Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Diabetes ; 71(8): 1679-1693, 2022 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-35587617

RESUMO

The pancreatic islet depends on blood supply to efficiently sense plasma glucose levels and deliver insulin and glucagon into the circulation. Long believed to be passive conduits of nutrients and hormones, islet capillaries were recently found to be densely covered with contractile pericytes with the capacity to locally control blood flow. Here, we determined the contribution of pericyte regulation of islet blood flow to plasma insulin and glucagon levels and glycemia. Selective optogenetic activation of pericytes in intraocular islet grafts contracted capillaries and diminished blood flow. In awake mice, acute light-induced stimulation of islet pericytes decreased insulin and increased glucagon plasma levels, producing hyperglycemic effects. Interestingly, pericytes are the targets of sympathetic nerves in the islet, suggesting that sympathetic control of hormone secretion may occur in part by modulating pericyte activity and blood flow. Indeed, in vivo activation of pericytes with the sympathetic agonist phenylephrine decreased blood flow in mouse islet grafts, lowered plasma insulin levels, and increased glycemia. We further show that islet pericytes and blood vessels in living human pancreas slices responded to sympathetic input. Our findings indicate that pericytes mediate vascular responses in the islet that are required for adequate hormone secretion and glucose homeostasis. Vascular and neuronal alterations that are commonly seen in the islets of people with diabetes may impair regulation of islet blood flow and thus precipitate islet dysfunction.


Assuntos
Glucagon , Ilhotas Pancreáticas , Animais , Glicemia , Glucose/farmacologia , Homeostase , Humanos , Insulina , Ilhotas Pancreáticas/irrigação sanguínea , Camundongos , Pericitos
2.
Gastroenterology ; 160(3): 875-888.e11, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33121946

RESUMO

BACKGROUND AND AIMS: Destroying visceral sensory nerves impacts pancreatic islet function, glucose metabolism, and diabetes onset, but how islet endocrine cells interact with sensory neurons has not been studied. METHODS: We characterized the anatomical pattern of pancreatic sensory innervation by combining viral tracing, immunohistochemistry, and reporter mouse models. To assess the functional interactions of ß-cells with vagal sensory neurons, we recorded Ca2+ responses in individual nodose neurons in vivo while selectively stimulating ß-cells with chemogenetic and pharmacologic approaches. RESULTS: We found that pancreatic islets are innervated by vagal sensory axons expressing Phox2b, substance P, calcitonin-gene related peptide, and the serotonin receptor 5-HT3R. Centrally, vagal neurons projecting to the pancreas terminate in the commissural nucleus of the solitary tract. Nodose neurons responded in vivo to chemogenetic stimulation of ß-cells and to pancreas infusion with serotonin, but were not sensitive to insulin. Responses to chemogenetic and pharmacologic stimulation of ß-cells were blocked by a 5-HT3R antagonist and were enhanced by increasing serotonin levels in ß-cells. We further confirmed directly in living pancreas slices that sensory terminals in the islet were sensitive to serotonin. CONCLUSIONS: Our study establishes that pancreatic ß-cells communicate with vagal sensory neurons, likely using serotonin signaling as a transduction mechanism. Serotonin is coreleased with insulin and may therefore convey information about the secretory state of ß-cells via vagal afferent nerves.


Assuntos
Vias Aferentes/fisiologia , Comunicação Celular , Células Secretoras de Insulina/fisiologia , Gânglio Nodoso/fisiologia , Células Receptoras Sensoriais/fisiologia , Animais , Feminino , Insulina/metabolismo , Microscopia Intravital , Masculino , Camundongos , Camundongos Transgênicos , Microscopia Confocal , Modelos Animais , Gânglio Nodoso/citologia , Serotonina/metabolismo , Transdução de Sinais/fisiologia
3.
Diabetes ; 69(6): 1206-1218, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32245801

RESUMO

Endocrine cells of the pancreatic islet interact with their microenvironment to maintain tissue homeostasis. Communication with local macrophages is particularly important in this context, but the homeostatic functions of human islet macrophages are not known. In this study, we show that the human islet contains macrophages in perivascular regions that are the main local source of the anti-inflammatory cytokine interleukin-10 (IL-10) and the metalloproteinase MMP9. Macrophage production and secretion of these homeostatic factors are controlled by endogenous purinergic signals. In obese and diabetic states, macrophage expression of purinergic receptors MMP9 and IL-10 is reduced. We propose that in those states, exacerbated ß-cell activity due to increased insulin demand and increased cell death produce high levels of ATP that downregulate purinergic receptor expression. Loss of ATP sensing in macrophages may reduce their secretory capacity.


Assuntos
Ilhotas Pancreáticas/citologia , Macrófagos/fisiologia , Purinas/metabolismo , Animais , Antígenos CD/genética , Antígenos CD/metabolismo , Cálcio/metabolismo , Citocinas , Citosol/química , Citosol/fisiologia , Diabetes Mellitus/metabolismo , Regulação para Baixo , Regulação da Expressão Gênica , Humanos , Ilhotas Pancreáticas/diagnóstico por imagem , Camundongos , Receptores Purinérgicos/metabolismo , Transdução de Sinais , Transcriptoma
4.
Cell Metab ; 31(2): 215-216, 2020 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-32023444

RESUMO

Neuromodulation is a promising new therapeutic avenue to treat chronic diseases. A paper that appeared recently in Nature Biotechnology (Guyot et al., 2019) now provides a roadmap for how to establish electrostimulation of nerves to delay or even prevent type 1 diabetes.


Assuntos
Diabetes Mellitus Tipo 1 , Terapia por Estimulação Elétrica , Humanos
5.
Diabetes ; 69(4): 550-558, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31882565

RESUMO

Secretion of glucagon from the pancreatic α-cells is conventionally seen as the first and most important defense against hypoglycemia. Recent findings, however, show that α-cell signals stimulate insulin secretion from the neighboring ß-cell. This article focuses on these seemingly counterintuitive local actions of α-cells and describes how they impact islet biology and glucose metabolism. It is mostly based on studies published in the last decade on the physiology of α-cells in human islets and incorporates results from rodents where appropriate. As this and the accompanying articles show, the emerging picture of α-cell function is one of increased complexity that needs to be considered when developing new therapies aimed at promoting islet function in the context of diabetes.


Assuntos
Diabetes Mellitus/metabolismo , Células Secretoras de Glucagon/metabolismo , Glucagon/metabolismo , Comunicação Parácrina/fisiologia , Animais , Humanos , Insulina/metabolismo , Secreção de Insulina/fisiologia
7.
Nat Commun ; 10(1): 3700, 2019 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-31420552

RESUMO

Little is known about the role of islet delta cells in regulating blood glucose homeostasis in vivo. Delta cells are important paracrine regulators of beta cell and alpha cell secretory activity, however the structural basis underlying this regulation has yet to be determined. Most delta cells are elongated and have a well-defined cell soma and a filopodia-like structure. Using in vivo optogenetics and high-speed Ca2+ imaging, we show that these filopodia are dynamic structures that contain a secretory machinery, enabling the delta cell to reach a large number of beta cells within the islet. This provides for efficient regulation of beta cell activity and is modulated by endogenous IGF-1/VEGF-A signaling. In pre-diabetes, delta cells undergo morphological changes that may be a compensation to maintain paracrine regulation of the beta cell. Our data provides an integrated picture of how delta cells can modulate beta cell activity under physiological conditions.


Assuntos
Ilhotas Pancreáticas/ultraestrutura , Comunicação Parácrina , Estado Pré-Diabético/patologia , Pseudópodes/ultraestrutura , Células Secretoras de Somatostatina/ultraestrutura , Animais , Glicemia/metabolismo , Humanos , Fator de Crescimento Insulin-Like I/metabolismo , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/ultraestrutura , Microscopia Intravital , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/patologia , Camundongos , Camundongos Transgênicos , Microscopia Eletrônica , Imagem Óptica , Optogenética , Estado Pré-Diabético/metabolismo , Pseudópodes/metabolismo , Células Secretoras de Somatostatina/citologia , Células Secretoras de Somatostatina/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
8.
Nat Metab ; 1(11): 1110-1126, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-32432213

RESUMO

Pancreatic beta cells synthesize and secrete the neurotransmitter γ-aminobutyric acid (GABA) as a paracrine and autocrine signal to help regulate hormone secretion and islet homeostasis. Islet GABA release has classically been described as a secretory vesicle-mediated event. Yet, a limitation of the hypothesized vesicular GABA release from islets is the lack of expression of a vesicular GABA transporter in beta cells. Consequentially, GABA accumulates in the cytosol. Here we provide evidence that the human beta cell effluxes GABA from a cytosolic pool in a pulsatile manner, imposing a synchronizing rhythm on pulsatile insulin secretion. The volume regulatory anion channel (VRAC), functionally encoded by LRRC8A or Swell1, is critical for pulsatile GABA secretion. GABA content in beta cells is depleted and secretion is disrupted in islets from type 1 and type 2 diabetic patients, suggesting that loss of GABA as a synchronizing signal for hormone output may correlate with diabetes pathogenesis.


Assuntos
Citosol/metabolismo , Células Secretoras de Insulina/metabolismo , Ácido gama-Aminobutírico/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Homeostase , Humanos , Frações Subcelulares/metabolismo
9.
Cell Metab ; 27(3): 549-558.e4, 2018 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-29514065

RESUMO

Every animal species has a signature blood glucose level or glycemic set point. These set points are different, and the normal glycemic levels (normoglycemia) of one species would be life threatening for other species. Mouse normoglycemia can be considered diabetic for humans. The biological determinants of the glycemic set point remain unclear. Here we show that the pancreatic islet imposes its glycemic set point on the organism, making it the bona fide glucostat in the body. Moreover, and in contrast to rodent islets, glucagon input from the alpha cell to the insulin-secreting beta cell is necessary to fine-tune the distinctive human set point. These findings affect transplantation and regenerative approaches to treat diabetes because restoring normoglycemia may require more than replacing only the beta cells. Furthermore, therapeutic strategies using glucagon receptor antagonists as hypoglycemic agents need to be reassessed, as they may reset the overall glucostat in the organism.


Assuntos
Células Secretoras de Glucagon/metabolismo , Glucagon/metabolismo , Glucose/metabolismo , Células Secretoras de Insulina/metabolismo , Animais , Diabetes Mellitus Experimental , Humanos , Hipoglicemiantes/farmacologia , Transplante das Ilhotas Pancreáticas , Macaca fascicularis , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Comunicação Parácrina , Receptores de Glucagon/antagonistas & inibidores
10.
Cell Metab ; 27(3): 630-644.e4, 2018 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-29514070

RESUMO

Efficient insulin secretion requires a well-functioning pancreatic islet microvasculature. The dense network of islet capillaries includes the islet pericyte, a cell that has barely been studied. Here we show that islet pericytes help control local blood flow by adjusting islet capillary diameter. Islet pericytes cover 40% of the microvasculature, are contractile, and are innervated by sympathetic axons. Sympathetic adrenergic input increases pericyte activity and reduces capillary diameter and local blood flow. By contrast, activating beta cells by increasing glucose concentration inhibits pericytes, dilates islet capillaries, and increases local blood flow. These effects on pericytes are mediated by endogenous adenosine, which is likely derived from ATP co-released with insulin. Pericyte coverage of islet capillaries drops drastically in type 2 diabetes, suggesting that, under diabetic conditions, islets lose this mechanism to control their own blood supply. This may lead to inadequate insulin release into the circulation, further deteriorating glycemic control.


Assuntos
Capilares , Glucose/metabolismo , Insulina/metabolismo , Ilhotas Pancreáticas/irrigação sanguínea , Pericitos , Adenosina/metabolismo , Adolescente , Adulto , Animais , Capilares/citologia , Capilares/inervação , Capilares/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Feminino , Humanos , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Masculino , Camundongos , Pessoa de Meia-Idade , Pericitos/citologia , Pericitos/metabolismo , Fluxo Sanguíneo Regional
11.
Diabetologia ; 61(1): 182-192, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28884198

RESUMO

AIMS/HYPOTHESIS: Tissue-resident macrophages sense the microenvironment and respond by producing signals that act locally to maintain a stable tissue state. It is now known that pancreatic islets contain their own unique resident macrophages, which have been shown to promote proliferation of the insulin-secreting beta cell. However, it is unclear how beta cells communicate with islet-resident macrophages. Here we hypothesised that islet macrophages sense changes in islet activity by detecting signals derived from beta cells. METHODS: To investigate how islet-resident macrophages respond to cues from the microenvironment, we generated mice expressing a genetically encoded Ca2+ indicator in myeloid cells. We produced living pancreatic slices from these mice and used them to monitor macrophage responses to stimulation of acinar, neural and endocrine cells. RESULTS: Islet-resident macrophages expressed functional purinergic receptors, making them exquisite sensors of interstitial ATP levels. Indeed, islet-resident macrophages responded selectively to ATP released locally from beta cells that were physiologically activated with high levels of glucose. Because ATP is co-released with insulin and is exclusively secreted by beta cells, the activation of purinergic receptors on resident macrophages facilitates their awareness of beta cell secretory activity. CONCLUSIONS/INTERPRETATION: Our results indicate that islet macrophages detect ATP as a proxy signal for the activation state of beta cells. Sensing beta cell activity may allow macrophages to adjust the secretion of factors to promote a stable islet composition and size.


Assuntos
Trifosfato de Adenosina/metabolismo , Macrófagos/metabolismo , Pâncreas/citologia , Pâncreas/metabolismo , Animais , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/citologia , Camundongos
12.
Cell Rep ; 20(6): 1490-1501, 2017 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-28793270

RESUMO

Blood flow regulation in pancreatic islets is critical for function but poorly understood. Here, we establish an in vivo imaging platform in a non-human primate where islets transplanted autologously into the anterior chamber of the eye are monitored non-invasively and longitudinally at single-cell resolution. Engrafted islets were vascularized and innervated and maintained the cytoarchitecture of in situ islets in the pancreas. Blood flow velocity in the engrafted islets was not affected by increasing blood glucose levels and/or the GLP-1R agonist liraglutide. However, islet blood flow was dynamic in nature and fluctuated in various capillaries. This was associated with vasoconstriction events resembling a sphincter-like action, most likely regulated by adrenergic signaling. These observations suggest a mechanism in primate islets that diverts blood flow to cell regions with higher metabolic demand. The described imaging technology applied in non-human primate islets may contribute to a better understanding of human islet pathophysiology.


Assuntos
Velocidade do Fluxo Sanguíneo , Ilhotas Pancreáticas/irrigação sanguínea , Animais , Glicemia/metabolismo , Capilares/fisiologia , Células Cultivadas , Hipoglicemiantes/farmacologia , Ilhotas Pancreáticas/efeitos dos fármacos , Liraglutida/farmacologia , Macaca fascicularis , Masculino , Fluxo Sanguíneo Regional , Vasoconstrição
13.
Lab Chip ; 17(5): 772-781, 2017 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-28157238

RESUMO

We report the design and fabrication of a robust fluidic platform built out of inert plastic materials and micromachined features that promote optimized convective fluid transport. The platform is tested for perfusion interrogation of rodent and human pancreatic islets, dynamic secretion of hormones, concomitant live-cell imaging, and optogenetic stimulation of genetically engineered islets. A coupled quantitative fluid dynamics computational model of glucose stimulated insulin secretion and fluid dynamics was first utilized to design device geometries that are optimal for complete perfusion of three-dimensional islets, effective collection of secreted insulin, and minimization of system volumes and associated delays. Fluidic devices were then fabricated through rapid prototyping techniques, such as micromilling and laser engraving, as two interlocking parts from materials that are non-absorbent and inert. Finally, the assembly was tested for performance using both rodent and human islets with multiple assays conducted in parallel, such as dynamic perfusion, staining and optogenetics on standard microscopes, as well as for integration with commercial perfusion machines. The optimized design of convective fluid flows, use of bio-inert and non-absorbent materials, reversible assembly, manual access for loading and unloading of islets, and straightforward integration with commercial imaging and fluid handling systems proved to be critical for perfusion assay, and particularly suited for time-resolved optogenetics studies.


Assuntos
Técnicas Citológicas , Ilhotas Pancreáticas , Técnicas Analíticas Microfluídicas , Modelos Biológicos , Animais , Células Cultivadas , Técnicas Citológicas/instrumentação , Técnicas Citológicas/métodos , Desenho de Equipamento , Humanos , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/metabolismo , Camundongos , Técnicas Analíticas Microfluídicas/instrumentação , Técnicas Analíticas Microfluídicas/métodos , Microscopia de Fluorescência
14.
Adv Exp Med Biol ; 938: 11-24, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27586419

RESUMO

Type 1 diabetes (T1D) patients who receive pancreatic islet transplant experience significant improvement in their quality-of-life. This comes primarily through improved control of blood sugar levels, restored awareness of hypoglycemia, and prevention of serious and potentially life-threatening diabetes-associated complications, such as kidney failure, heart and vascular disease, stroke, nerve damage, and blindness. Therefore, beta cell replacement through transplantation of isolated islets is an important option in the treatment of T1D. However, lasting success of this promising therapy depends on durable survival and efficacy of the transplanted islets, which are directly influenced by the islet isolation procedures. Thus, isolating pancreatic islets with consistent and reliable quality is critical in the clinical application of islet transplantation.Quality of isolated islets is important in pre-clinical studies as well, as efforts to advance and improve clinical outcomes of islet transplant therapy have relied heavily on animal models ranging from rodents, to pigs, to nonhuman primates. As a result, pancreatic islets have been isolated from these and other species and used in a variety of in vitro or in vivo applications for this and other research purposes. Protocols for islet isolation have been somewhat similar across species, especially, in mammals. However, given the increasing evidence about the distinct structural and functional features of human and mouse islets, using similar methods of islet isolation may contribute to inconsistencies in the islet quality, immunogenicity, and experimental outcomes. This may also contribute to the discrepancies commonly observed between pre-clinical findings and clinical outcomes. Therefore, it is prudent to consider the particular features of pancreatic islets from different species when optimizing islet isolation protocols.In this chapter, we explore the structural and functional features of pancreatic islets from mice, pigs, nonhuman primates, and humans because of their prevalent use in nonclinical, preclinical, and clinical applications.


Assuntos
Ilhotas Pancreáticas/fisiologia , Animais , Humanos , Ilhotas Pancreáticas/irrigação sanguínea , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/inervação , Comunicação Parácrina , Transdução de Sinais
15.
Cell Metab ; 23(3): 541-6, 2016 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-26876561

RESUMO

Incretin mimetics are frequently used in the treatment of type 2 diabetes because they potentiate ß cell response to glucose. Clinical evidence showing short-term benefits of such therapeutics (e.g., liraglutide) is abundant; however, there have been several recent reports of unexpected complications in association with incretin mimetic therapy. Importantly, clinical evidence on the potential effects of such agents on the ß cell and islet function during long-term, multiyear use remains lacking. We now show that prolonged daily liraglutide treatment of >200 days in humanized mice, transplanted with human pancreatic islets in the anterior chamber of the eye, is associated with compromised release of human insulin and deranged overall glucose homeostasis. These findings raise concern about the chronic potentiation of ß cell function through incretin mimetic therapy in diabetes.


Assuntos
Diabetes Mellitus Experimental/tratamento farmacológico , Hipoglicemiantes/farmacologia , Células Secretoras de Insulina/fisiologia , Liraglutida/farmacologia , Animais , Glicemia , Diabetes Mellitus Experimental/sangue , Avaliação Pré-Clínica de Medicamentos , Olho/patologia , Humanos , Células Secretoras de Insulina/efeitos dos fármacos , Transplante das Ilhotas Pancreáticas , Camundongos
16.
Best Pract Res Clin Endocrinol Metab ; 28(5): 745-56, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25256769

RESUMO

The autonomic nervous system affects glucose metabolism partly through its connection to the pancreatic islet. Since its discovery by Paul Langerhans, the precise innervation patterns of the islet has remained elusive, mainly because of technical limitations. Using 3-dimensional reconstructions of axonal terminal fields, recent studies have determined the innervation patterns of mouse and human islets. In contrast to the mouse islet, endocrine cells within the human islet are sparsely contacted by autonomic axons. Instead, the invading sympathetic axons preferentially innervate smooth muscle cells of blood vessels. This innervation pattern suggests that, rather than acting directly on endocrine cells, sympathetic nerves may control hormone secretion by modulating blood flow in human islets. In addition to autonomic efferent axons, islets also receive sensory innervation. These axons transmit sensory information to the brain but also have the ability to locally release neuroactive substances that have been suggested to promote diabetes pathogenesis. We discuss recent findings on islet innervation, the connections of the islet with the brain, and the role islet innervation plays during the progression of diabetes.


Assuntos
Sistema Nervoso Autônomo/metabolismo , Encéfalo/metabolismo , Diabetes Mellitus/metabolismo , Ilhotas Pancreáticas/metabolismo , Neurônios/fisiologia , Animais , Sistema Nervoso Autônomo/fisiopatologia , Diabetes Mellitus/fisiopatologia , Metabolismo Energético/fisiologia , Humanos , Ilhotas Pancreáticas/fisiopatologia , Camundongos
17.
Diabetes ; 63(8): 2714-26, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24658304

RESUMO

Acetylcholine regulates hormone secretion from the pancreatic islet and is thus crucial for glucose homeostasis. Little is known, however, about acetylcholine (cholinergic) signaling in the human islet. We recently reported that in the human islet, acetylcholine is primarily a paracrine signal released from α-cells rather than primarily a neural signal as in rodent islets. In this study, we demonstrate that the effects acetylcholine produces in the human islet are different and more complex than expected from studies conducted on cell lines and rodent islets. We found that endogenous acetylcholine not only stimulates the insulin-secreting ß-cell via the muscarinic acetylcholine receptors M3 and M5, but also the somatostatin-secreting δ-cell via M1 receptors. Because somatostatin is a strong inhibitor of insulin secretion, we hypothesized that cholinergic input to the δ-cell indirectly regulates ß-cell function. Indeed, when all muscarinic signaling was blocked, somatostatin secretion decreased and insulin secretion unexpectedly increased, suggesting a reduced inhibitory input to ß-cells. Endogenous cholinergic signaling therefore provides direct stimulatory and indirect inhibitory input to ß-cells to regulate insulin secretion from the human islet.


Assuntos
Acetilcolina/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Transdução de Sinais/fisiologia , Técnicas Biossensoriais , Cálcio/química , Cálcio/metabolismo , Citoplasma , Regulação da Expressão Gênica , Glucagon/metabolismo , Humanos , Secreção de Insulina , Receptores Muscarínicos/genética , Receptores Muscarínicos/metabolismo , Somatostatina/metabolismo
18.
J Physiol ; 592(16): 3413-7, 2014 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-24591573

RESUMO

In this symposium review we discuss the role of neurotransmitters as paracrine signals that regulate pancreatic islet function. A large number of neurotransmitters and their receptors has been identified in the islet, but relatively little is known about their involvement in islet biology. Interestingly, neurotransmitters initially thought to be present in autonomic axons innervating the islet are also present in endocrine cells of the human islet. These neurotransmitters can thus be released as paracrine signals to help control hormone release. Here we propose that the role of neurotransmitters may extend beyond controlling endocrine cell function to work as signals modulating vascular flow and immune responses within the islet.


Assuntos
Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Neurotransmissores/metabolismo , Comunicação Parácrina , Animais , Humanos , Secreção de Insulina , Ilhotas Pancreáticas/inervação
19.
Endocrinol Metab Clin North Am ; 42(1): 39-56, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23391238

RESUMO

The autonomic nervous system helps regulate glucose homeostasis by acting on pancreatic islets of Langerhans. Despite decades of research on the innervation of the pancreatic islet, the mechanisms used by the autonomic nervous input to influence islet cell biology have not been elucidated. This article discusses how these barriers can be overcome to study the role of the autonomic innervation of the pancreatic islet in glucose metabolism. It describes recent advances in microscopy and novel approaches to studying the effects of nervous input that may help clarify how autonomic axons regulate islet biology.


Assuntos
Sistema Nervoso Autônomo/fisiologia , Diabetes Mellitus/fisiopatologia , Ilhotas Pancreáticas/inervação , Animais , Axônios/fisiologia , Humanos
20.
Proc Natl Acad Sci U S A ; 109(52): 21456-61, 2012 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-23236142

RESUMO

The autonomic nervous system is thought to modulate blood glucose homeostasis by regulating endocrine cell activity in the pancreatic islets of Langerhans. The role of islet innervation, however, has remained elusive because the direct effects of autonomic nervous input on islet cell physiology cannot be studied in the pancreas. Here, we used an in vivo model to study the role of islet nervous input in glucose homeostasis. We transplanted islets into the anterior chamber of the eye and found that islet grafts became densely innervated by the rich parasympathetic and sympathetic nervous supply of the iris. Parasympathetic innervation was imaged intravitally by using transgenic mice expressing GFP in cholinergic axons. To manipulate selectively the islet nervous input, we increased the ambient illumination to increase the parasympathetic input to the islet grafts via the pupillary light reflex. This reduced fasting glycemia and improved glucose tolerance. These effects could be blocked by topical application of the muscarinic antagonist atropine to the eye, indicating that local cholinergic innervation had a direct effect on islet function in vivo. By using this approach, we found that parasympathetic innervation influences islet function in C57BL/6 mice but not in 129X1 mice, which reflected differences in innervation densities and may explain major strain differences in glucose homeostasis. This study directly demonstrates that autonomic axons innervating the islet modulate glucose homeostasis.


Assuntos
Sistema Nervoso Autônomo/fisiologia , Olho/inervação , Ilhotas Pancreáticas/fisiologia , Modelos Biológicos , Animais , Proteínas de Fluorescência Verde/metabolismo , Iris/inervação , Iris/fisiologia , Transplante das Ilhotas Pancreáticas , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fibras Nervosas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...