Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Exp Cell Res ; 350(1): 50-61, 2017 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-27836810

RESUMEN

Polycystin-2 (PC2, TRPP2) is a nonselective cation channel whose dysfunction is associated with the onset of autosomal dominant polycystic kidney disease (ADPKD). PC2 contributes to Ca2+ transport and cell signaling in renal epithelia and other tissues. Little is known however, as to the external Ca2+ regulation of PC2 channel function. In this study, we explored the effect of external Ca2+ on endogenous PC2 in wild type LLC-PK1 renal epithelial cells. We obtained whole cell currents at different external Ca2+ concentrations, and observed that the basal whole cell conductance in normal Ca2+(1.2mM), decreased by 30.2% in zero (nominal) Ca2+ and conversely, increased by 38% in high external Ca2+(6.2mM). The high Ca2+-increased whole cell currents were completely inhibited by either PC2 gene silencing, or intracellular dialysis with active, but not denatured by boiling, PC2 antibody. Exposure of cells to high Ca2+ was also associated with relocation of PC2 to the plasma membrane. To explore whether a Ca2+ sensing receptor (CaSR) was implicated in the external Ca2+ modulation of PC2 currents, we tested the effect of the CaSR agonists, spermine and the calcimimetic R-568, which largely mimicked the effect of high Ca2+ under Ca2+-free conditions. The CaSR agonist gentamicin also increased the PC2 currents in the presence of normal Ca2+. The presence of CaSR was confirmed by immunocytochemistry, which partially colocalized with the intracellular PC2 protein, in an external Ca2+-dependent manner. The data support a novel Ca2+ sensing mechanism for PC2 expression and functional regulation in renal epithelial cells.


Asunto(s)
Calcio/metabolismo , Células Epiteliales/metabolismo , Riñón/metabolismo , Canales Catiónicos TRPP/metabolismo , Animales , Membrana Celular/metabolismo , Porcinos
2.
Diabetes Obes Metab ; 19 Suppl 1: 90-94, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28880482

RESUMEN

After multiple decades of investigation, the precise mechanisms involved in fuel-stimulated insulin secretion are still being revealed. One avenue for gaining deeper knowledge is to apply emergent tools of "metabolomics," involving mass spectrometry and nuclear magnetic resonance-based profiling of islet cells in their fuel-stimulated compared with basal states. The current article summarizes recent insights gained from application of metabolomics tools to the specific process of glucose-stimulated insulin secretion, revealing 2 new mechanisms that may provide targets for improving insulin secretion in diabetes.


Asunto(s)
Investigación Biomédica/métodos , Islotes Pancreáticos/metabolismo , Metabolómica/métodos , Modelos Biológicos , Animales , Investigación Biomédica/tendencias , Exocitosis , Glucosa/metabolismo , Humanos , Insulina/metabolismo , Secreción de Insulina , Islotes Pancreáticos/enzimología , Metabolómica/tendencias , Vías Secretoras
3.
Diabetologia ; 58(7): 1513-22, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25874445

RESUMEN

AIMS/HYPOTHESIS: Precise regulation of insulin secretion by the pancreatic beta cell is essential for the maintenance of glucose homeostasis. Insulin secretory activity is initiated by the stepwise breakdown of ambient glucose to increase cellular ATP via glycolysis and mitochondrial respiration. Knockout of Lkb1, the gene encoding liver kinase B1 (LKB1) from the beta cell in mice enhances insulin secretory activity by an undefined mechanism. Here, we sought to determine the molecular basis for how deletion of Lkb1 promotes insulin secretion. METHODS: To explore the role of LKB1 on individual steps in the insulin secretion pathway, we used mitochondrial functional analyses, electrophysiology and metabolic tracing coupled with by gas chromatography and mass spectrometry. RESULTS: Beta cells lacking LKB1 surprisingly display impaired mitochondrial metabolism and lower ATP levels following glucose stimulation, yet compensate for this by upregulating both uptake and synthesis of glutamine, leading to increased production of citrate. Furthermore, under low glucose conditions, Lkb1(-/-) beta cells fail to inhibit acetyl-CoA carboxylase 1 (ACC1), the rate-limiting enzyme in lipid synthesis, and consequently accumulate NEFA and display increased membrane excitability. CONCLUSIONS/INTERPRETATION: Taken together, our data show that LKB1 plays a critical role in coupling glucose metabolism to insulin secretion, and factors in addition to ATP act as coupling intermediates between feeding cues and secretion. Our data suggest that beta cells lacking LKB1 could be used as a system to identify additional molecular events that connect metabolism to cellular excitation in the insulin secretion pathway.


Asunto(s)
Glucosa/metabolismo , Insulina/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Quinasas Activadas por AMP , Acetil-CoA Carboxilasa/metabolismo , Animales , Ácidos Grasos no Esterificados/sangre , Glucosa/deficiencia , Glucosa/farmacología , Glutamina/biosíntesis , Glutamina/metabolismo , Hipoglucemiantes/farmacología , Secreción de Insulina , Células Secretoras de Insulina , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Metabolómica , Ratones , Ratones Noqueados , Mitocondrias/metabolismo , ARN Interferente Pequeño/biosíntesis , ARN Interferente Pequeño/genética
4.
Diabetologia ; 58(7): 1503-12, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25930156

RESUMEN

AIMS/HYPOTHESIS: There are potential advantages to the low-temperature (-196 °C) banking of isolated islets, including the maintenance of viable islets for future research. We therefore assessed the in vitro and in vivo function of islets cryopreserved for nearly 20 years. METHODS: Human islets were cryopreserved from 1991 to 2001 and thawed between 2012 and 2014. These were characterised by immunostaining, patch-clamp electrophysiology, insulin secretion, transcriptome analysis and transplantation into a streptozotocin (STZ)-induced mouse model of diabetes. RESULTS: The cryopreservation time was 17.6 ± 0.4 years (n = 43). The thawed islets stained positive with dithizone, contained insulin-positive and glucagon-positive cells, and displayed levels of apoptosis and transcriptome profiles similar to those of freshly isolated islets, although their insulin content was lower. The cryopreserved beta cells possessed ion channels and exocytotic responses identical to those of freshly isolated beta cells. Cells from a subset of five donors demonstrated similar perifusion insulin secretion profiles pre- and post-cryopreservation. The transplantation of cryopreserved islets into the diabetic mice improved their glucose tolerance but did not completely normalise their blood glucose levels. Circulating human insulin and insulin-positive grafts were detectable at 10 weeks post-transplantation. CONCLUSIONS/INTERPRETATION: We have demonstrated the potential for long-term banking of human islets for research, which could enable the use of tissue from a large number of donors with future technologies to gain new insight into diabetes.


Asunto(s)
Criopreservación , Islotes Pancreáticos/fisiología , Bancos de Tejidos , Adulto , Animales , Diabetes Mellitus Experimental/terapia , Exocitosis/fisiología , Femenino , Proteínas de Homeodominio/genética , Humanos , Insulina/sangre , Insulina/metabolismo , Células Secretoras de Insulina/fisiología , Canales Iónicos/metabolismo , Trasplante de Islotes Pancreáticos , Masculino , Ratones , Ratones Noqueados , Técnicas de Placa-Clamp , Transcriptoma/genética
5.
Diabetologia ; 57(2): 383-91, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24190582

RESUMEN

AIMS/HYPOTHESIS: Genome-wide association studies have revealed an association of the transcription factor ETS variant gene 5 (ETV5) with human obesity. However, its role in glucose homeostasis and energy balance is unknown. METHODS: Etv5 knockout (KO) mice were monitored weekly for body weight (BW) and food intake. Body composition was measured at 8 and 16 weeks of age. Glucose metabolism was studied, and glucose-stimulated insulin secretion was measured in vivo and in vitro. RESULTS: Etv5 KO mice are smaller and leaner, and have a reduced BW and lower fat mass than their wild-type controls on a chow diet. When exposed to a high-fat diet, KO mice are resistant to diet-induced BW gain. Despite a greater insulin sensitivity, KO mice have profoundly impaired glucose tolerance associated with impaired insulin secretion. Morphometric analysis revealed smaller islets and a reduced beta cell size in the pancreatic islets of Etv5 KO mice. Knockdown of ETV5 in an insulin-secreting cell line or beta cells from human donors revealed intact mitochondrial and Ca(2+) channel activity, but reduced insulin exocytosis. CONCLUSION/INTERPRETATION: This work reveals a critical role for ETV5 in specifically regulating insulin secretion both in vitro and in vivo.


Asunto(s)
Péptido C/metabolismo , Proteínas de Unión al ADN/metabolismo , Exocitosis/fisiología , Glucosa/metabolismo , Homeostasis/fisiología , Insulina/metabolismo , Factores de Transcripción/metabolismo , Animales , Composición Corporal , Peso Corporal , Dieta Alta en Grasa , Ingestión de Alimentos , Estudio de Asociación del Genoma Completo , Prueba de Tolerancia a la Glucosa , Resistencia a la Insulina , Secreción de Insulina , Ratones , Ratones Noqueados
6.
J Physiol ; 592(17): 3715-26, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-24907310

RESUMEN

Post-translational modification by the small ubiquitin-like modifier-1 (SUMO1) limits insulin secretion from ß-cells by inhibiting insulin exocytosis and glucagon-like peptide-1 (GLP-1) receptor signalling. The secretion of glucagon from α-cells is regulated in a manner opposite to that of insulin; it is inhibited by elevated glucose and GLP-1, and increased by adrenergic signalling. We therefore sought to determine whether SUMO1 modulates mouse and human α-cell function. Action potentials (APs), ion channel function and exocytosis in single α-cells from mice and humans, identified by glucagon immunostaining, and glucagon secretion from intact islets were measured. The effects of SUMO1 on α-cell function and the respective inhibitory and stimulatory effects of exendin 4 and adrenaline were examined. Upregulation of SUMO1 increased α-cell AP duration, frequency and amplitude, in part as a result of increased Ca(2+) channel activity that led to elevated exocytosis. The ability of SUMO1 to enhance α-cell exocytosis was cAMP-dependent and resulted from an increased L-type Ca(2+) current and a shift away from exocytosis dependent on non-L-type channels, an effect that was mimicked by knockdown of the deSUMOylating enzyme sentrin/SUMO-specific protease-1 (SENP1). Finally, although SUMO1 prevented GLP-1 receptor-mediated inhibition of α-cell Na(+) channels and single-cell exocytosis, it failed to prevent the exendin 4-mediated inhibition of glucagon secretion. Consistent with its cAMP dependence, however, SUMO1 enhanced α-cell exocytosis and glucagon secretion stimulated by adrenaline. Thus, by contrast with its inhibitory role in ß-cell exocytosis, SUMO1 is a positive regulator of α-cell exocytosis and glucagon secretion under conditions of elevated cAMP.


Asunto(s)
AMP Cíclico/metabolismo , Exocitosis , Células Secretoras de Glucagón/metabolismo , Glucagón/metabolismo , Proteína SUMO-1/metabolismo , Potenciales de Acción , Animales , Canales de Calcio Tipo L/metabolismo , Células Cultivadas , Cisteína Endopeptidasas , Endopeptidasas/genética , Endopeptidasas/metabolismo , Receptor del Péptido 1 Similar al Glucagón , Células Secretoras de Glucagón/fisiología , Humanos , Ratones , Ratones Endogámicos C57BL , Receptores de Glucagón/agonistas , Receptores de Glucagón/antagonistas & inhibidores , Proteína SUMO-1/genética , Sodio/metabolismo
7.
J Biol Chem ; 288(1): 264-73, 2013 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-23161538

RESUMEN

Epithelial sodium channel (ENaC) in the kidneys is critical for Na(+) balance, extracellular volume, and blood pressure. Altered ENaC function is associated with respiratory disorders, pseudohypoaldosteronism type 1, and Liddle syndrome. ENaC is known to interact with components of the cytoskeleton, but the functional roles remain largely unclear. Here, we examined the interaction between ENaC and filamins, important actin filament components. We first discovered by yeast two-hybrid screening that the C termini of ENaC α and ß subunits bind filamin A, B, and C, and we then confirmed the binding by in vitro biochemical assays. We demonstrated by co-immunoprecipitation that ENaC, either overexpressed in HEK, HeLa, and melanoma A7 cells or natively expressed in LLC-PK1 and IMCD cells, is in the same complex with native filamin. Furthermore, the biotinylation and co-immunoprecipitation combined assays showed the ENaC-filamin interaction on the cell surface. Using Xenopus oocyte expression and two-electrode voltage clamp electrophysiology, we found that co-expression of an ENaC-binding domain of filamin substantially reduces ENaC channel function. Western blot and immunohistochemistry experiments revealed that the filamin A C terminus (FLNAC) modestly reduces the expression of the ENaC α subunit in oocytes and A7 cells. After normalizing the current by plasma membrane expression, we found that FLNAC results in ~50% reduction in the ENaC channel activity. The inhibitory effect of FLNAC was confirmed by lipid bilayer electrophysiology experiments using purified ENaC and FLNAC proteins, which showed that FLNAC substantially reduces ENaC single channel open probability. Taken together, our study demonstrated that filamin reduces ENaC channel function through direct interaction on the cell surface.


Asunto(s)
Proteínas Contráctiles/química , Canales Epiteliales de Sodio/química , Regulación de la Expresión Génica , Proteínas de Microfilamentos/química , Sodio/metabolismo , Animales , Línea Celular Tumoral , Membrana Celular/metabolismo , Proliferación Celular , Citoesqueleto/metabolismo , Perros , Filaminas , Glutatión Transferasa/metabolismo , Células HEK293 , Células HeLa , Homeostasis , Humanos , Riñón/metabolismo , Ratones , Oocitos/metabolismo , Mapeo de Interacción de Proteínas/métodos , Canales de Sodio/metabolismo , Porcinos , Técnicas del Sistema de Dos Híbridos , Xenopus
8.
Nat Commun ; 15(1): 334, 2024 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-38184650

RESUMEN

Pancreatic ß-cells respond to metabolic stress by upregulating insulin secretion, however the underlying mechanisms remain unclear. Here we show, in ß-cells from overweight humans without diabetes and mice fed a high-fat diet for 2 days, insulin exocytosis and secretion are enhanced without increased Ca2+ influx. RNA-seq of sorted ß-cells suggests altered metabolic pathways early following high fat diet, where we find increased basal oxygen consumption and proton leak, but a more reduced cytosolic redox state. Increased ß-cell exocytosis after 2-day high fat diet is dependent on this reduced intracellular redox state and requires the sentrin-specific SUMO-protease-1. Mice with either pancreas- or ß-cell-specific deletion of this fail to up-regulate exocytosis and become rapidly glucose intolerant after 2-day high fat diet. Mechanistically, redox-sensing by the SUMO-protease requires a thiol group at C535 which together with Zn+-binding suppresses basal protease activity and unrestrained ß-cell exocytosis, and increases enzyme sensitivity to regulation by redox signals.


Asunto(s)
Dieta Alta en Grasa , Exocitosis , Animales , Humanos , Ratones , Cisteína Endopeptidasas/genética , Citosol , Dieta Alta en Grasa/efectos adversos , Glucosa , Péptido Hidrolasas
9.
bioRxiv ; 2024 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-38948734

RESUMEN

Comprehensive molecular and cellular phenotyping of human islets can enable deep mechanistic insights for diabetes research. We established the Human Islet Data Analysis and Sharing (HI-DAS) consortium to advance goals in accessibility, usability, and integration of data from human islets isolated from donors with and without diabetes at the Alberta Diabetes Institute (ADI) IsletCore. Here we introduce HumanIslets.com, an open resource for the research community. This platform, which presently includes data on 547 human islet donors, allows users to access linked datasets describing molecular profiles, islet function and donor phenotypes, and to perform various statistical and functional analyses at the donor, islet and single-cell levels. As an example of the analytic capacity of this resource we show a dissociation between cell culture effects on transcript and protein expression, and an approach to correct for exocrine contamination found in hand-picked islets. Finally, we provide an example workflow and visualization that highlights links between type 2 diabetes status, SERCA3b Ca2+-ATPase levels at the transcript and protein level, insulin secretion and islet cell phenotypes. HumanIslets.com provides a growing and adaptable set of resources and tools to support the metabolism and diabetes research community.

10.
J Biol Chem ; 287(9): 6551-61, 2012 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-22174419

RESUMEN

Pkd2L1 (also called TRPP3) is a non-selective cation channel permeable to Ca(2+), Na(+), and K(+) and is activated by Ca(2+). It is also part of an acid-triggered off-response cation channel complex. We previously reported roles of the Pkd2L1 C-terminal fragments in its channel function, but the role of the N terminus remains unclear. Using a yeast two-hybrid screening, we found that the Pkd2L1 N terminus interacts with the receptor for activated C kinase 1 (RACK1), a scaffolding/anchoring protein implicated in various cellular functions. This interaction requires the last two Trp-Asp (WD) motifs of RACK1 and fragment Ala(19)-Pro(45) of Pkd2L1. The interaction was confirmed by GST pulldown, blot overlay, and co-immunoprecipitation assays. By (45)Ca tracer uptake and two-microelectrode voltage clamp electrophysiology, we found that in Xenopus oocytes with RACK1 overexpression Pkd2L1 channel activity is abolished or substantially reduced. Combining with oocyte surface biotinylation experiments, we demonstrated that RACK1 inhibits the function of Pkd2L1 channel on the plasma membrane in addition to reducing its total and plasma membrane expression. Overexpressing Pkd2L1 N- or C-terminal fragments as potential blocking peptides for the Pkd2L1-RACK1 interaction, we found that Pkd2L1 N-terminal fragment Met(1)-Pro(45), but not Ile(40)-Ile(97) or C-terminal fragments, abolishes the inhibition of Pkd2L1 channel by overexpressed and oocyte-native RACK1 likely through disrupting the Pkd2L1-RACK1 association. Taken together, our study demonstrated that RACK1 inhibits Pkd2L1 channel function through binding to domain Met(1)-Pro(45) of Pkd2L1. Thus, Pkd2L1 is a novel target channel whose function is regulated by the versatile scaffolding protein RACK1.


Asunto(s)
Canales de Calcio/química , Canales de Calcio/fisiología , Proteínas de Unión al GTP/química , Proteínas de Unión al GTP/fisiología , Proteínas de Neoplasias/química , Proteínas de Neoplasias/fisiología , Receptores de Superficie Celular/química , Receptores de Superficie Celular/fisiología , Animales , Sitios de Unión/fisiología , Calcio/metabolismo , Canales de Calcio/genética , Proteínas de Unión al GTP/genética , Células HEK293 , Humanos , Mutagénesis/fisiología , Proteínas de Neoplasias/genética , Oocitos/fisiología , Técnicas de Placa-Clamp , Dominios y Motivos de Interacción de Proteínas/fisiología , Estructura Terciaria de Proteína/fisiología , ARN Mensajero/farmacología , Receptores de Cinasa C Activada , Receptores de Superficie Celular/genética , Técnicas del Sistema de Dos Híbridos , Xenopus
11.
JCI Insight ; 8(24)2023 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-37943614

RESUMEN

HNF1A haploinsufficiency underlies the most common form of human monogenic diabetes (HNF1A-maturity onset diabetes of the young [HNF1A-MODY]), and hypomorphic HNF1A variants confer type 2 diabetes risk. But a lack of experimental systems for interrogating mature human islets has limited our understanding of how the transcription factor HNF1α regulates adult islet function. Here, we combined conditional genetic targeting in human islet cells, RNA-Seq, chromatin mapping with cleavage under targets and release using nuclease (CUT&RUN), and transplantation-based assays to determine HNF1α-regulated mechanisms in adult human pancreatic α and ß cells. Short hairpin RNA-mediated (shRNA-mediated) suppression of HNF1A in primary human pseudoislets led to blunted insulin output and dysregulated glucagon secretion after transplantation in mice, recapitulating phenotypes observed in patients with diabetes. These deficits corresponded with altered expression of genes encoding factors critical for hormone secretion, including calcium channel subunits, ATPase transporters, and extracellular matrix constituents. Additionally, HNF1A loss led to upregulation of transcriptional repressors, providing evidence for a mechanism of transcriptional derepression through HNF1α. CUT&RUN mapping of HNF1α DNA binding sites in primary human islets imputed a subset of HNF1α-regulated genes as direct targets. These data elucidate mechanistic links between HNF1A loss and diabetic phenotypes in mature human α and ß cells.


Asunto(s)
Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Animales , Humanos , Ratones , Diabetes Mellitus Tipo 2/metabolismo , Regulación de la Expresión Génica , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Páncreas/metabolismo
12.
Diabetes ; 72(9): 1277-1288, 2023 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-37364047

RESUMEN

Carboxypeptidase E (CPE) facilitates the conversion of prohormones into mature hormones and is highly expressed in multiple neuroendocrine tissues. Carriers of CPE mutations have elevated plasma proinsulin and develop severe obesity and hyperglycemia. We aimed to determine whether loss of Cpe in pancreatic ß-cells disrupts proinsulin processing and accelerates development of diabetes and obesity in mice. Pancreatic ß-cell-specific Cpe knockout mice (ßCpeKO; Cpefl/fl x Ins1Cre/+) lack mature insulin granules and have elevated proinsulin in plasma; however, glucose-and KCl-stimulated insulin secretion in ßCpeKO islets remained intact. High-fat diet-fed ßCpeKO mice showed weight gain and glucose tolerance comparable with those of Wt littermates. Notably, ß-cell area was increased in chow-fed ßCpeKO mice and ß-cell replication was elevated in ßCpeKO islets. Transcriptomic analysis of ßCpeKO ß-cells revealed elevated glycolysis and Hif1α-target gene expression. On high glucose challenge, ß-cells from ßCpeKO mice showed reduced mitochondrial membrane potential, increased reactive oxygen species, reduced MafA, and elevated Aldh1a3 transcript levels. Following multiple low-dose streptozotocin injections, ßCpeKO mice had accelerated development of hyperglycemia with reduced ß-cell insulin and Glut2 expression. These findings suggest that Cpe and proper proinsulin processing are critical in maintaining ß-cell function during the development of hyperglycemia. ARTICLE HIGHLIGHTS: Carboxypeptidase E (Cpe) is an enzyme that removes the carboxy-terminal arginine and lysine residues from peptide precursors. Mutations in CPE lead to obesity and type 2 diabetes in humans, and whole-body Cpe knockout or mutant mice are obese and hyperglycemic and fail to convert proinsulin to insulin. We show that ß-cell-specific Cpe deletion in mice (ßCpeKO) does not lead to the development of obesity or hyperglycemia, even after prolonged high-fat diet treatment. However, ß-cell proliferation rate and ß-cell area are increased, and the development of hyperglycemia induced by multiple low-dose streptozotocin injections is accelerated in ßCpeKO mice.


Asunto(s)
Carboxipeptidasa H , Diabetes Mellitus Tipo 2 , Hiperglucemia , Células Secretoras de Insulina , Islotes Pancreáticos , Animales , Ratones , Carboxipeptidasa H/genética , Carboxipeptidasa H/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Glucosa/metabolismo , Hiperglucemia/genética , Hiperglucemia/metabolismo , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Ratones Noqueados , Obesidad/metabolismo , Proinsulina/metabolismo , Estreptozocina
13.
bioRxiv ; 2023 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-36711922

RESUMEN

Altered function and gene regulation of pancreatic islet beta cells is a hallmark of type 2 diabetes (T2D), but a comprehensive understanding of mechanisms driving T2D is still missing. Here we integrate information from measurements of chromatin activity, gene expression and function in single beta cells with genetic association data to identify disease-causal gene regulatory changes in T2D. Using machine learning on chromatin accessibility data from 34 non-diabetic, pre-T2D and T2D donors, we robustly identify two transcriptionally and functionally distinct beta cell subtypes that undergo an abundance shift in T2D. Subtype-defining active chromatin is enriched for T2D risk variants, suggesting a causal contribution of subtype identity to T2D. Both subtypes exhibit activation of a stress-response transcriptional program and functional impairment in T2D, which is likely induced by the T2D-associated metabolic environment. Our findings demonstrate the power of multimodal single-cell measurements combined with machine learning for identifying mechanisms of complex diseases.

14.
Nat Genet ; 55(6): 984-994, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37231096

RESUMEN

Dysfunctional pancreatic islet beta cells are a hallmark of type 2 diabetes (T2D), but a comprehensive understanding of the underlying mechanisms, including gene dysregulation, is lacking. Here we integrate information from measurements of chromatin accessibility, gene expression and function in single beta cells with genetic association data to nominate disease-causal gene regulatory changes in T2D. Using machine learning on chromatin accessibility data from 34 nondiabetic, pre-T2D and T2D donors, we identify two transcriptionally and functionally distinct beta cell subtypes that undergo an abundance shift during T2D progression. Subtype-defining accessible chromatin is enriched for T2D risk variants, suggesting a causal contribution of subtype identity to T2D. Both beta cell subtypes exhibit activation of a stress-response transcriptional program and functional impairment in T2D, which is probably induced by the T2D-associated metabolic environment. Our findings demonstrate the power of multimodal single-cell measurements combined with machine learning for characterizing mechanisms of complex diseases.


Asunto(s)
Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Humanos , Diabetes Mellitus Tipo 2/genética , Multiómica , Células Secretoras de Insulina/metabolismo , Regulación de la Expresión Génica , Cromatina/metabolismo
15.
Front Mol Neurosci ; 15: 769725, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36090255

RESUMEN

Dendritic spines (DS) are tiny protrusions implicated in excitatory postsynaptic responses in the CNS. To achieve their function, DS concentrate a high density of ion channels and dynamic actin networks in a tiny specialized compartment. However, to date there is no direct information on DS ionic conductances. Here, we used several experimental techniques to obtain direct electrical information from DS of the adult mouse hippocampus. First, we optimized a method to isolate DS from the dissected hippocampus. Second, we used the lipid bilayer membrane (BLM) reconstitution and patch clamping techniques and obtained heretofore unavailable electrical phenotypes on ion channels present in the DS membrane. Third, we also patch clamped DS directly in cultured adult mouse hippocampal neurons, to validate the electrical information observed with the isolated preparation. Electron microscopy and immunochemistry of PDS-95 and NMDA receptors and intrinsic actin networks confirmed the enrichment of the isolated DS preparation, showing open and closed DS, and multi-headed DS. The preparation was used to identify single channel activities and "whole-DS" electrical conductance. We identified NMDA and Ca2+-dependent intrinsic electrical activity in isolated DS and in situ DS of cultured adult mouse hippocampal neurons. In situ recordings in the presence of local NMDA, showed that individual DS intrinsic electrical activity often back-propagated to the dendrite from which it sprouted. The DS electrical oscillations were modulated by changes in actin cytoskeleton dynamics by addition of the F-actin disrupter agent, cytochalasin D, and exogenous actin-binding proteins. The data indicate that DS are elaborate excitable electrical devices, whose activity is a functional interplay between ion channels and the underlying actin networks. The data argue in favor of the active contribution of individual DS to the electrical activity of neurons at the level of both the membrane conductance and cytoskeletal signaling.

16.
PLoS One ; 17(1): e0263005, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35081145

RESUMEN

The objective of this study is to optimize the cryopreservation of dissociated islet cells and obtain functional cells that can be used in single-cell transcriptome studies on the pathology and treatment of diabetes. Using an iterative graded freezing approach we obtained viable cells after cooling in 10% dimethyl sulfoxide and 6% hydroxyethyl starch at 1°C/min to -40°C, storage in liquid nitrogen, rapid thaw, and removal of cryoprotectants by serial dilution. The expression of epithelial cell adhesion molecule declined immediately after thaw, but recovered after overnight incubation, while that of an endocrine cell marker (HPi2) remained high after cryopreservation. Patch-clamp electrophysiology revealed differences in channel activities and exocytosis of various islet cell types; however, exocytotic responses, and the biophysical properties of voltage-gated Na+ and Ca2+ channels, are sustained after cryopreservation. Single-cell RNA sequencing indicates that overall transcriptome and crucial exocytosis genes are comparable between fresh and cryopreserved dispersed human islet cells. Thus, we report an optimized procedure for cryopreserving dispersed islet cells that maintained their membrane integrity, along with their molecular and functional phenotypes. Our findings will not only provide a ready source of cells for investigating cellular mechanisms in diabetes but also for bio-engineering pseudo-islets and islet sheets for modeling studies and potential transplant applications.


Asunto(s)
Criopreservación , Islotes Pancreáticos/metabolismo , Adolescente , Adulto , Anciano , Antígenos de Diferenciación/metabolismo , Canales de Calcio/metabolismo , Crioprotectores/farmacología , Femenino , Humanos , Islotes Pancreáticos/citología , Trasplante de Islotes Pancreáticos , Masculino , Persona de Mediana Edad , RNA-Seq , Análisis de la Célula Individual , Canales de Sodio/metabolismo
17.
Nat Commun ; 13(1): 735, 2022 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-35136059

RESUMEN

Insulin receptor (Insr) protein is present at higher levels in pancreatic ß-cells than in most other tissues, but the consequences of ß-cell insulin resistance remain enigmatic. Here, we use an Ins1cre knock-in allele to delete Insr specifically in ß-cells of both female and male mice. We compare experimental mice to Ins1cre-containing littermate controls at multiple ages and on multiple diets. RNA-seq of purified recombined ß-cells reveals transcriptomic consequences of Insr loss, which differ between female and male mice. Action potential and calcium oscillation frequencies are increased in Insr knockout ß-cells from female, but not male mice, whereas only male ßInsrKO islets have reduced ATP-coupled oxygen consumption rate and reduced expression of genes involved in ATP synthesis. Female ßInsrKO and ßInsrHET mice exhibit elevated insulin release in ex vivo perifusion experiments, during hyperglycemic clamps, and following i.p. glucose challenge. Deletion of Insr does not alter ß-cell area up to 9 months of age, nor does it impair hyperglycemia-induced proliferation. Based on our data, we adapt a mathematical model to include ß-cell insulin resistance, which predicts that ß-cell Insr knockout improves glucose tolerance depending on the degree of whole-body insulin resistance. Indeed, glucose tolerance is significantly improved in female ßInsrKO and ßInsrHET mice compared to controls at 9, 21 and 39 weeks, and also in insulin-sensitive 4-week old males. We observe no improved glucose tolerance in older male mice or in high fat diet-fed mice, corroborating the prediction that global insulin resistance obscures the effects of ß-cell specific insulin resistance. The propensity for hyperinsulinemia is associated with mildly reduced fasting glucose and increased body weight. We further validate our main in vivo findings using an Ins1-CreERT transgenic line and find that male mice have improved glucose tolerance 4 weeks after tamoxifen-mediated Insr deletion. Collectively, our data show that ß-cell insulin resistance in the form of reduced ß-cell Insr contributes to hyperinsulinemia in the context of glucose stimulation, thereby improving glucose homeostasis in otherwise insulin sensitive sex, dietary and age contexts.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Hiperinsulinismo/genética , Resistencia a la Insulina/genética , Células Secretoras de Insulina/metabolismo , Receptor de Insulina/genética , Animales , Conjuntos de Datos como Asunto , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Femenino , Técnicas de Sustitución del Gen , Técnicas de Inactivación de Genes , Glucosa/metabolismo , Humanos , Hiperinsulinismo/sangre , Hiperinsulinismo/metabolismo , Hiperinsulinismo/patología , Insulina/sangre , Insulina/metabolismo , Células Secretoras de Insulina/patología , Masculino , Ratones , Ratones Transgénicos , RNA-Seq , Receptor de Insulina/deficiencia , Factores Sexuales
18.
Cell Metab ; 34(2): 256-268.e5, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-35108513

RESUMEN

In diabetes, glucagon secretion from pancreatic α cells is dysregulated. The underlying mechanisms, and whether dysfunction occurs uniformly among cells, remain unclear. We examined α cells from human donors and mice using electrophysiological, transcriptomic, and computational approaches. Rising glucose suppresses α cell exocytosis by reducing P/Q-type Ca2+ channel activity, and this is disrupted in type 2 diabetes (T2D). Upon high-fat feeding of mice, α cells shift toward a "ß cell-like" electrophysiological profile in concert with indications of impaired identity. In human α cells we identified links between cell membrane properties and cell surface signaling receptors, mitochondrial respiratory chain complex assembly, and cell maturation. Cell-type classification using machine learning of electrophysiology data demonstrated a heterogenous loss of "electrophysiologic identity" in α cells from donors with type 2 diabetes. Indeed, a subset of α cells with impaired exocytosis is defined by an enrichment in progenitor and lineage markers and upregulation of an immature transcriptomic phenotype, suggesting important links between α cell maturation state and dysfunction.


Asunto(s)
Diabetes Mellitus Tipo 2 , Células Secretoras de Glucagón , Islotes Pancreáticos , Animales , Diabetes Mellitus Tipo 2/metabolismo , Exocitosis/fisiología , Glucagón/metabolismo , Células Secretoras de Glucagón/metabolismo , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Ratones
19.
JCI Insight ; 6(18)2021 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-34428183

RESUMEN

Islet-enriched transcription factors (TFs) exert broad control over cellular processes in pancreatic α and ß cells, and changes in their expression are associated with developmental state and diabetes. However, the implications of heterogeneity in TF expression across islet cell populations are not well understood. To define this TF heterogeneity and its consequences for cellular function, we profiled more than 40,000 cells from normal human islets by single-cell RNA-Seq and stratified α and ß cells based on combinatorial TF expression. Subpopulations of islet cells coexpressing ARX/MAFB (α cells) and MAFA/MAFB (ß cells) exhibited greater expression of key genes related to glucose sensing and hormone secretion relative to subpopulations expressing only one or neither TF. Moreover, all subpopulations were identified in native pancreatic tissue from multiple donors. By Patch-Seq, MAFA/MAFB-coexpressing ß cells showed enhanced electrophysiological activity. Thus, these results indicate that combinatorial TF expression in islet α and ß cells predicts highly functional, mature subpopulations.


Asunto(s)
Células Secretoras de Glucagón/metabolismo , Células Secretoras de Insulina/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Adulto , Fenómenos Electrofisiológicos , Expresión Génica , Células Secretoras de Glucagón/fisiología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/fisiología , Factores de Transcripción Maf de Gran Tamaño/genética , Factores de Transcripción Maf de Gran Tamaño/metabolismo , Factor de Transcripción MafB/genética , Factor de Transcripción MafB/metabolismo , Persona de Mediana Edad , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Transcriptoma , Adulto Joven
20.
Cell Metab ; 31(5): 1017-1031.e4, 2020 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-32302527

RESUMEN

Impaired function of pancreatic islet cells is a major cause of metabolic dysregulation and disease in humans. Despite this, it remains challenging to directly link physiological dysfunction in islet cells to precise changes in gene expression. Here we show that single-cell RNA sequencing combined with electrophysiological measurements of exocytosis and channel activity (patch-seq) can be used to link endocrine physiology and transcriptomes at the single-cell level. We collected 1,369 patch-seq cells from the pancreata of 34 human donors with and without diabetes. An analysis of function and gene expression networks identified a gene set associated with functional heterogeneity in ß cells that can be used to predict electrophysiology. We also report transcriptional programs underlying dysfunction in type 2 diabetes and extend this approach to cryopreserved cells from donors with type 1 diabetes, generating a valuable resource for understanding islet cell heterogeneity in health and disease.


Asunto(s)
Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Islotes Pancreáticos/metabolismo , Análisis de la Célula Individual , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 2/genética , Humanos , Análisis de Secuencia de ARN , Transcriptoma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA