Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Mol Metab ; 87: 101990, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39009220

RESUMO

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.


Assuntos
Senescência Celular , Células Secretoras de Glucagon , Células Secretoras de Insulina , Transcriptoma , Animais , Camundongos , Células Secretoras de Insulina/metabolismo , Senescência Celular/genética , Células Secretoras de Glucagon/metabolismo , Camundongos Endogâmicos C57BL , Regulação para Cima , Células Secretoras de Somatostatina/metabolismo , Masculino , Antígenos de Histocompatibilidade Classe I/genética , Antígenos de Histocompatibilidade Classe I/metabolismo , Envelhecimento/genética , Envelhecimento/metabolismo , Ilhotas Pancreáticas/metabolismo , Animais Recém-Nascidos , Antígenos CD/metabolismo , Antígenos CD/genética
2.
J Endocrinol ; 261(3)2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38593829

RESUMO

Pancreatic alpha cell activity and glucagon secretion lower as glucose levels increase. While part of the decrease is regulated by glucose itself, paracrine signaling by their neighboring beta and delta cells also plays an important role. Somatostatin from delta cells is an important local inhibitor of alpha cells at high glucose. Additionally, urocortin 3 (UCN3) is a hormone that is co-released from beta cells with insulin and acts locally to potentiate somatostatin secretion from delta cells. UCN3 thus inhibits insulin secretion via a negative feedback loop with delta cells, but its role with respect to alpha cells and glucagon secretion is not understood. We hypothesize that the somatostatin-driven glucagon inhibition at high glucose is regulated in part by UCN3 from beta cells. Here, we use a combination of live functional Ca2+ and cAMP imaging as well as direct glucagon secretion measurement, all from alpha cells in intact mouse islets, to determine the contributions of UCN3 to alpha cell behavior. Exogenous UCN3 treatment decreased alpha cell Ca2+ and cAMP levels and inhibited glucagon release. Blocking endogenous UCN3 signaling increased alpha cell Ca2+ by 26.8 ± 7.6%, but this did not result in increased glucagon release at high glucose. Furthermore, constitutive deletion of Ucn3 did not increase Ca2+ activity or glucagon secretion relative to controls. UCN3 is thus capable of inhibiting mouse alpha cells, but, given the subtle effects of endogenous UCN3 signaling on alpha cells, we propose that UCN3-driven somatostatin may serve to regulate local paracrine glucagon levels in the islet instead of inhibiting gross systemic glucagon release.


Assuntos
Células Secretoras de Glucagon , Glucagon , Comunicação Parácrina , Urocortinas , Animais , Urocortinas/metabolismo , Urocortinas/genética , Células Secretoras de Glucagon/metabolismo , Células Secretoras de Glucagon/efeitos dos fármacos , Camundongos , Glucagon/metabolismo , Glucose/metabolismo , Cálcio/metabolismo , Masculino , Camundongos Endogâmicos C57BL , AMP Cíclico/metabolismo , Somatostatina/farmacologia , Somatostatina/metabolismo
3.
Peptides ; 175: 171179, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38360354

RESUMO

Glucagon-like peptide-1 receptor (GLP1R) and glucose-dependent insulinotropic polypeptide receptor (GIPR) are transmembrane receptors involved in insulin, glucagon and somatostatin secretion from the pancreatic islet. Therapeutic targeting of GLP1R and GIPR restores blood glucose levels in part by influencing beta cell, alpha cell and delta cell function. Despite the importance of the incretin-mimetics for diabetes therapy, our understanding of GLP1R and GIPR expression patterns and signaling within the islet remain incomplete. Here, we present the evidence for GLP1R and GIPR expression in the major islet cell types, before addressing signaling pathway(s) engaged, as well as their influence on cell survival and function. While GLP1R is largely a beta cell-specific marker within the islet, GIPR is expressed in alpha cells, beta cells, and (possibly) delta cells. GLP1R and GIPR engage Gs-coupled pathways in most settings, although the exact outcome on hormone release depends on paracrine communication and promiscuous signaling. Biased agonism away from beta-arrestin is an emerging concept for improving therapeutic efficacy, and is also relevant for GLP1R/GIPR dual agonism. Lastly, dual agonists exert multiple effects on islet function through GIPR > GLP1R imbalance, increased GLP1R surface expression and cAMP signaling, as well as beneficial alpha cell-beta cell-delta cell crosstalk.


Assuntos
Células Secretoras de Glucagon , Receptores dos Hormônios Gastrointestinais , Células Secretoras de Somatostatina/metabolismo , Células Secretoras de Glucagon/metabolismo , Receptor do Peptídeo Semelhante ao Glucagon 1/genética , Receptores dos Hormônios Gastrointestinais/metabolismo , Polipeptídeo Inibidor Gástrico/genética , Polipeptídeo Inibidor Gástrico/metabolismo , Transdução de Sinais
4.
Front Endocrinol (Lausanne) ; 14: 1250023, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37772078

RESUMO

Manifest diabetes, but also conditions of increased insulin resistance such as pregnancy or obesity can lead to islet architecture remodeling. The contributing mechanisms are as poorly understood as the consequences of altered cell arrangement. For the quantification of the different cell types but also the frequency of different cell-cell contacts within the islets, different approaches exist. However, few methods are available to characterize islet cell distribution in a statistically valid manner. Here we describe PyCreas, an open-source tool written in Python that allows semi-automated analysis of islet cell distribution based on images of pancreatic sections stained by immunohistochemistry or immunofluorescence. To ensure that the PyCreas tool is suitable for quantitative analysis of cell distribution in the islets at different metabolic states, we studied the localization and distribution of alpha, beta, and delta cells during gestation and prediabetes. We compared the islet cell distribution of pancreatic islets from metabolically healthy NMRI mice with that of New Zealand obese (NZO) mice, which exhibit impaired glucose tolerance (IGT) both preconceptionally and during gestation, and from C57BL/6 N (B6) mice, which acquire this IGT only during gestation. Since substrain(s) of the NZO mice are known to show a variant in the Abcc8 gene, we additionally examined preconceptional SUR1 knock-out (SUR1-KO) mice. PyCreas provided quantitative evidence that alterations in the Abcc8 gene are associated with an altered distribution pattern of islet cells. Moreover, our data indicate that this cannot be a consequence of prolonged hyperglycemia, as islet architecture is already altered in the prediabetic state. Furthermore, the quantitative analysis suggests that states of transient IGT, such as during common gestational diabetes mellitus (GDM), are not associated with changes in islet architecture as observed during long-term IGT. PyCreas provides the ability to systematically analyze the localization and distribution of islet cells at different stages of metabolic disease to better understand the underlying pathophysiology.

5.
Diabetologia ; 66(5): 884-896, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36884057

RESUMO

AIMS/HYPOTHESIS: Transcriptome analyses revealed insulin-gene-derived transcripts in non-beta endocrine islet cells. We studied alternative splicing of human INS mRNA in pancreatic islets. METHODS: Alternative splicing of insulin pre-mRNA was determined by PCR analysis performed on human islet RNA and single-cell RNA-seq analysis. Antisera were generated to detect insulin variants in human pancreatic tissue using immunohistochemistry, electron microscopy and single-cell western blot to confirm the expression of insulin variants. Cytotoxic T lymphocyte (CTL) activation was determined by MIP-1ß release. RESULTS: We identified an alternatively spliced INS product. This variant encodes the complete insulin signal peptide and B chain and an alternative C-terminus that largely overlaps with a previously identified defective ribosomal product of INS. Immunohistochemical analysis revealed that the translation product of this INS-derived splice transcript was detectable in somatostatin-producing delta cells but not in beta cells; this was confirmed by light and electron microscopy. Expression of this alternatively spliced INS product activated preproinsulin-specific CTLs in vitro. The exclusive presence of this alternatively spliced INS product in delta cells may be explained by its clearance from beta cells by insulin-degrading enzyme capturing its insulin B chain fragment and a lack of insulin-degrading enzyme expression in delta cells. CONCLUSIONS/INTERPRETATION: Our data demonstrate that delta cells can express an INS product derived from alternative splicing, containing both the diabetogenic insulin signal peptide and B chain, in their secretory granules. We propose that this alternative INS product may play a role in islet autoimmunity and pathology, as well as endocrine or paracrine function or islet development and endocrine destiny, and transdifferentiation between endocrine cells. INS promoter activity is not confined to beta cells and should be used with care when assigning beta cell identity and selectivity. DATA AVAILABILITY: The full EM dataset is available via www.nanotomy.org (for review: http://www.nanotomy.org/OA/Tienhoven2021SUB/6126-368/ ). Single-cell RNA-seq data was made available by Segerstolpe et al [13] and can be found at https://sandberglab.se/pancreas . The RNA and protein sequence of INS-splice was uploaded to GenBank (BankIt2546444 INS-splice OM489474).


Assuntos
Insulisina , Ilhotas Pancreáticas , Humanos , Células Secretoras de Somatostatina/metabolismo , Insulisina/metabolismo , Insulina/genética , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , RNA , Sinais Direcionadores de Proteínas
6.
Mol Metab ; 60: 101467, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35240340

RESUMO

OBJECTIVES: Until recently, communication between neighboring cells in islets of Langerhans was overlooked by genomic technologies, which require rigorous tissue dissociation into single cells. METHODS: We utilize sorting of physically interacting cells (PICs) with single-cell RNA-sequencing to systematically map cellular interactions in the endocrine pancreas after pancreatectomy. RESULTS: The pancreas cellular landscape features pancreatectomy associated heterogeneity of beta-cells, including an interaction-specific program between paired beta and delta-cells. CONCLUSIONS: Our analysis suggests that the particular cluster of beta-cells that pairs with delta-cells benefits from stress protection, implying that the interaction between beta- and delta-cells might safeguard against pancreatectomy associated challenges. The work encourages testing the potential relevance of physically-interacting beta-delta-cells also in diabetes mellitus.


Assuntos
Células Secretoras de Insulina , Ilhotas Pancreáticas , Pâncreas , Pancreatectomia , Regeneração
7.
Mol Metab ; 42: 101060, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32763423

RESUMO

OBJECTIVES: The main endocrine cell types in pancreatic islets are alpha, beta, and delta cells. Although these cell types have distinct roles in the regulation of glucose homeostasis, inadequate purification methods preclude the study of cell type-specific effects. We developed a reliable approach that enables simultaneous sorting of live alpha, beta, and delta cells from mouse islets for downstream analyses. METHODS: We developed an antibody panel against cell surface antigens to enable isolation of highly purified endocrine subsets from mouse islets based on the specific differential expression of CD71 on beta cells and CD24 on delta cells. We rigorously demonstrated the reliability and validity of our approach using bulk and single cell qPCR, immunocytochemistry, reporter mice, and transcriptomics. RESULTS: Pancreatic alpha, beta, and delta cells can be separated based on beta cell-specific CD71 surface expression and high expression of CD24 on delta cells. We applied our new sorting strategy to demonstrate that CD71, which is the transferrin receptor mediating the uptake of transferrin-bound iron, is upregulated in beta cells during early postnatal weeks. We found that beta cells express higher levels of several other genes implicated in iron metabolism and iron deprivation significantly impaired beta cell function. In human beta cells, CD71 is similarly required for iron uptake and CD71 surface expression is regulated in a glucose-dependent manner. CONCLUSIONS: This study provides a novel and efficient purification method for murine alpha, beta, and delta cells, identifies for the first time CD71 as a postnatal beta cell-specific marker, and demonstrates a central role of iron metabolism in beta cell function.


Assuntos
Antígenos de Superfície/imunologia , Células Secretoras de Insulina/metabolismo , Ferro/metabolismo , Animais , Antígenos CD/imunologia , Antígenos de Superfície/isolamento & purificação , Antígenos de Superfície/metabolismo , Biomarcadores/metabolismo , Antígeno CD24/imunologia , Linhagem Celular , Feminino , Células Secretoras de Glucagon/imunologia , Células Secretoras de Glucagon/metabolismo , Células Secretoras de Glucagon/fisiologia , Humanos , Imuno-Histoquímica/métodos , Células Secretoras de Insulina/imunologia , Células Secretoras de Insulina/fisiologia , Ferro/fisiologia , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pâncreas/metabolismo , Pâncreas/fisiologia , Receptores da Transferrina/imunologia , Reprodutibilidade dos Testes , Células Secretoras de Somatostatina/imunologia , Células Secretoras de Somatostatina/metabolismo , Células Secretoras de Somatostatina/fisiologia
8.
Cell Rep ; 32(2): 107904, 2020 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-32668245

RESUMO

Numerous mammalian cells contain abundant Zn2+ in their secretory granules, yet available Zn2+ sensors lack the desired specificity and sensitivity for imaging granular Zn2+. We developed a fluorescent zinc granule indicator, ZIGIR, that possesses numerous desired properties for live cell imaging, including >100-fold fluorescence enhancement, membrane permeability, and selective enrichment to acidic granules. The combined advantages endow ZIGIR with superior sensitivity and specificity for imaging granular Zn2+. ZIGIR enables separation of heterogenous ß cells based on their insulin content and sorting of mouse islets into pure α cells and ß cells. In human islets, ZIGIR facilitates sorting of endocrine cells into highly enriched α cells and ß cells, reveals unexpectedly high Zn2+ activity in the somatostatin granule of some δ cells, and uncovers variation in the glucagon content among human α cells. We expect broad applications of ZIGIR for studying Zn2+ biology and Zn2+-rich secretory granules and for engineering ß cells with high insulin content for treating diabetes.


Assuntos
Grânulos Citoplasmáticos/metabolismo , Corantes Fluorescentes/metabolismo , Células Secretoras de Glucagon/metabolismo , Ilhotas Pancreáticas/metabolismo , Zinco/metabolismo , Adulto , Idoso , Animais , Células Cultivadas , Feminino , Fluorescência , Corantes Fluorescentes/química , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Masculino , Camundongos , Pessoa de Meia-Idade , Coloração e Rotulagem
9.
Mol Metab ; 40: 101021, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32446876

RESUMO

OBJECTIVES: Elevated plasma glucagon is an early symptom of diabetes, occurring in subjects with impaired glucose regulation. Here, we explored alpha-cell function in female mice fed a high-fat diet (HFD). METHODS: Female mice expressing the Ca2+ indicator GCaMP3 specifically in alpha-cells were fed a high-fat or control (CTL) diet. We then conducted in vivo phenotyping of these mice, as well as experiments on isolated (ex vivo) islets and in the in situ perfused pancreas. RESULTS: In HFD-fed mice, fed plasma glucagon levels were increased and glucagon secretion from isolated islets and in the perfused mouse pancreas was also elevated. In mice fed a CTL diet, increasing glucose reduced intracellular Ca2+ ([Ca2+]i) oscillation frequency and amplitude. This effect was also observed in HFD mice; however, both the frequency and amplitude of the [Ca2+]i oscillations were higher than those in CTL alpha-cells. Given that alpha-cells are under strong paracrine control from neighbouring somatostatin-secreting delta-cells, we hypothesised that this elevation of alpha-cell output was due to a lack of somatostatin (SST) secretion. Indeed, SST secretion in isolated islets from HFD-fed mice was reduced but exogenous SST also failed to suppress glucagon secretion and [Ca2+]i activity from HFD alpha-cells, in contrast to observations in CTL mice. CONCLUSIONS: These findings suggest that reduced delta-cell function, combined with intrinsic changes in alpha-cells including sensitivity to somatostatin, accounts for the hyperglucagonaemia in mice fed a HFD.


Assuntos
Células Secretoras de Glucagon/metabolismo , Glucagon/metabolismo , Somatostatina/metabolismo , Animais , Glicemia/metabolismo , Dieta Hiperlipídica/efeitos adversos , Feminino , Glucagon/genética , Glucose/metabolismo , Insulina/metabolismo , Secreção de Insulina/fisiologia , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Somatostatina/genética , Células Secretoras de Somatostatina/metabolismo
10.
Semin Cell Dev Biol ; 103: 14-19, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32081627

RESUMO

The islet of Langerhans contains at least five types of endocrine cells producing distinct hormones. In response to nutrient or neuronal stimulation, islet endocrine cells release biochemicals including peptide hormones to regulate metabolism and to control glucose homeostasis. It is now recognized that malfunction of islet cells, notably insufficient insulin release of ß-cells and hypersecretion of glucagon from α-cells, represents a causal event leading to hyperglycemia and frank diabetes, a disease that is increasing at an alarming rate to reach an epidemic level worldwide. Understanding the mechanisms regulating stimulus-secretion coupling and investigating how islet ß-cells maintain a robust secretory activity are important topics in islet biology and diabetes research. To facilitate such studies, a number of biological systems and assay platforms have been developed for the functional analysis of islet cells. These technologies have enabled detailed analyses of individual islets at the cellular level, either in vitro, in situ, or in vivo.


Assuntos
Diabetes Mellitus/metabolismo , Técnicas In Vitro/métodos , Dosimetria in Vivo/métodos , Ilhotas Pancreáticas/metabolismo , Humanos
11.
J Physiol ; 596(2): 197-215, 2018 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-28975620

RESUMO

KEY POINTS: We used a mouse expressing a light-sensitive ion channel in ß-cells to understand how α-cell activity is regulated by ß-cells. Light activation of ß-cells triggered a suppression of α-cell activity via gap junction-dependent activation of δ-cells. Mathematical modelling of human islets suggests that 23% of the inhibitory effect of glucose on glucagon secretion is mediated by ß-cells via gap junction-dependent activation of δ-cells/somatostatin secretion. ABSTRACT: Glucagon, the body's principal hyperglycaemic hormone, is released from α-cells of the pancreatic islet. Secretion of this hormone is dysregulated in type 2 diabetes mellitus but the mechanisms controlling secretion are not well understood. Regulation of glucagon secretion by factors secreted by neighbouring ß- and δ-cells (paracrine regulation) have been proposed to be important. In this study, we explored the importance of paracrine regulation by using an optogenetic strategy. Specific light-induced activation of ß-cells in mouse islets expressing the light-gated channelrhodopsin-2 resulted in stimulation of electrical activity in δ-cells but suppression of α-cell activity. Activation of the δ-cells was rapid and sensitive to the gap junction inhibitor carbenoxolone, whereas the effect on electrical activity in α-cells was blocked by CYN 154806, an antagonist of the somatostatin-2 receptor. These observations indicate that optogenetic activation of the ß-cells propagates to the δ-cells via gap junctions, and the consequential stimulation of somatostatin secretion inhibits α-cell electrical activity by a paracrine mechanism. To explore whether this pathway is important for regulating α-cell activity and glucagon secretion in human islets, we constructed computational models of human islets. These models had detailed architectures based on human islets and consisted of a collection of >500 α-, ß- and δ-cells. Simulations of these models revealed that this gap junctional/paracrine mechanism accounts for up to 23% of the suppression of glucagon secretion by high glucose.


Assuntos
Simulação por Computador , Junções Comunicantes/fisiologia , Células Secretoras de Glucagon/fisiologia , Células Secretoras de Insulina/fisiologia , Células Secretoras de Somatostatina/fisiologia , Animais , Cálcio/metabolismo , Comunicação Celular , Células Cultivadas , Feminino , Células Secretoras de Glucagon/citologia , Células Secretoras de Glucagon/metabolismo , Glucose/metabolismo , Insulina/metabolismo , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Masculino , Camundongos , Somatostatina/metabolismo , Células Secretoras de Somatostatina/citologia , Células Secretoras de Somatostatina/metabolismo
12.
Methods Mol Biol ; 1507: 95-111, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27832535

RESUMO

Epigenetic mechanisms have been proposed to contribute to multiple complex diseases, including diabetes mellitus. Dissecting the epigenomic landscape of normal and disease states has greatly expanded our understanding of the regulation of key players in disease pathogenesis and is thus opening doors to novel therapeutic avenues. The human pancreatic islet is a pivotal micro-organ that maintains global glucose homeostasis. The heterogeneity of the islet impedes meaningful in-depth epigenetic analysis using whole islet tissue, because important marks in one cell type are masked by signals from other cell types. We describe here a detailed protocol for the isolation of highly purified endocrine cell subtypes (beta, alpha, and delta cells) from human cadaveric islets, to perform cell-type-specific epigenomic analysis of histone modifications as well as global gene expression profiling.


Assuntos
Epigênese Genética , Perfilação da Expressão Gênica , Ilhotas Pancreáticas/metabolismo , Biomarcadores/metabolismo , Separação Celular , Células Cultivadas , Cromatina/isolamento & purificação , Imunoprecipitação da Cromatina , DNA/isolamento & purificação , Células Enteroendócrinas/metabolismo , Citometria de Fluxo , Histonas/isolamento & purificação , Histonas/metabolismo , Humanos , Ilhotas Pancreáticas/citologia , Processamento de Proteína Pós-Traducional , RNA/genética , RNA/isolamento & purificação , RNA/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Análise de Sequência de RNA
13.
Mol Metab ; 5(7): 449-458, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27408771

RESUMO

OBJECTIVE: Complex local crosstalk amongst endocrine cells within the islet ensures tight coordination of their endocrine output. This is illustrated by the recent demonstration that the negative feedback control by delta cells within pancreatic islets determines the homeostatic set-point for plasma glucose during mouse postnatal development. However, the close association of islet endocrine cells that facilitates paracrine crosstalk also complicates the distinction between effects mediated directly on beta cells from indirect effects mediated via local intermediates, such as somatostatin from delta cells. METHODS: To resolve this problem, we generated reporter mice that allow collection of pure pancreatic delta cells along with alpha and beta cells from the same islets and generated comprehensive transcriptomes for each islet endocrine cell type. These transcriptomes afford an unparalleled view of the receptors expressed by delta, alpha and beta cells, and allow the prediction of which signal targets which endocrine cell type with great accuracy. RESULTS: From these transcriptomes, we discovered that the ghrelin receptor is expressed exclusively by delta cells within the islet, which was confirmed by fluorescent in situ hybridization and qPCR. Indeed, ghrelin increases intracellular calcium in delta cells in intact mouse islets, measured by GCaMP6 and robustly potentiates glucose-stimulated somatostatin secretion on mouse and human islets in both static and perfusion assays. In contrast, des-acyl-ghrelin at the same dose had no effect on somatostatin secretion and did not block the actions of ghrelin. CONCLUSIONS: These results offer a straightforward explanation for the well-known insulinostatic actions of ghrelin. Rather than engaging beta cells directly, ghrelin engages delta cells to promote local inhibitory feedback that attenuates insulin release. These findings illustrate the power of our approach to resolve some of the long-standing conundrums with regard to the rich feedback that occurs within the islet that is integral to islet physiology and therefore highly relevant to diabetes.

14.
Artigo em Coreano | WPRIM (Pacífico Ocidental) | ID: wpr-720546

RESUMO

Hepatosplenic gamma delta-cell lymphoma is a rare histologic type of peripheral T-cell lymphomas, clinically characterized by predominant involvement of liver and spleen, no or little adenopathy, and an often aggressive course. We report a case of bone marrow involvement of hepatosplenicgamma delta-cell lymphoma in a 21- year-old woman who presented with fever, anemia, thrombocytopenia, and hepatosplenomegaly. A lymphoma was found subsequently by bone marrow biopsy and computed tomography scan of the abdomen and pelvis. Immunologic characterization of lymphoma cells in bone marrow revealed positivity for CD2, CD3, and CD16/56, and negativity for CD4, CD5, CD7, CD8, CD34, and terminal deoxynucleotidyl transferase (TdT). Conventional cytogenetic studies revealed the presence of isochromosome 7q. Using the PCR-SSCP technique, monoclonal gene rearrangement of the T-cell receptor gamma chain was demonstrated. Thus, we could make a confirmatory diagnosis as hepatosplenic gamma delta-cell lymphoma.


Assuntos
Feminino , Humanos , Abdome , Anemia , Biópsia , Medula Óssea , Citogenética , Diagnóstico , DNA Nucleotidilexotransferase , Febre , Rearranjo Gênico , Isocromossomos , Fígado , Linfoma , Linfoma de Células T Periférico , Pelve , Receptores de Antígenos de Linfócitos T , Baço , Trombocitopenia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA