Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
2.
Genome Biol ; 25(1): 151, 2024 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-38858759

RESUMEN

Deconvolution methods infer quantitative cell type estimates from bulk measurement of mixed samples including blood and tissue. DNA methylation sequencing measures multiple CpGs per read, but few existing deconvolution methods leverage this within-read information. We develop CelFiE-ISH, which extends an existing method (CelFiE) to use within-read haplotype information. CelFiE-ISH outperforms CelFiE and other existing methods, achieving 30% better accuracy and more sensitive detection of rare cell types. We also demonstrate the importance of marker selection and of tailoring markers for haplotype-aware methods. While here we use gold-standard short-read sequencing data, haplotype-aware methods will be well-suited for long-read sequencing.


Asunto(s)
Metilación de ADN , Haplotipos , Humanos , Modelos Estadísticos , Análisis de Secuencia de ADN/métodos , Islas de CpG
3.
Nucleic Acids Res ; 52(11): 6298-6316, 2024 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-38682582

RESUMEN

Senescent cells can influence the function of tissues in which they reside, and their propensity for disease. A portion of adult human pancreatic beta cells express the senescence marker p16, yet it is unclear whether they are in a senescent state, and how this affects insulin secretion. We analyzed single-cell transcriptome datasets of adult human beta cells, and found that p16-positive cells express senescence gene signatures, as well as elevated levels of beta-cell maturation genes, consistent with enhanced functionality. Senescent human beta-like cells in culture undergo chromatin reorganization that leads to activation of enhancers regulating functional maturation genes and acquisition of glucose-stimulated insulin secretion capacity. Strikingly, Interferon-stimulated genes are elevated in senescent human beta cells, but genes encoding senescence-associated secretory phenotype (SASP) cytokines are not. Senescent beta cells in culture and in human tissue show elevated levels of cytoplasmic DNA, contributing to their increased interferon responsiveness. Human beta-cell senescence thus involves chromatin-driven upregulation of a functional-maturation program, and increased responsiveness of interferon-stimulated genes, changes that could increase both insulin secretion and immune reactivity.


Asunto(s)
Senescencia Celular , Ensamble y Desensamble de Cromatina , Células Secretoras de Insulina , Interferones , Humanos , Células Secretoras de Insulina/metabolismo , Senescencia Celular/genética , Interferones/metabolismo , Interferones/genética , Secreción de Insulina , Insulina/metabolismo , Cromatina/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Células Cultivadas , Fenotipo Secretor Asociado a la Senescencia/genética , Transcriptoma , Análisis de la Célula Individual
4.
iScience ; 27(2): 108802, 2024 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-38318380

RESUMEN

Inflammation is consistently linked to dysmetabolism. In transgenic mice (Def+/+) model the neutrophilic peptide, alpha defensin, proved atherogenic. This phenotype occurred despite favorable cholesterol and glucose levels, and lower body weight and blood pressure. In this study, integration of metabolic&behavioral phenotyping system, endocrine, biochemical and mitochondrial assessment, pathological and immunohistochemical tests, and multiple challenge tests was established to explore the metabolic impact of alpha defensin. Compared to the control group, Def+/+ mice exhibited lower total energy expenditure and carbohydrate utilization, and higher fat oxidation. Their ACTH-cortisol and thyroid profiles were intact. Intriguingly, they had low levels of glucagon, with high ammonia, uric acid, triglyceride, and lactate. Mitochondrial evaluations were normal. Overall, defensin-induced hypoglucagonemia is associated with lipolysis, restricted glucose oxidation, and enhanced wasting. Def+/+ mice may be a useful model for studying the category of lean, apparently metabolically healthy, and atherosclerotic phenotype, with insight into a potential inflammatory-metabolic link.

5.
Diabetologia ; 66(10): 1925-1942, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37480416

RESUMEN

AIM/HYPOTHESIS: Hyperglycaemia is associated with alpha cell dysfunction, leading to dysregulated glucagon secretion in type 1 and type 2 diabetes; however, the mechanisms involved are still elusive. The nutrient sensor mammalian target of rapamycin complex 1 (mTORC1) plays a major role in the maintenance of alpha cell mass and function. We studied the regulation of alpha cell mTORC1 by nutrients and its role in the development of hyperglucagonaemia in diabetes. METHODS: Alpha cell mTORC1 activity was assessed by immunostaining for phosphorylation of its downstream target, the ribosomal protein S6, and glucagon, followed by confocal microscopy on pancreatic sections and flow cytometry on dispersed human and mouse islets and the alpha cell line, αTC1-6. Metabolomics and metabolic flux were studied by 13C glucose labelling in 2.8 or 16.7 mmol/l glucose followed by LC-MS analysis. To study the role of mTORC1 in mediating hyperglucagonaemia in diabetes, we generated an inducible alpha cell-specific Rptor knockout in the Akita mouse model of diabetes and tested the effects on glucose tolerance by IPGTT and on glucagon secretion. RESULTS: mTORC1 activity was increased in alpha cells from diabetic Akita mice in parallel to the development of hyperglycaemia and hyperglucagonaemia (two- to eightfold increase). Acute exposure of mouse and human islets to amino acids stimulated alpha cell mTORC1 (3.5-fold increase), whereas high glucose concentrations inhibited mTORC1 (1.4-fold decrease). The mTORC1 response to glucose was abolished in human and mouse diabetic alpha cells following prolonged islet exposure to high glucose levels, resulting in sustained activation of mTORC1, along with increased glucagon secretion. Metabolomics and metabolic flux analysis showed that exposure to high glucose levels enhanced glycolysis, glucose oxidation and the synthesis of glucose-derived amino acids. In addition, chronic exposure to high glucose levels increased the expression of Slc7a2 and Slc38a4, which encode amino acid transporters, as well as the levels of branched-chain amino acids and methionine cycle metabolites (~1.3-fold increase for both). Finally, conditional Rptor knockout in alpha cells from adult diabetic mice inhibited mTORC1, thereby inhibiting glucagon secretion (~sixfold decrease) and improving diabetes, despite persistent insulin deficiency. CONCLUSIONS/INTERPRETATION: Alpha cell exposure to hyperglycaemia enhances amino acid synthesis and transport, resulting in sustained activation of mTORC1, thereby increasing glucagon secretion. mTORC1 therefore plays a major role in mediating alpha cell dysfunction in diabetes. DATA AVAILABILITY: All sequencing data are available from the Gene Expression Omnibus (GEO) repository (accession no. GSE154126; https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE154126 ).


Asunto(s)
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2 , Hiperglucemia , Adulto , Humanos , Animales , Glucagón , Diana Mecanicista del Complejo 1 de la Rapamicina , Glucosa , Mamíferos
6.
Cell Rep ; 41(9): 111719, 2022 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-36450253

RESUMEN

Diabetogenic ablation of beta cells in mice triggers a regenerative response whereby surviving beta cells proliferate and euglycemia is regained. Here, we identify and characterize heterogeneity in response to beta cell ablation. Efficient beta cell elimination leading to severe hyperglycemia (>28 mmol/L), causes permanent diabetes with failed regeneration despite cell cycle engagement of surviving beta cells. Strikingly, correction of glycemia via insulin, SGLT2 inhibition, or a ketogenic diet for about 3 weeks allows partial regeneration of beta cell mass and recovery from diabetes, demonstrating regenerative potential masked by extreme glucotoxicity. We identify gene expression changes in beta cells exposed to extremely high glucose levels, pointing to metabolic stress and downregulation of key cell cycle genes, suggesting failure of cell cycle completion. These findings reconcile conflicting data on the impact of glucose on beta cell regeneration and identify a glucose threshold converting glycemic load from pro-regenerative to anti-regenerative.


Asunto(s)
Diabetes Mellitus , Hiperglucemia , Células Secretoras de Insulina , Animales , Ratones , Control Glucémico , Glucosa
7.
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
8.
Diabetologia ; 64(5): 1133-1143, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33558985

RESUMEN

AIMS/HYPOTHESIS: Acute hyperglycaemia stimulates pancreatic beta cell proliferation in the mouse whereas chronic hyperglycaemia appears to be toxic. We hypothesise that this toxic effect is mediated by increased beta cell workload, unrelated to hyperglycaemia per se. METHODS: To test this hypothesis, we developed a novel mouse model of cell-autonomous increased beta cell glycolytic flux caused by a conditional heterozygous beta cell-specific mutation that activates glucokinase (GCK), mimicking key aspects of the rare human genetic disease GCK-congenital hyperinsulinism. RESULTS: In the mutant mice, we observed random and fasting hypoglycaemia (random 4.5-5.4 mmol/l and fasting 3.6 mmol/l) that persisted for 15 months. GCK activation led to increased beta cell proliferation as measured by Ki67 expression (2.7% vs 1.5%, mutant and wild-type (WT), respectively, p < 0.01) that resulted in a 62% increase in beta cell mass in young mice. However, by 8 months of age, mutant mice developed impaired glucose tolerance, which was associated with decreased absolute beta cell mass from 2.9 mg at 1.5 months to 1.8 mg at 8 months of age, with preservation of individual beta cell function. Impaired glucose tolerance was further exacerbated by a high-fat/high-sucrose diet (AUC 1796 vs 966 mmol/l × min, mutant and WT, respectively, p < 0.05). Activation of GCK was associated with an increased DNA damage response and an elevated expression of Chop, suggesting metabolic stress as a contributor to beta cell death. CONCLUSIONS/INTERPRETATION: We propose that increased workload-driven biphasic beta cell dynamics contribute to decreased beta cell function observed in long-standing congenital hyperinsulinism and type 2 diabetes.


Asunto(s)
Hiperinsulinismo Congénito/patología , Glucoquinasa/genética , Células Secretoras de Insulina/patología , Animales , Recuento de Células , Hiperinsulinismo Congénito/genética , Hiperinsulinismo Congénito/metabolismo , Diabetes Mellitus Tipo 2/etiología , Diabetes Mellitus Tipo 2/patología , Modelos Animales de Enfermedad , Femenino , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/patología , Masculino , Ratones , Ratones Transgénicos , Mutación , Tamaño de los Órganos
9.
Mol Metab ; 42: 101057, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32739450

RESUMEN

OBJECTIVE: Dedifferentiation of pancreatic ß-cells may reduce islet function in type 2 diabetes (T2D). However, the prevalence, plasticity and functional consequences of this cellular state remain unknown. METHODS: We employed single-cell RNAseq to detail the maturation program of α- and ß-cells during human ontogeny. We also compared islets from non-diabetic and T2D individuals. RESULTS: Both α- and ß-cells mature in part by repressing non-endocrine genes; however, α-cells retain hallmarks of an immature state, while ß-cells attain a full ß-cell specific gene expression program. In islets from T2D donors, both α- and ß-cells have a less mature expression profile, de-repressing the juvenile genetic program and exocrine genes and increasing expression of exocytosis, inflammation and stress response signalling pathways. These changes are consistent with the increased proportion of ß-cells displaying suboptimal function observed in T2D islets. CONCLUSIONS: These findings provide new insights into the molecular program underlying islet cell maturation during human ontogeny and the loss of transcriptomic maturity that occurs in islets of type 2 diabetics.


Asunto(s)
Desdiferenciación Celular/genética , Diabetes Mellitus Tipo 2/metabolismo , Células Secretoras de Insulina/metabolismo , Desdiferenciación Celular/fisiología , Biología Computacional/métodos , Diabetes Mellitus Tipo 2/fisiopatología , Exocitosis/fisiología , Expresión Génica/genética , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica/genética , Células Secretoras de Glucagón/metabolismo , Células Secretoras de Glucagón/fisiología , Humanos , Inflamación/metabolismo , Células Secretoras de Insulina/fisiología , Islotes Pancreáticos/metabolismo , Páncreas/metabolismo , Cultivo Primario de Células , Transducción de Señal/fisiología , Análisis de la Célula Individual/métodos , Transcriptoma/genética
10.
Mol Metab ; 27S: S25-S32, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31500828

RESUMEN

BACKGROUND: Epigenetic processes control timing and level of gene expression throughout life, during development, differentiation, and aging, and are the link to adapting gene expression profiles to environmental cues. To qualify for the definition of 'epigenetic', a change to a gene's activity must be inherited through at least one mitotic division. Epigenetic mechanisms link changes in the environment to adaptions of the genome that do not rely on changes in the DNA sequence. In the past two decades, multiple studies have aimed to identify epigenetic mechanisms, and to define their role in development, differentiation and disease. SCOPE OF REVIEW: In this review, we will focus on the current knowledge of the epigenetic control of pancreatic beta cell maturation and dysfunction and its relationship to the development of islet cell failure in diabetes. Most of the data currently available have been obtained in mice, but we will summarize studies of human data as well. We will focus here on DNA methylation, as this is the most stable epigenetic mark, and least impacted by the variables inherent in islet procurement, isolation, and culture. MAJOR CONCLUSIONS: DNA methylation patterns of beta cell are dynamic during maturation and during the diabetic process. In both cases, the changes occur at cell specific regulatory regions such as enhancers, where the methylation profile is cell type specific. Frequently, the differentially methylated regulatory elements are associated with key function genes such as PDX1, NKX6-1 and TCF7L2. During maturation, enhancers tend to become demethylated in association with increased activation of beta cell function genes and increased functionality, as indicated by glucose stimulated insulin secretion. Likewise, the changes to the DNA methylome that are present in pancreatic islets from diabetic donors are enriched in regulatory regions as well.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Epigenoma , Células Secretoras de Insulina/metabolismo , Animales , Diferenciación Celular , Metilación de ADN , Diabetes Mellitus Tipo 2/patología , Humanos
11.
J Clin Invest ; 129(1): 209-214, 2019 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-30352048

RESUMEN

The loss of insulin-secreting ß cells is characteristic among type I and type II diabetes. Stimulating proliferation to expand sources of ß cells for transplantation remains a challenge because adult ß cells do not proliferate readily. The cell cycle inhibitor p57 has been shown to control cell division in human ß cells. Expression of p57 is regulated by the DNA methylation status of the imprinting control region 2 (ICR2), which is commonly hypomethylated in Beckwith-Wiedemann syndrome patients who exhibit massive ß cell proliferation. We hypothesized that targeted demethylation of the ICR2 using a transcription activator-like effector protein fused to the catalytic domain of TET1 (ICR2-TET1) would repress p57 expression and promote cell proliferation. We report here that overexpression of ICR2-TET1 in human fibroblasts reduces p57 expression levels and increases proliferation. Furthermore, human islets overexpressing ICR2-TET1 exhibit repression of p57 with concomitant upregulation of Ki-67 while maintaining glucose-sensing functionality. When transplanted into diabetic, immunodeficient mice, the epigenetically edited islets show increased ß cell replication compared with control islets. These findings demonstrate that epigenetic editing is a promising tool for inducing ß cell proliferation, which may one day alleviate the scarcity of transplantable ß cells for the treatment of diabetes.


Asunto(s)
Síndrome de Beckwith-Wiedemann/metabolismo , Proliferación Celular , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/biosíntesis , Desmetilación del ADN , Sitios Genéticos , Células Secretoras de Insulina/metabolismo , Regulación hacia Arriba , Síndrome de Beckwith-Wiedemann/patología , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Células Secretoras de Insulina/patología , Antígeno Ki-67/biosíntesis
12.
Diabetes ; 68(2): 337-348, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30425060

RESUMEN

The sustained expression of the MAFB transcription factor in human islet ß-cells represents a distinct difference in mice. Moreover, mRNA expression of closely related and islet ß-cell-enriched MAFA does not peak in humans until after 9 years of age. We show that the MAFA protein also is weakly produced within the juvenile human islet ß-cell population and that MafB expression is postnatally restricted in mouse ß-cells by de novo DNA methylation. To gain insight into how MAFB affects human ß-cells, we developed a mouse model to ectopically express MafB in adult mouse ß-cells using MafA transcriptional control sequences. Coexpression of MafB with MafA had no overt impact on mouse ß-cells, suggesting that the human adult ß-cell MAFA/MAFB heterodimer is functionally equivalent to the mouse MafA homodimer. However, MafB alone was unable to rescue the islet ß-cell defects in a mouse mutant lacking MafA in ß-cells. Of note, transgenic production of MafB in ß-cells elevated tryptophan hydroxylase 1 mRNA production during pregnancy, which drives the serotonin biosynthesis critical for adaptive maternal ß-cell responses. Together, these studies provide novel insight into the role of MAFB in human islet ß-cells.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Factores de Transcripción Maf de Gran Tamaño/metabolismo , Factor de Transcripción MafB/metabolismo , Animales , Células Cultivadas , Inmunoprecipitación de Cromatina , Cromosomas Artificiales Bacterianos/genética , Metilación de ADN/genética , Metilación de ADN/fisiología , Femenino , Humanos , Técnicas In Vitro , Factores de Transcripción Maf de Gran Tamaño/genética , Factor de Transcripción MafB/genética , Ratones , Ratones Transgénicos , Embarazo , Triptófano Hidroxilasa/genética , Triptófano Hidroxilasa/metabolismo
13.
Elife ; 72018 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-30412050

RESUMEN

Unresolved ER stress followed by cell death is recognized as the main cause of a multitude of pathologies including neonatal diabetes. A systematic analysis of the mechanisms of ß-cell loss and dysfunction in Akita mice, in which a mutation in the proinsulin gene causes a severe form of permanent neonatal diabetes, showed no increase in ß-cell apoptosis throughout life. Surprisingly, we found that the main mechanism leading to ß-cell dysfunction is marked impairment of ß-cell growth during the early postnatal life due to transient inhibition of mTORC1, which governs postnatal ß-cell growth and differentiation. Importantly, restoration of mTORC1 activity in neonate ß-cells was sufficient to rescue postnatal ß-cell growth, and to improve diabetes. We propose a scenario for the development of permanent neonatal diabetes, possibly also common forms of diabetes, where early-life events inducing ER stress affect ß-cell mass expansion due to mTOR inhibition.


Asunto(s)
Diabetes Mellitus/genética , Estrés del Retículo Endoplásmico/genética , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Proinsulina/genética , Animales , Animales Recién Nacidos/genética , Animales Recién Nacidos/crecimiento & desarrollo , Apoptosis/genética , Diabetes Mellitus/patología , Retículo Endoplásmico/genética , Humanos , Células Secretoras de Insulina/patología , Ratones , Mutación , Pliegue de Proteína
14.
Diabetes Obes Metab ; 19 Suppl 1: 147-152, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28880481

RESUMEN

While the ß-cells of the endocrine pancreas are defined as cells with high levels of insulin production and tight stimulus-secretion coupling, the existence of functional heterogeneity among them has been known for decades. Recent advances in molecular technologies, in particular single-cell profiling on both the protein and messenger RNA level, have uncovered that ß-cells exist in several antigenically and molecularly definable states. Using antibodies to cell surface markers or multidimensional clustering of ß-cells using more than 20 protein markers by mass cytometry, 4 distinct groups of ß-cells could be differentiated. However, whether these states represent permanent cell lineages or are readily interconvertible from one group to another remains to be determined. Nevertheless, future analysis of the pathogenesis of type 1 and type 2 diabetes will certainly benefit from a growing appreciation of ß-cell heterogeneity. Here, we aim to summarize concisely the recent advances in the field and their possible impact on our understanding of ß-cell physiology and pathophysiology.


Asunto(s)
Regulación de la Expresión Génica , Células Secretoras de Insulina/fisiología , Transcriptoma , Animales , Biomarcadores/metabolismo , Investigación Biomédica/métodos , Investigación Biomédica/tendencias , Linaje de la Célula , Diabetes Mellitus Tipo 1/patología , Diabetes Mellitus Tipo 1/fisiopatología , Diabetes Mellitus Tipo 2/patología , Diabetes Mellitus Tipo 2/fisiopatología , Perfilación de la Expresión Génica/tendencias , Humanos , Insulina/genética , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Análisis de la Célula Individual/tendencias , Especificidad de la Especie
15.
Diabetologia ; 60(8): 1363-1369, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28597073

RESUMEN

Beta cells are primarily defined by their ability to produce insulin and secrete it in response to appropriate stimuli. It has been known for some time, however, that beta cells are not functionally identical to each other and that the rates of insulin synthesis and release differ from cell to cell, although the functional significance of this variability remains unclear. Recent studies have used heterogeneous gene expression to isolate and evaluate different subpopulations of beta cells and to demonstrate alterations in these subpopulations in diabetes. In the last few years, novel technologies have emerged that permit the detailed evaluation of the proteome (e.g. time-of-flight mass spectroscopy, [CyTOF]) and transcriptome (e.g. massively parallel RNA sequencing) at the single-cell level, and tools for single beta cell metabolomics and epigenomics are quickly maturing. The first wave of single beta cell proteome and transcriptome studies were published in 2016, giving a glimpse into the power, but also the limitations, of these approaches. Despite this progress, it remains unclear if the observed heterogeneity of beta cells represents stable, distinct beta cell types or, alternatively, highly dynamic beta cell states. Here we provide a concise overview of recent developments in the emerging field of beta cell heterogeneity and the implications for our understanding of beta cell biology and pathology.


Asunto(s)
Diabetes Mellitus/metabolismo , Diabetes Mellitus/patología , Células Secretoras de Insulina/metabolismo , Animales , Epigenómica , Perfilación de la Expresión Génica , Humanos , Células Secretoras de Insulina/patología , Proteómica
16.
PLoS Pathog ; 13(5): e1006294, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28467504

RESUMEN

[This corrects the article DOI: 10.1371/journal.ppat.1002350.].

17.
J Clin Invest ; 127(1): 230-243, 2017 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-27941241

RESUMEN

Type 2 diabetes is thought to involve a compromised ß cell differentiation state, but the mechanisms underlying this dysfunction remain unclear. Here, we report a key role for the TF PAX6 in the maintenance of adult ß cell identity and function. PAX6 was downregulated in ß cells of diabetic db/db mice and in WT mice treated with an insulin receptor antagonist, revealing metabolic control of expression. Deletion of Pax6 in ß cells of adult mice led to lethal hyperglycemia and ketosis that were attributed to loss of ß cell function and expansion of α cells. Lineage-tracing, transcriptome, and chromatin analyses showed that PAX6 is a direct activator of ß cell genes, thus maintaining mature ß cell function and identity. In parallel, we found that PAX6 binds promoters and enhancers to repress alternative islet cell genes including ghrelin, glucagon, and somatostatin. Chromatin analysis and shRNA-mediated gene suppression experiments indicated a similar function of PAX6 in human ß cells. We conclude that reduced expression of PAX6 in metabolically stressed ß cells may contribute to ß cell failure and α cell dysfunction in diabetes.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Cetoacidosis Diabética/metabolismo , Células Secretoras de Glucagón/metabolismo , Hiperglucemia/metabolismo , Células Secretoras de Insulina/metabolismo , Factor de Transcripción PAX6/biosíntesis , Animales , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patología , Cetoacidosis Diabética/genética , Cetoacidosis Diabética/patología , Elementos de Facilitación Genéticos , Eliminación de Gen , Regulación de la Expresión Génica , Células Secretoras de Glucagón/patología , Hiperglucemia/genética , Hiperglucemia/patología , Células Secretoras de Insulina/patología , Ratones , Ratones Transgénicos , Factor de Transcripción PAX6/genética
18.
Diabetes ; 65(10): 3028-38, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27364731

RESUMEN

Human pancreatic islets consist of multiple endocrine cell types. To facilitate the detection of rare cellular states and uncover population heterogeneity, we performed single-cell RNA sequencing (RNA-seq) on islets from multiple deceased organ donors, including children, healthy adults, and individuals with type 1 or type 2 diabetes. We developed a robust computational biology framework for cell type annotation. Using this framework, we show that α- and ß-cells from children exhibit less well-defined gene signatures than those in adults. Remarkably, α- and ß-cells from donors with type 2 diabetes have expression profiles with features seen in children, indicating a partial dedifferentiation process. We also examined a naturally proliferating α-cell from a healthy adult, for which pathway analysis indicated activation of the cell cycle and repression of checkpoint control pathways. Importantly, this replicating α-cell exhibited activated Sonic hedgehog signaling, a pathway not previously known to contribute to human α-cell proliferation. Our study highlights the power of single-cell RNA-seq and provides a stepping stone for future explorations of cellular heterogeneity in pancreatic endocrine cells.


Asunto(s)
Islotes Pancreáticos/citología , Islotes Pancreáticos/metabolismo , Transcriptoma/genética , Ciclo Celular/genética , Ciclo Celular/fisiología , Proliferación Celular/genética , Proliferación Celular/fisiología , Biología Computacional/métodos , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Células Secretoras de Glucagón/citología , Células Secretoras de Glucagón/metabolismo , Humanos , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Microfluídica/métodos , Transducción de Señal/genética , Transducción de Señal/fisiología
19.
Cell Metab ; 22(4): 619-32, 2015 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-26321660

RESUMEN

Aging is driven by changes of the epigenetic state that are only partially understood. We performed a comprehensive epigenomic analysis of the pancreatic ß cell, key player in glucose homeostasis, in adolescent and very old mice. We observe a global methylation drift resulting in an overall more leveled methylome in old ß cells. Importantly, we discover targeted changes in the methylation status of ß cell proliferation and function genes that go against the global methylation drift, are specific to ß cells, and correlate with repression of the proliferation program and activation of metabolic regulators. These targeted alterations are associated with specific chromatin marks and transcription factor occupancy in young ß cells. Strikingly, we find ß cell function improved in aged mice, as predicted by the changes in methylome and transcriptome. Thus, aging of terminally differentiated cells in mammals is not always coupled to functional decline.


Asunto(s)
Envejecimiento , Cromatina/metabolismo , Células Secretoras de Insulina/metabolismo , Animales , Proliferación Celular , Células Cultivadas , Islas de CpG , Metilación de ADN , Elementos de Facilitación Genéticos/genética , Histonas/genética , Histonas/metabolismo , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas , Unión Proteica , Factores de Transcripción/química , Factores de Transcripción/metabolismo
20.
J Clin Invest ; 124(2): 670-4, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24430183

RESUMEN

Children with focal hyperinsulinism of infancy display a dramatic, non-neoplastic clonal expansion of ß cells that have undergone mitotic recombination, resulting in paternal disomy of part of chromosome 11. This disomic region contains imprinted genes, including the gene encoding the cell cycle inhibitor p57Kip2 (CDKN1C), which is silenced as a consequence of the recombination event. We hypothesized that targeting p57Kip2 could stimulate adult human ß cell replication. Indeed, when we suppressed CDKN1C expression in human islets obtained from deceased adult organ donors and transplanted them into hyperglycemic, immunodeficient mice, ß cell replication increased more than 3-fold. The newly replicated cells retained properties of mature ß cells, including the expression of ß cell markers such as insulin, PDX1, and NKX6.1. Importantly, these newly replicated cells demonstrated normal glucose-induced calcium influx, further indicating ß cell functionality. These findings provide a molecular explanation for the massive ß cell replication that occurs in children with focal hyperinsulinism. These data also provided evidence that ß cells from older humans, in which baseline replication is negligible, can be coaxed to re-enter and complete the cell cycle while maintaining mature ß cell properties. Thus, controlled manipulation of this pathway holds promise for the expansion of ß cells in patients with type 2 diabetes.


Asunto(s)
Inhibidor p57 de las Quinasas Dependientes de la Ciclina/metabolismo , Células Secretoras de Insulina/citología , Animales , Calcio/metabolismo , Ciclo Celular , Separación Celular , Trasplante de Células , Células Cultivadas , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Modelos Animales de Enfermedad , Citometría de Flujo , Silenciador del Gen , Glucosa/metabolismo , Células HEK293 , Proteínas de Homeodominio/metabolismo , Humanos , Hiperinsulinismo/metabolismo , Células Secretoras de Insulina/metabolismo , Lentivirus/metabolismo , Ratones , Ratones SCID , Microscopía Fluorescente , ARN Interferente Pequeño/metabolismo , Transactivadores/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA