Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Front Immunol ; 10: 68, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30774629

RESUMO

CLEC16A locus polymorphisms have been associated with several autoimmune diseases. We overexpressed CLEC16A in YTS natural killer (NK) cells and observed reduced NK cell cytotoxicity and IFN-γ release, delayed dendritic cell (DC) maturation, decreased conjugate formation, cell-surface receptor downregulation and increased autophagy. In contrast, siRNA mediated knockdown resulted in increased NK cell cytotoxicity, reversal of receptor expression and disrupted mitophagy. Subcellular localization studies demonstrated that CLEC16A is a cytosolic protein that associates with Vps16A, a subunit of class C Vps-HOPS complex, and modulates receptor expression via autophagy. Clec16a knockout (KO) in mice resulted in altered immune cell populations, increased splenic NK cell cytotoxicity, imbalance of dendritic cell subsets, altered receptor expression, upregulated cytokine and chemokine secretion. Taken together, our findings indicate that CLEC16A restrains secretory functions including cytokine release and cytotoxicity and that a delicate balance of CLEC16A is needed for NK cell function and homeostasis.


Assuntos
Células Matadoras Naturais/imunologia , Lectinas Tipo C/imunologia , Proteínas de Transporte de Monossacarídeos/imunologia , Animais , Doenças Autoimunes/genética , Linhagem Celular , Citotoxicidade Imunológica , Células Dendríticas/imunologia , Predisposição Genética para Doença , Humanos , Interferon gama/imunologia , Lectinas Tipo C/genética , Camundongos Knockout , Proteínas de Transporte de Monossacarídeos/genética , Baço/imunologia
2.
Diabetes ; 68(3): 598-608, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30552110

RESUMO

Although most patients with type 1 diabetes (T1D) continue to produce small amounts of insulin decades after disease onset, very few ß-cells persist within their pancreata. Consequently, the source of persistent insulin secretion within T1D remains unclear. We hypothesized that low-level insulin content within non-ß-cells could underlie persistent T1D insulin secretion. We tested for low levels of insulin (insulinlow) within a large cohort of JDRF Network for Pancreatic Organ Donors With Diabetes (nPOD) human pancreata across a wide range of ages and T1D disease durations. Long exposures, high-throughput imaging, and blinded parallel examiners allowed precise quantification of insulinlow cells. Of note, abundant islet endocrine cells with low quantities of insulin were present in most T1D pancreata. Insulinlow islet abundance and composition were not influenced by age, duration of diabetes, or age of onset. Insulinlow islets also contained ß-cell markers at variable levels, including Pdx1, Nkx6.1, GLUT1, and PC1/3. Most insulinlow cells contained abundant glucagon and other α-cell markers, suggesting that α-cells drive much of the insulinlow phenotype in T1D. However, pancreatic polypeptide, somatostatin, and ghrelin cells also contributed to the insulinlow cell population. Insulinlow cells represent a potential source of persistent insulin secretion in long-standing T1D and a possible target for regenerative therapies to expand ß-cell function in disease.


Assuntos
Diabetes Mellitus Tipo 1/metabolismo , Insulina/metabolismo , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Criança , Pré-Escolar , Feminino , Glucagon/metabolismo , Transportador de Glucose Tipo 1/metabolismo , Proteínas de Homeodomínio/metabolismo , Humanos , Imuno-Histoquímica , Lactente , Recém-Nascido , Ilhotas Pancreáticas/metabolismo , Masculino , Pessoa de Meia-Idade , Transativadores/metabolismo , Adulto Jovem
3.
Diabetes ; 67(4): 674-686, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29326366

RESUMO

The proliferative response of non-ß islet endocrine cells in response to type 1 diabetes (T1D) remains undefined. We quantified islet endocrine cell proliferation in a large collection of nondiabetic control and T1D human pancreata across a wide range of ages. Surprisingly, islet endocrine cells with abundant proliferation were present in many adolescent and young-adult T1D pancreata. But the proliferative islet endocrine cells were also present in similar abundance within control samples. We queried the proliferating islet cells with antisera against various islet hormones. Although pancreatic polypeptide, somatostatin, and ghrelin cells did not exhibit frequent proliferation, glucagon-expressing α-cells were highly proliferative in many adolescent and young-adult samples. Notably, α-cells only comprised a fraction (∼1/3) of the proliferative islet cells within those samples; most proliferative cells did not express islet hormones. The proliferative hormone-negative cells uniformly contained immunoreactivity for ARX (indicating α-cell fate) and cytoplasmic Sox9 (Sox9Cyt). These hormone-negative cells represented the majority of islet endocrine Ki67+ nuclei and were conserved from infancy through young adulthood. Our studies reveal a novel population of highly proliferative ARX+ Sox9Cyt hormone-negative cells and suggest the possibility of previously unrecognized islet development and/or lineage plasticity within adolescent and adult human pancreata.


Assuntos
Proliferação de Células , Diabetes Mellitus Tipo 1 , Células Secretoras de Glucagon/citologia , Adolescente , Adulto , Estudos de Casos e Controles , Linhagem da Célula , Plasticidade Celular , Criança , Pré-Escolar , Feminino , Grelina/metabolismo , Glucagon/metabolismo , Proteínas de Homeodomínio/metabolismo , Humanos , Lactente , Recém-Nascido , Ilhotas Pancreáticas/citologia , Antígeno Ki-67/metabolismo , Masculino , Polipeptídeo Pancreático/metabolismo , Fatores de Transcrição SOX9/metabolismo , Somatostatina/metabolismo , Fatores de Transcrição/metabolismo , Adulto Jovem
4.
Endocrinology ; 158(6): 1701-1714, 2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28323942

RESUMO

The impact of incretins upon pancreatic ß-cell expansion remains extremely controversial. Multiple studies indicate that incretin-based therapies can increase ß-cell proliferation, and incretins have been hypothesized to expand ß-cell mass. However, disagreement exists on whether incretins increase ß-cell mass. Moreover, some reports indicate that incretins may cause metaplastic changes in pancreatic histology. To resolve these questions, we treated a large cohort of mice with incretin-based therapies and carried out a rigorous analysis of ß-cell turnover and pancreatic histology using high-throughput imaging. Young mice received exenatide via osmotic pump, des-fluoro-sitagliptin, or glipizide compounded in diet for 2 weeks (short-term) on a low-fat diet (LFD) or 4.5 months (long-term) on a LFD or high-fat diet (HFD). Pancreata were quantified for ß-cell turnover and mass. Slides were examined for gross anatomical and microscopic changes to exocrine pancreas. Short-term des-fluoro-sitagliptin increased serum insulin and induced modest ß-cell proliferation but no change in ß-cell mass. Long-term incretin therapy in HFD-fed mice resulted in reduced weight gain, improved glucose homeostasis, and abrogated ß-cell mass expansion. No evidence for rapidly dividing progenitor cells was found in islets or pancreatic parenchyma, indicating that incretins do not induce islet neogenesis or pancreatic metaplasia. Contrasting prior reports, we found no evidence of ß-cell mass expansion after acute or chronic incretin therapy. Chronic incretin administration was not associated with histological abnormalities in pancreatic parenchyma; mice did not develop tumors, pancreatitis, or ductal hyperplasia. We conclude that incretin therapies do not generate ß-cells or alter pancreatic histology in young mice.


Assuntos
Proliferação de Células/efeitos dos fármacos , Hipoglicemiantes/uso terapêutico , Incretinas/uso terapêutico , Células Secretoras de Insulina/efeitos dos fármacos , Pâncreas/efeitos dos fármacos , Pâncreas/patologia , Animais , Contagem de Células , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/patologia , Dieta com Restrição de Gorduras , Dieta Hiperlipídica , Exenatida , Células Secretoras de Insulina/patologia , Células Secretoras de Insulina/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Peptídeos/uso terapêutico , Fosfato de Sitagliptina/uso terapêutico , Peçonhas/uso terapêutico
5.
PLoS One ; 11(7): e0159276, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27410263

RESUMO

The ß-cell mitogenic effects of ANGPTL8 have been subjected to substantial debate. The original findings suggested that ANGPTL8 overexpression in mice induced a 17-fold increase in ß-cell proliferation. Subsequent studies in mice contested this claim, but a more recent report in rats supported the original observations. These conflicting results might be explained by variable ANGPTL8 expression and differing methods of ß-cell quantification. To resolve the controversy, three independent labs collaborated on a blinded study to test the effects of ANGPTL8 upon ß-cell proliferation. Recombinant human betatrophin (hBT) fused to maltose binding protein (MBP) was delivered to mice by intravenous injection. The results demonstrate that ANGPTL8 does not stimulate significant ß-cell proliferation. Each lab employed different methods for ß-cell identification, resulting in variable quantification of ß-cell proliferation and suggests a need for standardizing practices for ß-cell quantification. We also observed a new action of ANGPTL8 in stimulating CD45+ hematopoietic-derived cell proliferation which may explain, in part, published discrepancies. Overall, the hypothesis that ANGPTL8 induces dramatic and specific ß-cell proliferation can no longer be supported. However, while ANGPTL8 does not stimulate robust ß-cell proliferation, the original experimental model using drug-induced (S961) insulin resistance was validated in subsequent studies, and thus still represents a robust system for studying signals that are either necessary or sufficient for ß-cell expansion. As an added note, we would like to commend collaborative group efforts, with repetition of results and procedures in multiple laboratories, as an effective method to resolve discrepancies in the literature.


Assuntos
Angiopoietinas/farmacologia , Linfócitos B/metabolismo , Proliferação de Células/efeitos dos fármacos , Proteínas Ligantes de Maltose/farmacologia , Mitógenos/farmacologia , Hormônios Peptídicos/farmacologia , Proteínas Recombinantes/farmacologia , Proteína 8 Semelhante a Angiopoietina , Proteínas Semelhantes a Angiopoietina , Angiopoietinas/metabolismo , Animais , Células Cultivadas , Masculino , Camundongos
6.
Proc Natl Acad Sci U S A ; 112(50): 15420-5, 2015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26627716

RESUMO

The great preclinical promise of the pancreatic endoplasmic reticulum kinase (PERK) inhibitors in neurodegenerative disorders and cancers is marred by pancreatic injury and diabetic syndrome observed in PERK knockout mice and humans lacking PERK function and suffering from Wolcott-Rallison syndrome. PERK mediates many of the unfolded protein response (UPR)-induced events, including degradation of the type 1 interferon (IFN) receptor IFNAR1 in vitro. Here we report that whole-body or pancreas-specific Perk ablation in mice leads to an increase in IFNAR1 protein levels and signaling in pancreatic tissues. Concurrent IFNAR1 deletion attenuated the loss of PERK-deficient exocrine and endocrine pancreatic tissues and prevented the development of diabetes. Experiments using pancreas-specific Perk knockouts, bone marrow transplantation, and cultured pancreatic islets demonstrated that stabilization of IFNAR1 and the ensuing increased IFN signaling in pancreatic tissues represents a major driver of injury triggered by Perk loss. Neutralization of IFNAR1 prevented pancreatic toxicity of PERK inhibitor, indicating that blocking the IFN pathway can mitigate human genetic disorders associated with PERK deficiency and help the clinical use of PERK inhibitors.


Assuntos
Interferon Tipo I/metabolismo , Pâncreas/enzimologia , Pâncreas/patologia , Receptor de Interferon alfa e beta/metabolismo , eIF-2 Quinase/antagonistas & inibidores , Animais , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Diabetes Mellitus Experimental/enzimologia , Diabetes Mellitus Experimental/patologia , Regulação para Baixo/efeitos dos fármacos , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Ativação Enzimática/efeitos dos fármacos , Imunofluorescência , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Camundongos , Pâncreas/efeitos dos fármacos , Inibidores de Proteínas Quinases/toxicidade , Transdução de Sinais/efeitos dos fármacos , Resposta a Proteínas não Dobradas , Regulação para Cima/efeitos dos fármacos , eIF-2 Quinase/metabolismo
7.
Diabetologia ; 58(7): 1523-31, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25917759

RESUMO

AIMS/HYPOTHESIS: The identification of novel targets that stimulate endogenous regeneration of beta cells would represent a significant advance in the treatment of patients with diabetes. The betatrophin hypothesis suggests that increased expression of angiopoietin-like protein 8 (ANGPTL8) induces dramatic and specific beta cell proliferation and subsequent beta cell mass expansion with improved glucose tolerance. In light of recent controversy, we further investigated the effects of ANGPTL8 overexpression on beta cell proliferation. METHODS: We performed hydrodynamic tail vein injections of green fluorescent protein (GFP) or Angptl8 (also known as Gm6484) DNA in multiple cohorts of mice of different ages. We employed state-of-the-art methods to comprehensively quantify beta cell mass and proliferation, controlling for mouse age, genetic strain, source of DNA injected, Angptl8 gene expression and proliferation markers. RESULTS: In two young and two aged cohorts of B6.129 mice, no substantial change in beta cell replication, mass or glucose homeostasis was observed following ANGPTL8 overexpression. Even in mice with extremely elevated Angptl8 expression (26-fold increase), beta cell replication was not significantly altered. Finally, we considered mice on the ICR background exactly as studied by Melton and colleagues, and still no beta cell mitogenic effect was detected following ANGPTL8 overexpression. CONCLUSION/INTERPRETATION: ANGPTL8 does not stimulate beta cell replication in young or old mice.


Assuntos
Angiopoietinas/biossíntese , Células Secretoras de Insulina/fisiologia , Hormônios Peptídicos/biossíntese , Envelhecimento/metabolismo , Proteína 8 Semelhante a Angiopoietina , Proteínas Semelhantes a Angiopoietina , Angiopoietinas/genética , Animais , Proliferação de Células , DNA/genética , Glucose/metabolismo , Metabolismo dos Lipídeos/genética , Metabolismo dos Lipídeos/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos ICR , Pancreatectomia , Hormônios Peptídicos/genética
8.
J Clin Invest ; 123(3): 990-5, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23454762

RESUMO

Preservation and regeneration of ß cell endocrine function is a long-sought goal in diabetes research. Defective insulin secretion from ß cells underlies both type 1 and type 2 diabetes, thus fueling considerable interest in molecules capable of rebuilding ß cell secretion capacity. Though early work in rodents suggested that regeneration might be possible, recent studies have revealed that aging powerfully restricts cell cycle entry of ß cells, which may limit regeneration capacity. Consequently, aging has emerged as an enigmatic challenge that might limit ß cell regeneration therapies. This Review summarizes recent data regarding the role of aging in ß cell regeneration and proposes models explaining these phenomena.


Assuntos
Envelhecimento , Diabetes Mellitus/patologia , Células Secretoras de Insulina/fisiologia , Animais , Ciclo Celular/genética , Inibidor p16 de Quinase Dependente de Ciclina/fisiologia , Epigênese Genética , Humanos , Insulina/metabolismo , Insulina/fisiologia , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Mitose , Pâncreas/patologia , Pâncreas/fisiopatologia , Regeneração
9.
Mol Cell Biol ; 32(24): 5129-39, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23071091

RESUMO

Germ line PERK mutations are associated with diabetes mellitus and growth retardation in both rodents and humans. In contrast, late embryonic excision of PERK permits islet development and was found to prevent onset of diabetes, suggesting that PERK may be dispensable in the adult pancreas. To definitively establish the functional role of PERK in adult pancreata, we generated mice harboring a conditional PERK allele in which excision is regulated by tamoxifen administration. Deletion of PERK in either young adult or mature adult mice resulted in hyperglycemia associated with loss of islet and ß cell architecture. PERK excision triggered intracellular accumulation of proinsulin and Glut2, massive endoplasmic reticulum (ER) expansion, and compensatory activation of the remaining unfolded-protein response (UPR) signaling pathways specifically in pancreatic tissue. Although PERK excision increased ß cell death, this was not a result of decreased proliferation as previously reported. In contrast, a significant and specific increase in ß cell proliferation was observed, a result reflecting increased cyclin D1 accumulation. This work demonstrates that contrary to expectations, PERK is required for secretory homeostasis and ß cell survival in adult mice.


Assuntos
Glucose/metabolismo , Pâncreas/metabolismo , eIF-2 Quinase/metabolismo , Animais , Sequência de Bases , Proliferação de Células , Ciclina D1/metabolismo , Diabetes Mellitus/etiologia , Diabetes Mellitus/genética , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patologia , Retículo Endoplasmático/metabolismo , Técnicas de Inativação de Genes , Homeostase , Hiperglicemia/etiologia , Hiperglicemia/genética , Hiperglicemia/metabolismo , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/patologia , Camundongos , Camundongos Knockout , Pâncreas/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Resposta a Proteínas não Dobradas , eIF-2 Quinase/deficiência , eIF-2 Quinase/genética
10.
Mol Cell ; 48(3): 353-64, 2012 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-23022383

RESUMO

MicroRNAs typically function at the level of posttranscriptional gene silencing within the cytoplasm; however, increasing evidence suggests that they may also function in nuclear, Argonaut-containing complexes, to directly repress target gene transcription. We have investigated the role of microRNAs in mediating endoplasmic reticulum (ER) stress responses. ER stress triggers the activation of three signaling molecules: Ire-1α/ß, PERK, and ATF6, whose function is to facilitate adaption to the ensuing stress. We demonstrate that PERK induces miR-211, which in turn attenuates stress-dependent expression of the proapoptotic transcription factor chop/gadd153. MiR-211 directly targets the proximal chop/gadd153 promoter, where it increases histone methylation and represses chop expression. Maximal chop accumulation ultimately correlates with miR-211 downregulation. Our data suggest a model in which PERK-dependent miR-211 induction prevents premature chop accumulation and thereby provides a window of opportunity for the cell to re-establish homeostasis prior to apoptotic commitment.


Assuntos
Regulação da Expressão Gênica , MicroRNAs/genética , Fator de Transcrição CHOP/genética , eIF-2 Quinase/genética , Fator 4 Ativador da Transcrição/genética , Fator 4 Ativador da Transcrição/metabolismo , Animais , Apoptose/genética , Linhagem Celular Tumoral , Sobrevivência Celular/genética , Células Cultivadas , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Estresse do Retículo Endoplasmático/genética , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Células HeLa , Histonas/metabolismo , Humanos , Metilação , Camundongos , Camundongos Knockout , MicroRNAs/metabolismo , Células NIH 3T3 , Fosforilação , Regiões Promotoras Genéticas/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tapsigargina/farmacologia , Fator de Transcrição CHOP/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , eIF-2 Quinase/metabolismo
12.
Mol Endocrinol ; 26(9): 1590-602, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22798294

RESUMO

The transcription factor HNF4α (hepatocyte nuclear factor-4α) is required for increased ß-cell proliferation during metabolic stress in vivo. We hypothesized that HNF4α could induce proliferation of human ß-cells. We employed adenoviral-mediated overexpression of an isoform of HNF4α (HNF4α8) alone, or in combination with cyclin-dependent kinase (Cdk)6 and Cyclin D3, in human islets. Heightened HNF4α8 expression led to a 300-fold increase in the number of ß-cells in early S-phase. When we overexpressed HNF4α8 together with Cdk6 and Cyclin D3, ß-cell cycle entry was increased even further. However, the punctate manner of bromodeoxyuridine incorporation into HNF4α(High) ß-cells indicated an uncoupling of the mechanisms that control the concise timing and execution of each cell cycle phase. Indeed, in HNF4α8-induced bromodeoxyuridine(+,punctate) ß-cells we observed signs of dysregulated DNA synthesis, cell cycle arrest, and activation of a double stranded DNA damage-associated cell cycle checkpoint mechanism, leading to the initiation of loss of ß-cell lineage fidelity. However, a substantial proportion of ß-cells stimulated to enter the cell cycle by Cdk6 and Cyclin D3 alone also exhibited a DNA damage response. HNF4α8 is a mitogenic signal in the human ß-cell but is not sufficient for completion of the cell cycle. The DNA damage response is a barrier to efficient ß-cell proliferation in vitro, and we suggest its evaluation in all attempts to stimulate ß-cell replication as an approach to diabetes treatment.


Assuntos
Fator 4 Nuclear de Hepatócito/metabolismo , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/citologia , Ciclo Celular , Divisão Celular , Células Cultivadas , Imunofluorescência , Glucose/farmacologia , Fator 4 Nuclear de Hepatócito/genética , Humanos , Marcação In Situ das Extremidades Cortadas , Insulina/metabolismo , Células Secretoras de Insulina/efeitos dos fármacos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
13.
Diabetes ; 60(3): 876-83, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21307078

RESUMO

OBJECTIVE: ß-Cell and islet endothelial cell destruction occurs during the progression of type 1 diabetes, but, paradoxically, ß-cell proliferation is increased during this period. Altered glucose tolerance may affect ß-cell mass and its association with endothelial cells. Our objective was to study the effects of glucose and inflammation on islet vascularity and on ß function, mass, and insulin in immunologically tolerant anti-CD3 monoclonal antibody (mAb)-treated and prediabetic NOD mice. RESEARCH DESIGN AND METHODS: The effects of phloridzin or glucose injections on ß-cells and endothelial cells were tested in prediabetic and previously diabetic NOD mice treated with anti-CD3 mAbs. Glucose tolerance, immunofluorescence staining, and examination of islet cultures ex vivo were evaluated. RESULTS: Islet endothelial cell density decreased in NOD mice and failed to recover after anti-CD3 mAb treatment despite baseline euglycemia. Glucose treatment of anti-CD3 mAb-treated mice showed increased islet vascular density and increased insulin content, which was associated with improved glucose tolerance. The increase in the vascular area was dependent on islet inflammation. Increased islet endothelial cell density was associated with increased production of vascular endothelial growth factor (VEGF) by islets from NOD mice. This response was recapitulated ex vivo by the transfer of supernatants from NOD islets cultured in high-glucose levels. CONCLUSIONS: Our results demonstrate a novel role for glucose and inflammation in the control of islet vasculature and insulin content of ß-cells in prediabetic and anti-CD3-treated NOD mice. VEGF production by the islets is affected by glucose levels and is imparted by soluble factors released by inflamed islets.


Assuntos
Diabetes Mellitus Tipo 1/metabolismo , Glucose/metabolismo , Inflamação/metabolismo , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/irrigação sanguínea , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Diabetes Mellitus Tipo 1/fisiopatologia , Imunofluorescência , Inflamação/fisiopatologia , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/fisiopatologia , Camundongos , Camundongos Endogâmicos NOD , Reação em Cadeia da Polimerase Via Transcriptase Reversa
14.
J Vis Exp ; (46)2010 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-21178965

RESUMO

Accurate measurement of cell division is a fundamental challenge in experimental biology that becomes increasingly complex when slowly dividing cells are analyzed. Established methods to detect cell division include direct visualization by continuous microscopy in cell culture, dilution of vital dyes such as carboxyfluorescein di-aetate succinimidyl ester (CFSE), immuno-detection of mitogenic antigens such as ki67 or PCNA, and thymidine analogues. Thymidine analogues can be detected by a variety of methods including radio-detection for tritiated thymidine, immuno-detection for bromo-deoxyuridine (BrdU), chloro-deoxyuridine (CldU) and iodo-deoxyuridine (IdU), and chemical detection for ethinyl-deoxyuridine (EdU). We have derived a strategy to detect sequential incorporation of different thymidine analogues (CldU and IdU) into tissues of adult mice. Our method allows investigators to accurately quantify two successive rounds of cell division. By optimizing immunostaining protocols our approach can detect very low dose thymidine analogues administered via the drinking water, safe to administer to mice for prolonged periods of time. Consequently, our technique can be used to detect cell turnover in very long-lived tissues. Optimal immunofluoresent staining results can be achieved in multiple tissue types, including pancreas, skin, gut, liver, adrenal, testis, ovary, thyroid, lymph node, and brain. We have also applied this technique to identify oncogenic transformation within tissues. We have further applied this technique to determine if transit-amplifying cells contribute to growth or renewal of tissues. In this sense, sequential administration of thymidine analogues represents a novel approach for studying the origins and survival of cells involved in tissue homeostasis.


Assuntos
Desoxiuridina/análogos & derivados , Idoxuridina/análise , Microscopia de Fluorescência/métodos , Timidina/análogos & derivados , Animais , Desoxiuridina/análise , Feminino , Processamento de Imagem Assistida por Computador/métodos , Camundongos , Pâncreas/química , Timidina/análise
15.
J Biol Chem ; 285(51): 40050-9, 2010 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-20943662

RESUMO

The calcium-regulated phosphatase calcineurin intersects with both calcium and cAMP-mediated signaling pathways in the pancreatic ß-cell. Pharmacologic calcineurin inhibition, necessary to prevent rejection in the setting of organ transplantation, is associated with post-transplant ß-cell failure. We sought to determine the effect of calcineurin inhibition on ß-cell replication and survival in rodents and in isolated human islets. Further, we assessed whether the GLP-1 receptor agonist and cAMP stimulus, exendin-4 (Ex-4), could rescue ß-cell replication and survival following calcineurin inhibition. Following treatment with the calcineurin inhibitor tacrolimus, human ß-cell apoptosis was significantly increased. Although we detected no human ß-cell replication, tacrolimus significantly decreased rodent ß-cell replication. Ex-4 nearly normalized both human ß-cell survival and rodent ß-cell replication when co-administered with tacrolimus. We found that tacrolimus decreased Akt phosphorylation, suggesting that calcineurin could regulate replication and survival via the PI3K/Akt pathway. We identify insulin receptor substrate-2 (Irs2), a known cAMP-responsive element-binding protein target and upstream regulator of the PI3K/Akt pathway, as a novel calcineurin target in ß-cells. Irs2 mRNA and protein are decreased by calcineurin inhibition in both rodent and human islets. The effect of calcineurin on Irs2 expression is mediated at least in part through the nuclear factor of activated T-cells (NFAT), as NFAT occupied the Irs2 promoter in a calcineurin-sensitive manner. Ex-4 restored Irs2 expression in tacrolimus-treated rodent and human islets nearly to baseline. These findings reveal calcineurin as a regulator of human ß-cell survival in part through regulation of Irs2, with implications for the pathogenesis and treatment of diabetes following organ transplantation.


Assuntos
Calcineurina/farmacologia , Proliferação de Células/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Calcineurina/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , AMP Cíclico/metabolismo , Diabetes Mellitus/etiologia , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patologia , Exenatida , Regulação da Expressão Gênica/efeitos dos fármacos , Receptor do Peptídeo Semelhante ao Glucagon 1 , Humanos , Hipoglicemiantes/farmacologia , Imunossupressores/farmacologia , Proteínas Substratos do Receptor de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Camundongos , Fatores de Transcrição NFATC/metabolismo , Transplante de Órgãos/efeitos adversos , Peptídeos/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Glucagon/agonistas , Receptores de Glucagon/metabolismo , Tacrolimo/farmacologia , Peçonhas/farmacologia
16.
Mol Endocrinol ; 23(11): 1865-75, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19628581

RESUMO

The molecular determinants of beta-cell mass expansion remain poorly understood. Cyclin D2 is the major D-type cyclin expressed in beta-cells, essential for adult beta-cell growth. We hypothesized that cyclin D2 could be actively regulated in beta-cells, which could allow mitogenic stimuli to influence beta-cell expansion. Cyclin D2 protein was sharply increased after partial pancreatectomy, but cyclin D2 mRNA was unchanged, suggesting posttranscriptional regulatory mechanisms influence cyclin D2 expression in beta-cells. Consistent with this hypothesis, cyclin D2 protein stability is powerfully regulated in fibroblasts. Threonine 280 of cyclin D2 is phosphorylated, and this residue critically limits D2 stability. We derived transgenic (tg) mice with threonine 280 of cyclin D2 mutated to alanine (T280A) or wild-type cyclin D2 under the control of the insulin promoter. Cyclin D2 T280A protein was expressed at much higher levels than wild-type cyclin D2 protein in beta-cells, despite equivalent expression of tg mRNAs. Cyclin D2 T280A tg mice exhibited a constitutively nuclear cyclin D2 localization in beta-cells, and increased cyclin D2 stability in islets. Interestingly, threonine 280-mutant cyclin D2 tg mice had greatly reduced beta-cell apoptosis, with suppressed expression of proapoptotic genes. Suppressed beta-cell apoptosis in threonine 280-mutant cyclin D2 tg mice resulted in greatly increased beta-cell area in aged mice. Taken together, these data indicate that cyclin D2 is regulated by protein stability in pancreatic beta-cells, that signals that act upon threonine 280 limit cyclin D2 stability in beta-cells, and that threonine 280-mutant cyclin D2 overexpression prolongs beta-cell survival and augments beta-cell mass expansion.


Assuntos
Ciclina D2/fisiologia , Regulação da Expressão Gênica , Células Secretoras de Insulina/citologia , Alanina/genética , Animais , Apoptose , Cruzamentos Genéticos , Ciclina D2/metabolismo , Exenatida , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Biológicos , Mutação , Peptídeos/metabolismo , RNA Mensageiro/metabolismo , Treonina/genética , Peçonhas/metabolismo
17.
Am J Pathol ; 174(1): 276-86, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19095950

RESUMO

Mutations in the phosphatase and tensin homologue (PTEN)/phosphatidylinositol-3 kinase-alpha (PI3K) signaling pathway are frequently found in human cancer. In addition, Pten(+/-) mice develop tumors in multiple organs because of the activation of the PI3K signaling cascade. Because activation of PI3K signaling leads to feedback inhibition of insulin receptor substrate-2 (IRS2) expression, an upstream activator of PI3K, we therefore anticipated that IRS2 expression would be low in tumors that lack PTEN. Surprisingly, however, an elevation of IRS2 was often detected in tumor samples in which PTEN levels were compromised. To determine the potential contribution of Irs2 to tumor progression, Pten(+/-) mice were crossed with Irs2(+/-) mice. Deletion of Irs2 did not affect the initiation of neoplasia found in Pten(+/-) mice but suppressed cancer cell growth, proliferation, and invasion through the basement membrane. Deletion of Irs2 also attenuated the expression of Myc in prostatic intraepithelial neoplasia in Pten(+/-) mice. In addition, the expression levels of IRS2 and MYC were highly correlated in human prostate cancer, and IRS2 could stimulate MYC expression in cultured cells. Our findings provide evidence that the PI3K-activating adaptor Irs2 contributes to tumor progression in Pten(+/-) mice by stimulating both Myc and DNA synthesis.


Assuntos
Proteínas Substratos do Receptor de Insulina/genética , Proteínas Substratos do Receptor de Insulina/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo , Animais , Western Blotting , Linhagem Celular Tumoral , Progressão da Doença , Neoplasias do Endométrio/genética , Neoplasias do Endométrio/metabolismo , Neoplasias do Endométrio/patologia , Feminino , Humanos , Imuno-Histoquímica , Hibridização In Situ , Masculino , Camundongos , Camundongos Mutantes , Neoplasias/patologia , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Proteínas Proto-Oncogênicas c-myc/metabolismo , Transfecção
18.
Endocrinology ; 148(11): 5136-44, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17673522

RESUMO

Immune modulators can arrest loss of insulin secretion in type 1 diabetes mellitus (T1DM), but they have not caused permanent disease remission or restored normal insulin secretion. We tested whether exendin-4, a glucagon-like peptide-1 receptor agonist, would enhance remission of T1DM in NOD mice treated with anti-CD3 monoclonal antibody (mAb) and studied the effects of exendin-4 treatment on cellular and metabolic responses of beta-cells. Diabetic NOD mice treated with anti-CD3 mAb and exendin-4 had a higher rate of remission (44%) than mice treated with anti-CD3 mAb alone (37%) or exendin-4 (0%) or insulin or IgG alone (0%) (P < 0.01). The effect of exendin-4 on reversal of diabetes after anti-CD3 mAb was greatest in mice with a glucose level of less than 350 mg/dl at diagnosis (63 vs. 39%, P < 0.05). Exendin-4 did not affect beta-cell area, replication, or apoptosis or reduce the frequency of diabetogenic or regulatory T cells or modulate the antigenicity of islet cells. Reversal of T1DM with anti-CD3 mAb was associated with recovery of insulin in glucose transporter-2(+)/insulin(-) islet cells that were identified at diagnosis. Glucose tolerance and insulin responses improved in mice treated with combination therapy, and exendin-4 increased insulin content and insulin release from beta-cells. We conclude that treatment with glucagon-like peptide-1 receptor agonist enhances remission of T1DM in NOD mice treated with anti-CD3 mAb by enhancing the recovery of the residual islets. This combinatorial approach may be useful in treatment of patients with new-onset T1DM.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Complexo CD3/imunologia , Diabetes Mellitus Tipo 1/terapia , Células Secretoras de Insulina/efeitos dos fármacos , Peptídeos/farmacologia , Peptídeos/uso terapêutico , Peçonhas/farmacologia , Peçonhas/uso terapêutico , Animais , Células Cultivadas , Terapia Combinada , Diabetes Mellitus Tipo 1/patologia , Exenatida , Feminino , Receptor do Peptídeo Semelhante ao Glucagon 1 , Imunoterapia , Células Secretoras de Insulina/patologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Receptores de Glucagon/agonistas , Indução de Remissão
19.
Nature ; 449(7159): 238-42, 2007 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-17728714

RESUMO

Stem cells are proposed to segregate chromosomes asymmetrically during self-renewing divisions so that older ('immortal') DNA strands are retained in daughter stem cells whereas newly synthesized strands segregate to differentiating cells. Stem cells are also proposed to retain DNA labels, such as 5-bromo-2-deoxyuridine (BrdU), either because they segregate chromosomes asymmetrically or because they divide slowly. However, the purity of stem cells among BrdU-label-retaining cells has not been documented in any tissue, and the 'immortal strand hypothesis' has not been tested in a system with definitive stem cell markers. Here we tested these hypotheses in haematopoietic stem cells (HSCs), which can be highly purified using well characterized markers. We administered BrdU to newborn mice, mice treated with cyclophosphamide and granulocyte colony-stimulating factor, and normal adult mice for 4 to 10 days, followed by 70 days without BrdU. In each case, less than 6% of HSCs retained BrdU and less than 0.5% of all BrdU-retaining haematopoietic cells were HSCs, revealing that BrdU has poor specificity and poor sensitivity as an HSC marker. Sequential administration of 5-chloro-2-deoxyuridine and 5-iodo-2-deoxyuridine indicated that all HSCs segregate their chromosomes randomly. Division of individual HSCs in culture revealed no asymmetric segregation of the label. Thus, HSCs cannot be identified on the basis of BrdU-label retention and do not retain older DNA strands during division, indicating that these are not general properties of stem cells.


Assuntos
Bromodesoxiuridina/metabolismo , Segregação de Cromossomos , Células-Tronco Hematopoéticas/citologia , Envelhecimento , Animais , Animais Recém-Nascidos , Células da Medula Óssea/metabolismo , Bromodesoxiuridina/farmacologia , Células Cultivadas , Segregação de Cromossomos/efeitos dos fármacos , Ciclofosfamida/farmacologia , Fator Estimulador de Colônias de Granulócitos/farmacologia , Células-Tronco Hematopoéticas/metabolismo , Camundongos , Processos Estocásticos , Fatores de Tempo
20.
Mol Cell Biol ; 27(18): 6484-96, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17636024

RESUMO

The pleckstrin homology domain-interacting protein (PHIP) was originally identified as a 902-amino-acid (aa) protein that regulates insulin receptor-stimulated GLUT4 translocation in skeletal-muscle cells. Immunoblotting and immunohistological analyses of pancreatic beta-cells reveal prominent expression of a 206-kDa PHIP isoform restricted to the nucleus. Herein, we report the cloning of this larger, 1,821-aa isoform of PHIP (PHIP1), which represents a novel WD40 repeat-containing protein. We demonstrate that PHIP1 overexpression stimulates insulin-like growth factor 1-dependent and -independent proliferation of beta-cells, an event which correlates with transcriptional upregulation of the cyclin D2 promoter and the accumulation of cyclin D2 protein. RNA interference knockdown of PHIP1 in INS-1 cells abrogates insulin receptor substrate 2 (IRS2)-mediated DNA synthesis, providing for a specific role for PHIP1 in the enhancement of IRS2-dependent signaling responses leading to beta-cell growth. Finally, we provide evidence that PHIP1 overexpression blocks free fatty acid-induced apoptosis in INS-1 cells, which is accompanied by marked activation of phosphoprotein kinase B (PKB)/AKT and the concomitant inhibition of caspase-9 and caspase-3 cleavage. Our finding that the restorative effect of PHIP1 on beta-cell lipotoxicity can be attenuated by the overexpression of dominant-negative PKB suggests a key role for PKB in PHIP1-mediated cytoprotection. Taken together, these findings provide strong support for PHIP1 as a novel positive regulator of beta-cell function. We suggest that PHIP1 may be involved in the induction of long-term gene expression programs to promote beta-cell mitogenesis and survival.


Assuntos
Células Secretoras de Insulina/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Sequências Repetitivas de Aminoácidos , Adenoviridae/genética , Sequência de Aminoácidos , Animais , Núcleo Celular/metabolismo , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Clonagem Molecular , DNA Complementar , Técnica Indireta de Fluorescência para Anticorpo , Corantes Fluorescentes , Regulação da Expressão Gênica , Genes Reporter , Indóis , Proteínas Substratos do Receptor de Insulina , Fator de Crescimento Insulin-Like I/metabolismo , Células Secretoras de Insulina/citologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Luciferases/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/genética , Fosfoproteínas/metabolismo , Isoformas de Proteínas , Interferência de RNA , Homologia de Sequência de Aminoácidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA