Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
1.
Cell Transplant ; 31: 9636897221096160, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35583214

RESUMEN

Patients fulfilling criteria for euthanasia can choose to donate their organs after circulatory death [donors after euthanasia (DCD V)]. This study assesses the outcome of islet cell isolation from DCD V pancreases. A procedure for DCD V procurement provided 13 pancreases preserved in Institut Georges Lopez-1 preservation solution and following acirculatory warm ischemia time under 10 minutes. Islet cell isolation outcomes are compared with those from reference donors after brain death (DBD, n = 234) and a cohort of donors after controlled circulatory death (DCD III, n = 29) procured under the same conditions. Islet cell isolation from DCD V organs resulted in better in vitro outcome than for selected DCD III or reference DBD organs. A 50% higher average beta cell number before and after culture and a higher average beta cell purity (35% vs 24% and 25%) was observed, which led to more frequent selection for our clinical protocol (77% of isolates vs 50%). The functional capacity of a DCD V islet cell preparation was illustrated by its in vivo effect following intraportal transplantation in a type 1 diabetes patient: injection of 2 million beta cells/kg body weight (1,900 IEQ/kg body weight) at 39% insulin purity resulted in an implant with functional beta cell mass that represented 30% of that in non-diabetic controls. In conclusion, this study describes procurement and preservation conditions for donor organs after euthanasia, which allow preparation of cultured islet cells, that more frequently meet criteria for clinical use than those from DBD or DCD III organs.


Asunto(s)
Células Secretoras de Insulina , Donantes de Tejidos , Peso Corporal , Muerte Encefálica , Eutanasia , Humanos , Células Secretoras de Insulina/trasplante , Páncreas
2.
Am J Transplant ; 22(3): 927-936, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34735732

RESUMEN

Intraportal (IP) islet cell transplants can restore metabolic control in type 1 diabetes patients, but limitations raise the need for establishing a functional beta cell mass (FBM) in a confined extrahepatic site. This study reports on function and composition of omental (OM) implants after placement of islet cell grafts with similar beta cell mass as in our IP-protocol (2-5.106 beta cells/kg body weight) on a scaffold. Four of seven C-peptide-negative recipients achieved low beta cell function (hyperglycemic clamp [HGC] 2-8 percent of controls) until laparoscopy, 2-6 months later, for OM-biopsy and concomitant IP-transplant with similar beta cell dose. This IP-transplant increased HGC-values to 15-40 percent. OM-biopsies reflected the composition of initial grafts, exhibiting varying proportions of endocrine-cell-enriched clusters with more beta than alpha cells and leucocyte pole, non-endocrine cytokeratin-positive clusters surrounded by leucocytes, and scaffold remnants with foreign body reaction. OM-implants on a polyglactin-thrombin-fibrinogen-scaffold presented larger endocrine clusters with infiltrating endothelial cells and corresponded to the higher HGC-values. No activation of cellular immunity to GAD/IA2 was measured post-OM-transplant. Establishment of a metabolically adequate FBM in omentum may require a higher beta cell number in grafts but also elimination of their immunogenic non-endocrine components as well as local conditioning that favors endocrine cell engraftment and function.


Asunto(s)
Diabetes Mellitus Tipo 1 , Trasplante de Islotes Pancreáticos , Islotes Pancreáticos , Diabetes Mellitus Tipo 1/cirugía , Células Endoteliales , Humanos , Trasplante de Islotes Pancreáticos/métodos , Epiplón/cirugía
3.
Cells ; 10(7)2021 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-34359863

RESUMEN

Ongoing beta cell death in type 1 diabetes (T1D) can be detected using biomarkers selectively discharged by dying beta cells into plasma. microRNA-375 (miR-375) ranks among the top biomarkers based on studies in animal models and human islet transplantation. Our objective was to identify additional microRNAs that are co-released with miR-375 proportionate to the amount of beta cell destruction. RT-PCR profiling of 733 microRNAs in a discovery cohort of T1D patients 1 h before/after islet transplantation indicated increased plasma levels of 22 microRNAs. Sub-selection for beta cell selectivity resulted in 15 microRNAs that were subjected to double-blinded multicenter analysis. This led to the identification of eight microRNAs that were consistently increased during early graft destruction: besides miR-375, these included miR-132/204/410/200a/429/125b, microRNAs with known function and enrichment in beta cells. Their potential clinical translation was investigated in a third independent cohort of 46 transplant patients by correlating post-transplant microRNA levels to C-peptide levels 2 months later. Only miR-375 and miR-132 had prognostic potential for graft outcome, and none of the newly identified microRNAs outperformed miR-375 in multiple regression. In conclusion, this study reveals multiple beta cell-enriched microRNAs that are co-released with miR-375 and can be used as complementary biomarkers of beta cell death.


Asunto(s)
Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/patología , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Trasplante de Islotes Pancreáticos , MicroARNs/genética , Biomarcadores/metabolismo , Recuento de Células , Estudios de Cohortes , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , MicroARNs/metabolismo , Curva ROC , Reproducibilidad de los Resultados , Tropismo
4.
PLoS One ; 16(5): e0251055, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33939760

RESUMEN

Organs from donors after controlled circulatory death (DCD III) exhibit a higher risk for graft dysfunction due to an initial period of warm ischemia. This procurement condition can also affect the yield of beta cells in islet isolates from donor pancreases, and hence their use for transplantation. The present study uses data collected and generated by our Beta Cell Bank to compare the number of beta cells in isolates from DCD III (n = 141) with that from donors after brain death (DBD, n = 609), before and after culture, and examines the influence of donor and procurement variables. Beta cell number per DCD III-organ was significantly lower (58 x 106 versus 84 x 106 beta cells per DBD-organ; p < 0.001) but their purity (24% insulin positive cells) and insulin content (17 µg / 106 beta cells in DCD III-organs versus 19 µg / 106 beta cells in DBD-organs) were similar. Beta cell number correlated negatively with duration of acirculatory warm ischemia time above 10 min; for shorter acirculatory warm ischemia time, DCD III-organs did not exhibit a lower beta cell yield (74 x 106 beta cells). Use of Institut Georges Lopez-1 cold preservation solution instead of University of Wisconsin solution or histidine-tryptophan-ketoglutarate also protected against the loss in beta cell yield from DCD III-organs (86 x 106 for IGL-1 versus 54 x 106 and 65 x 106 beta cells respectively, p = 0.042). Multivariate analysis indicates that both limitation of acirculatory warm ischemia time and use of IGL-1 prevent the reduced beta cell yield in islet cell isolates from DCD III-organs.


Asunto(s)
Muerte Encefálica/metabolismo , Muerte Encefálica/patología , Supervivencia de Injerto/fisiología , Células Secretoras de Insulina/fisiología , Soluciones Preservantes de Órganos/metabolismo , Adenosina/metabolismo , Adenosina/fisiología , Adulto , Alopurinol/metabolismo , Femenino , Glutaratos/metabolismo , Glutatión/metabolismo , Glutatión/fisiología , Histidina/metabolismo , Humanos , Insulina/metabolismo , Insulina/fisiología , Células Secretoras de Insulina/metabolismo , Trasplante de Hígado/métodos , Masculino , Persona de Mediana Edad , Rafinosa/metabolismo , Rafinosa/fisiología , Donantes de Tejidos , Obtención de Tejidos y Órganos/métodos , Triptófano/metabolismo , Isquemia Tibia/métodos
5.
Virchows Arch ; 479(2): 295-304, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33594586

RESUMEN

Autoantibodies against islet cell antigens are routinely used to identify subjects at increased risk of symptomatic type 1 diabetes, but their relation to the intra-islet pathogenetic process that leads to positivity for these markers is poorly understood. We screened 556 non-diabetic organ donors (3 months to 24 years) for five different autoantibodies and found positivity in 27 subjects, 25 single- and two double autoantibody-positive donors. Histopathological screening of pancreatic tissue samples showed lesion characteristic for recent-onset type 1 diabetes in the two organ donors with a high-risk profile, due to their positivity for multiple autoantibodies and HLA-inferred risk. Inflammatory infiltrates (insulitis) were found in a small fraction of islets (<5%) and consisted predominantly of CD3+CD8+ T-cells. Islets with insulitis were found in close proximity to islets devoid of insulin-positivity; such pseudo-atrophic islets were present in multiple small foci scattered throughout the pancreatic tissue or were found to be distributed with a lobular pattern. Relative beta cell area in both single and multiple autoantibody-positive donors was comparable to that in autoantibody-negative controls. In conclusion, in organ donors under age 25 years, insulitis and pseudo-atrophic islets were restricted to multiple autoantibody-positive individuals allegedly at high risk of developing symptomatic type 1 diabetes, in line with reports in older age groups. These observations may give further insight into the early pathogenetic events that may culminate in clinically overt disease.


Asunto(s)
Antígenos/inmunología , Autoanticuerpos/sangre , Linfocitos T CD8-positivos/inmunología , Diabetes Mellitus Tipo 1/inmunología , Células Secretoras de Insulina/inmunología , Trasplante de Islotes Pancreáticos , Donantes de Tejidos , Adolescente , Factores de Edad , Biomarcadores/sangre , Estudios de Casos y Controles , Proliferación Celular , Niño , Preescolar , Diabetes Mellitus Tipo 1/sangre , Diabetes Mellitus Tipo 1/patología , Selección de Donante , Femenino , Humanos , Lactante , Células Secretoras de Insulina/patología , Masculino , Adulto Joven
6.
Am J Transplant ; 21(6): 2090-2099, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33206461

RESUMEN

Detection of amyloid in intraportal islet implants of type 1 diabetes patients has been proposed as cause in their functional decline. The present study uses cultured adult human islets devoid of amyloid to examine conditions of its formation. After intraportal injection in patients, amyloid deposits <15 µm diameter were identified in 5%-12% of beta cell containing aggregates, 3-76 months posttransplant. Such deposits also formed in glucose-controlling islet implants in the kidney of diabetic mice but not in failing implants. Alginate-encapsulated islets formed amyloid during culture when functional, and in all intraperitoneal implants that corrected diabetes in mice, exhibiting larger sizes than in functioning nonencapsulated implants. After intraperitoneal injection in a patient, retrieved single capsules presented amyloid near living beta cells, whereas no amyloid occurred in clustered capsules with dead cells. Amyloid was also demonstrated in functional human stem cell-generated beta cell implants in subcutaneous devices of mice. Deposits up to 35 µm diameter were localized in beta cell-enriched regions and related to an elevated IAPP over insulin ratio in the newly generated beta cells. Amyloid in device-encapsulated human stem cell-generated beta cell implants marks the formation of a functional beta cell mass but also an imbalance between its activated state and its microenvironment.


Asunto(s)
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Islotes Pancreáticos , Adulto , Amiloide , Animales , Humanos , Polipéptido Amiloide de los Islotes Pancreáticos , Ratones , Células Madre
7.
Am J Transplant ; 20(12): 3662-3666, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32476268

RESUMEN

Patients with heterotaxy syndrome (HS) can present with an associated complete dorsal pancreas agenesis (DPA). They are considered to be at increased risk for developing diabetes due to a reduced functional beta cell mass (FBM) as well as for chronic pancreatitis leading to unmanageable pain. We report the case of a young woman with chronic pancreatitis due to HS and associated DPA. She presented with a severe persisting upper abdominal pain refractory to nonsurgical treatment. Unlike in previously reported cases, she had a high FBM (ie, 150% of normoglycemic controls) as determined by hyperglycemic clamp. She underwent a total pancreatectomy followed within 24 hours by an intraportal autologous islet cell transplant containing 4 × 106 beta cells (4700 islet equivalent)/kg body weight. After surgery, the pain resolved, eliminating the need for analgesics. The intraportal implant established an adequate FBM (72% of controls at posttransplant month 2), achieving glycemic control without need for insulin administration. A hyperglycemic clamp can assess the utility and efficacy of an intraportal islet cell autotransplant following total pancreatectomy in patients with HS and complete DPA.


Asunto(s)
Síndrome de Heterotaxia , Células Secretoras de Insulina , Trasplante de Islotes Pancreáticos , Pancreatitis Crónica , Autoinjertos , Femenino , Humanos , Páncreas/diagnóstico por imagen , Páncreas/cirugía , Pancreatectomía , Pancreatitis Crónica/cirugía , Trasplante Autólogo , Resultado del Tratamiento
8.
Transplantation ; 104(10): e295-e302, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32433237

RESUMEN

BACKGROUND: Clinical islet transplantation is generally conducted within 72 hours after isolating sufficient beta-cell mass. A preparation that does not meet the sufficient dose can be cultured until this is reached after combination with subsequent ones. This retrospective study examines whether metabolic outcome is influenced by culture duration. METHODS: Forty type 1 diabetes recipients of intraportal islet cell grafts under antithymocyte globulin induction and mycophenolate mofetil-tacrolimus maintenance immunosuppression were analyzed. One subgroup (n = 10) was transplanted with preparations cultured for ≥96 hours; in the other subgroup (n = 30) grafts contained similar beta-cell numbers but included isolates that were cultured for a shorter duration. Both subgroups were compared by numbers with plasma C-peptide ≥0.5 ng/mL, low glycemic variability associated with C-peptide ≥1.0 ng/mL, and with insulin independence. RESULTS: The subgroup with all cells cultured ≥96 hours exhibited longer C-peptide ≥0.5 ng/mL (103 versus 48 mo; P = 0.006), and more patients with low glycemic variability and C-peptide ≥1.0 ng/mL, at month 12 (9/10 versus 12/30; P = 0.005) and 24 (7/10 versus 6/30; P = 0.007). In addition, 9/10 became insulin-independent versus 15/30 (P = 0.03). Grafts with all cells cultured ≥96 hours did not contain more beta cells but a higher endocrine purity (49% versus 36%; P = 0.03). In multivariate analysis, longer culture duration and older recipient age were independently associated with longer graft function. CONCLUSIONS: Human islet isolates with insufficient beta-cell mass for implantation within 72 hours can be cultured for 96 hours and longer to combine multiple preparations in order to reach the desired beta-cell dose and therefore result in a better metabolic benefit.


Asunto(s)
Proliferación Celular , Diabetes Mellitus Tipo 1/cirugía , Células Secretoras de Insulina/trasplante , Trasplante de Islotes Pancreáticos , Adulto , Biomarcadores/sangre , Glucemia/metabolismo , Péptido C/sangre , Células Cultivadas , Diabetes Mellitus Tipo 1/sangre , Diabetes Mellitus Tipo 1/diagnóstico , Femenino , Humanos , Hipoglucemiantes/uso terapéutico , Insulina/uso terapéutico , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Factores de Tiempo , Técnicas de Cultivo de Tejidos , Resultado del Tratamiento
9.
Transplantation ; 104(1): 190-196, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31365472

RESUMEN

BACKGROUND: Europe is currently the most active region in the field of pancreatic islet transplantation, and many of the leading groups are actually achieving similar good outcomes. Further collaborative advances in the field require the standardization of islet cell product isolation processes, and this work aimed to identify differences in the human pancreatic islet isolation processes within European countries. METHODS: A web-based questionnaire about critical steps, including donor selection, pancreas processing, pancreas perfusion and digestion, islet counting and culture, islet quality evaluation, microbiological evaluation, and release criteria of the product, was completed by isolation facilities participating at the Ninth International European Pancreas and Islet Transplant Association (EPITA) Workshop on Islet-Beta Cell Replacement in Milan. RESULTS: Eleven islet isolation facilities completed the questionnaire. The facilities reported 445 and 53 islet isolations per year over the last 3 years from deceased organ donors and pancreatectomized patients, respectively. This activity resulted in 120 and 40 infusions per year in allograft and autograft recipients, respectively. Differences among facilities emerged in donor selection (age, cold ischemia time, intensive care unit length, amylase concentration), pancreas procurement, isolation procedures (brand and concentration of collagenase, additive, maximum acceptable digestion time), quality evaluation, and release criteria for transplantation (glucose-stimulated insulin secretion tests, islet numbers, and purity). Moreover, even when a high concordance about the relevance of one parameter was evident, thresholds for the acceptance were different among facilities. CONCLUSIONS: The result highlighted the presence of a heterogeneity in the islet cell product process and product release criteria.


Asunto(s)
Separación Celular/métodos , Selección de Donante/métodos , Trasplante de Islotes Pancreáticos/métodos , Islotes Pancreáticos/citología , Recolección de Tejidos y Órganos/métodos , Adolescente , Adulto , Factores de Edad , Anciano , Recuento de Células/normas , Recuento de Células/estadística & datos numéricos , Separación Celular/estadística & datos numéricos , Células Cultivadas/trasplante , Niño , Preescolar , Isquemia Fría/normas , Isquemia Fría/estadística & datos numéricos , Selección de Donante/normas , Selección de Donante/estadística & datos numéricos , Europa (Continente) , Humanos , Lactante , Recién Nacido , Trasplante de Islotes Pancreáticos/normas , Persona de Mediana Edad , Perfusión/métodos , Perfusión/estadística & datos numéricos , Guías de Práctica Clínica como Asunto , Cultivo Primario de Células/métodos , Cultivo Primario de Células/normas , Cultivo Primario de Células/estadística & datos numéricos , Encuestas y Cuestionarios/estadística & datos numéricos , Factores de Tiempo , Recolección de Tejidos y Órganos/normas , Recolección de Tejidos y Órganos/estadística & datos numéricos , Adulto Joven
10.
Diabetes ; 69(3): 401-412, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31843955

RESUMEN

M2 macrophages play an important role in tissue repair and regeneration. They have also been found to modulate ß-cell replication in mouse models of pancreatic injury and disease. We previously reported that ß-cell replication is strongly increased in a subgroup of human organ donors characterized by prolonged duration of stay in an intensive care unit (ICU) and increased number of leukocytes in the pancreatic tissue. In the present study we investigated the relationship between duration of stay in the ICU, M2 macrophages, vascularization, and pancreatic cell replication. Pancreatic organs from 50 donors without diabetes with different durations of stay in the ICU were analyzed by immunostaining and digital image analysis. The number of CD68+CD206+ M2 macrophages increased three- to sixfold from ≥6 days' duration of stay in the ICU onwards. This was accompanied by a threefold increased vascular density and a four- to ninefold increase in pancreatic cells positive for the replication marker Ki67. A strong correlation was observed between the number of M2 macrophages and ß-cell replication. These results show that a prolonged duration of stay in the ICU is associated with an increased M2 macrophage number, increased vascular density, and an overall increase in replication of all pancreatic cell types. Our data show evidence of marked levels of tissue repair in the human donor pancreas.


Asunto(s)
Proliferación Celular/fisiología , Unidades de Cuidados Intensivos , Tiempo de Internación , Macrófagos/patología , Páncreas/fisiología , Regeneración/fisiología , Donantes de Tejidos , Adolescente , Adulto , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Femenino , Humanos , Antígeno Ki-67/metabolismo , Lectinas Tipo C/metabolismo , Macrófagos/metabolismo , Masculino , Receptor de Manosa , Lectinas de Unión a Manosa/metabolismo , Persona de Mediana Edad , Neovascularización Fisiológica/fisiología , Páncreas/metabolismo , Páncreas/patología , Receptores de Superficie Celular/metabolismo , Adulto Joven
11.
Stem Cells Transl Med ; 8(12): 1296-1305, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31379140

RESUMEN

Device-encapsulated human stem cell-derived pancreatic endoderm (PE) can generate functional ß-cell implants in the subcutis of mice, which has led to the start of clinical studies in type 1 diabetes. Assessment of the formed functional ß-cell mass (FBM) and its correlation with in vivo metabolic markers can guide clinical translation. We recently reported ex vivo characteristics of device-encapsulated human embryonic stem cell-derived (hES)-PE implants in mice that had established a metabolically adequate FBM during 50-week follow-up. Cell suspensions from retrieved implants indicated a correlation with the number of formed ß cells and their maturation to a functional state comparable to human pancreatic ß cells. Variability in metabolic outcome was attributed to differences in number of PE-generated ß cells. This variability hinders studies on processes involved in FBM-formation. This study reports modifications that reduce variability. It is undertaken with device-encapsulated human induced pluripotent stem cell-derived-PE subcutaneously implanted in mice. Cell mass of each cell type was determined on intact tissue inside the device to obtain more precise data than following isolation and dispersion. Implants in a preformed pouch generated a glucose-controlling ß-cell mass within 20 weeks in over 60% of recipients versus less than 20% in the absence of a pouch, whether the same or threefold higher cell dose had been inserted. In situ analysis of implants indicated a role for pancreatic progenitor cell expansion and endocrine differentiation in achieving the size of ß- and α-cell mass that correlated with in vivo markers of metabolic control. Stem Cells Translational Medicine 2019;8:1296&1305.


Asunto(s)
Endodermo/citología , Glucosa/metabolismo , Células Madre Pluripotentes Inducidas/citología , Células Secretoras de Insulina/citología , Trasplante de Islotes Pancreáticos/instrumentación , Páncreas/citología , Animales , Técnicas de Cultivo de Célula , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Endodermo/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Secretoras de Insulina/metabolismo , Trasplante de Islotes Pancreáticos/métodos , Masculino , Ratones , Ratones SCID , Páncreas/metabolismo , Ingeniería de Tejidos
12.
PLoS One ; 14(2): e0212210, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30779812

RESUMEN

Cell therapy for diabetes could benefit from the identification of small-molecule compounds that increase the number of functional pancreatic beta cells. Using a newly developed screening assay, we previously identified glucocorticoids as potent stimulators of human and rat beta cell proliferation. We now compare the stimulatory action of these steroid hormones to a selection of checkpoint tyrosine kinase inhibitors that were also found to activate the cell cycle-in beta cells and analyzed their respective effects on DNA-synthesis, beta cell numbers and expression of cell cycle regulators. Our data using glucocorticoids in combination with a receptor antagonist, mifepristone, show that 48h exposure is sufficient to allow beta cells to pass the cell cycle restriction point and to become committed to cell division regardless of sustained glucocorticoid-signaling. To reach the end-point of mitosis another 40h is required. Within 14 days glucocorticoids stimulate up to 75% of the cells to undergo mitosis, which indicates that these steroid hormones act as proliferation competence-inducing factors. In contrast, by correlating thymidine-analogue incorporation to changes in absolute cell numbers, we show that the checkpoint kinase inhibitors, as compared to glucocorticoids, stimulate DNA-synthesis only during a short time-window in a minority of cells, insufficient to give a measurable increase of beta cell numbers. Glucocorticoids, but not the kinase inhibitors, were also found to induce changes in the expression of checkpoint regulators. Our data, using checkpoint kinase-specific inhibitors further point to a role for Chk1 and Cdk1 in G1/S transition and progression of beta cells through the cell cycle upon stimulation with glucocorticoids.


Asunto(s)
Fase G1/efectos de los fármacos , Glucocorticoides/farmacología , Células Secretoras de Insulina/metabolismo , Mitosis/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Fase S/efectos de los fármacos , Adulto , Anciano , Animales , Proteína Quinasa CDC2/antagonistas & inhibidores , Proteína Quinasa CDC2/metabolismo , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/antagonistas & inhibidores , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/metabolismo , Femenino , Humanos , Células Secretoras de Insulina/citología , Masculino , Persona de Mediana Edad , Ratas
13.
J Clin Endocrinol Metab ; 104(2): 451-460, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30203041

RESUMEN

Aim: Several biomarkers have been proposed to detect pancreatic ß cell destruction in vivo but so far have not been compared for sensitivity and significance. Methods: We used islet transplantation as a model to compare plasma concentrations of miR-375, 65-kDa subunit of glutamate decarboxylase (GAD65), and unmethylated insulin DNA, measured at subpicomolar sensitivity, and study their discharge kinetics, power for outcome prediction, and detection of graft loss during follow-up. Results: At 60 minutes after transplantation, GAD65 and miR-375 consistently showed near-equimolar and correlated increases proportional to the number of implanted ß cells. GAD65 and miR-375 showed comparable power to predict poor graft outcome at 2 months, with areas under the curve of 0.833 and 0.771, respectively (P = 0.53). Using receiver operating characteristic analysis, we defined likelihood ratios (LRs) for rationally selected result intervals. In GADA-negative recipients (n = 28), GAD65 <4.5 pmol/L (LR = 0.15) and >12.2 pmol/L (LR = ∞) predicted good and poor outcomes, respectively. miR-375 could be used in all recipients irrespective of GAD65 autoantibody status (n = 46), with levels <1.4 pmol/L (LR = 0.14) or >7.6 pmol/L (LR = 9.53) as dual thresholds. The posttransplant surge of unmethylated insulin DNA was inconsistent and unrelated to outcome. Combined measurement of these three biomarkers was also tested as liquid biopsy for ß cell death during 2-month follow-up; incidental surges of GAD65, miR-375, and (un)methylated insulin DNA, alone or combined, were confidently detected but could not be related to outcome. Conclusions: GAD65 and miR-375 performed equally well in quantifying early graft destruction and predicting graft outcome, outperforming unmethylated insulin DNA.


Asunto(s)
Diabetes Mellitus Tipo 1/cirugía , Glutamato Descarboxilasa/sangre , Rechazo de Injerto/diagnóstico , Insulina/sangre , Trasplante de Islotes Pancreáticos/efectos adversos , MicroARNs/sangre , Adulto , Biomarcadores , Metilación de ADN , Estudios de Seguimiento , Rechazo de Injerto/sangre , Humanos , Insulina/genética , Persona de Mediana Edad , Periodo Posoperatorio , Pronóstico
14.
Diabetes ; 67(12): 2640-2649, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30305364

RESUMEN

Alginate (Alg)-encapsulated porcine islet cell grafts are developed to overcome limitations of human islet transplantation. They can generate functional implants in animals when prepared from fetal, perinatal, and adult pancreases. Implants have not yet been examined for efficacy to establish sustained, metabolically adequate functional ß-cell mass (FBM) in comparison with human islet cells. This study in immune-compromised mice demonstrates that subcutaneous implants of Alg-encapsulated porcine prenatal islet cells with 4 × 105 ß-cells form, over 10 weeks, a FBM that results in glucose-induced plasma C-peptide >2 ng/mL and metabolic control over the following 10 weeks, with higher efficiency than nonencapsulated, while failing in peritoneum. This intracapsular FBM formation involves ß-cell replication, increasing number fourfold, and maturation toward human adult ß-cells. Subcutaneous Alg-encapsulated human islet cells with similar ß-cell number establish implants with plasma C-peptide >2 ng/mL for the first 10 weeks, with nonencapsulated cells failing; their ß-cells do not replicate but progressively die (>70%), explaining C-peptide decline and insufficient metabolic control. An Alg matrix thus helps establish ß-cell functions in subcutis. It allows formation of sustained metabolically adequate FBM by immature porcine ß-cells with proliferative activity but not by human adult islet cells. These findings define conditions for evaluating its immune-protecting properties.


Asunto(s)
Alginatos , Péptido C/sangre , Células Secretoras de Insulina/metabolismo , Insulina/sangre , Trasplante de Islotes Pancreáticos/métodos , Animales , Cápsulas , Humanos , Ratones , Porcinos
15.
Diabetologia ; 61(7): 1623-1632, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29679103

RESUMEN

AIMS/HYPOTHESIS: HLA-A*24 carriership hampers achievement of insulin independence in islet allograft recipients. However, less than half of those who fail to achieve insulin independence carry the allele. We investigated whether genetic polymorphism at the recipients' zinc transporter 8-encoding SLC30A8 gene (rs13266634) could complement their HLA-A*24 status in predicting functional graft outcome. METHODS: We retrospectively analysed data of a hospital-based patient cohort followed for 18 months post transplantation. Forty C-peptide-negative type 1 diabetic individuals who received >2 million beta cells (>4000 islet equivalents) per kg body weight in one or two intraportal implantations under similar immunosuppression were genotyped for SLC30A8. Outcome measurements included achievement and maintenance of graft function. Metabolic benefit was defined as <25% CV of fasting glycaemia in the presence of >331 pmol/l C-peptide, in addition to achievement of insulin independence and maintenance of C-peptide positivity. RESULTS: In multivariate analysis, HLA-A*24 positivity, presence of SLC30A8 CT or TT genotypes and BMI more than or equal to the group median (23.9 kg/m2) were independently associated with failure to achieve insulin independence (p = 0.015-0.046). The risk increased with the number of factors present (p < 0.001). High BMI interacted with SLC30A8 T allele carriership to independently predict difficulty in achieving graft function with metabolic benefit (p = 0.015). Maintenance of C-peptide positivity was mainly associated with older age at the time of implantation. Only HLA-A*24 carriership independently predicted failure to maintain acceptable graft function once achieved (p = 0.012). CONCLUSIONS/INTERPRETATION: HLA-A*24, the SLC30A8 T allele and high BMI are associated with poor graft outcome and should be considered in the interpretation of future transplantation trials. TRIAL REGISTRATION: ClinicalTrials.gov NCT00798785 and NCT00623610.


Asunto(s)
Glucemia/metabolismo , Índice de Masa Corporal , Diabetes Mellitus Tipo 1/cirugía , Antígeno HLA-A24/genética , Células Secretoras de Insulina/trasplante , Trasplante de Islotes Pancreáticos/efectos adversos , Polimorfismo Genético , Transportador 8 de Zinc/genética , Aloinjertos , Biomarcadores/sangre , Glucemia/efectos de los fármacos , Ensayos Clínicos como Asunto , Diabetes Mellitus Tipo 1/sangre , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/inmunología , Femenino , Antígeno HLA-A24/inmunología , Humanos , Hipoglucemiantes/uso terapéutico , Insulina/uso terapéutico , Células Secretoras de Insulina/inmunología , Células Secretoras de Insulina/metabolismo , Masculino , Persona de Mediana Edad , Recuperación de la Función , Estudios Retrospectivos , Factores de Riesgo , Resultado del Tratamiento
16.
PLoS One ; 13(3): e0193670, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29518118

RESUMEN

A disproportional increase of circulating GAD65 within hours from an intraportal islet allotransplantation has been validated as biomarker of beta cell loss and poor functional outcome. More sensitive assays are, however, needed to allow detection of episodes of subtle beta cell loss during late-stage graft rejection or in the peri-onset period of type 1 diabetes. We applied the same sandwich monoclonal antibody couple reactive towards the C- and N-terminus of GAD65 on three advanced immunoassay platforms-the Cytometric Bead Array (CBA, Becton, Dickinson and Company), ElectroChemiLuminescence ImmunoAssay (ECLIA, Meso Scale Discovery) and digital ELISA technology (Single Molecule Array-SIMOA, Quanterix. We then compared analytical performance (linearity, imprecision, limit of detection and functional sensitivity), correlation of results, and practicality. All evaluated techniques showed linearity up to at least 500 ng/dL (76.9 pmol/L). SIMOA achieved the lowest imprecision. The 3 platforms correlate well with each other and could all detect subpicomolar concentrations of GAD65 in plasma, but only SIMOA and CBA could quantify down to that range. SIMOA can achieve the highest sample throughput. The three methods tested allow sensitive detection of GAD65, but SIMOA appears best suited for automated quantification of subpicomolar concentrations.


Asunto(s)
Glutamato Descarboxilasa/análisis , Glutamato Descarboxilasa/sangre , Inmunoensayo/instrumentación , Biomarcadores/sangre , Análisis Químico de la Sangre/instrumentación , Ensayo de Inmunoadsorción Enzimática/instrumentación , Humanos , Proteínas Recombinantes/análisis , Proteínas Recombinantes/sangre , Sensibilidad y Especificidad
17.
Stem Cell Reports ; 10(3): 739-750, 2018 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-29503087

RESUMEN

Human stem cells represent a potential source for implants that replace the depleted functional beta cell mass (FBM) in diabetes patients. Human embryonic stem cell-derived pancreatic endoderm (hES-PE) can generate implants with glucose-responsive beta cells capable of reducing hyperglycemia in mice. This study with device-encapsulated hES-PE (4 × 106 cells/mouse) determines the biologic characteristics at which implants establish metabolic control during a 50-week follow-up. A metabolically adequate FBM was achieved by (1) formation of a sufficient beta cell number (>0.3 × 106/mouse) at >50% endocrine purity and (2) their maturation to a functional state comparable with human pancreatic beta cells, as judged by their secretory responses during perifusion, their content in typical secretory vesicles, and their nuclear NKX6.1-PDX1-MAFA co-expression. Assessment of FBM in implants and its correlation with in vivo metabolic markers will guide clinical translation of stem cell-derived grafts in diabetes.


Asunto(s)
Células Madre Embrionarias/metabolismo , Células Madre Embrionarias/fisiología , Endodermo/metabolismo , Endodermo/fisiología , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/fisiología , Animales , Línea Celular , Glucosa/metabolismo , Proteínas de Homeodominio/metabolismo , Humanos , Factores de Transcripción Maf de Gran Tamaño/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Transactivadores/metabolismo
18.
Transplantation ; 101(9): 2218-2227, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-27779572

RESUMEN

BACKGROUND: Induction therapy with a T cell-depleting agent followed by mycophenolate mofetil and tacrolimus is presently the most frequently used immune suppression (IS) regimen in islet transplantation. This study assesses its safety and tolerability in nonuremic type 1 diabetic recipients. METHODS: Fifty-one patients (age, between 29 and 63 years) with high glycemic variability and problematic hypoglycemia received intraportal islet grafts under anti-thymocyte globulin-mycophenolate mofetil-tacrolimus protocol. They were followed up for over 48 months for function of the implant and adverse events. RESULTS: Severe hypoglycemia and diabetic ketoacidosis were absent in patients with functioning graft. Immune suppressive therapy was maintained for 48 months in 29 recipients with sustained function (group A), whereas 16 patients stopped earlier due to graft failure (group B) and in 6 for other reasons. Group A was significantly older at the time of implantation and achieved higher graft function at posttransplantation month 6 under similar dose of IS. Prevalence of IS-related side effects was similar in groups A and B, occurring predominantly during the first year posttransplantation. IS-related serious adverse events (SAE) were reported in 47% of patients, with 4 presenting with cytomegalovirus infection and 4 (age, 42-59 years) diagnosed with cancer. Except in 1 patient with cancer, all SAEs resolved after appropriate treatment. CONCLUSIONS: These risk/benefit data serve as a basis for clinical decision-making before entering an intraportal islet transplantation protocol. A longer benefit is observed in recipients of higher age (≥40 years), but it is not associated with more side effects and SAE.


Asunto(s)
Suero Antilinfocítico/uso terapéutico , Diabetes Mellitus Tipo 1/cirugía , Inmunosupresores/uso terapéutico , Trasplante de Islotes Pancreáticos , Ácido Micofenólico/uso terapéutico , Tacrolimus/uso terapéutico , Adulto , Suero Antilinfocítico/efectos adversos , Biomarcadores/sangre , Glucemia/metabolismo , Péptido C/sangre , Diabetes Mellitus Tipo 1/sangre , Quimioterapia Combinada , Femenino , Humanos , Inmunosupresores/efectos adversos , Trasplante de Islotes Pancreáticos/efectos adversos , Masculino , Persona de Mediana Edad , Ácido Micofenólico/efectos adversos , Complicaciones Posoperatorias/etiología , Medición de Riesgo , Factores de Riesgo , Tacrolimus/efectos adversos , Factores de Tiempo , Trasplante Homólogo , Resultado del Tratamiento
19.
Cell Transplant ; 26(1): 1-9, 2017 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-27729094

RESUMEN

Transplantation of islet allografts into type 1 diabetic recipients usually requires multiple pancreas donors to achieve insulin independence. This adds to the challenges of immunological monitoring of islet transplantation currently relying on surrogate immune markers in peripheral blood. We investigated donor origin and infiltration of islets transplanted in the liver of a T1D patient who died of hemorrhagic stroke 4 months after successful transplantation with two intraportal islet grafts combining six donors. Immunohistological staining for donor HLA using a unique panel of human monoclonal HLA-specific alloantibodies was performed on liver cryosections after validation on cryopreserved kidney, liver, and pancreas and compared with auto- and alloreactive T-cell immunity in peripheral blood. HLA-specific staining intensity and signal-to-noise ratio varied between tissues from very strong on kidney glomeruli, less in liver, kidney tubuli, and endocrine pancreas to least in exocrine pancreas, complicating the staining of inflamed islets in an HLA-disparate liver. Nonetheless, five islets from different liver lobes could be attributed to donors 1, 2, and 5 by staining patterns with multiple HLA types. All islets showed infiltration with CD8+ cytotoxic T cells that was mirrored by progressive alloreactive responses in peripheral blood mononuclear cells (PBMCs) to donors 1, 2, and 5 after transplantation. Stably low rates of peripheral islet autoreactive T-cell responses after islet infusion fit with a complete HLA mismatch between grafts and recipient and exclude the possibility that the islet-infiltrating CD8 T cells were autoreactive. HLA-specific immunohistochemistry can identify donor origin in situ and differentiate graft dysfunction and immunological destruction.


Asunto(s)
Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/cirugía , Trasplante de Islotes Pancreáticos/inmunología , Donantes de Tejidos , Autoinmunidad/inmunología , Linfocitos T CD8-positivos/metabolismo , Diabetes Mellitus Tipo 1/inmunología , Femenino , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Hígado/metabolismo , Persona de Mediana Edad , Páncreas/inmunología , Páncreas/metabolismo , Trasplante Homólogo
20.
Am J Physiol Endocrinol Metab ; 311(4): E698-E705, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27555297

RESUMEN

Compounds that increase ß-cell number can serve as ß-cell replacement therapies in diabetes. In vitro studies have identified several agents that can activate DNA synthesis in primary ß-cells but only in small percentages of cells and without demonstration of increases in cell number. We used whole well multiparameter imaging to first screen a library of 1,280 compounds for their ability to recruit adult rat ß-cells into DNA synthesis and then assessed influences of stimulatory agents on the number of living cells. The four compounds with highest ß-cell recruitment were glucocorticoid (GC) receptor ligands. The GC effect occurred in glucose-activated ß-cells and was associated with increased glucose utilization and oxidation. Hydrocortisone and methylprednisolone almost doubled the number of ß-cells in 2 wk. The expanded cell population provided an increased functional ß-cell mass for transplantation in diabetic animals. These effects are age dependent; they did not occur in neonatal rat ß-cells, where GC exposure suppressed basal replication and was cytotoxic. We concluded that GCs can induce the replication of adult rat ß-cells through a direct action, with intercellular differences in responsiveness that have been related to differences in glucose activation and in age. These influences can explain variability in GC-induced activation of DNA synthesis in rat and human ß-cells. Our study also demonstrated that ß-cells can be expanded in vitro to increase the size of metabolically adequate grafts.


Asunto(s)
Glucocorticoides/farmacología , Glucosa/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Trasplante de Islotes Pancreáticos/métodos , Animales , Animales Recién Nacidos , Recuento de Células , Separación Celular , ADN/biosíntesis , ADN/genética , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratas , Ratas Wistar , Receptores de Glucocorticoides/biosíntesis , Receptores de Glucocorticoides/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...