Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Diabetologia ; 66(11): 2042-2061, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37537395

RESUMO

AIMS/HYPOTHESIS: Increased circulating levels of incompletely processed insulin (i.e. proinsulin) are observed clinically in type 1 and type 2 diabetes. Previous studies have suggested that Ca2+ signalling within beta cells regulates insulin processing and secretion; however, the mechanisms that link impaired Ca2+ signalling with defective insulin maturation remain incompletely understood. METHODS: We generated mice with beta cell-specific sarcoendoplasmic reticulum Ca2+ ATPase-2 (SERCA2) deletion (ßS2KO mice) and used an INS-1 cell line model of SERCA2 deficiency. Whole-body metabolic phenotyping, Ca2+ imaging, RNA-seq and protein processing assays were used to determine how loss of SERCA2 impacts beta cell function. To test key findings in human model systems, cadaveric islets were treated with diabetogenic stressors and prohormone convertase expression patterns were characterised. RESULTS: ßS2KO mice exhibited age-dependent glucose intolerance and increased plasma and pancreatic levels of proinsulin, while endoplasmic reticulum (ER) Ca2+ levels and glucose-stimulated Ca2+ synchronicity were reduced in ßS2KO islets. Islets isolated from ßS2KO mice and SERCA2-deficient INS-1 cells showed decreased expression of the active forms of the proinsulin processing enzymes PC1/3 and PC2. Additionally, immunofluorescence staining revealed mis-location and abnormal accumulation of proinsulin and proPC2 in the intermediate region between the ER and the Golgi (i.e. the ERGIC) and in the cis-Golgi in beta cells of ßS2KO mice. Treatment of islets from human donors without diabetes with high glucose and palmitate concentrations led to reduced expression of the active forms of the proinsulin processing enzymes, thus phenocopying the findings observed in ßS2KO islets and SERCA2-deficient INS-1 cells. Similar findings were observed in wild-type mouse islets treated with brefeldin A, a compound that perturbs ER-to-Golgi trafficking. CONCLUSIONS/INTERPRETATION: Taken together, these data highlight an important link between ER Ca2+ homeostasis and proinsulin processing in beta cells. Our findings suggest a model whereby chronic ER Ca2+ depletion due to SERCA2 deficiency impairs the spatial regulation of prohormone trafficking, processing and maturation within the secretory pathway. DATA AVAILABILITY: RNA-seq data have been deposited in the Gene Expression Omnibus (GEO; accession no.: GSE207498).


Assuntos
Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Ilhotas Pancreáticas , Camundongos , Humanos , Animais , Proinsulina/genética , Proinsulina/metabolismo , Células Secretoras de Insulina/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/genética , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Insulina/metabolismo , Glucose/metabolismo , Ilhotas Pancreáticas/metabolismo
2.
Eur J Pharmacol ; 920: 174843, 2022 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-35196519

RESUMO

Diabetes mellitus (DM) has recently become one of the major diseases that have received attention. Cordycepin (molecular formula: C10H13N5O3), is one of the major bioactive components of Cordyceps militaris, decreases blood glucose levels. In this study, the effect and mechanism of cordycepin in normal and oxidative-damaged INS-1 cells were explored by using cell and molecular biology methods. Results showed that cordycepin could enhance insulin synthesis and secretion. The mechanism is possibly related to the elevated ATP content induced membrane depolarisation and increased Ca2+ concentration. At the genetic level, cordycepin upregulated the mRNA level of insulin, pancreatic duodenal homeobox factor-1 (PDX-1) and glucose transporter 1 (GLUT1). At the protein level, cordycepin promoted the expression of PDX-1, GLUT1, serine threonine kinase (Akt) and phosphorylated Akt (P-Akt). These effects may also contribute to the enhancement of insulin synthesis and secretion. Further analysis revealed that cordycepin protected against H2O2-induced damage on INS-1 cells and improved their viability and insulin synthesis/secretion. This effect should be attributed to the reduced intracellular reactive oxygen species (ROS), enhanced mitochondrial membrane potential (MMP), increased activity of superoxide dismutase (SOD) and upregulated genetic and protein expression of catalase (CAT), PDX-1, GLUT1 and P-Akt. In conclusion, cordycepin promotes insulin synthesis and secretion in normal islet ß cells and improves this function in oxidative-damaged islet ß cells. Given that islet ß cells are vulnerable to oxidative stress, the improving effect of cordycepin on the antioxidant capacity and insulin synthesis/secretion of INS-1 cells may be an important mechanism for its hypoglycaemic effect.


Assuntos
Células Secretoras de Insulina , Insulina , Desoxiadenosinas , Glucose/metabolismo , Peróxido de Hidrogênio/farmacologia , Insulina/metabolismo , Estresse Oxidativo
3.
Acta Pharm Sin B ; 11(9): 2719-2725, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34589392

RESUMO

Insulin derivatives such as insulin detemir and insulin degludec are U.S. Food and Drug Administration (FDA)-approved long-acting insulin currently used by millions of people with diabetes. These derivatives are modified in C-terminal B29 lysine to retain insulin bioactivity. New and efficient methods for facile synthesis of insulin derivatives may lead to new discovery of therapeutic insulin. Herein, we report a new method using sortase A (SrtA)-mediated ligation for the synthesis of insulin derivatives with high efficiency and functional group tolerance in the C-terminal B chain. This new insulin molecule (Ins-SA) with an SrtA-recognizing motif can be conjugated to diverse groups with N-terminal oligoglycines to generate new insulin derivatives. We further demonstrated that a new insulin derivative synthesized by this SrtA-mediated ligation shows strong cellular and in vivo bioactivity. This enzymatic method can therefore be used for future insulin design and development.

4.
EXCLI J ; 19: 1227-1245, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33088259

RESUMO

Nitric oxide (NO) is a gas that serves as a ubiquitous signaling molecule participating in physiological activities of various organ systems. Nitric oxide is produced in the endocrine pancreas and contributes to synthesis and secretion of insulin. The potential role of NO in insulin secretion is disputable - both stimulatory and inhibitory effects have been reported. Available data indicate that effects of NO critically depend on its concentration. Different isoforms of NO synthase (NOS) control this and have the potential to decrease or increase insulin secretion. In this review, the role of NO in insulin secretion as well as the possible reasons for discrepant findings are discussed. A better understanding of the role of NO system in the regulation of insulin secretion may facilitate the development of new therapeutic strategies in the management of diabetes.

5.
J Biol Chem ; 295(41): 14164-14177, 2020 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-32788214

RESUMO

Success or failure of pancreatic beta cell adaptation to ER stress is a determinant of diabetes susceptibility. The ATF6 and IRE1/XBP1 pathways are separate ER stress-response effectors important to beta cell health and function. ATF6α. and XBP1 direct overlapping transcriptional responses in some cell types. However, the signaling dynamics and interdependence of ATF6α and XBP1 in pancreatic beta cells have not been explored. To assess pathway-specific signal onset, we performed timed exposures of primary mouse islet cells to ER stressors and measured the early transcriptional response. Comparing the time course of induction of ATF6 and XBP1 targets suggested that the two pathways have similar response dynamics. The role of ATF6α in target induction was assessed by acute knockdown using islet cells from Atf6α flox/flox mice transduced with adenovirus expressing Cre recombinase. Surprisingly, given the mild impact of chronic deletion in mice, acute ATF6α knockdown markedly reduced ATF6-pathway target gene expression under both basal and stressed conditions. Intriguingly, although ATF6α knockdown did not alter Xbp1 splicing dynamics or intensity, it did reduce induction of XBP1 targets. Inhibition of Xbp1 splicing did not decrease induction of ATF6α targets. Taken together, these data suggest that the XBP1 and ATF6 pathways are simultaneously activated in islet cells in response to acute stress and that ATF6α is required for full activation of XBP1 targets, but XBP1 is not required for activation of ATF6α targets. These observations improve understanding of the ER stress transcriptional response in pancreatic islets.


Assuntos
Fator 6 Ativador da Transcrição/metabolismo , Estresse do Retículo Endoplasmático , Células Secretoras de Insulina/metabolismo , Transdução de Sinais , Transcrição Gênica , Proteína 1 de Ligação a X-Box/metabolismo , Fator 6 Ativador da Transcrição/genética , Animais , Técnicas de Silenciamento de Genes , Camundongos , Camundongos Transgênicos
6.
J Biol Chem ; 294(35): 13040-13050, 2019 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-31300553

RESUMO

An early hallmark of type 2 diabetes is a failure of proinsulin-to-insulin processing in pancreatic ß-cells, resulting in hyperproinsulinemia. Proinsulin processing is quite sensitive to nutrient flux, and ß-cell-specific deletion of the nutrient-sensing protein modifier OGlcNAc transferase (ßOGTKO) causes ß-cell failure and diabetes, including early development of hyperproinsulinemia. The mechanisms underlying this latter defect are unknown. Here, using several approaches, including site-directed mutagenesis, Click O-GlcNAc labeling, immunoblotting, and immunofluorescence and EM imaging, we provide the first evidence for a relationship between the O-GlcNAcylation of eukaryotic translation initiation factor 4γ1 (eIF4G1) and carboxypeptidase E (CPE)-dependent proinsulin processing in ßOGTKO mice. We first established that ßOGTKO hyperproinsulinemia is independent of age, sex, glucose levels, and endoplasmic reticulum-CCAAT enhancer-binding protein homologous protein (CHOP)-mediated stress status. Of note, OGT loss was associated with a reduction in ß-cell-resident CPE, and genetic reconstitution of CPE in ßOGTKO islets rescued the dysfunctional proinsulin-to-insulin ratio. We show that although CPE is not directly OGlcNAc modified in islets, overexpression of the suspected OGT target eIF4G1, previously shown to regulate CPE translation in ß-cells, increases islet CPE levels, and fully reverses ßOGTKO islet-induced hyperproinsulinemia. Furthermore, our results reveal that OGT O-GlcNAc-modifies eIF4G1 at Ser-61 and that this modification is critical for eIF4G1 protein stability. Together, these results indicate a direct link between nutrient-sensitive OGT and insulin processing, underscoring the importance of post-translational O-GlcNAc modification in general cell physiology.


Assuntos
Carboxipeptidase H/metabolismo , Diabetes Mellitus/metabolismo , Fator de Iniciação Eucariótico 4G/metabolismo , Células Secretoras de Insulina/metabolismo , N-Acetilglucosaminiltransferases/metabolismo , Animais , Modelos Animais de Doenças , Camundongos , Camundongos Knockout , N-Acetilglucosaminiltransferases/deficiência
7.
Diabetes Obes Metab ; 20 Suppl 2: 20-27, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30230180

RESUMO

Efficient and accurate protein translation is essential to producing insulin in pancreatic ß-cells. Transfer RNA (tRNA) is known as the key component of the protein translational machinery. Interestingly, tRNA contains a wide variety of chemical modifications, which are posttranscriptionally catalysed by tRNA modifying enzymes. Recent advances in genome-sequencing technology have unveiled a number of genetic variations that are associated with the development of type 2 diabetes (T2D). Some of these mutations are located in the genes of tRNA modifying enzymes. Using cellular and animal models, it has been showed that dysregulation of tRNA modification impairs protein translation in pancreatic ß-cells and leads to aberrant insulin production. In this review, we discuss the recent findings in the molecular functions of tRNA modifications and their involvement in the development of T2D.


Assuntos
Células Secretoras de Insulina/fisiologia , RNA de Transferência/fisiologia , Processamento Alternativo/genética , Animais , Glicemia/metabolismo , Quinase 5 Dependente de Ciclina/genética , Quinase 5 Dependente de Ciclina/fisiologia , Metilação de DNA/fisiologia , Diabetes Mellitus Tipo 2/genética , Humanos , Insulina/biossíntese , Secreção de Insulina/fisiologia , Camundongos Knockout , Oxirredução , Fenótipo , Polimorfismo de Nucleotídeo Único/fisiologia , Transdução de Sinais/fisiologia , tRNA Metiltransferases/deficiência , tRNA Metiltransferases/genética , tRNA Metiltransferases/fisiologia
8.
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
9.
EBioMedicine ; 30: 295-302, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29631902

RESUMO

Transcription factor 7-like 2 (TCF7L2) is the main susceptibility gene for type 2 diabetes, primarily through impairing the insulin secretion by pancreatic ß cells. However, the exact in vivo mechanisms remain poorly understood. We performed a family study and determined if the T risk allele of the rs7903146 in the TCF7L2 gene increases the risk of type 2 diabetes based on real-time stable isotope measurements of insulin synthesis during an Oral Glucose Tolerance Test. In addition, we performed oral minimal model (OMM) analyses to assess insulin sensitivity and ß cell function indices. Compared to unaffected relatives, individuals with type 2 diabetes had lower OMM indices and a higher level of insulin synthesis. We found a T allele-dosage effect on insulin synthesis and on glucose tolerance status, therefore insulin synthesis was higher among T-allele carriers with type 2 diabetes than in wild-type individuals. These results suggest that hyperinsulinemia is not only an adaptation to insulin resistance, but also a direct cause of type 2 diabetes.


Assuntos
Diabetes Mellitus Tipo 2/genética , Insulina/biossíntese , Proteína 2 Semelhante ao Fator 7 de Transcrição/genética , Adulto , Alelos , Peptídeo C/metabolismo , Feminino , Glucose/metabolismo , Teste de Tolerância a Glucose , Humanos , Masculino , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único/genética , Análise de Regressão
10.
Diabetologia ; 59(8): 1609-15, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27207082

RESUMO

Recent results suggest that insulin is synthesised by a subpopulation of neurons in the cerebral cortex and neural progenitor cells of the hippocampus. Supplementing the slow supply of insulin to the brain by pancreatic beta cells, the insulin locally released by neurons provides a rapid means of regulating local microcircuits, effectively modulating synaptic transmission and on-demand energy homeostasis of neural networks. Modulation of insulin production by brain neurons via glucagon-like peptide 1 (GLP-1) agonists might be useful in counteracting diabetes, obesity and neurodegenerative diseases. Replacement of lost pancreatic beta cells by autologous transplantation of insulin-producing neural progenitor cells could be a viable therapy for diabetes.


Assuntos
Córtex Cerebral/metabolismo , Insulina/metabolismo , Animais , Peptídeo 1 Semelhante ao Glucagon/agonistas , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Humanos
11.
J Mol Endocrinol ; 56(2): R33-54, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26576641

RESUMO

The inability of pancreatic ß-cells to make sufficient insulin to control blood sugar is a central feature of the aetiology of most forms of diabetes. In this review we focus on the deleterious effects of oxidative stress and endoplasmic reticulum (ER) stress on ß-cell insulin biosynthesis and secretion and on inflammatory signalling and apoptosis with a particular emphasis on type 2 diabetes (T2D). We argue that oxidative stress and ER stress are closely entwined phenomena fundamentally involved in ß-cell dysfunction by direct effects on insulin biosynthesis and due to consequences of the ER stress-induced unfolded protein response. We summarise evidence that, although these phenomenon can be driven by intrinsic ß-cell defects in rare forms of diabetes, in T2D ß-cell stress is driven by a range of local environmental factors including increased drivers of insulin biosynthesis, glucolipotoxicity and inflammatory cytokines. We describe our recent findings that a range of inflammatory cytokines contribute to ß-cell stress in diabetes and our discovery that interleukin 22 protects ß-cells from oxidative stress regardless of the environmental triggers and can correct much of diabetes pathophysiology in animal models. Finally we summarise evidence that ß-cell dysfunction is reversible in T2D and discuss therapeutic opportunities for relieving oxidative and ER stress and restoring glycaemic control.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Estresse do Retículo Endoplasmático , Células Secretoras de Insulina/metabolismo , Estresse Oxidativo , Animais , Apoptose , Citocinas/fisiologia , Diabetes Mellitus Tipo 2/patologia , Diabetes Mellitus Tipo 2/terapia , Humanos , Insulina/biossíntese , Insulina/metabolismo , Secreção de Insulina , Resposta a Proteínas não Dobradas
12.
J Mol Endocrinol ; 54(3): 315-24, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25917831

RESUMO

The relationship between protein arginine methyltransferases (PRMTs) and insulin synthesis in ß cells is not yet well understood. In the present study, we showed that PRMT4 expression was increased in INS-1 and HIT-T15 pancreatic ß cells under high-glucose conditions. In addition, asymmetric dimethylation of Arg17 in histone H3 was significantly increased in both cell lines in the presence of glucose. The inhibition or knockdown of PRMT4 suppressed glucose-induced insulin gene expression in INS-1 cells by 81.6 and 79% respectively. Additionally, the overexpression of mutant PRMT4 also significantly repressed insulin gene expression. Consistently, insulin secretion induced in response to high levels of glucose was decreased by both PRMT4 inhibition and knockdown. Moreover, the inhibition of PRMT4 blocked high-glucose-induced insulin gene expression and insulin secretion in primary pancreatic islets. These results indicate that PRMT4 might be a key regulator of high-glucose-induced insulin secretion from pancreatic ß cells via H3R17 methylation.


Assuntos
Histonas/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Proteína-Arginina N-Metiltransferases/metabolismo , Animais , Linhagem Celular Tumoral , Cricetinae , Indução Enzimática , Glucose/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/enzimologia , Masculino , Metilação , Camundongos , Camundongos Endogâmicos C57BL , Processamento de Proteína Pós-Traducional , Proteína-Arginina N-Metiltransferases/genética , Ratos
13.
J Biol Chem ; 290(12): 7647-57, 2015 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-25645923

RESUMO

The murine Mafa transcription factor is a key regulator of postnatal islet ß-cell activity, affecting insulin transcription, insulin secretion, and ß-cell mass. Human MAFA expression is also markedly decreased in islet ß-cells of type 2 diabetes mellitus (T2DM) patients. Moreover, levels are profoundly reduced in db/db islet ß-cells, a mouse model of T2DM. To examine the significance of this key islet ß-cell-enriched protein to glycemic control under diabetic conditions, we generated transgenic mice that conditionally and specifically produced Mafa in db/db islet ß-cells. Sustained expression of Mafa resulted in significantly lower plasma glucose levels, higher plasma insulin, and augmented islet ß-cell mass. In addition, there was increased expression of insulin, Slc2a2, and newly identified Mafa-regulated genes involved in reducing ß-cell stress, like Gsta1 and Gckr. Importantly, the levels of human GSTA1 were also compromised in T2DM islets. Collectively, these results illustrate how consequential the reduction in Mafa activity is to islet ß-cell function under pathophysiological conditions.


Assuntos
Glicemia/metabolismo , Diabetes Mellitus Experimental/metabolismo , Ilhotas Pancreáticas/metabolismo , Fatores de Transcrição Maf Maior/metabolismo , Animais , Sequência de Bases , Primers do DNA , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase em Tempo Real
14.
J Biol Chem ; 290(6): 3647-53, 2015 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-25533471

RESUMO

We used cre-lox technology to test whether the inducible expression of Cre minimize the deleterious effect of the enzyme on beta cell function. We studied mice in which Cre is linked to a modified estrogen receptor (ER), and its expression is controlled by the rat insulin promoter (RIP). Following the injection of tamoxifen (TM), CreER- migrates to the nucleus and promotes the appearance of a reporter protein, enhanced yellow fluorescent protein (EYFP), in cells. Immunocytochemical analysis indicated that 46.6 ± 2.1% insulin cells of adult RIPCreER- EYFP expressed EYFP. RIPCreER-EYFP (+TM) mice were normoglycemic throughout the study, and their glucose tolerance test results were similar to control CD-1 mice. However, an extended exposure to reagents that stimulate insulin synthesis was detrimental to the survival of IN+EYFP+cells. The administration of an inhibitor of the enzyme dipeptidyl-peptidase (DPP4i), which prevents the cleavage of glucagon-like peptide (GLP-1), to adult RIPCreER-EYFP mice lead to a decrease in the percentage of IN+EYFP+ to 17.5 ± 1.73 and a significant increase in apoptotic cells in islets. Similarly, a 2-week administration of the GLP-1 analog exendin 4 (ex-4) induced an almost complete ablation of IN+ expressing a different reporter protein and a significant decrease in the beta cell mass and rate of beta cell proliferation. Since normal beta cells do not die when induced to increase insulin synthesis, our observations indicate that insulin cells expressing an inducible RIPCre transgene are functionally deficient. Studies employing these mice should carefully consider the pitfalls of the Cre-Lox technique.


Assuntos
Células Secretoras de Insulina/metabolismo , Insulina/genética , Fenótipo , Regiões Promotoras Genéticas , Animais , Proliferação de Células , Dipeptidil Peptidase 4/metabolismo , Exenatida , Genes Reporter , Receptor do Peptídeo Semelhante ao Glucagon 1 , Intolerância à Glucose/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Hipoglicemiantes/farmacologia , Insulina/metabolismo , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/fisiologia , Integrases/genética , Integrases/metabolismo , Camundongos , Peptídeos/farmacologia , Inibidores de Proteases/farmacologia , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo , Receptores de Glucagon/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Moduladores Seletivos de Receptor Estrogênico/farmacologia , Tamoxifeno/farmacologia , Peçonhas/farmacologia
15.
R Soc Open Sci ; 2(12): 150306, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27019722

RESUMO

Insulin production is the central feature of functionally mature and differentiated pancreatic ß-cells. Reduced insulin transcription and dedifferentiation have been implicated in type 2 diabetes, making drugs that could reverse these processes potentially useful. We have previously established ratiometric live-cell imaging tools to identify factors that increase insulin promoter activity and promote ß-cell differentiation. Here, we present a single vector imaging tool with eGFP and mRFP, driven by the Pdx1 and Ins1 promoters, respectively, targeted to the nucleus to enhance identification of individual cells in a high-throughput manner. Using this new approach, we screened 1120 off-patent drugs for factors that regulate Ins1 and Pdx1 promoter activity in MIN6 ß-cells. We identified a number of compounds that positively modulate Ins1 promoter activity, including several drugs known to modulate ion channels. Carbamazepine was selected for extended follow-up, as our previous screen also identified this use-dependent sodium channel inhibitor as a positive modulator of ß-cell survival. Indeed, carbamazepine increased Ins1 and Ins2 mRNA in primary mouse islets at lower doses than were required to protect ß-cells. We validated the role of sodium channels in insulin production by examining Nav1.7 (Scn9a) knockout mice and remarkably islets from these animals had dramatically elevated insulin content relative to wild-type controls. Collectively, our experiments provide a starting point for additional studies aimed to identify drugs and molecular pathways that control insulin production and ß-cell differentiation status. In particular, our unbiased screen identified a novel role for a ß-cell sodium channel gene in insulin production.

16.
Am J Physiol Endocrinol Metab ; 307(9): E838-46, 2014 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-25205822

RESUMO

ß-Cells generated from large-scale sources can overcome current shortages in clinical islet cell grafts provided that they adequately respond to metabolic variations. Pancreatic (non)endocrine cells can develop from human embryonic stem (huES) cells following in vitro derivation to pancreatic endoderm (PE) that is subsequently implanted in immune-incompetent mice for further differentiation. Encapsulation of PE increases the proportion of endocrine cells in subcutaneous implants, with enrichment in ß-cells when they are placed in TheraCyte-macrodevices and predominantly α-cells when they are alginate-microencapsulated. At posttransplant (PT) weeks 20-30, macroencapsulated huES implants presented higher glucose-responsive plasma C-peptide levels and a lower proinsulin-over-C-peptide ratio than human islet cell implants under the kidney capsule. Their ex vivo analysis showed the presence of single-hormone-positive α- and ß-cells that exhibited rapid secretory responses to increasing and decreasing glucose concentrations, similar to isolated human islet cells. However, their insulin secretory amplitude was lower, which was attributed in part to a lower cellular hormone content; it was associated with a lower glucose-induced insulin biosynthesis, but not with lower glucagon-induced stimulation, which together is compatible with an immature functional state of the huES-derived ß-cells at PT weeks 20-30. These data support the therapeutic potential of macroencapsulated huES implants but indicate the need for further functional analysis. Their comparison with clinical-grade human islet cell grafts sets references for future development and clinical translation.


Assuntos
Células Imobilizadas/transplante , Diabetes Mellitus Tipo 1/cirurgia , Células-Tronco Embrionárias/transplante , Implantes Experimentais/efeitos adversos , Transplante das Ilhotas Pancreáticas/efeitos adversos , Transplante Heterólogo/efeitos adversos , Transplante Heterotópico/efeitos adversos , Animais , Peptídeo C/sangue , Peptídeo C/metabolismo , Diferenciação Celular , Linhagem Celular , Células Imobilizadas/citologia , Células Imobilizadas/metabolismo , Cruzamentos Genéticos , Diabetes Mellitus Tipo 1/sangue , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patologia , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Células Secretoras de Glucagon/citologia , Células Secretoras de Glucagon/metabolismo , Humanos , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Rim , Membranas , Camundongos Endogâmicos NOD , Camundongos SCID , Proinsulina/sangue , Proinsulina/metabolismo , Tela Subcutânea , Alicerces Teciduais/efeitos adversos
17.
J Biol Chem ; 289(39): 26904-26913, 2014 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-25122762

RESUMO

Oxidative folding of (pro)insulin is crucial for its assembly and biological function. This process takes place in the endoplasmic reticulum (ER) and is accomplished by protein disulfide isomerase and ER oxidoreductin 1ß, generating stoichiometric amounts of hydrogen peroxide (H2O2) as byproduct. During insulin resistance in the prediabetic state, increased insulin biosynthesis can overwhelm the ER antioxidative and folding capacity, causing an imbalance in the ER redox homeostasis and oxidative stress. Peroxiredoxin 4 (Prdx4), an ER-specific antioxidative peroxidase can utilize luminal H2O2 as driving force for reoxidizing protein disulfide isomerase family members, thus efficiently contributing to disulfide bond formation. Here, we examined the functional significance of Prdx4 on ß-cell function with emphasis on insulin content and secretion during stimulation with nutrient secretagogues. Overexpression of Prdx4 in glucose-responsive insulin-secreting INS-1E cells significantly metabolized luminal H2O2 and improved the glucose-induced insulin secretion, which was accompanied by the enhanced proinsulin mRNA transcription and insulin content. This ß-cell beneficial effect was also observed upon stimulation with the nutrient insulin secretagogue combination of leucine plus glutamine, indicating that the effect is not restricted to glucose. However, knockdown of Prdx4 had no impact on H2O2 metabolism or ß-cell function due to the fact that Prdx4 expression is negligibly low in pancreatic ß-cells. Moreover, we provide evidence that the constitutively low expression of Prdx4 is highly susceptible to hyperoxidation in the presence of high glucose. Overall, these data suggest an important role of Prdx4 in maintaining insulin levels and improving the ER folding capacity also under conditions of a high insulin requirement.


Assuntos
Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Glucose/farmacologia , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Peroxirredoxinas/biossíntese , Edulcorantes/farmacologia , Retículo Endoplasmático/genética , Retículo Endoplasmático/metabolismo , Regulação Enzimológica da Expressão Gênica/fisiologia , Técnicas de Silenciamento de Genes , Glucose/metabolismo , Células Hep G2 , Humanos , Peróxido de Hidrogênio/metabolismo , Insulina/genética , Secreção de Insulina , Células Secretoras de Insulina/citologia , Oxirredução/efeitos dos fármacos , Peroxirredoxinas/genética , Dobramento de Proteína/efeitos dos fármacos , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Edulcorantes/metabolismo , Transcrição Gênica/efeitos dos fármacos , Transcrição Gênica/fisiologia
18.
Cytokine ; 69(1): 39-46, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25022960

RESUMO

Macrophage migration inhibitory factor (MIF) is a molecule with plethora of functions such as regulation of immune response, hormone-like, enzymatic and chaperone-like activity. Further, MIF is a major participant in glucose homeostasis since it is an autocrine stimulator of insulin secretion. MIF absence in male knockout mice (MIF-KO) results in development of glucose intolerance, while sensitivity to insulin is fully preserved. Since our results confirm that beta cells from MIF-KO mice express, produce and secrete insulin similarly to beta cells of their wild type (WT) counterparts C57BL/6 mice, we hypothesize that MIF-KO-derived insulin is less active. Indeed, insulin from MIF-KO islets is unable to significantly induce glucose uptake into hepatocytes and to efficiently promote insulin-triggered Akt phosphorylation determined by immunoblot. However, MIF's tautomerase function is not crucial for insulin biosynthesis since MIF inhibitors had no impact on WT insulin activity. Importantly, MIF recognition by anti-MIF antibody (ELISA) after in vitro co-incubation with purified insulin was significantly lower suggesting that insulin covers MIF immunodominant epitope. In addition, MIF binds insulin within beta cell as confirmed by co-immunoprecipitation. WT and MIF-KO-derived insulin exhibited different cleavage patterns suggesting different protein conformations. Finally, pre-incubation of recombinant MIF with insulin promotes formation of insulin hexamers. These results imply that MIF probably enables proper insulin folding what results in insulin full activity. This newly discovered feature of the cytokine MIF could be potentially important for commercially produced insulin, for increasing its stability and/or bioavailability.


Assuntos
Intolerância à Glucose/genética , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Oxirredutases Intramoleculares/metabolismo , Fatores Inibidores da Migração de Macrófagos/metabolismo , Dobramento de Proteína/efeitos dos fármacos , Animais , Anticorpos/imunologia , Linhagem Celular , Epitopos/imunologia , Células Hep G2 , Hepatócitos/metabolismo , Humanos , Oxirredutases Intramoleculares/genética , Oxirredutases Intramoleculares/farmacologia , Fatores Inibidores da Migração de Macrófagos/genética , Fatores Inibidores da Migração de Macrófagos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação , Ligação Proteica , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Recombinantes/farmacologia
19.
J Biol Chem ; 289(23): 16290-302, 2014 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-24770419

RESUMO

Among the defects in the early events of insulin biosynthesis, proinsulin misfolding and endoplasmic reticulum (ER) stress have drawn increasing attention as causes of ß cell failure. However, no studies have yet addressed potential defects at the cytosolic entry point of preproinsulin into the secretory pathway. Here, we provide the first evidence that inefficient translocation of preproinsulin (caused by loss of a positive charge in the n region of its signal sequence) contributes to ß cell failure and diabetes. Specifically, we find that, after targeting to the ER membrane, preproinsulin signal peptide (SP) mutants associated with autosomal dominant late-onset diabetes fail to be fully translocated across the ER membrane. The newly synthesized, untranslocated preproinsulin remains strongly associated with the ER membrane, exposing its proinsulin moiety to the cytosol. Rather than accumulating in the ER and inducing ER stress, untranslocated preproinsulin accumulates in a juxtanuclear compartment distinct from the Golgi complex, induces the expression of heat shock protein 70 (HSP70), and promotes ß cell death. Restoring an N-terminal positive charge to the mutant preproinsulin SP significantly improves the translocation defect. These findings not only reveal a novel molecular pathogenesis of ß cell failure and diabetes but also provide the first evidence of the physiological and pathological significance of the SP n region positive charge of secretory proteins.


Assuntos
Diabetes Mellitus/metabolismo , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Precursores de Proteínas/metabolismo , Sequência de Aminoácidos , Animais , Diabetes Mellitus/patologia , Retículo Endoplasmático/metabolismo , Humanos , Insulina/química , Ilhotas Pancreáticas/patologia , Camundongos , Dados de Sequência Molecular , Precursores de Proteínas/química , Transporte Proteico , Ratos , Homologia de Sequência de Aminoácidos
20.
J Biol Chem ; 288(50): 35824-39, 2013 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-24187134

RESUMO

Glucokinase acts as a glucose sensor in pancreatic beta cells. Its posttranslational regulation is important but not yet fully understood. Therefore, a pancreatic islet yeast two-hybrid library was produced and searched for glucokinase-binding proteins. A protein sequence containing a full-length ubiquitin-like domain was identified to interact with glucokinase. Mammalian two-hybrid and fluorescence resonance energy transfer analyses confirmed the interaction between glucokinase and the ubiquitin-like domain in insulin-secreting MIN6 cells and revealed the highest binding affinity at low glucose. Overexpression of parkin, an ubiquitin E3 ligase exhibiting an ubiquitin-like domain with high homology to the identified, diminished insulin secretion in MIN6 cells but had only some effect on glucokinase activity. Overexpression of the elucidated ubiquitin-like domain or midnolin, containing exactly this ubiquitin-like domain, significantly reduced both intrinsic glucokinase activity and glucose-induced insulin secretion. Midnolin has been to date classified as a nucleolar protein regulating mouse development. However, we could not confirm localization of midnolin in nucleoli. Fluorescence microscopy analyses revealed localization of midnolin in nucleus and cytoplasm and co-localization with glucokinase in pancreatic beta cells. In addition we could show that midnolin gene expression in pancreatic islets is up-regulated at low glucose and that the midnolin protein is highly expressed in pancreatic beta cells and also in liver, muscle, and brain of the adult mouse and cell lines of human and rat origin. Thus, the results of our study suggest that midnolin plays a role in cellular signaling of adult tissues and regulates glucokinase enzyme activity in pancreatic beta cells.


Assuntos
Glucoquinase/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Ubiquitina/química , Sequência de Aminoácidos , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Glucose/farmacologia , Humanos , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Masculino , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Especificidade de Órgãos , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Transporte Proteico/efeitos dos fármacos , Ratos , Homologia de Sequência de Aminoácidos , Especificidade da Espécie , Ubiquitina-Proteína Ligases/química , Ubiquitina-Proteína Ligases/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA