Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 424
Filtrar
Mais filtros

País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Physiol Rev ; 98(3): 1143-1167, 2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29717931

RESUMO

Diabetes mellitus results from disturbed glucose homeostasis due to an absolute (type 1) or relative (type 2) deficiency of insulin, a peptide hormone almost exclusively produced by the beta cells of the endocrine pancreas in a tightly regulated manner. Current therapy only delays disease progression through insulin injection and/or oral medications that increase insulin secretion or sensitivity, decrease hepatic glucose production, or promote glucosuria. These drugs have turned diabetes into a chronic disease as they do not solve the underlying beta cell defects or entirely prevent the long-term complications of hyperglycemia. Beta cell replacement through islet transplantation is a more physiological therapeutic alternative but is severely hampered by donor shortage and immune rejection. A curative strategy should combine newer approaches to immunomodulation with beta cell replacement. Success of this approach depends on the development of practical methods for generating beta cells, either in vitro or in situ through beta cell replication or beta cell differentiation. This review provides an overview of human beta cell generation.


Assuntos
Técnicas de Cultura de Células , Células Secretoras de Insulina/fisiologia , Regeneração , Animais , Homeostase , Humanos , Células Secretoras de Insulina/transplante
2.
Proc Natl Acad Sci U S A ; 117(1): 448-453, 2020 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-31871187

RESUMO

Voltage-gated calcium 3.1 (CaV3.1) channels are absent in healthy mouse ß cells and mediate minor T-type Ca2+ currents in healthy rat and human ß cells but become evident under diabetic conditions. Whether more active CaV3.1 channels affect insulin secretion and glucose homeostasis remains enigmatic. We addressed this question by enhancing de novo expression of ß cell CaV3.1 channels and exploring the consequent impacts on dynamic insulin secretion and glucose homeostasis as well as underlying molecular mechanisms with a series of in vitro and in vivo approaches. We now demonstrate that a recombinant adenovirus encoding enhanced green fluorescent protein-CaV3.1 subunit (Ad-EGFP-CaV3.1) efficiently transduced rat and human islets as well as dispersed islet cells. The resulting CaV3.1 channels conducted typical T-type Ca2+ currents, leading to an enhanced basal cytosolic-free Ca2+ concentration ([Ca2+]i). Ad-EGFP-CaV3.1-transduced islets released significantly less insulin under both the basal and first phases following glucose stimulation and could no longer normalize hyperglycemia in recipient rats rendered diabetic by streptozotocin treatment. Furthermore, Ad-EGFP-CaV3.1 transduction reduced phosphorylated FoxO1 in the cytoplasm of INS-1E cells, elevated FoxO1 nuclear retention, and decreased syntaxin 1A, SNAP-25, and synaptotagmin III. These effects were prevented by inhibiting CaV3.1 channels or the Ca2+-dependent phosphatase calcineurin. Enhanced expression of ß cell CaV3.1 channels therefore impairs insulin release and glucose homeostasis by means of initial excessive Ca2+ influx, subsequent activation of calcineurin, consequent dephosphorylation and nuclear retention of FoxO1, and eventual FoxO1-mediated down-regulation of ß cell exocytotic proteins. The present work thus suggests an elevated expression of CaV3.1 channels plays a significant role in diabetes pathogenesis.


Assuntos
Canais de Cálcio Tipo T/metabolismo , Diabetes Mellitus Experimental/metabolismo , Secreção de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Adolescente , Adulto , Animais , Células COS , Bloqueadores dos Canais de Cálcio/farmacologia , Bloqueadores dos Canais de Cálcio/uso terapêutico , Canais de Cálcio Tipo T/genética , Núcleo Celular/metabolismo , Chlorocebus aethiops , Citosol/metabolismo , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/tratamento farmacológico , Exocitose/efeitos dos fármacos , Estudos de Viabilidade , Feminino , Proteínas de Fluorescência Verde/genética , Células HEK293 , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/transplante , Masculino , Pessoa de Meia-Idade , Fosforilação , Cultura Primária de Células , Ratos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Estreptozocina/toxicidade , Proteínas de Transporte Vesicular/metabolismo , Adulto Jovem
3.
Stem Cells ; 39(5): 522-535, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33497522

RESUMO

Strategies to mitigate the pathologies from diabetes range from simply administering insulin to prescribing complex drug/biologic regimens combined with lifestyle changes. There is a substantial effort to better understand ß-cell physiology during diabetes pathogenesis as a means to develop improved therapies. The convergence of multiple fields ranging from developmental biology to microfluidic engineering has led to the development of new experimental systems to better study complex aspects of diabetes and ß-cell biology. Here we discuss the available insulin-secreting cell types used in research, ranging from primary human ß-cells, to cell lines, to pluripotent stem cell-derived ß-like cells. Each of these sources possess inherent strengths and weaknesses pertinent to specific applications, especially in the context of engineered platforms. We then outline how insulin-expressing cells have been used in engineered platforms and how recent advances allow for better mimicry of in vivo conditions. Chief among these conditions are ß-cell interactions with other endocrine organs. This facet is beginning to be thoroughly addressed by the organ-on-a-chip community, but holds enormous potential in the development of novel diabetes therapeutics. Furthermore, high throughput strategies focused on studying ß-cell biology, improving ß-cell differentiation, or proliferation have led to enormous contributions in the field and will no doubt be instrumental in bringing new diabetes therapeutics to the clinic.


Assuntos
Diabetes Mellitus/terapia , Células Secretoras de Insulina/metabolismo , Insulina/biossíntese , Células-Tronco Pluripotentes/metabolismo , Comunicação Celular/genética , Diferenciação Celular/genética , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patologia , Humanos , Insulina/genética , Células Secretoras de Insulina/transplante , Dispositivos Lab-On-A-Chip , Células-Tronco Pluripotentes/transplante
4.
Biochem Biophys Res Commun ; 534: 1053-1058, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-33160622

RESUMO

High mobility group (HMGB1) is an alarmin known to be harmful to pancreatic beta cells and associated with diabetes mellitus pathogenesis and pancreatic islet graft failure. It has been long thought that the suppression of HMGB1 molecule is beneficial to the beta cells. However, recent studies have indicated that cytoplasmic HMGB1 (cHMGB1) could function as a modulator to relieve cells from apoptotic stress by autophagy induction. Particularly, pancreatic beta cells have been known to utilize the autophagy-to-apoptosis switch when exposed to hypoxia or lipotoxicity. This study aimed to investigate the beta cells under hypoxic and lipotoxic stress while utilizing a small molecule inhibitor of HMGB1, inflachromene (ICM) which can suppress cHMGB1 accumulation. It was revealed that under cellular stress, blockade of cHMGB1 accumulation decreased the viability of islet grafts, primary islets and MIN6 cells. MIN6 cells under cHMGB1 blockade along with lipotoxic stress showed decreased autophagic flux and increased apoptosis. Moreover, cHMGB1 blockade in HFD-fed mice produced unfavorable outcomes on their glucose tolerance. In sum, these results suggested the role of cHMGB1 within beta cell autophagy/apoptosis checkpoint. Given the importance of autophagy in beta cells under apoptotic stresses, this study might provide further insights regarding HMGB1 and diabetes.


Assuntos
Proteína HMGB1/antagonistas & inibidores , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Hipóxia Celular , Sobrevivência Celular/efeitos dos fármacos , Proteína HMGB1/metabolismo , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/transplante , Masculino , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Suínos
5.
Proc Natl Acad Sci U S A ; 115(15): 3924-3929, 2018 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-29599125

RESUMO

Type 1 diabetes is characterized by autoimmune destruction of ß cells located in pancreatic islets. However, tractable in vivo models of human pancreatic ß cells have been limited. Here, we generated xenogeneic human pancreatic ß-like cells in the mouse pancreas by orthotopic transplantation of stem cell-derived ß (SC-ß) cells into the pancreas of neonatal mice. The engrafted ß-like cells expressed ß cell transcription factors and markers associated with functional maturity. Engrafted human cells recruited mouse endothelial cells, suggesting functional integration. Human insulin was detected in the blood circulation of transplanted mice for months after transplantation and increased upon glucose stimulation. In addition to ß-like cells, human cells expressing markers for other endocrine pancreas cell types, acinar cells, and pancreatic ductal cells were identified in the pancreata of transplanted mice, indicating that this approach allows studying other human pancreatic cell types in the mouse pancreas. Our results demonstrate that orthotopic transplantation of human SC-ß cells into neonatal mice is an experimental platform that allows the generation of mice with human pancreatic ß-like cells in the endogenous niche.


Assuntos
Diabetes Mellitus Tipo 1/terapia , Células Secretoras de Insulina/citologia , Pâncreas/citologia , Células-Tronco Pluripotentes/transplante , Células Acinares/metabolismo , Animais , Diferenciação Celular , Proliferação de Células , Terapia Baseada em Transplante de Células e Tecidos , Células Cultivadas , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/fisiopatologia , Glucose/metabolismo , Humanos , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/transplante , Camundongos , Pâncreas/metabolismo , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo
6.
J Am Chem Soc ; 142(7): 3430-3439, 2020 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-32040300

RESUMO

Pancreatic ß cells are responsible for insulin secretion and are important for glucose regulation in a healthy body and diabetic disease patient without prelabeling of islets. While the conventional biomarkers for diabetes have been glucose and insulin concentrations in the blood, the direct determination of the pancreatic ß cell mass would provide critical information for the disease status and progression. By combining fluorination and diversity-oriented fluorescence library strategy, we have developed a multimodal pancreatic ß cell probe PiF for both fluorescence and for PET (positron emission tomography). By simple tail vein injection, PiF stains pancreatic ß cells specifically and allows intraoperative fluorescent imaging of pancreatic islets. PiF-injected pancreatic tissue even facilitated an antibody-free islet analysis within 2 h, dramatically accelerating the day-long histological procedure without any fixing and dehydration step. Not only islets in the pancreas but also the low background of PiF in the liver allowed us to monitor the intraportal transplanted islets, which is the first in vivo visualization of transplanted human islets without a prelabeling of the islets. Finally, we could replace the built-in fluorine atom in PiF with radioactive 18F and successfully demonstrate in situ PET imaging for pancreatic islets.


Assuntos
Corantes Fluorescentes/química , Células Secretoras de Insulina/citologia , Xantenos/química , Animais , Diabetes Mellitus Experimental/patologia , Fluorescência , Corantes Fluorescentes/síntese química , Corantes Fluorescentes/farmacocinética , Corantes Fluorescentes/toxicidade , Humanos , Células Secretoras de Insulina/transplante , Transplante das Ilhotas Pancreáticas , Fígado/citologia , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Tomografia por Emissão de Pósitrons , Ratos , Xantenos/síntese química , Xantenos/farmacocinética , Xantenos/toxicidade
7.
Am J Physiol Endocrinol Metab ; 319(2): E388-E400, 2020 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-32543944

RESUMO

Replacement of islets/ß-cells that provide long-lasting glucose-sensing and insulin-releasing functions has the potential to restore extended glycemic control in individuals with type 1 diabetes. Unfortunately, persistent challenges preclude such therapies from widespread clinical use, including cumbersome administration via portal vein infusion, significant loss of functional islet mass upon administration, limited functional longevity, and requirement for systemic immunosuppression. Previously, fibril-forming type I collagen (oligomer) was shown to support subcutaneous injection and in situ encapsulation of syngeneic islets within diabetic mice, with rapid (<24 h) reversal of hyperglycemia and maintenance of euglycemia for beyond 90 days. Here, we further evaluated this macroencapsulation strategy, defining effects of islet source (allogeneic and xenogeneic) and dose (500 and 800 islets), injection microenvironment (subcutaneous and intraperitoneal), and macrocapsule format (injectable and preformed implantable) on islet functional longevity and recipient immune response. We found that xenogeneic rat islets functioned similarly to or better than allogeneic mouse islets, with only modest improvements in longevity noted with dosage. Additionally, subcutaneous injection led to more consistent encapsulation outcomes along with improved islet health and longevity, compared with intraperitoneal administration, whereas no significant differences were observed between subcutaneous injectable and preformed implantable formats. Collectively, these results document the benefits of incorporating natural collagen for islet/ß-cell replacement therapies.


Assuntos
Encapsulamento de Células/métodos , Colágeno , Diabetes Mellitus Tipo 1/terapia , Transplante das Ilhotas Pancreáticas/métodos , Aloenxertos , Animais , Glicemia/análise , Sobrevivência Celular , Colágeno/química , Diabetes Mellitus Experimental/terapia , Diabetes Mellitus Tipo 1/sangue , Sobrevivência de Enxerto , Xenoenxertos , Injeções Intraperitoneais , Injeções Subcutâneas , Células Secretoras de Insulina/fisiologia , Células Secretoras de Insulina/transplante , Ilhotas Pancreáticas/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Ratos , Ratos Sprague-Dawley
8.
Lancet ; 394(10205): 1274-1285, 2019 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-31533905

RESUMO

The main goal of treatment for type 1 diabetes is to control glycaemia with insulin therapy to reduce disease complications. For some patients, technological approaches to insulin delivery are inadequate, and allogeneic islet transplantation is a safe alternative for those patients who have had severe hypoglycaemia complicated by impaired hypoglycaemia awareness or glycaemic lability, or who already receive immunosuppressive drugs for a kidney transplant. Since 2000, intrahepatic islet transplantation has proven efficacious in alleviating the burden of labile diabetes and preventing complications related to diabetes, whether or not a previous kidney transplant is present. Age, body-mass index, renal status, and cardiopulmonary status affect the choice between pancreas or islet transplantation. Access to transplantation is limited by the number of deceased donors and the necessity of immunosuppression. Future approaches might include alternative sources of islets (eg, xenogeneic tissue or human stem cells), extrahepatic sites of implantation (eg, omental, subcutaneous, or intramuscular), and induction of immune tolerance or encapsulation of islets.


Assuntos
Diabetes Mellitus Tipo 1/cirurgia , Células Secretoras de Insulina/transplante , Rejeição de Enxerto/prevenção & controle , Humanos , Terapia de Imunossupressão , Resultado do Tratamento
9.
Clin Exp Immunol ; 201(2): 222-230, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32293025

RESUMO

T regulatory type 1 (Tr1) cells are a class of regulatory T cells (Tregs ) participating in peripheral tolerance, hence the rationale behind their testing in clinical trials in different disease settings. One of their applications is tolerance induction to allogeneic islets for long-term diabetes-free survival. Currently the cellular and molecular mechanisms that promote Tr1-cell induction in vivo remain poorly understood. We employed a mouse model of transplant tolerance where treatment with granulocyte colony-stimulating factor (G-CSF)/rapamycin induces permanent engraftment of allogeneic pancreatic islets in C57BL/6 mice via Tr1 cells. The innate composition of graft and spleen cells in tolerant mice was analyzed by flow cytometry. Graft phagocytic cells were co-cultured with CD4+ T cells in vitro to test their ability to induce Tr1-cell induction. Graft phagocytic cells were depleted in vivo at different time-points during G-CSF/rapamycin treatment, to identify their role in Tr1-cell induction and consequently in graft survival. In the spleen, the site of Tr1-cell induction, no differences in the frequencies of macrophages or dendritic cells (DC) were observed. In the graft, the site of antigen uptake, a high proportion of macrophages and not DC was detected in tolerant but not in rejecting mice. Graft-infiltrating macrophages of G-CSF/rapamycin-treated mice had an M2 phenotype, characterized by higher CD206 expression and interleukin (IL)-10 production, whereas splenic macrophages only had an increased CD206 expression. Graft-infiltrating cells from G-CSF/rapamycin-treated mice-induced Tr1-cell expansion in vitro. Furthermore, Tr1-cell induction was perturbed upon in-vivo depletion of phagocytic cells, early and not late during treatment, leading to graft loss suggesting that macrophages play a key role in tolerance induction mediated by Tr1 cells. Taken together, in this mouse model of Tr1-cell induced tolerance to allogeneic islets, M2 macrophages infiltrating the graft upon G-CSF/rapamycin treatment are key for Tr1-cell induction. This work provides mechanistic insight into pharmacologically induced Tr1-cell expansion in vivo in this stringent model of allogeneic transplantation.


Assuntos
Diabetes Mellitus Experimental/imunologia , Células Secretoras de Insulina/citologia , Transplante das Ilhotas Pancreáticas , Macrófagos/imunologia , Linfócitos T Reguladores/imunologia , Células Th1/imunologia , Animais , Células Cultivadas , Modelos Animais de Doenças , Fator Estimulador de Colônias de Granulócitos/metabolismo , Humanos , Células Secretoras de Insulina/transplante , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Sirolimo/metabolismo , Tolerância ao Transplante , Transplante Homólogo
10.
Diabet Med ; 37(4): 580-592, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31705689

RESUMO

Despite major advances in structured education, insulin delivery and glucose monitoring, diabetes self-management remains an unremitting challenge. Insulin therapy is inextricably linked to risk of dangerous hypoglycaemia and sustained hyperglycaemia remains a leading cause of renal failure. This review sets out to demystify transplantation for diabetes multidisciplinary teams, facilitating consideration and incorporation within holistic overall person-centred management. Deceased and living donor kidney, whole pancreas and isolated islet transplant procedures, indications and potential benefits are described, in addition to outcomes within the integrated UK transplant programme.


Assuntos
Diabetes Mellitus/terapia , Células Secretoras de Insulina/transplante , Transplante de Rim/métodos , Humanos , Células Secretoras de Insulina/fisiologia , Transplante das Ilhotas Pancreáticas/métodos , Doadores Vivos , Transplante de Pâncreas/métodos , Doadores de Tecidos/provisão & distribuição
11.
Int J Mol Sci ; 21(19)2020 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-33019671

RESUMO

There are presently no reliable ways to quantify human pancreatic beta cell mass (BCM) in vivo, which prevents an accurate understanding of the progressive beta cell loss in diabetes or following islet transplantation. Furthermore, the lack of beta cell imaging hampers the evaluation of the impact of new drugs aiming to prevent beta cell loss or to restore BCM in diabetes. We presently discuss the potential value of BCM determination as a cornerstone for individualized therapies in diabetes, describe the presently available probes for human BCM evaluation, and discuss our approach for the discovery of novel beta cell biomarkers, based on the determination of specific splice variants present in human beta cells. This has already led to the identification of DPP6 and FXYD2ga as two promising targets for human BCM imaging, and is followed by a discussion of potential safety issues, the role for radiochemistry in the improvement of BCM imaging, and concludes with an overview of the different steps from pre-clinical validation to a first-in-man trial for novel tracers.


Assuntos
Diabetes Mellitus Tipo 1/diagnóstico por imagem , Diabetes Mellitus Tipo 2/diagnóstico por imagem , Células Secretoras de Insulina/ultraestrutura , Transplante das Ilhotas Pancreáticas/diagnóstico por imagem , Compostos Radiofarmacêuticos/química , Anticorpos de Domínio Único/química , 5-Hidroxitriptofano/química , 5-Hidroxitriptofano/farmacocinética , Animais , Biomarcadores/análise , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patologia , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Dipeptidil Peptidases e Tripeptidil Peptidases/genética , Dipeptidil Peptidases e Tripeptidil Peptidases/metabolismo , Exenatida/química , Exenatida/farmacocinética , Radioisótopos de Flúor/química , Radioisótopos de Flúor/farmacocinética , Humanos , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/transplante , Imageamento por Ressonância Magnética/métodos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada/métodos , Canais de Potássio/genética , Canais de Potássio/metabolismo , Compostos Radiofarmacêuticos/farmacocinética , Anticorpos de Domínio Único/metabolismo , ATPase Trocadora de Sódio-Potássio/genética , ATPase Trocadora de Sódio-Potássio/metabolismo , Tecnécio/química , Tecnécio/metabolismo , Tetrabenazina/análogos & derivados , Tetrabenazina/química , Tetrabenazina/farmacocinética , Tomografia Computadorizada de Emissão de Fóton Único/métodos
12.
Diabetologia ; 62(11): 1961-1968, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31414144

RESUMO

Reciprocal signalling between the endothelium and the pancreatic epithelium is crucial for coordinated differentiation of the embryonic endocrine and exocrine pancreas. In the adult pancreas, islets depend on their dense capillary network to adequately respond to changes in plasma glucose levels. Vascular changes contribute to the onset and progression of both type 1 and type 2 diabetes. Impaired revascularisation of islets transplanted in individuals with type 1 diabetes is linked to islet graft failure and graft loss. This review summarises our understanding of the role of vascular endothelial growth factor-A (VEGF-A) and endothelial cells in beta cell development, physiology and disease. In addition, the therapeutic potential of modulating VEGF-A levels in beta and beta-like cells for transplantation is discussed.


Assuntos
Vasos Sanguíneos/metabolismo , Células Endoteliais/citologia , Células Secretoras de Insulina/citologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Glicemia/análise , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Progressão da Doença , Homeostase , Humanos , Células Secretoras de Insulina/transplante , Ilhotas Pancreáticas/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Neovascularização Fisiológica , Pâncreas/metabolismo , Ratos , Regeneração , Transdução de Sinais
13.
J Cell Physiol ; 234(7): 10196-10204, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30387142

RESUMO

Diabetes mellitus is an autoimmune and chronic disorder that is rapidly expanding worldwide due to increasing obesity. In the current study, we were able to design a reliable 3-dimensional differentiation process of human Wharton's jelly mesenchymal stem cells into pancreatic beta cell precursors (PBCPs) and detected that transplanted PBCPs could improve hyperglycemia in a diabetes-induced model in mice. Polylactic acid/chitosan nanofibrous scaffold was prepared using an electrospinning method. Quantitative real-time reverse transcription-polymerase chain reaction and immunocytochemistry analysis were carried out to assess pancreatic marker expression in the differentiated cells. PBCPs were transplanted under the kidney capsule of diabetic mice that induced streptozotocin injection 14 days before the transplantation. Moreover, an intraperitoneal glucose tolerance test (ipGTT) was carried out 2 and 4 weeks after the transplantation to measure the reaction to a sudden increase of the blood glucose level in the transplanted animals. The results indicated that the expression of SRY (sex determining region Y)-box (Sox17), forkhead box A2 (FoxA2), pancreatic and duodenal homeobox 1 (Pdx1), neurogenin 3 (Ngn3), hepatic nuclear factor 4, alpha (Hnf4α), and NK2 homeobox 2 (Nkx2.2) were increased significantly in the differentiated cells compared with that of the control group. In the current study, the diabetic disease was confirmed by measuring blood glucose and proved by conducting some other behavioral tests. After the PBCPs transplantation in a diabetic model, the ipGTT and hyperglycemia investigation during the determinant times confirmed the disease's significant improvement in the experimental models. In this study, some preclinical data suggested that the transplantation of PBCPs associated with appropriate nanofiber scaffold can be utilized for the treatment of diabetes models. In addition, studies are required to elucidate the molecular mechanism of PBCPs acting in diabetes models before being used for patients with diabetes.


Assuntos
Diabetes Mellitus Tipo 1 , Células Secretoras de Insulina/transplante , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/citologia , Engenharia Tecidual/métodos , Alicerces Teciduais , Animais , Diferenciação Celular , Quitosana , Diabetes Mellitus Experimental , Proteína Homeobox Nkx-2.2 , Proteínas de Homeodomínio , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Nanofibras , Proteínas Nucleares , Poliésteres , Alicerces Teciduais/química , Fatores de Transcrição
14.
J Cell Physiol ; 234(6): 7811-7827, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30480819

RESUMO

Pancreatic ß-cells are destroyed by the immune system, in type 1 diabetes (T1D) and are impaired by glucose insensitivity in type 2 diabetes (T2D). Islet-cells transplantation is a promising therapeutic approach based on in vitro differentiation of pluripotent stem cells (PSCs) to insulin-producing cells (IPCs). According to evolutionary stages in ß-cell development, there are several distinct checkpoints; each one has a unique characteristic, including definitive endoderm (DE), primitive gut (PG), posterior foregut (PF), pancreatic epithelium (PE), endocrine precursor (EP), and immature ß-cells up to functional ß-cells. A better understanding of the gene regulatory networks (GRN) and associated transcription factors in each specific developmental stage, guarantees the achievement of the next successful checkpoints and ensures an efficient ß-cell differentiation procedure. The new findings in signaling pathways, related to the development of the pancreas are discussed here, including Wnt, Activin/Nodal, FGF, BMP, retinoic acid (RA), sonic hedgehog (Shh), Notch, and downstream regulators, required for ß-cell commitment. We also summarized different approaches in the IPCs protocol to conceptually define a standardized system, leading to the creation of a reproducible method for ß-cell differentiation. To normalize blood glucose level in diabetic mice, the replacement therapy in the early differentiation stage, such as EP stages was associated with better outcome when compared with the fully differentiated ß-cells' graft.


Assuntos
Diferenciação Celular/genética , Células Secretoras de Insulina/metabolismo , Pâncreas/crescimento & desenvolvimento , Células-Tronco Pluripotentes/metabolismo , Animais , Glicemia/genética , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/terapia , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/terapia , Redes Reguladoras de Genes/genética , Humanos , Células Secretoras de Insulina/transplante , Transplante das Ilhotas Pancreáticas , Camundongos , Camundongos Endogâmicos NOD , Pâncreas/metabolismo , Pâncreas/patologia , Células-Tronco Pluripotentes/transplante
15.
EMBO J ; 34(7): 841-55, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-25733347

RESUMO

The discovery of insulin more than 90 years ago introduced a life-saving treatment for patients with type 1 diabetes, and since then, significant progress has been made in clinical care for all forms of diabetes. However, no method of insulin delivery matches the ability of the human pancreas to reliably and automatically maintain glucose levels within a tight range. Transplantation of human islets or of an intact pancreas can in principle cure diabetes, but this approach is generally reserved for cases with simultaneous transplantation of a kidney, where immunosuppression is already a requirement. Recent advances in cell reprogramming and beta cell differentiation now allow the generation of personalized stem cells, providing an unlimited source of beta cells for research and for developing autologous cell therapies. In this review, we will discuss the utility of stem cell-derived beta cells to investigate the mechanisms of beta cell failure in diabetes, and the challenges to develop beta cell replacement therapies. These challenges include appropriate quality controls of the cells being used, the ability to generate beta cell grafts of stable cellular composition, and in the case of type 1 diabetes, protecting implanted cells from autoimmune destruction without compromising other aspects of the immune system or the functionality of the graft. Such novel treatments will need to match or exceed the relative safety and efficacy of available care for diabetes.


Assuntos
Reprogramação Celular/imunologia , Terapia de Imunossupressão , Células Secretoras de Insulina , Transplante das Ilhotas Pancreáticas/imunologia , Imunologia de Transplantes , Animais , Autoenxertos , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/patologia , Diabetes Mellitus Tipo 1/terapia , Humanos , Células Secretoras de Insulina/imunologia , Células Secretoras de Insulina/transplante , Transplante de Rim
16.
EMBO J ; 34(13): 1759-72, 2015 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-25908839

RESUMO

Directed differentiation of human pluripotent stem cells into functional insulin-producing beta-like cells holds great promise for cell replacement therapy for patients suffering from diabetes. This approach also offers the unique opportunity to study otherwise inaccessible aspects of human beta cell development and function in vitro. Here, we show that current pancreatic progenitor differentiation protocols promote precocious endocrine commitment, ultimately resulting in the generation of non-functional polyhormonal cells. Omission of commonly used BMP inhibitors during pancreatic specification prevents precocious endocrine formation while treatment with retinoic acid followed by combined EGF/KGF efficiently generates both PDX1(+) and subsequent PDX1(+)/NKX6.1(+) pancreatic progenitor populations, respectively. Precise temporal activation of endocrine differentiation in PDX1(+)/NKX6.1(+) progenitors produces glucose-responsive beta-like cells in vitro that exhibit key features of bona fide human beta cells, remain functional after short-term transplantation, and reduce blood glucose levels in diabetic mice. Thus, our simplified and scalable system accurately recapitulates key steps of human pancreas development and provides a fast and reproducible supply of functional human beta-like cells.


Assuntos
Técnicas de Cultura de Células/métodos , Diferenciação Celular , Células-Tronco Embrionárias/fisiologia , Células Secretoras de Insulina/fisiologia , Pâncreas/citologia , Animais , Glicemia/metabolismo , Células Cultivadas , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Experimental/terapia , Células-Tronco Embrionárias/citologia , Glucose/farmacologia , Humanos , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/transplante , Camundongos , Camundongos SCID , Camundongos Transgênicos , Estreptozocina
17.
J Intern Med ; 286(3): 309-316, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31108016

RESUMO

Since the establishment of the Seldinger technique for secure entry to the vascular system, there has been a rapid evolution in imaging and catheters that has made the arteries and veins internal routes to any place in the body for interventions. It is curious that a general exit from the vasculature in a similar manner has not been proposed earlier. Possibly, the simplest reason is that accidental perforation of the vasculature by guide wire or catheter is a feared adverse event in endovascular intervention. Most places in the body can be reached by ultrasonography or computed tomography-guided intervention. Some organs such as the central nervous system, the heart and pancreas are harder to access and, in some organs, like the kidney, repeated percutaneous punctions to cover large areas is not suitable. We present a new general purpose micro-endovascular device creating a working channel to these 'hard to reach' organs by an inverted Seldinger technique. This review details this trans-vessel wall technique, which has been studied in pancreas for transplantation of insulin-producing cells, for injection of contrast agent to the heart and to the brain, bowels and kidney in rat, rabbit, swine and macaque monkeys with up to one year of follow-up without adverse events. Furthermore, the payloads that can be given through such a system are briefly discussed. Drugs, cells, gene vectors and other therapeutic substances may be injected directly to the tissue to increase efficacy and decrease risk of off-site adverse effects.


Assuntos
Procedimentos Endovasculares/instrumentação , Células Secretoras de Insulina/transplante , Transplante de Órgãos/métodos , Animais , Vasos Sanguíneos , Desenho de Equipamento , Humanos , Macaca , Coelhos , Instrumentos Cirúrgicos , Suínos
18.
Small ; 15(50): e1904290, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31595687

RESUMO

Islet transplantation has been one promising strategy in diabetes treatment, which can maintain patient's insulin level long-term and avoid periodical insulin injections. However, donor shortage and temporal mismatch between donors and recipients has limited its widespread use. Therefore, searching for islet substitutes and developing efficient cryopreservation technology (providing potential islet bank for transplantation on demand) is in great need. Herein, a novel cryopreservation method is developed for islet ß cells by combining microfluidic encapsulation and cold-responsive nanocapsules (CR-NCs). The cryopreserved cell-laden hydrogels (calcium alginate hydrogel, CAH) can be transplanted for diabetes treatment. During the freezing process, trehalose is released inside ß cells through the CR-NCs and serves as the sole cryoprotectant (CPA). Additionally, CAH helps cells to survive the freeze-thaw process and provide cells with a natural immune barrier in vivo. Different from traditional cryopreservation methods, this method combining the CR-NCs and hydrogel encapsulation replaces the toxic CPAs with natural trehalose. Great preservation results are obtained and transplantation experiments of diabetic rats further prove the excellent glucose regulation ability of such ß cell-laden hydrogels post cryopreservation. This novel cryopreservation method helps to establish a reliable and ready-to-use bank of biological samples for transplantation therapy and other biomedical applications.


Assuntos
Temperatura Baixa , Criopreservação , Crioprotetores/farmacologia , Diabetes Mellitus Experimental/terapia , Hidrogéis/farmacologia , Células Secretoras de Insulina/transplante , Nanocápsulas/química , Trealose/farmacologia , Animais , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Células Secretoras de Insulina/efeitos dos fármacos , Masculino , Nanocápsulas/ultraestrutura , Ratos Sprague-Dawley , Testes de Toxicidade
19.
Diabet Med ; 36(3): 297-307, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30362170

RESUMO

Many people with Type 1 diabetes struggle with the burden of self-management and are unable to achieve optimal glycaemic control without risk of hypoglycaemia. Future therapies with the potential to reduce the risk for short- and long-term complications while simultaneously reducing the burden of diabetes are therefore attractive. ß-cell replacement is one strategy which might achieve this. Islet transplantation is limited by organ supply and the risks of long-term immunosuppression. Encapsulated stem-cell-derived ß cells have the potential to address both of these issues and phase I/II clinical trials of encapsulated pancreatic progenitors have begun. A significant risk associated with the translation of stem-cell science to the clinical management of Type 1 diabetes is an underestimation of the complexity of the process and a mismatch between the hype and the expectations of both people with Type 1 diabetes and the public. We provide an update on progress in clinical trials of encapsulated stem-cell-derived ß cells and propose a road map for the design and conduct of future trials to facilitate the translation of this exciting science to clinical care.


Assuntos
Ensaios Clínicos como Assunto/métodos , Diabetes Mellitus Tipo 1/terapia , Transplante de Células-Tronco/métodos , Ensaios Clínicos como Assunto/organização & administração , Ensaios Clínicos como Assunto/normas , Humanos , Células Secretoras de Insulina/transplante , Transplante das Ilhotas Pancreáticas , Projetos de Pesquisa/normas
20.
Curr Diab Rep ; 19(9): 75, 2019 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-31375935

RESUMO

PURPOSE OF REVIEW: Type 1 diabetes impacts 1.3 million people in the USA with a total direct lifetime medical cost of $133.7 billion. Management requires a mix of daily exogenous insulin administration and frequent glucose monitoring. Decision-making by the individual can be burdensome. RECENT FINDINGS: Beta-cell replacement, which involves devices protecting cells from autoimmunity and allo-rejection, aims at restoring physiological glucose regulation and improving clinical outcomes in patients. Given the significant burden of T1D in the healthcare systems, cost-effectiveness analyses can drive innovation and policymaking in the area. This review presents the health economics analyses performed for donor-derived islet transplantation and the possible outcomes of stem cell-derived beta cells. Long-term cost-effectiveness of islet transplantation depends on the engraftment of these transplants, and the expenses and thresholds assumed by healthcare systems in different countries. Early health technology assessment analyses for stem cell-derived beta-cell replacement suggest manufacturing optimization is necessary to reduce upfront costs.


Assuntos
Diabetes Mellitus Tipo 1/cirurgia , Células Secretoras de Insulina/transplante , Transplante das Ilhotas Pancreáticas/economia , Transplante das Ilhotas Pancreáticas/métodos , Glicemia/análise , Automonitorização da Glicemia , Análise Custo-Benefício , Diabetes Mellitus Tipo 1/sangue , Diabetes Mellitus Tipo 1/economia , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA