransplant HOMA-ß using insulin and C-peptide and evaluations of proposed insulin independence by SUITO and BETA-2 graft function equations. These were compared with oral glucose tolerance tests and pancreas histological samples taken at the time of transplant, examined for ß-cell maturity markers. Results: Pre-PIAT, HOMA-ß (60%-100%) associated with post-PIAT insulin independence. This association was only moderately supported by post-PIAT SUITO threshold scores (≥26) but robustly by BETA-2 scores (≥16.2). Appropriate posttransplant oral glucose tolerance test curves were found in those patients with normal pretransplant HOMA-ß values. Preoperative low serological ß-cell function was displayed by concurrent evidence of ß-cell identity alterations including colocalization of insulin and glucagon, loss of urocortin-3, and increased intra-islet vimentin in patients who were insulin-dependent post-PIAT. Conclusions: These data encourage HOMA-ß assessment before PIAT for estimating posttransplant insulin independence.

2.
Biomaterials ; 257: 120232, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32768727

RESUMEN

Cell encapsulation is an attractive transplantation strategy to treat endocrine disorders. Transplanted cells offer a dynamic and stimulus-responsive system that secretes therapeutics based on patient need. Despite significant advancements, a challenge in allogeneic cell encapsulation is maintaining sufficient oxygen and nutrient exchange, while providing protection from the host immune system. To this end, we developed a subcutaneously implantable dual-reservoir encapsulation system integrating in situ prevascularization and local immunosuppressant delivery, termed NICHE. NICHE structure is 3D-printed in biocompatible polyamide 2200 and comprises of independent cell and drug reservoirs separated by a nanoporous membrane for sustained local release of immunosuppressant. Here we present the development and characterization of NICHE, as well as efficacy validation for allogeneic cell transplantation in an immunocompetent rat model. We established biocompatibility and mechanical stability of NICHE. Further, NICHE vascularization was achieved with the aid of mesenchymal stem cells. Our study demonstrated sustained local elution of immunosuppressant (CTLA4Ig) into the cell reservoir protected transcutaneously-transplanted allogeneic Leydig cells from host immune destruction during a 31-day study, and reduced systemic drug exposure by 12-fold. In summary, NICHE is the first encapsulation platform achieving both in situ vascularization and immunosuppressant delivery, presenting a viable strategy for allogeneic cell transplantation.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Preparaciones Farmacéuticas , Animales , Encapsulación Celular , Inmunosupresores , Masculino , Ratas , Trasplante Homólogo
3.
Endocrinol Diabetes Metab ; 3(1): e00104, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31922031

RESUMEN

BACKGROUND: We explored the association of C-peptide (marker of secreted insulin), proinsulin and proinsulin /C-peptide ratio (PI/C) (markers of beta-cell endoplasmic reticulum [ER] stress) with undercarboxylated (uOC) and carboxylated osteocalcin (cOC) and their ratio (uOC/cOC) in children with recently diagnosed type 1 (T1D) or type 2 diabetes (T2D), and the correlation of these variables with partial remission (PR) in children with T1D. METHODS: Demographic and clinical data of children with new-onset diabetes (n = 68; median age = 12.2 years; 33.8% non-Hispanic White, 45.6% Hispanic/Latino, 16.2% African American and 4.4% other) were collected at diagnosis and during the first (V1), second (V2) and third clinical visits at 9.0, 32.0 and 175.7 weeks, respectively. Serum proinsulin, C-peptide, uOC and cOC values were measured 7.0 weeks after diagnosis. PR was defined as insulin dose-adjusted HbA1c (IDAA1c) ≤9. RESULTS: In children with new-onset T1D with DKA (33.3%) or T2D (29.4%), Spearman's correlation coefficient revealed a positive association between the C-peptide levels and both uOC and uOC/cOC ratio. In T1D (n = 48), both higher serum C-peptide levels and low PI:C ratio were associated with higher BMI percentile (ß = 0.02, P = .001; ß = -0.01, P = .02, respectively) and older age at diagnosis (ß = 0.13, P = .001; ß = -0.12, P = .001, respectively). Furthermore, in children with T1D, C-peptide levels at V1 correlated with IDAA1c ≤ 9 at V1 (P = .04). CONCLUSION: C-peptide levels are associated with a higher uOC and uOC/cOC ratio in paediatric diabetes. In new-onset T1D children, older age and higher BMI were associated with lower beta-cell stress and higher preserved function, which was predictive of PR on follow-up.

4.
Am J Transplant ; 19(5): 1568-1576, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30372580

RESUMEN

Beta-cell dedifferentiation as shown by cellular colocalization of insulin with glucagon and/or vimentin, and decreased expression of MAFA and/or urocortin3 has been suggested to contribute to metabolic decompensation in type 2 diabetes, and was recently described postimplantation in islet allotransplant patients. Dysglycaemia and diabetes mellitus are often encountered preoperatively in patients undergoing pancreatectomy and islet autotransplantation (PIAT). In this series of case reports, we document variation in islet phenotypic identity in three patients with chronic pancreatitis (CP) without diabetes or significant insulin resistance who subsequently underwent PIAT. Pancreas histology was examined using colocalization of endocrine hormones, mesenchymal and pan-endocrine markers in islets, and the relative expression of MAFA and urocortin3 in insulin-expressing cells as compared to that of nondiabetic and type 2 diabetic donors. We present results of pre- and posttransplant clinical metabolic testing. Varying degrees of islet-cell dedifferentiation are identified in nondiabetic patients with CP at the time of PIAT, and may need further investigation.


Asunto(s)
Células Endocrinas/patología , Trasplante de Islotes Pancreáticos/métodos , Islotes Pancreáticos/citología , Pancreatitis Crónica/terapia , Adulto , Biomarcadores/metabolismo , Células Endocrinas/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Pronóstico , Trasplante Autólogo , Adulto Joven
5.
J Exp Med ; 214(9): 2591-2610, 2017 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-28765400

RESUMEN

Although a large proportion of patients with type 2 diabetes (T2D) accumulate misfolded aggregates composed of the islet amyloid polypeptide (IAPP), its role in the disease is unknown. Here, we show that pancreatic IAPP aggregates can promote the misfolding and aggregation of endogenous IAPP in islet cultures obtained from transgenic mouse or healthy human pancreas. Islet homogenates immunodepleted with anti-IAPP-specific antibodies were not able to induce IAPP aggregation. Importantly, intraperitoneal inoculation of pancreatic homogenates containing IAPP aggregates into transgenic mice expressing human IAPP dramatically accelerates IAPP amyloid deposition, which was accompanied by clinical abnormalities typical of T2D, including hyperglycemia, impaired glucose tolerance, and a substantial reduction on ß cell number and mass. Finally, induction of IAPP deposition and diabetic abnormalities were also induced in vivo by administration of IAPP aggregates prepared in vitro using pure, synthetic IAPP. Our findings suggest that some of the pathologic and clinical alterations of T2D might be transmissible through a similar mechanism by which prions propagate in prion diseases.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/metabolismo , Animales , Diabetes Mellitus Tipo 2/etiología , Diabetes Mellitus Tipo 2/patología , Femenino , Humanos , Islotes Pancreáticos/patología , Masculino , Ratones , Ratones Transgénicos , Priones/metabolismo , Agregado de Proteínas , Deficiencias en la Proteostasis/metabolismo
6.
Biotechnol J ; 12(9)2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28734022

RESUMEN

Transplantation of pancreatic islets or stem cell derived insulin secreting cells is an attractive treatment strategy for diabetes. However, islet transplantation is associated with several challenges including function-loss associated with dispersion and limited vascularization as well as the need for continuous immunosuppression. To overcome these limitations, here we present a novel 3D printed and functionalized encapsulation system for subcutaneous engraftment of islets or islet like cells. The devices were 3D printed with polylactic acid and the surfaces treated and patterned to increase the hydrophilicity, cell attachment, and proliferation. Surface treated encapsulation systems were implanted with growth factor enriched platelet gel, which helped to create a vascularized environment before loading human islets. The device protected the encapsulated islets from acute hypoxia and kept them functional. The adaptability of the encapsulation system was demonstrated by refilling some of the experimental groups transcutaneously with additional islets.


Asunto(s)
Trasplante de Islotes Pancreáticos/métodos , Islotes Pancreáticos , Impresión Tridimensional , Animales , Bioimpresión , Diabetes Mellitus , Femenino , Histocitoquímica , Humanos , Insulina/análisis , Insulina/metabolismo , Islotes Pancreáticos/irrigación sanguínea , Islotes Pancreáticos/citología , Ratones Desnudos , Ingeniería de Tejidos
7.
Pediatr Diabetes ; 18(8): 869-873, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28093839

RESUMEN

BACKGROUND: Osteocalcin (OC), a hormone secreted by osteoblasts, improves beta-cell function in vitro and in vivo. We aimed to understand the relationship between OC and hemoglobin A1c (HbA1c) in pediatric diabetes. METHODS: Children (n = 70; mean [SD] age = 11.8 years [3.1]; 34.3% non-Hispanic white, 46.3% Hispanic, 14.9% African-American, 4.5% other) newly diagnosed with diabetes (69.1% type 1 diabetes [T1D], 30.9% type 2 diabetes [T2D]) were studied. We collected clinical data at diagnosis and first clinical visit (V1) 9 weeks later (interquartile range [IQR] = 7.9-12.0). Serum undercarboxylated OC (uOC) and carboxylated OC (cOC) were measured 7.0 weeks (IQR 4.3-8.9) after diagnosis. RESULTS: Mean [SD] uOC was 20.3 (19.6) ng/mL, cOC 29.7 [13.7] ng/mL and u/cOC 0.68 [0.81]. uOC, cOC, or u/cOC were not different by gender, race/ethnicity, age, diabetes type, BMI percentile, or random C-peptide, glucose or HbA1c at diagnosis. However, among 61 children with V1 within 4 months of diagnosis, uOC was higher in those with V1 HbA1c < 7.5% (HbA1c < 58 mmol/mol) (uOC=33.1 [22.0]) compared with children with HbA1c ≥ 7.5% (uOC=17.4 [2.3], P = .0004). The difference was larger among patients with T2D (34.6 and 4.7 ng/mL, respectively, P = .0001) than T1D (32.2 and 19.3, P = .0169), and in males (36.1 and 17.4, P = .018) than females (27.6 and 17.3, P = .072). Analysis for u/cOC were similar while there were no differences in cOC. uOC was inversely correlated with HbA1c at V1 (Spearman's rho = -0.29, P = .02). CONCLUSION: Our findings suggest that serum uOC is inversely related to HbA1c shortly after diagnosis of pediatric diabetes. This potentially modifiable factor of glucose metabolism warrants further studies.


Asunto(s)
Diabetes Mellitus Tipo 1/sangre , Osteocalcina/sangre , Péptido C/sangre , Niño , Femenino , Hemoglobina Glucada/metabolismo , Humanos , Masculino , Estudios Prospectivos
8.
J Tissue Eng ; 7: 2041731416638198, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27152147

RESUMEN

Diabetes is one of the most prevalent, costly, and debilitating diseases in the world. Pancreas and islet transplants have shown success in re-establishing glucose control and reversing diabetic complications. However, both are limited by donor availability, need for continuous immunosuppression, loss of transplanted tissue due to dispersion, and lack of vascularization. To overcome the limitations of poor islet availability, here, we investigate the potential of bone marrow-derived mesenchymal stem cells differentiated into islet-like insulin-producing aggregates. Islet-like insulin-producing aggregates, characterized by gene expression, are shown to be similar to pancreatic islets and display positive immunostaining for insulin and glucagon. To address the limits of current encapsulation systems, we developed a novel three-dimensional printed, scalable, and potentially refillable polymeric construct (nanogland) to support islet-like insulin-producing aggregates' survival and function in the host body. In vitro studies showed that encapsulated islet-like insulin-producing aggregates maintained viability and function, producing steady levels of insulin for at least 4 weeks. Nanogland-islet-like insulin-producing aggregate technology here investigated as a proof of concept holds potential as an effective and innovative approach for diabetes cell therapy.

9.
Lab Chip ; 13(18): 3675-88, 2013 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-23884326

RESUMEN

Despite the clinical success of pancreatic islet transplantation, graft function is frequently lost over time due to islet dispersion, lack of neovascularization, and loss of physiological architecture. To address these problems, islet encapsulation strategies including scaffolds and devices have been developed, which produced encouraging results in preclinical models. However, islet loss from such architectures could represent a significant limitation to clinical use. Here, we developed and characterized a novel islet encapsulation silicon device, the NanoGland, to overcome islet loss, while providing a physiological-like environment for long-term islet viability and revascularization. NanoGlands, microfabricated with a channel size ranging from 3.6 nm to 60 µm, were mathematically modeled to predict the kinetics of the response of encapsulated islets to glucose stimuli, based on different channel sizes, and to rationally select membranes for further testing. The model was validated in vitro using static and perifusion testing, during which insulin secretion and functionality were demonstrated for over 30-days. In vitro testing also showed 70-83% enhanced islet retention as compared to porous scaffolds, here simulated through a 200 µm channel membrane. Finally, evidence of in vivo viability of human islets subcutaneously transplanted within NanoGlands was shown in mice for over 120 days. In this context, mouse endothelial cell infiltration suggesting neovascularization from the host were identified in the retrieved grafts. The NanoGland represents a novel, promising approach for the autotransplantation of human islets.


Asunto(s)
Trasplante de Islotes Pancreáticos , Islotes Pancreáticos/citología , Nanoestructuras/química , Nanotecnología/instrumentación , Animales , Movimiento Celular , Difusión , Células Endoteliales , Glucosa/farmacología , Humanos , Insulina/metabolismo , Secreción de Insulina , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Ratones , Modelos Teóricos , Neovascularización Fisiológica , Silicio/química , Trasplante Heterólogo
10.
Cell Transplant ; 21(12): 2805-14, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22863057

RESUMEN

Culture of human islets before clinical transplantation or distribution for research purposes is standard practice. At the time the Edmonton protocol was introduced, clinical islet manufacturing did not include culture, and human serum albumin (HSA), instead of fetal bovine serum (FBS), was used during other steps of the process to avoid the introduction of xenogeneic material. When culture was subsequently introduced, HSA was also used for medium supplementation instead of FBS, which was typically used for research islet culture. The use of HSA as culture supplement was not evaluated before this implementation. We performed a retrospective analysis of 103 high-purity islet preparations (76 research preparations, all with FBS culture supplementation, and 27 clinical preparations, all with HSA supplementation) for oxygen consumption rate per DNA content (OCR/DNA; a measure of viability) and diabetes reversal rate in diabetic nude mice (a measure of potency). After 2-day culture, research preparations exhibited an average OCR/DNA 51% higher (p < 0.001) and an average diabetes reversal rate 54% higher (p < 0.05) than clinical preparations, despite 87% of the research islet preparations having been derived from research-grade pancreata that are considered of lower quality. In a prospective paired study on islets from eight research preparations, OCR/DNA was, on average, 27% higher with FBS supplementation than that with HSA supplementation (p < 0.05). We conclude that the quality of clinical islet preparations can be improved when culture is performed in media supplemented with serum instead of albumin.


Asunto(s)
Medios de Cultivo , Islotes Pancreáticos/efectos de los fármacos , Albúmina Sérica/farmacología , Suero , Animales , Bovinos , Técnicas de Cultivo de Célula , Células Cultivadas , Medios de Cultivo/farmacología , Diabetes Mellitus Experimental/mortalidad , Diabetes Mellitus Experimental/cirugía , Humanos , Islotes Pancreáticos/citología , Trasplante de Islotes Pancreáticos , Estimación de Kaplan-Meier , Ratones , Ratones Desnudos , Consumo de Oxígeno , Estudios Retrospectivos , Trasplante Heterólogo
11.
Cell Transplant ; 17(7): 785-92, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19044205

RESUMEN

The ability to isolate high-yield pure and viable islets from human cadaver pancreas donors is dependent on donor factor as well as isolation factors. The aim of this study was to examine factors influencing islets recovery and in vivo function with an emphasis on donor and isolation methods as well as to compare the effectiveness of Liberase, widely used in clinical islet isolation, with Serva for the isolation of pure functional islets. The results of 123 islet isolations using Liberase for digestion were compared with those of 113 isolations with Serva. Islet equivalents per gram of tissue were similar between Liberase and Serva (3620 +/- 1858 vs. 4132 +/- 2104, p < 0.2) as well as the percent purity (75 +/- 16 vs. 74 +/- 15, p < 0.9). In vivo function of islets from 71 isolations (Liberase = 45, Serva = 26) were further tested by transplantation into NOD-SCID mice following short-term culture (< 6 days, n = 71). Our data show that both Liberase- and Serva-isolated islets showed similar function results following short-term culture. These data demonstrate that there is no difference in islet yield, purity, and function between the two enzymes. However, when these 71 isolations were analyzed for in vivo function with emphasis on donor factors, cold ischemia time (12.0 +/- 5.3 vs. 15.0 +/- 5.7, p < 0.04), islet integrity (1.6 +/- 0.7 vs. 1.3 +/- 0.5, p < 0.05), and female gender were the only factors that correlated with in vivo function. We also compared the mechanical-shaking method for islets isolation with hand-shaking methods. Our results show that although there is no different in islet yield, purity, and integrity between different enzymes using the same method, hand-shaking method yields more islets with better integrity than mechanical-shaking method.


Asunto(s)
Separación Celular/métodos , Islotes Pancreáticos , Donantes de Tejidos , Adolescente , Adulto , Anciano , Animales , Péptido C/metabolismo , Cadáver , Supervivencia Celular , Colagenasas/metabolismo , Femenino , Humanos , Insulina/metabolismo , Islotes Pancreáticos/citología , Islotes Pancreáticos/metabolismo , Ratones , Persona de Mediana Edad , Termolisina/metabolismo , Adulto Joven
12.
Cell Transplant ; 16(8): 775-85, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-18087998

RESUMEN

Transforming growth factor-beta1 (TGF-beta1) is a pleotropic cytokine that promotes angiogenesis and extracellular matrix protein synthesis in addition to its immunosuppressive effects. The purpose of this study is to identify optimal conditions for in vivo expression of TGF-beta1 by human islets to exploit the possible beneficial effects and minimize undesirable side effects. We transduced human islets with adenoviral vectors encoding the active form of Ad-TGF-beta1 or Ad-LacZ to test the effects of TGF-beta1 gene expression on islet in vivo function following their transplantation into a NOD-SCID mouse model. Islets were transduced with multiplicity of infection (MOI) of 20, 10, 5, and 2.5 per islet cell. At a MOI ranging from 2.5 to 20, expression of TGF-beta1 in islet supernatant persisted for 1-2 months and ranged from 153 +/- 5 to 2574 +/- 1299 pg/ml, respectively. Transduction with the lowest MOI (2.5) did not compromise the in vivo production of human C-peptide. We conclude that TGF-beta1 expression in transplanted islets does not compromise viability and that adenoviral transduction with the TGF-beta1 gene has a dose-dependent effect, with larger MOIs being deleterious. The data also indicate that in vitro culture system and the in vivo NOD-SCID model could be used successfully to evaluate the nonimmune effects of gene transduction.


Asunto(s)
Apoptosis/fisiología , Islotes Pancreáticos/metabolismo , Factor de Crecimiento Transformador beta/fisiología , Adenoviridae/genética , Animales , Apoptosis/genética , Péptido C/metabolismo , Supervivencia Celular/genética , Supervivencia Celular/fisiología , Fragmentación del ADN , Ensayo de Inmunoadsorción Enzimática , Vectores Genéticos/genética , Humanos , Etiquetado Corte-Fin in Situ , Islotes Pancreáticos/citología , Trasplante de Islotes Pancreáticos/métodos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Factores de Tiempo , Transfección , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Trasplante Heterólogo
13.
Prog Transplant ; 16(4): 350-4, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17183943

RESUMEN

BACKGROUND: A major problem in the islet field is the high selectivity exercised in accepting cadaveric pancreas for islet isolation. This practice is based on experience that indicates that islet yield and posttransplant function are related to donor demographics and injury mechanisms. OBJECTIVE: To examine factors influencing islets recovery and in vivo function with emphasis on donor-related factors. METHODS: Islets were isolated from 99 human donor pancreata, and islet yield was reported as islet equivalent per gram pancreatic tissue. Donor, procurement, and isolation factors were collected for each isolation and correlation statistics were performed between these variables and islet yield. RESULTS: Results indicated a differential effect of enzyme mixes on yield with Collagenase P digestion most suitable for increased ischemic time (R2 = 0.1; P < .08), Liberase with small donor pancreas size and elevated preprocurement glucose (R2 = 0.15; P < .02), and Serva with female donors (R2 = 0.17; P < .06). Islets from 29 isolations were further tested by transplantation under the kidney capsule of immune-deficient NOD-SCID mice. Although all 29 preparations had acceptable in vitro perfusion parameters indicating viability, only 19 functioned in vivo with serum levels of insulin >5 U/mL and C peptide >1.5 ng/mL. No significant differences in donor, procurement, and isolation factors were evident between the islet preparations that functioned in vivo and those that were nonfunctional. CONCLUSIONS: These data demonstrate that although yield is affected by a variety of donor factors and enzyme mixes, these factors do not affect islet in vivo function.


Asunto(s)
Trasplante de Islotes Pancreáticos , Islotes Pancreáticos/citología , Donantes de Tejidos , Recolección de Tejidos y Órganos/métodos , Adulto , Animales , Separación Celular , Supervivencia Celular , Colagenasas , Femenino , Humanos , Islotes Pancreáticos/crecimiento & desarrollo , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Análisis Multivariante , Análisis de Regresión , Termolisina
14.
Transplantation ; 80(8): 1003-11, 2005 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-16278578

RESUMEN

BACKGROUND: An in vitro method to assess human islets could prevent transplantation of nonviable islets and facilitate the optimization of islet preparation. We hypothesize that glucose-stimulated cytochrome c reduction and oxygen consumption by human islets can be used as predictors of transplant success. METHODS: Isolated human islets were obtained from research-grade pancreata. Using a previously developed islet flow culture system, the response of cytochrome c reduction and oxygen consumption to glucose was compared to the ability of islets transplanted into nondiabetic NOD-SCID mice to secrete C-peptide in response to a glucose tolerance test conducted 7 days following transplant (n=10). RESULTS: In vitro responses by human islets were qualitatively similar to those seen in rat islets: glucose increased both oxygen consumption and cytochrome c reduction. However, the responses were smaller in magnitude and quite variable. Scatter plots of C-peptide and quantiles for ln(C-peptide) indicated that 12 ng/ml could be used as threshold of transplant success with which to evaluate the diagnostic potential of cytochrome c and oxygen consumption. Data was analyzed by generating receiver operating curves and the area under the curve was 0.889 (95% CI: 0.645-1.000) and 0.738 (95% CI: 0.413-1.000) for cytochrome c reduction and oxygen consumption respectively (1 indicates absolute predictive capability and 0.5 indicates no predictive capability). CONCLUSIONS: The detection of glucose-stimulated cytochrome c reduction and oxygen consumption may have utility as criteria for the assessment of human islet quality.


Asunto(s)
Citocromos c/metabolismo , Glucosa/farmacología , Trasplante de Islotes Pancreáticos , Islotes Pancreáticos/efectos de los fármacos , Consumo de Oxígeno/efectos de los fármacos , Animales , Células Cultivadas , Citocromos c/análisis , Humanos , Insulina/metabolismo , Secreción de Insulina , Islotes Pancreáticos/enzimología , Islotes Pancreáticos/metabolismo , Masculino , Ratones , Ratones SCID , Oxidación-Reducción , Ratas , Ratas Endogámicas F344
16.
Transplantation ; 77(8): 1147-54, 2004 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-15114076

RESUMEN

BACKGROUND: Culturing human islets in Memphis serum-free media (M-SFM) is associated with excellent postculture recovery, in vitro function, and in vivo survival. The authors investigate the possibility of preserving islet function for extended periods (6 months) in culture and describe the in vitro and in vivo functional outcomes associated with these extended culture times. METHODS: Human islets isolated from three cadaveric donor organs were cultured in M-SFM for 1, 3, or 6 months before transplantation under the kidney capsule of nonobese diabetic (NOD)-severe combined immunodeficiency (SCID) mice. In vitro function was measured by static incubation at the time of transplantation. In vivo function was assessed by measuring human insulin and C-peptide production, and by the ability of 6-month cultured islets to cure streptozotocin-induced diabetes in this mouse model. RESULTS: Islet recovery ratios after 1 month in culture ranged from 85% to 88% and declined to 28% to 53% after 6 months of culture (P <0.01). Insulin stimulation indices did not differ among the fresh or the 6-month cultured preparations. All preparations cultured for 1 to 3 months functioned in the NOD-SCID mice. After 6 months of culture, two of the three preparations demonstrated in vivo function and were able to cure streptozotocin-induced diabetes. CONCLUSIONS: These data demonstrate that human islets can be cultured in M-SFM for extended periods and still retain in vitro and in vivo function and the ability to cure experimental diabetes. The ability to maintain islets in culture for prolonged periods is an important step toward the development of islet tissue repositories and distribution centers.


Asunto(s)
Islotes Pancreáticos/fisiología , Conservación de Tejido/métodos , Animales , Péptido C/biosíntesis , Medio de Cultivo Libre de Suero , Técnicas de Cultivo , Diabetes Mellitus Experimental/fisiopatología , Diabetes Mellitus Experimental/terapia , Supervivencia de Injerto , Humanos , Insulina/biosíntesis , Trasplante de Islotes Pancreáticos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Factores de Tiempo , Trasplante Heterólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...