Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 136
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 119(11): e2113074119, 2022 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-35254894

RESUMO

SignificanceWith obesity on the rise, there is a growing appreciation for intracellular lipid droplet (LD) regulation. Here, we show how saturated fatty acids (SFAs) reduce fat storage-inducing transmembrane protein 2 (FIT2)-facilitated, pancreatic ß cell LD biogenesis, which in turn induces ß cell dysfunction and death, leading to diabetes. This mechanism involves direct acylation of FIT2 cysteine residues, which then marks the FIT2 protein for endoplasmic reticulum (ER)-associated degradation. Loss of ß cell FIT2 and LDs reduces insulin secretion, increases intracellular ceramides, stimulates ER stress, and exacerbates diet-induced diabetes in mice. While palmitate and stearate degrade FIT2, unsaturated fatty acids such as palmitoleate and oleate do not, results of which extend to nutrition and diabetes.


Assuntos
Diabetes Mellitus/etiologia , Diabetes Mellitus/metabolismo , Células Secretoras de Insulina/metabolismo , Gotículas Lipídicas/metabolismo , Metabolismo dos Lipídeos , Proteínas de Membrana/genética , Animais , Linhagem Celular , Estresse do Retículo Endoplasmático , Ácidos Graxos/metabolismo , Glucose/metabolismo , Intolerância à Glucose , Proteínas de Membrana/metabolismo , Camundongos , Mutação , Palmitatos/metabolismo , Estearatos/metabolismo
2.
J Biol Chem ; 299(8): 104803, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37172723

RESUMO

Interleukin-1ß is one of the most potent inducers of beta cell inflammation in the lead-up to type 1 diabetes. We have previously reported that IL1ß-stimulated pancreatic islets from mice with genetic ablation of stress-induced pseudokinase TRB3(TRB3KO) show attenuated activation kinetics for the MAP3K MLK3 and JNK stress kinases. However, JNK signaling constitutes only a portion of the cytokine-induced inflammatory response. Here we report that TRB3KO islets also show a decrease in amplitude and duration of IL1ß-induced phosphorylation of TAK1 and IKK, kinases that drive the potent NF-κB proinflammatory signaling pathway. We observed that TRB3KO islets display decreased cytokine-induced beta cell death, preceded by a decrease in select downstream NF-κB targets, including iNOS/NOS2 (inducible nitric oxide synthase), a mediator of beta cell dysfunction and death. Thus, loss of TRB3 attenuates both pathways required for a cytokine-inducible, proapoptotic response in beta cells. In order to better understand the molecular basis of TRB3-enhanced, post-receptor IL1ß signaling, we interrogated the TRB3 interactome using coimmunoprecipitation followed by mass spectrometry to identify immunomodulatory protein Flightless homolog 1 (Fli1) as a novel, TRB3-interacting protein. We show that TRB3 binds and disrupts Fli1-dependent sequestration of MyD88, thereby increasing availability of this most proximal adaptor required for IL1ß receptor-dependent signaling. Fli1 sequesters MyD88 in a multiprotein complex resulting in a brake on the assembly of downstream signaling complexes. By interacting with Fli1, we propose that TRB3 lifts the brake on IL1ß signaling to augment the proinflammatory response in beta cells.


Assuntos
Proteínas de Ciclo Celular , Interleucina-1beta , Transdução de Sinais , Animais , Camundongos , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Citocinas/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , NF-kappa B/genética , NF-kappa B/metabolismo , Transdução de Sinais/genética , Inibidores Enzimáticos/farmacologia , Apoptose/efeitos dos fármacos , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/fisiologia , Ativação Transcricional/genética
3.
Rev Endocr Metab Disord ; 25(2): 259-278, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38048021

RESUMO

Undernutrition is still a recurring nutritional problem in low and middle-income countries. It is directly associated with the social and economic sphere, but it can also negatively impact the health of the population. In this sense, it is believed that undernourished individuals may be more susceptible to the development of non-communicable diseases, such as diabetes mellitus, throughout life. This hypothesis was postulated and confirmed until today by several studies that demonstrate that experimental models submitted to protein undernutrition present alterations in glycemic homeostasis linked, in part, to the reduction of insulin secretion. Therefore, understanding the changes that lead to a reduction in the secretion of this hormone is essential to prevent the development of diabetes in undernourished individuals. This narrative review aims to describe the main molecular changes already characterized in pancreatic ß cells that will contribute to the reduction of insulin secretion in protein undernutrition. So, it will provide new perspectives and targets for postulation and action of therapeutic strategies to improve glycemic homeostasis during this nutritional deficiency.


Assuntos
Diabetes Mellitus Tipo 2 , Diabetes Mellitus , Desnutrição , Distúrbios Nutricionais , Humanos , Secreção de Insulina , Insulina/metabolismo
4.
Diabetes Obes Metab ; 2024 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-39044309

RESUMO

Obesity has increased dramatically worldwide. Being overweight or obese can lead to various conditions, including dyslipidaemia, hypertension, glucose intolerance and metabolic syndrome (MetS), which may further lead to type 2 diabetes mellitus (T2DM). Previous studies have identified a link between ß-cell dysfunction and the severity of MetS, with multiple organs and tissues affected. Identifying the associations between pancreatic ß-cell dysfunction and organs is critical. Research has focused on the interaction between the liver, gut and pancreatic ß-cells. However, the mechanisms and related core targets are still not perfectly elucidated. The aims of this review were to summarize the mechanisms of ß-cell dysfunction and to explore the potential pathogenic pathways and targets that connect the liver, gut, adipose tissue, muscle, and brain to pancreatic ß-cell dysfunction.

5.
Exp Cell Res ; 422(1): 113406, 2023 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-36332684

RESUMO

The reduction of insulin secretion due to pancreatic ß cell injury caused by autoimmune reaction is the pathological basis of Type 1 diabetes mellitus (T1DM). Therefore, seeking new molecular targets for alleviating pancreatic ß cell injury will provide experimental basis for the prevention and treatment of T1DM. SRY-box 9 (Sox9) is not only an important molecule regulating the development of various organs, but also its high expression can aggravate the pathological process of various diseases. In addition, Sox9+ cells are also pancreatic progenitor cells, participating in pancreatic repair reaction induced by injury. In our study, elevated blood glucose and lack of pancreatic ß cells almost returned to normal over time after streptozotocin (STZ)-induced pancreatic ß cell damage, implying that pancreatic ß cells were regenerated after STZ-induced injury. In particular, the expression of Sox9 was significantly elevated during pancreatic ß cell regeneration. On this basis, we conducted in vitro experiments to verify whether overexpression of Sox9 could inhibit the damage of pancreatic ß cells by inflammatory factors. Our results showed that overexpression of Sox9 alleviated the damage of pancreatic ß cells by inflammatory factors and improved the inhibitory effect of inflammatory factors on insulin secretion of pancreatic ß cells. Unsurprising, blood glucose levels, insulin content and pancreatic ß cell number failed to return to near-normal levels timely after pancreatic ß cells specific knockout Sox9 mice were treated with STZ, further confirming the importance of Sox9 in facilitating pancreatic ß cell repair or regeneration. Our study indicate that enhanced Sox9 activity might protect pancreatic ß cells from autoimmune induced damage and thus improve the pathological process of T1DM.


Assuntos
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 1 , Células Secretoras de Insulina , Camundongos , Animais , Células Secretoras de Insulina/metabolismo , Glicemia/metabolismo , Diabetes Mellitus Tipo 1/patologia , Diabetes Mellitus Experimental/metabolismo , Estreptozocina/farmacologia , Insulina/metabolismo , Camundongos Knockout
6.
Biochem Biophys Res Commun ; 658: 80-87, 2023 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-37027908

RESUMO

Wfs1 is an endoplasmic reticulum (ER) membrane located protein highly expressed in pancreatic ß cells and brain. Wfs1 deficiency causes adult pancreatic ß cells dysfunction following ß cells apoptosis. Previous studies mainly focus on the Wfs1 function in adult mouse pancreatic ß cells. However, whether Wfs1 loss-of-function impairs mouse pancreatic ß cell from its early development is unknown. In our study, Wfs1 deficiency disrupts the composition of mouse pancreatic endocrine cells from early postnatal day 0 (P0) to 8 weeks old, with decreased percentage of ß cells and increased percentage of α and δ cells. Meanwhile, Wfs1 loss-of-function leads to reduced intracellular insulin content. Notably, Wfs1 deficiency impairs Glut2 localization and causes the accumulation of Glut2 in mouse pancreatic ß cell cytoplasm. In Wfs1-deficient mice, glucose homeostasis is disturbed from early 3 weeks old to 8 weeks old. This work reveals that Wfs1 is significantly required for the composition of pancreatic endocrine cells and is essential for Glut2 localization in mouse pancreatic ß cells.


Assuntos
Células Secretoras de Insulina , Proteínas de Membrana , Síndrome de Wolfram , Animais , Camundongos , Retículo Endoplasmático/metabolismo , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Síndrome de Wolfram/metabolismo , Proteínas de Membrana/genética , Mutação com Perda de Função
7.
Cell Tissue Res ; 394(1): 131-144, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37474621

RESUMO

Pancreatic ß cell clusters produce insulin and play a central role in glucose homeostasis. The regenerative capacity of mammalian ß cells is limited and the loss of ß cells causes diabetes. In contrast, zebrafish ß cell clusters have a high regenerative capacity, making them an attractive model to study ß cell cluster regeneration. How zebrafish ß cell clusters regenerate, when the regeneration process is complete, and the identification of the cellular source of regeneration are fundamental questions that require investigation. Here, using larval and adult zebrafish, we demonstrate that pancreatic ß cell clusters undergo a two-step regeneration process, regenerating functionality and then ß cell numbers. Additionally, we found that all regenerating pancreatic ß cells arose from Neurod1-expressing cells and that cells from different lineages contribute to both functional and ß cell number recovery throughout their life. Furthermore, we found that during development and neogenesis, as well as regeneration, all ß cells undergo Neurod1expression in zebrafish. Together, these results shed light on the fundamental cellular mechanisms underlying ß cell cluster development, neogenesis, and regeneration.


Assuntos
Diabetes Mellitus , Células Secretoras de Insulina , Animais , Linhagem da Célula , Insulina , Mamíferos , Peixe-Zebra , Proteínas de Peixe-Zebra/metabolismo
8.
Gastroenterology ; 160(5): 1662-1678.e18, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33421511

RESUMO

BACKGROUND & AIMS: Interstitial cells of Cajal (ICCs) and pancreatic ß cells require receptor tyrosine kinase (KIT) to develop and function properly. Degeneration of ICCs is linked to diabetic gastroparesis. The mechanisms linking diabetes and gastroparesis are unclear, but may involve microRNA (miRNA)-mediated post-transcriptional gene silencing in KIT+ cells. METHODS: We performed miRNA-sequencing analysis from isolated ICCs in diabetic mice and plasma from patients with idiopathic and diabetic gastroparesis. miR-10b-5p target genes were identified and validated in mouse and human cell lines. For loss-of-function studies, we used KIT+ cell-restricted mir-10b knockout mice and KIT+ cell depletion mice. For gain-of-function studies, a synthetic miR-10b-5p mimic was injected in multiple diabetic mouse models. We compared the efficacy of miR-10b-5p mimic treatment vs antidiabetic and prokinetic medicines. RESULTS: miR-10b-5p is highly expressed in ICCs from healthy mice, but drastically depleted in ICCs from diabetic mice. A conditional knockout of mir-10b in KIT+ cells or depletion of KIT+ cells in mice leads to degeneration of ß cells and ICCs, resulting in diabetes and gastroparesis. miR-10b-5p targets the transcription factor Krüppel-like factor 11 (KLF11), which negatively regulates KIT expression. The miR-10b-5p mimic or Klf11 small interfering RNAs injected into mir-10b knockout mice, diet-induced diabetic mice, and TALLYHO polygenic diabetic mice rescue the diabetes and gastroparesis phenotype for an extended period of time. Furthermore, the miR-10b-5p mimic is more effective in improving glucose homoeostasis and gastrointestinal motility compared with common antidiabetic and prokinetic medications. CONCLUSIONS: miR-10b-5p is a key regulator in diabetes and gastrointestinal dysmotility via the KLF11-KIT pathway. Restoration of miR-10b-5p may provide therapeutic benefits for these disorders.


Assuntos
Glicemia/metabolismo , Diabetes Mellitus/prevenção & controle , Esvaziamento Gástrico , Trânsito Gastrointestinal , Gastroparesia/prevenção & controle , Células Secretoras de Insulina/metabolismo , Células Intersticiais de Cajal/metabolismo , MicroRNAs/metabolismo , Adulto , Idoso , Animais , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Diabetes Mellitus/genética , Diabetes Mellitus/metabolismo , Modelos Animais de Doenças , Feminino , Gastroparesia/genética , Gastroparesia/metabolismo , Gastroparesia/fisiopatologia , Células HEK293 , Humanos , Células Secretoras de Insulina/patologia , Células Intersticiais de Cajal/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , MicroRNAs/genética , Pessoa de Meia-Idade , Células NIH 3T3 , Proteínas Proto-Oncogênicas c-kit/genética , Proteínas Proto-Oncogênicas c-kit/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Adulto Jovem
9.
Biochem Biophys Res Commun ; 605: 90-96, 2022 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-35316768

RESUMO

Patients with type 2 diabetes often exhibit impairments in both glucose-induced insulin secretion (GIIS) and incretin-induced insulin secretion (IIIS). These phenotypes are associated with altered glucose metabolism in pancreatic ß-cells, although the molecular mechanisms remain unclear. Here, we used MIN6-K8 pancreatic ß-cell lines as a model to examine the effect of O-linked N-acetylglucosamine glycosylation (O-GlcNAcylation), a glucose-induced protein posttranslational modification, on insulin secretion. O-GlcNAcylation was enhanced in high-glucose-treated MIN6-K8 cells, and high levels of O-GlcNAcylation attenuated PKA-dependent phosphorylation, suggesting that the two protein modifications may compete with each other. Immunoprecipitation proteomic analysis identified six candidate proteins that were O-GlcNAcylated by high-glucose treatment, whereas the O-GlcNAcylations were removed by treatment with an incretin mimetic, exendin-4. Among these proteins, knockdown of myocyte enhancer factor 2D (Mef2d) enhanced insulin secretion, and high-glucose treatment increased the level of O-GlcNAcylation of Mef2d in MIN6-K8 cells. Furthermore, knockout of Mef2d promoted GIIS in MIN6-K8 cells, whereas adenovirus-mediated rescue of Mef2d decreased GIIS in the knockout cells. These results suggest that Mef2d negatively regulates insulin secretion through O-GlcNAcylation.


Assuntos
Diabetes Mellitus Tipo 2 , Acetilglucosamina/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Glucose/metabolismo , Humanos , Incretinas , Secreção de Insulina , Fatores de Transcrição MEF2/metabolismo , Processamento de Proteína Pós-Traducional , Proteômica
10.
Calcif Tissue Int ; 110(2): 244-259, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34417862

RESUMO

A bidirectional and complex relationship exists between bone and glycemia. Persons with type 2 diabetes (T2D) are at risk for bone loss and fracture, however, heightened osteoanabolism may ameliorate T2D-induced deficits in glycemia as bone-forming osteoblasts contribute to energy metabolism via increased glucose uptake and cellular glycolysis. Mice globally lacking nuclear matrix protein 4 (Nmp4), a transcription factor expressed in all tissues and conserved between humans and rodents, are healthy and exhibit enhanced bone formation in response to anabolic osteoporosis therapies. To test whether loss of Nmp4 similarly impacted bone deficits caused by diet-induced obesity, male wild-type and Nmp4-/- mice (8 weeks) were fed either low-fat diet or high-fat diet (HFD) for 12 weeks. Endpoint parameters included bone architecture, structural and estimated tissue-level mechanical properties, body weight/composition, glucose-stimulated insulin secretion, glucose tolerance, insulin tolerance, and metabolic cage analysis. HFD diminished bone architecture and ultimate force and stiffness equally in both genotypes. Unexpectedly, the Nmp4-/- mice exhibited deficits in pancreatic ß-cell function and were modestly glucose intolerant under normal diet conditions. Despite the ß-cell deficits, the Nmp4-/- mice were less sensitive to HFD-induced weight gain, increases in % fat mass, and decreases in glucose tolerance and insulin sensitivity. We conclude that Nmp4 supports pancreatic ß-cell function but suppresses peripheral glucose utilization, perhaps contributing to its suppression of induced skeletal anabolism. Selective disruption of Nmp4 in peripheral tissues may provide a strategy for improving both induced osteoanabolism and energy metabolism in comorbid patients.


Assuntos
Diabetes Mellitus Tipo 2 , Resistência à Insulina , Animais , Dieta Hiperlipídica/efeitos adversos , Humanos , Insulina , Secreção de Insulina , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Associadas à Matriz Nuclear/metabolismo , Hormônio Paratireóideo , Fatores de Transcrição/metabolismo
11.
J Biochem Mol Toxicol ; 36(4): e23004, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35191145

RESUMO

Saponin gypenoside A (GP) has shown its potential to handle diabetes mellitus. MicroRNA-150-3p (miR-150-3p) is closely related to the dysfunction of pancreatic ß cells by targeting PDX1. Given the function of GP is related to its regulation on different miRs, the current study assessed the role of miR-150-3p as a therapeutic target for the hypoglycemic effects of GP. Pancreatic ß cell dysfunction was induced in mice using the high-fatty diet (HFD) method and then handled with GP. Changes in insulin release and resistance and the activity of the miR-150-3p/PDX1 axis were detected. The expression of miR-150-3p was induced to confirm its central in the effects of GP. The results of in vivo tests were then validated with in vitro assays. HFD administration suppressed glucose tolerance, delayed insulin release, and induced insulin resistance and pancreas apoptosis in mice, which was indicative of the dysfunction of ß pancreatic cells. Changes in pancreatic ß function were associated with the increased expression of miR-150-3p and suppressed expression of PDX1. After the administration of GP, the impairments of the pancreas were alleviated and the expression of miR-150-3p was inhibited, contributing to the restored level of PDX1. The injection of miR-150-3p agomir counteracted the protective effects of GP. In in vitro assays, the pretransfection of miR-150-3p mimetics also counteracted the protective effects of GP on pancreatic ß cells against palmitic acid. Collectively, miR-150-3p played a key role in the protective effects of GP against pancreatic ß cell dysfunction by inhibiting PDX1 expression.


Assuntos
Células Secretoras de Insulina , MicroRNAs , Animais , Gynostemma , Proteínas de Homeodomínio/metabolismo , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Extratos Vegetais , Transdução de Sinais , Transativadores/metabolismo
12.
Bioorg Chem ; 129: 106194, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36244321

RESUMO

Pancreatic islet transplantation is an ideal treatment strategy for type 1 diabetes mellitus (T1DM), but hypoxia-induced pancreatic ß cell death after islet transplantation is the huge obstacle that causes failure of this therapy. Thus, it become necessary to improve pancreatic ß cell viability under hypoxic conditions. In the present study, we designed mesenchymal stem cells (MSCs)-derived hypoxia-inducible factor 1α (HIF-1α)-overexpressed extracellular vesicle (EVs) (HIF-1α-EVs) and found that HIF-1α-EVs was effectively to promote cell viability and autophagy, and suppress cell apoptosis and senescence in the hypoxia-treated pancreatic ß cells. In addition, blockage of autophagy by its inhibitor 3-methyladenine (3-MA) abrogated the rescuing effects of HIF-1α-EVs on hypoxia-induced pancreatic ß cell death. Then, the potential underlying mechanisms by which HIF-1α-EVs triggered protective autophagy were uncovered, and we found that HIF-1α-EVs upregulated YTHDF1, resulting in the upregulation of autophagy-associated proteins (ATG5, ATG2A and ATG14), which were abrogated by deleting m6A writer METTL3. Finally, we verified that HIF-1α-EVs rescued cell viability, and reversed hypoxia-induced pancreatic ß cell apoptosis and senescence in a YTHDF1-dependent manner. Collectively, we concluded that MSCs-derived HIF-1α-EVs activated YTHDF1-mediated protective autophagy to promote pancreatic ß cell survival under hypoxic conditions, and HIF-1α-EVs could be used as candidate treatment strategy to increase the success rate of islet transplantation.


Assuntos
Vesículas Extracelulares , Células Secretoras de Insulina , Células-Tronco Mesenquimais , Humanos , Células Secretoras de Insulina/metabolismo , Hipóxia Celular , Autofagia , Apoptose , Vesículas Extracelulares/metabolismo , Células-Tronco Mesenquimais/metabolismo , Hipóxia/metabolismo , Metiltransferases/metabolismo , Proteínas de Ligação a RNA/metabolismo
13.
Clin Exp Pharmacol Physiol ; 49(1): 104-112, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34448213

RESUMO

Dysfunction of pancreatic ß cell insulin secretion is related to the pathogenesis of type 2 diabetes (T2D). Rab proteins have been shown to be key players in insulin secretion by pancreatic ß cells, and phogrin is a marker for the processes of exocytosis and insulin secretion. The purposes of this study were to clarify the regulatory role of Rab35 in insulin secretion and analyse the Rab35/phogrin interaction mechanism in ß-TC-6 cells. We studied the effects of Rab35 gene overexpression and interference on insulin secretion and phogrin expression and levels in ß-TC-6 cells. The Rab35/phogrin interaction was verified by GST pulldown, co-IP and co-localisation experiments. Here, we report that Rab35 is mainly distributed in the ß-TC-6-cell plasma membrane and cytoplasm. Rab35 overexpression promotes insulin secretion and decreases phogrin expression in ß-TC-6 cells, whereas its silencing significantly inhibits insulin secretion, promotes phogrin expression (p < 0.05) and causes phogrin redistribution. Furthermore, Rab35 silencing suppresses exocytosis of insulin. Rab35 interacts with phogrin, and both proteins co-localise in the plasma membranes and cytoplasm of ß-TC-6 cells. Our study presents novel evidence that Rab35 regulates insulin secretion by inhibiting phogrin expression and causing intracellular phogrin redistribution in pancreatic ß cells.


Assuntos
Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Proteínas Tirosina Fosfatases Classe 8 Semelhantes a Receptores/fisiologia , Proteínas rab de Ligação ao GTP/fisiologia , Células HEK293 , Humanos , Células Secretoras de Insulina/fisiologia , Proteínas Tirosina Fosfatases Classe 8 Semelhantes a Receptores/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo
14.
Lipids Health Dis ; 21(1): 73, 2022 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-35982452

RESUMO

Syntaxin regulates pancreatic ß cell mass and participates in insulin secretion by regulating insulin exocytosis. In addition, syntaxin 4 reduces IFNγ and TNF-α signaling via NF-ĸB in islet ß-cells that facilitates plasma glucose sensing and appropriate insulin secretion. Arachidonic acid (AA) has potent anti-inflammatory actions and prevents the cytotoxic actions of alloxan and streptozotocin (STZ) against pancreatic ß cells and thus, prevents the development of type 1 diabetes mellitus (induced by alloxan and STZ) and by virtue of its anti-inflammatory actions protects against the development of type 2 diabetes mellitus (DM) induced by STZ in experimental animals that are models of type 1 and type 2 DM in humans. AA has been shown to interact with syntaxin and thus, potentiate exocytosis. AA enhances cell membrane fluidity, increases the expression of GLUT and insulin receptors, and brings about its anti-inflammatory actions at least in part by enhancing the formation of its metabolite lipoxin A4 (LXA4). Prostaglandin E2 (PGE2), the pro-inflammatory metabolite of AA, activates ventromedial hypothalamus (VMH) neurons of the hypothalamus and inhibits insulin secretion leading to reduced glucose tolerance and decreases insulin sensitivity in the skeletal muscle and liver. This adverse action of PGE2 on insulin release and action can be attributed to its (PGE2) pro-inflammatory action and inhibitory action on vagal tone (vagus nerve and its principal neurotransmitter acetylcholine has potent anti-inflammatory actions). High fat diet fed animals have hypothalamic inflammation due to chronic elevation of PGE2. Patients with type 2 DM show low plasma concentrations of AA and LXA4 and elevated levels of PGE2. Administration of AA enhances LXA4 formation without altering or reducing PGE2 levels and thus, tilts the balance more towards anti-inflammatory events. These results suggest that administration of AA is useful in the prevention and management of DM by enhancing the action of syntaxin, increasing cell membrane fluidity, and reducing VMH inflammation. Docosahexaenoic acid (DHA) has actions like AA: it increases cell membrane fluidity; has anti-inflammatory actions by enhancing the formation of its anti-inflammatory metabolites resolvins, protectins and maresins; interacts with syntaxin and enhance exocytosis in general and of insulin. But the DHA content of cell membrane is lower compared to AA and its content in brain is significant. Hence, it is likely DHA is important in neurotransmitters secretion and regulating hypothalamic inflammation. It is likely that a combination of AA and DHA can prevent DM.


Assuntos
Diabetes Mellitus Tipo 2 , Insulinas , Aloxano , Animais , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Ácido Araquidônico/farmacologia , Diabetes Mellitus Tipo 2/tratamento farmacológico , Dinoprostona , Ácidos Docosa-Hexaenoicos/farmacologia , Humanos , Inflamação , Insulinas/efeitos adversos , Proteínas Qa-SNARE , Estreptozocina
15.
Int J Mol Sci ; 23(8)2022 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-35457121

RESUMO

Calcium (Ca2+) is a ubiquitous and fundamental signaling component that is utilized by cells to regulate a diverse range of cellular functions, such as insulin secretion from pancreatic ß-cells of the islets of Langerhans. Cyclic ADP-ribose (cADPR), synthesized from NAD+ by ADP-ribosyl cyclase family proteins, such as the mammalian cluster of differentiation 38 (CD38), is important for intracellular Ca2+ mobilization for cell functioning. cADPR induces Ca2+ release from endoplasmic reticulum via the ryanodine receptor intracellular Ca2+ channel complex, in which the FK506-binding protein 12.6 works as a cADPR-binding regulatory protein. Recently, involvements of the CD38-cADPR signal system in several human diseases and animal models have been reported. This review describes the biochemical and molecular biological basis of the CD38-cADPR signal system and the diseases caused by its abnormalities.


Assuntos
Antígenos CD , ADP-Ribose Cíclica , ADP-Ribosil Ciclase 1/metabolismo , Animais , Antígenos CD/metabolismo , Antígenos de Diferenciação/metabolismo , Cálcio/metabolismo , Sinalização do Cálcio , ADP-Ribose Cíclica/metabolismo , Mamíferos/metabolismo , Glicoproteínas de Membrana/metabolismo
16.
J Environ Sci (China) ; 115: 162-172, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-34969446

RESUMO

Per- and polyfluoroalkyl substances (PFAS) are found to have multiple adverse outcomes on human health. Recently, epidemiological and toxicological studies showed that exposure to PFAS had adverse impacts on pancreas and showed association with insulin abnormalities. To explore how PFAS may contribute to diabetes, we studied impacts of perfluorooctane sulfonate (PFOS) on cell viability and insulin release capacity of pancreatic ß cells by using in vivo and in vitro methods. We found that 28-day administration with PFOS (10 mg/(kg body weight•day)) caused reductions of pancreas weight and islet size in male mice. PFOS administration also led to lower serum insulin level both in fasting state and after glucose infusion among male mice. For cell-based in vitro bioassay, we used mouse ß-TC-6 cancer cells and found 48-hr exposure to PFOS decreased the cell viability at 50 µmol/L. By measuring insulin content in supernatant, 48-hr pretreatment of PFOS (100 µmol/L) decreased the insulin release capacity of ß-TC-6 cells after glucose stimulation. Although these concentrations were higher than the environmental concentration of PFOS, it might be reasonable for high concentration of PFOS to exert observable toxic effects in mice considering mice had a faster removal efficiency of PFOS than human. PFOS exposure (50 µmol/L) to ß-TC-6 cells induced intracellular accumulation of reactive oxidative specie (ROS). Excessive ROS induced the reactive toxicity of cells, which eventually invoke apoptosis and necrosis. Results in this study provide evidence for the possible causal link of exposure to PFOS and diabetes risk.


Assuntos
Ácidos Alcanossulfônicos , Poluentes Ambientais , Fluorocarbonos , Células Secretoras de Insulina , Ácidos Alcanossulfônicos/toxicidade , Animais , Sobrevivência Celular , Fluorocarbonos/toxicidade , Insulina , Masculino , Camundongos
17.
J Transl Med ; 19(1): 137, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33794915

RESUMO

Type 1 diabetes affects millions of people globally and requires careful management to avoid serious long-term complications, including heart and kidney disease, stroke, and loss of sight. The type 1 diabetes patient cohort is highly heterogeneous, with individuals presenting with disease at different stages and severities, arising from distinct etiologies, and overlaying varied genetic backgrounds. At present, the "one-size-fits-all" treatment for type 1 diabetes is exogenic insulin substitution therapy, but this approach fails to achieve optimal blood glucose control in many individuals. With advances in our understanding of early-stage diabetes development, diabetes stratification, and the role of genetics, type 1 diabetes is a promising candidate for a personalized medicine approach, which aims to apply "the right therapy at the right time, to the right patient". In the case of type 1 diabetes, great efforts are now being focused on risk stratification for diabetes development to enable pre-clinical detection, and the application of treatments such as gene therapy, to prevent pancreatic destruction in a sub-set of patients. Alongside this, breakthroughs in stem cell therapies hold great promise for the regeneration of pancreatic tissues in some individuals. Here we review the recent initiatives in the field of personalized medicine for type 1 diabetes, including the latest discoveries in stem cell and gene therapy for the disease, and current obstacles that must be overcome before the dream of personalized medicine for all type 1 diabetes patients can be realized.


Assuntos
Diabetes Mellitus Tipo 1 , Diabetes Mellitus Tipo 1/diagnóstico , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/terapia , Humanos , Insulina , Pâncreas , Medicina de Precisão , Transplante de Células-Tronco
18.
Rev Cardiovasc Med ; 22(3): 769-777, 2021 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-34565075

RESUMO

The renin-angiotensin system (RAS) helps to regulate cardiovascular function, the maintenance of electrolyte and fluid balance, and blood pressure. The RAS contains two axes; the angiotensin-converting enzyme/angiotensin II/Ang II type 1 receptors (ACE/Ang II/AT1) classic axis, which has a role in regulating blood pressure, vascular oxidative stress, coagulation, and cellular proliferation. The other is the angiotensin-converting enzyme 2/angiotensin-(1-7)/Mas receptors (ACE2/Ang-(1-7)/Mas) axis, which can inhibit the former axis, improve fat metabolism, reduce inflammation and oxidative stress, and enhance glucose tolerance and insulin sensitivity. The ACE2/Ang-(1-7)/Mas axis is found in blood vessels, kidneys, liver, pancreas and the brain. It can protect the body from abnormalities in glucose metabolism. The ACE2/Ang-(1-7)/Mas axis can enhance glucose tolerance and improve insulin sensitivity by protecting pancreatic ß cells, increasing insulin secretion, improving glucose metabolism in adipose tissue, enhancing glucose uptake by skeletal muscle, and inhibiting hepatic gluconeogenesis. This article reviews the main characteristics and functions of the ACE2/Ang-(1-7)/Mas axis and its regulation of glucose metabolism in order to demonstrate its potential as a target for the treatment of metabolic diseases such as diabetes.


Assuntos
Angiotensina I , Enzima de Conversão de Angiotensina 2 , Glucose , Humanos , Fragmentos de Peptídeos
19.
FASEB J ; 34(3): 3915-3931, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31944392

RESUMO

So far, the mechanism that links mitochondrial dysfunction to PDX1 inhibition in the pathogenesis of pancreatic ß cell dysfunction under diabetic condition remains largely unclear. This study determined the role of mitochondrial protein FAM3A in regulating PDX1 expression in pancreatic ß cells using gain- and loss-of function methods in vitro and in vivo. Within pancreas, FAM3A is highly expressed in ß, α, δ, and pp cells of islets. Islet FAM3A expression was correlated with insulin expression under physiological and diabetic conditions. Mice with specific knockout of FAM3A in islet ß cells exhibited markedly blunted insulin secretion and glucose intolerance. FAM3A-deficient islets showed significant decrease in PDX1 expression, and insulin expression and secretion. FAM3A overexpression upregulated PDX1 and insulin expressions, and augmented insulin secretion in cultured islets and ß cells. Mechanistically, FAM3A enhanced ATP production to elevate cellular Ca2+ level and promote insulin secretion. Furthermore, FAM3A-induced ATP release activated CaM to function as a co-activator of FOXA2, stimulating PDX1 gene transcription. In conclusion, FAM3A plays crucial roles in controlling PDX1 and insulin expressions in pancreatic ß cells. Inhibition of FAM3A will trigger mitochondrial dysfunction to repress PDX1 and insulin expressions.


Assuntos
Citocinas/metabolismo , Proteínas de Homeodomínio/metabolismo , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Transativadores/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Cálcio/metabolismo , Linhagem Celular , Células Cultivadas , Citocinas/genética , Glucose/metabolismo , Fator 3-beta Nuclear de Hepatócito , Proteínas de Homeodomínio/genética , Humanos , Immunoblotting , Imunoprecipitação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Transativadores/genética
20.
Int J Mol Sci ; 22(4)2021 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-33546200

RESUMO

Diabetes is a chronic metabolic disorder characterized by inappropriately elevated glucose levels as a result of impaired pancreatic ß cell function and insulin resistance. Extensive studies have been conducted to elucidate the mechanism involved in the development of ß cell failure and death under diabetic conditions such as hyperglycemia, hyperlipidemia, and inflammation. Of the plethora of proposed mechanisms, endoplasmic reticulum (ER) stress, mitochondrial dysfunction, and oxidative stress have been shown to play a central role in promoting ß cell dysfunction. It has become more evident in recent years that these 3 factors are closely interrelated and importantly aggravate each other. Oxidative stress in particular is of great interest to ß cell health and survival as it has been shown that ß cells exhibit lower antioxidative capacity. Therefore, this review will focus on discussing factors that contribute to the development of oxidative stress in pancreatic ß cells and explore the downstream effects of oxidative stress on ß cell function and health. Furthermore, antioxidative capacity of ß cells to counteract these effects will be discussed along with new approaches focused on preserving ß cells under oxidative conditions.


Assuntos
Diabetes Mellitus/fisiopatologia , Células Secretoras de Insulina/fisiologia , Estresse Oxidativo , Animais , Diabetes Mellitus/metabolismo , Humanos , Células Secretoras de Insulina/metabolismo , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA