Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Cell Mol Gastroenterol Hepatol ; 14(6): 1269-1294, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35970323

RESUMO

BACKGROUND & AIMS: Nonalcoholic fatty liver disease (NAFLD) is a major health problem with complex pathogenesis. Although sex differences in NAFLD pathogenesis have been reported, the mechanisms underlying such differences remain understudied. Interleukin (IL)22 is a pleiotropic cytokine with both protective and/or pathogenic effects during liver injury. IL22 was shown to be hepatoprotective in NAFLD-related liver injury. However, these studies relied primarily on exogenous administration of IL22 and did not examine the sex-dependent effect of IL22. Here, we sought to characterize the role of endogenous IL22-receptor signaling during NAFLD-induced liver injury in males and females. METHODS: We used immunofluorescence, flow cytometry, histopathologic assessment, and gene expression analysis to examine IL22 production and characterize the intrahepatic immune landscape in human subjects with NAFLD (n = 20; 11 men and 9 women) and in an in vivo Western high-fat diet-induced NAFLD model in IL22RA knock out mice and their wild-type littermates. RESULTS: Examination of publicly available data sets from 2 cohorts with NAFLD showed increased hepatic IL22 gene expression in females compared with males. Furthermore, our immunofluorescence analysis of liver sections from NAFLD subjects (n = 20) showed increased infiltration of IL22-producing cells in females. Similarly, IL22-producing cells were increased in wild-type female mice with NAFLD and the hepatic IL22/IL22 binding protein messenger RNA ratio correlated with expression of anti-apoptosis genes. The lack of endogenous IL22-receptor signaling (IL22RA knockout) led to exacerbated liver damage, inflammation, apoptosis, and liver fibrosis in female, but not male, mice with NAFLD. CONCLUSIONS: Our data suggest a sex-dependent hepatoprotective antiapoptotic effect of IL22-receptor signaling during NAFLD-related liver injury in females.


Assuntos
Hepatopatia Gordurosa não Alcoólica , Feminino , Humanos , Masculino , Camundongos , Animais , Receptores de Interleucina/genética , Transdução de Sinais , Cirrose Hepática , Camundongos Knockout
2.
Diabetes ; 70(12): 2850-2859, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34610983

RESUMO

Controllable genetic manipulation is an indispensable tool in research, greatly advancing our understanding of cell biology and physiology. However in ß-cells, transgene silencing, low inducibility, ectopic expression, and off-targets effects are persistent challenges. In this study, we investigated whether an inducible Tetracycline (Tet)-Off system with ß-cell-specific mouse insulin promoter (MIP)-itTA-driven expression of tetracycline operon (TetO)-CreJaw/J could circumvent previous issues of specificity and efficacy. Following assessment of tissue-specific gene recombination, ß-cell architecture, in vitro and in vivo glucose-stimulated insulin secretion, and whole-body glucose homeostasis, we discovered that expression of any tetracycline-controlled transactivator (e.g., improved itTA, reverse rtTA, or tTA) in ß-cells significantly reduced Insulin gene expression and decreased insulin content. This translated into lower pancreatic insulin levels and reduced insulin secretion in mice carrying any tTA transgene, independent of Cre recombinase expression or doxycycline exposure. Our study echoes ongoing challenges faced by fundamental researchers working with ß-cells and highlights the need for consistent and comprehensive controls when using the tetracycline-controlled transactivator systems (Tet-On or Tet-Off) for genome editing.


Assuntos
Células Secretoras de Insulina/metabolismo , Insulina/genética , Insulina/metabolismo , Animais , Células Cultivadas , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Integrases/genética , Integrases/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Regiões Promotoras Genéticas/efeitos dos fármacos , Regiões Promotoras Genéticas/genética , Tetraciclina/farmacologia , Transativadores/efeitos dos fármacos , Transativadores/genética , Transgenes/efeitos dos fármacos
3.
Diabetes ; 70(2): 492-503, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33277337

RESUMO

FURIN is a proprotein convertase (PC) responsible for proteolytic activation of a wide array of precursor proteins within the secretory pathway. It maps to the PRC1 locus, a type 2 diabetes susceptibility locus, but its specific role in pancreatic ß-cells is largely unknown. The aim of this study was to determine the role of FURIN in glucose homeostasis. We show that FURIN is highly expressed in human islets, whereas PCs that potentially could provide redundancy are expressed at considerably lower levels. ß-cell-specific Furin knockout (ßFurKO) mice are glucose intolerant as a result of smaller islets with lower insulin content and abnormal dense-core secretory granule morphology. mRNA expression analysis and differential proteomics on ßFurKO islets revealed activation of activating transcription factor 4 (ATF4), which was mediated by mammalian target of rapamycin C1 (mTORC1). ßFurKO cells show impaired cleavage or shedding of vacuolar-type ATPase (V-ATPase) subunits Ac45 and prorenin receptor, respectively, and impaired lysosomal acidification. Blocking V-ATPase pharmacologically in ß-cells increased mTORC1 activity, suggesting involvement of the V-ATPase proton pump in the phenotype. Taken together, these results suggest a model of mTORC1-ATF4 hyperactivation and impaired lysosomal acidification in ß-cells lacking Furin, causing ß-cell dysfunction.


Assuntos
Fator 4 Ativador da Transcrição/metabolismo , Furina/metabolismo , Células Secretoras de Insulina/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Animais , Diabetes Mellitus Tipo 2/metabolismo , Furina/genética , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Transdução de Sinais/fisiologia
4.
Mol Metab ; 34: 72-84, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32180561

RESUMO

OBJECTIVE: The liver is regularly exposed to changing metabolic and inflammatory environments. It must sense and adapt to metabolic need while balancing resources required to protect itself from insult. Peroxisome proliferator activated receptor gamma coactivator-1 alpha (PGC-1α) is a transcriptional coactivator expressed as multiple, alternatively spliced variants transcribed from different promoters that coordinate metabolic adaptation and protect against inflammation. It is not known how PGC-1α integrates extracellular signals to balance metabolic and anti-inflammatory outcomes. METHODS: Primary mouse hepatocytes were used to evaluate the role(s) of different PGC-1α proteins in regulating hepatic metabolism and inflammatory signaling downstream of tumor necrosis factor alpha (TNFα). Gene expression and signaling analysis were combined with biochemical measurement of apoptosis using gain- and loss-of-function in vitro and in vivo. RESULTS: Hepatocytes expressed multiple isoforms of PGC-1α, including PGC-1α4, which microarray analysis showed had common and isoform-specific functions linked to metabolism and inflammation compared with canonical PGC-1α1. Whereas PGC-1α1 primarily impacted gene programs of nutrient metabolism and mitochondrial biology, TNFα signaling showed several pathways related to innate immunity and cell death downstream of PGC-1α4. Gain- and loss-of-function models illustrated that PGC-1α4 uniquely enhanced expression of anti-apoptotic gene programs and attenuated hepatocyte apoptosis in response to TNFα or lipopolysaccharide (LPS). This was in contrast to PGC-1α1, which decreased the expression of a wide inflammatory gene network but did not prevent hepatocyte death in response to cytokines. CONCLUSIONS: PGC-1α variants have distinct, yet complementary roles in hepatic responses to metabolism and inflammation, and we identify PGC-1α4 as an important mitigator of apoptosis.


Assuntos
Apoptose , Hepatócitos/metabolismo , Inflamação/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Animais , Linhagem Celular , Feminino , Hepatócitos/patologia , Inflamação/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/deficiência , Isoformas de Proteínas/deficiência , Isoformas de Proteínas/metabolismo
5.
J Biol Chem ; 293(32): 12516-12524, 2018 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-29941454

RESUMO

One feature of diabetes is the failure of pancreatic ß cells to produce insulin, but the molecular mechanisms leading to this failure remain unclear. Increasing evidence supports a role for protein kinase R-like endoplasmic reticulum kinase (PERK) in the development and function of healthy pancreatic ß cells. Previously, our group identified the adaptor protein Nck1 as a negative regulator of PERK. Indeed, we demonstrated that Nck1, by directly binding PERK autophosphorylated on Tyr561, limits PERK activation and signaling. Accordingly, we found that stable depletion of Nck1 in ß cells promotes PERK activation and signaling, increases insulin biosynthesis, and improves cell viability in response to diabetes-related stresses. Herein, we explored the therapeutic potential of abrogating the interaction between Nck and PERK to improve ß-cell function and survival. To do so, we designed and used a peptide containing the minimal PERK sequence involved in binding Nck1 conjugated to the cell-permeable protein transduction domain from the HIV protein TAT. In the current study, we confirm that the synthetic TAT-Tyr(P)561 phosphopeptide specifically binds the SH2 domain of Nck and prevents Nck interaction with PERK, thereby promoting basal PERK activation. Moreover, we report that treatment of ß cells with TAT-Tyr(P)561 inhibits glucolipotoxicity-induced apoptosis, whereas it enhances insulin production and secretion. Taken together, our results support the potential of sequestering Nck using a synthetic peptide to enhance basal PERK activation and create more robust ß cells.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Diabetes Mellitus/fisiopatologia , Células Secretoras de Insulina/efeitos dos fármacos , Insulina/biossíntese , Insulinoma/prevenção & controle , Proteínas Oncogênicas/metabolismo , Fragmentos de Peptídeos/farmacologia , Substâncias Protetoras/farmacologia , eIF-2 Quinase/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Apoptose , Células Cultivadas , Intolerância à Glucose/genética , Intolerância à Glucose/metabolismo , Intolerância à Glucose/prevenção & controle , Células Secretoras de Insulina/patologia , Insulinoma/genética , Insulinoma/metabolismo , Camundongos , Proteínas Oncogênicas/genética , Fosforilação , Transdução de Sinais , Estresse Fisiológico
6.
Exp Cell Res ; 360(1): 19-23, 2017 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-28351751

RESUMO

The pancreas is a centralized organ vital for whole body metabolic control. Recent advances in the field of metabolism have reinforced its importance for orchestrating endocrine hormone secretion in response to several nutrients including glucose, lipids and amino acids, in addition to hormones and inflammatory signals. Cell types within the pancreas, in particular the insulin-producing ß cells, control nutrient breakdown and energy production and are essential to maintain not only efficient hormone secretion, but also cell integrity, survival, and the ability to sense and adapt to changing metabolic environments. The present review highlights recent research advances on how glucolipotoxicity, mitochondrial dysfunction, and systemic inflammation affects pancreatic metabolism, and how new technologies and more advanced research models are improving our ability to study this organ system. Taken together, careful characterization and understanding of the importance of nutrient metabolism within this important, yet complex organ, will help us to better understand pathologies intimately associated with the pancreas and possibly discover new and more effective therapeutic strategies.


Assuntos
Glucose/metabolismo , Homeostase/fisiologia , Pâncreas/metabolismo , Animais , Humanos
7.
Endocrinology ; 157(9): 3388-90, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27580804

RESUMO

Aging affects every species and tissue but not in equal ways. Human pancreatic ß-cells lose their ability to replicate, regenerate, and secrete insulin as one gets older. This natural process increases risk of developing diabetes as you age and is a concern for donor islets and stem cells obtained from older subjects destined for transplantation or emerging regenerative therapies. Using fluidic sorting and RNA sequencing on single cells, Xin et al describe a transcriptional signature of mouse ß-cell aging between adulthood and a very old age. Amazingly, expression levels of more than 99% of genes do not change over time, despite the long lifespan of this specialized tissue. They identify a novel set of transcription factors that can explain decreases in cell survival and proliferation genes and potentially drive age-associated decline in regenerative capacity. Yet somehow, mouse ß-cells maintain pathways regulating glucose metabolism and ß-cell function despite experiencing challenges commonly associated with old age, including increased weight and fat mass. The authors conclude that ß-cells of old mice are overall strikingly similar to young ß-cells, implying that mechanisms may exist to resist aging and maintain their 'youth'. These new discoveries have interesting implications for efforts to preserve or improve function of human ß-cells, providing potential clues toward prolonging the life and health of donor tissues or islets of people with diabetes.


Assuntos
Envelhecimento/imunologia , Linfócitos B/fisiologia , Animais , Humanos
8.
Mol Metab ; 4(11): 811-22, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26629405

RESUMO

OBJECTIVES: Peroxisome proliferator-activated receptor γ coactivator 1 (PPARGCA1, PGC-1) transcriptional coactivators control gene programs important for nutrient metabolism. Islets of type 2 diabetic subjects have reduced PGC-1α expression and this is associated with decreased insulin secretion, yet little is known about why this occurs or what role it plays in the development of diabetes. Our goal was to delineate the role and importance of PGC-1 proteins to ß-cell function and energy homeostasis. METHODS: We investigated how nutrient signals regulate coactivator expression in islets and the metabolic consequences of reduced PGC-1α and PGC-1ß in primary and cultured ß-cells. Mice with inducible ß-cell specific double knockout of Pgc-1α/Pgc-1ß (ßPgc-1 KO) were created to determine the physiological impact of reduced Pgc1 expression on glucose homeostasis. RESULTS: Pgc-1α and Pgc-1ß expression was increased in primary mouse and human islets by acute glucose and palmitate exposure. Surprisingly, PGC-1 proteins were dispensable for the maintenance of mitochondrial mass, gene expression, and oxygen consumption in response to glucose in adult ß-cells. However, islets and mice with an inducible, ß-cell-specific PGC-1 knockout had decreased insulin secretion due in large part to loss of the potentiating effect of fatty acids. Consistent with an essential role for PGC-1 in lipid metabolism, ß-cells with reduced PGC-1s accumulated acyl-glycerols and PGC-1s controlled expression of key enzymes in lipolysis and the glycerolipid/free fatty acid cycle. CONCLUSIONS: These data highlight the importance of PGC-1s in coupling ß-cell lipid metabolism to promote efficient insulin secretion.

9.
Diabetes ; 64(11): 3798-807, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26153246

RESUMO

There is growing concern over confounding artifacts associated with ß-cell-specific Cre-recombinase transgenic models, raising questions about their general usefulness in research. The inducible ß-cell-specific transgenic (MIP-CreERT(1Lphi)) mouse was designed to circumvent many of these issues, and we investigated whether this tool effectively addressed concerns of ectopic expression and disruption of glucose metabolism. Recombinase activity was absent from the central nervous system using a reporter line and high-resolution microscopy. Despite increased pancreatic insulin content, MIP-CreERT mice on a chow diet exhibited normal ambient glycemia, glucose tolerance and insulin sensitivity, and appropriate insulin secretion in response to glucose in vivo and in vitro. However, MIP-CreERT mice on different genetic backgrounds were protected from high-fat/ streptozotocin (STZ)-induced hyperglycemia that was accompanied by increased insulin content and islet density. Ectopic human growth hormone (hGH) was highly expressed in MIP-CreERT islets independent of tamoxifen administration. Circulating insulin levels remained similar to wild-type controls, whereas STZ-associated increases in α-cell number and serum glucagon were significantly blunted in MIP-CreERT(1Lphi) mice, possibly due to paracrine effects of hGH-induced serotonin expression. These studies reveal important new insight into the strengths and limitations of the MIP-CreERT mouse line for ß-cell research.


Assuntos
Diabetes Mellitus Experimental/metabolismo , Hormônio do Crescimento Humano/metabolismo , Hiperglicemia/metabolismo , Células Secretoras de Insulina/metabolismo , Fenótipo , Animais , Glicemia/metabolismo , Diabetes Mellitus Experimental/genética , Homeostase/fisiologia , Hormônio do Crescimento Humano/genética , Humanos , Hiperglicemia/genética , Insulina/sangue , Masculino , Camundongos , Camundongos Transgênicos
10.
Curr Opin Immunol ; 25(4): 428-35, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23751490

RESUMO

Many of the pathogens that cause human infectious diseases do not infect rodents or other mammalian species. Small animal models that allow studies of the pathogenesis of these agents and evaluation of drug efficacy are critical for identifying ways to prevent and treat human infectious diseases. Immunodeficient mice engrafted with functional human cells and tissues, termed 'humanized' mice, represent a critical pre-clinical bridge for in vivo studies of human pathogens. Recent advances in the development of humanized mice have allowed in vivo studies of multiple human infectious agents providing novel insights into their pathogenesis that was otherwise not possible.


Assuntos
Doenças Transmissíveis/imunologia , Animais , Doenças Transmissíveis/microbiologia , Doenças Transmissíveis/parasitologia , Modelos Animais de Doenças , Humanos , Camundongos , Pesquisa Translacional Biomédica
11.
Biochem Biophys Res Commun ; 413(2): 248-53, 2011 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-21884683

RESUMO

In Saccharomyces cerevisiae, the immunosuppressor rapamycin engenders the degradation of excessive RNA polymerase II leading to growth arrest but the regulation of this process is not known yet. Here, we show that this mechanism is dependent on the peptidyl prolyl cis/trans isomerase Rrd1. Strikingly this degradation is independent of RNA polymerase II polyubiquitylation and does not require the elongation factor Elc1. Our data reveal that there are at least two alternative pathways to degrade RNA polymerase II that depend on different type of stresses.


Assuntos
Imunossupressores/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Peptidilprolil Isomerase/metabolismo , RNA Polimerase II/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/efeitos dos fármacos , Sirolimo/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptidilprolil Isomerase/genética , Saccharomyces cerevisiae/enzimologia , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Ubiquitinação
12.
BMC Mol Biol ; 11: 92, 2010 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-21129186

RESUMO

BACKGROUND: In Saccharomyces cerevisiae, the immunosuppressant rapamycin engenders a profound modification in the transcriptional profile leading to growth arrest. Mutants devoid of Rrd1, a protein possessing in vitro peptidyl prolyl cis/trans isomerase activity, display striking resistance to the drug, although how Rrd1 activity is linked to the biological responses has not been elucidated. RESULTS: We now provide evidence that Rrd1 is associated with the chromatin and it interacts with RNA polymerase II. Circular dichroism revealed that Rrd1 mediates structural changes onto the C-terminal domain (CTD) of the large subunit of RNA polymerase II (Rpb1) in response to rapamycin, although this appears to be independent of the overall phosphorylation status of the CTD. In vitro experiments, showed that recombinant Rrd1 directly isomerizes purified GST-CTD and that it releases RNA polymerase II from the chromatin. Consistent with this, we demonstrated that Rrd1 is required to alter RNA polymerase II occupancy on rapamycin responsive genes. CONCLUSION: We propose as a mechanism, that upon rapamycin exposure Rrd1 isomerizes Rpb1 to promote its dissociation from the chromatin in order to modulate transcription.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Peptidilprolil Isomerase/metabolismo , RNA Polimerase II/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Sirolimo/farmacologia , Cromatina/metabolismo , Dicroísmo Circular , Isomerismo , Fosforilação , Estrutura Terciária de Proteína , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo , RNA Polimerase II/química , Saccharomyces cerevisiae/enzimologia , Saccharomyces cerevisiae/metabolismo
13.
J Biol Chem ; 283(45): 30632-41, 2008 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-18776186

RESUMO

Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) has diverse biological functions including its nuclear translocation in response to oxidative stress. We show that GAPDH physically associates with APE1, an essential enzyme involved in the repair of abasic sites in damaged DNA, as well as in the redox regulation of several transcription factors. This interaction allows GAPDH to convert the oxidized species of APE1 to the reduced form, thereby reactivating its endonuclease activity to cleave abasic sites. The GAPDH variants C152G and C156G retain the ability to interact with but are unable to reactivate APE1, implicating these cysteines in catalyzing the reduction of APE1. Interestingly, GAPDH-small interfering RNA knockdown sensitized the cells to methyl methane sulfonate and bleomycin, which generate lesions that are repaired by APE1, but showed normal sensitivity to 254-nm UV. Moreover, the GAPDH knockdown cells exhibited an increased level of spontaneous abasic sites in the genomic DNA as a result of diminished APE1 endonuclease activity. Thus, the nuclear translocation of GAPDH during oxidative stress constitutes a protective mechanism to safeguard the genome by preventing structural inactivation of APE1.


Assuntos
Núcleo Celular/enzimologia , Reparo do DNA/fisiologia , DNA Liase (Sítios Apurínicos ou Apirimidínicos)/metabolismo , Genoma Humano/fisiologia , Gliceraldeído-3-Fosfato Desidrogenases/metabolismo , Estresse Oxidativo/fisiologia , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Transporte Ativo do Núcleo Celular/efeitos da radiação , Antibióticos Antineoplásicos/farmacologia , Bleomicina/farmacologia , Linhagem Celular Tumoral , Cisteína/metabolismo , Reparo do DNA/efeitos dos fármacos , Reparo do DNA/efeitos da radiação , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Ativação Enzimática/efeitos da radiação , Gliceraldeído-3-Fosfato Desidrogenases/antagonistas & inibidores , Humanos , Metanossulfonato de Metila/farmacologia , Mutagênicos/farmacologia , Oxirredução/efeitos dos fármacos , Oxirredução/efeitos da radiação , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/efeitos da radiação , RNA Interferente Pequeno/farmacologia , Raios Ultravioleta
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA