Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Immunity ; 45(4): 831-846, 2016 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-27760339

RESUMO

T follicular helper (Tfh) cells are a CD4+ T cell subset critical for long-lived humoral immunity. We hypothesized that integrins play a decisive role in Tfh cell biology. Here we show that Tfh cells expressed a highly active form of leukocyte function-associated antigen-1 (LFA-1) that was required for their survival within the germinal center niche. In addition, LFA-1 promoted expression of Bcl-6, a transcriptional repressor critical for Tfh cell differentiation, and inhibition of LFA-1 abolished Tfh cell generation and prevented protective humoral immunity to intestinal helminth infection. Furthermore, we demonstrated that expression of Talin-1, an adaptor protein that regulates LFA-1 affinity, dictated Tfh versus Th2 effector cell differentiation. Collectively, our results define unique functions for LFA-1 in the Tfh cell effector program and suggest that integrin activity is important in lineage decision-making events in the adaptive immune system.


Assuntos
Diferenciação Celular/imunologia , Ativação Linfocitária/imunologia , Antígeno-1 Associado à Função Linfocitária/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Animais , Células Cultivadas , Centro Germinativo/imunologia , Humanos , Imunidade Humoral/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Proto-Oncogênicas c-bcl-6/imunologia
2.
J Immunol ; 199(1): 244-252, 2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28533444

RESUMO

IgE production plays a crucial role in protective as well as pathogenic type 2 immune responses. Although the cytokine IL-4 is required for the development of IgE-producing plasma cells, the source of IL-4 and cellular requirements for optimal IgE responses remain unclear. Recent evidence suggests that T follicular helper (Tfh) cells are the primary producer of IL-4 in the reactive lymph node during type 2 immune responses. As Tfh cells are also required for the development of plasmablasts derived from germinal center and extrafollicular sources, we hypothesized that this cell subset is essential for the IgE plasmablast response. In this study, we show that during intestinal helminth infection, IL-4 derived from Tfh cells is required for IgE class switching and plasmablast formation. Notably, early IgE class switching did not require germinal center formation. Additionally, Tfh cell-derived IL-4 was required to maintain the Th2 response in the mesenteric lymph nodes of infected mice. Collectively, our results indicate that IL-4-producing Tfh cells are central orchestrators of the type 2 immune response in the reactive lymph nodes during parasitic helminth infection.


Assuntos
Helmintíase/imunologia , Imunoglobulina E/biossíntese , Interleucina-4/imunologia , Enteropatias Parasitárias/imunologia , Nematospiroides dubius , Infecções por Strongylida/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Animais , Linfócitos B/imunologia , Diferenciação Celular , Centro Germinativo/citologia , Centro Germinativo/imunologia , Helmintíase/parasitologia , Switching de Imunoglobulina , Imunoglobulina E/imunologia , Imunoglobulina G/imunologia , Enteropatias Parasitárias/parasitologia , Linfonodos/citologia , Linfonodos/imunologia , Camundongos , Plasmócitos/imunologia , Subpopulações de Linfócitos T
3.
J Biol Chem ; 288(34): 24825-33, 2013 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-23853095

RESUMO

In pancreatic ß-cells, glucose induces the binding of the transcription factor pancreatic duodenal homeobox-1 (PDX-1) to the insulin gene promoter to activate insulin gene transcription. At low glucose levels, glycogen synthase kinase 3ß (GSK3ß) is known to phosphorylate PDX-1 on C-terminal serine residues, which triggers PDX-1 proteasomal degradation. We previously showed that the serine/threonine Per-Arnt-Sim domain-containing kinase (PASK) regulates insulin gene transcription via PDX-1. However, the mechanisms underlying this regulation are unknown. In this study, we aimed to identify the role of PASK in the regulation of PDX-1 phosphorylation, protein expression, and stability in insulin-secreting cells and isolated rodent islets of Langerhans. We observed that glucose induces a decrease in overall PDX-1 serine phosphorylation and that overexpression of WT PASK mimics this effect. In vitro, PASK directly phosphorylates GSK3ß on its inactivating phosphorylation site Ser(9). Overexpression of a kinase-dead (KD), dominant negative version of PASK blocks glucose-induced Ser(9) phosphorylation of GSK3ß. Accordingly, GSK3ß Ser(9) phosphorylation is reduced in islets from pask-null mice. Overexpression of WT PASK or KD GSK3ß protects PDX-1 from degradation and results in increased PDX-1 protein abundance. Conversely, overexpression of KD PASK blocks glucose-induction of PDX-1 protein. We conclude that PASK phosphorylates and inactivates GSK3ß, thereby preventing PDX-1 serine phosphorylation and alleviating GSK3ß-mediated PDX-1 protein degradation in pancreatic ß-cells.


Assuntos
Quinase 3 da Glicogênio Sintase/metabolismo , Proteínas de Homeodomínio/metabolismo , Células Secretoras de Insulina/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transativadores/metabolismo , Animais , Glucose/farmacologia , Quinase 3 da Glicogênio Sintase/genética , Glicogênio Sintase Quinase 3 beta , Células Hep G2 , Proteínas de Homeodomínio/genética , Humanos , Células Secretoras de Insulina/citologia , Masculino , Camundongos , Mutação , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , Proteínas Serina-Treonina Quinases/genética , Estabilidade Proteica/efeitos dos fármacos , Ratos , Ratos Wistar , Edulcorantes/farmacologia , Transativadores/genética , Transcrição Gênica/efeitos dos fármacos , Transcrição Gênica/fisiologia
4.
Mucosal Immunol ; 16(6): 801-816, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37659724

RESUMO

Cluster of differentiation (CD4+) T cells consist of multiple subtypes, defined by expression of lineage-specific transcription factors, that contribute to the control of infectious diseases by providing help to immune and nonimmune target cells. In the current study, we examined the role of B cell lymphoma (Bcl)-6, a transcriptional repressor and master regulator of T follicular helper cell differentiation, in T cell-mediated host defense against intestinal and systemic parasitic infections. We demonstrate that while Bcl-6 expression by CD4+ T cells is critical for antibody-mediated protective immunity against secondary infection with the nematode Heligmosoides polygyrus bakeri, it paradoxically compromises worm expulsion during primary infection by limiting the generation of interleukin-10 (IL-10)-producing Gata3+ T helper 2 cells. Enhanced worm expulsion in the absence of Bcl-6 expressing T cells was associated with amplified intestinal goblet cell differentiation and increased generation of alternatively activated macrophages, effects that were reversed by neutralization of IL-10 signals. An increase in IL-10 production by Bcl-6-deficient CD4+ T cells was also evident in the context of systemic Leishmania donovani infection, but in contrast to Heligmosoides polygyrus bakeri infection, compromised T helper 1-mediated liver macrophage activation and increased susceptibility to this distinct parasitic challenge. Collectively, our studies suggest that host defense pathways that protect against parasite superinfection and lethal systemic protozoal infections can be engaged at the cost of compromised primary resistance to well-tolerated helminths.


Assuntos
Nematoides , Doenças Parasitárias , Animais , Interleucina-10 , Células Th2
5.
J Proteome Res ; 11(7): 3520-32, 2012 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-22578083

RESUMO

The prevalence of diabetes mellitus is increasing dramatically throughout the world, and the disease has become a major public health issue. The most common form of the disease, type 2 diabetes, is characterized by insulin resistance and insufficient insulin production from the pancreatic beta-cell. Since glucose is the most potent regulator of beta-cell function under physiological conditions, identification of the insulin secretory defect underlying type 2 diabetes requires a better understanding of glucose regulation of human beta-cell function. To this aim, a bottom-up LC-MS/MS-based proteomics approach was used to profile pooled islets from multiple donors under basal (5 mM) or high (15 mM) glucose conditions. Our analysis discovered 256 differentially abundant proteins (∼p < 0.05) after 24 h of high glucose exposure from more than 4500 identified in total. Several novel glucose-regulated proteins were elevated under high glucose conditions, including regulators of mRNA splicing (pleiotropic regulator 1), processing (retinoblastoma binding protein 6), and function (nuclear RNA export factor 1), in addition to neuron navigator 1 and plasminogen activator inhibitor 1. Proteins whose abundances markedly decreased during incubation at 15 mM glucose included Bax inhibitor 1 and synaptotagmin-17. Up-regulation of dicer 1 and SLC27A2 and down-regulation of phospholipase Cß4 were confirmed by Western blots. Many proteins found to be differentially abundant after high glucose stimulation are annotated as uncharacterized or hypothetical. These findings expand our knowledge of glucose regulation of the human islet proteome and suggest many hitherto unknown responses to glucose that require additional studies to explore novel functional roles.


Assuntos
Glucose/fisiologia , Ilhotas Pancreáticas/metabolismo , Proteoma/metabolismo , Acetiltransferases/genética , Acetiltransferases/isolamento & purificação , Acetiltransferases/metabolismo , Cromatografia por Troca Iônica , Cromatografia de Fase Reversa , Análise por Conglomerados , Coenzima A Ligases/metabolismo , RNA Helicases DEAD-box/metabolismo , Elongases de Ácidos Graxos , Regulação da Expressão Gênica , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/isolamento & purificação , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Mitocôndrias/enzimologia , Mapeamento de Peptídeos , Fosfolipase C beta/metabolismo , Proteoma/genética , Proteoma/isolamento & purificação , Proteômica , Ribonuclease III/metabolismo , Espectrometria de Massas em Tandem , Técnicas de Cultura de Tecidos
6.
J Biol Chem ; 286(51): 44005-44014, 2011 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-22065581

RESUMO

PAS kinase (PASK) is a glucose-regulated protein kinase involved in the control of pancreatic islet hormone release and insulin sensitivity. We aimed here to identify mutations in the PASK gene that may be associated with young-onset diabetes in humans. We screened 18 diabetic probands with unelucidated maturity-onset diabetes of the young (MODY). We identified two rare nonsynonymous mutations in the PASK gene (p.L1051V and p.G1117E), each of which was found in a single MODY family. Wild type or mutant PASKs were expressed in HEK 293 cells. Kinase activity of the affinity-purified proteins was assayed as autophosphorylation at amino acid Thr307 or against an Ugp1p-derived peptide. Whereas the PASK p.G1117E mutant displayed a ∼25% increase with respect to wild type PASK in the extent of autophosphorylation, and a ∼2-fold increase in kinase activity toward exogenous substrates, the activity of the p.L1051V mutant was unchanged. Amino acid Gly1117 is located in an α helical region opposing the active site of PASK and may elicit either: (a) a conformational change that increases catalytic efficiency or (b) a diminished inhibitory interaction with the PAS domain. Mouse islets were therefore infected with adenoviruses expressing wild type or mutant PASK and the regulation of insulin secretion was examined. PASK p.G1117E-infected islets displayed a 4-fold decrease in glucose-stimulated (16.7 versus 3 mM) insulin secretion, chiefly reflecting a 4.5-fold increase in insulin release at low glucose. In summary, we have characterized a rare mutation (p.G1117E) in the PASK gene from a young-onset diabetes family, which modulates glucose-stimulated insulin secretion.


Assuntos
Glucose/metabolismo , Insulina/metabolismo , Ilhotas Pancreáticas/citologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Adulto , Animais , Linhagem Celular , Diabetes Mellitus/metabolismo , Genômica , Glucagon/metabolismo , Células HEK293 , Proteínas de Choque Térmico HSP70/metabolismo , Humanos , Secreção de Insulina , Masculino , Proteínas de Membrana/metabolismo , Modelos Genéticos , Mutagênese , Fosforilação , Ratos , Ratos Wistar , Proteínas Recombinantes/metabolismo
7.
J Exp Med ; 219(9)2022 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-35938990

RESUMO

Enteric helminths form intimate physical connections with the intestinal epithelium, yet their ability to directly alter epithelial stem cell fate has not been resolved. Here we demonstrate that infection of mice with the parasite Heligmosomoides polygyrus bakeri (Hpb) reprograms the intestinal epithelium into a fetal-like state marked by the emergence of Clusterin-expressing revival stem cells (revSCs). Organoid-based studies using parasite-derived excretory-secretory products reveal that Hpb-mediated revSC generation occurs independently of host-derived immune signals and inhibits type 2 cytokine-driven differentiation of secretory epithelial lineages that promote their expulsion. Reciprocally, type 2 cytokine signals limit revSC differentiation and, consequently, Hpb fitness, indicating that helminths compete with their host for control of the intestinal stem cell compartment to promote continuation of their life cycle.


Assuntos
Nematospiroides dubius , Infecções por Strongylida , Animais , Citocinas , Mucosa Intestinal , Intestinos , Camundongos , Células-Tronco
8.
Cell Rep ; 38(10): 110502, 2022 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-35235831

RESUMO

Since the vast majority of species solely rely on innate immunity for host defense, it stands to reason that a critical evolutionary trait like immunological memory evolved in this primitive branch of our immune system. There is ample evidence that vaccines such as bacillus Calmette-Guérin (BCG) induce protective innate immune memory responses (trained immunity) against heterologous pathogens. Here we show that while BCG vaccination significantly reduces morbidity and mortality against influenza A virus (IAV), it fails to provide protection against severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). In contrast to IAV, SARS-CoV-2 infection leads to unique pulmonary vasculature damage facilitating viral dissemination to other organs, including the bone marrow (BM), a central site for BCG-mediated trained immunity. Finally, monocytes from BCG-vaccinated individuals mount an efficient cytokine response to IAV infection, while this response is minimal following SARS-CoV-2. Collectively, our data suggest that the protective capacity of BCG vaccination is contingent on viral pathogenesis and tissue tropism.


Assuntos
COVID-19 , Vírus da Influenza A , Vacina BCG , COVID-19/prevenção & controle , Humanos , Imunidade Inata , SARS-CoV-2 , Vacinação
9.
Biochim Biophys Acta ; 1801(3): 289-98, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19715772

RESUMO

The concept of glucolipotoxicity refers to the combined, deleterious effects of elevated glucose and fatty acid levels on pancreatic beta-cell function and survival. Significant progress has been made in recent years towards a better understanding of the cellular and molecular basis of glucolipotoxicity in the beta cell. The permissive effect of elevated glucose on the detrimental actions of fatty acids stems from the influence of glucose on intracellular fatty acid metabolism, promoting the synthesis of cellular lipids. The combination of excessive levels of fatty acids and glucose therefore leads to decreased insulin secretion, impaired insulin gene expression, and beta-cell death by apoptosis, all of which probably have distinct underlying mechanisms. Recent studies from our laboratory have identified several pathways implicated in fatty acid inhibition of insulin gene expression, including the extracellular-regulated kinase (ERK1/2) pathway, the metabolic sensor Per-Arnt-Sim kinase (PASK), and the ATF6 branch of the unfolded protein response. We have also confirmed in vivo in rats that the decrease in insulin gene expression is an early defect which precedes any detectable abnormality in insulin secretion. While the role of glucolipotoxicity in humans is still debated, the inhibitory effects of chronically elevated fatty acid levels has been clearly demonstrated in several studies, at least in individuals genetically predisposed to developing type 2 diabetes. It is therefore likely that glucolipotoxicity contributes to beta-cell failure in type 2 diabetes as well as to the decline in beta-cell function observed after the onset of the disease.


Assuntos
Ácidos Graxos/metabolismo , Glucose/metabolismo , Células Secretoras de Insulina/metabolismo , Transtornos do Metabolismo dos Lipídeos/metabolismo , Animais , Feminino , Humanos , Resistência à Insulina , Masculino , Obesidade/metabolismo
10.
Cell Rep ; 36(4): 109456, 2021 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-34320346

RESUMO

Interleukin-17 (IL-17)-producing γδ (γδ17) T cells are innate-like lymphocytes that contribute to protective anti-microbial responses but are also implicated in pathogenic inflammation at barrier sites. Understanding tissue-specific signals that regulate this subset is important to boost host defense mechanisms, but also to mitigate immunopathology. Here, we demonstrate that prostaglandin E2 (PGE2), a cyclooxygenase-dependent member of the eicosanoid family, directly enhances cytokine production by circulating and tissue-specific γδ17 T cells in vitro. Gain- and loss-of-function in vivo approaches further reveal that although provision of PGE2 amplifies psoriasiform inflammation, ablation of host mPGES1-dependent PGE2 synthesis is dispensable for cutaneous γδ17 T cell activation. By contrast, loss of endogenous PGE2 production or depletion of the gut microbiota compromises intestinal γδ17 T cell responses and increases disease severity during experimental colitis. Together, our results demonstrate how a lipid mediator can synergize with tissue-specific signals to enhance innate lymphocyte production of IL-17 during barrier inflammation.


Assuntos
Dinoprostona/farmacologia , Inflamação/metabolismo , Interleucina-17/biossíntese , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Pele/patologia , Animais , Colite/induzido quimicamente , Sulfato de Dextrana , Feminino , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/imunologia , Masculino , Camundongos Endogâmicos C57BL , Prostaglandina-E Sintases/metabolismo , Receptores de Antígenos de Linfócitos T gama-delta/efeitos dos fármacos
11.
Mucosal Immunol ; 13(2): 357-370, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31776431

RESUMO

Parasitic helminths cause significant damage as they migrate through host tissues to complete their life cycle. While chronic helminth infections are characterized by a well-described Type 2 immune response, the early, tissue-invasive stages are not well understood. Here we investigate the immune pathways activated during the early stages of Heligmosomoides polygyrus bakeri (Hpb), a natural parasitic roundworm of mice. In contrast to the Type 2 immune response present at later stages of infection, a robust Type 1 immune signature including IFNg production was dominant at the time of parasite invasion and granuloma formation. This early response was associated with an accumulation of activated Natural Killer (NK) cells, with no increase of other innate lymphoid cell populations. Parabiosis and confocal microscopy studies indicated that NK cells were recruited from circulation to the small intestine, where they surrounded parasitic larvae. NK cell recruitment required IFNγ receptor signaling, but was independent of CXCR3 expression. The depletion of tissue-infiltrating NK cells altered neither worm burden nor parasite fitness, but increased vascular injury, suggesting a role for NK cells in mediating tissue protection. Together, these data identify an unexpected role for NK cells in promoting disease tolerance during the invasive stage of an enteric helminth infection.


Assuntos
Trato Gastrointestinal/imunologia , Vigilância Imunológica , Intestinos/imunologia , Células Matadoras Naturais/imunologia , Nematospiroides dubius/fisiologia , Infecções por Strongylida/imunologia , Células Th1/metabolismo , Lesões do Sistema Vascular/imunologia , Animais , Movimento Celular , Feminino , Imunidade Inata , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Parabiose , Receptores CXCR/metabolismo , Receptores de Interferon/genética , Receptores de Interferon/metabolismo , Transdução de Sinais , Proteínas com Domínio T/metabolismo , Células Th1/imunologia , Receptor de Interferon gama
12.
Mucosal Immunol ; 12(3): 772-783, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30783183

RESUMO

Current treatments for tuberculosis (TB) are effective in controlling Mycobacterium tuberculosis (Mtb) growth, yet have significant side effects and do not prevent reinfection. Therefore, it is critical to understand why our host defense system is unable to generate permanent immunity to Mtb despite prolonged anti-tuberculosis therapy (ATT). Here, we demonstrate that treatment of mice with the most widely used anti-TB drugs, rifampicin (RIF) or isoniazid (INH) and pyrazinamide (PYZ), significantly altered the composition of the gut microbiota. Unexpectedly, treatment of mice with the pro-Mtb drugs INH and PYZ, but not RIF, prior to Mtb infection resulted in an increased bacterial burden, an effect that was reversible by fecal transplantation from untreated animals. Mechanistically, susceptibility of INH/PYZ-treated mice was associated with impaired metabolism of alveolar macrophages and defective bactericidal activity. Collectively, these data indicate that dysbiosis induced by ATT administered to millions of individuals worldwide may have adverse effects on the anti-Mtb response of alveolar macrophages.


Assuntos
Antibióticos Antituberculose/uso terapêutico , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/imunologia , Disbiose/imunologia , Microbioma Gastrointestinal/fisiologia , Macrófagos Alveolares/metabolismo , Mycobacterium tuberculosis/fisiologia , Tuberculose Pulmonar/tratamento farmacológico , Animais , Modelos Animais de Doenças , Disbiose/etiologia , Microbioma Gastrointestinal/efeitos dos fármacos , Interações Hospedeiro-Patógeno , Humanos , Imunidade Inata/efeitos dos fármacos , Imunomodulação , Isoniazida/uso terapêutico , Macrófagos Alveolares/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Pirazinamida/uso terapêutico , Rifampina/uso terapêutico
13.
Cell Rep ; 29(2): 391-405.e5, 2019 10 08.
Artigo em Inglês | MEDLINE | ID: mdl-31597099

RESUMO

Interleukin-17-producing γδ T (γδ17) cells play a central role in protective and pathogenic immune responses. However, the tissue-specific mechanisms that control the activation of these innate lymphocytes are not known. Here, we demonstrate that CD109, a glycosylphosphatidylinositol (GPI)-anchored protein highly expressed by keratinocytes, is an important regulator of skin homeostasis and γδ17 cell activation. Genetic deletion of CD109 results in spontaneous epidermal hyperplasia, aberrant accumulation of dermal-derived γδ17 cells, and enhanced susceptibility to psoriasiform inflammation. In this context, γδ17 activation requires interleukin (IL)-23 signals and is reversed by transient depletion of the skin microbiota. Mechanistically, CD109 restrains γδ17 cell activation in a cell-extrinsic manner by fortifying skin barrier integrity. Collectively, our data provide insight into the regulation of the skin IL-23/IL-17 immune axis and how homeostasis is maintained at this important barrier site.


Assuntos
Antígenos CD/metabolismo , Interleucina-17/biossíntese , Microbiota , Proteínas de Neoplasias/metabolismo , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Pele/imunologia , Células Th17/metabolismo , Animais , Epiderme/metabolismo , Feminino , Deleção de Genes , Humanos , Inflamação/patologia , Interleucina-23/metabolismo , Ativação Linfocitária/imunologia , Masculino , Camundongos Endogâmicos C57BL , Proteínas de Neoplasias/deficiência , Especificidade de Órgãos , Psoríase/imunologia , Psoríase/patologia , Pele/patologia
14.
Ann N Y Acad Sci ; 1070: 161-6, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16888159

RESUMO

Miniglucagon (glucagon 19-29) is the ultimate processing product of proglucagon, present in the glucagon-secreting granules of the alpha cells, at a close vicinity of the insulin-secreting beta cells. Co-released with glucagon and thanks to its original mode of action and its huge potency, it suppresses, inside the islet of Langerhans, the detrimental effect of glucagon on insulin secretion, while it leaves untouched the beneficial effect of glucagon on glucose competence of the beta cell. At the periphery, miniglucagon is processed at the surface of glucagon- and insulin-sensitive cells from circulating glucagon. At that level, it acts via a cellular pathway which uses initial molecular steps distinct from that of insulin which, when impaired, are involved in insulin resistence. This bypass allows miniglucagon to act as an insulin-like component, a characteristic which makes this peptide of particular interest from a pathophysiological and pharmacological point of views in understanding and treating metabolic diseases, such as the type 2 diabetes.


Assuntos
Glucagon/metabolismo , Animais , Transporte Biológico , Glucagon/química , Glucose/metabolismo , Metaloendopeptidases/metabolismo , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo
15.
Endocrinology ; 157(3): 1299-306, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26671180

RESUMO

Mouse ß-cell-specific reporter lines have played a key role in diabetes research. Although the rat provides several advantages, its use has lagged behind the mouse due to the relative paucity of genetic models. In this report we describe the generation and characterization of transgenic rats expressing a Renilla luciferase (RLuc)-enhanced yellow fluorescent protein (YFP) fusion under control of a 9-kb genomic fragment from the rat ins2 gene (RIP7-RLuc-YFP). Analysis of RLuc luminescence and YFP fluorescence revealed that reporter expression is restricted to ß-cells in the adult rat. Physiological characteristics including body weight, fat and lean mass, fasting and fed glucose levels, glucose and insulin tolerance, and ß-cell mass were similar between two RIP7-RLuc-YFP lines and wild-type littermates. Glucose-induced insulin secretion in isolated islets was indistinguishable from controls in one of the lines, whereas surprisingly, insulin secretion was defective in the second line. Consequently, subsequent studies were limited to the former line. We asked whether transgene activity was responsive to glucose as shown previously for the ins2 gene. Exposing islets ex vivo to high glucose (16.7 mM) or in vivo infusion of glucose for 24 hours increased luciferase activity in islets, whereas the fraction of YFP-positive ß-cells after glucose infusion was unchanged. Finally, we showed that fluorescence-activated cell sorting of YFP-positive islet cells can be used to enrich for ß-cells. Overall, this transgenic line will enable for the first time the application of both fluorescence and bioluminescence/luminescence-based approaches for the study of rat ß-cells.


Assuntos
Proteínas de Bactérias/genética , Genes Reporter/genética , Células Secretoras de Insulina/metabolismo , Insulina/genética , Luciferases de Renilla/genética , Proteínas Luminescentes/genética , Modelos Animais , Animais , Fusão Gênica Artificial , Proteínas de Bactérias/efeitos dos fármacos , Glicemia/metabolismo , Citometria de Fluxo , Genes Reporter/efeitos dos fármacos , Glucose/farmacologia , Teste de Tolerância a Glucose , Células Secretoras de Insulina/efeitos dos fármacos , Luciferases de Renilla/efeitos dos fármacos , Proteínas Luminescentes/efeitos dos fármacos , Masculino , Regiões Promotoras Genéticas , Ratos , Ratos Transgênicos
16.
Endocrinology ; 146(2): 702-12, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15539558

RESUMO

Miniglucagon (MG), the C-terminal glucagon fragment, processed from glucagon by the MG-generating endopeptidase (MGE) at the Arg17-Arg18 dibasic site, displays biological effects opposite to that of the mother-hormone. This secondary processing occurs in the glucagon- and MG-producing alpha-cells of the islets of Langerhans and from circulating glucagon. We first characterized the enzymatic activities of MGE in culture media from glucagon and MG-secreting alphaTC1.6 cells as made of a metalloendoprotease and an aminopeptidase. We observed that glucagon is a substrate for N-arginine dibasic convertase (NRDc), a metalloendoprotease, and that aminopeptidase B cleaves in vitro the intermediate cleavage products sequentially, releasing mature MG. Furthermore, immunodepletion of either enzyme resulted in the disappearance of the majority of MGE activity from the culture medium. We found RNAs and proteins corresponding to both enzymes in different cell lines containing a MGE activity (mouse alphaTC1.6 cells, rat hepatic FaO, and rat pituitary GH4C1). Using confocal microscopy, we observed a granular immunostaining of both enzymes in the alphaTC1.6 and native rat alpha-cells from islets of Langerhans. By immunogold electron microscopy, both enzymes were found in the mature secretory granules of alpha-cells, close to their substrate (glucagon) and their product (MG). Finally, we found NRDc only in the fractions from perfused pancreas that contain glucagon and MG after stimulation by hypoglycemia. We conclude that MGE is composed of NRDc and aminopeptidase B acting sequentially, providing a molecular basis for this uncommon regulatory process, which should be now addressed in both physiological and pathophysiological situations.


Assuntos
Aminopeptidases/metabolismo , Glucagon/biossíntese , Glucagon/metabolismo , Ilhotas Pancreáticas/enzimologia , Metaloendopeptidases/metabolismo , Fragmentos de Peptídeos/biossíntese , Aminopeptidases/antagonistas & inibidores , Aminopeptidases/genética , Animais , Cálcio/farmacologia , Células Cultivadas , Cobalto/farmacologia , Endopeptidases/genética , Endopeptidases/metabolismo , Hipoglicemia/metabolismo , Metaloendopeptidases/genética , Camundongos , Fenantrolinas/farmacologia , Inibidores de Proteases/farmacologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Especificidade por Substrato , Zinco/farmacologia
17.
Diabetes ; 51(2): 406-12, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11812748

RESUMO

Miniglucagon, the COOH-terminal (19-29) fragment processed from glucagon, is a potent and efficient inhibitor of insulin secretion from the MIN 6 beta-cell line. Using the rat isolated-perfused pancreas, we investigated the inhibitory effect of miniglucagon on insulin secretion and evaluated the existence of an inhibitory tone exerted by this peptide inside the islet. Miniglucagon dose-dependently inhibited insulin secretion stimulated by 8.3 mol/l glucose, with no change in the perfusion flow rate. A concentration of 1 nmol/l miniglucagon had a significant inhibitory effect on a 1 nmol/l glucagon-like peptide 1 (7-36) amide-potentiated insulin secretion. A decrease in extracellular glucose concentration simultaneously stimulated glucagon and miniglucagon secretion from pancreatic alpha-cells. Using confocal and electron microscopy analysis, we observed that miniglucagon is colocalized with glucagon in mature secretory granules of alpha-cells. Perfusion of an anti-miniglucagon antiserum directed against the biologically active moiety of the peptide resulted in a more pronounced effect of a glucose challenge on insulin secretion, indicating that miniglucagon exerts a local inhibitory tone on beta-cells. We concluded that miniglucagon is a novel local regulator of the pancreatic islet physiology and that any abnormal inhibitory tone exerted by this peptide on the beta-cell would result in an impaired insulin secretion, as observed in type 2 diabetes.


Assuntos
Glucagon/fisiologia , Ilhotas Pancreáticas/fisiologia , Fragmentos de Peptídeos/fisiologia , Animais , Imunofluorescência , Glucagon/imunologia , Glucagon/metabolismo , Soros Imunes/farmacologia , Técnicas In Vitro , Insulina/metabolismo , Secreção de Insulina , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/ultraestrutura , Masculino , Microscopia Eletrônica , Fragmentos de Peptídeos/imunologia , Fragmentos de Peptídeos/metabolismo , Ratos , Ratos Wistar
18.
Diabetes ; 64(12): 4112-22, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26283735

RESUMO

Cystic fibrosis (CF) is the result of mutations in the cystic fibrosis transmembrane conductance regulator (CFTR). CF-related diabetes affects 50% of adult CF patients. How CFTR deficiency predisposes to diabetes is unknown. Herein, we examined the impact of the most frequent cftr mutation in humans, deletion of phenylalanine at position 508 (ΔF508), on glucose homeostasis in mice. We compared ΔF508 mutant mice with wild-type (WT) littermates. Twelve-week-old male ΔF508 mutants had lower body weight, improved oral glucose tolerance, and a trend toward higher insulin tolerance. Glucose-induced insulin secretion was slightly diminished in ΔF508 mutant islets, due to reduced insulin content, but ΔF508 mutant islets were not more sensitive to proinflammatory cytokines than WT islets. Hyperglycemic clamps confirmed an increase in insulin sensitivity with normal ß-cell function in 12- and 18-week-old ΔF508 mutants. In contrast, 24-week-old ΔF508 mutants exhibited insulin resistance and reduced ß-cell function. ß-Cell mass was unaffected at 11 weeks of age but was significantly lower in ΔF508 mutants versus controls at 24 weeks. This was not associated with gross pancreatic pathology. We conclude that the ΔF508 CFTR mutation does not lead to an intrinsic ß-cell secretory defect but is associated with insulin resistance and a ß-cell mass deficit in aging mutants.


Assuntos
Envelhecimento , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Resistência à Insulina , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Mutação , Animais , Cruzamentos Genéticos , Fibrose Cística/genética , Fibrose Cística/metabolismo , Fibrose Cística/patologia , Fibrose Cística/fisiopatologia , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Diabetes Mellitus/etiologia , Regulação para Baixo , Feminino , Humanos , Imuno-Histoquímica , Insulina/sangue , Secreção de Insulina , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/patologia , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/patologia , Masculino , Camundongos Endogâmicos , Camundongos Mutantes , Técnicas de Cultura de Tecidos
19.
Diabetes ; 63(3): 982-93, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24194502

RESUMO

The cellular and molecular mechanisms underpinning the compensatory increase in ß-cell mass in response to insulin resistance are essentially unknown. We previously reported that a 72-h coinfusion of glucose and Intralipid (GLU+IL) induces insulin resistance and a marked increase in ß-cell proliferation in 6-month-old, but not in 2-month-old, Wistar rats. The aim of the current study was to identify the mechanisms underlying nutrient-induced ß-cell proliferation in this model. A transcriptomic analysis identified a central role for the forkhead transcription factor FOXM1 and its targets, and for heparin-binding epidermal growth factor (EGF)-like growth factor (HB-EGF), a ligand of the EGF receptor (EGFR), in nutrient-induced ß-cell proliferation. Phosphorylation of ribosomal S6 kinase, a mammalian target of rapamycin (mTOR) target, was increased in islets from GLU+IL-infused 6-month-old rats. HB-EGF induced proliferation of insulin-secreting MIN6 cells and isolated rat islets, and this effect was blocked in MIN6 cells by the EGFR inhibitor AG1478 or the mTOR inhibitor rapamycin. Coinfusion of either AG1478 or rapamycin blocked the increase in FOXM1 signaling, ß-cell proliferation, and ß-cell mass and size in response to GLU+IL infusion in 6-month-old rats. We conclude that chronic nutrient excess promotes ß-cell mass expansion via a pathway that involves EGFR signaling, mTOR activation, and FOXM1-mediated cell proliferation.


Assuntos
Proliferação de Células , Receptores ErbB/fisiologia , Fatores de Transcrição Forkhead/fisiologia , Células Secretoras de Insulina/fisiologia , Transdução de Sinais/fisiologia , Serina-Treonina Quinases TOR/fisiologia , Animais , Ciclo Celular , Células Cultivadas , Proteína Forkhead Box M1 , Perfilação da Expressão Gênica , Resistência à Insulina , Células Secretoras de Insulina/citologia , Masculino , Quinazolinas/farmacologia , Ratos , Ratos Wistar , Tirfostinas/farmacologia
20.
J Vis Exp ; (78)2013 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-23979115

RESUMO

Chronic exposure to excessive levels of nutrients is postulated to affect the function of several organs and tissues and to contribute to the development of the many complications associated with obesity and the metabolic syndrome, including type 2 diabetes. To study the mechanisms by which excessive levels of glucose and fatty acids affect the pancreatic beta-cell and the secretion of insulin, we have established a chronic nutrient infusion model in the rat. The procedure consists of catheterizing the right jugular vein and left carotid artery under general anesthesia; allowing a 7-day recuperation period; connecting the catheters to the pumps using a swivel and counterweight system that enables the animal to move freely in the cage; and infusing glucose and/or Intralipid (a soybean oil emulsion which generates a mixture of approximately 80% unsaturated/20% saturated fatty acids when infused with heparin) for 72 hr. This model offers several advantages, including the possibility to finely modulate the target levels of circulating glucose and fatty acids; the option to co-infuse pharmacological compounds; and the relatively short time frame as opposed to dietary models. It can be used to examine the mechanisms of nutrient-induced dysfunction in a variety of organs and to test the effectiveness of drugs in this context.


Assuntos
Nutrição Enteral/métodos , Glucose/administração & dosagem , Modelos Animais , Fosfolipídeos/administração & dosagem , Óleo de Soja/administração & dosagem , Animais , Glicemia/análise , Glicemia/metabolismo , Cateterismo Venoso Central/métodos , Emulsões/administração & dosagem , Nutrição Enteral/efeitos adversos , Fosfolipídeos/sangue , Ratos , Óleo de Soja/sangue
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA