Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
1.
Cell ; 132(2): 197-207, 2008 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-18243096

RESUMO

Novel strategies in diabetes therapy would obviously benefit from the use of beta (beta) cell stem/progenitor cells. However, whether or not adult beta cell progenitors exist is one of the most controversial issues in today's diabetes research. Guided by the expression of Neurogenin 3 (Ngn3), the earliest islet cell-specific transcription factor in embryonic development, we show that beta cell progenitors can be activated in injured adult mouse pancreas and are located in the ductal lining. Differentiation of the adult progenitors is Ngn3 dependent and gives rise to all islet cell types, including glucose responsive beta cells that subsequently proliferate, both in situ and when cultured in embryonic pancreas explants. Multipotent progenitor cells thus exist in the pancreas of adult mice and can be activated cell autonomously to increase the functional beta cell mass by differentiation and proliferation rather than by self-duplication of pre-existing beta cells only.


Assuntos
Células Secretoras de Insulina/citologia , Pâncreas/citologia , Pâncreas/lesões , Células-Tronco/citologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/isolamento & purificação , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular , Núcleo Celular/metabolismo , Proliferação de Células , Expressão Gênica , Genes Reporter , Vetores Genéticos , Proteínas de Fluorescência Verde/metabolismo , Imuno-Histoquímica , Insulina/análise , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Queratinas/metabolismo , Lentivirus/genética , Ligadura , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/isolamento & purificação , Proteínas do Tecido Nervoso/metabolismo , Técnicas de Cultura de Órgãos , Ductos Pancreáticos/cirurgia , Células-Tronco/metabolismo , Fatores de Tempo , beta-Galactosidase/metabolismo
2.
Am J Transplant ; 22(3): 927-936, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34735732

RESUMO

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.


Assuntos
Diabetes Mellitus Tipo 1 , Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas , Diabetes Mellitus Tipo 1/cirurgia , Células Endoteliais , Humanos , Transplante das Ilhotas Pancreáticas/métodos , Omento/cirurgia
3.
Am J Transplant ; 21(6): 2090-2099, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33206461

RESUMO

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.


Assuntos
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Ilhotas Pancreáticas , Adulto , Amiloide , Animais , Humanos , Polipeptídeo Amiloide das Ilhotas Pancreáticas , Camundongos , Células-Tronco
4.
Am J Transplant ; 20(12): 3662-3666, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32476268

RESUMO

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.


Assuntos
Síndrome de Heterotaxia , Células Secretoras de Insulina , Transplante das Ilhotas Pancreáticas , Pancreatite Crônica , Autoenxertos , Feminino , Humanos , Pâncreas/diagnóstico por imagem , Pâncreas/cirurgia , Pancreatectomia , Pancreatite Crônica/cirurgia , Transplante Autólogo , Resultado do Tratamento
5.
Diabetologia ; 61(7): 1623-1632, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29679103

RESUMO

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.


Assuntos
Glicemia/metabolismo , Índice de Massa Corporal , Diabetes Mellitus Tipo 1/cirurgia , Antígeno HLA-A24/genética , Células Secretoras de Insulina/transplante , Transplante das Ilhotas Pancreáticas/efeitos adversos , Polimorfismo Genético , Transportador 8 de Zinco/genética , Aloenxertos , Biomarcadores/sangue , Glicemia/efeitos dos fármacos , Ensaios Clínicos como Assunto , Diabetes Mellitus Tipo 1/sangue , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/imunologia , Feminino , Antígeno HLA-A24/imunologia , Humanos , Hipoglicemiantes/uso terapêutico , Insulina/uso terapêutico , Células Secretoras de Insulina/imunologia , Células Secretoras de Insulina/metabolismo , Masculino , Pessoa de Meia-Idade , Recuperação de Função Fisiológica , Estudos Retrospectivos , Fatores de Risco , Resultado do Tratamento
6.
Am J Physiol Endocrinol Metab ; 311(4): E698-E705, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27555297

RESUMO

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.


Assuntos
Glucocorticoides/farmacologia , Glucose/farmacologia , Células Secretoras de Insulina/efeitos dos fármacos , Transplante das Ilhotas Pancreáticas/métodos , Animais , Animais Recém-Nascidos , Contagem de Células , Separação Celular , DNA/biossíntese , DNA/genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos NOD , Ratos , Ratos Wistar , Receptores de Glucocorticoides/biossíntese , Receptores de Glucocorticoides/genética
7.
Am J Physiol Endocrinol Metab ; 307(9): E838-46, 2014 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-25205822

RESUMO

ß-Cells generated from large-scale sources can overcome current shortages in clinical islet cell grafts provided that they adequately respond to metabolic variations. Pancreatic (non)endocrine cells can develop from human embryonic stem (huES) cells following in vitro derivation to pancreatic endoderm (PE) that is subsequently implanted in immune-incompetent mice for further differentiation. Encapsulation of PE increases the proportion of endocrine cells in subcutaneous implants, with enrichment in ß-cells when they are placed in TheraCyte-macrodevices and predominantly α-cells when they are alginate-microencapsulated. At posttransplant (PT) weeks 20-30, macroencapsulated huES implants presented higher glucose-responsive plasma C-peptide levels and a lower proinsulin-over-C-peptide ratio than human islet cell implants under the kidney capsule. Their ex vivo analysis showed the presence of single-hormone-positive α- and ß-cells that exhibited rapid secretory responses to increasing and decreasing glucose concentrations, similar to isolated human islet cells. However, their insulin secretory amplitude was lower, which was attributed in part to a lower cellular hormone content; it was associated with a lower glucose-induced insulin biosynthesis, but not with lower glucagon-induced stimulation, which together is compatible with an immature functional state of the huES-derived ß-cells at PT weeks 20-30. These data support the therapeutic potential of macroencapsulated huES implants but indicate the need for further functional analysis. Their comparison with clinical-grade human islet cell grafts sets references for future development and clinical translation.


Assuntos
Células Imobilizadas/transplante , Diabetes Mellitus Tipo 1/cirurgia , Células-Tronco Embrionárias/transplante , Implantes Experimentais/efeitos adversos , Transplante das Ilhotas Pancreáticas/efeitos adversos , Transplante Heterólogo/efeitos adversos , Transplante Heterotópico/efeitos adversos , Animais , Peptídeo C/sangue , Peptídeo C/metabolismo , Diferenciação Celular , Linhagem Celular , Células Imobilizadas/citologia , Células Imobilizadas/metabolismo , Cruzamentos Genéticos , Diabetes Mellitus Tipo 1/sangue , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patologia , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Células Secretoras de Glucagon/citologia , Células Secretoras de Glucagon/metabolismo , Humanos , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Rim , Membranas , Camundongos Endogâmicos NOD , Camundongos SCID , Proinsulina/sangue , Proinsulina/metabolismo , Tela Subcutânea , Alicerces Teciduais/efeitos adversos
8.
Gastroenterology ; 141(2): 731-41, 741.e1-4, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21703267

RESUMO

BACKGROUND & AIMS: Animal studies have indicated that pancreatic exocrine acinar cells have phenotypic plasticity. In rodents, acinar cells can differentiate into ductal precursors that can be converted to pancreatic ductal adenocarcinoma or insulin-producing endocrine cells. However, little is known about human acinar cell plasticity. We developed nongenetic and genetic lineage tracing methods to study the fate of human acinar cells in culture. METHODS: Human exocrine tissue was obtained from organ donors, dissociated, and cultured. Cell proliferation and survival were measured, and cell phenotypes were analyzed by immunocytochemistry. Nongenetic tracing methods were developed based on selective binding and uptake by acinar cells of a labeled lectin (Ulex europaeus agglutinin 1). Genetic tracing methods were developed based on adenoviral introduction of a Cre-lox reporter system, controlled by the amylase promoter. RESULTS: Both tracing methods showed that human acinar cells can transdifferentiate into cells that express specific ductal markers, such as cytokeratin 19, hepatocyte nuclear factor 1ß, SOX9, CD133, carbonic anhydrase II, and cystic fibrosis transmembrane conductance regulator. Within 1 week of culture, all surviving acinar cells had acquired a ductal phenotype. This transdifferentiation was decreased by inhibiting mitogen-activated protein kinase signaling. CONCLUSIONS: Human acinar cells have plasticity similar to that described in rodent cells. These results might be used to develop therapeutic strategies for patients with diabetes or pancreatic cancer.


Assuntos
Linhagem da Célula/genética , Transdiferenciação Celular/genética , Pâncreas Exócrino/citologia , Ductos Pancreáticos/citologia , RNA Mensageiro/metabolismo , Transdução de Sinais/fisiologia , Antígeno AC133 , Antígenos CD/metabolismo , Biomarcadores/metabolismo , Anidrase Carbônica II/metabolismo , Linhagem da Célula/fisiologia , Proliferação de Células , Sobrevivência Celular , Transdiferenciação Celular/fisiologia , Células Cultivadas , Quimotripsina/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Genes Reporter , Glicoproteínas/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Fator 1-beta Nuclear de Hepatócito/metabolismo , Humanos , Queratina-19/metabolismo , Antígeno Ki-67/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Pâncreas Exócrino/metabolismo , Ductos Pancreáticos/metabolismo , Peptídeos/metabolismo , Fenótipo , Lectinas de Plantas/farmacocinética , Regiões Promotoras Genéticas , Fatores de Transcrição SOX9/metabolismo , Transdução de Sinais/genética , Transdução Genética
9.
Cell Transplant ; 31: 9636897221096160, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35583214

RESUMO

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.


Assuntos
Células Secretoras de Insulina , Doadores de Tecidos , Peso Corporal , Morte Encefálica , Eutanásia , Humanos , Células Secretoras de Insulina/transplante , Pâncreas
10.
Cell Metab ; 4(5): 391-406, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17084712

RESUMO

Perturbation of endoplasmic reticulum (ER) homeostasis impairs insulin biosynthesis, beta cell survival, and glucose homeostasis. We show that a murine model of diabetes is associated with the development of ER stress in beta cells and that treatment with the GLP-1R agonist exendin-4 significantly reduced biochemical markers of islet ER stress in vivo. Exendin-4 attenuated translational downregulation of insulin and improved cell survival in purified rat beta cells and in INS-1 cells following induction of ER stress in vitro. GLP-1R agonists significantly potentiated the induction of ATF-4 by ER stress and accelerated recovery from ER stress-mediated translational repression in INS-1 beta cells in a PKA-dependent manner. The effects of exendin-4 on the induction of ATF-4 were mediated via enhancement of ER stress-stimulated ATF-4 translation. Moreover, exendin-4 reduced ER stress-associated beta cell death in a PKA-dependent manner. These findings demonstrate that GLP-1R signaling directly modulates the ER stress response leading to promotion of beta cell adaptation and survival.


Assuntos
Fator 4 Ativador da Transcrição/farmacologia , Retículo Endoplasmático/fisiologia , Células Secretoras de Insulina/fisiologia , Peptídeos/farmacologia , Receptores de Glucagon/metabolismo , Estresse Fisiológico , Peçonhas/farmacologia , eIF-2 Quinase/fisiologia , Animais , Sobrevivência Celular , Células Cultivadas , Retículo Endoplasmático/efeitos dos fármacos , Exenatida , Feminino , Receptor do Peptídeo Semelhante ao Glucagon 1 , Glucose/metabolismo , Homeostase , Hipoglicemiantes/farmacologia , Células Secretoras de Insulina/efeitos dos fármacos , Camundongos , Ratos , Ratos Wistar , Receptores de Glucagon/fisiologia , Regulação para Cima
11.
PLoS One ; 16(5): e0251055, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33939760

RESUMO

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.


Assuntos
Morte Encefálica/metabolismo , Morte Encefálica/patologia , Sobrevivência de Enxerto/fisiologia , Células Secretoras de Insulina/fisiologia , Soluções para Preservação de Órgãos/metabolismo , Adenosina/metabolismo , Adenosina/fisiologia , Adulto , Alopurinol/metabolismo , Feminino , Glutaratos/metabolismo , Glutationa/metabolismo , Glutationa/fisiologia , Histidina/metabolismo , Humanos , Insulina/metabolismo , Insulina/fisiologia , Células Secretoras de Insulina/metabolismo , Transplante de Fígado/métodos , Masculino , Pessoa de Meia-Idade , Rafinose/metabolismo , Rafinose/fisiologia , Doadores de Tecidos , Obtenção de Tecidos e Órgãos/métodos , Triptofano/metabolismo , Isquemia Quente/métodos
12.
Cells ; 10(7)2021 07 04.
Artigo em Inglês | MEDLINE | ID: mdl-34359863

RESUMO

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.


Assuntos
Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/patologia , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Transplante das Ilhotas Pancreáticas , MicroRNAs/genética , Biomarcadores/metabolismo , Contagem de Células , Estudos de Coortes , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , MicroRNAs/metabolismo , Curva ROC , Reprodutibilidade dos Testes , Tropismo
13.
Virchows Arch ; 479(2): 295-304, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-33594586

RESUMO

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.


Assuntos
Antígenos/imunologia , Autoanticorpos/sangue , Linfócitos T CD8-Positivos/imunologia , Diabetes Mellitus Tipo 1/imunologia , Células Secretoras de Insulina/imunologia , Transplante das Ilhotas Pancreáticas , Doadores de Tecidos , Adolescente , Fatores Etários , Biomarcadores/sangue , Estudos de Casos e Controles , Proliferação de Células , Criança , Pré-Escolar , Diabetes Mellitus Tipo 1/sangue , Diabetes Mellitus Tipo 1/patologia , Seleção do Doador , Feminino , Humanos , Lactente , Células Secretoras de Insulina/patologia , Masculino , Adulto Jovem
14.
Transplantation ; 104(10): e295-e302, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32433237

RESUMO

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.


Assuntos
Proliferação de Células , Diabetes Mellitus Tipo 1/cirurgia , Células Secretoras de Insulina/transplante , Transplante das Ilhotas Pancreáticas , Adulto , Biomarcadores/sangue , Glicemia/metabolismo , Peptídeo C/sangue , Células Cultivadas , Diabetes Mellitus Tipo 1/sangue , Diabetes Mellitus Tipo 1/diagnóstico , Feminino , Humanos , Hipoglicemiantes/uso terapêutico , Insulina/uso terapêutico , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Fatores de Tempo , Técnicas de Cultura de Tecidos , Resultado do Tratamento
15.
Diabetes ; 69(3): 401-412, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31843955

RESUMO

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.


Assuntos
Proliferação de Células/fisiologia , Unidades de Terapia Intensiva , Tempo de Internação , Macrófagos/patologia , Pâncreas/fisiologia , Regeneração/fisiologia , Doadores de Tecidos , Adolescente , Adulto , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Feminino , Humanos , Antígeno Ki-67/metabolismo , Lectinas Tipo C/metabolismo , Macrófagos/metabolismo , Masculino , Receptor de Manose , Lectinas de Ligação a Manose/metabolismo , Pessoa de Meia-Idade , Neovascularização Fisiológica/fisiologia , Pâncreas/metabolismo , Pâncreas/patologia , Receptores de Superfície Celular/metabolismo , Adulto Jovem
16.
Transplantation ; 104(1): 190-196, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31365472

RESUMO

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.


Assuntos
Separação Celular/métodos , Seleção do Doador/métodos , Transplante das Ilhotas Pancreáticas/métodos , Ilhotas Pancreáticas/citologia , Coleta de Tecidos e Órgãos/métodos , Adolescente , Adulto , Fatores Etários , Idoso , Contagem de Células/normas , Contagem de Células/estatística & dados numéricos , Separação Celular/estatística & dados numéricos , Células Cultivadas/transplante , Criança , Pré-Escolar , Isquemia Fria/normas , Isquemia Fria/estatística & dados numéricos , Seleção do Doador/normas , Seleção do Doador/estatística & dados numéricos , Europa (Continente) , Humanos , Lactente , Recém-Nascido , Transplante das Ilhotas Pancreáticas/normas , Pessoa de Meia-Idade , Perfusão/métodos , Perfusão/estatística & dados numéricos , Guias de Prática Clínica como Assunto , Cultura Primária de Células/métodos , Cultura Primária de Células/normas , Cultura Primária de Células/estatística & dados numéricos , Inquéritos e Questionários/estatística & dados numéricos , Fatores de Tempo , Coleta de Tecidos e Órgãos/normas , Coleta de Tecidos e Órgãos/estatística & dados numéricos , Adulto Jovem
17.
PLoS One ; 14(2): e0212210, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30779812

RESUMO

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.


Assuntos
Fase G1/efeitos dos fármacos , Glucocorticoides/farmacologia , Células Secretoras de Insulina/metabolismo , Mitose/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Fase S/efeitos dos fármacos , Adulto , Idoso , Animais , Proteína Quinase CDC2/antagonistas & inibidores , Proteína Quinase CDC2/metabolismo , Quinase 1 do Ponto de Checagem/antagonistas & inibidores , Quinase 1 do Ponto de Checagem/metabolismo , Feminino , Humanos , Células Secretoras de Insulina/citologia , Masculino , Pessoa de Meia-Idade , Ratos
18.
Stem Cells Transl Med ; 8(12): 1296-1305, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31379140

RESUMO

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.


Assuntos
Endoderma/citologia , Glucose/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Células Secretoras de Insulina/citologia , Transplante das Ilhotas Pancreáticas/instrumentação , Pâncreas/citologia , Animais , Técnicas de Cultura de Células , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Endoderma/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células Secretoras de Insulina/metabolismo , Transplante das Ilhotas Pancreáticas/métodos , Masculino , Camundongos , Camundongos SCID , Pâncreas/metabolismo , Engenharia Tecidual
19.
J Clin Endocrinol Metab ; 104(2): 451-460, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30203041

RESUMO

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.


Assuntos
Diabetes Mellitus Tipo 1/cirurgia , Glutamato Descarboxilase/sangue , Rejeição de Enxerto/diagnóstico , Insulina/sangue , Transplante das Ilhotas Pancreáticas/efeitos adversos , MicroRNAs/sangue , Adulto , Biomarcadores , Metilação de DNA , Seguimentos , Rejeição de Enxerto/sangue , Humanos , Insulina/genética , Pessoa de Meia-Idade , Período Pós-Operatório , Prognóstico
20.
Transplantation ; 85(2): 256-63, 2008 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-18212631

RESUMO

BACKGROUND: One year survival of islet cell grafts has been reproducibly achieved under combination immune therapy including tacrolimus (TAC). However, the use of TAC causes beta-cell and renal toxicity. Because sirolimus (SIR) monotherapy was successful in kidney transplantation under antithymocyte globulin (ATG), we undertook a pilot study comparing SIR monotherapy with SIR-TAC combination therapy. METHODS: Nonuremic type 1 diabetics received a cultured beta-cell graft under ATG and were randomly assigned to SIR or SIR-TAC-maintenance therapy; a second graft was implanted during posttransplantation month 3 without ATG. The planned number of patients per group (n=10) was reduced to five in view of the observed side effects. RESULTS: At posttransplant month 6, three SIR-patients had lost graft function and two presented marginal function; among SIR-TAC-patients, there were two early graft failures but three became insulin-independent. These three patients maintained metabolically relevant function (C-peptide >1 ng/ml and coefficient of variation fasting glycemia <25%) for more than 2 years but low-dose insulin therapy was needed from 8, 18, and 26 months posttransplant; this was still the case in two of them after reducing and stopping TAC dose. In both groups, incapacitating adverse events were attributed to sirolimus requiring its discontinuation in 4 of 10 patients; in the 3 patients with pretransplant microalbuminuria, macroalbuminuria developed which resolved when sirolimus was stopped. CONCLUSIONS: SIR monotherapy is not sufficient to suppress rejection after transplantation under ATG, but it can maintain survival of established beta-cell grafts. However, the risk for a SIR-induced proteinuria remains a concern.


Assuntos
Transplante de Células/métodos , Diabetes Mellitus Tipo 1/cirurgia , Transplante das Ilhotas Pancreáticas/imunologia , Ilhotas Pancreáticas/citologia , Sirolimo/uso terapêutico , Tacrolimo/uso terapêutico , Adulto , Albuminúria/epidemiologia , Autoanticorpos/sangue , Peptídeo C/sangue , Transplante de Células/efeitos adversos , Quimioterapia Combinada , Feminino , Sobrevivência de Enxerto/imunologia , Humanos , Imunossupressores/uso terapêutico , Ilhotas Pancreáticas/imunologia , Transplante das Ilhotas Pancreáticas/efeitos adversos , Contagem de Linfócitos , Masculino , Pessoa de Meia-Idade , Complicações Pós-Operatórias/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA