Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Diabetologia ; 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38743124

RESUMEN

AIMS/HYPOTHESIS: Regulatory factor X 6 (RFX6) is crucial for pancreatic endocrine development and differentiation. The RFX6 variant p.His293LeufsTer7 is significantly enriched in the Finnish population, with almost 1:250 individuals as a carrier. Importantly, the FinnGen study indicates a high predisposition for heterozygous carriers to develop type 2 and gestational diabetes. However, the precise mechanism of this predisposition remains unknown. METHODS: To understand the role of this variant in beta cell development and function, we used CRISPR technology to generate allelic series of pluripotent stem cells. We created two isogenic stem cell models: a human embryonic stem cell model; and a patient-derived stem cell model. Both were differentiated into pancreatic islet lineages (stem-cell-derived islets, SC-islets), followed by implantation in immunocompromised NOD-SCID-Gamma mice. RESULTS: Stem cell models of the homozygous variant RFX6-/- predictably failed to generate insulin-secreting pancreatic beta cells, mirroring the phenotype observed in Mitchell-Riley syndrome. Notably, at the pancreatic endocrine stage, there was an upregulation of precursor markers NEUROG3 and SOX9, accompanied by increased apoptosis. Intriguingly, heterozygous RFX6+/- SC-islets exhibited RFX6 haploinsufficiency (54.2% reduction in protein expression), associated with reduced beta cell maturation markers, altered calcium signalling and impaired insulin secretion (62% and 54% reduction in basal and high glucose conditions, respectively). However, RFX6 haploinsufficiency did not have an impact on beta cell number or insulin content. The reduced insulin secretion persisted after in vivo implantation in mice, aligning with the increased risk of variant carriers to develop diabetes. CONCLUSIONS/INTERPRETATION: Our allelic series isogenic SC-islet models represent a powerful tool to elucidate specific aetiologies of diabetes in humans, enabling the sensitive detection of aberrations in both beta cell development and function. We highlight the critical role of RFX6 in augmenting and maintaining the pancreatic progenitor pool, with an endocrine roadblock and increased cell death upon its loss. We demonstrate that RFX6 haploinsufficiency does not affect beta cell number or insulin content but does impair function, predisposing heterozygous carriers of loss-of-function variants to diabetes. DATA AVAILABILITY: Ultra-deep bulk RNA-seq data for pancreatic differentiation stages 3, 5 and 7 of H1 RFX6 genotypes are deposited in the Gene Expression Omnibus database with accession code GSE234289. Original western blot images are deposited at Mendeley ( https://data.mendeley.com/datasets/g75drr3mgw/2 ).

2.
Cell Rep ; 42(8): 112970, 2023 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-37556323

RESUMEN

Pancreatic islets regulate blood glucose homeostasis through the controlled release of insulin; however, current metabolic models of glucose-sensitive insulin secretion are incomplete. A comprehensive understanding of islet metabolism is integral to studies of endocrine cell development as well as diabetic islet dysfunction. Human pluripotent stem cell-derived islets (SC-islets) are a developmentally relevant model of human islet function that have great potential in providing a cure for type 1 diabetes. Using multiple 13C-labeled metabolic fuels, we demonstrate that SC-islets show numerous divergent patterns of metabolite trafficking in proposed insulin release pathways compared with primary human islets but are still reliant on mitochondrial aerobic metabolism to derive function. Furthermore, reductive tricarboxylic acid cycle activity and glycolytic metabolite cycling occur in SC-islets, suggesting that non-canonical coupling factors are also present. In aggregate, we show that many facets of SC-islet metabolism overlap with those of primary islets, albeit with a retained immature signature.

3.
Nat Biotechnol ; 40(7): 1042-1055, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35241836

RESUMEN

Transplantation of pancreatic islet cells derived from human pluripotent stem cells is a promising treatment for diabetes. Despite progress in the generation of stem-cell-derived islets (SC-islets), no detailed characterization of their functional properties has been conducted. Here, we generated functionally mature SC-islets using an optimized protocol and benchmarked them comprehensively against primary adult islets. Biphasic glucose-stimulated insulin secretion developed during in vitro maturation, associated with cytoarchitectural reorganization and the increasing presence of alpha cells. Electrophysiology, signaling and exocytosis of SC-islets were similar to those of adult islets. Glucose-responsive insulin secretion was achieved despite differences in glycolytic and mitochondrial glucose metabolism. Single-cell transcriptomics of SC-islets in vitro and throughout 6 months of engraftment in mice revealed a continuous maturation trajectory culminating in a transcriptional landscape closely resembling that of primary islets. Our thorough evaluation of SC-islet maturation highlights their advanced degree of functionality and supports their use in further efforts to understand and combat diabetes.


Asunto(s)
Trasplante de Islotes Pancreáticos , Islotes Pancreáticos , Células Madre Pluripotentes , Animales , Glucosa/metabolismo , Humanos , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Trasplante de Islotes Pancreáticos/métodos , Ratones , Células Madre Pluripotentes/metabolismo
4.
Elife ; 72018 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-30412052

RESUMEN

Insulin gene mutations are a leading cause of neonatal diabetes. They can lead to proinsulin misfolding and its retention in endoplasmic reticulum (ER). This results in increased ER-stress suggested to trigger beta-cell apoptosis. In humans, the mechanisms underlying beta-cell failure remain unclear. Here we show that misfolded proinsulin impairs developing beta-cell proliferation without increasing apoptosis. We generated induced pluripotent stem cells (iPSCs) from people carrying insulin (INS) mutations, engineered isogenic CRISPR-Cas9 mutation-corrected lines and differentiated them to beta-like cells. Single-cell RNA-sequencing analysis showed increased ER-stress and reduced proliferation in INS-mutant beta-like cells compared with corrected controls. Upon transplantation into mice, INS-mutant grafts presented reduced insulin secretion and aggravated ER-stress. Cell size, mTORC1 signaling, and respiratory chain subunits expression were all reduced in INS-mutant beta-like cells, yet apoptosis was not increased at any stage. Our results demonstrate that neonatal diabetes-associated INS-mutations lead to defective beta-cell mass expansion, contributing to diabetes development.


Asunto(s)
Diabetes Mellitus/genética , Estrés del Retículo Endoplásmico/genética , Células Madre Pluripotentes Inducidas/química , Proinsulina/genética , Animales , Apoptosis/genética , Sistemas CRISPR-Cas/genética , Diferenciación Celular/genética , Proliferación Celular/genética , Diabetes Mellitus/patología , Retículo Endoplásmico/genética , Femenino , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Recién Nacido , Células Secretoras de Insulina/química , Células Secretoras de Insulina/metabolismo , Masculino , Ratones , Mutación , Proinsulina/química , Pliegue de Proteína , Análisis de Secuencia de ARN , Transducción de Señal , Análisis de la Célula Individual
5.
Diabetologia ; 61(10): 2202-2214, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30032427

RESUMEN

AIMS/HYPOTHESIS: There is a great need to identify factors that could protect pancreatic beta cells against apoptosis or stimulate their replication and thus prevent or reverse the development of diabetes. One potential candidate is mesencephalic astrocyte-derived neurotrophic factor (MANF), an endoplasmic reticulum (ER) stress inducible protein. Manf knockout mice used as a model of diabetes develop the condition because of increased apoptosis and reduced proliferation of beta cells, apparently related to ER stress. Given this novel association between MANF and beta cell death, we studied the potential of MANF to protect human beta cells against experimentally induced ER stress. METHODS: Primary human islets were challenged with proinflammatory cytokines, with or without MANF. Cell viability was analysed and global transcriptomic analysis performed. Results were further validated using the human beta cell line EndoC-ßH1. RESULTS: There was increased expression and secretion of MANF in human beta cells in response to cytokines. Addition of recombinant human MANF reduced cytokine-induced cell death by 38% in human islets (p < 0.05). MANF knockdown in EndoC-ßH1 cells led to increased ER stress after cytokine challenge. Mechanistic studies showed that the protective effect of MANF was associated with repression of the NF-κB signalling pathway and amelioration of ER stress. MANF also increased the proliferation of primary human beta cells twofold when TGF-ß signalling was inhibited (p < 0.01). CONCLUSIONS/INTERPRETATION: Our studies show that exogenous MANF protein can provide protection to human beta cells against death induced by inflammatory stress. The antiapoptotic and mitogenic properties of MANF make it a potential therapeutic agent for beta cell protection.


Asunto(s)
Estrés del Retículo Endoplásmico , Retículo Endoplásmico/metabolismo , Células Secretoras de Insulina/citología , Factores de Crecimiento Nervioso/metabolismo , Astrocitos/metabolismo , Muerte Celular/efectos de los fármacos , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Citocinas/metabolismo , Humanos , Inflamación , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/citología , FN-kappa B/metabolismo , ARN Interferente Pequeño/metabolismo , Proteínas Recombinantes/metabolismo , Transducción de Señal , Transcriptoma
6.
Cell Rep ; 19(2): 281-294, 2017 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-28402852

RESUMEN

Activating germline mutations in STAT3 were recently identified as a cause of neonatal diabetes mellitus associated with beta-cell autoimmunity. We have investigated the effect of an activating mutation, STAT3K392R, on pancreatic development using induced pluripotent stem cells (iPSCs) derived from a patient with neonatal diabetes and pancreatic hypoplasia. Early pancreatic endoderm differentiated similarly from STAT3K392R and healthy-control cells, but in later stages, NEUROG3 expression was upregulated prematurely in STAT3K392R cells together with insulin (INS) and glucagon (GCG). RNA sequencing (RNA-seq) showed robust NEUROG3 downstream targets upregulation. STAT3 mutation correction with CRISPR/Cas9 reversed completely the disease phenotype. STAT3K392R-activating properties were not explained fully by altered DNA-binding affinity or increased phosphorylation. Instead, reporter assays demonstrated NEUROG3 promoter activation by STAT3 in pancreatic cells. Furthermore, proteomic and immunocytochemical analyses revealed increased nuclear translocation of STAT3K392R. Collectively, our results demonstrate that the STAT3K392R mutation causes premature endocrine differentiation through direct induction of NEUROG3 expression.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Diferenciación Celular/genética , Diabetes Mellitus/genética , Proteínas del Tejido Nervioso/biosíntesis , Factor de Transcripción STAT3/genética , Autoinmunidad/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Sistemas CRISPR-Cas , Línea Celular , Diabetes Mellitus/etiología , Diabetes Mellitus/patología , Regulación del Desarrollo de la Expresión Génica , Glucagón/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Insulina/genética , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Mutación , Proteínas del Tejido Nervioso/genética , Regiones Promotoras Genéticas , Factor de Transcripción STAT3/biosíntesis
7.
Sci Rep ; 6: 24992, 2016 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-27114183

RESUMEN

Protection or restoration of pancreatic ß-cell mass as a therapeutic treatment for type 1 diabetes requires understanding of the mechanisms that drive the specification and development of pancreatic endocrine cells. Septins are filamentous small GTPases that function in the regulation of cell division, cytoskeletal organization and membrane remodeling, and are involved in various tissue-specific developmental processes. However, their role in pancreatic endocrine cell differentiation remains unknown. Here we show by functional manipulation techniques in transgenic zebrafish lines that suppression of sept7b, the zebrafish ortholog of human SEPT7, profoundly increases the number of endocrine progenitors but limits their differentiation, leading to reduction in ß- and α-cell mass. Furthermore, we discovered that shh (sonic hedgehog) expression in the endoderm, essential for the development of pancreatic progenitors of the dorsal pancreatic bud, is absent in larvae depleted of sept7b. We also discovered that sept7b is important for the differentiation of ventral pancreatic bud-derived cells: sept7b-depleted larvae exhibit downregulation of Notch receptors notch1a and notch1b and show precocious differentiation of NeuroD-positive endocrine cells in the intrapancreatic duct and gut epithelium. Collectively, this study provides a novel insight into the development of pancreatic endocrine progenitors, revealing an essential role for sept7b in endocrine progenitor differentiation.


Asunto(s)
Islotes Pancreáticos/citología , Septinas/genética , Septinas/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Pez Cebra/crecimiento & desarrollo , Animales , Animales Modificados Genéticamente , Diferenciación Celular , Regulación del Desarrollo de la Expresión Génica , Técnicas de Inactivación de Genes , Células Secretoras de Glucagón/citología , Células Secretoras de Glucagón/metabolismo , Proteínas Hedgehog/metabolismo , Proteínas de Homeodominio/metabolismo , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptor Notch1/metabolismo , Pez Cebra/genética , Pez Cebra/metabolismo
8.
Biomed Res Int ; 2015: 102969, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26000279

RESUMEN

The transgenic E1-DN mice express a kinase-negative epidermal growth factor receptor in their pancreatic islets and are diabetic from two weeks of age due to impaired postnatal growth of ß-cell mass. Here, we characterize the development of hyperglycaemia-induced renal injury in the E1-DN mice. Homozygous mice showed increased albumin excretion rate (AER) at the age of 10 weeks; the albuminuria increased over time and correlated with blood glucose. Morphometric analysis of PAS-stained histological sections and electron microscopy images revealed mesangial expansion in homozygous E1-DN mice, and glomerular sclerosis was observed in the most hyperglycaemic mice. The albuminuric homozygous mice developed also other structural changes in the glomeruli, including thickening of the glomerular basement membrane and widening of podocyte foot processes that are typical for diabetic nephropathy. Increased apoptosis of podocytes was identified as one mechanism contributing to glomerular injury. In addition, nephrin expression was reduced in the podocytes of albuminuric homozygous E1-DN mice. Tubular changes included altered epithelial cell morphology and increased proliferation. In conclusion, hyperglycaemic E1-DN mice develop albuminuria and glomerular and tubular injury typical of human diabetic nephropathy and can serve as a new model to study the mechanisms leading to the development of diabetic nephropathy.


Asunto(s)
Nefropatías Diabéticas/etiología , Albuminuria/etiología , Animales , Proteínas de la Cápside , Nefropatías Diabéticas/patología , Nefropatías Diabéticas/fisiopatología , Modelos Animales de Enfermedad , Receptores ErbB/genética , Homocigoto , Humanos , Glomérulos Renales/patología , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Podocitos/patología
9.
Cell Rep ; 7(2): 366-375, 2014 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-24726366

RESUMEN

All forms of diabetes mellitus (DM) are characterized by the loss of functional pancreatic ß cell mass, leading to insufficient insulin secretion. Thus, identification of novel approaches to protect and restore ß cells is essential for the development of DM therapies. Mesencephalic astrocyte-derived neurotrophic factor (MANF) is an endoplasmic reticulum (ER)-stress-inducible protein, but its physiological role in mammals has remained obscure. We generated MANF-deficient mice that strikingly develop severe diabetes due to progressive postnatal reduction of ß cell mass, caused by decreased proliferation and increased apoptosis. Additionally, we show that lack of MANF in vivo in mouse leads to chronic unfolded protein response (UPR) activation in pancreatic islets. Importantly, MANF protein enhanced ß cell proliferation in vitro and overexpression of MANF in the pancreas of diabetic mice enhanced ß cell regeneration. We demonstrate that MANF specifically promotes ß cell proliferation and survival, thereby constituting a therapeutic candidate for ß cell protection and regeneration.


Asunto(s)
Proliferación Celular , Diabetes Mellitus Experimental/metabolismo , Células Secretoras de Insulina/metabolismo , Factores de Crecimiento Nervioso/metabolismo , Animales , Apoptosis , Supervivencia Celular , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patología , Células Secretoras de Insulina/fisiología , Ratones , Ratones Endogámicos C57BL , Factores de Crecimiento Nervioso/genética , Especificidad de Órganos , Respuesta de Proteína Desplegada
10.
PLoS One ; 9(4): e93651, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24695557

RESUMEN

Placental lactogen (PL) induced serotonergic signaling is essential for gestational ß-cell mass expansion. We have previously shown that intact Epidermal growth factor -receptor (EGFR) function is a crucial component of this pathway. We now explored more specifically the link between EGFR and pregnancy-induced ß-cell mass compensation. Islets were isolated from wild-type and ß-cell-specific EGFR-dominant negative mice (E1-DN), stimulated with PL and analyzed for ß-cell proliferation and expression of genes involved in gestational ß-cell growth. ß-cell mass dynamics were analyzed both with traditional morphometrical methods and three-dimensional optical projection tomography (OPT) of whole-mount insulin-stained pancreata. Insulin-positive volume analyzed with OPT increased 1.4-fold at gestational day 18.5 (GD18.5) when compared to non-pregnant mice. Number of islets peaked by GD13.5 (680 vs 1134 islets per pancreas, non-pregnant vs. GD13.5). PL stimulated beta cell proliferation in the wild-type islets, whereas the proliferative response was absent in the E1-DN mouse islets. Serotonin synthesizing enzymes were upregulated similarly in both the wild-type and E1-DN mice. However, while survivin (Birc5) mRNA was upregulated 5.5-fold during pregnancy in the wild-type islets, no change was seen in the E1-DN pregnant islets. PL induced survivin expression also in isolated islets and this was blocked by EGFR inhibitor gefitinib, mTOR inhibitor rapamycin and MEK inhibitor PD0325901. Our 3D-volumetric analysis of ß-cell mass expansion during murine pregnancy revealed that islet number increases during pregnancy. In addition, our results suggest that EGFR signaling is required for lactogen-induced survivin expression via MAPK and mTOR pathways.


Asunto(s)
Proliferación Celular , Receptores ErbB/metabolismo , Proteínas Inhibidoras de la Apoptosis/metabolismo , Islotes Pancreáticos/citología , Proteínas Represoras/metabolismo , Transducción de Señal , Animales , Femenino , Islotes Pancreáticos/metabolismo , Quinasas Quinasa Quinasa PAM/metabolismo , Ratones , Embarazo , Reacción en Cadena en Tiempo Real de la Polimerasa , Survivin , Serina-Treonina Quinasas TOR/metabolismo , Regulación hacia Arriba
11.
Diabetologia ; 57(5): 970-9, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24493201

RESUMEN

AIMS/HYPOTHESIS: EGF receptor (EGFR) signalling is required for normal beta cell development and postnatal beta cell proliferation. We tested whether beta cell proliferation can be triggered by EGFR activation at any age and whether this can protect beta cells against apoptosis induced by diabetogenic insults in a mouse model. METHODS: We generated transgenic mice with doxycycline-inducible expression of constitutively active EGFR (L858R) (CA-EGFR) under the insulin promoter. Mice were given doxycycline at various ages for different time periods, and beta cell proliferation and mass were analysed. Mice were also challenged with streptozotocin and isolated islets exposed to cytokines. RESULTS: Expression of EGFR (L858R) led to increased phosphorylation of EGFR and Akt in pancreatic islets. CA-EGFR expression during pancreatic development (embryonic day [E]12.5 to postnatal day [P]1) increased beta cell proliferation and mass in newborn mice. However, CA-EGFR expression in adult mice did not affect beta cell mass. Expression of the transgene improved glycaemia and markedly inhibited beta cell apoptosis after a single high dose, as well as after multiple low doses of streptozotocin. In vitro mechanistic studies showed that CA-EGFR protected isolated islets from cytokine-mediated beta cell death, possibly by repressing the proapoptotic protein BCL2-like 11 (BIM). CONCLUSIONS/INTERPRETATION: Our findings show that the expression of CA-EGFR in the developing, but not in the adult pancreas stimulates beta cell replication and leads to increased beta cell mass. Importantly, CA-EGFR protects beta cells against streptozotocin- and cytokine-induced death.


Asunto(s)
Diabetes Mellitus/genética , Receptores ErbB/genética , Células Secretoras de Insulina/metabolismo , Páncreas/embriología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Apoptosis , Glucemia/análisis , Muerte Celular , Proliferación Celular , Supervivencia Celular , Diabetes Mellitus/enzimología , Activación Enzimática , Homeostasis , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Fosforilación , Transducción de Señal
12.
PLoS One ; 8(10): e76205, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24098444

RESUMEN

Correct interactions with extracellular matrix are essential to human pluripotent stem cells (hPSC) to maintain their pluripotent self-renewal capacity during in vitro culture. hPSCs secrete laminin 511/521, one of the most important functional basement membrane components, and they can be maintained on human laminin 511 and 521 in defined culture conditions. However, large-scale production of purified or recombinant laminin 511 and 521 is difficult and expensive. Here we have tested whether a commonly available human choriocarcinoma cell line, JAR, which produces high quantities of laminins, supports the growth of undifferentiated hPSCs. We were able to maintain several human pluripotent stem cell lines on decellularized matrix produced by JAR cells using a defined culture medium. The JAR matrix also supported targeted differentiation of the cells into neuronal and hepatic directions. Importantly, we were able to derive new human induced pluripotent stem cell (hiPSC) lines on JAR matrix and show that adhesion of the early hiPSC colonies to JAR matrix is more efficient than to matrigel. In summary, JAR matrix provides a cost-effective and easy-to-prepare alternative for human pluripotent stem cell culture and differentiation. In addition, this matrix is ideal for the efficient generation of new hiPSC lines.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes/citología , Técnicas de Cultivo de Célula , Diferenciación Celular , Línea Celular , Coriocarcinoma/genética , Coriocarcinoma/metabolismo , Coriocarcinoma/patología , Células Madre Embrionarias , Matriz Extracelular/metabolismo , Células Nutrientes , Hepatocitos/citología , Hepatocitos/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Laminina/metabolismo , Neuronas/citología , Neuronas/metabolismo , Células Madre Pluripotentes/metabolismo
13.
Diabetes ; 62(11): 3797-806, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23903354

RESUMEN

Loss-of-function mutations in the KATP channel genes KCNJ11 and ABCC8 cause neonatal hyperinsulinism in humans. Dominantly inherited mutations cause less severe disease, which may progress to glucose intolerance and diabetes in later life (e.g., SUR1-E1506K). We generated a mouse expressing SUR1-E1506K in place of SUR1. KATP channel inhibition by MgATP was enhanced in both homozygous (homE1506K) and heterozygous (hetE1506K) mutant mice, due to impaired channel activation by MgADP. As a consequence, mutant ß-cells showed less on-cell KATP channel activity and fired action potentials in glucose-free solution. HomE1506K mice exhibited enhanced insulin secretion and lower fasting blood glucose within 8 weeks of birth, but reduced insulin secretion and impaired glucose tolerance at 6 months of age. These changes correlated with a lower insulin content; unlike wild-type or hetE1506K mice, insulin content did not increase with age in homE1506K mice. There was no difference in the number and size of islets or ß-cells in the three types of mice, or evidence of ß-cell proliferation. We conclude that the gradual development of glucose intolerance in patients with the SUR1-E1506K mutation might, as in the mouse model, result from impaired insulin secretion due a failure of insulin content to increase with age.


Asunto(s)
Hiperinsulinismo/genética , Islotes Pancreáticos/fisiopatología , Receptores de Sulfonilureas/genética , Envejecimiento/fisiología , Animales , Glucemia/metabolismo , Calcio/metabolismo , Modelos Animales de Enfermedad , Heterocigoto , Homocigoto , Humanos , Insulina/metabolismo , Secreción de Insulina , Canales KATP/fisiología , Ratones , Bloqueadores de los Canales de Potasio/farmacología
14.
Exp Cell Res ; 319(17): 2535-44, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-23954819

RESUMEN

Activin/Nodal and Wnt signaling are known to play important roles in the regional specification of endoderm. Here we have investigated the effect of the length of stimulation with Activin A plus Wnt3a on the development of hepatic and pancreatic progenitors from the definitive endoderm (DE) cells derived from human pluripotent stem cells (hPSC). We show that DE-cells derived from hPSC with 3 days high Activin A and Wnt3a treatment were able to differentiate further into both tested endodermal lineages. When prolonging the DE-induction protocol from 3 to 5 or 7 days, almost pure DE-marker positive cell populations were obtained. However, these cells had an impaired pancreatic differentiation capacity, while they still developed into hepatocyte-like cells. Further propagation of the DE-cells in the presence of Wnt3a and Activin A led to the complete loss of differentiation capacity into hepatic or pancreatic lineages. When Wnt3a was removed after 24h from the initiation of the differentiation, the cells were able to differentiate into PDX1+/NKX6.1+ pancreatic progenitors even with longer DE induction time while efficiency of hepatic differentiation was lower. Our results suggest that both the length and the timing of Wnt3a treatment during DE induction are crucial for the final differentiation outcome. Although it is possible to derive apparently pure DE cells with prolonged Activin A/Wnt-stimulation, their progenitor capacity is restricted to a limited time window.


Asunto(s)
Activinas/farmacología , Diferenciación Celular/efectos de los fármacos , Endodermo/citología , Proteína Wnt3A/farmacología , Linaje de la Célula , Inducción Embrionaria , Hepatocitos/citología , Hepatocitos/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Páncreas/citología , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Transactivadores/genética , Transactivadores/metabolismo
15.
Pediatr Res ; 62(5): 570-5, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17805209

RESUMEN

Concomitant use of hydrocortisone and the nonspecific cyclo-oxygenase (COX)-inhibitor indomethacin increases the risk for intestinal perforations in preterm infants. We determined whether this was associated with insufficient epidermal growth factor receptor (EGF-R) signaling. We tested the effect of EGF, hydrocortisone, and indomethacin on its activation, cell proliferation and migration, COX-2 expression, and prostaglandin E2 (PGE2) production. Human small intestine epithelial cell line FHsInt74 and EGF-R-deficient mice [EGF-R (-/-)] were used as models. The data revealed that EGF-R signaling had a bimodal positive effect on fetal enterocyte: 1) it increased cell proliferation and migration synergistically with hydrocortisone and 2) up-regulated COX-2 mRNA expression and subsequent PGE2 production. Correlating with this, COX-2 protein expression was down-regulated in EGF-R (-/-) intestine. Despite a positive effect on cell proliferation with EGF, hydrocortisone blunted the stimulatory effect of EGF on COX-2 expression and PGE2 production. Addition of indomethacin even further inhibited the EGF-stimulated PGE2 synthesis. The data suggest that concomitant use of indomethacin and hydrocortisone on preterm infants, who physiologically synthesize only low levels of EGF-R ligands, may lead to intestinal problems related to failure in cytoprotective and regenerative events.


Asunto(s)
Inhibidores de la Ciclooxigenasa/farmacología , Enterocitos/efectos de los fármacos , Receptores ErbB/metabolismo , Hidrocortisona/farmacología , Indometacina/farmacología , Intestino Delgado/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Animales , Línea Celular , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Dinoprostona/metabolismo , Enterocitos/enzimología , Enterocitos/metabolismo , Factor de Crecimiento Epidérmico/metabolismo , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Humanos , Intestino Delgado/citología , Intestino Delgado/embriología , Intestino Delgado/metabolismo , Ratones , Ratones Noqueados , ARN Mensajero/metabolismo , Regulación hacia Arriba
16.
Mol Cell Endocrinol ; 264(1-2): 28-34, 2007 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-17116362

RESUMEN

The long-term function of human pancreatic islet grafts may depend on the neogenesis of beta cells from epithelial precursors within the grafted tissue. We have developed an in vitro model for human islet neogenesis. In this study, we have investigated the morphological signs of maturation in cultivated human islet buds (CHIBs) before and after transplantation. Clusterin is a molecule associated with beta-cell differentiation in rodents. In adult human islets, clusterin expression was located only in alpha- and PP-cells, but in CHIBs and human fetal islets, it was distributed in all four types of endocrine cells. Some immature endocrine cells in the CHIBs co-expressed insulin and glucagon. After transplantation, CHIBs became mature with one type of hormone per endocrine cell, and clusterin expression became restricted in alpha-cells. Cells co-expressing endocrine markers and cytokeratin 19, as a sign of ductal to endocrine cell transition, were frequently detected in both fresh islets and CHIBs after transplantation. We conclude that clusterin may be involved in the development of islets, and the in vitro-derived islets become mature after transplantation into nude mice. Ductal cell differentiation into endocrine cells may be an important factor in sustaining the long-term function of islet transplants.


Asunto(s)
Antígenos de Diferenciación/biosíntesis , Diferenciación Celular/fisiología , Regulación de la Expresión Génica/fisiología , Trasplante de Islotes Pancreáticos , Islotes Pancreáticos/metabolismo , Animales , Clusterina/biosíntesis , Glucagón/biosíntesis , Humanos , Insulina/biosíntesis , Islotes Pancreáticos/citología , Queratina-19/biosíntesis , Ratones , Ratones Desnudos , Ratas , Factores de Tiempo , Trasplante Heterólogo
17.
Diabetes ; 55(12): 3299-308, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17130473

RESUMEN

Epidermal growth factor receptor (EGF-R) signaling is essential for proper fetal development and growth of pancreatic islets, and there is also evidence for its involvement in beta-cell signal transduction in the adult. To study the functional roles of EGF-R in beta-cell physiology in postnatal life, we have generated transgenic mice that carry a mutated EGF-R under the pancreatic duodenal homeobox-1 promoter (E1-DN mice). The transgene was expressed in islet beta- and delta-cells but not in alpha-cells, as expected, and it resulted in an approximately 40% reduction in pancreatic EGF-R, extracellular signal-related kinase, and Akt phosphorylation. Homozygous E1-DN mice were overtly diabetic after the age of 2 weeks. The hyperglycemia was more pronounced in male than in female mice. The relative beta-cell surface area of E1-DN mice was highly reduced at the age of 2 months, while alpha-cell surface area was not changed. This defect was essentially postnatal, since the differences in beta-cell area of newborn mice were much smaller. An apparent explanation for this is impaired postnatal beta-cell proliferation; the normal surge of beta-cell proliferation during 2 weeks after birth was totally abolished in the transgenic mice. Heterozygous E1-DN mice were glucose intolerant in intraperitoneal glucose tests. This was associated with a reduced insulin response. However, downregulation of EGF-R signaling had no influence on the insulinotropic effect of glucagon-like peptide-1 analog exendin-4. In summary, our results show that even a modest attenuation of EGF-R signaling leads to a severe defect in postnatal growth of the beta-cells, which leads to the development of diabetes.


Asunto(s)
Receptores ErbB/fisiología , Células Secretoras de Insulina/fisiología , Islotes Pancreáticos/fisiología , Animales , Regulación hacia Abajo , Receptores ErbB/deficiencia , Receptores ErbB/genética , Humanos , Células Secretoras de Insulina/patología , Hígado/fisiología , Ratones , Ratones Transgénicos , Transducción de Señal
18.
BMC Dev Biol ; 6: 40, 2006 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-16895598

RESUMEN

BACKGROUND: Individual differences between human embryonic stem cell (hESC) lines are poorly understood. Here, we describe the derivation of five hESC lines (called FES 21, 22, 29, 30 and 61) from frozen-thawed human embryos and compare their individual differentiation characteristic. RESULTS: The cell lines were cultured either on human or mouse feeder cells. The cells grew significantly faster and could be passaged enzymatically only on mouse feeders. However, this was found to lead to chromosomal instability after prolonged culture. All hESC lines expressed the established markers of pluripotent cells as well as several primordial germ cell (PGC) marker genes in a uniform manner. However, the cell lines showed distinct features in their spontaneous differentiation patterns. The embryoid body (EB) formation frequency of FES 30 cell line was significantly lower than that of other lines and cells within the EBs differentiated less readily. Likewise, teratomas derived from FES 30 cells were constantly cystic and showed only minor solid tissue formation with a monotonous differentiation pattern as compared with the other lines. CONCLUSION: hESC lines may differ substantially in their differentiation properties although they appear similar in the undifferentiated state.


Asunto(s)
Diferenciación Celular , Línea Celular , Embrión de Mamíferos/citología , Inducción Embrionaria/fisiología , Células Madre/metabolismo , Animales , Biomarcadores/análisis , Técnicas de Cultivo de Célula/métodos , Células Germinativas/metabolismo , Células Germinativas/ultraestructura , Humanos , Cariotipificación , Masculino , Ratones , Células Madre/ultraestructura , Teratoma/patología
19.
Diabetes ; 52(8): 2007-15, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12882917

RESUMEN

We have reproduced a previously described method for the in vitro generation of endocrine cells in adult human pancreatic tissue culture. The aim of this study was to characterize the nature of pancreatic progenitor cells and to identify the factors necessary for their differentiation in this model. During monolayer expansion, two types of cells proliferated sequentially; first cytokeratin 19 (CK19)-positive ductal epithelial cells and then nestin-positive fibroblastoid cells. After the bromodeoxyuridine-labeled cells were traced in differentiated islet buds, some of the proliferating ductal cells had differentiated into endocrine cells, whereas nestin-positive cells could not give rise to endocrine tissue. Serum-free culture was found to be an absolute requirement for the endocrine differentiation to occur. Also, overlay of the cells with Matrigel was essential, whereas nicotinamide had a potentiating effect. The in vitro-generated islet buds released insulin in response to glucose nearly as efficiently as native islets. When transplanted under the kidney capsule of nude mice, only one of five grafts demonstrated further growth with foci of both endocrine and exocrine differentiation. Our results support the previous notion that pancreatic progenitor cells represent a subpopulation of ductal epithelial cells. No evidence was found for the development of endocrine cells from nestin-positive stem cells.


Asunto(s)
Trasplante de Islotes Pancreáticos , Islotes Pancreáticos/citología , Proteínas del Tejido Nervioso , Células Madre/citología , Adulto , Animales , Materiales Biocompatibles , Diferenciación Celular/fisiología , División Celular/fisiología , Células Cultivadas , Colágeno , Combinación de Medicamentos , Células Epiteliales/química , Células Epiteliales/citología , Expresión Génica/fisiología , Glucagón/genética , Humanos , Insulina/genética , Insulina/metabolismo , Proteínas de Filamentos Intermediarios/análisis , Islotes Pancreáticos/química , Islotes Pancreáticos/metabolismo , Queratinas/análisis , Laminina , Ratones , Ratones Desnudos , Nestina , Proteoglicanos , Células Madre/química
20.
Diabetes ; 52(1): 199-204, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12502513

RESUMEN

We have identified patients in whom strenuous physical exercise leads to hypoglycemia caused by inappropriate insulin release (exercise-induced hyperinsulinism [EIHI]). The aim of the present study was to test the hypothesis that the increased levels of lactate and/or pyruvate during anaerobic exercise would trigger the aberrant insulin secretion in these patients. A total of 12 patients (8 women and 4 men from two families) were diagnosed with EIHI, based on hypoglycemia and a more than threefold increase in plasma insulin induced by a 10-min bicycle exercise test. The mode of inheritance was autosomal dominant in these families. The acute response of insulin release to a bolus of intravenous pyruvate (13.9 mmol/1.73 m(2)) was studied in the patients and eight healthy control subjects. Insulin secretion did not respond to the pyruvate bolus in healthy control subjects. However, all EIHI patients responded to pyruvate, displaying a brisk increase in plasma insulin. The 1 + 3-min peak response was 5.6-fold in the patients and 0.9-fold in the control subjects (P < 0.001). To test the hypothesis that the pathogenesis of EIHI would involve monocarboxylate transport or metabolism in the beta-cell, we sequenced the genes encoding the known monocarboxylate transporter proteins and tested the transport of pyruvate into patient fibroblasts. The results revealed normal coding sequences and pyruvate transport. In conclusion, EIHI represents a new autosomal-dominant hyperinsulinemia syndrome that may be more common than has been realized. The pyruvate test provides a simple, safe, and specific diagnostic test for this condition.


Asunto(s)
Ejercicio Físico/fisiología , Fibroblastos/metabolismo , Genes Dominantes , Hiperinsulinismo/sangre , Hiperinsulinismo/genética , Hipoglucemia/sangre , Hipoglucemia/genética , Insulina/metabolismo , Ácido Pirúvico/metabolismo , Adulto , Transporte Biológico , Prueba de Esfuerzo , Femenino , Humanos , Secreción de Insulina , L-Lactato Deshidrogenasa/metabolismo , Masculino , Persona de Mediana Edad , Transportadores de Ácidos Monocarboxílicos/genética , Linaje
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA