Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Anal Chem ; 91(9): 5794-5801, 2019 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-30843680

RESUMEN

Comprehensive phosphoproteomic analysis of small populations of cells remains a daunting task due primarily to the insufficient MS signal intensity from low concentrations of enriched phosphopeptides. Isobaric labeling has a unique multiplexing feature where the "total" peptide signal from all channels (or samples) triggers MS/MS fragmentation for peptide identification, while the reporter ions provide quantitative information. In light of this feature, we tested the concept of using a "boosting" sample (e.g., a biological sample mimicking the study samples but available in a much larger quantity) in multiplexed analysis to enable sensitive and comprehensive quantitative phosphoproteomic measurements with <100 000 cells. This simple boosting to amplify signal with isobaric labeling (BASIL) strategy increased the overall number of quantifiable phosphorylation sites more than 4-fold. Good reproducibility in quantification was demonstrated with a median CV of 15.3% and Pearson correlation coefficient of 0.95 from biological replicates. A proof-of-concept experiment demonstrated the ability of BASIL to distinguish acute myeloid leukemia cells based on the phosphoproteome data. Moreover, in a pilot application, this strategy enabled quantitative analysis of over 20 000 phosphorylation sites from human pancreatic islets treated with interleukin-1ß and interferon-γ. Together, this signal boosting strategy provides an attractive solution for comprehensive and quantitative phosphoproteome profiling of relatively small populations of cells where traditional phosphoproteomic workflows lack sufficient sensitivity.


Asunto(s)
Interferón gamma/farmacología , Interleucina-1beta/farmacología , Islotes Pancreáticos/metabolismo , Fosfopéptidos/metabolismo , Fosfoproteínas/metabolismo , Coloración y Etiquetado/métodos , Espectrometría de Masas en Tándem/métodos , Antivirales/farmacología , Humanos , Islotes Pancreáticos/citología , Islotes Pancreáticos/efectos de los fármacos , Fosforilación
2.
J Biol Chem ; 292(43): 17598-17608, 2017 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-28860191

RESUMEN

Preservation of insulin-secreting ß-cells is an important goal for therapies aimed at restoring normoglycemia in patients with diabetes. One approach, the inhibition of histone deacetylases (HDACs), has been reported to suppress pancreatic islet inflammation and ß-cell apoptosis in vitro In this report, we demonstrate the efficacy of HDAC inhibitors (HDACi) in vivo We show that daily administration of BRD3308, an isoform-selective HDAC3 inhibitor, for 2 weeks to female nonobese diabetic (NOD) mice, beginning at 3 weeks of age, followed by twice-weekly injections until age 25 weeks, protects the animals from diabetes. The preservation of ß-cells was because of a significant decrease in islet infiltration of mononuclear cells. Moreover, the BRD3308 treatment increased basal insulin secretion from islets cultured in vitro All metabolic tissues tested in vehicle- or BRD3308-treated groups showed virtually no sign of immune cell infiltration, except minimal infiltration in white adipose tissue in animals treated with the highest BRD3308 dose (10 mg/kg), providing additional evidence of protection from immune attack in the treated groups. Furthermore, pancreata from animals treated with 10 mg/kg BRD3308 exhibited significantly decreased numbers of apoptotic ß-cells compared with those treated with vehicle or low-dose BRD3308. Finally, animals treated with 1 or 10 mg/kg BRD3308 had enhanced ß-cell proliferation. These in vivo results point to the potential use of selective HDAC3 inhibitors as a therapeutic approach to suppress pancreatic islet infiltration and prevent ß-cell death with the long-term goal of limiting the progression of type 1 diabetes.


Asunto(s)
Apoptosis/efectos de los fármacos , Diabetes Mellitus Experimental/prevención & control , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/metabolismo , Células Secretoras de Insulina/enzimología , Tejido Adiposo Blanco/enzimología , Tejido Adiposo Blanco/patología , Animales , Diabetes Mellitus Experimental/enzimología , Diabetes Mellitus Experimental/patología , Femenino , Células Secretoras de Insulina/patología , Ratones , Ratones Endogámicos NOD
3.
Proc Natl Acad Sci U S A ; 111(3): 1192-7, 2014 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-24395790

RESUMEN

In insulin resistant states such as type 2 diabetes, there is a high demand on the ß-cell to synthesize and secrete insulin, which challenges the ability of the endoplasmic reticulum (ER) to synthesize and fold nascent proteins. This creates a state of ER stress that triggers a coordinated program referred to as the unfolded protein response (UPR) that attempts to restore ER homeostasis. We identified a role for the p85α regulatory subunit of PI3K to modulate the UPR by promoting the nuclear localization of X-box binding protein 1, a transcription factor central to the UPR. In the present study we demonstrate that reducing p85α expression in ß-cells can markedly delay the onset and severity of the diabetic phenotype observed in Akita(+/-) mice, which express a mutant insulin molecule. This is due to a decrease in activation of ER stress-dependent apoptotic pathways and a preservation of ß-cell mass and function. These data demonstrate that modulation of p85α can protect pancreatic ß-cells from ER stress, pointing to a potentially therapeutic target in diabetic states.


Asunto(s)
Apoptosis , Fosfatidilinositol 3-Quinasa Clase Ia/deficiencia , Estrés del Retículo Endoplásmico , Retículo Endoplásmico/metabolismo , Células Secretoras de Insulina/citología , Alelos , Animales , Diabetes Mellitus Tipo 2/metabolismo , Regulación Enzimológica de la Expresión Génica , Genotipo , Glucosa/metabolismo , Hiperglucemia/metabolismo , Insulina/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Tamaño de los Órganos , Estrés Oxidativo , Páncreas/fisiología , Fenotipo , Desnaturalización Proteica , Pliegue de Proteína , Factores de Tiempo
4.
J Proteome Res ; 14(8): 3111-3122, 2015 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-26151086

RESUMEN

Compensatory islet response is a distinct feature of the prediabetic insulin-resistant state in humans and rodents. To identify alterations in the islet proteome that characterize the adaptive response, we analyzed islets from 5 month old male control, high-fat diet fed (HFD), or obese ob/ob mice by LC-MS/MS and quantified ~1100 islet proteins (at least two peptides) with a false discovery rate < 1%. Significant alterations in abundance were observed for ~350 proteins among groups. The majority of alterations were common to both models, and the changes of a subset of ~40 proteins and 12 proteins were verified by targeted quantification using selected reaction monitoring and western blots, respectively. The insulin-resistant islets in both groups exhibited reduced expression of proteins controlling energy metabolism, oxidative phosphorylation, hormone processing, and secretory pathways. Conversely, an increased expression of molecules involved in protein synthesis and folding suggested effects in endoplasmic reticulum stress response, cell survival, and proliferation in both insulin-resistant models. In summary, we report a unique comparison of the islet proteome that is focused on the compensatory response in two insulin-resistant rodent models that are not overtly diabetic. These data provide a valuable resource of candidate proteins to the scientific community to undertake further studies aimed at enhancing ß-cell mass in patients with diabetes. The data are available via the MassIVE repository, under accession no. MSV000079093.


Asunto(s)
Resistencia a la Insulina , Islotes Pancreáticos/metabolismo , Proteoma/metabolismo , Proteómica/métodos , Secuencia de Aminoácidos , Animales , Western Blotting , Cromatografía Liquida , Dieta Alta en Grasa , Masculino , Ratones Endogámicos C57BL , Ratones Obesos , Datos de Secuencia Molecular , Espectrometría de Masas en Tándem
5.
J Biol Chem ; 288(8): 5353-6, 2013 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-23306198

RESUMEN

Maturity onset diabetes of the young (MODY) is an autosomal dominant disease. Despite extensive research, the mechanism by which a mutant MODY gene results in monogenic diabetes is not yet clear due to the inaccessibility of patient samples. Induced pluripotency and directed differentiation toward the pancreatic lineage are now viable and attractive methods to uncover the molecular mechanisms underlying MODY. Here we report, for the first time, the derivation of human induced pluripotent stem cells (hiPSCs) from patients with five types of MODY: MODY1 (HNF4A), MODY2 (GCK), MODY3 (HNF1A), MODY5 (HNF1B), and MODY8 (CEL) with a polycistronic lentiviral vector expressing a Cre-excisable human "stem cell cassette" containing the four reprogramming factors OCT4, KLF4, SOX2, and CMYC. These MODY-hiPSCs morphologically resemble human pluripotent stem cells (hPSCs), express pluripotency markers OCT4, SOX2, NANOG, SSEA-4, and TRA-1-60, give rise to derivatives of the three germ layers in a teratoma assay, and are karyotypically normal. Overall, our MODY-hiPSCs serve as invaluable tools to dissect the role of MODY genes in the development of pancreas and islet cells and to evaluate their significance in regulating beta cell function. This knowledge will aid future attempts aimed at deriving functional mature beta cells from hPSCs.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Diabetes Mellitus Tipo 2/metabolismo , Regulación de la Expresión Génica , Células Madre Pluripotentes Inducidas/citología , Adolescente , Adulto , Biopsia/métodos , Niño , Femenino , Fibroblastos/citología , Humanos , Células Secretoras de Insulina/citología , Islotes Pancreáticos/citología , Cariotipificación , Factor 4 Similar a Kruppel , Masculino , Persona de Mediana Edad , Modelos Genéticos , Páncreas/metabolismo , Linaje , Piel/metabolismo , Factores de Transcripción/metabolismo
6.
Am J Physiol Endocrinol Metab ; 307(10): E906-18, 2014 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-25249504

RESUMEN

Studies in both humans and rodents suggest that maternal diabetes leads to a higher risk of the fetus developing impaired glucose tolerance and obesity during adulthood. However, the impact of hyperinsulinemia in the mother on glucose homeostasis in the offspring has not been fully explored. We aimed to determine the consequences of maternal insulin resistance on offspring metabolism and endocrine pancreas development using the LIRKO mouse model, which exhibits sustained hyperinsulinemia and transient increase in blood glucose concentrations during pregnancy. We examined control offspring born to either LIRKO or control mothers on embryonic days 13.5, 15.5, and 17.5 and postpartum days 0, 4, and 10. Control offspring born to LIRKO mothers displayed low birth weights and subsequently rapidly gained weight, and their blood glucose and plasma insulin concentrations were higher than offspring born to control mothers in early postnatal life. In addition, concentrations of plasma leptin, glucagon, and active GLP-1 were higher in control pups from LIRKO mothers. Analyses of the endocrine pancreas revealed significantly reduced ß-cell area in control offspring of LIRKO mothers shortly after birth. ß-Cell proliferation and total islet number were also lower in control offspring of LIRKO mothers during early postnatal days. Together, these data indicate that maternal hyperinsulinemia and the transient hyperglycemia impair endocrine pancreas development in the control offspring and induce multiple metabolic alterations in early postnatal life. The relatively smaller ß-cell mass/area and ß-cell proliferation in these control offspring suggest cell-autonomous epigenetic mechanisms in the regulation of islet growth and development.


Asunto(s)
Glucemia/metabolismo , Diabetes Gestacional , Hiperglucemia/metabolismo , Hiperinsulinismo/metabolismo , Resistencia a la Insulina , Islotes Pancreáticos/patología , Complicaciones del Embarazo/metabolismo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Animales , Animales Recién Nacidos , Proliferación Celular , Modelos Animales de Enfermedad , Femenino , Glucagón/sangre , Péptido 1 Similar al Glucagón/sangre , Hiperglucemia/sangre , Hiperglucemia/patología , Hiperinsulinismo/sangre , Hiperinsulinismo/patología , Insulina/sangre , Células Secretoras de Insulina/citología , Leptina/sangre , Ratones , Tamaño de los Órganos , Fenotipo , Embarazo , Complicaciones del Embarazo/sangre , Complicaciones del Embarazo/patología , Efectos Tardíos de la Exposición Prenatal/sangre , Efectos Tardíos de la Exposición Prenatal/patología , Aumento de Peso
7.
Obesity (Silver Spring) ; 31(4): 1024-1037, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36854900

RESUMEN

OBJECTIVE: G-protein coupled receptor 75 (GPR75) has been identified as the high-affinity receptor of 20-hydroxyeicosatetraenoic acid (20-HETE), a vasoactive and proinflammatory lipid, and mice overproducing 20-HETE have been shown to develop insulin resistance when fed a high-fat diet (HFD), which was prevented by a 20-HETE receptor blocker. Simultaneously, a large-scale exome sequencing of 640,000 subjects identified an association between loss-of-function GPR75 variants and protection against obesity. METHODS: Wild-type (WT) and Gpr75-deficient mice were placed on HFD for 14 weeks, and their obesity phenotype was examined. RESULTS: Male and female Gpr75 null (knockout [KO]) and heterozygous mice gained less weight than WT mice when placed on HFD. KO mice maintained the same level of energy expenditure during HFD feeding, whereas WT mice showed a significant reduction in energy expenditure. Diet-driven adiposity and adipocyte hypertrophy were greatly lessened in Gpr75-deficient mice. HFD-fed KO mice did not develop insulin resistance. Adipose tissue from Gpr75-deficient mice had increased expression of thermogenic genes and decreased levels of inflammatory markers. Moreover, insulin signaling, which was impaired in HFD-fed WT mice, was unchanged in KO mice. CONCLUSIONS: These findings suggest that GPR75 is an important player in the control of metabolism and glucose homeostasis and a likely novel therapeutic target to combat obesity-driven metabolic disorders.


Asunto(s)
Dieta Alta en Grasa , Resistencia a la Insulina , Ratones , Masculino , Femenino , Animales , Dieta Alta en Grasa/efectos adversos , Resistencia a la Insulina/genética , Obesidad/genética , Obesidad/prevención & control , Obesidad/metabolismo , Tejido Adiposo/metabolismo , Ratones Noqueados , Ratones Endogámicos C57BL
8.
Adv Pharmacol ; 97: 229-255, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37236760

RESUMEN

Vascular function is dynamically regulated and dependent on a bevy of cell types and factors that work in concert across the vasculature. The vasoactive eicosanoid, 20-Hydroxyeicosatetraenoic acid (20-HETE) is a key player in this system influencing the sensitivity of the vasculature to constrictor stimuli, regulating endothelial function, and influencing the renin angiotensin system (RAS), as well as being a driver of vascular remodeling independent of blood pressure elevations. Several of these bioactions are accomplished through the ligand-receptor pairing between 20-HETE and its high-affinity receptor, GPR75. This 20-HETE axis is at the root of various vascular pathologies and processes including ischemia induced angiogenesis, arteriogenesis, septic shock, hypertension, atherosclerosis, myocardial infarction and cardiometabolic diseases including diabetes and insulin resistance. Pharmacologically, several preclinical tools have been developed to disrupt the 20-HETE axis including 20-HETE synthesis inhibitors (DDMS and HET0016), synthetic 20-HETE agonist analogues (20-5,14-HEDE and 20-5,14-HEDGE) and 20-HETE receptor blockers (AAA and 20-SOLA). Systemic or cell-specific therapeutic targeting of the 20-HETE-GPR75 axis continues to be an invaluable approach as studies examine the molecular underpinnings activated by 20-HETE under various physiological settings. In particular, the development and characterization of 20-HETE receptor blockers look to be a promising new class of compounds that can provide a considerable benefit to patients suffering from these cardiovascular pathologies.


Asunto(s)
Hipertensión , Sistema Renina-Angiotensina , Humanos , Ácidos Hidroxieicosatetraenoicos/metabolismo , Ácidos Hidroxieicosatetraenoicos/farmacología , Hipertensión/metabolismo , Remodelación Vascular , Receptores Acoplados a Proteínas G/metabolismo
9.
JCI Insight ; 7(16)2022 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-35819843

RESUMEN

Adaptation to increased insulin demand is mediated by ß cell proliferation and neogenesis, among other mechanisms. Although it is known that pancreatic ß cells can arise from ductal progenitors, these observations have been limited mostly to the neonatal period. We have recently reported that the duct is a source of insulin-secreting cells in adult insulin-resistant states. To further explore the signaling pathways underlying the dynamic ß cell reserve during insulin resistance, we undertook human islet and duct transplantations under the kidney capsule of immunodeficient NOD/SCID-γ (NSG) mouse models that were pregnant, were insulin-resistant, or had insulin resistance superimposed upon pregnancy (insulin resistance + pregnancy), followed by single-nucleus RNA-Seq (snRNA-Seq) on snap-frozen graft samples. We observed an upregulation of proliferation markers (e.g., NEAT1) and expression of islet endocrine cell markers (e.g., GCG and PPY), as well as mature ß cell markers (e.g., INS), in transplanted human duct grafts in response to high insulin demand. We also noted downregulation of ductal cell identity genes (e.g., KRT19 and ONECUT2) coupled with upregulation of ß cell development and insulin signaling pathways. These results indicate that subsets of ductal cells are able to gain ß cell identity and reflect a form of compensation during the adaptation to insulin resistance in both physiological and pathological states.


Asunto(s)
Resistencia a la Insulina , Adulto , Animales , Femenino , Proteínas de Homeodominio , Humanos , Recién Nacido , Insulina/metabolismo , Resistencia a la Insulina/genética , Ratones , Ratones Endogámicos NOD , Ratones Desnudos , Ratones SCID , Embarazo , RNA-Seq , Factores de Transcripción
10.
Cell Chem Biol ; 29(9): 1368-1380.e5, 2022 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-35998625

RESUMEN

Analogs of the incretin hormones Gip and Glp-1 are used to treat type 2 diabetes and obesity. Findings in experimental models suggest that manipulating several hormones simultaneously may be more effective. To identify small molecules that increase the number of incretin-expressing cells, we established a high-throughput in vivo chemical screen by using the gip promoter to drive the expression of luciferase in zebrafish. All hits increased the numbers of neurogenin 3-expressing enteroendocrine progenitors, Gip-expressing K-cells, and Glp-1-expressing L-cells. One of the hits, a dual-specificity tyrosine phosphorylation-regulated kinase (DYRK) inhibitor, additionally decreased glucose levels in both larval and juvenile fish. Knock-down experiments indicated that nfatc4, a downstream mediator of DYRKs, regulates incretin+ cell number in zebrafish, and that Dyrk1b regulates Glp-1 expression in an enteroendocrine cell line. DYRK inhibition also increased the number of incretin-expressing cells in diabetic mice, suggesting a conserved reinforcement of the enteroendocrine system, with possible implications for diabetes.


Asunto(s)
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2 , Animales , Descubrimiento de Drogas , Polipéptido Inhibidor Gástrico/metabolismo , Polipéptido Inhibidor Gástrico/uso terapéutico , Péptido 1 Similar al Glucagón/genética , Péptido 1 Similar al Glucagón/metabolismo , Péptido 1 Similar al Glucagón/uso terapéutico , Glucosa/metabolismo , Incretinas/metabolismo , Incretinas/uso terapéutico , Ratones , Tirosina , Pez Cebra/metabolismo
11.
Nat Metab ; 4(1): 76-89, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35058633

RESUMEN

MODY8 (maturity-onset diabetes of the young, type 8) is a dominantly inherited monogenic form of diabetes associated with mutations in the carboxyl ester lipase (CEL) gene expressed by pancreatic acinar cells. MODY8 patients develop childhood-onset exocrine pancreas dysfunction followed by diabetes during adulthood. However, it is unclear how CEL mutations cause diabetes. In the present study, we report the transfer of CEL proteins from acinar cells to ß-cells as a form of cross-talk between exocrine and endocrine cells. Human ß-cells show a relatively higher propensity for internalizing the mutant versus the wild-type CEL protein. After internalization, the mutant protein forms stable intracellular aggregates leading to ß-cell secretory dysfunction. Analysis of pancreas sections from a MODY8 patient reveals the presence of CEL protein in the few extant ß-cells. The present study provides compelling evidence for the mechanism by which a mutant gene expressed specifically in acinar cells promotes dysfunction and loss of ß-cells to cause diabetes.


Asunto(s)
Comunicación Celular , Diabetes Mellitus Tipo 2/etiología , Diabetes Mellitus Tipo 2/metabolismo , Células Secretoras de Insulina/metabolismo , Páncreas Exocrino/metabolismo , Células Acinares/metabolismo , Animales , Humanos , Inmunohistoquímica , Islotes Pancreáticos/metabolismo , Trasplante de Islotes Pancreáticos , Lipasa/química , Lipasa/genética , Lipasa/metabolismo , Ratones , Mutación , Páncreas/metabolismo , Páncreas/patología , Transporte de Proteínas , Solubilidad
12.
Diabetes ; 71(5): 945-960, 2022 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-35212729

RESUMEN

Growing evidence indicates an important link between gut microbiota, obesity, and metabolic syndrome. Alterations in exocrine pancreatic function are also widely present in patients with diabetes and obesity. To examine this interaction, C57BL/6J mice were fed a chow diet, a high-fat diet (HFD), or an HFD plus oral vancomycin or metronidazole to modify the gut microbiome. HFD alone leads to a 40% increase in pancreas weight, decreased glucagon-like peptide 1 and peptide YY levels, and increased glucose-dependent insulinotropic peptide in the plasma. Quantitative proteomics identified 138 host proteins in fecal samples of these mice, of which 32 were significantly changed by the HFD. The most significant of these were the pancreatic enzymes. These changes in amylase and elastase were reversed by antibiotic treatment. These alterations could be reproduced by transferring gut microbiota from donor C57BL/6J mice to germ-free mice. By contrast, antibiotics had no effect on pancreatic size or exocrine function in C57BL/6J mice fed the chow diet. Further, 1 week vancomycin administration significantly increased amylase and elastase levels in obese men with prediabetes. Thus, the alterations in gut microbiota in obesity can alter pancreatic growth, exocrine function, and gut endocrine function and may contribute to the alterations observed in patients with obesity and diabetes.


Asunto(s)
Microbioma Gastrointestinal , Amilasas , Animales , Dieta Alta en Grasa/efectos adversos , Péptido 1 Similar al Glucagón , Humanos , Ratones , Ratones Endogámicos C57BL , Obesidad/metabolismo , Páncreas/metabolismo , Elastasa Pancreática , Vancomicina/farmacología
13.
Diabetes Metab Res Rev ; 27(6): 575-83, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21584921

RESUMEN

BACKGROUND: To increase the success rate in xenogeneic islet transplantation, proper assessment of graft mass is required following transplantation. For this reason, we aimed to develop a suitable fluorescence imaging system to monitor islet xenograft survival in diabetic mice. METHODS: Adenovirus vector encoding enhanced green fluorescent protein-transduced rat pancreatic islets were transplanted under the renal capsule of streptozotocin-induced diabetic mice and the fluorescence signal was quantified over time using a cooled charge-coupled device. Non-fasting blood glucose levels were recorded during the same period. Insulin release from transduced and control islets was detected via enzyme-linked immunosorbent assay. RESULTS: Adenovirus vector encoding enhanced green fluorescent protein infection did not alter the function or survival of pancreatic islets post transduction. A direct correlation was found between the number of islets (250-750) transplanted under the kidney capsule and the blood glucose recovery. CONCLUSIONS: Fluorescence imaging appears to be a useful tool for quantitative assessment of islet cell viability post transplantation and could permit earlier detection of graft rejection.


Asunto(s)
Supervivencia de Injerto/fisiología , Proteínas Fluorescentes Verdes , Trasplante de Islotes Pancreáticos/métodos , Trasplante Heterólogo , Adenoviridae/genética , Animales , Glucemia/metabolismo , Supervivencia Celular , Diabetes Mellitus Experimental , Femenino , Fluorescencia , Masculino , Ratones , Ratones Endogámicos BALB C , Ratas , Ratas Wistar
14.
Genome Med ; 13(1): 128, 2021 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-34376240

RESUMEN

BACKGROUND: Human pancreatic islets are a central focus of research in metabolic studies. Transcriptomics is frequently used to interrogate alterations in cultured human islet cells using single-cell RNA-sequencing (scRNA-seq). We introduce single-nucleus RNA-sequencing (snRNA-seq) as an alternative approach for investigating transplanted human islets. METHODS: The Nuclei EZ protocol was used to obtain nuclear preparations from fresh and frozen human islet cells. Such preparations were first used to generate snRNA-seq datasets and compared to scRNA-seq output obtained from cells from the same donor. Finally, we employed snRNA-seq to obtain the transcriptomic profile of archived human islets engrafted in immunodeficient animals. RESULTS: We observed virtually complete concordance in identifying cell types and gene proportions as well as a strong association of global and islet cell type gene signatures between scRNA-seq and snRNA-seq applied to fresh and frozen cultured or transplanted human islet samples. CONCLUSIONS: We propose snRNA-seq as a reliable strategy to probe transcriptomic profiles of freshly harvested or frozen sources of transplanted human islet cells especially when scRNA-seq is not ideal.


Asunto(s)
Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Islotes Pancreáticos/metabolismo , Análisis de la Célula Individual , Transcriptoma , Animales , Biomarcadores , Humanos , Islotes Pancreáticos/citología , Trasplante de Islotes Pancreáticos , Ratones , Análisis de Secuencia de ADN , Análisis de la Célula Individual/métodos , Secuenciación del Exoma
15.
J Mol Endocrinol ; 66(4): 325-338, 2021 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-33875613

RESUMEN

Strategies to increase functional pancreatic beta cell mass is of great interest in diabetes-related research. TNF-related apoptosis-inducing ligand (TRAIL) is well known to promote proliferation and survival in various cell types, including vascular smooth muscle and endothelial cells. Correlation between the protective nature of TRAIL on these cells and its proliferative effect is noteworthy. TRAIL's seemingly protective/therapeutic effect in diabetes prompted us to question whether it may act as an inducer of proliferation in pancreatic beta cells. We used rat primary islet cells and MIN6 mouse beta cell line to investigate TRAIL-induced proliferation. Cell viability and/or death was analyzed by MTT, WST-1, and Annexin-V/PI assays, while proliferation rates and pathways were assessed via immunocytochemical and Western blot analyses. Receptor neutralization antibodies identified the mediator receptors. Recombinant soluble TRAIL (sTRAIL) treatment led to 1.6-fold increased proliferation in insulin-positive cells in dispersed rat islets compared to the untreated group, while adenovirus-mediated overexpression of TRAIL increased the number of proliferating beta cells up to more than six-fold. sTRAIL or adenoviral vector-mediated TRAIL overexpression induced proliferation in MIN6 cells also. TRAIL's proliferative effect was mediated via AKT activation, which was suppressed upon specific inhibition. Neutralization of each TRAIL receptor reversed the proliferative effect to some degree, with the highest level of inhibition in death receptor 5 (DR5) blockage in MIN6 cells and in decoy receptor 1 (DcR1) blockage in primary rat beta cells. Thus, TRAIL induces proliferation in rodent pancreatic beta cells through activation of the AKT pathway.


Asunto(s)
Diabetes Mellitus/genética , Células Secretoras de Insulina/metabolismo , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/genética , Miembro 10c de Receptores del Factor de Necrosis Tumoral/genética , Ligando Inductor de Apoptosis Relacionado con TNF/genética , Animales , Apoptosis/genética , Proliferación Celular/genética , Supervivencia Celular/genética , Diabetes Mellitus/patología , Células Endoteliales/metabolismo , Células Endoteliales/patología , Regulación de la Expresión Génica/genética , Humanos , Células Secretoras de Insulina/patología , Ratones , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Proteínas Proto-Oncogénicas c-akt/genética , Ratas
16.
Life Sci Alliance ; 4(4)2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33514654

RESUMEN

Highly sensitive approaches to target insulin-expressing cells would allow more effective imaging, sorting, and analysis of pancreatic ß-cells. Here, we introduce the use of a reaction-based probe, diacetylated Zinpyr1 (DA-ZP1), to image pancreatic ß-cells and ß-like cells derived from human pluripotent stem cells. We harness the high intracellular zinc concentration of ß-cells to induce a fluorescence signal in cells after administration of DA-ZP1. Given its specificity and rapid uptake by cells, we used DA-ZP1 to purify live stem cell-derived ß-like cells as confirmed by immunostaining analysis. We tested the ability of DA-ZP1 to image transplanted human islet grafts and endogenous mouse pancreatic islets in vivo after its systemic administration into mice. Thus, DA-ZP1 enables purification of insulin-secreting ß-like cells for downstream applications, such as functional studies, gene-expression, and cell-cell interaction analyses and can be used to label engrafted human islets and endogenous mouse islets in vivo.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Imagen Molecular/métodos , Sondas Moleculares , Animales , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Colorantes Fluorescentes/química , Humanos , Islotes Pancreáticos/citología , Islotes Pancreáticos/metabolismo , Ratones , Sondas Moleculares/química , Estructura Molecular
17.
Mol Metab ; 53: 101260, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34023484

RESUMEN

The regulation of glucose-stimulated insulin secretion and glucose excursion has a sensory component that operates in a sex-dependent manner. OBJECTIVE: Here, we aim to dissect the basis of the sexually dimorphic interaction between sensory neurons and pancreatic ß cells and its overall impact on insulin release and glucose homeostasis. METHODS: We used viral retrograde tracing techniques, surgical and chemodenervation models, and primary cell-based co-culture systems to uncover the biology underlying sex differences in sensory modulation of pancreatic ß-cell activity. RESULTS: Retrograde transsynaptic labeling revealed a sex difference in the density of sensory innervation in the pancreas. The number of sensory neurons emanating from the dorsal root and nodose ganglia that project in the pancreas is higher in male than in female mice. Immunostaining and confocal laser scanning microscopy confirmed the higher abundance of peri-islet sensory axonal tracts in the male pancreas. Capsaicin-induced sensory chemodenervation concomitantly enhanced glucose-stimulated insulin secretion and glucose clearance in male mice. These metabolic benefits were blunted when mice were orchidectomized prior to the ablation of sensory nerves. Interestingly, orchidectomy also lowered the density of peri-islet sensory neurons. In female mice, capsaicin treatment did not affect glucose-induced insulin secretion nor glucose excursion and ovariectomy did not modify these outcomes. Interestingly, same- and opposite-sex sensory-islet co-culture paradigms unmasked the existence of potential gonadal hormone-independent mechanisms mediating the male-female difference in sensory modulation of islet ß-cell activity. CONCLUSION: Taken together, these data suggest that the sex-biased nature of the sensory control of islet ß-cell activity is a result of a combination of neurodevelopmental inputs, sex hormone-dependent mechanisms and the potential action of somatic molecules encoded by the sex chromosome complement.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Células Receptoras Sensoriales/metabolismo , Animales , Glucemia/metabolismo , Femenino , Homeostasis , Secreción de Insulina , Masculino , Ratones , Ratones Endogámicos C57BL , Caracteres Sexuales
18.
Science ; 373(6550)2021 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-34210852

RESUMEN

Large-scale human exome sequencing can identify rare protein-coding variants with a large impact on complex traits such as body adiposity. We sequenced the exomes of 645,626 individuals from the United Kingdom, the United States, and Mexico and estimated associations of rare coding variants with body mass index (BMI). We identified 16 genes with an exome-wide significant association with BMI, including those encoding five brain-expressed G protein-coupled receptors (CALCR, MC4R, GIPR, GPR151, and GPR75). Protein-truncating variants in GPR75 were observed in ~4/10,000 sequenced individuals and were associated with 1.8 kilograms per square meter lower BMI and 54% lower odds of obesity in the heterozygous state. Knock out of Gpr75 in mice resulted in resistance to weight gain and improved glycemic control in a high-fat diet model. Inhibition of GPR75 may provide a therapeutic strategy for obesity.


Asunto(s)
Índice de Masa Corporal , Exoma/genética , Obesidad/genética , Receptores Acoplados a Proteínas G/genética , Animales , Variación Genética , Humanos , Ratones , Ratones Noqueados , Análisis de Secuencia de ADN , Aumento de Peso/genética
19.
Artículo en Inglés | MEDLINE | ID: mdl-32111717

RESUMEN

OBJECTIVE: Poor maternal and paternal environments increase the risk for obesity and diabetes in offspring, whereas maternal and paternal exercise in mice can improve offspring metabolic health. We determined the effects of combined maternal and paternal exercise on offspring health and the effects of parental exercise on offspring pancreas phenotype, a major tissue regulating glucose homeostasis. RESEARCH DESIGN AND METHODS: Breeders were high fat fed and housed±running wheels before breeding (males) and before and during gestation (females). Offspring groups were: both parents sedentary (Sed); maternal exercise only (Mat Ex); paternal exercise only (Pat Ex); and maternal+paternal exercise (Mat+Pat Ex). Offspring were sedentary, chow fed, and studied at weaning, 12, 20 and 52 weeks. RESULTS: While there was no effect of parental exercise on glucose tolerance at younger ages, at 52 weeks, offspring of Mat Ex, Pat Ex and Mat+Pat Ex displayed lower glycemia and improved glucose tolerance. The greatest effects were in offspring from parents that both exercised (Mat+Pat Ex). Offspring from Mat Ex, Pat Ex, and Mat+Pat Ex had decreased beta cell size, whereas islet size and beta cell mass only decreased in Mat+Pat Ex offspring. CONCLUSIONS: Maternal and paternal exercise have additive effects to improve glucose tolerance in offspring as they age, accompanied by changes in the offspring endocrine pancreas. These findings have important implications for the prevention and treatment of type 2 diabetes.


Asunto(s)
Glucemia/análisis , Padre , Homeostasis/fisiología , Células Secretoras de Insulina/metabolismo , Madres , Fenotipo , Condicionamiento Físico Animal/métodos , Animales , Diabetes Mellitus Tipo 2/prevención & control , Dieta Alta en Grasa , Femenino , Prueba de Tolerancia a la Glucosa , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/prevención & control , Embarazo , Conducta Sedentaria , Destete
20.
J Clin Invest ; 130(5): 2391-2407, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32250344

RESUMEN

The prevalence of nonalcoholic fatty liver disease (NAFLD) is increasing worldwide. Although gene-environment interactions have been implicated in the etiology of several disorders, the impact of paternal and/or maternal metabolic syndrome on the clinical phenotypes of offspring and the underlying genetic and epigenetic contributors of NAFLD have not been fully explored. To this end, we used the liver-specific insulin receptor knockout (LIRKO) mouse, a unique nondietary model manifesting 3 hallmarks that confer high risk for the development of NAFLD: hyperglycemia, insulin resistance, and dyslipidemia. We report that parental metabolic syndrome epigenetically reprograms members of the TGF-ß family, including neuronal regeneration-related protein (NREP) and growth differentiation factor 15 (GDF15). NREP and GDF15 modulate the expression of several genes involved in the regulation of hepatic lipid metabolism. In particular, NREP downregulation increases the protein abundance of 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR) and ATP-citrate lyase (ACLY) in a TGF-ß receptor/PI3K/protein kinase B-dependent manner, to regulate hepatic acetyl-CoA and cholesterol synthesis. Reduced hepatic expression of NREP in patients with NAFLD and substantial correlations between low serum NREP levels and the presence of steatosis and nonalcoholic steatohepatitis highlight the clinical translational relevance of our findings in the context of recent preclinical trials implicating ACLY in NAFLD progression.


Asunto(s)
Epigénesis Genética , Regulación Enzimológica de la Expresión Génica , Metabolismo de los Lípidos , Síndrome Metabólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Animales , Femenino , Síndrome Metabólico/genética , Síndrome Metabólico/patología , Ratones , Ratones Noqueados , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/patología , Embarazo , Efectos Tardíos de la Exposición Prenatal/genética , Efectos Tardíos de la Exposición Prenatal/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA