RESUMEN
Insulin secretion is impaired in individuals with cystic fibrosis (CF), contributing to high rates of CF-related diabetes (CFRD) and substantially increasing disease burden. To develop improved therapies for CFRD, better knowledge of pancreatic pathology in CF is needed. Glucagon like peptide-1 (GLP-1) from islet α cells potentiates insulin secretion by binding GLP-1 receptors (GLP-1Rs) on ß cells. We determined whether expression of GLP-1 and/or its signaling components are reduced in CFRD, thereby contributing to impaired insulin secretion. Immunohistochemistry of pancreas from humans with CFRD versus no-CF/no-diabetes revealed no difference in GLP-1 immunoreactivity per islet area, whereas GLP-1R immunoreactivity per islet area or per insulin-positive islet area was reduced in CFRD. Using spatial transcriptomics, we observed several differentially expressed α- and/or ß-cell genes between CFRD and control pancreas. In CFRD, we found upregulation of α-cell PCSK1 which encodes the enzyme (PC1/3) that generates GLP-1, and downregulation of α-cell PCSK1N which inhibits PC1/3. Gene set enrichment analysis also revealed α and ß cell-specific pathway dysregulation in CFRD. Together, our data suggest intra-islet GLP-1 is not limiting in CFRD, but its action may be restricted due to reduced GLP-1R protein levels. Thus, restoring ß-cell GLP-1R protein expression may improve ß-cell function in CFRD.
Asunto(s)
Fibrosis Quística , Receptor del Péptido 1 Similar al Glucagón , Humanos , Fibrosis Quística/metabolismo , Fibrosis Quística/genética , Fibrosis Quística/complicaciones , Fibrosis Quística/patología , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Receptor del Péptido 1 Similar al Glucagón/genética , Masculino , Femenino , Adulto , Diabetes Mellitus/metabolismo , Diabetes Mellitus/genética , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Islotes Pancreáticos/metabolismo , Células Secretoras de Glucagón/metabolismo , Células Secretoras de Glucagón/patología , Péptido 1 Similar al Glucagón/metabolismo , Adulto Joven , Regulación de la Expresión Génica , Adolescente , Insulina/metabolismoRESUMEN
Following the near-total depletion of pancreatic beta-cells with streptozotocin (STZ), a partial recovery of beta-cell mass (BCM) can occur, in part due to the alpha- to beta-cell transdifferentiation with an intermediary insulin/glucagon bi-hormonal cell phenotype. However, human type 2 diabetes typically involves only a partial reduction in BCM and it is not known if recovery after therapeutic intervention involves islet cell transdifferentiation, or how this varies with age. Here, we used transgenic mouse models to examine if islet cell transdifferentiation contributes to BCM recovery following only a partial depletion of BCM. Cell lineage tracing was employed using Glucagon-Cre/yellow fluorescent protein (YFP) transgenic mice treated with STZ (25 mg/kg-neonates; 70 mg/kg-adults) or vehicle alone on 3 consecutive days. Mice were euthanized 2-30 days later with a prior glucose tolerance test on day 30, and immunofluorescence histology performed on the pancreata. Beta-cell abundance was reduced by 30-40% two days post STZ in both neonates and adults, and subsequently partially recovered in adult but not neonatal mice. Glucose tolerance recovered in adult females, but not in males or neonates. Bi-hormonal cell abundance increased 2-3-fold in STZ-treated mice vs. controls in both neonates and adults, as did transdifferentiated cells expressing insulin and the YFP lineage tag, but not glucagon. Transdifferentiated cell presence was an order of magnitude lower than that of bi-hormonal cells. We conclude that alpha- to beta-cell transdifferentiation occurs in mice following only a moderate depletion in BCM, and that this was accompanied by a partial recovery of BCM in adults.
Asunto(s)
Transdiferenciación Celular , Diabetes Mellitus Experimental , Células Secretoras de Glucagón , Células Secretoras de Insulina , Ratones Transgénicos , Estreptozocina , Animales , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/citología , Ratones , Células Secretoras de Glucagón/metabolismo , Células Secretoras de Glucagón/citología , Masculino , Femenino , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Glucagón/metabolismo , Animales Recién Nacidos , Insulina/metabolismo , Páncreas/metabolismo , Páncreas/citología , Prueba de Tolerancia a la Glucosa , Proteínas Luminiscentes/metabolismo , Proteínas Luminiscentes/genéticaRESUMEN
Introduction: Alpha-cell hyperplasia (ACH) is a rare pancreatic endocrine condition. Three types of ACH have been described: functional or nonglucagonoma hyperglucagonemic glucagonoma syndrome, reactive or secondary to defective glucagon signaling, and non-functional. Few cases of ACH with concomitant pancreatic neuroendocrine tumors (pNETs) have been reported and its etiology remains poorly understood. A case report of non-functional ACH with glucagon-producing NET is herein presented. Case report: A 72-year-old male was referred to our institution for a 2 cm single pNET incidentally found during imaging for acute cholecystitis. The patient's past medical history included type 2 diabetes (T2D) diagnosed 12 years earlier, for which he was prescribed metformin, dapagliflozin, and semaglutide. The pNET was clinically and biochemically non-functioning, apart from mildly elevated glucagon 217 pg/ml (<209), and 68Ga-SSTR PET/CT positive uptake was only found at the pancreatic tail (SUVmax 11.45). The patient underwent a caudal pancreatectomy and the post-operative 68Ga-SSTR PET/CT was negative. A multifocal well-differentiated NET G1, pT1N0M0R0 (mf) strongly staining for glucagon on a background neuroendocrine alpha-cell hyperplasia with some degree of acinar fibrosis was identified on pathology analysis. Discussion and conclusion: This case reports the incidental finding of a clinically non-functioning pNET in a patient with T2D and elevated glucagon levels, unexpectedly diagnosed as glucagon-producing NET and ACH. A high level of suspicion was required to conduct the glucagon immunostaining, which is not part of the pathology routine for a clinically non-functioning pNET, and was key for the diagnosis that otherwise would have been missed. This case highlights the need to consider the diagnosis of glucagon-producing pNET on an ACH background even in the absence of glucagonoma syndrome.
Asunto(s)
Células Secretoras de Glucagón , Glucagón , Hiperplasia , Tumores Neuroendocrinos , Neoplasias Pancreáticas , Humanos , Masculino , Anciano , Hiperplasia/metabolismo , Hiperplasia/patología , Células Secretoras de Glucagón/metabolismo , Células Secretoras de Glucagón/patología , Glucagón/metabolismo , Tumores Neuroendocrinos/metabolismo , Tumores Neuroendocrinos/patología , Tumores Neuroendocrinos/diagnóstico , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/diagnóstico , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/complicacionesRESUMEN
Diabetes mellitus affects 537 million adults around the world. Adropin is expressed in different cell types. Our aim was to investigate the cellular localization in the endocrine pancreas and its effect on modulating pancreatic endocrine hormone release in streptozotocin (STZ)-induced diabetic rats. Adropin expression in the pancreas was investigated in normal and diabetic rats using immunohistochemistry and immunoelectron microscopy. Serum levels of insulin, glucagon pancreatic polypeptide (PP), and somatostatin were measured using a Luminex® χMAP (Magpix®) analyzer. Pancreatic endocrine hormone levels in INS-1 832/3 rat insulinoma cells, as well as pancreatic tissue fragments of normal and diabetic rats treated with different concentrations of adropin (10-6, 10-9, and 10-12 M), were measured using ELISA. Adropin was colocalized with cells producing either insulin, glucagon, or PP. Adropin treatment reduced the number of glucagon-secreting alpha cells and suppressed glucagon release from the pancreas. The serum levels of GLP-1 and amylin were significantly increased after treatment with adropin. Our study indicates a potential role of adropin in modulating glucagon secretion in animal models of diabetes mellitus.
Asunto(s)
Diabetes Mellitus Experimental , Glucagón , Insulina , Islotes Pancreáticos , Animales , Glucagón/metabolismo , Glucagón/sangre , Diabetes Mellitus Experimental/metabolismo , Ratas , Masculino , Islotes Pancreáticos/metabolismo , Insulina/metabolismo , Insulina/sangre , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Péptido 1 Similar al Glucagón/metabolismo , Péptido 1 Similar al Glucagón/sangre , Células Secretoras de Glucagón/metabolismo , Somatostatina/metabolismo , Polipéptido Pancreático/metabolismo , Polipéptido Pancreático/sangre , Ratas Sprague-Dawley , Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo , Polipéptido Amiloide de los Islotes Pancreáticos/genética , Proteínas Sanguíneas , PéptidosRESUMEN
Pancreatic δ cells act locally to repress both insulin and glucagon secretion. Because they are a rare cell type, experimentation examining δ-cell function and control has lagged that of the more abundant α and ß cells. Emerging evidence, enabled partly by developing single-cell technology, demonstrates that δ-cell function is, in part, directed by δ cells but that δ cells also have intrinsic control. The contribution of these cells to overall glucose homeostasis and diabetes onset and progression is still unclear. However, they regulate both α and ß cells, both of which are dysfunctional in diabetes, and their numbers are disrupted in humans with diabetes and in multiple animal models of diabetes, suggesting δ cells are a pivotal character in both health and disease.
Asunto(s)
Células Secretoras de Insulina , Humanos , Animales , Células Secretoras de Insulina/fisiología , Diabetes Mellitus , Células Secretoras de Somatostatina/metabolismo , Insulina/metabolismo , Células Secretoras de Glucagón/metabolismo , Glucagón/metabolismoRESUMEN
Processing of proglucagon into glucagon-like peptide-1 (GLP-1) and GLP-2 in intestinal L cells is mediated by the prohormone convertase 1/3 (PC1/3) while PC2 is responsible for the synthesis of glucagon in pancreatic alpha cells. While GLP-1 is also produced by alpha cells, the identity of the convertase involved in its synthesis is still unsettled. It also remains to be determined whether all alpha cells produce the incretin. The aims of this study were first, to elucidate the identity of the proconvertase responsible for GLP-1 production in human alpha cells, and second, to ascertain whether the number of glucagon cells expressing GLP-1 increase during diabetes. To answer these questions, sections of pancreas from donors' non-diabetic controls, type 1 and type 2 diabetes were processed for double-labelled immunostaining of glucagon and GLP-1 and of each hormone and either PC1 or PC2. Stained sections were examined by confocal microscopy. It was found that all alpha cells of islets from those three groups expressed GLP-1 and PC2 but not PC1/3. This observation supports the view that PC2 is the convertase involved in GLP-1 synthesis in all human glucagon cells and suggests that the regulation of its activity may have important clinical application in diabetes.
Asunto(s)
Péptido 1 Similar al Glucagón , Células Secretoras de Glucagón , Proproteína Convertasa 1 , Proproteína Convertasa 2 , Humanos , Péptido 1 Similar al Glucagón/metabolismo , Células Secretoras de Glucagón/metabolismo , Proproteína Convertasa 1/metabolismo , Proproteína Convertasa 1/genética , Proproteína Convertasa 2/metabolismo , Proproteína Convertasa 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patología , Glucagón/metabolismo , Adulto , Masculino , Persona de Mediana Edad , FemeninoRESUMEN
SCOPE: Garlic extract (GE) has been shown to ameliorate hyperglycemia in diabetic rats (DRs) by increasing insulin production. However, the mechanism through which it exerts its effects remains unclear. Here, it investigates the molecular process and the origin of regenerating ß-cell in rats with streptozotocin (STZ)-induced diabetes in response to GE. METHODS AND RESULTS: In this study, quantitative RT-PCR (qRT-PCR), western blotting, and immunohistochemical analysis are carried out after pancreas isolation. These findings show that 1 week of GE treatment increases the expression of the endocrine progenitor cell markers Neurogenin3 (Neurog3), pancreatic and duodenal homeobox 1 (Pdx1), neurogenic differentiation factor 1 (Neurod1), paired box proteins (Pax)4, V-maf musculoaponeurotic fibrosarcoma oncogene homolog B (Mafb), and NK homeobox factors (Nkx)6-1 in STZ-induced DRs. Continuation with GE treatment for 8 weeks causes the expression of the mature ß-cell markers insulin(Ins)2, urocortin3 (Ucn3), and glucose transporter 2 (Glut2) to peak. Comprehensive examination of the islet through immunohistochemical analysis reveals the presence of a heterogeneous cell population including INS+/GLUT2- and INS+/GLUT2+ ß-cell subpopulations with few bihormonal INS+/GCG+ cells after 4 weeks. By week 8, islet architecture is reestablished, and glucose-stimulated insulin secretion was restored through the upregulation of Ucn3. CONCLUSION: GE induces ß-cell neogenesis in DRs and restores islet architecture. The newly formed mature ß-like cells could have originated through the differentiation of endocrine progenitor cells as well as α- to ß-cell transdifferentiation.
Asunto(s)
Transdiferenciación Celular , Diabetes Mellitus Experimental , Ajo , Células Secretoras de Insulina , Extractos Vegetales , Animales , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Diabetes Mellitus Experimental/tratamiento farmacológico , Extractos Vegetales/farmacología , Transdiferenciación Celular/efectos de los fármacos , Masculino , Ajo/química , Insulina/metabolismo , Células Secretoras de Glucagón/efectos de los fármacos , Células Secretoras de Glucagón/metabolismo , Ratas Wistar , Homeostasis/efectos de los fármacos , Glucemia/metabolismo , Glucemia/efectos de los fármacos , Ratas , Glucosa/metabolismo , Proteínas de Homeodominio/metabolismo , Proteínas de Homeodominio/genética , Proteína Doblecortina , Proteínas del Tejido Nervioso , Factores de Transcripción con Motivo Hélice-Asa-Hélice BásicoRESUMEN
G6PC2 encodes a glucose-6-phosphatase catalytic subunit that opposes the action of glucokinase in pancreatic islets, thereby modulating the sensitivity of insulin and glucagon secretion to glucose. In mice, G6pc2 is expressed at ~20-fold higher levels in ß-cells than in α-cells, whereas in humans G6PC2 is expressed at only ~5-fold higher levels in ß-cells. We therefore hypothesize that G6PC2 likely influences glucagon secretion to a greater degree in humans. With a view to generating a humanized mouse that recapitulates augmented G6PC2 expression levels in α-cells, we sought to identify the genomic regions that confer differential mouse G6pc2 expression in α-cells versus ß-cells as well as the evolutionary changes that have altered this ratio in humans. Studies in islet-derived cell lines suggest that the elevated G6pc2 expression in mouse ß-cells versus α-cells is mainly due to a difference in the relative activity of the proximal G6pc2 promoter in these cell types. Similarly, the smaller difference in G6PC2 expression between α-cells and ß-cells in humans is potentially explained by a change in relative proximal G6PC2 promoter activity. However, we show that both glucocorticoid levels and multiple differences in the relative activity of eight transcriptional enhancers between mice and humans likely contribute to differential G6PC2 expression. Finally, we show that a mouse-specific non-coding RNA, Gm13613, whose expression is controlled by G6pc2 enhancer I, does not regulate G6pc2 expression, indicating that altered expression of Gm13613 in a humanized mouse that contains both the human promoter and enhancers should not affect G6PC2 function.
Asunto(s)
Elementos de Facilitación Genéticos , Células Secretoras de Glucagón , Glucosa-6-Fosfatasa , Regiones Promotoras Genéticas , Animales , Humanos , Regiones Promotoras Genéticas/genética , Glucosa-6-Fosfatasa/genética , Glucosa-6-Fosfatasa/metabolismo , Ratones , Células Secretoras de Glucagón/metabolismo , Células Secretoras de Insulina/metabolismo , Regulación de la Expresión Génica , Línea CelularRESUMEN
BACKGROUND & AIMS: The pancreas is composed of endocrine and exocrine parts, and its interlacing structure indicates potential interaction between endocrine and exocrine cells. Although the tumor microenvironment of pancreatic ductal adenocarcinoma (PDAC) has been well characterized, the role of pancreatic endocrine cells during carcinogenesis is relatively understudied. METHODS: The changes of endocrine cells in PDAC by single-cell transcriptome sequencing, spatial transcriptome sequencing, and multiplex immunohistochemistry were depicted. After that, the interaction between pancreatic carcinogenesis and endocrine changes was explored in orthotopic transplantation mice, KrasLSL-G12DPdx1-Cre mice, and KrasLSL-G12Dp53LoxPPdx1-CreER mice. Finally, we proved the mechanism of the interaction between endocrine and exocrine parts of the pancreas through islet isolation, co-culture in vitro and co-injection in vivo. RESULTS: Pancreatic endocrine cells displayed significantly different transcriptomic characteristics and increased interaction with exocrine part in PDAC. Specifically, among all of the changes, pancreatic polypeptide-positive cells showed a sharp increment accompanied by the progression of the cancer lesion, which might be derived from the transdifferentiation of α and ß cells. Interestingly, it was proved that PDAC cells were able to induce the transdifferentiation of pancreatic α cells and ß cells into glucagon-pancreatic polypeptide and insulin-pancreatic polypeptide double-positive cells, which further promoted carcinogenesis and development of PDAC in a paracrine-dependent manner and formed a reciprocal interaction. CONCLUSIONS: This study systematically maps the alteration of pancreatic endocrine cells in PDAC and elucidates the potential endocrine-exocrine interaction mechanisms during PDAC carcinogenesis. In addition, cancer-associated endocrine cells are defined and characterized, thereby further broadening the composition of PDAC microenvironment.
Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Microambiente Tumoral , Animales , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Carcinoma Ductal Pancreático/patología , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Ratones , Humanos , Técnicas de Cocultivo , Análisis de la Célula Individual , Transdiferenciación Celular , Células Secretoras de Insulina/patología , Células Secretoras de Insulina/metabolismo , Transcriptoma , Línea Celular Tumoral , Células Secretoras de Glucagón/patología , Células Secretoras de Glucagón/metabolismo , Carcinogénesis/patología , Carcinogénesis/genética , Regulación Neoplásica de la Expresión Génica , Transformación Celular Neoplásica/patología , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/genética , Perfilación de la Expresión Génica , Modelos Animales de Enfermedad , Ratones TransgénicosRESUMEN
OBJECTIVES: This study aimed to evaluate the efficacy of a purification method developed for isolating alpha, beta, and delta cells from pancreatic islets of adult mice, extending its application to islets from newborn and aged mice. Furthermore, it sought to examine transcriptome dynamics in mouse pancreatic endocrine islet cells throughout postnatal development and to validate age-related alterations within these cell populations. METHODS: We leveraged the high surface expression of CD71 on beta cells and CD24 on delta cells to FACS-purify alpha, beta, and delta cells from newborn (1-week-old), adult (12-week-old), and old (18-month-old) mice. Bulk RNA sequencing was conducted on these purified cell populations, and subsequent bioinformatic analyses included differential gene expression, overrepresentation, and intersection analysis. RESULTS: Alpha, beta, and delta cells from newborn and aged mice were successfully FACS-purified using the same method employed for adult mice. Our analysis of the age-related transcriptional changes in alpha, beta, and delta cell populations revealed a decrease in cell cycling and an increase in neuron-like features processes during the transition from newborn to adult mice. Progressing from adult to old mice, we identified an inflammatory gene signature related to aging (inflammaging) encompassing an increase in ß-2 microglobulin and major histocompatibility complex (MHC) Class I expression. CONCLUSIONS: Our study demonstrates the effectiveness of our cell sorting technique in purifying endocrine subsets from mouse islets at different ages. We provide a valuable resource for better understanding endocrine pancreas aging and identified an inflammaging gene signature with increased ß-2 microglobulin and MHC Class I expression as a common hallmark of old alpha, beta, and delta cells, with potential implications for immune response regulation and age-related diabetes.
Asunto(s)
Senescencia Celular , Células Secretoras de Glucagón , Células Secretoras de Insulina , Transcriptoma , Animales , Ratones , Células Secretoras de Insulina/metabolismo , Senescencia Celular/genética , Células Secretoras de Glucagón/metabolismo , Ratones Endogámicos C57BL , Regulación hacia Arriba , Células Secretoras de Somatostatina/metabolismo , Masculino , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/metabolismo , Envejecimiento/genética , Envejecimiento/metabolismo , Islotes Pancreáticos/metabolismo , Animales Recién Nacidos , Antígenos CD/metabolismo , Antígenos CD/genéticaRESUMEN
Diabetes involves the death or dysfunction of pancreatic ß-cells. Analysis of bulk sequencing from human samples and studies using in vitro and in vivo models suggest that endoplasmic reticulum and inflammatory signaling play an important role in diabetes progression. To better characterize cell type-specific stress response, we perform multiplexed single-cell RNA sequencing to define the transcriptional signature of primary human islet cells exposed to endoplasmic reticulum and inflammatory stress. Through comprehensive pair-wise analysis of stress responses across pancreatic endocrine and exocrine cell types, we define changes in gene expression for each cell type under different diabetes-associated stressors. We find that ß-, α-, and ductal cells have the greatest transcriptional response. We utilize stem cell-derived islets to study islet health through the candidate gene CIB1, which was upregulated under stress in primary human islets. Our findings provide insights into cell type-specific responses to diabetes-associated stress and establish a resource to identify targets for diabetes therapeutics.
Asunto(s)
Estrés del Retículo Endoplásmico , Células Secretoras de Insulina , Islotes Pancreáticos , Humanos , Estrés del Retículo Endoplásmico/genética , Islotes Pancreáticos/metabolismo , Células Secretoras de Insulina/metabolismo , Proteínas de Unión al Calcio/metabolismo , Proteínas de Unión al Calcio/genética , Análisis de la Célula Individual , Células Secretoras de Glucagón/metabolismo , Análisis de Secuencia de ARN , Transcriptoma , Estrés FisiológicoRESUMEN
PURPOSE: Over the past 20 years, much of the research on diabetes has focused on pancreatic beta cells. In the last 10 years, interest in the important role of pancreatic alpha cells in the pathogenesis of diabetes, which had previously received little attention, has grown. We aimed to summarize and visualize the hotspot and development trends of pancreatic alpha cells through bibliometric analysis and to provide research direction and future ideas for the treatment of diabetes and other islet-related diseases. METHODS: We used two scientometric software packages (CiteSpace 6.1.R6 and VOSviewer1.6.18) to visualize the information and connection of countries, institutions, authors, and keywords in this field. RESULTS: A total of 532 publications, published in 752 institutions in 46 countries and regions, were included in this analysis. The United States showed the highest output, accounting for 39.3% of the total number of published papers. The most active institution was Vanderbilt University, and the authors with highest productivity came from Ulster University. In recent years, research hotspots have concentrated on transdifferentiation, gene expression, and GLP-1 regulatory function. Visualization analysis shows that research hotspots mainly focus on clinical diseases as well as physiological and pathological mechanisms and related biochemical indicators. CONCLUSIONS: This study provides a review and summary of the literature on pancreatic alpha cells through bibliometric and visual methods and shows research hotspot and development trends, which can guide future directions for research.
Asunto(s)
Bibliometría , Células Secretoras de Glucagón , Humanos , Células Secretoras de Glucagón/metabolismo , Investigación Biomédica/tendencias , Animales , Diabetes MellitusRESUMEN
Blood amino acid levels are maintained in a narrow physiological range. The pancreatic α cells have emerged as the primary aminoacidemia regulator through glucagon secretion to promote hepatic amino acid catabolism. Interruption of glucagon signaling disrupts the liver-α cells axis leading to hyperaminoacidemia, which triggers a compensatory rise in glucagon secretion and α cell hyperplasia. The mechanisms of hyperaminoacidemia-induced α cell hyperplasia remain incompletely understood. Using a mouse α cell line and in vivo studies in zebrafish and mice, we found that hyperaminoacidemia-induced α cell hyperplasia requires ErbB3 signaling. In addition to mechanistic target of rapamycin complex 1, another ErbB3 downstream effector signal transducer and activator of transcription 3 also plays a role in α cell hyperplasia. Mechanistically, ErbB3 may partner with ErbB2 to stimulate cyclin D2 and suppress p27 via mechanistic target of rapamycin complex 1 and signal transducer and activator of transcription 3. Our study identifies ErbB3 as a new regulator for hyperaminoacidemia-induced α cell proliferation and a critical component of the liver-α cells axis that regulates aminoacidemia.
Asunto(s)
Ciclina D2 , Células Secretoras de Glucagón , Hiperplasia , Diana Mecanicista del Complejo 1 de la Rapamicina , Receptor ErbB-3 , Pez Cebra , Animales , Humanos , Ratones , Aminoácidos/metabolismo , Línea Celular , Proliferación Celular , Ciclina D2/metabolismo , Ciclina D2/genética , Células Secretoras de Glucagón/metabolismo , Células Secretoras de Glucagón/patología , Hiperplasia/metabolismo , Hiperplasia/patología , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Receptor ErbB-2/metabolismo , Receptor ErbB-2/genética , Receptor ErbB-3/metabolismo , Receptor ErbB-3/genética , Transducción de Señal , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT3/genéticaRESUMEN
The T allele at rs7903146 in TCF7L2 increases the rate of conversion from prediabetes to type 2 diabetes. This has been associated with impaired ß-cell function and with defective suppression of α-cell secretion by glucose. However, the temporal relationship of these abnormalities is uncertain. To study the longitudinal changes in islet function, we recruited 128 subjects, with 67 homozygous for the diabetes-associated allele (TT) at rs7903146 and 61 homozygous for the protective allele. Subjects were studied on two occasions, 3 years apart, using an oral 75-g glucose challenge. The oral minimal model was used to quantitate ß-cell function; the glucagon secretion rate was estimated from deconvolution of glucagon concentrations. Glucose tolerance worsened in subjects with the TT genotype. This was accompanied by impaired postchallenge glucagon suppression but appropriate ß-cell responsivity to rising glucose concentrations. These data suggest that α-cell abnormalities associated with the TT genotype (rs7903146) occur early and may precede ß-cell dysfunction in people as they develop glucose intolerance and type 2 diabetes.
Asunto(s)
Diabetes Mellitus Tipo 2 , Glucagón , Prueba de Tolerancia a la Glucosa , Células Secretoras de Insulina , Proteína 2 Similar al Factor de Transcripción 7 , Humanos , Proteína 2 Similar al Factor de Transcripción 7/genética , Proteína 2 Similar al Factor de Transcripción 7/metabolismo , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Glucagón/metabolismo , Masculino , Femenino , Adulto , Persona de Mediana Edad , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/fisiología , Estudios Longitudinales , Células Secretoras de Glucagón/metabolismo , Islotes Pancreáticos/metabolismo , Genotipo , Glucemia/metabolismo , Intolerancia a la Glucosa/genética , Intolerancia a la Glucosa/metabolismo , AlelosRESUMEN
Glucagon is critical for the maintenance of blood glucose, however nutrient regulation of pancreatic α-cells remains poorly understood. Here, we identified a role of leucine, a well-known ß-cell fuel, in the α-cell-intrinsic regulation of glucagon release. In islet perifusion assays, physiologic concentrations of leucine strongly inhibited alanine- and arginine-stimulated glucagon secretion from human and mouse islets under hypoglycemic conditions. Mechanistically, leucine dose-dependently reduced α-cell cAMP, independently of Ca2+, ATP/ADP, or fatty acid oxidation. Leucine also reduced α-cell cAMP in islets treated with somatostatin receptor 2 antagonists or diazoxide, compounds that limit paracrine signaling from ß/δ-cells. Studies in dispersed mouse islets confirmed an α-cell-intrinsic effect. The inhibitory effect of leucine on cAMP was mimicked by glucose, α-ketoisocaproate, succinate, and the glutamate dehydrogenase activator BCH and blocked by cyanide, indicating a mechanism dependent on mitochondrial metabolism. Glucose dose-dependently reduced the impact of leucine on α-cell cAMP, indicating an overlap in function; however, leucine was still effective at suppressing glucagon secretion in the presence of elevated glucose, amino acids, and the incretin GIP. Taken together, these findings show that leucine plays an intrinsic role in limiting the α-cell secretory tone across the physiologic range of glucose levels, complementing the inhibitory paracrine actions of ß/δ-cells.
Asunto(s)
AMP Cíclico , Células Secretoras de Glucagón , Glucagón , Leucina , Comunicación Paracrina , Animales , Glucagón/metabolismo , AMP Cíclico/metabolismo , Células Secretoras de Glucagón/metabolismo , Células Secretoras de Glucagón/efectos de los fármacos , Ratones , Humanos , Leucina/farmacología , Comunicación Paracrina/efectos de los fármacos , Glucosa/metabolismo , Cetoácidos/farmacología , Masculino , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Ratones Endogámicos C57BL , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismoRESUMEN
Glucagon, a hormone released from pancreatic α-cells, is critical for maintaining euglycemia and plays a key role in the pathophysiology of diabetes. To stimulate the development of new classes of therapeutic agents targeting glucagon release, key α-cell signaling pathways that regulate glucagon secretion need to be identified. Here, we focused on the potential importance of α-cell Gs signaling on modulating α-cell function. Studies with α-cell-specific mouse models showed that activation of α-cell Gs signaling causes a marked increase in glucagon secretion. We also found that intra-islet adenosine plays an unexpected autocrine/paracrine role in promoting glucagon release via activation of α-cell Gs-coupled A2A adenosine receptors. Studies with α-cell-specific Gαs knockout mice showed that α-cell Gs also plays an essential role in stimulating the activity of the Gcg gene, thus ensuring proper islet glucagon content. Our data suggest that α-cell enriched Gs-coupled receptors represent potential targets for modulating α-cell function for therapeutic purposes.
Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gs , Células Secretoras de Glucagón , Glucagón , Ratones Noqueados , Transducción de Señal , Glucagón/metabolismo , Animales , Células Secretoras de Glucagón/metabolismo , Ratones , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Adenosina/metabolismo , Receptor de Adenosina A2A/metabolismo , Receptor de Adenosina A2A/genética , Masculino , Ratones Endogámicos C57BL , Islotes Pancreáticos/metabolismoRESUMEN
AIMS/INTRODUCTION: Imeglimin is a recently approved oral antidiabetic agent that improves insulin resistance, and promotes insulin secretion from pancreatic ß-cells. Here, we investigated the effects of imeglimin on glucagon secretion from pancreatic α-cells. MATERIALS AND METHODS: Experiments were carried out in high-fat, high-sucrose diet-fed mice. The effects of imeglimin were examined using insulin and glucose tolerance tests, glucose clamp studies, and measurements of glucagon secretion from isolated islets. Glucagon was measured using both the standard and the sequential protocol of Mercodia sandwich enzyme-linked immunosorbent assay; the latter eliminates cross-reactivities with other proglucagon-derived peptides. RESULTS: Plasma glucagon, insulin and glucagon-like peptide-1 levels were increased by imeglimin administration in high-fat, high-sucrose diet-fed mice. Glucose clamp experiments showed that the glucagon increase was not caused by reduced blood glucose levels. After both single and long-term administration of imeglimin, glucagon secretions were significantly enhanced during glucose tolerance tests. Milder enhancement was observed when using the sequential protocol. Long-term administration of imeglimin did not alter α-cell mass. Intraperitoneal imeglimin administration did not affect glucagon secretion, despite significantly decreased blood glucose levels. Imeglimin did not enhance glucagon secretion from isolated islets. Imeglimin administration improved fatty liver by suppressing de novo lipogenesis through decreasing sterol regulatory element binding protein-1c and carbohydrate response element binding protein and their target genes, while enhancing fatty acid oxidation through increasing carnitine palmitoyltransferase I. CONCLUSIONS: Overall, the present results showed that imeglimin enhances glucagon secretion through an indirect mechanism. Our findings also showed that glucagon secretion promoted by imeglimin could contribute to improvement of fatty liver through suppressing de novo lipogenesis and enhancing fatty acid oxidation.
Asunto(s)
Dieta Alta en Grasa , Hígado Graso , Glucagón , Animales , Dieta Alta en Grasa/efectos adversos , Glucagón/metabolismo , Ratones , Masculino , Hígado Graso/metabolismo , Hígado Graso/tratamiento farmacológico , Ratones Endogámicos C57BL , Insulina/metabolismo , Glucemia/análisis , Células Secretoras de Glucagón/metabolismo , Células Secretoras de Glucagón/efectos de los fármacos , Prueba de Tolerancia a la Glucosa , Péptido 1 Similar al Glucagón/metabolismo , Sacarosa en la Dieta , Hipoglucemiantes/farmacología , Resistencia a la Insulina , TriazinasRESUMEN
Human islets of Langerhans are composed mostly of glucagon-secreting α cells and insulin-secreting ß cells closely intermingled one another. Current methods for identifying α and ß cells involve either fixing islets and using immunostaining or disaggregating islets and employing flow cytometry for classifying α and ß cells based on their size and autofluorescence. Neither approach, however, allows investigating the dynamic behavior of α and ß cells in a living and intact islet. To tackle this issue, we present a machine-learning-based strategy for identification α and ß cells in label-free infrared micrographs of living human islets without immunostaining. Intrinsic autofluorescence is stimulated by infrared light and collected both in intensity and lifetime in the visible range, dominated by NAD(P)H and lipofuscin signals. Descriptive parameters are derived from micrographs for ~ 103 cells. These parameters are used as input for a boosted decision-tree model (XGBoost) pre-trained with immunofluorescence-derived cell-type information. The model displays an optimized-metrics performance of 0.86 (i.e. area under a ROC curve), with an associated precision of 0.94 for the recognition of ß cells and 0.75 for α cells. This tool promises to enable longitudinal studies on the dynamic behavior of individual cell types at single-cell resolution within the intact tissue.
Asunto(s)
Células Secretoras de Insulina , Aprendizaje Automático , Humanos , Células Secretoras de Insulina/metabolismo , Células Secretoras de Glucagón/metabolismo , Islotes Pancreáticos/metabolismo , Rayos InfrarrojosRESUMEN
Dysregulation of α cells results in hyperglycemia and hyperglucagonemia in type 2 diabetes mellitus (T2DM). Mesenchymal stromal cell (MSC)-based therapy increases oxygen consumption of islets and enhances insulin secretion. However, the underlying mechanism for the protective role of MSCs in α-cell mitochondrial dysfunction remains unclear. Here, human umbilical cord MSCs (hucMSCs) were used to treat 2 kinds of T2DM mice and αTC1-6 cells to explore the role of hucMSCs in improving α-cell mitochondrial dysfunction and hyperglucagonemia. Plasma and supernatant glucagon were detected by enzyme-linked immunosorbent assay (ELISA). Mitochondrial function of α cells was assessed by the Seahorse Analyzer. To investigate the underlying mechanisms, Sirtuin 1 (SIRT1), Forkhead box O3a (FoxO3a), glucose transporter type1 (GLUT1), and glucokinase (GCK) were assessed by Western blotting analysis. In vivo, hucMSC infusion improved glucose and insulin tolerance, as well as hyperglycemia and hyperglucagonemia in T2DM mice. Meanwhile, hucMSC intervention rescued the islet structure and decreased α- to ß-cell ratio. Glucagon secretion from αTC1-6 cells was consistently inhibited by hucMSCs in vitro. Meanwhile, hucMSC treatment activated intracellular SIRT1/FoxO3a signaling, promoted glucose uptake and activation, alleviated mitochondrial dysfunction, and enhanced ATP production. However, transfection of SIRT1 small interfering RNA (siRNA) or the application of SIRT1 inhibitor EX-527 weakened the therapeutic effects of hucMSCs on mitochondrial function and glucagon secretion. Our observations indicate that hucMSCs mitigate mitochondrial dysfunction and glucagon hypersecretion of α cells in T2DM via SIRT1/FoxO3a signaling, which provides novel evidence demonstrating the potential for hucMSCs in treating T2DM.
Asunto(s)
Diabetes Mellitus Tipo 2 , Proteína Forkhead Box O3 , Glucagón , Células Madre Mesenquimatosas , Mitocondrias , Transducción de Señal , Sirtuina 1 , Sirtuina 1/metabolismo , Animales , Células Madre Mesenquimatosas/metabolismo , Proteína Forkhead Box O3/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/terapia , Mitocondrias/metabolismo , Ratones , Humanos , Glucagón/metabolismo , Trasplante de Células Madre Mesenquimatosas/métodos , Masculino , Células Secretoras de Glucagón/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/terapia , Ratones Endogámicos C57BLRESUMEN
BACKGROUND: This study explored the correlation between pancreatic islet α cell function, as reflected by the plasma glucagon levels, and Diabetic Peripheral Neuropathy (DPN) in patients with Type 2 Diabetes Mellitus (T2DM). METHODS: A total of 358 patients with T2DM were retrospectively enrolled in this study and divided into the Non-DPN (NDPN) group (n = 220) and the DPN group (n = 138). All patients underwent an oral glucose tolerance test to detect levels of blood glucose, insulin and glucagon, and the Area Under the Curve (AUC) for Glucagon (AUCglu) was used to estimate the overall glucagon level. The Peripheral Nerve Conduction Velocity (PNCV), Amplitude (PNCA) and Latency (PNCL) were obtained with electromyography, and their Z scores were calculated. RESULTS: There were significant differences regarding the age, disease duration, serum levels of alanine aminotransferase, aspartate aminotransferase, urea nitrogen, high-density lipoprotein, and 2h-C peptide between these two groups (p < 0.05). The NDPN group had higher glucagon levels at 30, 60 and 120 min and AUCglu (p < 0.05). The Z-scores of PNCV and PNCA showed an increasing trend (p < 0.05), while the Z-score of PNCL showed a decreasing trend (p < 0.05). The glucagon levels were positively correlated with PNCV and PNCA, but negatively correlated with PNCL, with Gluca30min having the strongest correlation (p < 0.05). Gluca30min was independently related to PNCV, PNCL, PNCA and DPN, respectively (p < 0.05). The function of pancreatic α islet cells, as reflected by the plasma glucagon level, is closely related to the occurrence of DPN in T2DM patients. CONCLUSION: Gluca30min may be a potentially valuable independent predictor for the occurrence of DPN.