Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
2.
Bioengineering (Basel) ; 9(12)2022 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-36550929

RESUMO

Type 1 diabetes results from the loss of pancreatic ß cells, reduced insulin secretion and dysregulated blood glucose levels. Replacement of these lost ß cells with stem cell-derived ß cells, and protecting these cells within macro-device implants is a promising approach to restore glucose homeostasis. However, to achieve this goal of restoration of glucose balance requires work to optimise ß cell function within implants. We know that native ß cell function is enhanced by cell-cell and cell-extracellular matrix interactions within the islets of Langerhans. Reproducing these interactions in 2D, such as culture on matrix proteins, does enhance insulin secretion. However, the impact of matrix proteins on the 3D organoids that would be in implants has not been widely studied. Here, we use native ß cells that are dispersed from islets and reaggregated into small spheroids. We show these ß cell spheroids have enhanced glucose-dependent insulin secretion when embedded into softer alginate hydrogels conjugated with RGD peptide (a common motif in extracellular matrix proteins). Embedding into alginate-RGD causes activation of integrin responses and repositioning of liprin, a protein that controls insulin secretion. We conclude that insulin secretion from ß cell spheroids can be enhanced through manipulation of the surrounding environment.

3.
Elife ; 112022 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-35559734

RESUMO

A developing understanding suggests that spatial compartmentalisation in pancreatic ß cells is critical in controlling insulin secretion. To investigate the mechanisms, we have developed live-cell subcellular imaging methods using the mouse organotypic pancreatic slice. We demonstrate that the organotypic pancreatic slice, when compared with isolated islets, preserves intact ß-cell structure, and enhances glucose-dependent Ca2+ responses and insulin secretion. Using the slice technique, we have discovered the essential role of local activation of integrins and the downstream component, focal adhesion kinase (FAK), in regulating ß cells. Integrins and FAK are exclusively activated at the ß-cell capillary interface and using in situ and in vitro models we show their activation both positions presynaptic scaffold proteins, like ELKS and liprin, and regulates glucose-dependent Ca2+ responses and insulin secretion. We conclude that FAK orchestrates the final steps of glucose-dependent insulin secretion within the restricted domain where ß-cell contact the islet capillaries.


Assuntos
Células Secretoras de Insulina , Ilhotas Pancreáticas , Animais , Cálcio/metabolismo , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Glucose/metabolismo , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Integrinas/metabolismo , Ilhotas Pancreáticas/metabolismo , Camundongos , Proteínas de Transporte Vesicular/metabolismo
4.
Front Endocrinol (Lausanne) ; 13: 842937, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35370948

RESUMO

We present a case of an obese 22-year-old man with activating GCK variant who had neonatal hypoglycemia, re-emerging with hypoglycemia later in life. We investigated him for asymptomatic hypoglycemia with a family history of hypoglycemia. Genetic testing yielded a novel GCK missense class 3 variant that was subsequently found in his mother, sister and nephew and reclassified as a class 4 likely pathogenic variant. Glucokinase enables phosphorylation of glucose, the rate-limiting step of glycolysis in the liver and pancreatic ß cells. It plays a crucial role in the regulation of insulin secretion. Inactivating variants in GCK cause hyperglycemia and activating variants cause hypoglycemia. Spleen-preserving distal pancreatectomy revealed diffuse hyperplastic islets, nuclear pleomorphism and periductular islets. Glucose stimulated insulin secretion revealed increased insulin secretion in response to glucose. Cytoplasmic calcium, which triggers exocytosis of insulin-containing granules, revealed normal basal but increased glucose-stimulated level. Unbiased gene expression analysis using 10X single cell sequencing revealed upregulated INS and CKB genes and downregulated DLK1 and NPY genes in ß-cells. Further studies are required to see if alteration in expression of these genes plays a role in the metabolic and histological phenotype associated with glucokinase pathogenic variant. There were more large islets in the patient's pancreas than in control subjects but there was no difference in the proportion of ß cells in the islets. His hypoglycemia was persistent after pancreatectomy, was refractory to diazoxide and improved with pasireotide. This case highlights the variable phenotype of GCK mutations. In-depth molecular analyses in the islets have revealed possible mechanisms for hyperplastic islets and insulin hypersecretion.


Assuntos
Glucoquinase , Hipoglicemia , Adulto , Glucoquinase/genética , Glucoquinase/metabolismo , Glucose , Humanos , Hipoglicemia/genética , Insulina/metabolismo , Secreção de Insulina , Masculino
5.
Cell Calcium ; 104: 102585, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35405569

RESUMO

Exocytotic release of hormones from endocrine cells must encompass mechanisms that direct the hormone into the blood stream. Increasing evidence indicates an intimate link between pancreatic ß-cells and the capillary bed of islets of Langerhans in both mouse and human. Integrins are exclusively activated at the region where ß-cells contact extracellular matrix proteins that surround the islet capillaries; furthermore, insulin granule exocytosis is targeted to this same region, therefore delivering hormone directly into the blood stream. In this review we discuss evidence suggesting that the capillary interface of ß-cells forms a specialised domain that is analogous to the presynaptic active zone of neurones. Pancreatic ß-cells possess many of the same proteins as found in the neuronal active zone, including several key presynaptic scaffold proteins. These scaffold proteins are enriched at the capillary interface of ß-cells and some have also been shown to control insulin secretion. We present a model that suggests this active zone-like domain in ß-cells may anchor key components of the stimulus secretion cascade, to not only target granule exocytosis to this region but also function as a significant regulator of insulin secretion.


Assuntos
Células Secretoras de Insulina , Ilhotas Pancreáticas , Animais , Exocitose/fisiologia , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Camundongos
6.
iScience ; 24(10): 103099, 2021 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-34622154

RESUMO

Pancreatic islets are essential for maintaining physiological blood glucose levels, and declining islet function is a hallmark of type 2 diabetes. We employ mass spectrometry-based proteomics to systematically analyze islets from 9 genetic or diet-induced mouse models representing a broad cross-section of metabolic health. Quantifying the islet proteome to a depth of >11,500 proteins, this study represents the most detailed analysis of mouse islet proteins to date. Our data highlight that the majority of islet proteins are expressed in all strains and diets, but more than half of the proteins vary in expression levels, principally due to genetics. Associating these varied protein expression levels on an individual animal basis with individual phenotypic measures reveals islet mitochondrial function as a major positive indicator of metabolic health regardless of strain. This compendium of strain-specific and dietary changes to mouse islet proteomes represents a comprehensive resource for basic and translational islet cell biology.

7.
Mol Pharm ; 18(12): 4428-4436, 2021 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-34649437

RESUMO

Type 1 diabetes develops in childhood and adolescence, with peak incidence in the early teenage years. There is an urgent need for an accurate method to detect insulin-producing ß-cells in patients that is not affected by alterations in ß-cell function. As part of our research program to design specific probes to measure ß-cell mass, we recently developed a novel insulin-binding peptide probe (IBPP) for the detection of ß-cells in vivo. Here, we applied our innovative method to show specific labeling of this IBPP to human and mouse fixed ß-cells in pancreatic islets. Importantly, we showed staining of human and mouse islets in culture without any negative functional or cell viability impact. Moreover, the IBPP-stained mouse islets after tail vein injection in vivo, albeit with batch differences in staining efficiency. In conclusion, we provide evidence showing that the IBPP can be used for future accurate detection of ß-cell mass in a variety of preclinical models of diabetes.


Assuntos
Diabetes Mellitus Tipo 1/diagnóstico por imagem , Células Secretoras de Insulina/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Animais , Células Cultivadas , Humanos , Insulina/análise , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Coloração e Rotulagem
8.
Metabolites ; 11(6)2021 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-34200432

RESUMO

Pancreatic ß cells secrete the hormone insulin into the bloodstream and are critical in the control of blood glucose concentrations. ß cells are clustered in the micro-organs of the islets of Langerhans, which have a rich capillary network. Recent work has highlighted the intimate spatial connections between ß cells and these capillaries, which lead to the targeting of insulin secretion to the region where the ß cells contact the capillary basement membrane. In addition, ß cells orientate with respect to the capillary contact point and many proteins are differentially distributed at the capillary interface compared with the rest of the cell. Here, we set out to develop an automated image analysis approach to identify individual ß cells within intact islets and to determine if the distribution of insulin across the cells was polarised. Our results show that a U-Net machine learning algorithm correctly identified ß cells and their orientation with respect to the capillaries. Using this information, we then quantified insulin distribution across the ß cells to show enrichment at the capillary interface. We conclude that machine learning is a useful analytical tool to interrogate large image datasets and analyse sub-cellular organisation.

9.
Front Endocrinol (Lausanne) ; 12: 667170, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33981287

RESUMO

Insulin-producing ß-cells constitute the majority of the cells in the pancreatic islets. Dysfunction of these cells is a key factor in the loss of glucose regulation that characterizes type 2 diabetes. The regulation of many of the functions of ß-cells relies on their close interaction with the intra-islet microvasculature, comprised of endothelial cells and pericytes. In addition to providing islet blood supply, cells of the islet vasculature directly regulate ß-cell activity through the secretion of growth factors and other molecules. These factors come from capillary mural pericytes and endothelial cells, and have been shown to promote insulin gene expression, insulin secretion, and ß-cell proliferation. This review focuses on the intimate crosstalk of the vascular cells and ß-cells and its role in glucose homeostasis and diabetes.


Assuntos
Diabetes Mellitus Tipo 2/patologia , Endotélio Vascular/fisiopatologia , Células Secretoras de Insulina/patologia , Microvasos , Neovascularização Patológica/fisiopatologia , Animais , Diabetes Mellitus Tipo 2/etiologia , Humanos
10.
Diabetologia ; 64(3): 618-629, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33399909

RESUMO

AIMS/HYPOTHESIS: We hypothesised that human beta cells are structurally and functional polarised with respect to the islet capillaries. We set out to test this using confocal microscopy to map the 3D spatial arrangement of key proteins and live-cell imaging to determine the distribution of insulin granule fusion around the cells. METHODS: Human pancreas samples were rapidly fixed and processed using the pancreatic slice technique, which maintains islet structure and architecture. Slices were stained using immunofluorescence for polarity markers (scribble, discs large [Dlg] and partitioning defective 3 homologue [Par3]) and presynaptic markers (liprin, Rab3-interacting protein [RIM2] and piccolo) and imaged using 3D confocal microscopy. Isolated human islets were dispersed and cultured on laminin-511-coated coverslips. Live 3D two-photon microscopy was used on cultured cells to image exocytic granule fusion events upon glucose stimulation. RESULTS: Assessment of the distribution of endocrine cells across human islets found that, despite distinct islet-to-islet complexity and variability, including multi-lobular islets, and intermixing of alpha and beta cells, there is still a striking enrichment of alpha cells at the islet mantle. Measures of cell position demonstrate that most beta cells contact islet capillaries. Subcellularly, beta cells consistently position polar determinants, such as Par3, Dlg and scribble, with a basal domain towards the capillaries and apical domain at the opposite face. The capillary interface/vascular face is enriched in presynaptic scaffold proteins, such as liprin, RIM2 and piccolo. Interestingly, enrichment of presynaptic scaffold proteins also occurs where the beta cells contact peri-islet capillaries, suggesting functional interactions. We also observed the same polarisation of synaptic scaffold proteins in islets from type 2 diabetic patients. Consistent with polarised function, isolated beta cells cultured onto laminin-coated coverslips target insulin granule fusion to the coverslip. CONCLUSIONS/INTERPRETATION: Structural and functional polarisation is a defining feature of human pancreatic beta cells and plays an important role in the control of insulin secretion.


Assuntos
Diabetes Mellitus Tipo 2/patologia , Células Secretoras de Insulina/patologia , Ilhotas Pancreáticas/irrigação sanguínea , Ilhotas Pancreáticas/patologia , Doadores de Tecidos , Biomarcadores/metabolismo , Grânulos Citoplasmáticos/metabolismo , Grânulos Citoplasmáticos/patologia , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Microscopia Confocal , Microscopia de Fluorescência por Excitação Multifotônica , Fenótipo , Vesículas Secretórias/metabolismo , Vesículas Secretórias/patologia , Técnicas de Cultura de Tecidos
11.
Stem Cells Transl Med ; 10(3): 492-505, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33145960

RESUMO

The differentiation of human stem cells into insulin secreting beta-like cells holds great promise to treat diabetes. Current protocols drive stem cells through stages of directed differentiation and maturation and produce cells that secrete insulin in response to glucose. Further refinements are now needed to faithfully phenocopy the responses of normal beta cells. A critical factor in normal beta cell behavior is the islet microenvironment which plays a central role in beta cell survival, proliferation, gene expression and secretion. One important influence on native cell responses is the capillary basement membrane. In adult islets, each beta cell makes a point of contact with basement membrane protein secreted by vascular endothelial cells resulting in structural and functional polarization. Interaction with basement membrane proteins triggers local activation of focal adhesions, cell orientation, and targeting of insulin secretion. This study aims to identifying the role of basement membrane proteins on the structure and function of human embryonic stem cell and induced pluripotent stem cell-derived beta cells. Here, we show that differentiated human stem cells-derived spheroids do contain basement membrane proteins as a diffuse web-like structure. However, the beta-like cells within the spheroid do not polarize in response to this basement membrane. We demonstrate that 2D culture of the differentiated beta cells on to basement membrane proteins enforces cell polarity and favorably alters glucose dependent insulin secretion.


Assuntos
Matriz Extracelular , Células Secretoras de Insulina , Células-Tronco Pluripotentes , Diferenciação Celular , Células Endoteliais , Glucose , Humanos , Insulina , Células Secretoras de Insulina/citologia , Células-Tronco Pluripotentes/citologia
12.
ACS Appl Mater Interfaces ; 12(51): 56908-56923, 2020 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-33314916

RESUMO

Encapsulation devices are an emerging barrier technology designed to prevent the immunorejection of replacement cells in regenerative therapies for intractable diseases. However, traditional polymers used in current devices are poor substrates for cell attachment and induce fibrosis upon implantation, impacting long-term therapeutic cell viability. Bioactivation of polymer surfaces improves local host responses to materials, and here we make the first step toward demonstrating the utility of this approach to improve cell survival within encapsulation implants. Using therapeutic islet cells as an exemplar cell therapy, we show that internal surface coatings improve islet cell attachment and viability, while distinct external coatings modulate local foreign body responses. Using plasma surface functionalization (plasma immersion ion implantation (PIII)), we employ hollow fiber semiporous poly(ether sulfone) (PES) encapsulation membranes and coat the internal surfaces with the extracellular matrix protein fibronectin (FN) to enhance islet cell attachment. Separately, the external fiber surface is coated with the anti-inflammatory cytokine interleukin-4 (IL-4) to polarize local macrophages to an M2 (anti-inflammatory) phenotype, muting the fibrotic response. To demonstrate the power of our approach, bioluminescent murine islet cells were loaded into dual FN/IL-4-coated fibers and evaluated in a mouse back model for 14 days. Dual FN/IL-4 fibers showed striking reductions in immune cell accumulation and elevated levels of the M2 macrophage phenotype, consistent with the suppression of fibrotic encapsulation and enhanced angiogenesis. These changes led to markedly enhanced islet cell survival and importantly to functional integration of the implant with the host vasculature. Dual FN/IL-4 surface coatings drive multifaceted improvements in islet cell survival and function, with significant implications for improving clinical translation of therapeutic cell-containing macroencapsulation implants.


Assuntos
Sobrevivência Celular/efeitos dos fármacos , Materiais Revestidos Biocompatíveis/química , Fibrose/prevenção & controle , Ilhotas Pancreáticas/metabolismo , Polímeros/química , Sulfonas/química , Animais , Adesão Celular/efeitos dos fármacos , Fibronectinas/química , Fibronectinas/farmacologia , Luciferina de Vaga-Lumes/farmacologia , Interleucina-4/química , Interleucina-4/farmacologia , Ilhotas Pancreáticas/diagnóstico por imagem , Ilhotas Pancreáticas/efeitos dos fármacos , Transplante das Ilhotas Pancreáticas/instrumentação , Transplante das Ilhotas Pancreáticas/métodos , Luciferases de Vaga-Lume/genética , Luciferases de Vaga-Lume/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Neovascularização Fisiológica/efeitos dos fármacos , Imagem Óptica , Próteses e Implantes , Células RAW 264.7
13.
J Biol Chem ; 295(27): 8901-8911, 2020 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-32341128

RESUMO

Within the pancreatic ß-cells, insulin secretory granules (SGs) exist in functionally distinct pools, displaying variations in motility as well as docking and fusion capability. Current therapies that increase insulin secretion do not consider the existence of these distinct SG pools. Accordingly, these approaches are effective only for a short period, with a worsening of glycemia associated with continued decline in ß-cell function. Insulin granule age is underappreciated as a determinant for why an insulin granule is selected for secretion and may explain why newly synthesized insulin is preferentially secreted from ß-cells. Here, using a novel fluorescent timer protein, we aimed to investigate the preferential secretion model of insulin secretion and identify how granule aging is affected by variation in the ß-cell environment, such as hyperglycemia. We demonstrate the use of a fluorescent timer construct, syncollin-dsRedE5TIMER, which changes its fluorescence from green to red over 18 h, in both microscopy and fluorescence-assisted organelle-sorting techniques. We confirm that the SG-targeting construct localizes to insulin granules in ß-cells and does not interfere with normal insulin SG behavior. We visualize insulin SG aging behavior in MIN6 and INS1 ß-cell lines and in primary C57BL/6J mouse and nondiabetic human islet cells. Finally, we separated young and old insulin SGs, revealing that preferential secretion of younger granules occurs in glucose-stimulated insulin secretion. We also show that SG population age is modulated by the ß-cell environment in vivo in the db/db mouse islets and ex vivo in C57BL/6J islets exposed to different glucose environments.


Assuntos
Secreção de Insulina/fisiologia , Insulina/metabolismo , Vesículas Secretórias/metabolismo , Animais , Linhagem Celular , Exocitose/fisiologia , Corantes Fluorescentes/química , Glucose/metabolismo , Humanos , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/fisiologia , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/fisiologia , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência/métodos , Fatores de Tempo
14.
J Cell Sci ; 133(6)2020 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-32079655

RESUMO

F-actin dynamics are known to control insulin secretion, but the point of intersection with the stimulus-secretion cascade is unknown. Here, using multiphoton imaging of ß cells isolated from Lifeact-GFP transgenic mice, we show that glucose stimulation does not cause global changes in subcortical F-actin. Instead, we observe spatially discrete and transient F-actin changes around each fusing granule. This F-actin remodelling is dependent on actin nucleation and is observed for granule fusion induced by either glucose or high potassium stimulation. Using GFP-labelled proteins, we identify local enrichment of Arp3, dynamin 2 and clathrin, all occurring after granule fusion, suggesting early recruitment of an endocytic complex to the fusing granules. Block of Arp2/3 activity with drugs or shRNA inhibits F-actin coating, traps granules at the cell membrane and reduces insulin secretion. Block of formin-mediated actin nucleation also blocks F-actin coating, but has no effect on insulin secretion. We conclude that local Arp2/3-dependent actin nucleation at the sites of granule fusion plays an important role in post-fusion granule dynamics and in the regulation of insulin secretion.


Assuntos
Complexo 2-3 de Proteínas Relacionadas à Actina , Actinas , Células Secretoras de Insulina , Complexo 2-3 de Proteínas Relacionadas à Actina/fisiologia , Actinas/genética , Actinas/metabolismo , Animais , Exocitose , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Camundongos
15.
EMBO Rep ; 21(3): e48692, 2020 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-32072744

RESUMO

Dysregulation of lipid homeostasis is intimately associated with defects in insulin secretion, a key feature of type 2 diabetes. Here, we explore the role of the putative lipid transporter ABCA12 in regulating insulin secretion from ß-cells. Mice with ß-cell-specific deletion of Abca12 display impaired glucose-stimulated insulin secretion and eventual islet inflammation and ß-cell death. ABCA12's action in the pancreas is independent of changes in the abundance of two other cholesterol transporters, ABCA1 and ABCG1, or of changes in cellular cholesterol or ceramide content. Instead, loss of ABCA12 results in defects in the genesis and fusion of insulin secretory granules and increases in the abundance of lipid rafts at the cell membrane. These changes are associated with dysregulation of the small GTPase CDC42 and with decreased actin polymerisation. Our findings establish a new, pleiotropic role for ABCA12 in regulating pancreatic lipid homeostasis and insulin secretion.


Assuntos
Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Transportador 1 de Cassete de Ligação de ATP/genética , Transportador 1 de Cassete de Ligação de ATP/metabolismo , Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Diabetes Mellitus Tipo 2/metabolismo , Glucose/metabolismo , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Camundongos
16.
J Mol Biol ; 432(5): 1407-1418, 2020 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-31711959

RESUMO

The islets of Langerhans or pancreatic islets are pivotal in the control of blood glucose and are complex microorgans embedded within the larger volume of the exocrine pancreas. Humans can have ~3.2 million islets [1] which, to our current knowledge, function in a similar manner to sense circulating blood glucose levels and respond with the secretion of a mix of different hormones that act to maintain glucose concentrations around a specific set point [2]. At a cellular level, the control of hormone secretion by glucose and other secretagogues is well-understood [3]. The key signal cascades have been identified and many details of the secretory process are known. However, if we shift focus from single cells and consider cells within intact islets, we do not have a comprehensive model as to how the islet environment influences cell function and how the islets work as a whole. This is important because there is overwhelming evidence that the structure and function of the individual endocrine cells are dramatically affected by the islet environment [4,5]. Uncovering the influence of this islet niche might drive future progress in treatments for Type 2 diabetes [6] and cell replacement therapies for Type 1 diabetes [7]. In this review, we focus on the insulin secreting beta cells and their interactions with the immediate environment that surrounds them including endocrine-endocrine interactions and contacts with capillaries.


Assuntos
Células Secretoras de Insulina , Ilhotas Pancreáticas/citologia , Animais , Capilares , Comunicação Celular , Diabetes Mellitus Tipo 1 , Diabetes Mellitus Tipo 2 , Matriz Extracelular/fisiologia , Glucose/metabolismo , Humanos , Secreção de Insulina/fisiologia , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/fisiologia , Ilhotas Pancreáticas/metabolismo , Transdução de Sinais
17.
Cell Rep ; 24(11): 2819-2826.e3, 2018 09 11.
Artigo em Inglês | MEDLINE | ID: mdl-30208309

RESUMO

The extracellular matrix (ECM) critically affects ß cell functions via integrin activation. But whether these ECM actions drive the spatial organization of ß cells, as they do in epithelial cells, is unknown. Here, we show that within islets of Langerhans, focal adhesion activation in ß cells occurs exclusively where they contact the capillary ECM (vascular face). In cultured ß cells, 3D mapping shows enriched insulin granule fusion where the cells contact ECM-coated coverslips, which depends on ß1 integrin receptor activation. Culture on micro-contact printed stripes of E-cadherin and fibronectin shows that ß cell contact at the fibronectin stripe selectively activates focal adhesions and enriches exocytic machinery and insulin granule fusion. Culture of cells in high glucose, as a model of glucotoxicity, abolishes granule targeting. We conclude that local integrin activation targets insulin secretion to the islet capillaries. This mechanism might be important for islet function and may change in disease.


Assuntos
Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Animais , Caderinas/metabolismo , Adesão Celular/fisiologia , Linhagem Celular Tumoral , Exocitose/fisiologia , Matriz Extracelular/metabolismo , Adesões Focais/metabolismo , Insulina/metabolismo , Secreção de Insulina/fisiologia , Integrinas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL
18.
Gastroenterology ; 154(6): 1805-1821.e5, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29360461

RESUMO

BACKGROUND & AIMS: Pancreatic acinar cells are polarized epithelial cells that store enzymes required for digestion as inactive zymogens, tightly packed at the cell apex. Stimulation of acinar cells causes the zymogen granules to fuse with the apical membrane, and the cells undergo exocytosis to release proteases into the intestinal lumen. Autophagy maintains homeostasis of pancreatic acini. Syntaxin 2 (STX2), an abundant soluble N-ethyl maleimide sensitive factor attachment protein receptor in pancreatic acini, has been reported to mediate apical exocytosis. Using human pancreatic tissues and STX2-knockout (KO) mice, we investigated the functions of STX2 in zymogen granule-mediated exocytosis and autophagy. METHODS: We obtained pancreatic tissues from 5 patients undergoing surgery for pancreatic cancer and prepared 80-µm slices; tissues were exposed to supramaximal cholecystokinin octapeptide (CCK-8) or ethanol and a low concentration of CCK-8 and analyzed by immunoblot and immunofluorescence analyses. STX2-KO mice and syntaxin 2+/+ C57BL6 mice (controls) were given intraperitoneal injections of supramaximal caerulein (a CCK-8 analogue) or fed ethanol and then given a low dose of caerulein to induce acute pancreatitis, or saline (controls); pancreata were isolated and analyzed by histology and immunohistochemistry. Acini were isolated from mice, incubated with CCK-8, and analyzed by immunofluorescence microscopy or used in immunoprecipitation experiments. Exocytosis was quantified using live-cell exocytosis and Ca2+ imaging analyses and based on formation of exocytotic soluble N-ethyl maleimide sensitive factor attachment protein receptor complexes. Dysregulations in autophagy were identified using markers, electron and immunofluorescence microscopy, and protease activation assays. RESULTS: Human pancreatic tissues and dispersed pancreatic acini from control mice exposed to CCK-8 or ethanol plus CCK-8 were depleted of STX2. STX2-KO developed more severe pancreatitis after administration of supramaximal caerulein or a 6-week ethanol diet compared with control. Acini from STX2-KO mice had increased apical exocytosis after exposure to CCK-8, as well as increased basolateral exocytosis, which led to ectopic release of proteases. These increases in apical and basolateral exocytosis required increased formation of fusogenic soluble N-ethyl maleimide sensitive factor attachment protein receptor complexes, mediated by STX3 and STX4. STX2 bound ATG16L1 and prevented it from binding clathrin. Deletion of STX2 from acini increased binding of AT16L1 to clathrin, increasing formation of pre-autophagosomes and inducing autophagy. Induction of autophagy promoted the CCK-8-induced increase in autolysosome formation and the activation of trypsinogen. CONCLUSIONS: In studies of human pancreatic tissues and pancreata from STX2-KO and control mice, we found STX2 to block STX3- and STX4-mediated fusion of zymogen granules with the plasma membrane and exocytosis and prevent binding of ATG16L1 to clathrin, which contributes to induction of autophagy. Exposure of pancreatic tissues to CCK-8 or ethanol depletes acinar cells of STX2, increasing basolateral exocytosis and promoting autophagy induction, leading to activation of trypsinogen.


Assuntos
Autofagia/genética , Exocitose/genética , Pâncreas/citologia , Pancreatite/genética , Sintaxina 1/metabolismo , Células Acinares/metabolismo , Animais , Membrana Celular/metabolismo , Ceruletídeo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pâncreas/patologia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/cirurgia , Pancreatite/induzido quimicamente , Vesículas Secretórias/fisiologia , Tripsinogênio/metabolismo
19.
J Biol Chem ; 293(7): 2510-2522, 2018 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-29284677

RESUMO

Epithelial pancreatic acinar cells perform crucial functions in food digestion, and acinar cell homeostasis required for secretion of digestive enzymes relies on SNARE-mediated exocytosis. The ubiquitously expressed Sec1/Munc18 protein mammalian uncoordinated-18c (Munc18c) regulates membrane fusion by activating syntaxin-4 (STX-4) to bind cognate SNARE proteins to form a SNARE complex that mediates exocytosis in many cell types. However, in the acinar cell, Munc18c's functions in exocytosis and homeostasis remain inconclusive. Here, we found that pancreatic acini from Munc18c-depleted mice (Munc18c+/-) and human pancreas (lenti-Munc18c-shRNA-treated) exhibit normal apical exocytosis of zymogen granules (ZGs) in response to physiologic stimulation with the intestinal hormone cholecystokinin (CCK-8). However, when stimulated with supraphysiologic CCK-8 levels to mimic pancreatitis, Munc18c-depleted (Munc18c+/-) mouse acini exhibited a reduction in pathological basolateral exocytosis of ZGs resulting from a decrease in fusogenic STX-4 SNARE complexes. This reduced basolateral exocytosis in part explained the less severe pancreatitis observed in Munc18c+/- mice after hyperstimulation with the CCK-8 analog caerulein. Likely as a result of this secretory blockade, Munc18c-depleted acini unexpectedly activated a component of the endoplasmic reticulum (ER) stress response that contributed to autophagy induction, resulting in downstream accumulation of autophagic vacuoles and autolysosomes. We conclude that Munc18c's role in mediating ectopic basolateral membrane fusion of ZGs contributes to the initiation of CCK-induced pancreatic injury, and that blockade of this secretory process could increase autophagy induction.


Assuntos
Ceruletídeo/efeitos adversos , Proteínas Munc18/metabolismo , Pancreatite/metabolismo , Idoso , Animais , Ceruletídeo/metabolismo , Colecistocinina/efeitos adversos , Colecistocinina/metabolismo , Retículo Endoplasmático/genética , Retículo Endoplasmático/metabolismo , Exocitose , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Proteínas Munc18/genética , Pâncreas/metabolismo , Pancreatite/genética , Proteínas SNARE/genética , Proteínas SNARE/metabolismo
20.
J Biol Chem ; 292(14): 5957-5969, 2017 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-28242761

RESUMO

A genuine understanding of human exocrine pancreas biology and pathobiology has been hampered by a lack of suitable preparations and reliance on rodent models employing dispersed acini preparations. We have developed an organotypic slice preparation of the normal portions of human pancreas obtained from cancer resections. The preparation was assessed for physiologic and pathologic responses to the cholinergic agonist carbachol (Cch) and cholecystokinin (CCK-8), including 1) amylase secretion, 2) exocytosis, 3) intracellular Ca2+ responses, 4) cytoplasmic autophagic vacuole formation, and 5) protease activation. Cch and CCK-8 both dose-dependently stimulated secretory responses from human pancreas slices similar to those previously observed in dispersed rodent acini. Confocal microscopy imaging showed that these responses were accounted for by efficient apical exocytosis at physiologic doses of both agonists and by apical blockade and redirection of exocytosis to the basolateral plasma membrane at supramaximal doses. The secretory responses and exocytotic events evoked by CCK-8 were mediated by CCK-A and not CCK-B receptors. Physiologic agonist doses evoked oscillatory Ca2+ increases across the acini. Supraphysiologic doses induced formation of cytoplasmic autophagic vacuoles and activation of proteases (trypsin, chymotrypsin). Maximal atropine pretreatment that completely blocked all the Cch-evoked responses did not affect any of the CCK-8-evoked responses, indicating that rather than acting on the nerves within the pancreas slice, CCK cellular actions directly affected human acinar cells. Human pancreas slices represent excellent preparations to examine pancreatic cell biology and pathobiology and could help screen for potential treatments for human pancreatitis.


Assuntos
Exocitose , Técnicas de Preparação Histocitológica/métodos , Modelos Biológicos , Pâncreas Exócrino/metabolismo , Pancreatite/metabolismo , Adulto , Idoso , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Pâncreas Exócrino/patologia , Pancreatite/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA