Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
J Diabetes Investig ; 15(6): 649-655, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38436511

RESUMEN

The ß-cell relies predominantly on glucose utilization to generate adenosine triphosphate, which is crucial for both cell viability and insulin secretion. The ß-cell has evolved remarkable metabolic flexibility to productively respond to shifts in environmental conditions and changes in glucose availability. Although these adaptive responses are important for maintaining optimal cellular function, there is emerging evidence that the resulting changes in cellular metabolites can impact the epigenome, causing transient and lasting alterations in gene expression. This review explores the intricate interplay between metabolism and the epigenome, providing valuable insights into the molecular mechanisms leading to ß-cell dysfunction in diabetes. Understanding these mechanisms will be critical for developing targeted therapeutic strategies to preserve and enhance ß-cell function, offering potential avenues for interventions to improve glycemic control in individuals with diabetes.


Asunto(s)
Glucosa , Células Secretoras de Insulina , Humanos , Células Secretoras de Insulina/metabolismo , Glucosa/metabolismo , Animales , Epigenómica , Epigénesis Genética , Epigenoma , Diabetes Mellitus/metabolismo , Diabetes Mellitus/genética
2.
Diabetes ; 73(3): 434-447, 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38015772

RESUMEN

Protein tyrosine phosphatase N2 (PTPN2) is a type 1 diabetes (T1D) candidate gene identified from human genome-wide association studies. PTPN2 is highly expressed in human and murine islets and becomes elevated upon inflammation and models of T1D, suggesting that PTPN2 may be important for ß-cell survival in the context of T1D. To test whether PTPN2 contributed to ß-cell dysfunction in an inflammatory environment, we generated a ß-cell-specific deletion of Ptpn2 in mice (PTPN2-ß knockout [ßKO]). Whereas unstressed animals exhibited normal metabolic profiles, low- and high-dose streptozotocin-treated PTPN2-ßKO mice displayed hyperglycemia and accelerated death, respectively. Furthermore, cytokine-treated Ptpn2-KO islets resulted in impaired glucose-stimulated insulin secretion, mitochondrial defects, and reduced glucose-induced metabolic flux, suggesting ß-cells lacking Ptpn2 are more susceptible to inflammatory stress associated with T1D due to maladaptive metabolic fitness. Consistent with the phenotype, proteomic analysis identified an important metabolic enzyme, ATP-citrate lyase, as a novel PTPN2 substrate.


Asunto(s)
Diabetes Mellitus Tipo 1 , Ratones , Humanos , Animales , Diabetes Mellitus Tipo 1/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 2/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 2/metabolismo , Monoéster Fosfórico Hidrolasas , Estudio de Asociación del Genoma Completo , Proteómica , Glucosa , Ratones Noqueados
3.
Nat Commun ; 14(1): 7732, 2023 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-38007492

RESUMEN

Insulin secretion is a tightly regulated process that is vital for maintaining blood glucose homeostasis. Although the molecular components of insulin granule trafficking and secretion are well established, how they are regulated to rapidly fine-tune secretion in response to changing environmental conditions is not well characterized. Recent studies have determined that dysregulation of RNA-binding proteins (RBPs) and aberrant mRNA splicing occurs at the onset of diabetes. We demonstrate that the RBP, RBFOX2, is a critical regulator of insulin secretion through the alternative splicing of genes required for insulin granule docking and exocytosis. Conditional mutation of Rbfox2 in the mouse pancreas results in decreased insulin secretion and impaired blood glucose homeostasis. Consistent with defects in secretion, we observe reduced insulin granule docking and corresponding splicing defects in the SNARE complex components. These findings identify an additional mechanism for modulating insulin secretion in both healthy and dysfunctional pancreatic ß cells.


Asunto(s)
Empalme Alternativo , Células Secretoras de Insulina , Ratones , Animales , Secreción de Insulina , Glucemia/metabolismo , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Exocitosis/fisiología , Factores de Empalme de ARN/genética , Factores de Empalme de ARN/metabolismo
5.
Mol Psychiatry ; 28(10): 4474-4484, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37648779

RESUMEN

Mitochondrial dysfunction has been implicated in Parkinson's Disease (PD) progression; however, the mitochondrial factors underlying the development of PD symptoms remain unclear. One candidate is CR6-interacting factor1 (CRIF1), which controls translation and membrane insertion of 13 mitochondrial proteins involved in oxidative phosphorylation. Here, we found that CRIF1 mRNA and protein expression were significantly reduced in postmortem brains of elderly PD patients compared to normal controls. To evaluate the effect of Crif1 deficiency, we produced mice lacking the Crif1 gene in dopaminergic neurons (DAT-CRIF1-KO mice). From 5 weeks of age, DAT-CRIF1-KO mice began to show decreased dopamine production with progressive neuronal degeneration in the nigral area. At ~10 weeks of age, they developed PD-like behavioral deficits, including gait abnormalities, rigidity, and resting tremor. L-DOPA, a medication used to treat PD, ameliorated these defects at an early stage, although it was ineffective in older mice. Taken together, the observation that CRIF1 expression is reduced in human PD brains and deletion of CRIF1 in dopaminergic neurons leads to early-onset PD with stepwise PD progression support the conclusion that CRIF1-mediated mitochondrial function is important for the survival of dopaminergic neurons.


Asunto(s)
Neuronas Dopaminérgicas , Enfermedad de Parkinson , Humanos , Ratones , Animales , Anciano , Neuronas Dopaminérgicas/metabolismo , Enfermedad de Parkinson/genética , Levodopa/farmacología , Dopamina/metabolismo , Encéfalo/metabolismo , Proteínas de Ciclo Celular/genética
6.
Mol Metab ; 66: 101632, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36347424

RESUMEN

OBJECTIVE: Zinc transporter 8 (ZnT8) is a major humoral target in human type 1 diabetes (T1D). Polymorphic variants of Slc30A8, which encodes ZnT8, are also associated with protection from type 2 diabetes (T2D). The current study examined whether ZnT8 might play a role beyond simply being a target of autoimmunity in the pathophysiology of T1D. METHODS: The phenotypes of NOD mice with complete or partial global loss of ZnT8 were determined using a combination of disease incidence, histological, transcriptomic, and metabolic analyses. RESULTS: Unexpectedly, while complete loss of ZnT8 accelerated spontaneous T1D, heterozygosity was partially protective. In vivo and in vitro studies of ZnT8 deficient NOD.SCID mice suggested that the accelerated disease was due to more rampant autoimmunity. Conversely, beta cells in heterozygous animals uniquely displayed increased mitochondrial fitness under mild proinflammatory conditions. CONCLUSIONS: In pancreatic beta cells and immune cell populations, Zn2+ plays a key role as a regulator of redox signaling and as an independent secondary messenger. Importantly, Zn2+ also plays a major role in maintaining mitochondrial homeostasis. Our results suggest that regulating mitochondrial fitness by altering intra-islet zinc homeostasis may provide a novel mechanism to modulate T1D pathophysiology.


Asunto(s)
Proteínas de Transporte de Catión , Diabetes Mellitus Tipo 1 , Diabetes Mellitus Tipo 2 , Humanos , Ratones , Animales , Transportador 8 de Zinc/genética , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Haploinsuficiencia/genética , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Ratones Endogámicos NOD , Ratones SCID , Respiración
7.
Exp Mol Med ; 54(7): 932-945, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35804190

RESUMEN

Genetic variations in mitoribosomal subunits and mitochondrial transcription factors are related to type 2 diabetes. However, the role of islet mitoribosomes in the development of type 2 diabetes has not been determined. We investigated the effects of the mitoribosomal gene on ß-cell function and glucose homeostasis. Mitoribosomal gene expression was analyzed in datasets from the NCBI GEO website (GSE25724, GSE76894, and GSE76895) and the European Nucleotide Archive (ERP017126), which contain the transcriptomes of type 2 diabetic and nondiabetic organ donors. We found deregulation of most mitoribosomal genes in islets from individuals with type 2 diabetes, including partial downregulation of CRIF1. The phenotypes of haploinsufficiency in a single mitoribosomal gene were examined using ß-cell-specific Crif1 (Mrpl59) heterozygous-deficient mice. Crif1beta+/- mice had normal glucose tolerance, but their islets showed a loss of first-phase glucose-stimulated insulin secretion. They also showed increased ß-cell mass associated with higher expression of Reg family genes. However, Crif1beta+/- mice showed earlier islet failure in response to high-fat feeding, which was exacerbated by aging. Haploinsufficiency of a single mitoribosomal gene predisposes rodents to glucose intolerance, which resembles the early stages of type 2 diabetes in humans.


Asunto(s)
Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Islotes Pancreáticos , Animales , Proteínas de Ciclo Celular/metabolismo , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Glucosa/metabolismo , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Ratones , Ribosomas Mitocondriales/metabolismo
8.
Biomolecules ; 11(4)2021 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-33808310

RESUMEN

The pancreatic beta cell is a highly specialized cell type whose primary function is to secrete insulin in response to nutrients to maintain glucose homeostasis in the body. As such, the beta cell has developed unique metabolic characteristics to achieve functionality; in healthy beta cells, the majority of glucose-derived carbons are oxidized and enter the mitochondria in the form of pyruvate. The pyruvate is subsequently metabolized to induce mitochondrial ATP and trigger the downstream insulin secretion response. Thus, in beta cells, mitochondria play a pivotal role in regulating glucose stimulated insulin secretion (GSIS). In type 2 diabetes (T2D), mitochondrial impairment has been shown to play an important role in beta cell dysfunction and loss. In type 1 diabetes (T1D), autoimmunity is the primary trigger of beta cell loss; however, there is accumulating evidence that intrinsic mitochondrial defects could contribute to beta cell susceptibility during proinflammatory conditions. Furthermore, there is speculation that dysfunctional mitochondrial responses could contribute to the formation of autoantigens. In this review, we provide an overview of mitochondrial function in the beta cells, and discuss potential mechanisms by which mitochondrial dysfunction may contribute to T1D pathogenesis.


Asunto(s)
Autoinmunidad/fisiología , Diabetes Mellitus Tipo 1/patología , Células Secretoras de Insulina/patología , Mitocondrias/metabolismo , Animales , Autofagia , Senescencia Celular , Diabetes Mellitus Tipo 1/inmunología , Epítopos , Humanos , Secreción de Insulina , Células Secretoras de Insulina/inmunología , Mitocondrias/patología , Mitofagia
9.
Diabetologia ; 63(4): 837-852, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31925461

RESUMEN

AIMS/HYPOTHESIS: Mitochondrial oxidative phosphorylation (OxPhos) is essential for energy production and survival. However, the tissue-specific and systemic metabolic effects of OxPhos function in adipocytes remain incompletely understood. METHODS: We used adipocyte-specific Crif1 (also known as Gadd45gip1) knockout (AdKO) mice with decreased adipocyte OxPhos function. AdKO mice fed a normal chow or high-fat diet were evaluated for glucose homeostasis, weight gain and energy expenditure (EE). RNA sequencing of adipose tissues was used to identify the key mitokines affected in AdKO mice, which included fibroblast growth factor 21 (FGF21) and growth differentiation factor 15 (GDF15). For in vitro analysis, doxycycline was used to pharmacologically decrease OxPhos in 3T3L1 adipocytes. To identify the effects of GDF15 and FGF21 on the metabolic phenotype of AdKO mice, we generated AdKO mice with global Gdf15 knockout (AdGKO) or global Fgf21 knockout (AdFKO). RESULTS: Under high-fat diet conditions, AdKO mice were resistant to weight gain and exhibited higher EE and improved glucose tolerance. In vitro pharmacological and in vivo genetic inhibition of OxPhos in adipocytes significantly upregulated mitochondrial unfolded protein response-related genes and secretion of mitokines such as GDF15 and FGF21. We evaluated the metabolic phenotypes of AdGKO and AdFKO mice, revealing that GDF15 and FGF21 differentially regulated energy homeostasis in AdKO mice. Both mitokines had beneficial effects on obesity and insulin resistance in the context of decreased adipocyte OxPhos, but only GDF15 regulated EE in AdKO mice. CONCLUSIONS/INTERPRETATION: The present study demonstrated that the adipose tissue adaptive mitochondrial stress response affected systemic energy homeostasis via cell-autonomous and non-cell-autonomous pathways. We identified novel roles for adipose OxPhos and adipo-mitokines in the regulation of systemic glucose homeostasis and EE, which facilitated adaptation of an organism to local mitochondrial stress.


Asunto(s)
Adipocitos/metabolismo , Proteínas de Ciclo Celular/genética , Metabolismo Energético/genética , Obesidad/genética , Adipocitos/patología , Animales , Proteínas de Ciclo Celular/metabolismo , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Obesos , Obesidad/metabolismo , Obesidad/prevención & control , Especificidad de Órganos/genética , Fosforilación Oxidativa
10.
Diabetes ; 69(3): 355-368, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31848151

RESUMEN

Loss of functional ß-cell mass is an essential feature of type 2 diabetes, and maintaining mature ß-cell identity is important for preserving a functional ß-cell mass. However, it is unclear how ß-cells achieve and maintain their mature identity. Here we demonstrate a novel function of protein arginine methyltransferase 1 (PRMT1) in maintaining mature ß-cell identity. Prmt1 knockout in fetal and adult ß-cells induced diabetes, which was aggravated by high-fat diet-induced metabolic stress. Deletion of Prmt1 in adult ß-cells resulted in the immediate loss of histone H4 arginine 3 asymmetric dimethylation (H4R3me2a) and the subsequent loss of ß-cell identity. The expression levels of genes involved in mature ß-cell function and identity were robustly downregulated as soon as Prmt1 deletion was induced in adult ß-cells. Chromatin immunoprecipitation sequencing and assay for transposase-accessible chromatin sequencing analyses revealed that PRMT1-dependent H4R3me2a increases chromatin accessibility at the binding sites for CCCTC-binding factor (CTCF) and ß-cell transcription factors. In addition, PRMT1-dependent open chromatin regions may show an association with the risk of diabetes in humans. Together, our results indicate that PRMT1 plays an essential role in maintaining ß-cell identity by regulating chromatin accessibility.


Asunto(s)
Cromatina/metabolismo , Regulación de la Expresión Génica , Intolerancia a la Glucosa/genética , Código de Histonas/genética , Histonas/metabolismo , Secreción de Insulina/genética , Células Secretoras de Insulina/metabolismo , Proteína-Arginina N-Metiltransferasas/genética , Animales , Factor de Unión a CCCTC/metabolismo , Diferenciación Celular/genética , Secuenciación de Inmunoprecipitación de Cromatina , Regulación hacia Abajo , Técnicas de Inactivación de Genes , Metilación , Ratones , Ratones Noqueados , RNA-Seq
11.
Nat Commun ; 9(1): 1551, 2018 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-29674655

RESUMEN

Oxidative functions of adipose tissue macrophages control the polarization of M1-like and M2-like phenotypes, but whether reduced macrophage oxidative function causes systemic insulin resistance in vivo is not clear. Here, we show that mice with reduced mitochondrial oxidative phosphorylation (OxPhos) due to myeloid-specific deletion of CR6-interacting factor 1 (Crif1), an essential mitoribosomal factor involved in biogenesis of OxPhos subunits, have M1-like polarization of macrophages and systemic insulin resistance with adipose inflammation. Macrophage GDF15 expression is reduced in mice with impaired oxidative function, but induced upon stimulation with rosiglitazone and IL-4. GDF15 upregulates the oxidative function of macrophages, leading to M2-like polarization, and reverses insulin resistance in ob/ob mice and HFD-fed mice with myeloid-specific deletion of Crif1. Thus, reduced macrophage oxidative function controls systemic insulin resistance and adipose inflammation, which can be reversed with GDF15 and leads to improved oxidative function of macrophages.


Asunto(s)
Resistencia a la Insulina , Macrófagos/metabolismo , Obesidad/metabolismo , Fosforilación Oxidativa , Tejido Adiposo , Animales , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Factor 15 de Diferenciación de Crecimiento/genética , Factor 15 de Diferenciación de Crecimiento/metabolismo , Interleucina-4/genética , Interleucina-4/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Obesidad/genética , Estrés Oxidativo
12.
Diabetes ; 66(11): 2774-2788, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28874416

RESUMEN

T-helper type 2 (Th2) cytokines, including interleukin (IL)-13 and IL-4, produced in adipose tissue, are critical regulators of intra-adipose and systemic lipid and glucose metabolism. Furthermore, IL-13 is a potential therapy for insulin resistance in obese mouse models. Here, we examined mediators produced by adipocytes that are responsible for regulating systemic glucose homeostasis in response to Th2 cytokines. We used RNA sequencing data analysis of cultured adipocytes to screen factors secreted in response to recombinant IL-13. Recombinant IL-13 induced expression of growth differentiation factor 15 (GDF15) via the Janus kinase-activated STAT6 pathway. In vivo administration of α-galactosylceramide or IL-33 increased IL-4 and IL-13 production, thereby increasing GDF15 levels in adipose tissue and in plasma of mice; however, these responses were abrogated in STAT6 knockout mice. Moreover, administration of recombinant IL-13 to wild-type mice fed a high-fat diet (HFD) improved glucose intolerance; this was not the case for GDF15 knockout mice fed the HFD. Taken together, these data suggest that GDF15 is required for IL-13-induced improvement of glucose intolerance in mice fed an HFD. Thus, beneficial effects of Th2 cytokines on systemic glucose metabolism and insulin sensitivity are mediated by GDF15. These findings open up a potential pharmacological route for reversing insulin resistance associated with obesity.


Asunto(s)
Glucemia/fisiología , Glucosa/metabolismo , Factor 15 de Diferenciación de Crecimiento/metabolismo , Células Th2/fisiología , Células 3T3-L1 , Animales , Dieta Alta en Grasa , Intolerancia a la Glucosa , Factor 15 de Diferenciación de Crecimiento/genética , Interleucina-13/genética , Interleucina-13/metabolismo , Interleucina-13/fisiología , Interleucina-33/administración & dosificación , Interleucina-33/farmacología , Interleucina-4/genética , Interleucina-4/metabolismo , Interleucina-4/fisiología , Quinasas Janus/genética , Quinasas Janus/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Proteína Disulfuro Reductasa (Glutatión) , Interferencia de ARN , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Proteínas Recombinantes/farmacología , Factor de Transcripción STAT6/genética , Factor de Transcripción STAT6/metabolismo
13.
J Endocrinol ; 233(1): 105-118, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28184000

RESUMEN

Recent studies revealed that the inhibition of mitochondrial oxidative phosphorylation (OXPHOS) is coupled with the mitochondrial unfolded protein response, thereby stimulating the secretion of non-cell autonomous factors, which may control systemic energy metabolism and longevity. However, the nature and roles of non-cell autonomous factors induced in adipose tissue in response to reduced OXPHOS function remain to be clarified in mammals. CR6-interacting factor 1 (CRIF1) is an essential mitoribosomal protein for the intramitochondrial production of mtDNA-encoded OXPHOS subunits. Deficiency of CRIF1 impairs the proper formation of the OXPHOS complex, resulting in reduced function. To determine which secretory factors are induced in response to reduced mitochondrial OXPHOS function, we analyzed gene expression datasets in Crif1-depleted mouse embryonic fibroblasts. Crif1 deficiency preferentially increased the expression of angiopoietin-like 6 (Angptl6) and did not affect other members of the ANGPTL family. Moreover, treatment with mitochondrial OXPHOS inhibitors increased the expression of Angptl6 in cultured adipocytes. To confirm Angptl6 induction in vivo, we generated a murine model of reduced mitochondrial OXPHOS function using adipose tissue-specific Crif1-deficient mice and verified the upregulation of Angptl6 and fibroblast growth factor 21 (Fgf21) in white adipose tissue. Treatment with recombinant ANGPTL6 protein increased oxygen consumption and Pparα expression through the extracellular signal-regulated kinase/mitogen-activated protein kinase pathway in cultured adipocytes. Furthermore, the ANGPTL6-mediated increase in Pparα expression resulted in increased FGF21 expression, thereby promoting ß-oxidation. In conclusion, mitochondrial OXPHOS function governs the expression of ANGPTL6, which is an essential factor for FGF21 production in adipose tissue and cultured adipocytes.


Asunto(s)
Tejido Adiposo/metabolismo , Angiopoyetinas/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Mitocondrias/metabolismo , Células 3T3-L1 , Adipocitos/metabolismo , Proteína 6 similar a la Angiopoyetina , Proteínas Similares a la Angiopoyetina , Angiopoyetinas/genética , Animales , Fibroblastos/metabolismo , Hepatocitos/metabolismo , Ratones , Ratones Transgénicos , Fosforilación Oxidativa , Consumo de Oxígeno/fisiología
14.
J Cell Biol ; 216(1): 149-165, 2017 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-27986797

RESUMEN

Reduced mitochondrial electron transport chain activity promotes longevity and improves energy homeostasis via cell-autonomous and -non-autonomous factors in multiple model systems. This mitohormetic effect is thought to involve the mitochondrial unfolded protein response (UPRmt), an adaptive stress-response pathway activated by mitochondrial proteotoxic stress. Using mice with skeletal muscle-specific deficiency of Crif1 (muscle-specific knockout [MKO]), an integral protein of the large mitoribosomal subunit (39S), we identified growth differentiation factor 15 (GDF15) as a UPRmt-associated cell-non-autonomous myomitokine that regulates systemic energy homeostasis. MKO mice were protected against obesity and sensitized to insulin, an effect associated with elevated GDF15 secretion after UPRmt activation. In ob/ob mice, administration of recombinant GDF15 decreased body weight and improved insulin sensitivity, which was attributed to elevated oxidative metabolism and lipid mobilization in the liver, muscle, and adipose tissue. Thus, GDF15 is a potent mitohormetic signal that safeguards against the onset of obesity and insulin resistance.


Asunto(s)
Tejido Adiposo/metabolismo , Metabolismo Energético , Factor 15 de Diferenciación de Crecimiento/metabolismo , Hígado/efectos de los fármacos , Músculo Esquelético/metabolismo , Obesidad/metabolismo , Células 3T3-L1 , Tejido Adiposo/efectos de los fármacos , Animales , Proteínas de Ciclo Celular/deficiencia , Proteínas de Ciclo Celular/genética , Metabolismo Energético/efectos de los fármacos , Predisposición Genética a la Enfermedad , Factor 15 de Diferenciación de Crecimiento/deficiencia , Factor 15 de Diferenciación de Crecimiento/genética , Factor 15 de Diferenciación de Crecimiento/farmacología , Homeostasis , Resistencia a la Insulina , Leptina/deficiencia , Leptina/genética , Lipólisis , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Obesos , Mitocondrias Hepáticas/metabolismo , Mitocondrias Musculares/metabolismo , Músculo Esquelético/efectos de los fármacos , Obesidad/genética , Obesidad/prevención & control , Oxidación-Reducción , Fosforilación Oxidativa , Fenotipo , Interferencia de ARN , Proteínas Recombinantes/farmacología , Transducción de Señal , Factores de Tiempo , Factor de Transcripción CHOP/genética , Factor de Transcripción CHOP/metabolismo , Transfección , Respuesta de Proteína Desplegada , Aumento de Peso
15.
J Hepatol ; 66(1): 132-141, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27663419

RESUMEN

BACKGROUND & AIMS: To date, no pharmacological therapy has been approved for non-alcoholic fatty liver disease (NAFLD). The aim of the present study was to evaluate the therapeutic potential of poly ADP-ribose polymerase (PARP) inhibitors in mouse models of NAFLD. METHODS: As poly ADP-ribosylation (PARylation) of proteins by PARPs consumes nicotinamide adenine dinucleotide (NAD+), we hypothesized that overactivation of PARPs drives NAD+ depletion in NAFLD. Therefore, we assessed the effectiveness of PARP inhibition to replenish NAD+ and activate NAD+-dependent sirtuins, hence improving hepatic fatty acid oxidation. To do this, we examined the preventive and therapeutic benefits of the PARP inhibitor (PARPi), olaparib, in different models of NAFLD. RESULTS: The induction of NAFLD in C57BL/6J mice using a high-fat high-sucrose (HFHS)-diet increased PARylation of proteins by PARPs. As such, increased PARylation was associated with reduced NAD+ levels and mitochondrial function and content, which was concurrent with elevated hepatic lipid content. HFHS diet supplemented with PARPi reversed NAFLD through repletion of NAD+, increasing mitochondrial biogenesis and ß-oxidation in liver. Furthermore, PARPi reduced reactive oxygen species, endoplasmic reticulum stress and fibrosis. The benefits of PARPi treatment were confirmed in mice fed with a methionine- and choline-deficient diet and in mice with lipopolysaccharide-induced hepatitis; PARP activation was attenuated and the development of hepatic injury was delayed in both models. Using Sirt1hep-/- mice, the beneficial effects of a PARPi-supplemented HFHS diet were found to be Sirt1-dependent. CONCLUSIONS: Our study provides a novel and practical pharmacological approach for treating NAFLD, fueling optimism for potential clinical studies. LAY SUMMARY: Non-alcoholic fatty liver disease (NAFLD) is now considered to be the most common liver disease in the Western world and has no approved pharmacological therapy. PARP inhibitors given as a treatment in two different mouse models of NAFLD confer a protection against its development. PARP inhibitors may therefore represent a novel and practical pharmacological approach for treating NAFLD.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico , Ftalazinas/farmacología , Piperazinas/farmacología , Animales , Modelos Animales de Enfermedad , Metabolismo de los Lípidos , Hígado/metabolismo , Hígado/patología , Ratones , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Oxidación-Reducción , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Poli(ADP-Ribosa) Polimerasas/metabolismo
16.
Diabetologia ; 58(4): 771-80, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25660120

RESUMEN

AIM/HYPOTHESIS: Although mitochondrial oxidative phosphorylation (OxPhos) dysfunction is believed to be responsible for beta cell dysfunction in insulin resistance and mitochondrial diabetes, the mechanisms underlying progressive beta cell failure caused by defective mitochondrial OxPhos are largely unknown. METHODS: We examined the in vivo phenotypes of beta cell dysfunction in beta cell-specific Crif1 (also known as Gadd45gip1)-deficient mice. CR6-interacting factor-1 (CRIF1) is a mitochondrial protein essential for the synthesis and formation of the OxPhos complex in the inner mitochondrial membrane. RESULTS: Crif1(beta-/-) mice exhibited impaired glucose tolerance with defective insulin secretion as early as 4 weeks of age without defects in islet structure. At 11 weeks of age, Crif1(beta-/-) mice displayed characteristic ultrastructural mitochondrial abnormalities as well as severe glucose intolerance. Furthermore, islet area and insulin content was decreased by approximately 50% compared with wild-type mice. Treatment with the glucoregulatory drug exenatide, a glucagon-like peptide-1 (GLP-1) agonist, was not sufficient to preserve beta cell function in Crif1(beta-/-) mice. CONCLUSIONS/INTERPRETATION: Our results indicate that mitochondrial OxPhos dysfunction triggers progressive beta cell failure that is not halted by treatment with a GLP-1 agonist. The Crif1(beta-/-) mouse is a useful model for the study of beta cell failure caused by mitochondrial OxPhos dysfunction.


Asunto(s)
Proteínas de Ciclo Celular/deficiencia , Diabetes Mellitus/metabolismo , Células Secretoras de Insulina/metabolismo , Mitocondrias/metabolismo , Fosforilación Oxidativa , Factores de Edad , Animales , Autofagia , Glucemia/metabolismo , Proteínas de Ciclo Celular/genética , Línea Celular , Diabetes Mellitus/tratamiento farmacológico , Diabetes Mellitus/genética , Diabetes Mellitus/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Exenatida , Genotipo , Receptor del Péptido 1 Similar al Glucagón/agonistas , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Hipoglucemiantes/farmacología , Incretinas/farmacología , Insulina/sangre , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/ultraestructura , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/efectos de los fármacos , Mitocondrias/ultraestructura , Péptidos/farmacología , Fenotipo , Factores de Tiempo , Ponzoñas/farmacología
17.
Endocrinology ; 156(3): 1181-93, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25555091

RESUMEN

Adult thyroid dysfunction is a common endocrine disorder associated with an increased risk of cardiovascular disease and mortality. A recent epidemiologic study revealed a link between obesity and increased prevalence of hypothyroidism. It is conceivable that excessive adiposity in obesity might lead to expansion of the interfollicular adipose (IFA) depot or steatosis in thyroid follicular cells (thyroid steatosis, TS). In this study, we investigated the morphological and functional changes in thyroid glands of obese humans and animal models, diet-induced obese (DIO), ob/ob, and db/db mice. Expanded IFA depot and TS were observed in obese patients. Furthermore, DIO mice showed increased expression of lipogenesis-regulation genes, such as sterol regulatory element binding protein 1 (SREBP-1), peroxisome proliferator-activated receptor γ (PPARγ), acetyl coenzyme A carboxylase (ACC), and fatty acid synthetase (FASN) in the thyroid gland. Steatosis and ultrastructural changes, including distension of the endoplasmic reticulum (ER) and mitochondrial distortion in thyroid follicular cells, were uniformly observed in DIO mice and genetically obese mouse models, ob/ob and db/db mice. Obese mice displayed a variable degree of primary thyroid hypofunction, which was not corrected by PPARγ agonist administration. We propose that systemically increased adiposity is associated with characteristic IFA depots and TS and may cause or influence the development of primary thyroid failure.


Asunto(s)
Tejido Adiposo/patología , Hipotiroidismo/patología , Obesidad/metabolismo , Glándula Tiroides/citología , Glándula Tiroides/patología , Animales , Grasas de la Dieta/efectos adversos , Humanos , Hipotiroidismo/metabolismo , Lípidos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Obesos
18.
Liver Int ; 35(4): 1341-53, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25443620

RESUMEN

BACKGROUND & AIMS: Nonalcoholic steatohepatitis (NASH) is associated with cirrhosis and hepatocellular carcinoma. Reactive oxygen species (ROS) and reactive nitrogen species (RNS) play key roles in the development of the disease. However, the therapeutic target of NASH has not been fully defined and new treatments are needed. We investigated the protective effects of the antioxidant indole-derived NecroX-7 in a NASH mouse model using leptin-deficient ob/ob and methionine- and choline-deficient (MCD) diet-fed ob/ob mice. METHODS: Six-week-old male mice were divided into three groups: ob/+ mice, ob/ob mice treated with vehicle and ob/ob mice treated daily with NecroX-7 (20 mg/kg) for 4 weeks. To study the effects of NecroX-7 in a fibrosis model, NASH was induced by feeding ob/ob mice an MCD diet. The effects of NecroX-7 on NASH progression were evaluated using biochemical, histological and molecular markers. RESULTS: NecroX-7-treated ob/ob mice had a marked decrease in serum aspartate aminotransferase and alanine transaminase compared with vehicle-treated controls. Interestingly, hepatic steatosis and lipid peroxidation were significantly improved by NecroX-7 treatment. NecroX-7 inhibited tert-butylhydroperoxide- and H2 O2 -induced mitochondrial ROS/RNS in primary hepatocytes and attenuated mitochondrial dysfunction in vitro and in vivo. Furthermore, NecroX-7-treated mice exhibited fewer infiltrating macrophages and reduced hepatic tumour necrosis factor-alpha expression. Hepatic fibrosis in MCD-fed ob/ob mice was significantly decreased by NecroX-7 treatment. CONCLUSIONS: NecroX-7 treatment improved hepatic steatosis and fibrosis in murine NASH models. These effects occurred through the suppression of whole-cell ROS/RNS and inflammatory responses and suggest that NecroX-7 has a potential therapeutic benefit in steatohepatitis.


Asunto(s)
Antioxidantes/farmacología , Inflamación/tratamiento farmacológico , Hígado/efectos de los fármacos , Mitocondrias Hepáticas/efectos de los fármacos , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Compuestos Orgánicos/farmacología , Estrés Oxidativo/efectos de los fármacos , Especies de Nitrógeno Reactivo/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Citoprotección , Metabolismo Energético/efectos de los fármacos , Células Hep G2 , Humanos , Inflamación/metabolismo , Inflamación/patología , Mediadores de Inflamación/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Cirrosis Hepática Experimental/tratamiento farmacológico , Cirrosis Hepática Experimental/metabolismo , Cirrosis Hepática Experimental/patología , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones Obesos , Mitocondrias Hepáticas/metabolismo , Mitocondrias Hepáticas/patología , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
19.
Yonsei Med J ; 54(5): 1158-67, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23918565

RESUMEN

PURPOSE: NAD(P)H:Quinone Oxidoreductase 1 (NQO1) C609T missense variant (NQO1*2) and 29 basepair (bp)-insertion/deletion (I29/D) polymorphism of the NRH:Quinone Oxidoreductase 2 (NQO2) gene promoter have been proposed as predictive and prognostic factors for cancer development and progression. The purpose of this study is to investigate the relationship between NQO1/NQO2 genotype and clinico-pathological features of papillary thyroid microcarcinoma (PTMC). MATERIALS AND METHODS: Genomic DNA was isolated from 243 patients; and clinical data were retrospectively analyzed. NQO1*2 and tri-allelic polymorphism of NQO2 were investigated by polymerase chain reaction (PCR) and restriction fragment length polymorphism (RFLP) analysis. RESULTS: PTMC with NQO1*2 frequently exhibited extra-thyroidal extension as compared to PTMC with wild-type NQO1 (p=0.039). There was a significant relationship between I29/I29 homozygosity of NQO2 and lymph node metastasis (p=0.042). Multivariate analysis showed that the I29/I29 genotype was associated with an increased risk of lymph node metastasis (OR, 2.24; 95% CI, 1.10-4.56; p=0.026). CONCLUSION: NQO1*2 and I29 allele of the NQO2 are associated with aggressive clinical phenotypes of PTMC, and the I29 allele represents a putative prognostic marker for PTMC.


Asunto(s)
Carcinoma Papilar/genética , NAD(P)H Deshidrogenasa (Quinona)/genética , Neoplasias de la Tiroides/genética , Adulto , Carcinoma Papilar/patología , Análisis Mutacional de ADN , Femenino , Predisposición Genética a la Enfermedad , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Análisis Multivariante , Mutagénesis Insercional , Mutación Missense , NAD(P)H Deshidrogenasa (Quinona)/química , Fenotipo , Polimorfismo Genético , Pronóstico , Regiones Promotoras Genéticas , Estudios Retrospectivos , Análisis de Secuencia de Proteína , Eliminación de Secuencia , Neoplasias de la Tiroides/patología
20.
J Ethnopharmacol ; 148(2): 505-14, 2013 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-23665312

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Myelophil is composed of Astragali Radix and Salviae Miltiorrhizae Radix, according to the long traditional pharmacological practices, and it has been used for patients with chronic fatigue-associated symptoms including concentration problem or memory loss. AIM OF THE STUDY: This study aimed to evaluate the clinical relevance of Myelophil on brain oxidative damage using a chronic cold stress mice model. MATERIAL AND METHODS: Balb/c mice were subjected to cold stress (4°C for 4h) six times per week for 2 weeks with or without oral administration of Myelophil (50, 100, or 200mg/kg), or ascorbic acid (50mg/kg). RESULTS: Chronic cold stress induced histopathological hippocampal apoptosis with drastically increased serum levels of total reactive oxygen species and nitric oxide, as well as brain lipid peroxidation levels, protein carbonyl, and caspase-3/7 activity. These alterations were significantly ameliorated by Myelophil treatment. Myelophil administration significantly recovered the depleted glutathione and its enzymes, superoxide dismutase activity, and catalase protein and gene expression levels. Serum levels of corticosterone, dopamine, and adrenaline were notably altered by chronic cold stress but were significantly ameliorated by Myelophil treatment. Myelophil also normalized alterations in tumor necrosis factor-α, interleukin (IL)-1ß, and IL-10 gene expression and protein levels. Chronic cold stress up-regulated gene expression levels of phenylethanolamine N-methyltransferase and monoamine oxidase-B, and glucocorticoid receptors in the hypothalamus and hippocampus, respectively, whereas Myelophil treatment completely normalized these levels. CONCLUSIONS: These results suggest that Myelophil has potent pharmaceutical effects against chronic cold-stress-induced brain damage by relieving oxidative stress and inflammation and regulating stress hormones in mice.


Asunto(s)
Lesiones Encefálicas/tratamiento farmacológico , Medicamentos Herbarios Chinos/farmacología , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Sistema Hipófiso-Suprarrenal/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Ácido Ascórbico/farmacología , Lesiones Encefálicas/sangre , Lesiones Encefálicas/metabolismo , Caspasa 3/metabolismo , Caspasa 7/metabolismo , Corticosterona/sangre , Modelos Animales de Enfermedad , Dopamina/sangre , Epinefrina/sangre , Glutatión/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Sistema Hipotálamo-Hipofisario/metabolismo , Interleucina-10/metabolismo , Interleucina-1beta/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos BALB C , Monoaminooxidasa/metabolismo , Óxido Nítrico/metabolismo , Feniletanolamina N-Metiltransferasa/metabolismo , Sistema Hipófiso-Suprarrenal/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptores de Glucocorticoides/metabolismo , Superóxido Dismutasa/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA