Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Metabolomics ; 18(3): 15, 2022 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-35179657

RESUMEN

INTRODUCTION: Patients with hepatocyte nuclear factor-1 beta (HNF1B) mutations present a variable phenotype with two main symptoms: maturity onset diabetes of the young (MODY) and polycystic kidney disease (PKD). OBJECTIVES: Identification of serum metabolites specific for HNF1Bmut and evaluation of their role in disease pathogenesis. METHODS: We recruited patients with HNF1Bmut (N = 10), HNF1Amut (N = 10), PKD: non-dialyzed and dialyzed (N = 8 and N = 13); and healthy controls (N = 12). Serum fingerprinting was performed by LC-QTOF-MS. Selected metabolite was validated by ELISA (enzyme-linked immunosorbent assay) measurements and then biologically connected with HNF1B by in silico analysis. HepG2 were stimulated with lysophosphatidic acid (LPA) and HNF1B gene was knocked down (kd) by small interfering RNA. Transcriptomic analysis with microarrays and western blot measurements were performed. RESULTS: Serum levels of six metabolites including: arachidonic acid, hydroxyeicosatetraenoic acid, linoleamide and three LPA (18:1, 18:2 and 20:4), had AUC (the area under the curve) > 0.9 (HNF1Bmut vs comparative groups). The increased level of LPA was confirmed by ELISA measurements. In HepG2HNF1Bkd cells LPA stimulation lead to downregulation of many pathways associated with cell cycle, lipid metabolism, and upregulation of steroid hormone metabolism and Wnt signaling. Also, increased intracellular protein level of autotaxin was detected in the cells. GSK-3alpha/beta protein level and its phosphorylated ratio were differentially affected by LPA stimulation in HNF1Bkd and control cells. CONCLUSIONS: LPA is elevated in sera of patients with HNF1Bmut. LPA contributes to the pathogenesis of HNF1B-MODY by affecting Wnt/GSK-3 signaling.


Asunto(s)
Glucógeno Sintasa Quinasa 3 , Enfermedades Renales Quísticas , Glucógeno Sintasa Quinasa 3/genética , Factor Nuclear 1-beta del Hepatocito/genética , Humanos , Lisofosfolípidos , Metabolómica , Mutación/genética
2.
Int J Mol Sci ; 22(13)2021 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-34202179

RESUMEN

The progression of non-alcoholic fatty liver (NAFL) into non-alcoholic steatohepatitis implicates multiple mechanisms, chief of which is mitochondrial dysfunction. However, the sequence of events underlying mitochondrial failure are still poorly clarified. In this work, male C57BL/6J mice were fed with a high-fat plus high-sucrose diet for 16, 20, 22, and 24 weeks to induce NAFL. Up to the 20th week, an early mitochondrial remodeling with increased OXPHOS subunits levels and higher mitochondrial respiration occurred. Interestingly, a progressive loss of mitochondrial respiration along "Western diet" feeding was identified, accompanied by higher susceptibility to mitochondrial permeability transition pore opening. Importantly, our findings prove that mitochondrial alterations and subsequent impairment are independent of an excessive mitochondrial reactive oxygen species (ROS) generation, which was found to be progressively diminished along with disease progression. Instead, increased peroxisomal abundance and peroxisomal fatty acid oxidation-related pathway suggest that peroxisomes may contribute to hepatic ROS generation and oxidative damage, which may accelerate hepatic injury and disease progression. We show here for the first time the sequential events of mitochondrial alterations involved in non-alcoholic fatty liver disease (NAFLD) progression and demonstrate that mitochondrial ROS are not one of the first hits that cause NAFLD progression.


Asunto(s)
Mitocondrias/metabolismo , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Antioxidantes/metabolismo , Autofagia , Ésteres del Colesterol/metabolismo , Biología Computacional/métodos , Susceptibilidad a Enfermedades , Fibrosis , Hepatocitos/metabolismo , Metabolismo de los Lípidos , Hígado/metabolismo , Masculino , Ratones , Mitocondrias/genética , Enfermedad del Hígado Graso no Alcohólico/patología , Oxidación-Reducción , Estrés Oxidativo , Triglicéridos/metabolismo
3.
Int J Mol Sci ; 21(18)2020 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-32961871

RESUMEN

Metabolic stress, such as lipotoxicity, affects the DNA methylation profile in pancreatic ß-cells and thus contributes to ß-cell failure and the progression of type 2 diabetes (T2D). Stearoyl-CoA desaturase 1 (SCD1) is a rate-limiting enzyme that is involved in monounsaturated fatty acid synthesis, which protects pancreatic ß-cells against lipotoxicity. The present study found that SCD1 is also required for the establishment and maintenance of DNA methylation patterns in ß-cells. We showed that SCD1 inhibition/deficiency caused DNA hypomethylation and changed the methyl group distribution within chromosomes in ß-cells. Lower levels of DNA methylation in SCD1-deficient ß-cells were followed by lower levels of DNA methyltransferase 1 (DNMT1). We also found that the downregulation of SCD1 in pancreatic ß-cells led to the activation of adenosine monophosphate-activated protein kinase (AMPK) and an increase in the activity of the NAD-dependent deacetylase sirtuin-1 (SIRT1). Furthermore, the physical association between DNMT1 and SIRT1 stimulated the deacetylation of DNMT1 under conditions of SCD1 inhibition/downregulation, suggesting a mechanism by which SCD1 exerts control over DNMT1. We also found that SCD1-deficient ß-cells that were treated with compound c, an inhibitor of AMPK, were characterized by higher levels of both global DNA methylation and DNMT1 protein expression compared with untreated cells. Therefore, we found that activation of the AMPK/SIRT1 signaling pathway mediates the effect of SCD1 inhibition/deficiency on DNA methylation status in pancreatic ß-cells. Altogether, these findings suggest that SCD1 is a gatekeeper that protects ß-cells against the lipid-derived loss of DNA methylation and provide mechanistic insights into the mechanism by which SCD1 regulates DNA methylation patterns in ß-cells and T2D-relevant tissues.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasa 1/metabolismo , Metilación de ADN , Células Secretoras de Insulina/metabolismo , Estearoil-CoA Desaturasa/metabolismo , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Proteínas Quinasas Activadas por AMP/metabolismo , Acetilación , Animales , Línea Celular , Metilación de ADN/efectos de los fármacos , Regulación hacia Abajo , Silenciador del Gen , Histonas/metabolismo , Células Secretoras de Insulina/enzimología , Masculino , Ratones , Ratones Endogámicos C57BL , ARN Interferente Pequeño , Ratas , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Sirtuina 1/metabolismo , Espectrometría Raman , Estearoil-CoA Desaturasa/antagonistas & inhibidores , Estearoil-CoA Desaturasa/genética , Regulación hacia Arriba
4.
Postepy Biochem ; 64(2): 166-174, 2018 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-30656899

RESUMEN

During long-standing obesity and insulin resistance, pancreatic ß-cells adapt in order to meet the growing demand of the periphery for insulin. The development of type 2 diabetes requires parallel pathological processes, during which ß-cells are continuously exposed to an overabundant supply of specific lipid derivatives. The metabolic events that lead to inevitable ß-cell damage are not completely uncovered, and for the time being, our understanding of the dynamic endothelium-adipose tissue-ß-cell interactions is limited. Here, we explore various links between continuous obesity, adipose tissue spillover, a dysfunctional endothelium, and defects in islet angioarchitecture to elucidate the crosstalk between signaling systems, cellular mediators, and cell types that contribute to ß-cell failure through diverse actions of fatty acids. These molecular and biochemical mechanisms initiate critical rearrangements of the pancreatic vasculature, intraorgan lipid storage capacity, and inflammatory status that subsequently have severe repercussions on ß-cell function and promote diabetes.


Asunto(s)
Tejido Adiposo/metabolismo , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/metabolismo , Endotelio/metabolismo , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Obesidad/complicaciones , Diabetes Mellitus Tipo 2/patología , Endotelio/patología , Humanos , Resistencia a la Insulina , Obesidad/metabolismo
5.
Int J Mol Sci ; 18(7)2017 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-28726733

RESUMEN

Studying organelles in isolation has been proven to be indispensable for deciphering the underlying mechanisms of molecular cell biology. However, observing organelles in intact cells with the use of microscopic techniques reveals a new set of different junctions and contact sites between them that contribute to the control and regulation of various cellular processes, such as calcium and lipid exchange or structural reorganization of the mitochondrial network. In recent years, many studies focused their attention on the structure and function of contacts between mitochondria and other organelles. From these studies, findings emerged showing that these contacts are involved in various processes, such as lipid synthesis and trafficking, modulation of mitochondrial morphology, endoplasmic reticulum (ER) stress, apoptosis, autophagy, inflammation and Ca 2 + handling. In this review, we focused on the physical interactions of mitochondria with the endoplasmic reticulum and plasma membrane and summarized present knowledge regarding the role of mitochondria-associated membranes in calcium homeostasis and lipid metabolism.


Asunto(s)
Calcio/metabolismo , Membrana Celular/metabolismo , Retículo Endoplásmico/metabolismo , Homeostasis , Metabolismo de los Lípidos , Mitocondrias/metabolismo , Animales , Apoptosis , Transporte Biológico , Membrana Celular/ultraestructura , Susceptibilidad a Enfermedades , Retículo Endoplásmico/ultraestructura , Humanos , Mitocondrias/ultraestructura , Dinámicas Mitocondriales , Transporte de Proteínas
6.
Diabetologia ; 59(7): 1463-1473, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27059371

RESUMEN

AIMS/HYPOTHESIS: We aimed to identify microRNAs (miRNAs) under transcriptional control of the HNF1ß transcription factor, and investigate whether its effect manifests in serum. METHODS: The Polish cohort (N = 60) consisted of 11 patients with HNF1B-MODY, 17 with HNF1A-MODY, 13 with GCK-MODY, an HbA1c-matched type 1 diabetic group (n = 9) and ten healthy controls. Replication was performed in 61 clinically-matched British patients mirroring the groups in the Polish cohort. The Polish cohort underwent miRNA serum level profiling with quantitative real-time PCR (qPCR) arrays to identify differentially expressed miRNAs. Validation was performed using qPCR. To determine whether serum content reflects alterations at a cellular level, we quantified miRNA levels in a human hepatocyte cell line (HepG2) with small interfering RNA knockdowns of HNF1α or HNF1ß. RESULTS: Significant differences (adjusted p < 0.05) were noted for 11 miRNAs. Five of them differed between HNF1A-MODY and HNF1B-MODY, and, amongst those, four (miR-24, miR-27b, miR-223 and miR-199a) showed HNF1B-MODY-specific expression levels in the replication group. In all four cases the miRNA expression level was lower in HNF1B-MODY than in all other tested groups. Areas under the receiver operating characteristic curves ranged from 0.79 to 0.86, with sensitivity and specificity reaching 91.7% (miR-24) and 82.1% (miR-199a), respectively. The cellular expression pattern of miRNA was consistent with serum levels, as all were significantly higher in HNF1α- than in HNF1ß-deficient HepG2 cells. CONCLUSIONS/INTERPRETATION: We have shown that expression of specific miRNAs depends on HNF1ß function. The impact of HNF1ß deficiency was evidenced at serum level, making HNF1ß-dependent miRNAs potentially applicable in the diagnosis of HNF1B-MODY.


Asunto(s)
Factor Nuclear 1-alfa del Hepatocito/metabolismo , Factor Nuclear 1-beta del Hepatocito/metabolismo , MicroARNs/sangre , MicroARNs/genética , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/fisiología , Hemoglobina Glucada/genética , Hemoglobina Glucada/metabolismo , Células Hep G2 , Factor Nuclear 1-alfa del Hepatocito/genética , Factor Nuclear 1-beta del Hepatocito/genética , Humanos , ARN Interferente Pequeño/genética , Curva ROC , Reacción en Cadena en Tiempo Real de la Polimerasa
7.
J Lipid Res ; 56(10): 1901-11, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26293158

RESUMEN

Autophagy is indispensable for the proper architecture and flawless functioning of pancreatic ß-cells. A growing body of evidence indicates reciprocal communication between autophagic pathways, apoptosis, and intracellular lipids. The way in which elevated levels of free saturated or unsaturated FAs contribute to progressive ß-cell failure remains incompletely understood. Stearoyl-CoA desaturase (SCD)1, a key regulatory enzyme in biosynthesis of MUFAs, was shown to play an important role in regulation of ß-cell function. Here, we investigated whether SCD1 activity is engaged in palmitate-induced pancreatic ß-cell autophagy. We found augmented apoptosis and diminished autophagy upon cotreatment of INS-1E cells with palmitate and an SCD1 inhibitor. Furthermore, we found that additional treatment of the cells with monensin, an inhibitor of autophagy at the step of fusion, exacerbates palmitate-induced apoptosis. Accordingly, diminished SCD1 activity affected the accumulation, composition, and saturation status of cellular membrane phospholipids and neutral lipids. Such an effect was accompanied by aberrant endoplasmic reticulum stress, mitochondrial injury, and decreases in insulin secretion and cell proliferation. Our data reveal a novel mechanism by which the inhibition of SCD1 activity affects autophagosome-lysosome fusion because of perturbations in cellular membrane integrity, thus leading to an aberrant stress response and ß-cell failure.


Asunto(s)
Autofagia/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Palmitatos/farmacología , Estearoil-CoA Desaturasa/antagonistas & inhibidores , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Insulina/farmacología , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/enzimología , Células Secretoras de Insulina/metabolismo , Insulinoma , Lisosomas/metabolismo , Fusión de Membrana/efectos de los fármacos , Ácido Palmítico/farmacología , Fosfolípidos/metabolismo , Ratas , Estearoil-CoA Desaturasa/genética , Estearoil-CoA Desaturasa/metabolismo
8.
Biochem Biophys Res Commun ; 460(3): 491-6, 2015 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-25843796

RESUMEN

Obesity-related type 2 diabetes develops in individuals with the onset of ß-cell dysfunction. Pancreatic islet lipotoxicity is now recognized as a primary reason for the onset and progression of the disease. Such dysfunction is reflected by the aberrant secretory capacity and detrimental loss of ß-cell mass and survival. Elevated circulating serum fatty acid levels and disordered lipid metabolism management are particularly interesting in the search for biologically relevant triggers of ß-cell demise. Herein, we review various types of toxic lipid metabolites that may play a significant role in pancreatic islet failure. The lipotoxic effect on ß-cells depends on the type of lipid mediator (e.g., long-chain fatty acids, diacylglycerols, ceramides, phospholipids), cellular location of its action (e.g., endoplasmic reticulum, mitochondria), and associated-organelle conditions (e.g., membranes, vesicles). We also discuss various aspects of lipid action in ß-cells, including effects on metabolic pathways, stress responses (e.g., oxidative stress, endoplasmic reticulum stress, and autophagy), and gene expression.


Asunto(s)
Diabetes Mellitus Tipo 2/fisiopatología , Islotes Pancreáticos/fisiopatología , Autofagia , Ceramidas/metabolismo , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/fisiología , Humanos , Mitocondrias/fisiología
9.
J Biol Chem ; 288(45): 32685-32699, 2013 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-24089517

RESUMEN

Endocannabinoid signaling has been implicated in modulating insulin release from ß cells of the endocrine pancreas. ß Cells express CB1 cannabinoid receptors (CB1Rs), and the enzymatic machinery regulating anandamide and 2-arachidonoylglycerol bioavailability. However, the molecular cascade coupling agonist-induced cannabinoid receptor activation to insulin release remains unknown. By combining molecular pharmacology and genetic tools in INS-1E cells and in vivo, we show that CB1R activation by endocannabinoids (anandamide and 2-arachidonoylglycerol) or synthetic agonists acutely or after prolonged exposure induces insulin hypersecretion. In doing so, CB1Rs recruit Akt/PKB and extracellular signal-regulated kinases 1/2 to phosphorylate focal adhesion kinase (FAK). FAK activation induces the formation of focal adhesion plaques, multimolecular platforms for second-phase insulin release. Inhibition of endocannabinoid synthesis or FAK activity precluded insulin release. We conclude that FAK downstream from CB1Rs mediates endocannabinoid-induced insulin release by allowing cytoskeletal reorganization that is required for the exocytosis of secretory vesicles. These findings suggest a mechanistic link between increased circulating and tissue endocannabinoid levels and hyperinsulinemia in type 2 diabetes.


Asunto(s)
Exocitosis , Quinasa 1 de Adhesión Focal/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Receptor Cannabinoide CB1/metabolismo , Vesículas Secretoras/metabolismo , Animales , Ácidos Araquidónicos/farmacología , Agonistas de Receptores de Cannabinoides/farmacología , Línea Celular , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Endocannabinoides/genética , Endocannabinoides/metabolismo , Endocannabinoides/farmacología , Activación Enzimática/efectos de los fármacos , Activación Enzimática/genética , Quinasa 1 de Adhesión Focal/genética , Glicéridos/farmacología , Humanos , Hiperinsulinismo/genética , Hiperinsulinismo/metabolismo , Hiperinsulinismo/patología , Insulina/genética , Secreción de Insulina , Ratones , Ratones Noqueados , Alcamidas Poliinsaturadas/farmacología , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor Cannabinoide CB1/agonistas , Receptor Cannabinoide CB1/genética , Vesículas Secretoras/genética
10.
Biochim Biophys Acta Mol Basis Dis ; 1869(6): 166711, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37054998

RESUMEN

The accelerating accumulation of surplus lipids in the pancreas triggers structural and functional changes in type 2 diabetes-affected islets. Pancreatic ß-cells exhibit a restricted capacity to store fat reservoirs in lipid droplets (LDs), which act as transient buffers to prevent lipotoxic stress. With the increasing incidence of obesity, growing interest has been seen in the intracellular regulation of LD metabolism for ß-cell function. Stearoyl-CoA desaturase 1 (SCD1) is critical for producing unsaturated fatty acyl moieties for fluent storage into and out of LDs, likely affecting the overall rate of ß-cell survival. We explored LD-associated composition and remodeling in SCD1-deprived INS-1E cells and in pancreatic islets in wildtype and SCD1-/- mice in the lipotoxic milieu. Deficiency in the enzymatic activity of SCD1 led to decrease in the size and number of LDs and the lower accumulation of neutral lipids. This occurred in parallel with a higher compactness and lipid order inside LDs, followed by changes in the saturation status and composition of fatty acids within core lipids and the phospholipid coat. The lipidome of LDs was enriched in 18:2n-6 and 20:4n-6 in ß-cells and pancreatic islets. These rearrangements markedly contributed to differences in protein association with the LD surface. Our findings highlight an unexpected molecular mechanism by which SCD1 activity affects the morphology, composition and metabolism of LDs. We demonstrate that SCD1-dependent disturbances in LD enrichment can impact pancreatic ß-cells and islet susceptibility to palmitate, which may have considerable diagnostic and methodological value for the characterization of LDs in human ß-cells in type 2 diabetes patients.


Asunto(s)
Diabetes Mellitus Tipo 2 , Palmitatos , Animales , Humanos , Ratones , Diabetes Mellitus Tipo 2/metabolismo , Ácidos Grasos/metabolismo , Gotas Lipídicas/metabolismo , Estearoil-CoA Desaturasa/genética , Estearoil-CoA Desaturasa/metabolismo
11.
Mol Metab ; 67: 101659, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36529318

RESUMEN

Abnormalities that characterize the pathophysiology of type 2 diabetes (T2D) include deficiencies of ß-cells and the expansion of α-cells in pancreatic islets, manifested by lower insulin release and glucagon oversecretion. The molecular mechanisms that determine intra-islet interactions between pancreatic α- and ß-cells are still not fully understood. The present study showed that stearoyl-coenzyme A (CoA) desaturase 1 (SCD1), an enzyme that is implicated in fatty acid metabolism, serves as a checkpoint in the control of endocrine cell equilibrium in pancreatic islets. Our data showed that SCD1 activity is essential for proper α-cell and ß-cell lineage determination during morphogenesis of the pancreas and the maintenance of mature ß-cell identity. The inhibition of SCD1 expression/activity led to both a decrease in the expression of ß-cell signature genes (e.g., Pdx1, Nkx6.1, MafA, and Neurod1, among others) and induction of the expression of the dedifferentiation marker Sox9 in mature pancreatic islets. The transcriptional repression of Pdx1 and MafA in SCD1-deficient ß-cells was related to the excessive methylation of promoter regions of these transcription factors. In contrast, SCD1 ablation favored the formation of α-cells over ß-cells throughout pancreas organogenesis and did not compromise α-cell identity in adult pancreatic islets. Such molecular changes that were caused by SCD1 downregulation resulted in the mislocalization of α-cells within the core of islets and increased the ratio of pancreatic α- to ß-cell mass. This was followed by islet dysfunction, including impairments in glucose-stimulated insulin release, simultaneously with elevations of basal glucagon secretion. Altogether, these findings provide additional mechanistic insights into the role of SCD1 in the pathogenesis of T2D.


Asunto(s)
Diabetes Mellitus Tipo 2 , Células Secretoras de Glucagón , Islotes Pancreáticos , Ratones , Animales , Estearoil-CoA Desaturasa/genética , Estearoil-CoA Desaturasa/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Glucagón/metabolismo , Islotes Pancreáticos/metabolismo , Insulina/metabolismo , Células Secretoras de Glucagón/metabolismo , Morfogénesis
12.
Int J Biochem Cell Biol ; 152: 106295, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36113832

RESUMEN

Diacylglycerol kinase-ε (DGKε) phosphorylates DAG to phosphatidic acid with unique specificity toward 18:0/20:4 DAG (SAG). SAG is a typical backbone of phosphatidylinositol and its derivatives, therefore DGKε activity is crucial for the turnover of these signaling lipids. Malfunction of DGKε contributes to several pathophysiological conditions, including atypical hemolytic uremic syndrome (aHUS) linked with DGKE mutations. In the present study we analyzed the role of a zinc finger motif of the C1B domain of DGKε, as some aHUS-linked mutations affect this ill-defined part of the kinase. For this, we introduce a novel fluorescent assay for determination of DGKε activity which relies on the use of NBD-SAG in mixed micelles as a substrate, followed by TLC separation of NBD-phosphatidic acid formed. The assay reliably determines the activity of purified human GST-DGKε, also endogenous DGKε or overexpressed mouse DGKε-Myc in cell lysates, homogenates, and kinase immunoprecipitates. Using the above assay we found that four amino acids, Cys135, Cys138, His161 and Cys164, forming the zinc finger motif in the C1B domain are required for the DGKε-Myc activity and stability. Substitution of any of these amino acids with Ala or Trp in DGKε-Myc abolished its activity and led to its proteasomal degradation, possibly assisted by Hsp70/90/40 chaperones. Inhibition of the 26S proteasome prevented the degradation but the mutated proteins were inactive. The present data on the deleterious effect of the zinc finger motif disruption contribute to the understanding of the DGKε-linked aHUS, as the Cys164Trp substitution in mouse DGKε corresponds to the Cys167Trp one in human DGKε found in some aHUS patients.


Asunto(s)
Síndrome Hemolítico Urémico Atípico , Diacilglicerol Quinasa , Animales , Humanos , Ratones , Aminoácidos , Diacilglicerol Quinasa/química , Diacilglicerol Quinasa/genética , Diacilglicerol Quinasa/metabolismo , Mutación , Ácidos Fosfatidicos , Transducción de Señal/fisiología , Síndrome Hemolítico Urémico Atípico/genética , Síndrome Hemolítico Urémico Atípico/metabolismo
13.
Redox Biol ; 55: 102400, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35863265

RESUMEN

Non-alcoholic fatty liver disease (NAFLD) is a health concern affecting 24% of the population worldwide. Although the pathophysiologic mechanisms underlying disease are not fully clarified, mitochondrial dysfunction and oxidative stress are key players in disease progression. Consequently, efforts to develop more efficient pharmacologic strategies targeting mitochondria for NAFLD prevention/treatment are underway. The conjugation of caffeic acid anti-oxidant moiety with an alkyl linker and a triphenylphosphonium cation (TPP+), guided by structure-activity relationships, led to the development of a mitochondria-targeted anti-oxidant (AntiOxCIN4) with remarkable anti-oxidant properties. Recently, we described that AntiOxCIN4 improved mitochondrial function, upregulated anti-oxidant defense systems, and cellular quality control mechanisms (mitophagy/autophagy) via activation of the Nrf2/Keap1 pathway, preventing fatty acid-induced cell damage. Despite the data obtained, AntiOxCIN4 effects on cellular and mitochondrial energy metabolism in vivo were not studied. In the present work, we proposed that AntiOxCIN4 (2.5 mg/day/animal) may prevent non-alcoholic fatty liver (NAFL) phenotype development in a C57BL/6J mice fed with 30% high-fat, 30% high-sucrose diet for 16 weeks. HepG2 cells treated with AntiOxCIN4 (100 µM, 48 h) before the exposure to supraphysiologic free fatty acids (FFAs) (250 µM, 24 h) were used for complementary studies. AntiOxCIN4 decreased body (by 43%), liver weight (by 39%), and plasma hepatocyte damage markers in WD-fed mice. Hepatic-related parameters associated with a reduction of fat liver accumulation (by 600%) and the remodeling of fatty acyl chain composition compared with the WD-fed group were improved. Data from human HepG2 cells confirmed that a reduction of lipid droplets size and number can be a result from AntiOxCIN4-induced stimulation of fatty acid oxidation and mitochondrial OXPHOS remodeling. In WD-fed mice, AntiOxCIN4 also induced a hepatic metabolism remodeling by upregulating mitochondrial OXPHOS, anti-oxidant defense system and phospholipid membrane composition, which is mediated by the PGC-1α-SIRT3 axis. AntiOxCIN4 prevented lipid accumulation-driven autophagic flux impairment, by increasing lysosomal proteolytic capacity. AntiOxCIN4 improved NAFL phenotype of WD-fed mice, via three main mechanisms: a) increase mitochondrial function (fatty acid oxidation); b) stimulation anti-oxidant defense system (enzymatic and non-enzymatic) and; c) prevent the impairment in autophagy. Together, the findings support the potential use of AntiOxCIN4 in the prevention/treatment of NAFLD.

14.
Biosens Bioelectron ; 183: 113215, 2021 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-33845292

RESUMEN

Type 2 diabetes is currently one of the most common metabolic diseases, affecting all ages worldwide. As the incidence of type 2 diabetes increases, a growing number of studies focus on islets of Langerhans. A three-dimensional research model that maps islet morphology and maintains hormonal balance in vivo is still needed. In this work, we present an Islet-on-a-chip system, specifically a micropillar-based microfluidic platform for three-dimensional pancreatic islet cell culture and analysis. The microfluidic system consisted of two culture chambers that were equipped with 15 circular microtraps each, which were built with seven round micropillars each. Micropillars in the structure of microtraps supported cell aggregation by limiting the growth surface and minimizing wall shear stress, thereby ensuring proper medium diffusion and optimal culture conditions for cell aggregates. Our system is compatible with microwell plate readers and confocal laser scanning microscopes. Because of optimization of the immunostaining method, the appropriate cell distribution and high viability and proliferation up to 72 h of culture were confirmed. Enzyme-linked immunosorbent assays were performed to measure insulin and glucagon secretion after stimulation with different glucose concentrations. To our knowledge, this is the first Lab-on-a-chip system which enables the formation and three-dimensional culture of cell aggregates composed of commercially available α and ß pancreatic islet cells. The specific composition and arrangement of cells in the obtained model corresponds to the arrangement of the cells in rodent pancreatic islets in vivo. This Islet-on-a-chip system may be utilized to test pathogenic effectors and future therapeutic agents.


Asunto(s)
Técnicas Biosensibles , Diabetes Mellitus Tipo 2 , Islotes Pancreáticos , Biomimética , Técnicas de Cultivo de Célula , Glucosa , Humanos , Insulina , Dispositivos Laboratorio en un Chip , Microfluídica
15.
Biosens Bioelectron ; 167: 112451, 2020 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-32777682

RESUMEN

Cases of type 2 diabetes mellitus have significantly increased in recent years. Researchers worldwide are combining their knowledge of biology, medicine, tissue engineering, and microtechnology to develop new effective treatments. An important aspect of current research is to develop of a complete model of three-dimensional pancreatic islets to test various factors that affect disease development and evaluate new therapies and drugs. Several methods have allowed the development of three-dimensional research models. The use of Lab-on-a-chip systems with appropriate microstructure geometry is a promising solution to macroscale problems. Such a device allows the development of a complete platform reflecting conditions that prevail in the body. Organ-on-a-chip platforms are successfully used mainly in studies of lung, heart, and liver diseases. This review presents the current state of knowledge on the creation of three-dimensional pancreatic islet structures in both microscale and microfluidic systems. We highlight the most important aspects of developing the geometry of such devices. We also discuss analytical detection methods that are suitable for detecting hormones that are secreted from pancreatic islets and, in combination with appropriate Lab-on-a-chip systems, can be used as a Micro Total Analysis System (µTAS).


Asunto(s)
Técnicas Biosensibles , Diabetes Mellitus Tipo 2 , Islotes Pancreáticos , Humanos , Dispositivos Laboratorio en un Chip , Medicina Regenerativa
16.
Antioxidants (Basel) ; 9(10)2020 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-33076261

RESUMEN

Nonalcoholic fatty liver disease (NAFLD) is characterized by the development of steatosis, which can ultimately compromise liver function. Mitochondria are key players in obesity-induced metabolic disorders; however, the distinct role of hypercaloric diet constituents in hepatic cellular oxidative stress and metabolism is unknown. Male mice were fed either a high-fat (HF) diet, a high-sucrose (HS) diet or a combined HF plus HS (HFHS) diet for 16 weeks. This study shows that hypercaloric diets caused steatosis; however, the HFHS diet induced severe fibrotic phenotype. At the mitochondrial level, lipidomic analysis showed an increased cardiolipin content for all tested diets. Despite this, no alterations were found in the coupling efficiency of oxidative phosphorylation and neither in mitochondrial fatty acid oxidation (FAO). Consistent with unchanged mitochondrial function, no alterations in mitochondrial-induced reactive oxygen species (ROS) and antioxidant capacity were found. In contrast, the HF and HS diets caused lipid peroxidation and provoked altered antioxidant enzyme levels/activities in liver tissue. Our work provides evidence that hepatic oxidative damage may be caused by augmented levels of peroxisomes and consequently higher peroxisomal FAO-induced ROS in the early NAFLD stage. Hepatic damage is also associated with autophagic flux impairment, which was demonstrated to be diet-type dependent. The HS diet induced a reduction in autophagosomal formation, while the HF diet reduced levels of cathepsins. The accumulation of damaged organelles could instigate hepatocyte injuries and NAFLD progression.

17.
Biochim Biophys Acta Mol Basis Dis ; 1865(10): 2753-2764, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31255704

RESUMEN

In the setting of metabolic overload, chronic elevations of free fatty acids in blood and tissues are associated with pancreatic ß-cell lipotoxicity and failure. Ultimately, obesity combined with insulin resistance increases the dysfunctional demand of ß-cells and contributes to the development of type 2 diabetes. Forkhead box O1 (FoxO1) is a potent transcriptional regulator of pancreatic ß-cell function and tolerance to lipid stress. The present study examined the effects of stearoyl-CoA desaturase 1 (SCD1)-metabolized precursors and products, notably oleic acid, on the compensatory capacity of ß-cells and their relationship with regulation of the FoxO1 and Wnt pathways. The trioleate-induced compromise of insulin sensitivity blunted the compensatory response of pancreatic ß-cells in primary rat islets. These events were associated with increases in the nuclear accumulation and transcriptional activity of FoxO1. Such effects were also observed in INS-1E cells that were subjected to oleate treatment. The overexpression of human SCD1 that was accompanied by endogenously generated oleic acid also led to an increase in the nuclear abundance of FoxO1. The mechanism of the oleate-mediated subcellular localization of FoxO1 was independent of the fatty acid receptor GPR40. Instead, the mechanism involved diversion of the active ß-catenin pool from an interaction with transcription factor 7-like 2 toward FoxO1-mediated transcription in ß-cells. Our findings identify a unique role for oleic acid in the compensatory response of pancreatic ß-cells and emphasize the importance of FoxO1 in ß-cell failure in obesity-induced insulin resistance.


Asunto(s)
Proteína Forkhead Box O1/metabolismo , Regulación de la Expresión Génica , Células Secretoras de Insulina/metabolismo , Ácido Oléico/metabolismo , Transporte de Proteínas/fisiología , beta Catenina/metabolismo , Animales , Núcleo Celular , Diabetes Mellitus Tipo 2/metabolismo , Proteínas de Homeodominio , Masculino , Proteínas del Tejido Nervioso/metabolismo , Ratas , Ratas Wistar , Receptores Acoplados a Proteínas G , Estearoil-CoA Desaturasa/metabolismo , Transactivadores , Proteína 2 Similar al Factor de Transcripción 7/metabolismo
18.
Nutrients ; 11(12)2019 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-31771244

RESUMEN

Nonalcoholic fatty liver disease (NAFLD) is a common disease in Western society and ranges from steatosis to steatohepatitis to end-stage liver disease such as cirrhosis and hepatocellular carcinoma. The molecular mechanisms that are involved in the progression of steatosis to more severe liver damage in patients are not fully understood. A deeper investigation of NAFLD pathogenesis is possible due to the many different animal models developed recently. In this review, we present a comparative overview of the most common dietary NAFLD rodent models with respect to their metabolic phenotype and morphological manifestation. Moreover, we describe similarities and controversies concerning the effect of NAFLD-inducing diets on mitochondria as well as mitochondria-derived oxidative stress in the progression of NAFLD.


Asunto(s)
Ácidos Grasos/metabolismo , Metabolismo de los Lípidos , Mitocondrias Hepáticas/fisiología , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/fisiopatología , Animales , Deficiencia de Colina , Diabetes Mellitus Tipo 2/complicaciones , Dieta Alta en Grasa/efectos adversos , Dieta Occidental/efectos adversos , Azúcares de la Dieta/efectos adversos , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Hígado Graso/complicaciones , Ratones , Enfermedad del Hígado Graso no Alcohólico/complicaciones , Estrés Oxidativo , Fenotipo , Ratas , Especies Reactivas de Oxígeno , Roedores
19.
Cell Death Dis ; 9(3): 332, 2018 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-29491385

RESUMEN

Sites of close contact between mitochondria and the endoplasmic reticulum (ER) are known as mitochondria-associated membranes (MAM) or mitochondria-ER contacts (MERCs), and play an important role in both cell physiology and pathology. A growing body of evidence indicates that changes observed in the molecular composition of MAM and in the number of MERCs predisposes MAM to be considered a dynamic structure. Its involvement in processes such as lipid biosynthesis and trafficking, calcium homeostasis, reactive oxygen species production, and autophagy has been experimentally confirmed. Recently, MAM have also been studied in the context of different pathologies, including Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, type 2 diabetes mellitus and GM1-gangliosidosis. An underappreciated amount of data links MAM with aging or senescence processes. In the present review, we summarize the current knowledge of basic MAM biology, composition and action, and discuss the potential connections supporting the idea that MAM are significant players in longevity.


Asunto(s)
Envejecimiento/metabolismo , Retículo Endoplásmico/metabolismo , Mitocondrias/metabolismo , Membranas Mitocondriales/metabolismo , Animales , Senescencia Celular , Humanos
20.
Sci Rep ; 6: 31553, 2016 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-27527335

RESUMEN

Wnt signaling molecules are associated with obesity, hyperlipidemia, and type 2 diabetes (T2D). Here, we show that two Wnt proteins, WNT3a and WNT4, are specifically secreted by skeletal muscle and adipose tissue during the development of insulin resistance and play an important role in cross-talk between insulin-resistant tissues and pancreatic beta cells. The activation of Frizzled receptor and Wnt signaling in pancreatic islets via circulating WNT3a in blood resulted in higher insulin secretion and an increase in beta cell proliferation, thus leading to islet adaptation in a pre-diabetic state. Interestingly, in fully developed T2D, the expression profiles of Wnt3a and Wnt4 in adipose tissue and muscle cells and blood plasma levels of these proteins were opposite to the pre-diabetic state, thus favoring the downregulation of Wnt signaling in beta cells and resulting in dysfunctional pancreatic islets. These results demonstrate that alterations in the secretion profile of a canonical Wnt activator (WNT3a) and inhibitor (WNT4) from insulin-resistant tissues during the development of T2D are responsible for triggering progression from a pre-diabetic to a diabetic state. We also show here that WNT3a and WNT4 are potent myokines, and their expression and secretion are regulated in response to nutritional and metabolic changes.


Asunto(s)
Tejido Adiposo/metabolismo , Islotes Pancreáticos/fisiopatología , Músculo Esquelético/metabolismo , Proteína Wnt3A/metabolismo , Proteína Wnt4/metabolismo , Células 3T3-L1 , Animales , Medios de Cultivo Condicionados , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patología , Dieta Alta en Grasa , Progresión de la Enfermedad , Resistencia a la Insulina , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/patología , Masculino , Ratones , Ratas , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA