Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 299(8): 104986, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37392854

RESUMEN

Congenital hyperinsulinism of infancy (CHI) can be caused by a deficiency of the ubiquitously expressed enzyme short-chain 3-hydroxyacyl-CoA dehydrogenase (SCHAD). To test the hypothesis that SCHAD-CHI arises from a specific defect in pancreatic ß-cells, we created genetically engineered ß-cell-specific (ß-SKO) or hepatocyte-specific (L-SKO) SCHAD knockout mice. While L-SKO mice were normoglycemic, plasma glucose in ß-SKO animals was significantly reduced in the random-fed state, after overnight fasting, and following refeeding. The hypoglycemic phenotype was exacerbated when the mice were fed a diet enriched in leucine, glutamine, and alanine. Intraperitoneal injection of these three amino acids led to a rapid elevation in insulin levels in ß-SKO mice compared to controls. Consistently, treating isolated ß-SKO islets with the amino acid mixture potently enhanced insulin secretion compared to controls in a low-glucose environment. RNA sequencing of ß-SKO islets revealed reduced transcription of ß-cell identity genes and upregulation of genes involved in oxidative phosphorylation, protein metabolism, and Ca2+ handling. The ß-SKO mouse offers a useful model to interrogate the intra-islet heterogeneity of amino acid sensing given the very variable expression levels of SCHAD within different hormonal cells, with high levels in ß- and δ-cells and virtually absent α-cell expression. We conclude that the lack of SCHAD protein in ß-cells results in a hypoglycemic phenotype characterized by increased sensitivity to amino acid-stimulated insulin secretion and loss of ß-cell identity.


Asunto(s)
3-Hidroxiacil-CoA Deshidrogenasa , Aminoácidos , Hiperinsulinismo Congénito , Hipoglucemia , Secreción de Insulina , Células Secretoras de Insulina , Animales , Ratones , Aminoácidos/metabolismo , Aminoácidos/farmacología , Hipoglucemia/enzimología , Hipoglucemia/genética , Insulina/metabolismo , Secreción de Insulina/efectos de los fármacos , Ratones Noqueados , 3-Hidroxiacil-CoA Deshidrogenasa/deficiencia , 3-Hidroxiacil-CoA Deshidrogenasa/genética , Células Secretoras de Insulina/enzimología , Hiperinsulinismo Congénito/genética
2.
Int J Mol Sci ; 25(9)2024 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-38731945

RESUMEN

The main hallmark in the development of both type 1 and type 2 diabetes is a decline in functional ß-cell mass. This decline is predominantly attributed to ß-cell death, although recent findings suggest that the loss of ß-cell identity may also contribute to ß-cell dysfunction. This phenomenon is characterized by a reduced expression of key markers associated with ß-cell identity. This review delves into the insights gained from single-cell omics research specifically focused on ß-cell identity. It highlights how single-cell omics based studies have uncovered an unexpected level of heterogeneity among ß-cells and have facilitated the identification of distinct ß-cell subpopulations through the discovery of cell surface markers, transcriptional regulators, the upregulation of stress-related genes, and alterations in chromatin activity. Furthermore, specific subsets of ß-cells have been identified in diabetes, such as displaying an immature, dedifferentiated gene signature, expressing significantly lower insulin mRNA levels, and expressing increased ß-cell precursor markers. Additionally, single-cell omics has increased insight into the detrimental effects of diabetes-associated conditions, including endoplasmic reticulum stress, oxidative stress, and inflammation, on ß-cell identity. Lastly, this review outlines the factors that may influence the identification of ß-cell subpopulations when designing and performing a single-cell omics experiment.


Asunto(s)
Células Secretoras de Insulina , Análisis de la Célula Individual , Células Secretoras de Insulina/metabolismo , Humanos , Análisis de la Célula Individual/métodos , Animales , Genómica/métodos , Estrés del Retículo Endoplásmico/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patología
3.
Cytotherapy ; 23(6): 510-520, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33736932

RESUMEN

BACKGROUND AIMS: The authors aimed to observe ß-cell dedifferentiation in type 2 diabetes mellitus (T2DM) and investigate the reversal effect of umbilical cord-derived mesenchymal stem cells (UC-MSCs) on early- and late-stage ß-cell dedifferentiation. METHODS: In high-fat diet (HFD)/streptozotocin (STZ)-induced T2DM mice, the authors examined the predominant role of ß-cell dedifferentiation over apoptosis in the development of T2DM and observed the reversion of ß-cell dedifferentiation by UC-MSCs. Next, the authors used db/db mice to observe the progress of ß-cell dedifferentiation from early to late stage, after which UC-MSC infusions of the same amount were performed in the early and late stages of dedifferentiation. Improvement in metabolic indices and restoration of ß-cell dedifferentiation markers were examined. RESULTS: In HFD/STZ-induced T2DM mice, the proportion of ß-cell dedifferentiation was much greater than that of apoptosis, demonstrating that ß-cell dedifferentiation was the predominant contributor to T2DM. UC-MSC infusions significantly improved glucose homeostasis and reversed ß-cell dedifferentiation. In db/db mice, UC-MSC infusions in the early stage significantly improved glucose homeostasis and reversed ß-cell dedifferentiation. In the late stage, UC-MSC infusions mildly improved glucose homeostasis and partially reversed ß-cell dedifferentiation. Combining with other studies, the authors found that the reversal effect of UC-MSCs on ß-cell dedifferentiation relied on the simultaneous relief of glucose and lipid metabolic disorders. CONCLUSIONS: UC-MSC therapy is a promising strategy for reversing ß-cell dedifferentiation in T2DM, and the reversal effect is greater in the early stage than in the late stage of ß-cell dedifferentiation.


Asunto(s)
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2 , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Animales , Desdiferenciación Celular , Diabetes Mellitus Experimental/terapia , Diabetes Mellitus Tipo 2/terapia , Humanos , Ratones , Cordón Umbilical
4.
BMC Endocr Disord ; 21(1): 47, 2021 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-33711989

RESUMEN

BACKGROUND: NKX6.1 is a transcription factor for insulin, as well as a marker for ß cell maturity. Abnormal NKX6.1 expression in ß cells, such as translocation from the nucleus to cytoplasm or lost expression, has been shown as a marker for ß cell dedifferentiation. METHODS: We obtained pancreatic sections from organ donors and immunofluorescence staining with NKX6.1 and insulin was performed to characterize NKX6.1 expression in subjects with or without type 2 diabetes mellitus (T2DM). RESULTS: Our results showed that cells with insulin expression but no nucleic NKX6.1 expression (NKX6.1Nuc-Ins+), and cells with cytoplasmic NKX6.1 expression but no insulin expression (NKX6.1cytIns-) were significantly increased in T2DM subjects and positively correlated with glycated hemoglobin (HbA1c), indicating the elevated ß cell dedifferentiation with NKX6.1 inactivation in T2DM. To investigate whether ß cell dedifferentiation has initiated in subjects with higher risks for T2DM, we next analyzed the association between ß-cell dedifferentiation level in ND subjects with different ages, body mass index, and HbA1c. The results showed the absolute number and percentage of dedifferentiated ß cells with NKX6.1 inactivation did not significantly change in subjects with advanced aging, obesity, or modest hyperglycemia, indicating that the ß cell dedifferentiation might mainly occur after T2DM was diagnosed. CONCLUSION: Our results suggested that NKX6.1 expression in ß cells was changed in type 2 diabetic subjects, evidenced by significantly increased NKX6.1Nuc-Ins+ and NKX6.1cytIns- cells. This abnormality did not occur more frequently in subjects with a higher risk for T2DM, suggesting that ß cell dedifferentiation might be secondary to the pathological changes in T2DM.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Proteínas de Homeodominio/metabolismo , Células Secretoras de Insulina/metabolismo , Estado Prediabético/metabolismo , Adulto , Anciano , Autopsia , Estudios de Casos y Controles , Recuento de Células , Diferenciación Celular , Diabetes Mellitus Tipo 2/patología , Femenino , Hemoglobina Glucada/metabolismo , Humanos , Células Secretoras de Insulina/patología , Células Secretoras de Insulina/fisiología , Masculino , Persona de Mediana Edad , Estado Prediabético/patología , Factores de Riesgo
5.
Mol Med ; 24(1): 43, 2018 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-30134927

RESUMEN

BACKGROUND: Type 2 diabetes mellitus (T2DM) is characterized by pancreatic ß-cell failure, which arises from metabolic stress and results in ß cell dedifferentiation, leading to ß-cell death. Pathological activation of the renin-angiotensin system (RAS) contributes to increase cell stress, while RAS intervention reduces the onset of T2DM in high-risk populations and promotes insulin secretion in rodents. In this study, we investigated whether and how RAS induces ß-cell dedifferentiation and the mechanism underlying this process. METHODS: In vitro, with the methods of quantitative real-time reverse transcriptase-PCR (qRT-PCR) and western blotting, we examined the change of cell identity-related gene expression, progenitor like gene expression, cellular function, and nuclear factor kappa b (NF-κb) signaling activity in ß cell lines after exposure to angiotensin II (AngII) and disruption of RAS. In vivo, parallel studies were performed using db/db mice. Related protein expression was detected by Immunofluorescence analysis. RESULT: Activation of RAS induced dedifferentiation and impaired insulin secretion, eventually leading to ß-cell failure. Mechanistically, Angll induced ß-cell dedifferentiation via NF-κb signaling, while treatment with lrbesartan and sc-514 reversed the progenitor state of ß cells. CONCLUSION: The present study found that RAS might induce ß-cell dedifferentiation via angiotensin II receptor type 1 activation, which was promoted by NF-κb signaling. Therefore, blocking RAS or NF-kb signaling efficiently reversed the dedifferentiated status of ß cells, suggesting a potential therapy for patients with type 2 diabetes.


Asunto(s)
Angiotensina II/farmacología , Desdiferenciación Celular/efectos de los fármacos , Células Secretoras de Insulina/efectos de los fármacos , FN-kappa B/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Diabetes Mellitus Tipo 2/metabolismo , Expresión Génica/efectos de los fármacos , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Masculino , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Ratas , Sistema Renina-Angiotensina , Transducción de Señal/efectos de los fármacos
6.
FASEB J ; 31(11): 4985-4997, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28765173

RESUMEN

Adenosine, a key extracellular signaling mediator, regulates several aspects of metabolism by activating 4 G-protein-coupled receptors, the A1, A2A, A2B, and A3 adenosine receptors (ARs). The role of A2AARs in regulating high-fat-diet (HFD)-induced metabolic derangements is unknown. To evaluate the role of A2AARs in regulating glucose and insulin homeostasis in obesity, we fed A2AAR-knockout (KO) and control mice an HFD for 16 wk to initiate HFD-induced metabolic disorder. We found that genetic deletion of A2AARs caused impaired glucose tolerance in mice fed an HFD. This impaired glucose tolerance was caused by a decrease in insulin secretion but not in insulin sensitivity. Islet size and insulin content in pancreata of A2AAR-deficient mice were decreased compared with control mice after consuming an HFD. A2AAR-KO mice had decreased expression of the ß-cell-specific markers pdx1, glut2, mafA, and nkx6.1 and increased expression of the dedifferentiation markers sox2 and hes1. Ex vivo islet experiments confirmed the role of A2AARs in protecting against decreased insulin content and release caused by HFD. Other experiments with bone marrow chimeras revealed that inflammation was not the primary cause of decreased insulin secretion in A2AAR-KO mice. Altogether, our data showed that A2AARs control pancreatic dysfunction in HFD-induced obesity.-Csóka, B., Töro, G., Vindeirinho, J., Varga, Z. V., Koscsó, B., Németh, Z. H., Kókai, E., Antonioli, L., Suleiman, M., Marchetti, P., Cseri, K., Deák, Á., Virág, L., Pacher, P., Bai, P., Haskó, G. A2A adenosine receptors control pancreatic dysfunction in high-fat-diet-induced obesity.


Asunto(s)
Grasas de la Dieta/efectos adversos , Células Secretoras de Insulina/metabolismo , Obesidad/metabolismo , Enfermedades Pancreáticas/metabolismo , Receptor de Adenosina A2A/metabolismo , Animales , Antígenos de Diferenciación/biosíntesis , Antígenos de Diferenciación/genética , Grasas de la Dieta/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Células Secretoras de Insulina/patología , Ratones , Ratones Noqueados , Obesidad/inducido químicamente , Obesidad/genética , Obesidad/patología , Enfermedades Pancreáticas/inducido químicamente , Enfermedades Pancreáticas/genética , Enfermedades Pancreáticas/patología , Receptor de Adenosina A2A/genética
7.
Am J Physiol Endocrinol Metab ; 311(3): E554-63, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27406742

RESUMEN

Limitations in ß-cell regeneration potential in middle-aged animals could contribute to the increased risk to develop diabetes associated with aging. We investigated ß-cell regeneration of middle-aged Wistar rats in response to two different regenerative stimuli: partial pancreatectomy (Px + V) and gastrin administration (Px + G). Pancreatic remnants were analyzed 3 and 14 days after surgery. ß-Cell mass increased in young animals after Px and was further increased after gastrin treatment. In contrast, ß-cell mass did not change after Px or after gastrin treatment in middle-aged rats. ß-Cell replication and individual ß-cell size were similarly increased after Px in young and middle-aged animals, and ß-cell apoptosis was not modified. Nuclear immunolocalization of neurog3 or nkx6.1 in regenerative duct cells, markers of duct cell plasticity, was increased in young but not in middle-aged Px rats. The pancreatic progenitor-associated transcription factors neurog3 and sox9 were upregulated in islet ß-cells of middle-aged rats and further increased after Px. The percentage of chromogranin A+/hormone islet cells was significantly increased in the pancreases of middle-aged Px rats. In summary, the potential for compensatory ß-cell hyperplasia and hypertrophy was retained in middle-aged rats, but ß-cell dedifferentiation and impaired duct cell plasticity limited ß-cell regeneration.


Asunto(s)
Desdiferenciación Celular/fisiología , Células Secretoras de Insulina/fisiología , Conductos Pancreáticos/citología , Conductos Pancreáticos/fisiología , Regeneración/fisiología , Envejecimiento/fisiología , Animales , Apoptosis/efectos de los fármacos , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Plasticidad de la Célula , Tamaño de la Célula , Gastrinas/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Masculino , Proteínas del Tejido Nervioso/metabolismo , Pancreatectomía , Ratas , Ratas Wistar , Factor de Transcripción SOX9/metabolismo , Regulación hacia Arriba
8.
Genes (Basel) ; 15(2)2024 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-38397173

RESUMEN

Serotonin emerges as a pivotal factor influencing the growth and functionality of ß-cells. Psilocybin, a natural compound derived from mushrooms of the Psilocybe genus, exerts agonistic effects on the serotonin 5-HT2A and 5-HT2B receptors, thereby mimicking serotonin's behavior. This study investigates the potential impacts of psilocybin on ß-cell viability, dedifferentiation, and function using an in vitro system. The INS-1 832/13 Rat Insulinoma cell line underwent psilocybin pretreatment, followed by exposure to high glucose-high lipid (HG-HL) conditions for specific time periods. After being harvested from treated cells, total transcript and cellular protein were utilized for further investigation. Our findings implied that psilocybin administration effectively mitigates HG-HL-stimulated ß-cell loss, potentially mediated through the modulation of apoptotic biomarkers, which is possibly related to the mitigation of TXNIP, STAT-1, and STAT-3 phosphorylation. Furthermore, psilocybin exhibits the capacity to modulate the expression of key genes associated with ß-cell dedifferentiation, including Pou5f1 and Nanog, indicating its potential in attenuating ß-cell dedifferentiation. This research lays the groundwork for further exploration into the therapeutic potential of psilocybin in Type II diabetes intervention.


Asunto(s)
Diabetes Mellitus Tipo 2 , Psilocibina , Animales , Ratas , Psilocibina/farmacología , Supervivencia Celular , Serotonina , Glucosa/farmacología , Lípidos , Proteínas de Ciclo Celular
9.
Nutrition ; 119: 112284, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38118383

RESUMEN

OBJECTIVE: The aim of this study was to explore the effect of the ketogenic diet (KD) on ß-cell dedifferentiation and hepatic lipid accumulation in db/db mice. METHODS: After a 3-wk habituation, male db/db mice ages 8 wk were assigned into one of three groups: normal diet (ND), KD, and 75% calorie restriction (CR) group. Free access to a standard diet, a KD, and 75% of a standard diet, respectively, were given to each group. Additionally, sex-matched 8-wk-old C57BL/6 mice were used to construct a control (C) group. After a 4-wk dietary intervention, mouse body weight, fasting blood glucose (FBG), blood lipids, fasting insulin (FINS), glucose tolerance, and ß-hydroxybutyric acid level were measured. The morphologies of the islet and liver were observed by hematoxylin and eosin staining. Positive expressions of ß-cell-specific transcription factors in mouse islets were determined by double immunofluorescence staining. The size and number of lipid droplets in mouse liver were examined by Oil Red O staining. Real-time quantitative reverse transcription polymerase chain reaction detected relative levels of adipogenesis-associated and lipolysis-associated genes in mouse liver. Additionally, expressions of CD36 protein in the mouse liver were determined by immunohistochemical staining and Western blot. RESULTS: After a 4-wk dietary intervention, FBG, FINS, and glucose area under the curve in the KD group became significantly lower than in the ND group (all P < 0.05). Regular morphology of mouse islets was observed in the KD group, with an increased number of islet cells. The KD significantly reversed the decrease in ß-cell number, disarrangement of ß-cells, decline of ß/α-cell ratio, and downregulation of ß-cell-specific transcription factors in db/db mice. Serum levels of triacylglycerol, total cholesterol, and low-density lipoprotein cholesterol were comparable between the ND and KD groups. In contrast, serum triacylglycerol levels were significantly lower in the CR group than in the ND group (P < 0.05). Vacuolar degeneration and lipid accumulation in the liver were more prominent in the KD group than in the ND and CR groups. The mRNA levels of Pparα and Acox1 in the KD group were lower than those in the ND group, although no significant differences were detected. Relative levels of Cd36 and inflammatory genes in the mouse liver were significantly higher in the KD group than in the ND group (all P < 0.05). CONCLUSION: The KD significantly reduced FBG and FINS and improved glucose tolerance in db/db mice by upregulating ß-cell-specific transcription factors and reversing ß-cell dedifferentiation. However, the KD also induced hepatic lipid accumulation and aggravated inflammatory response in the liver of db/db mice.


Asunto(s)
Dieta Cetogénica , Masculino , Ratones , Animales , Desdiferenciación Celular , Ratones Endogámicos C57BL , Hígado/metabolismo , Glucosa/metabolismo , Triglicéridos , Lípidos , Colesterol , Factores de Transcripción/metabolismo , Glucemia/metabolismo
10.
Artículo en Inglés | MEDLINE | ID: mdl-39133811

RESUMEN

CONTEXT: ß-cell dedifferentiation ratio is increased in type 2 diabetes; but its direct link to in vivo ß-cell function in human remains unclear. OBJECTIVE: The present study was designed to investigate whether ß-cell dedifferentiation in situ was closely associated with ß-cell function in vivo and to identify targets crucial for ß-cell dedifferentiation/function in human. METHODS: We acquired HOMA-ß values, calculated the number of hormone-negative endocrine cells and evaluated important markers and novel candidates for ß-cell dedifferentiation/function on paraneoplastic pancreatic tissues from 13 patients with benign pancreatic cystic neoplasm (PCN) or intrapancreatic accessory spleen. RESULTS: Both ß-cell dedifferentiation ratio and dedifferentiation marker (Aldh1a3) were inversely related with in vivo ß-cell function (HOMA-ß) and in situ ß-cell functional markers Glut2 and Ucn3 in human. Moreover, the islets from HOMA-ßlow subjects were manifested as 1) increased ß-cell dedifferentiation ratio, 2) enriched dedifferentiation maker Aldh1a3, and 3) lower expression of Glut2 and Ucn3, compared to those from HOMA-ßhigh subjects. We found that basic leucine zipper transcription factor 2 (Bach2) expression was significantly induced in islets from HOMA-ßlow patients and was positively correlated with the ratio of ß-cell dedifferentiation in human. CONCLUSIONS: Our findings emphasize the contribution of ß-cell dedifferentiation to ß-cell dysfunction in human. The Bach2 induction in ß-cells with higher frequency of dedifferentiation observed in HOMA-ßlow subjects reinforce its distinctive role as a pharmaceutical target of ß-cell dedifferentiation for the treatment of human diabetes.

11.
J Ethnopharmacol ; 321: 117481, 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38007164

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Modified Da Chaihu decoction (MDCH) is a traditional Chinese herbal prescription that has been used in the clinic to treat type 2 diabetes (T2D). Previous studies have confirmed that MDCH improves glycemic and lipid metabolism, enhances pancreatic function, and alleviates insulin resistance in patients with T2D and diabetic rats. Evidence has demonstrated that MDCH protects pancreatic ß cells via regulating the gene expression of sirtuin 1 (SIRT1) and forkhead box protein O1 (FOXO1). However, the detailed mechanism remains unclear. AIM OF THE STUDY: Dedifferentiation of pancreatic ß cells mediated by FOXO1 has been recognized as the main pathogenesis of T2D. This study aims to investigate the therapeutic effects of MDCH on T2D in vitro and in vivo to elucidate the potential molecular mechanisms. MATERIALS AND METHODS: To predict the key targets of MDCH in treating T2D, network pharmacology methods were used. A T2D model was induced in diet-induced obese (DIO) C57BL/6 mice with a single intraperitoneal injection of streptozotocin. Glucose metabolism indicators (oral glucose tolerance test, insulin tolerance test), lipid metabolism indicators (total cholesterol, triglyceride, low-density lipoprotein cholesterol, high-density lipoprotein cholesterol), inflammatory factors (C-reactive protein, interleukin 6, tumor necrosis factor alpha), oxidative stress indicators (total antioxidant capacity, superoxide dismutase, malondialdehyde), and hematoxylin and eosin staining were analyzed to evaluate the therapeutic effect of MDCH on T2D. Immunofluorescence staining and quantification of FOXO1, pancreatic and duodenal homeobox 1 (PDX1), NK6 homeobox 1 (NKX6.1), octamer-binding protein 4 (OCT4), neurogenin 3 (Ngn3), insulin, and SIRT1, and Western blot analysis of insulin, SIRT1, and FOXO1 were performed to investigate the mechanism by which MDCH inhibited pancreatic ß-cell dedifferentiation. RESULTS: The chemical ingredients identified in MDCH were predicted to be important for signaling pathways related to lipid metabolism and insulin resistance, including lipids in atherosclerosis, the advanced glycation end product receptor of the advanced glycation end product signaling pathway, and the FOXO signaling pathway. Experimental studies showed that MDCH improved glucose and lipid metabolism in T2D mice, alleviated inflammation and oxidative stress damage, and reduced pancreatic pathological damage. Furthermore, MDCH upregulated the expression levels of SIRT1, FOXO1, PDX1, and NKX6.1, while downregulating the expression levels of OCT4 and Ngn3, which indicated that MDCH inhibited pancreatic dedifferentiation of ß cells. CONCLUSIONS: MDCH has therapeutic effects on T2D, through regulating the SIRT1/FOXO1 signaling pathway to inhibit pancreatic ß-cell dedifferentiation, which has not been reported previously.


Asunto(s)
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2 , Resistencia a la Insulina , Células Secretoras de Insulina , Humanos , Ratas , Ratones , Animales , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Experimental/metabolismo , Desdiferenciación Celular , Sirtuina 1/metabolismo , Farmacología en Red , Ratones Endogámicos C57BL , Insulina/metabolismo , Colesterol/metabolismo
12.
Nutrients ; 16(7)2024 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-38613031

RESUMEN

In diabetes, pancreatic ß-cells gradually lose their ability to secrete insulin with disease progression. ß-cell dysfunction is a contributing factor to diabetes severity. Recently, islet cell heterogeneity, exemplified by ß-cell dedifferentiation and identified in diabetic animals, has attracted attention as an underlying molecular mechanism of ß-cell dysfunction. Previously, we reported ß-cell dedifferentiation suppression by calorie restriction, not by reducing hyperglycemia using hypoglycemic agents (including sodium-glucose cotransporter inhibitors), in an obese diabetic mice model (db/db). Here, to explore further mechanisms of the effects of food intake on ß-cell function, db/db mice were fed either a high-carbohydrate/low-fat diet (db-HC) or a low-carbohydrate/high-fat diet (db-HF) using similar calorie restriction regimens. After one month of intervention, body weight reduced, and glucose intolerance improved to a similar extent in the db-HC and db-HF groups. However, ß-cell dedifferentiation did not improve in the db-HC group, and ß-cell mass compensatory increase occurred in this group. More prominent fat accumulation occurred in the db-HC group livers. The expression levels of genes related to lipid metabolism, mainly regulated by peroxisome proliferator-activated receptor α and γ, differed significantly between groups. In conclusion, the fat/carbohydrate ratio in food during calorie restriction in obese mice affected both liver lipid metabolism and ß-cell dedifferentiation.


Asunto(s)
Restricción Calórica , Diabetes Mellitus Experimental , Animales , Ratones , Ratones Obesos , Dieta Alta en Grasa/efectos adversos , Desdiferenciación Celular , Dieta Baja en Carbohidratos , Hígado , Carbohidratos , Obesidad
13.
Biochim Biophys Acta Mol Basis Dis ; 1867(12): 166261, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34455055

RESUMEN

Rapamycin insensitive companion of mechanistic target of Rapamycin (Rictor), the key component of mTOR complex 2 (mTORC2), controls both ß-cell proliferation and function. We sought to study whether long chain acyl-CoA synthetase 4 (Acsl4) worked downstream of Rictor/mTORC2 to maintain ß-cell functional mass. We found Acsl4 was positively regulated by Rictor at transcriptional and posttranslational levels in mouse ß-cell. Infecting adenovirus expressing Acsl4 in ß-cell-specific-Rictor-knockout (ßRicKO) islets and Min6 cells knocking down Rictor with lentivirus-expressing siRNA-oligos targeting Rictor(siRic), recovered the ß-cell dysplasia but not dysfunction. Cell bioenergetic experiment performed with Seahorse XF showed that Acsl4 could not rescue the dampened glucose oxidation in Rictor-lacking ß-cell, but further promoted lipid oxidation. Transposase-Accessible Chromatin (ATAC) and H3K27Ac chromatin immunoprecipitation (ChIP) sequencing studies reflected the epigenetic elevated molecular signature for ß-cell dedifferentiation and mitigated oxidative defense/response. These results were confirmed by the observations of elevated acetylation and ubiquitination of FoxO1, increased protein levels of Gpx1 and Hif1an, excessive reactive oxygen species (ROS) production and diminished MafA in Acsl4 overexpressed Rictor-lacking ß-cells. In these cells, antioxidant treatment significantly recovered MafA level and insulin content. Inducing lipid oxidation alone could not mimic the effect of Acsl4 in Rictor lacking ß-cell. Our study suggested that Acsl4 function in ß-cell was context dependent and might facilitate ß-cell dedifferentiation with attenuated Rictor/mTORC2 activity or insulin signaling via posttranslational inhibiting FoxO1 and epigenetically enhancing ROS induced MafA degradation.


Asunto(s)
Desdiferenciación Celular/genética , Coenzima A Ligasas/genética , Proteína Forkhead Box O1/genética , Células Secretoras de Insulina/metabolismo , Proteína Asociada al mTOR Insensible a la Rapamicina/genética , Animales , Proliferación Celular/genética , Epigenómica , Regulación de la Expresión Génica/genética , Glutatión Peroxidasa/genética , Humanos , Insulina/genética , Insulina/metabolismo , Células Secretoras de Insulina/patología , Metabolismo de los Lípidos/genética , Diana Mecanicista del Complejo 2 de la Rapamicina/genética , Ratones , Oxigenasas de Función Mixta/genética , Especies Reactivas de Oxígeno/metabolismo , Glutatión Peroxidasa GPX1
14.
Nutrients ; 13(5)2021 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-34068827

RESUMEN

Diabetes is a metabolic disease characterized by hyperglycemia. Over 90% of patients with diabetes have type 2 diabetes. Pancreatic ß-cells are endocrine cells that produce and secrete insulin, an essential endocrine hormone that regulates blood glucose levels. Deficits in ß-cell function and mass play key roles in the onset and progression of type 2 diabetes. Apoptosis has been considered as the main contributor of ß-cell dysfunction and decrease in ß-cell mass for a long time. However, recent studies suggest that ß-cell failure occurs mainly due to increased ß-cell dedifferentiation rather than limited ß-cell proliferation or increased ß-cell death. In this review, we summarize the current advances in the understanding of the pancreatic ß-cell dedifferentiation process including potential mechanisms. A better understanding of ß-cell dedifferentiation process will help to identify novel therapeutic targets to prevent and/or reverse ß-cell loss in type 2 diabetes.


Asunto(s)
Desdiferenciación Celular/fisiología , Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina/fisiología , Animales , Apoptosis , Proliferación Celular , Citocinas , Diabetes Mellitus Tipo 2/patología , Estrés del Retículo Endoplásmico , Humanos , Inflamación , Insulina/metabolismo , MicroARNs/metabolismo , Estrés Oxidativo , ARN Largo no Codificante
15.
Stem Cell Res Ther ; 12(1): 449, 2021 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-34380570

RESUMEN

BACKGROUND: Mesenchymal stem cells (MSCs) show promising therapeutic potential in treating type 2 diabetes mellitus (T2DM) in clinical studies. Accumulating evidence has suggested that the therapeutic effects of MSCs are not due to their direct differentiation into functional ß-cells but are instead mediated by their paracrine functions. Among them, exosomes, nano-sized extracellular vesicles, are important substances that exert paracrine functions. However, the underlying mechanisms of exosomes in ameliorating T2DM remain largely unknown. METHODS: Bone marrow mesenchymal stem cell (bmMSC)-derived exosomes (bmMDEs) were administrated to T2DM rats and high-glucose-treated primary islets in order to detect their effects on ß-cell dedifferentiation. Differential miRNAs were then screened via miRNA sequencing, and miR-146a was isolated after functional verification. TargetScan, reporter gene detection, insulin secretion assays, and qPCR validation were used to predict downstream target genes and involved signaling pathways of miR-146a. RESULTS: Our results showed that bmMDEs reversed diabetic ß-cell dedifferentiation and improved ß-cell insulin secretion both in vitro and in vivo. Results of miRNA sequencing in bmMDEs and subsequent functional screening demonstrated that miR-146a, a highly conserved miRNA, improved ß-cell function. We further found that miR-146a directly targeted Numb, a membrane-bound protein involved in cell fate determination, leading to activation of ß-catenin signaling in ß-cells. Exosomes derived from miR-146a-knockdown bmMSCs lost the ability to improve ß-cell function. CONCLUSIONS: These findings demonstrate that bmMSC-derived exosomal miR-146a protects against diabetic ß-cell dysfunction by acting on the NUMB/ß-catenin signaling pathway, which may represent a novel therapeutic strategy for T2DM.


Asunto(s)
Diabetes Mellitus Tipo 2 , Exosomas , Células Madre Mesenquimatosas , MicroARNs , Animales , Desdiferenciación Celular , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/terapia , Exosomas/genética , MicroARNs/genética , Ratas
16.
Int Rev Cell Mol Biol ; 359: 357-402, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33832653

RESUMEN

Type 2 diabetes (T2D), a heterogeneous disorder derived from metabolic dysfunctions, leads to a glucose overflow in the circulation due to both defective insulin secretion and peripheral insulin resistance. One of the critical risk factor for T2D is obesity, which represents a global epidemic that has nearly tripled since 1975. Obesity is characterized by chronically elevated free fatty acid (FFA) levels, which cause deleterious effects on glucose homeostasis referred to as lipotoxicity. Here, we review the physiological FFA roles onto glucose-stimulated insulin secretion (GSIS) and the pathological ones affecting many steps of the mechanisms and modulation of GSIS. We also describe in vitro and in vivo experimental evidences addressing lipotoxicity in ß-cells and the role of saturation and chain length of FFA on the potency of GSIS stimulation. The molecular mechanisms underpinning lipotoxic-ß-cell dysfunction are also reviewed. Among them, endoplasmic reticulum stress, oxidative stress and mitochondrial dysfunction, inflammation, impaired autophagy and ß-cell dedifferentiation. Finally therapeutic strategies for the ß-cells dysfunctions such as the use of metformin, glucagon-like peptide 1, thiazolidinediones, anti-inflammatory drugs, chemical chaperones and weight are discussed.


Asunto(s)
Células Secretoras de Insulina/patología , Lípidos/toxicidad , Animales , Glucosa/metabolismo , Humanos , Secreción de Insulina/efectos de los fármacos , Células Secretoras de Insulina/efectos de los fármacos , Metaboloma/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
17.
Mol Metab ; 53: 101267, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34116231

RESUMEN

OBJECTIVE: The miR-200-Zeb1 axis regulates the epithelial-to-mesenchymal transition (EMT), differentiation, and resistance to apoptosis. A better understanding of these processes in diabetes is highly relevant, as ß-cell dedifferentiation and apoptosis contribute to the loss of functional ß-cell mass and diabetes progression. Furthermore, EMT promotes the loss of ß-cell identity in the in vitro expansion of human islets. Though the miR-200 family has previously been identified as a regulator of ß-cell apoptosis in vivo, studies focusing on Zeb1 are lacking. The aim of this study was thus to investigate the role of Zeb1 in ß-cell function and survival in vivo. METHODS: miR-200 and Zeb1 are involved in a double-negative feedback loop. We characterized a mouse model in which miR-200 binding sites in the Zeb1 3'UTR are mutated (Zeb1200), leading to a physiologically relevant upregulation of Zeb1 mRNA expression. The role of Zeb1 was investigated in this model via metabolic tests and analysis of isolated islets. Further insights into the distinct contributions of the miR-200 and Zeb1 branches of the feedback loop were obtained by crossing the Zeb1200 allele into a background of miR-141-200c overexpression. RESULTS: Mild Zeb1 derepression in vivo led to broad transcriptional changes in islets affecting ß-cell identity, EMT, insulin secretion, cell-cell junctions, the unfolded protein response (UPR), and the response to ER stress. The aggregation and insulin secretion of dissociated islets of mice homozygous for the Zeb1200 mutation (Zeb1200M) were impaired, and Zeb1200M islets were resistant to thapsigargin-induced ER stress ex vivo. Zeb1200M mice had increased circulating proinsulin levels but no overt metabolic phenotype, reflecting the strong compensatory ability of islets to maintain glucose homeostasis. CONCLUSIONS: This study signifies the importance of the miR-200-Zeb1 axis in regulating key aspects of ß-cell function and survival. A better understanding of this axis is highly relevant in developing therapeutic strategies for inducing ß-cell redifferentiation and maintaining ß-cell identity in in vitro islet expansion.


Asunto(s)
Células Secretoras de Insulina/metabolismo , MicroARNs/metabolismo , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo , Animales , Apoptosis , Células Cultivadas , Estrés del Retículo Endoplásmico , Secreción de Insulina , Ratones , Ratones Noqueados , MicroARNs/genética , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/deficiencia , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/genética
18.
Front Endocrinol (Lausanne) ; 11: 614123, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33613449

RESUMEN

Unfolded protein response (UPR) is a process conserved from yeasts to mammals and, based on the generally accepted dogma, helps the secretory performance of a cell, by improving its capacity to cope with a burden in the endoplasmic reticulum (ER). The ER of ß-cells, "professional secretory cells", has to manage tremendous amounts of insulin, which elicits a strong pressure on the ER intrinsic folding capacity. Thus, the constant demand for insulin production results in misfolded proinsulin, triggering a physiological upregulation of UPR to restore homeostasis. Most diabetic disorders are characterized by the loss of functional ß-cells, and the pathological side of UPR plays an instrumental role. The transition from a homeostatic to a pathological UPR that ultimately leads to insulin-producing ß-cell decay entails complex cellular processes and molecular mechanisms which remain poorly described so far. Here, we summarize important processes that are coupled with or driven by UPR in ß-cells, such as proliferation, inflammation and dedifferentiation. We conclude that the UPR comes in different "flavors" and each of them is correlated with a specific outcome for the cell, for survival, differentiation, proliferation as well as cell death. All these greatly depend on the way UPR is triggered, however what exactly is the switch that favors the activation of one UPR as opposed to others is largely unknown. Substantial work needs to be done to progress the knowledge in this important emerging field as this will help in the development of novel and more efficient therapies for diabetes.


Asunto(s)
Diabetes Mellitus/metabolismo , Estrés del Retículo Endoplásmico/fisiología , Células Secretoras de Insulina/metabolismo , Respuesta de Proteína Desplegada/fisiología , Animales , Diferenciación Celular/fisiología , Diabetes Mellitus/patología , Homeostasis/fisiología , Humanos , Células Secretoras de Insulina/patología
19.
Aging Dis ; 10(4): 719-730, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31440379

RESUMEN

A strategic approach is urgently needed to curb the growing global epidemic of diabetes. In this study, we investigated the effects and mechanisms of salsalate (SAL), an anti-inflammatory drug with anti-diabetic properties, assessing its potential to prevent diabetes in Otsuka Long-Evans Tokushima Fatty rats (OLETF). All animals in our placebo group developed diabetes, whereas none in the SAL test group did so, and only 25% of SAL-treated rats displayed impaired glucose tolerance (IGT). SAL lowered levels of glucagon and raised levels of insulin in plasma, while improving both insulin sensitivity and ß-cell function. The protective effect of SAL is likely due to diminished ß-cell dedifferentiation, manifested as relative declines in Neurogenin 3+/insulin- cells and synaptophysin+/islet hormone- cells and increased expression of ß-cell-specific transcription factor Foxo1. Both Notch1-siRNA and N-[N-(3,5-difluorophenacetyl)-1-alanyl]-S-phenylglycine t-butyl ester (DAPT; an indirect inhibitor of the Notch1 pathway) were shown to prevent ß-cell dedifferentiation. Similar to DAPT, SAL effectively reduced ß-cell dedifferentiation, significantly suppressing Notch1 pathway activation in INS-1 cells. The inhibitory role of SAL in ß-cell dedifferentiation may thus be attributable to Notch1 pathway suppression.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA