Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Atherosclerosis ; 387: 117383, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-38061313

RESUMEN

BACKGROUND AND AIMS: Deficiency in the thiol transferase glutaredoxin 1 (Grx1) in aging mice promotes, in a sexually dimorphic manner, dysregulation of macrophages and atherogenesis. However, the underlying mechanisms are not known. Here we tested the hypothesis that macrophage-restricted overexpression of Grx1 protects atherosclerosis-prone mice against macrophage reprogramming and dysfunction induced by a high-calorie diet (HCD) and thereby reduces the severity of atherosclerosis. METHODS: We generated lentiviral vectors carrying cluster of differentiation 68 (CD68) promoter-driven enhanced green fluorescent protein (EGFP) or Grx1 constructs and conducted bone marrow (BM) transplantation studies to overexpress Grx1 in a macrophage-specific manner in male and female atherosclerosis-prone LDLR-/- mice, and fed these mice a HCD to induce atherogenesis. Atherosclerotic lesion size was determined in both the aortic root and the aorta. We isolated BM-derived macrophages (BMDM) to assess protein S-glutathionylation levels and loss of mitogen-activated protein kinase phosphatase 1 (MKP-1) activity as measures of HCD-induced thiol oxidative stress. We also conducted gene profiling on these BMDM to determine the impact of Grx1 activity on HCD-induced macrophage reprogramming. RESULTS: Overexpression of Grx1 protected macrophages against HCD-induced protein S-glutathionylation, reduced monocyte chemotaxis in vivo, limited macrophage recruitment into atherosclerotic lesions, and was sufficient to reduce the severity of atherogenesis in both male and female mice. Gene profiling revealed major sex differences in the transcriptional reprogramming of macrophages induced by HCD feeding, but Grx1 overexpression only partially reversed HCD-induced transcriptional reprogramming of macrophages. CONCLUSIONS: Macrophage Grx1 plays a major role in protecting mice atherosclerosis mainly by maintaining the thiol redox state of the macrophage proteome and preventing macrophage dysfunction.


Asunto(s)
Aterosclerosis , Glutarredoxinas , Animales , Femenino , Masculino , Ratones , Aterosclerosis/genética , Aterosclerosis/prevención & control , Aterosclerosis/metabolismo , Glutarredoxinas/genética , Glutarredoxinas/metabolismo , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Nutrientes , Compuestos de Sulfhidrilo
2.
Cardiovasc Res ; 118(18): 3586-3601, 2023 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-35704032

RESUMEN

AIMS: To test the hypothesis that the activation of the growth hormone-releasing hormone (GHRH) receptor signalling pathway within the myocardium both prevents and reverses diastolic dysfunction and pathophysiologic features consistent with heart failure with preserved ejection fraction (HFpEF). Impaired myocardial relaxation, fibrosis, and ventricular stiffness, among other multi-organ morbidities, characterize the phenotype underlying the HFpEF syndrome. Despite the rapidly increasing prevalence of HFpEF, few effective therapies have emerged. Synthetic agonists of the GHRH receptors reduce myocardial fibrosis, cardiomyocyte hypertrophy, and improve performance in animal models of ischaemic cardiomyopathy, independently of the growth hormone axis. METHODS AND RESULTS: CD1 mice received 4- or 8-week continuous infusion of angiotensin-II (Ang-II) to generate a phenotype with several features consistent with HFpEF. Mice were administered either vehicle or a potent synthetic agonist of GHRH, MR-356 for 4-weeks beginning concurrently or 4-weeks following the initiation of Ang-II infusion. Ang-II-treated animals exhibited diastolic dysfunction, ventricular hypertrophy, interstitial fibrosis, and normal ejection fraction. Cardiomyocytes isolated from these animals exhibited incomplete relaxation, depressed contractile responses, altered myofibrillar protein phosphorylation, and disturbed calcium handling mechanisms (ex vivo). MR-356 both prevented and reversed the development of the pathological phenotype in vivo and ex vivo. Activation of the GHRH receptors increased cAMP and cGMP in cardiomyocytes isolated from control animals but only cAMP in cardiac fibroblasts, suggesting that GHRH-A exert differential effects on cardiomyocytes and fibroblasts. CONCLUSION: These findings indicate that the GHRH receptor signalling pathway(s) represents a new molecular target to counteract dysfunctional cardiomyocyte relaxation by targeting myofilament phosphorylation and fibrosis. Accordingly, activation of GHRH receptors with potent, synthetic GHRH agonists may provide a novel therapeutic approach to management of the myocardial alterations associated with the HFpEF syndrome.


Asunto(s)
Cardiomiopatías , Insuficiencia Cardíaca , Ratones , Animales , Insuficiencia Cardíaca/metabolismo , Volumen Sistólico/fisiología , Cardiomiopatías/metabolismo , Cardiomegalia/metabolismo , Miocitos Cardíacos/metabolismo , Hormona Liberadora de Hormona del Crecimiento/metabolismo , Fibrosis
3.
Expert Opin Biol Ther ; 22(4): 449-463, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-34882517

RESUMEN

INTRODUCTION: Mesenchymal stromal cells (MSCs; AKA mesenchymal stem cells) stimulate healing and reduce inflammation. Promising therapeutic responses are seen in many late-phase clinical trials, but others have not satisfied their primary endpoints, making translation of MSCs into clinical practice difficult. These inconsistencies may be related to the route of MSC delivery, lack of product optimization, or varying background therapies received in clinical trials over time. AREAS COVERED: Here we discuss the different routes of MSC delivery, highlighting the proposed mechanism(s) of therapeutic action as well as potential safety concerns. PubMed search criteria used: MSC plus: local administration; routes of administration; delivery methods; mechanism of action; therapy in different diseases. EXPERT OPINION: Direct injection of MSCs using a controlled local delivery approach appears to have benefits in certain disease states, but further studies are required to make definitive conclusions regarding the superiority of one delivery method over another.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Humanos , Trasplante de Células Madre Mesenquimatosas/efectos adversos , Células Madre Mesenquimatosas/fisiología , Cicatrización de Heridas
4.
Artículo en Inglés | MEDLINE | ID: mdl-34790975

RESUMEN

INTRODUCTION: Induced pluripotent stem cells (iPSCs) provide a model of cardiomyocyte (CM) maturation. Nitric oxide signaling promotes CM differentiation and maturation, although the mechanisms remain controversial. AIM: The study tested the hypothesis that in the absence of S-nitrosoglutathione reductase (GSNOR), a denitrosylase regulating protein S-nitrosylation, the resultant increased S-nitrosylation accelerates the differentiation and maturation of iPSC-derived cardiomyocytes (CMs). METHODS AND RESULTS: iPSCs derived from mice lacking GSNOR (iPSCGSNOR-/-) matured faster than wildtype iPSCs (iPSCWT) and demonstrated transient increases in expression of murine Snail Family Transcriptional Repressor 1 gene (Snail), murine Snail Family Transcriptional Repressor 2 gene (Slug) and murine Twist Family BHLH Transcription Factor 1 gene (Twist), transcription factors that promote epithelial-to-mesenchymal transition (EMT) and that are regulated by Glycogen Synthase Kinase 3 Beta (GSK3ß). Murine Glycogen Synthase Kinase 3 Beta (Gsk3ß) gene exhibited much greater S-nitrosylation, but lower expression in iPSCGSNOR-/-. S-nitrosoglutathione (GSNO)-treated iPSCWT and human (h)iPSCs also demonstrated reduced expression of GSK3ß. Nkx2.5 expression, a CM marker, was increased in iPSCGSNOR-/- upon directed differentiation toward CMs on Day 4, whereas murine Brachyury (t), Isl1, and GATA Binding Protein (Gata4) mRNA were decreased, compared to iPSCWT, suggesting that GSNOR deficiency promotes CM differentiation beginning immediately following cell adherence to the culture dish-transitioning from mesoderm to cardiac progenitor. CONCLUSION: Together these findings suggest that increased S-nitrosylation of Gsk3ß promotes CM differentiation and maturation from iPSCs. Manipulating the post-translational modification of GSK3ß may provide an important translational target and offers new insight into understanding of CM differentiation from pluripotent stem cells. ONE SENTENCE SUMMARY: Deficiency of GSNOR or addition of GSNO accelerates early differentiation and maturation of iPSC-cardiomyocytes.

5.
Front Cardiovasc Med ; 8: 667298, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34322524

RESUMEN

Investigations into the regulatory mechanisms controlling cholesterol homeostasis have proven fruitful in identifying low-density lipoprotein (LDL)-lowering therapies to reduce the risk of atherosclerotic cardiovascular disease. A major advance was the discovery of proprotein convertase subtilisin/kexin type 9 (PCSK9), a secreted protein that binds the LDL receptor (LDLR) on the cell surface and internalizes it for degradation, thereby blunting its ability to take up circulating LDL. The discovery that loss-of-function mutations in PCSK9 lead to lower plasma levels of LDL cholesterol and protection from cardiovascular disease led to the therapeutic development of PCSK9 inhibitors at an unprecedented pace. However, there remain many gaps in our understanding of PCSK9 regulation and biology, including its posttranscriptional control by microRNAs. Using a high-throughput region(3'-UTR) of human microRNA library screen, we identified microRNAs targeting the 3' untranslated region of human PCSK9. The top 35 hits were confirmed by large-format PCSK9 3'-UTR luciferase assays, and 10 microRNAs were then selected for further validation in hepatic cells, including effects on PCSK9 secretion and LDLR cell surface expression. These studies identified seven novel microRNAs that reduce PCSK9 expression, including miR-221-5p, miR-342-5p, miR-363-5p, miR-609, miR-765, and miR-3165. Interestingly, several of these microRNAs were also found to target other genes involved in LDLR regulation and potently upregulate LDLR cell surface expression in hepatic cells. Together, these data enhance our understanding of post-transcriptional regulators of PCSK9 and their potential for therapeutic manipulation of hepatic LDLR expression.

6.
Biomolecules ; 11(1)2021 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-33430172

RESUMEN

Endothelial dysfunction precedes atherosclerosis and is an independent predictor of cardiovascular events. Cholesterol levels and oxidative stress are key contributors to endothelial damage, whereas high levels of plasma high-density lipoproteins (HDL) could prevent it. Cholesteryl ester transfer protein (CETP) is one of the most potent endogenous negative regulators of HDL-cholesterol. However, whether and to what degree CETP expression impacts endothelial function, and the molecular mechanisms underlying the vascular effects of CETP on endothelial cells, have not been addressed. Acetylcholine-induced endothelium-dependent relaxation of aortic rings was impaired in human CETP-expressing transgenic mice, compared to their non-transgenic littermates. However, endothelial nitric oxide synthase (eNOS) activation was enhanced. The generation of superoxide and hydrogen peroxide was increased in aortas from CETP transgenic mice, while silencing CETP in cultured human aortic endothelial cells effectively decreased oxidative stress promoted by all major sources of ROS: mitochondria and NOX2. The endoplasmic reticulum stress markers, known as GADD153, PERK, and ARF6, and unfolded protein response effectors, were also diminished. Silencing CETP reduced endothelial tumor necrosis factor (TNF) α levels, intercellular cell adhesion molecule-1 (ICAM-1), and vascular cell adhesion molecule-1 (VCAM-1) expression, diminishing monocyte adhesion. These results support the notion that CETP expression negatively impacts endothelial cell function, revealing a new mechanism that might contribute to atherosclerosis.


Asunto(s)
Proteínas de Transferencia de Ésteres de Colesterol/metabolismo , Células Endoteliales/metabolismo , Endotelio Vascular/patología , Endotelio Vascular/fisiopatología , Estrés Oxidativo , Animales , Caveolinas/metabolismo , Moléculas de Adhesión Celular/metabolismo , Proteínas de Transferencia de Ésteres de Colesterol/antagonistas & inhibidores , Proteínas de Transferencia de Ésteres de Colesterol/genética , Estrés del Retículo Endoplásmico , Activación Enzimática , Humanos , Ratones Transgénicos , NADPH Oxidasas/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fosforilación , ARN Mensajero/genética , ARN Mensajero/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Células THP-1 , Vasodilatación
7.
Front Cardiovasc Med ; 7: 81, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32528976

RESUMEN

MicroRNAs (miRNA) have emerged as important post-transcriptional regulators of metabolic pathways that contribute to cellular and systemic lipoprotein homeostasis. Here, we identify two conserved miRNAs, miR-224, and miR-520d, which target gene networks regulating hepatic expression of the low-density lipoprotein (LDL) receptor (LDLR) and LDL clearance. In silico prediction of miR-224 and miR-520d target gene networks showed that they each repress multiple genes impacting the expression of the LDLR, including the chaperone molecules PCSK9 and IDOL that limit LDLR expression at the cell surface and the rate-limiting enzyme for cholesterol synthesis HMGCR, which is the target of LDL-lowering statin drugs. Using gain- and loss-of-function studies, we tested the role of miR-224 and miR-520d in the regulation of those predicted targets and their impact on LDLR expression. We show that overexpression of miR-224 or miR-520d dose-dependently reduced the activity of PCSK9, IDOL, and HMGCR 3'-untranslated region (3'-UTR)-luciferase reporter constructs and that this repression was abrogated by mutation of the putative miR-224 or miR-520d response elements in the PCSK9, IDOL, and HMGCR 3'-UTRs. Compared to a control miRNA, overexpression of miR-224 or miR-520d in hepatocytes inhibited PCSK9, IDOL, and HMGCR mRNA and protein levels and decreased PCSK9 secretion. Furthermore, miR-224 and miR-520d repression of PCSK9, IDOL, and HMGCR was associated with an increase in LDLR protein levels and cell surface expression, as well as enhanced LDL binding. Notably, the effects of miR-224 and miR-520d were additive to the effects of statins in upregulating LDLR expression. Finally, we show that overexpression of miR-224 in the livers of Ldlr +/- mice using lipid nanoparticle-mediated delivery resulted in a 15% decrease in plasma levels of LDL cholesterol, compared to a control miRNA. Together, these findings identify roles for miR-224 and miR-520d in the posttranscriptional control of LDLR expression and function.

8.
Front Cardiovasc Med ; 7: 618651, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33521067

RESUMEN

The COVID-19 pandemic caused by the SARS-CoV-2 coronavirus requires reliable assays for studying viral entry mechanisms which remains poorly understood. This knowledge is important for the development of therapeutic approaches to control SARS-CoV-2 infection by permitting the screening for neutralizing antibodies and other agents that can block infection. This is particularly important for patients who are at high risk for severe outcomes related to COVID-19. The production of pseudotyped viral particles may seem like a daunting task for a non-virology laboratory without experience in the two most commonly used pseudotyping systems, namely retro/lentiviruses and vesicular stomatitis virus (VSV) which lacks the VSV envelope glycoprotein (VSVΔG). By incorporating the most up-to-date knowledge, we have developed a detailed, easy-to-follow novel protocol for producing SARS-CoV-2 spike-bearing pseudovirus using the VSV-ΔG system. We describe the infection assay which uses GFP fluorescence as a measure of infection in a 24-well live imaging system. We present results of our optimization of the system to enhance viral infection levels through the over-expression of human ACE2 receptor and the overexpression of at least one of two proteases - TMPRSS2 or Furin, as well as, supplementation with Poloxamer 407 (P407) and Prostaglandin E2 (PGE2) as adjuvants. We show that the system works efficiently in three unrelated, clinically relevant cell lines: human 293T (renal epithelial) cells, human Calu-3 (lung epithelial) cells, and the non-human primate (African Green Monkey) cell line, Vero-E6 (renal epithelial) cells. In addition, we have used this system to show infection of human induced pluripotent stem cell-derived cardiomyocytes (iPS-CMs). This system is efficient (virus generation, titration, and infection assays can be performed in 1 week), quantitative, inexpensive, and readily scalable for application in drug development and therapeutic screening approaches.

9.
Biochem J ; 476(24): 3769-3789, 2019 12 23.
Artículo en Inglés | MEDLINE | ID: mdl-31803904

RESUMEN

The atherosclerosis prone LDL receptor knockout mice (Ldlr-/-, C57BL/6J background) carry a deletion of the NADP(H)-transhydrogenase gene (Nnt) encoding the mitochondrial enzyme that catalyzes NADPH synthesis. Here we hypothesize that both increased NADPH consumption (due to increased steroidogenesis) and decreased NADPH generation (due to Nnt deficiency) in Ldlr-/- mice contribute to establish a macrophage oxidative stress and increase atherosclerosis development. Thus, we compared peritoneal macrophages and liver mitochondria from three C57BL/6J mice lines: Ldlr and Nnt double mutant, single Nnt mutant and wild-type. We found increased oxidants production in both mitochondria and macrophages according to a gradient: double mutant > single mutant > wild-type. We also observed a parallel up-regulation of mitochondrial biogenesis (PGC1a, TFAM and respiratory complexes levels) and inflammatory (iNOS, IL6 and IL1b) markers in single and double mutant macrophages. When exposed to modified LDL, the single and double mutant cells exhibited significant increases in lipid accumulation leading to foam cell formation, the hallmark of atherosclerosis. Nnt deficiency cells showed up-regulation of CD36 and down-regulation of ABCA1 transporters what may explain lipid accumulation in macrophages. Finally, Nnt wild-type bone marrow transplantation into LDLr-/- mice resulted in reduced diet-induced atherosclerosis. Therefore, Nnt plays a critical role in the maintenance of macrophage redox, inflammatory and cholesterol homeostasis, which is relevant for delaying the atherogenesis process.


Asunto(s)
Aterosclerosis/metabolismo , Macrófagos Peritoneales/metabolismo , NADP/metabolismo , Estrés Oxidativo , Transportador 1 de Casete de Unión a ATP/genética , Transportador 1 de Casete de Unión a ATP/metabolismo , Animales , Aterosclerosis/inducido químicamente , Aterosclerosis/genética , Biomarcadores , Antígenos CD36/metabolismo , Dieta Alta en Grasa , Regulación de la Expresión Génica , Genotipo , Glutatión/metabolismo , Inflamación , Masculino , Ratones , Ratones Noqueados , Mitocondrias/metabolismo , Mutación , NADP Transhidrogenasas , Receptores de LDL/genética , Superóxidos/metabolismo
10.
J Cell Physiol ; 234(7): 11047-11059, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30536661

RESUMEN

New onset of diabetes is associated with the use of statins. We have recently demonstrated that pravastatin-treated hypercholesterolemic LDL receptor knockout (LDLr-/- ) mice exhibit reductions in insulin secretion and increased islet cell death and oxidative stress. Here, we hypothesized that these diabetogenic effects of pravastatin could be counteracted by treatment with the antioxidant coenzyme Q 10 (CoQ 10 ), an intermediate generated in the cholesterol synthesis pathway. LDLr -/- mice were treated with pravastatin and/or CoQ 10 for 2 months. Pravastatin treatment resulted in a 75% decrease of liver CoQ 10 content. Dietary CoQ 10 supplementation of pravastatin-treated mice reversed fasting hyperglycemia, improved glucose tolerance (20%) and insulin sensitivity (>2-fold), and fully restored islet glucose-stimulated insulin secretion impaired by pravastatin (40%). Pravastatin had no effect on insulin secretion of wild-type mice. In vitro, insulin-secreting INS1E cells cotreated with CoQ 10 were protected from cell death and oxidative stress induced by pravastatin. Simvastatin and atorvastatin were more potent in inducing dose-dependent INS1E cell death (10-15-fold), which were also attenuated by CoQ 10 cotreatment. Together, these results demonstrate that statins impair ß-cell redox balance, function and viability. However, CoQ 10 supplementation can protect the statins detrimental effects on the endocrine pancreas.


Asunto(s)
Hipercolesterolemia/tratamiento farmacológico , Células Secretoras de Insulina/efectos de los fármacos , Pravastatina/efectos adversos , Receptores de LDL/metabolismo , Ubiquinona/análogos & derivados , Animales , Línea Celular , Supervivencia Celular , Diabetes Mellitus/inducido químicamente , Suplementos Dietéticos , Femenino , Prueba de Tolerancia a la Glucosa , Peróxido de Hidrógeno , Insulina , Hígado/metabolismo , Ratones , Ratones Noqueados , Pravastatina/uso terapéutico , Receptores de LDL/genética , Ubiquinona/farmacología
11.
Artículo en Inglés | MEDLINE | ID: mdl-28278125

RESUMEN

Ractopamine (RAC), is a ß-adrenergic agonist increasingly used in the swine and cattle industry. This compound redirects nutrients to favour leanness rather than fat deposition, improves growth and carcass traits gaining higher economic benefit to producers. Countries around the world are split over whether to allow the use of RAC in meat production. Clenbuterol (CLB) and salbutamol (SLB) are anillinic and phenolic ß-agonists, respectively, with the same capacity of producing economic benefits for the meat sector. However, they are prohibited because of the potentially adverse reactions they can cause in consumers. The three ß-agonist compounds have been included in the Brazilian National Regulatory Survey and consequentially there is an eminent need for reliable methods capable of detecting those substances at the same time and reduce analytical costs. Therefore, an LC-MS/MS method for the simultaneous determination of residual RAC, CLB and SAL in swine and cattle muscle was developed and validated with quantification levels respecting the action levels established for Brazil which are 0.1, 0.2 and 5 µg kg-1 for RAC, CLB and SAL, respectively. Samples were quantified using RAC-d5, CLB-d9 and SLB-d6 as internal standards. The validation was performed according to European Union Decision 2002/657, which includes criteria (CCα, CCß, recovery, repeatability, reproducibility and calibration curve). The method meets the Brazilian regulatory requirement that establishes criteria and procedures for the determination of parameters such as CCα, CCß, precision and recovery. CCα values were 0.02, 0.21 and 5.42 µg kg-1 for RAC, CLB and SAL, respectively, in bovine and swine muscle samples; CCß values were 0.03, 0.22 and 5.8 µg kg-1 for RAC, CLB and SAL, respectively, in bovine and swine muscle samples. Average recoveries fortified with 0.05-7.5 µg kg-1 of the studied ß-agonist leads around 95%. The method was demonstrated to be suitable for the determination of RAC, CLB and SLB in swine and cattle muscle samples.


Asunto(s)
Albuterol/análisis , Clenbuterol/análisis , Músculos/química , Fenetilaminas/análisis , Animales , Brasil , Bovinos , Cromatografía Líquida de Alta Presión , Laboratorios , Límite de Detección , Porcinos , Espectrometría de Masas en Tándem
12.
Toxicology ; 344-346: 42-52, 2016 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-26875785

RESUMEN

We have previously demonstrated that hypercholesterolemic LDL receptor knockout (LDLr(-/-)) mice secrete less insulin than wild-type mice. Removing cholesterol from isolated islets using methyl-beta-cyclodextrin reversed this defect. In this study, we hypothesized that in vivo treatment of LDLr(-/-) mice with the HMGCoA reductase inhibitor pravastatin would improve glucose-stimulated insulin secretion. Female LDLr(-/-) mice were treated with pravastatin (400mg/L) for 1-3 months. Isolated pancreatic islets were assayed for insulin secretion rates, intracellular calcium oscillations, cholesterol levels, NAD(P)H and SNARE protein levels, apoptosis indicators and lipidomic profile. Two months pravastatin treatment reduced cholesterol levels in plasma, liver and islets by 35%, 25% and 50%, respectively. Contrary to our hypothesis, pravastatin treatment increased fasting and fed plasma levels of glucose and decreased markedly (40%) fed plasma levels of insulin. In addition, ex vivo glucose stimulated insulin secretion was significantly reduced after two and three months (36-48%, p<0.05) of pravastatin treatment. Although reducing insulin secretion and insulinemia, two months pravastatin treatment did not affect glucose tolerance because it improved global insulin sensitivity. Pravastatin induced islet dysfunction was associated with marked reductions of exocytosis-related SNARE proteins (SNAP25, Syntaxin 1A, VAMP2) and increased apoptosis markers (Bax/Bcl2 protein ratio, cleaved caspase-3 and lower NAD(P)H production rates) observed in pancreatic islets from treated mice. In addition, several oxidized phospholipids, tri- and diacylglycerols and the proapoptotic lipid molecule ceramide were identified as markers of pravastatin-treated islets. Cell death and oxidative stress (H2O2 production) were confirmed in insulin secreting INS-1E cells treated with pravastatin. These results indicate that chronic treatment with pravastatin impairs the insulin exocytosis machinery and increases ß-cell death. These findings suggest that prolonged use of statins may have a diabetogenic effect.


Asunto(s)
Exocitosis/efectos de los fármacos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/toxicidad , Hipercolesterolemia/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Insulina/metabolismo , Pravastatina/toxicidad , Animales , Esquema de Medicación , Exocitosis/fisiología , Femenino , Hipercolesterolemia/genética , Hipercolesterolemia/patología , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de LDL/deficiencia , Receptores de LDL/genética
13.
Circ Res ; 116(11): 1744-52, 2015 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-25868464

RESUMEN

RATIONALE: Coronary artery disease, the direct result of atherosclerosis, is the most common cause of death in Western societies. Vascular smooth muscle cell (VSMC) apoptosis occurs during the progression of atherosclerosis and in advanced lesions and promotes plaque necrosis, a common feature of high-risk/vulnerable atherosclerotic plaques. Akt1, a serine/threonine protein kinase, regulates several key endothelial cell and VSMC functions including cell growth, migration, survival, and vascular tone. Although global deficiency of Akt1 results in impaired angiogenesis and massive atherosclerosis, the specific contribution of VSMC Akt1 remains poorly characterized. OBJECTIVE: To investigate the contribution of VSMC Akt1 during atherogenesis and in established atherosclerotic plaques. METHODS AND RESULTS: We generated 2 mouse models in which Akt1 expression can be suppressed specifically in VSCMs before (Apoe(-/-)Akt1(fl/fl)Sm22α(CRE)) and after (Apoe(-/-)Akt1(fl/fl)SM-MHC-CreER(T2E)) the formation of atherosclerotic plaques. This approach allows us to interrogate the role of Akt1 during the initial and late steps of atherogenesis. The absence of Akt1 in VSMCs during the progression of atherosclerosis results in larger atherosclerotic plaques characterized by bigger necrotic core areas, enhanced VSMC apoptosis, and reduced fibrous cap and collagen content. In contrast, VSMC Akt1 inhibition in established atherosclerotic plaques does not influence lesion size but markedly reduces the relative fibrous cap area in plaques and increases VSMC apoptosis. CONCLUSIONS: Akt1 expression in VSMCs influences early and late stages of atherosclerosis. The absence of Akt1 in VSMCs induces features of plaque vulnerability including fibrous cap thinning and extensive necrotic core areas. These observations suggest that interventions enhancing Akt1 expression specifically in VSMCs may lessen plaque progression.


Asunto(s)
Aterosclerosis/fisiopatología , Miocitos del Músculo Liso/metabolismo , Placa Aterosclerótica/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Apoptosis/genética , Aterosclerosis/genética , Aterosclerosis/metabolismo , Western Blotting , Proliferación Celular/genética , Células Cultivadas , Citocinas/genética , Progresión de la Enfermedad , Técnica del Anticuerpo Fluorescente , Expresión Génica , Macrófagos/metabolismo , Macrófagos/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Placa Aterosclerótica/genética , Placa Aterosclerótica/patología , Proteínas Proto-Oncogénicas c-akt/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo
14.
FASEB J ; 29(2): 597-610, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25392271

RESUMEN

Atherosclerosis is the major cause of death and disability in diabetic and obese subjects with insulin resistance. Akt2, a phosphoinositide-dependent serine-threonine protein kinase, is highly express in insulin-responsive tissues; however, its role during the progression of atherosclerosis remains unknown. Thus, we aimed to investigate the contribution of Akt2 during the progression of atherosclerosis. We found that germ-line Akt2-deficient mice develop similar atherosclerotic plaques as wild-type mice despite higher plasma lipids and glucose levels. It is noteworthy that transplantation of bone marrow cells isolated from Akt2(-/-) mice to Ldlr(-/-) mice results in marked reduction of the progression of atherosclerosis compared with Ldlr(-/-) mice transplanted with wild-type bone marrow cells. In vitro studies indicate that Akt2 is required for macrophage migration in response to proatherogenic cytokines (monocyte chemotactic protein-1 and macrophage colony-stimulating factor). Moreover, Akt2(-/-) macrophages accumulate less cholesterol and have an alternative activated or M2-type phenotype when stimulated with proinflammatory cytokines. Together, these results provide evidence that macrophage Akt2 regulates migration, the inflammatory response and cholesterol metabolism and suggest that targeting Akt2 in macrophages might be beneficial for treating atherosclerosis.


Asunto(s)
Aterosclerosis/fisiopatología , Proteínas Proto-Oncogénicas c-akt/deficiencia , Proteínas Proto-Oncogénicas c-akt/genética , Animales , Glucemia/metabolismo , Células de la Médula Ósea/citología , Trasplante de Médula Ósea , Movimiento Celular , Colesterol/metabolismo , Citocinas/metabolismo , Progresión de la Enfermedad , Inflamación , Insulina/química , Leucocitos/citología , Lípidos/sangre , Lipoproteínas LDL/metabolismo , Macrófagos/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal , Microscopía Fluorescente , Placa Aterosclerótica , Receptores de LDL/genética
15.
J AOAC Int ; 97(5): 1476-80, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25903003

RESUMEN

Boldenone, an androgenic steroid, is forbidden for use in meat production in most countries worldwide. Residues of this drug in food present a potential risk to consumers. A sensitive LC/MS/MS method for analysis of 17ß-boldenone using boldenone-d3 as an internal standard was developed. An enzymatic hydrolysis and extraction using ethyl acetate, methanol, and hexane were performed in the sample preparation. Parameters such as decision limit (CCα), detection capability (CCß), precision, recovery, and ruggedness were evaluated according to the Brazilian Regulation 24/2009 (equivalent to European Union Decision 2002/657/EC) and International Organization for Standardization/International Electrotechnical Commission 17025:2005. CCα and CCß were determined to be 0.17 and 0.29 µg/kg, respectively. Average recoveries from bovine liver samples fortified with 1, 1.5, and 2 µg/kg were around 100%. A complete statistical analysis was performed on the results obtained, including an estimation of the method uncertainty. The method is considered robust after being subjected to day-to-day analytical variations and has been used as a standard method in Brazil to report boldenone levels in bovine liver.


Asunto(s)
Cromatografía Liquida/métodos , Residuos de Medicamentos/análisis , Hígado/química , Espectrometría de Masas en Tándem/métodos , Testosterona/análogos & derivados , Animales , Bovinos , Testosterona/análisis
16.
Artículo en Inglés | MEDLINE | ID: mdl-23701281

RESUMEN

Nitroimidazoles are a class of veterinary drugs used for the treatment and prevention of certain bacterial and protozoal diseases in poultry, swine dysentery and genital trichomoniasis in cattle. Since the safety assessment of nitroimidazoles showed them to be genotoxic, carcinogenic and mutagenic, a number of nitroimidazoles have been banned for therapeutic purposes in different countries. Moreover, nitroimidazoles have also been extensively used as growth promoters in food-producing animals. Due to their efficacious improvement in meat production and feed conversion, deliberate use still exists. Therefore, the illegal use of nitroimidazoles in animal husbandry must be monitored. A sensitive method based on LC-MS/MS for the simultaneous determination and confirmation of five banned nitroimidazole drugs including metronidazole, ronidazole, dimetridazole, metronidazole-OH (metabolite of metronidazole), and 2-hydroxymethyl-1-methyl-5-nitroimidazole (metabolite of ronidazole and dimetridazole) in bovine muscle, using ronidazole-d3 as an internal standard, was developed and validated. After extraction with ethyl acetate and evaporation, the nitroimidazoles were reconstituted in petroleum ether and purified, and LC-MS/MS analysis was performed. The method was validated according to Brazilian Regulation 24/2009 (equivalent to European Union Decision 2002/657/EC). Parameters such as decision limit (CCα), detection capability (CCß), precision, accuracy, uncertaincy and ruggedness were determined. Average accuracy of the five nitroimidazoles from bovine muscle fortified at 5 levels (0.5, 1.0, 1.5, 2.0 and 2.5 µg kg(-1)) ranged from 96% to 103%. The calculated CCα ranged from 0.0 to 0.17 µg kg(-1); CCß ranged from 0.08 to 0.41 µg kg(-1). A complete statistical analysis was performed and the results indicate that the method is robust when subjected to day-to-day analytical variations.


Asunto(s)
Antiinfecciosos/análisis , Cromatografía Liquida/métodos , Residuos de Medicamentos/análisis , Músculos/química , Espectrometría de Masas en Tándem/métodos , Animales , Bovinos , Límite de Detección , Estándares de Referencia , Reproducibilidad de los Resultados
17.
J AOAC Int ; 96(1): 212-5, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23513979

RESUMEN

Sulfonamides are one class of antimicrobial agents used in aquaculture production. Sulfonamides are often overused because they are inexpensive and readily available. Their presence at a concentration above the legal limits is a potential hazard to human health. Brazilian authorities have included in the National Regulatory Monitoring Program the control of the three most widely used sulfonamides in aquaculture production, i.e., sulfathiazole, sulfamethazine, and sulfadimethoxine. An LC method with UV detection for the determination of residual sulfonamides in fish muscle, using sulfapyridine as an internal standard has been developed and validated. The validation was performed according to the Brazilian Regulation 24/2009 (equivalent to European Union Decision 2002/657/EC). The method meets the Brazilian regulatory requirement that establishes criteria and procedures for determination of parameters such as decision limit (CCalpha), detection capability (CCbeta), precision, and recovery. For fish muscle, CCalpha was determined at 3.63, 2.91, and 7.46 microg/kg for sulfathiazole, sulfamethazine, and sulfadimethoxine, respectively. CCbeta was 9.39, 14.54, and 9.39 microg/kg for sulfathiazole, sulfamethazine, and sulfadimethoxine, respectively. For shrimp, CCalpha was 11.5, 8.67, and 4.46 microg/kg for sulfathiazole, sulfamethazine, and sulfadimethoxine, respectively. CCbeta was 18, 11.93, and 5.24 microg/kg for sulfathiazole, sulfamethazine, and sulfadimethoxine, respectively. A complete statistical analysis was performed on the results obtained. The results indicate that the method is robust when subjected to day-to-day analytical variations.


Asunto(s)
Cromatografía Liquida/métodos , Residuos de Medicamentos/análisis , Sulfonamidas/análisis , Animales , Unión Europea , Peces , Músculos/química , Penaeidae , Espectrofotometría Ultravioleta
18.
Drug Test Anal ; 4 Suppl 1: 125-9, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22851369

RESUMEN

Florfenicol, a derivative of thiamphenicol in which the hydroxyl group at C-3 has been replaced with fluorine, is listed by the World Health Organization as an antibacterial agent for human medicine that is critically important in risk management strategies for non-human use. AOAC International has also identified it as an important molecule for the development of effective methods for the seafood sector. Following inspection missions from the European Union and United States of America, it was introduced in the Brazilian Residues Control Program to fulfill export and internal national requirements with a Maximum Residue Limit of 800 µg/kg. A high performance liquid chromatography method with ultraviolet detection at a wavelength of 230 nm (λ = 230 nm) for the detection of florfenicol in fish muscle was developed and validated according to the Brazilian Regulation 24/2009 (equivalent to European Union Decision 2002/657/EC). Fish samples were extracted with ethyl acetate and hexane followed by C18 solid phase clean-up and chromatographic separation on a reversed-phase C18 LC column with acetonitrile:water as mobile phase. The method results were also was compared with those obtained using liquid chromatography-tandem quadrupole mass spectrometry. The method meets the Brazilian regulatory requirements with a decision limit (CCα) of 840 µg/kg and detection capability (CCß) of 879 µg/kg. This method is easy to use and has been implemented into Brazil's residue control program, with liquid chromatography-tandem mass spectrometry (LC-MS/MS) confirmation of any suspect samples using the same method.


Asunto(s)
Antibacterianos/análisis , Residuos de Medicamentos/análisis , Peces/metabolismo , Alimentos Marinos/análisis , Espectrometría de Masas en Tándem/métodos , Tianfenicol/análogos & derivados , Animales , Antibacterianos/aislamiento & purificación , Brasil , Cromatografía Liquida/métodos , Residuos de Medicamentos/aislamiento & purificación , Músculos/metabolismo , Reproducibilidad de los Resultados , Extracción en Fase Sólida/métodos , Tianfenicol/análisis , Tianfenicol/aislamiento & purificación
19.
Cell Cycle ; 11(5): 922-33, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22333591

RESUMEN

Cholesterol metabolism is tightly regulated at the cellular level and is essential for cellular growth. microRNAs (miRNAs), a class of noncoding RNAs, have emerged as critical regulators of gene expression, acting predominantly at posttranscriptional level. Recent work from our group and others has shown that hsa-miR-33a and hsa-miR-33b, miRNAs located within intronic sequences of the Srebp genes, regulate cholesterol and fatty acid metabolism in concert with their host genes. Here, we show that hsa-miR-33 family members modulate the expression of genes involved in cell cycle regulation and cell proliferation. MiR-33 inhibits the expression of the cyclin-dependent kinase 6 (CDK6) and cyclin D1 (CCND1), thereby reducing cell proliferation and cell cycle progression. Overexpression of miR-33 induces a significant G 1 cell cycle arrest in Huh7 and A549 cell lines. Most importantly, inhibition of miR-33 expression using 2'fluoro/methoxyethyl-modified (2'F/MOE-modified) phosphorothioate backbone antisense oligonucleotides improves liver regeneration after partial hepatectomy (PH) in mice, suggesting an important role for miR-33 in regulating hepatocyte proliferation during liver regeneration. Altogether, these results suggest that Srebp/miR-33 locus may cooperate to regulate cell proliferation, cell cycle progression and may also be relevant to human liver regeneration.


Asunto(s)
MicroARNs/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ciclina D1/metabolismo , Quinasa 6 Dependiente de la Ciclina/metabolismo , Puntos de Control de la Fase G1 del Ciclo Celular , Células HeLa , Humanos , Regeneración Hepática/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Oligonucleótidos Antisentido/química , Oligonucleótidos Antisentido/farmacología , Fosfatos/química
20.
Arterioscler Thromb Vasc Biol ; 31(11): 2707-14, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21885853

RESUMEN

OBJECTIVE: The ATP-binding cassette transporter A1 (ABCA1) is a major regulator of macrophage cholesterol efflux and protects cells from excess intracellular cholesterol accumulation; however, the mechanism involved in posttranscriptional regulation of ABCA1 is poorly understood. We previously showed that microRNA-33 (miR-33) is 1 regulator. Here, we investigated the potential contribution of other microRNAs (miRNAs) to posttranscriptional regulation of ABCA1 and macrophage cholesterol efflux. METHODS AND RESULTS: We performed a bioinformatic analysis for identifying miRNA target prediction sites in ABCA1 gene and an unbiased genome-wide screen to identify miRNAs modulated by cholesterol excess in mouse peritoneal macrophages. Quantitative real-time reverse transcription-polymerase chain reaction confirmed that miR-758 is repressed in cholesterol-loaded macrophages. Under physiological conditions, high dietary fat excess in mice repressed miR-758 both in peritoneal macrophages and, to a lesser extent, in the liver. In mouse and human cells in vitro, miR-758 repressed the expression of ABCA1, and conversely, the inhibition of this miRNA by using anti-miR-758 increased ABCA1 expression. In mouse cells, miR-758 reduced cellular cholesterol efflux to apolipoprotein A1 (apoA1), and anti-miR-758 increased it. miR-758 directly targets the 3'-untranslated region of Abca1 as assessed by 3'-untranslated region luciferase reporter assays. Interestingly, miR-758 is highly expressed in the brain, where it also targets several genes involved in neurological functions, including Slc38a1, Ntm, Epha7, and Mytl1. CONCLUSION: We identified miR-758 as a novel miRNA that posttranscriptionally controls ABCA1 levels in different cells and regulates macrophage cellular cholesterol efflux to apoA1, opening new avenues to increase apoA1 and raise high-density lipoprotein levels.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Colesterol/metabolismo , MicroARNs/metabolismo , Procesamiento Postranscripcional del ARN/fisiología , Regiones no Traducidas 3'/genética , Regiones no Traducidas 3'/fisiología , Transportador 1 de Casete de Unión a ATP , Transportadoras de Casetes de Unión a ATP/genética , Animales , Apolipoproteína A-I/metabolismo , Astrocitos/metabolismo , Astrocitos/patología , Línea Celular , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , Metabolismo de los Lípidos/fisiología , Macrófagos Peritoneales/metabolismo , Macrófagos Peritoneales/patología , Ratones , MicroARNs/farmacología , Modelos Animales
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...