Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 204
Filtrar
1.
Biochim Biophys Acta Mol Cell Res ; 1869(5): 119235, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35151663

RESUMEN

Glucose homeostasis is maintained by hormones secreted from different types of pancreatic islets and its dysregulation can result in diseases including diabetes mellitus. The secretion of hormones from pancreatic islets is highly complex and tightly controlled by G protein-coupled receptors (GPCRs). Moreover, GPCR signaling may play a role in enhancing islet cell replication and proliferation. Thus, targeting GPCRs offers a promising strategy for regulating the functionality of pancreatic islets. Here, available RNAseq datasets from human and mouse islets were used to identify the GPCR expression profile and the impact of GPCR signaling for normal islet functionality is discussed.


Asunto(s)
Islotes Pancreáticos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Células Secretoras de Glucagón/citología , Células Secretoras de Glucagón/metabolismo , Humanos , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/citología , Islotes Pancreáticos/crecimiento & desarrollo , Células Secretoras de Polipéptido Pancreático/citología , Células Secretoras de Polipéptido Pancreático/metabolismo , Receptores Acoplados a Proteínas G/genética , Transducción de Señal , Transcriptoma
2.
Nutrients ; 13(7)2021 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-34209449

RESUMEN

Interactions between endocrine α and ß cells are critical to their secretory function in vivo. The interactions are highly regulated, although yet to be fully understood. In this study, we aim to assess the impact of α and ß cell co-culture on hormone secretion. Mouse clonal cell lines α-TC6-1 (α cell line) and MIN-6 (ß cell line) were cultured independently or in combination in a medium containing 5.5, 11.1, or 25 mM glucose, respectively. After 72 h, hormone release was measured using insulin and glucagon secretion assays, the cell distribution was visualized by inverted microscopy and an immunocytochemistry assay, and changes in gene expressions were assessed using the RT-PCR technique. The co-culture of the two cell lines caused a decrease in glucagon secretion from α-TC1-6 cells, while no effect on insulin secretion from MIN-6 cells was revealed. Both types of cells were randomly scattered throughout the culture flask, unlike in mice islets in vivo where ß cells cluster in the core and α cells are localized at the periphery. During the α-ß cell co-culture, the gene expression of glucagon (Gcg) decreased significantly. We conclude that islet ß cells suppress glucagon secretion from α cells, apparently via direct cell-to-cell contact, of which the molecular mechanism needs further verification.


Asunto(s)
Comunicación Celular , Células Secretoras de Glucagón/citología , Células Secretoras de Glucagón/metabolismo , Glucagón/metabolismo , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Animales , Supervivencia Celular/efectos de los fármacos , Regulación de la Expresión Génica , Glucosa/farmacología , Secreción de Insulina , Ratones
3.
Biochem Biophys Res Commun ; 568: 158-166, 2021 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-34217973

RESUMEN

The lactate dehydrogenase isoform A (LDHA) is a key metabolic enzyme that preferentially catalyzes the conversion of pyruvate to lactate. Whereas LDHA is highly expressed in many tissues, its expression is turned off in the differentiated adult ß-cell within the pancreatic islets. The repression of LDHA under normal physiological condition and its inappropriate upregulation under a diabetogenic environment is well-documented in rodent islets/ß-cells but little is known about LDHA expression in human islet cells and whether its abundance is altered under diabetic conditions. Analysis of public single-cell RNA-seq (sc-RNA seq) data as well as cell type-specific immunolabeling of human pancreatic islets showed that LDHA was mainly localized in human α-cells while it is expressed at a very low level in ß-cells. Furthermore, LDHA, both at mRNA and protein, as well as lactate production is upregulated in human pancreatic islets exposed to chronic high glucose treatment. Microscopic analysis of stressed human islets and autopsy pancreases from individuals with type 2 diabetes (T2D) showed LDHA upregulation mainly in human α-cells. Pharmacological inhibition of LDHA in isolated human islets enhanced insulin secretion under physiological conditions but did not significantly correct the deregulated secretion of insulin or glucagon under diabetic conditions.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Células Secretoras de Glucagón/metabolismo , L-Lactato Deshidrogenasa/genética , Células Cultivadas , Diabetes Mellitus Tipo 2/metabolismo , Células Secretoras de Glucagón/citología , Glucosa/metabolismo , Humanos , Secreción de Insulina , L-Lactato Deshidrogenasa/análisis , L-Lactato Deshidrogenasa/metabolismo , ARN Mensajero/análisis , ARN Mensajero/genética , Regulación hacia Arriba
4.
Diabetes ; 70(7): 1508-1518, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33906911

RESUMEN

In contrast to the skin and the gut, where somatic stem cells and their niche are well characterized, a definitive pancreatic multipotent cell population in the adult pancreas has yet to be revealed. Of particular interest is whether such cells may be endogenous in patients with diabetes, and if so, can they be used for therapeutic purposes? In the current study, we used two separate reporter lines to target Cre-recombinase expression to the Lgr5- or glucagon-expressing cells in the pancreas. We provide evidence for the existence of a population of cells within and in the proximity of the ducts that transiently express the stem-cell marker Lgr5 during late gestational stages. Careful timing of tamoxifen treatment in Lgr5EGFP-IRES-CreERT2 ;R26 Tomato mice allowed us to show that these Lgr5-expressing progenitor cells can differentiate into α-cells during pregnancy. Furthermore, we report on a spontaneous lineage conversion of α- to ß-cells specifically after parturition. The contribution of Lgr5 progeny to the ß-cell compartment through an α-cell intermediate phase early after pregnancy appears to be part of a novel mechanism that would counterbalance against excessive ß-cell mass reduction during ß-cell involution.


Asunto(s)
Linaje de la Célula , Células Secretoras de Glucagón/citología , Células Secretoras de Insulina/citología , Páncreas/citología , Periodo Posparto/metabolismo , Receptores Acoplados a Proteínas G/fisiología , Células Madre/citología , Animales , Apoptosis , Diferenciación Celular , Femenino , Ratones , Ratones Endogámicos C57BL
5.
Front Endocrinol (Lausanne) ; 12: 633625, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33716983

RESUMEN

Enzymatically stable and specific neuropeptide Y1 receptor (NPYR1) agonists, such as sea lamprey PYY(1-36) (SL-PYY(1-36)), are believed to improve glucose regulation in diabetes by targeting pancreatic islets. In this study, streptozotocin (STZ) diabetic transgenic GluCreERT2 ;ROSA26-eYFP and Ins1Cre/+;Rosa26-eYFP mouse models have been used to study effects of sustained NPYR1 activation on islet cell composition and alpha- and beta-cell lineage transitioning. STZ induced a particularly severe form of diabetes in Ins1Cre/+;Rosa26-eYFP mice, but twice-daily administration (25 nmol/kg) of SL-PYY(1-36) for 11 days consistently improved metabolic status. Blood glucose was decreased (p < 0.05 - p < 0.001) and both fasted plasma and pancreatic insulin significantly increased by SL-PYY(1-36). In both GluCreERT2 ;ROSA26-eYFP and Ins1Cre/+; Rosa26-eYFP mice, STZ provoked characteristic losses (p < 0.05 - p < 0.001) of islet numbers, beta-cell and pancreatic islet areas together with increases in area and central islet location of alpha-cells. With exception of alpha-cell area, these morphological changes were fully, or partially, returned to non-diabetic control levels by SL-PYY(1-36). Interestingly, STZ apparently triggered decreased (p < 0.001) alpha- to beta-cell transition in GluCreERT2 ;ROSA26-eYFP mice, together with increased loss of beta-cell identity in Ins1Cre/+;Rosa26-eYFP mice, but both effects were significantly (p < 0.001) reversed by SL-PYY(1-36). SL-PYY(1-36) also apparently reduced (p < 0.05) beta- to alpha-cell conversion in Ins1Cre/+;Rosa26-eYFP mice and glucagon expressing alpha-cells in GluCreERT2 ;ROSA26-eYFP mice. These data indicate that islet benefits of prolonged NPY1R activation, and especially restoration of beta-cell mass, are observed irrespective of diabetes status, being linked to cell lineage alterations including transdifferentiation of alpha- to beta-cells.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Células Secretoras de Glucagón/citología , Células Secretoras de Insulina/citología , Islotes Pancreáticos/metabolismo , Neuropéptido Y/metabolismo , Neuropéptido Y/farmacología , Animales , Proteínas Bacterianas/química , Glucemia/metabolismo , Diferenciación Celular , Glucagón/farmacología , Insulina/farmacología , Proteínas Luminiscentes/química , Ratones , Ratones Endogámicos C57BL , Péptidos , Petromyzon , Estreptozocina , Transgenes
6.
Exp Biol Med (Maywood) ; 246(5): 617-628, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33231513

RESUMEN

Maternal pancreatic beta-cell mass (BCM) increases during pregnancy to compensate for relative insulin resistance. If BCM expansion is suboptimal, gestational diabetes mellitus can develop. Alpha-cell mass (ACM) also changes during pregnancy, but there is a lack of information about α-cell plasticity in pregnancy and whether α- to ß-cell transdifferentiation can occur. To investigate this, we used a mouse model of gestational glucose intolerance induced by feeding low-protein (LP) diet from conception until weaning and compared pregnant female offspring to control diet-fed animals. Control and LP pancreata were collected for immunohistochemical analysis and serum glucagon levels were measured. In order to lineage trace α- to ß-cell conversion, we utilized transgenic mice expressing yellow fluorescent protein behind the proglucagon gene promoter (Gcg-Cre/YFP) and collected pancreata for histology at various gestational timepoints. Alpha-cell proliferation increased significantly at gestational day (GD) 9.5 in control pregnancies resulting in an increased ACM at GD18.5, and this was significantly reduced in LP animals. Despite these changes, serum glucagon was higher in LP mice at GD18.5. Pregnant Gcg-Cre/YFP mice showed no increase in the abundance of insulin+YFP+glucagon- cells (phenotypic ß-cells). A second population of insulin+YFP+glucagon+ cells was identified which also did not alter during pregnancy. However, there was an altered anatomical distribution within islets with fewer insulin+YFP+glucagon- cells but more insulin+YFP+glucagon+ cells being present in the islet mantle at GD18.5. These findings demonstrate that dynamic changes in ACM occur during normal pregnancy and were altered in glucose-intolerant pregnancies.


Asunto(s)
Transdiferenciación Celular , Células Secretoras de Glucagón/citología , Células Secretoras de Insulina/citología , Animales , Proliferación Celular , Femenino , Glucagón/sangre , Glucagón/metabolismo , Células Secretoras de Glucagón/metabolismo , Intolerancia a la Glucosa/patología , Insulina/sangre , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Masculino , Ratones Endogámicos C57BL , Embarazo
7.
Genes Dev ; 34(23-24): 1650-1665, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33184223

RESUMEN

Circadian clocks in pancreatic islets participate in the regulation of glucose homeostasis. Here we examined the role of these timekeepers in ß-cell regeneration after the massive ablation of ß cells by doxycycline-induced expression of diphtheria toxin A (DTA) in Insulin-rtTA/TET-DTA mice. Since we crossed reporter genes expressing α- and ß-cell-specific fluorescent proteins into these mice, we could follow the fate of α- and ß cells separately. As expected, DTA induction resulted in an acute hyperglycemia, which was accompanied by dramatic changes in gene expression in residual ß cells. In contrast, only temporal alterations of gene expression were observed in α cells. Interestingly, ß cells entered S phase preferentially during the nocturnal activity phase, indicating that the diurnal rhythm also plays a role in the orchestration of ß-cell regeneration. Indeed, in arrhythmic Bmal1-deficient mice, which lack circadian clocks, no compensatory ß-cell proliferation was observed, and the ß-cell ablation led to aggravated hyperglycemia, hyperglucagonemia, and fatal diabetes.


Asunto(s)
Factores de Transcripción ARNTL/genética , Factores de Transcripción ARNTL/metabolismo , Células Secretoras de Insulina/citología , Páncreas/fisiología , Regeneración/genética , Animales , Proliferación Celular/genética , Ritmo Circadiano , Células Secretoras de Glucagón/citología , Ratones , Transcriptoma
8.
Cell Transplant ; 29: 963689720958655, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33086892

RESUMEN

The transcription factor Pax4 plays an essential role in the development of insulin-producing ß cells in pancreatic islets. Ectopic Pax4 expression not only promotes ß cell survival but also induces α-to-ß cell transdifferentiation. This dual functionality of Pax4 makes it an appealing therapeutic target for the treatment of insulin-deficient type 1 diabetes (T1D). In this study, we demonstrated that Pax4 gene delivery by an adenoviral vector, Ad5.Pax4, improved ß cell function of mouse and human islets by promoting islet cell survival and α-to-ß cell transdifferentiation, as assessed by multiple viability assays and lineage-tracing analysis. We then explored the therapeutic benefits of Pax4 gene delivery in the context of islet transplantation using T1D mouse models. Both mouse-to-mouse and human-to-mouse islet transplantation, via either kidney capsule or portal vein, were examined. In all settings, Ad5.Pax4-treated donor islets (mouse or human) showed substantially better therapeutic outcomes. These results suggest that Pax4 gene delivery into donor islets may be considered as an adjunct therapy for islet transplantation, which can either improve the therapeutic outcome of islet transplantation using the same amount of donor islets or allow the use of fewer donor islets to achieve normoglycemia.


Asunto(s)
Transdiferenciación Celular , Técnicas de Transferencia de Gen , Células Secretoras de Glucagón/citología , Proteínas de Homeodominio/genética , Células Secretoras de Insulina/citología , Trasplante de Islotes Pancreáticos , Factores de Transcripción Paired Box/genética , Animales , Linaje de la Célula , Supervivencia Celular , Diabetes Mellitus Tipo 1/terapia , Femenino , Regulación de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Humanos , Masculino , Ratones Endogámicos NOD , Ratones SCID , Factores de Transcripción Paired Box/metabolismo , Resultado del Tratamiento
9.
Int J Mol Sci ; 21(16)2020 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-32824212

RESUMEN

Diabetes is one of the leading causes of death globally. Currently, the donor pancreas is the only source of human islets, placing extreme constraints on supply. Hence, it is imperative to develop renewable islets for diabetes research and treatment. To date, extensive efforts have been made to derive insulin-secreting cells from human pluripotent stem cells with substantial success. However, the in vitro generation of functional islet organoids remains a challenge due in part to our poor understanding of the signaling molecules indispensable for controlling differentiation pathways towards the self-assembly of functional islets from stem cells. Since this process relies on a variety of signaling molecules to guide the differentiation pathways, as well as the culture microenvironments that mimic in vivo physiological conditions, this review highlights extracellular matrix proteins, growth factors, signaling molecules, and microenvironments facilitating the generation of biologically functional pancreatic endocrine cells from human pluripotent stem cells. Signaling pathways involved in stepwise differentiation that guide the progression of stem cells into the endocrine lineage are also discussed. The development of protocols enabling the generation of islet organoids with hormone release capacities equivalent to native adult islets for clinical applications, disease modeling, and diabetes research are anticipated.


Asunto(s)
Diferenciación Celular , Células Secretoras de Glucagón/metabolismo , Células Secretoras de Insulina/metabolismo , Células Madre Pluripotentes/metabolismo , Transducción de Señal , Animales , Técnicas de Reprogramación Celular/métodos , Células Secretoras de Glucagón/citología , Humanos , Células Secretoras de Insulina/citología , Células Madre Pluripotentes/citología , Nicho de Células Madre
10.
Biochem Biophys Res Commun ; 530(1): 266-272, 2020 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-32828297

RESUMEN

Glucagon is a peptide hormone generated by pancreatic α cells. It is the counterpart of insulin and plays an essential role in the regulation of blood glucose level. Therefore, a tight regulation of glucagon levels is pivotal to maintain homeostasis of blood glucose. However, little is known about the mechanisms regulating glucagon biosynthesis. In this study, we demonstrate that the RNA-binding protein HuD regulates glucagon expression in pancreatic α cells. HuD was found in α cells from mouse pancreatic islet and mouse glucagonoma αTC1 cell line. Ribonucleoprotein immunoprecipitation analysis, followed by RT-qPCR showed the association of HuD with glucagon mRNA. Knockdown of HuD resulted in a reduction in both proglucagon expression and cellular glucagon level by decreasing its de novo synthesis. Reporter analysis using the EGFP reporter containing 3' untranslated region (3'UTR) of glucagon mRNA showed that HuD regulates proglucagon expression via its 3'UTR. In addition, the relative level of glucagon in the islets and plasma was lower in HuD knockout (KO) mice compared to age-matched control mice. Taken together, these results suggest that HuD is a novel factor regulating the biosynthesis of proglucagon in pancreatic α cells.


Asunto(s)
Proteína 4 Similar a ELAV/metabolismo , Células Secretoras de Glucagón/metabolismo , Proglucagón/metabolismo , Animales , Vías Biosintéticas , Línea Celular , Línea Celular Tumoral , Regulación hacia Abajo , Proteína 4 Similar a ELAV/genética , Técnicas de Silenciamiento del Gen , Células Secretoras de Glucagón/citología , Ratones , Proglucagón/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo
11.
Metabolism ; 109: 154290, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32522488

RESUMEN

BACKGROUND: Males absent on the first (Mof) is implicated in gene control of diverse biological processes, such as cell growth, differentiation, apoptosis and autophagy. However, the relationship between glucose regulation and Mof-mediated transcription events remains unexplored. We aimed to unravel the role of Mof in glucose regulation by using global and pancreatic α-cell-specific Mof-deficient mice in vivo and α-TC1-6 cell line in vitro. METHODS: We used tamoxifen-induced temporal Mof-deficient mice first to show Mof regulate glucose homeostasis, islet cell proportions and hormone secretion. Then we used α-cell-specific Mof-deficient mice to clarify how α-cell subsets and ß-cell mass were regulated and corresponding hormone level alterations. Ultimately, we used small interfering RNA (siRNA) to knockdown Mof in α-TC1-6 and unravel the mechanism regulating α-cell mass and glucagon secretion. RESULTS: Mof was mainly expressed in α-cells. Global Mof deficiency led to lower glucose levels, attributed by decreased α/ß-cell ratio and glucagon secretion. α-cell-specific Mof-deficient mice exhibited similar alterations, with more reduced prohormone convertase 2 (PC2)-positive α-cell mass, responsible for less glucagon, and enhanced prohormone convertase 1 (PC1/3)-positive α-cell mass, leading to more glucagon-like peptide-1 (GLP-1) secretion, thus increased ß-cell mass and insulin secretion. In vitro, increased DNA damage, dysregulated autophagy, enhanced apoptosis and altered cell fate factors expressions upon Mof knockdown were observed. Genes and pathways linked to impaired glucagon secretion were uncovered through transcriptome sequencing. CONCLUSION: Mof is a potential interventional target for glucose regulation, from the aspects of both α-cell subset mass and glucagon, intra-islet GLP-1 secretion. Upon Mof deficiency, Up-regulated PC1/3 but down-regulated PC2-positive α-cell mass, leads to more GLP-1 and insulin but less glucagon secretion, and contributed to lower glucose level.


Asunto(s)
Glucemia/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Células Secretoras de Glucagón/citología , Glucagón/metabolismo , Histona Acetiltransferasas/fisiología , Homeostasis , Animales , Línea Celular , Histona Acetiltransferasas/deficiencia , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Ratones , Proproteína Convertasa 1/metabolismo , Proproteína Convertasa 2/metabolismo
12.
J Biol Chem ; 295(29): 9879-9892, 2020 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-32439805

RESUMEN

Type 2 diabetes is a chronic metabolic disease characterized by pancreatic ß-cell dysfunction and peripheral insulin resistance. Among individuals with type 2 diabetes, ∼30% exhibit hypomagnesemia. Hypomagnesemia has been linked to insulin resistance through reduced tyrosine kinase activity of the insulin receptor; however, its impact on pancreatic ß-cell function is unknown. In this study, through analysis of several single-cell RNA-sequencing data sets in tandem with quantitative PCR validation in both murine and human islets, we identified NIPAL1 (NIPA-like domain containing 1), encoding a magnesium influx transporter, as an islet-enriched gene. A series of immunofluorescence experiments confirmed NIPAL1's magnesium-dependent expression and that it specifically localizes to the Golgi in Min6-K8 cells, a pancreatic ß-cell-like cell line (mouse insulinoma 6 clone K8). Under varying magnesium concentrations, NIPAL1 knockdown decreased both basal insulin secretion and total insulin content; in contrast, its overexpression increased total insulin content. Although the expression, distribution, and magnesium responsiveness of NIPAL1 in α-TC6 glucagonoma cells (a pancreatic α-cell line) were similar to the observations in Min6-K8 cells, no effect was observed on glucagon secretion in α-TC6 cells under the conditions studied. Overall, these results suggest that NIPAL1 expression is regulated by extracellular magnesium and that down-regulation of this transporter decreases glucose-stimulated insulin secretion and intracellular insulin content, particularly under conditions of hypomagnesemia.


Asunto(s)
Proteínas de Transporte de Catión/biosíntesis , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Magnesio/metabolismo , Animales , Proteínas de Transporte de Catión/genética , Línea Celular Tumoral , Regulación de la Expresión Génica , Células Secretoras de Glucagón/citología , Células Secretoras de Glucagón/metabolismo , Células Secretoras de Insulina/citología , Masculino , Ratones
13.
Life Sci ; 255: 117810, 2020 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-32473248

RESUMEN

AIMS: The aim of the present study was to clarify if in utero exposure to DEX would affect the development of different types of pancreatic endocrine cells during postnatal life. MAIN METHODS: We investigated morphological and transcriptional features of both pancreatic ß- and α-cell populations within the pancreatic islets during the early postnatal life of rats born to mothers treated with DEX (0.1 mg/kg) from day 14 to 19 of pregnancy. Untreated pregnant Wistar rats of the same age (12-week-old) were used as control (CTL). Pups were euthanized on the 1st, 3rd and 21st (PND1, PND3 and PND21, respectively) days of life, regardless of sex. Serum insulin and glucagon levels were also evaluated. KEY FINDINGS: Rats born to DEX-treated mothers exhibited increased pancreatic α-cell mass, circulating glucagon levels and Gcg, Pax6, MafB and Nkx2.2 expression. Rats born to DEX-treated mothers also presented a rise in serum insulin levels on the PND3 that was paralleled by reduced ß-cell mass. Such increase in serum insulin levels, instead, was associated with increased expression of genes associated to insulin secretion such as Gck and Slc2a2. SIGNIFICANCE: Altogether, the present data reveals yet unknown changes in endocrine pancreas during early postnatal life of rats exposed to DEX in utero. Such data may contribute to the understanding of the metabolic features of rats born to DEX-treated mothers.


Asunto(s)
Dexametasona/toxicidad , Células Secretoras de Glucagón/efectos de los fármacos , Glucocorticoides/toxicidad , Células Secretoras de Insulina/efectos de los fármacos , Animales , Dexametasona/administración & dosificación , Femenino , Regulación de la Expresión Génica , Glucagón/sangre , Células Secretoras de Glucagón/citología , Glucocorticoides/administración & dosificación , Proteína Homeobox Nkx-2.2 , Insulina/sangre , Secreción de Insulina/fisiología , Células Secretoras de Insulina/citología , Masculino , Embarazo , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Ratas , Ratas Wistar
14.
JCI Insight ; 5(10)2020 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-32352931

RESUMEN

Pancreatic islets secrete insulin from ß cells and glucagon from α cells, and dysregulated secretion of these hormones is a central component of diabetes. Thus, an improved understanding of the pathways governing coordinated ß and α cell hormone secretion will provide insight into islet dysfunction in diabetes. However, the 3D multicellular islet architecture, essential for coordinated islet function, presents experimental challenges for mechanistic studies of intracellular signaling pathways in primary islet cells. Here, we developed an integrated approach to study the function of primary human islet cells using genetically modified pseudoislets that resemble native islets across multiple parameters. Further, we developed a microperifusion system that allowed synchronous acquisition of GCaMP6f biosensor signal and hormone secretory profiles. We demonstrate the utility of this experimental approach by studying the effects of Gi and Gq GPCR pathways on insulin and glucagon secretion by expressing the designer receptors exclusively activated by designer drugs (DREADDs) hM4Di or hM3Dq. Activation of Gi signaling reduced insulin and glucagon secretion, while activation of Gq signaling stimulated glucagon secretion but had both stimulatory and inhibitory effects on insulin secretion, which occur through changes in intracellular Ca2+. The experimental approach of combining pseudoislets with a microfluidic system allowed the coregistration of intracellular signaling dynamics and hormone secretion and demonstrated differences in GPCR signaling pathways between human ß and α cells.


Asunto(s)
Técnicas Biosensibles , Células Secretoras de Glucagón/metabolismo , Células Secretoras de Insulina/metabolismo , Dispositivos Laboratorio en un Chip , Técnicas Analíticas Microfluídicas , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Femenino , Células Secretoras de Glucagón/citología , Humanos , Secreción de Insulina , Células Secretoras de Insulina/citología , Masculino
15.
Nat Commun ; 11(1): 2241, 2020 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-32382023

RESUMEN

The generation of pancreatic cell types from renewable cell sources holds promise for cell replacement therapies for diabetes. Although most effort has focused on generating pancreatic beta cells, considerable evidence indicates that glucagon secreting alpha cells are critically involved in disease progression and proper glucose control. Here we report on the generation of stem cell-derived human pancreatic alpha (SC-alpha) cells from pluripotent stem cells via a transient pre-alpha cell intermediate. These pre-alpha cells exhibit a transcriptional profile similar to mature alpha cells and although they produce proinsulin protein, they do not secrete significant amounts of processed insulin. Compound screening identified a protein kinase c activator that promotes maturation of pre-alpha cells into SC-alpha cells. The resulting SC-alpha cells do not express insulin, share an ultrastructure similar to cadaveric alpha cells, express and secrete glucagon in response to glucose and some glucagon secretagogues, and elevate blood glucose upon transplantation in mice.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Células Secretoras de Glucagón/citología , Células Secretoras de Insulina/efectos de los fármacos , Células Madre Pluripotentes/citología , Western Blotting , Diferenciación Celular/fisiología , Línea Celular , Electrofisiología , Técnica del Anticuerpo Fluorescente , Humanos , Páncreas/citología
16.
Artículo en Inglés | MEDLINE | ID: mdl-32296396

RESUMEN

Insulin and glucagon are hormones secreted by pancreatic ß and α cells, respectively, which together regulate glucose homeostasis. Dysregulation of insulin or glucagon can result in loss of blood glucose control, characterized by hyperglycemia or hypoglycemia. To better understand the endocrine physiology of cetaceans, we cloned and characterized the insulin and glucagon genes from pygmy sperm whale (Kogia breviceps). We obtained the complete coding sequences of the preproinsulin and preproglucagon genes, which encodes the preproinsulin protein of 110 amino acid (aa) residues and encodes the preproglucagon protein of 179 aa residues, respectively. Sequence comparison and phylogenetic analyses demonstrate that protein structures were similar to other mammalian orthologs. Immunohistochemistry and immunofluorescence staining using insulin, glucagon, and somatostatin antibodies allowed analysis of pygmy sperm whale islet distribution, architecture, and composition. Our results showed the pygmy sperm whale islet was irregularly shaped and randomly distributed throughout the pancreas. The architecture of α, ß, and δ cells of the pygmy sperm whale was similar to that of artiodactyls species. This is the first report about insulin and glucagon genes in cetaceans, which provides new information about the structural conservation of the insulin and glucagon genes. Furthermore, offers novel information on the properties of endocrine cells in cetacean for further studies.


Asunto(s)
Células Endocrinas/metabolismo , Glucagón/genética , Insulina/genética , Ballenas , Animales , Clonación Molecular , Células Endocrinas/citología , Femenino , Glucagón/metabolismo , Células Secretoras de Glucagón/citología , Células Secretoras de Glucagón/metabolismo , Células Secretoras de Glucagón/fisiología , Insulina/metabolismo , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Páncreas/citología , Páncreas/metabolismo , Filogenia , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Análisis de Secuencia de ADN/veterinaria , Ballenas/genética , Ballenas/metabolismo
17.
Development ; 147(6)2020 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-32108026

RESUMEN

Reliance on rodents for understanding pancreatic genetics, development and islet function could limit progress in developing interventions for human diseases such as diabetes mellitus. Similarities of pancreas morphology and function suggest that porcine and human pancreas developmental biology may have useful homologies. However, little is known about pig pancreas development. To fill this knowledge gap, we investigated fetal and neonatal pig pancreas at multiple, crucial developmental stages using modern experimental approaches. Purification of islet ß-, α- and δ-cells followed by transcriptome analysis (RNA-seq) and immunohistology identified cell- and stage-specific regulation, and revealed that pig and human islet cells share characteristic features that are not observed in mice. Morphometric analysis also revealed endocrine cell allocation and architectural similarities between pig and human islets. Our analysis unveiled scores of signaling pathways linked to native islet ß-cell functional maturation, including evidence of fetal α-cell GLP-1 production and signaling to ß-cells. Thus, the findings and resources detailed here show how pig pancreatic islet studies complement other systems for understanding the developmental programs that generate functional islet cells, and that are relevant to human pancreatic diseases.


Asunto(s)
Diferenciación Celular/genética , Células Secretoras de Insulina/fisiología , Islotes Pancreáticos/embriología , Islotes Pancreáticos/crecimiento & desarrollo , Porcinos , Animales , Animales Recién Nacidos , Células Cultivadas , Embrión de Mamíferos , Femenino , Feto/metabolismo , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Células Secretoras de Glucagón/citología , Células Secretoras de Glucagón/fisiología , Humanos , Islotes Pancreáticos/citología , Ratones , Organogénesis/genética , Embarazo , Porcinos/embriología , Porcinos/genética , Porcinos/crecimiento & desarrollo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcriptoma
18.
Metabolism ; 102: 153963, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31593706

RESUMEN

BACKGROUND: Pregnancy represents a major metabolic challenge for the mother, and involves a compensatory response of the pancreatic beta-cell to maintain normoglycemia. However, although pancreatic alpha-cells play a key role in glucose homeostasis and seem to be involved in gestational diabetes, there is no information about their potential adaptations or changes during pregnancy. MATERIAL AND METHODS: Non-pregnant (controls) and pregnant C57BL/6 mice at gestational day 18.5 (G18.5) and their isolated pancreatic islets were used for in vivo and ex vivo studies, respectively. The effect of pregnancy hormones was tested in glucagon-secreting α-TC1.9 cells. Immunohistochemical analysis was performed in pancreatic slices. Glucagon gene expression was monitored by RT-qPCR. Glucagon secretion and plasma hormones were measured by ELISA. RESULTS: Pregnant mice on G18.5 exhibited alpha-cell hypertrophy as well as augmented alpha-cell area and mass. This alpha-cell mass expansion was mainly due to increased proliferation. No changes in alpha-cell apoptosis, ductal neogenesis, or alpha-to-beta transdifferentiation were found compared with controls. Pregnant mice on G18.5 exhibited hypoglucagonemia. Additionally, in vitro glucagon secretion at low glucose levels was decreased in isolated islets from pregnant animals. Glucagon content was also reduced. Experiments in α-TC1.9 cells indicated that, unlike estradiol and progesterone, placental lactogens and prolactin stimulated alpha-cell proliferation. Placental lactogens, prolactin and estradiol also inhibited glucagon release from α-TC1.9 cells at low glucose levels. CONCLUSIONS: The pancreatic alpha-cell in mice undergoes several morphofunctional changes during late pregnancy, which may contribute to proper glucose homeostasis. Gestational hormones are likely involved in these processes.


Asunto(s)
Adaptación Fisiológica/fisiología , Edad Gestacional , Células Secretoras de Glucagón/citología , Células Secretoras de Glucagón/fisiología , Animales , Recuento de Células , Tamaño de la Célula , Células Cultivadas , Femenino , Glucagón/metabolismo , Islotes Pancreáticos/citología , Islotes Pancreáticos/fisiología , Ratones , Ratones Endogámicos C57BL , Hormonas Placentarias/fisiología , Embarazo
19.
Sci Rep ; 9(1): 9515, 2019 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-31266981

RESUMEN

Most studies in type 1 diabetes (T1D) have focused on the loss of the pancreatic beta-cell population. However, despite the involvement of the alpha-cell in the aetiology and complications of T1D, little is known about the regulation of the pancreatic alpha-cell mass in this disease. The need for a better understanding of this process is further emphasized by recent findings suggesting that alpha-cells may constitute a potential reservoir for beta-cell regeneration. In this study, we characterized the pancreatic alpha-cell mass and its regulatory processes in the transgenic RIP-B7.1 mice model of experimental autoimmune diabetes (EAD). Diabetic mice presented insulitis, hyperglycaemia, hypoinsulinemia and hyperglucagonemia along with lower pancreatic insulin content. While alpha-cell mass and pancreatic glucagon content were preserved at the early-onset of EAD, both parameters were reduced in the advanced phase. At both stages, alpha-cell size, proliferation and ductal neogenesis were up-regulated, whereas apoptosis was almost negligible. Interestingly, we found an increase in the proportion of glucagon-containing cells positive for insulin or the beta-cell transcription factor PDX1. Our findings suggest that pancreatic alpha-cell renewal mechanisms are boosted during the natural course of EAD, possibly as an attempt to maintain the alpha-cell population and/or to increase beta-cell regeneration via alpha-cell transdifferentiation.


Asunto(s)
Diabetes Mellitus Experimental/patología , Animales , Antígeno B7-1/deficiencia , Antígeno B7-1/genética , Proliferación Celular , Transdiferenciación Celular , Diabetes Mellitus Experimental/complicaciones , Modelos Animales de Enfermedad , Glucagón/análisis , Glucagón/metabolismo , Células Secretoras de Glucagón/citología , Células Secretoras de Glucagón/metabolismo , Proteínas de Homeodominio/metabolismo , Hiperglucemia/complicaciones , Hiperglucemia/patología , Insulina/análisis , Insulina/sangre , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Transactivadores/metabolismo
20.
Development ; 146(12)2019 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-31160419

RESUMEN

During pancreatic development, endocrine cells appear from the pancreatic epithelium when Neurog3-positive cells delaminate and differentiate into α-, ß-, γ- and δ-cells. The mechanisms involved in this process are still incompletely understood. We characterized the temporal, lineage-specific developmental programs during pancreatic development by sequencing the transcriptome of thousands of individual pancreatic cells from E12.5 to E18.5 in mice, and identified all known cell types that are present in the embryonic pancreas, but focused specifically on α- and ß-cell differentiation by enrichment of a MIP-GFP reporter. We characterized transcriptomic heterogeneity in the tip domain based on proliferation, and characterized two endocrine precursor clusters marked by expression of Neurog3 and Fev Pseudotime analysis revealed specific branches for developing α- and ß-cells, which allowed identification of specific gene regulation patterns. These include some known and many previously unreported genes that appear to define pancreatic cell fate transitions. This resource allows dynamic profiling of embryonic pancreas development at single cell resolution and reveals novel gene signatures during pancreatic differentiation into α- and ß-cells.


Asunto(s)
Linaje de la Célula , Regulación del Desarrollo de la Expresión Génica , Células Secretoras de Glucagón/citología , Células Secretoras de Insulina/citología , Páncreas/embriología , Transcriptoma , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular , Separación Celular , Citometría de Flujo , Biblioteca de Genes , Proteínas Fluorescentes Verdes/metabolismo , Ratones , Proteínas del Tejido Nervioso/metabolismo , Organogénesis , Células Madre/citología , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...