Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Diabetes Obes Metab ; 25(1): 261-271, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36097728

RESUMEN

AIMS: To evaluate effectiveness and healthcare resource utilization (HCRU) of empagliflozin versus dipeptidyl peptidase-4 inhibitors (DPP-4i) in Swedish clinical practice, as part of the EMPRISE EU study (EUPAS27606, NCT03817463). MATERIALS AND METHODS: A non-interventional, cohort study using retrospectively collected data from Swedish national registries. Adults with type 2 diabetes newly initiated on empagliflozin or DPP-4i from May 2014 to December 2018 were matched 1:1 using propensity scores based on >180 covariates. Cardiovascular outcomes included hospitalization for heart failure (HHF), all-cause mortality (ACM), myocardial infarction (MI), stroke and cardiovascular mortality (CVM), as well as their composite outcomes. Renal outcomes included end-stage renal disease (ESRD), estimated glomerular filtration rate (eGFR) decline to <60 ml/min/1.73 m2 and progression to micro/macroalbuminuria. HCRU outcomes were also assessed. Comparisons were done using Cox proportional hazards and Poisson regression models. RESULTS: Overall, 15,785 matched-pairs were identified, with a mean follow-up of 6.4 and 9.7 months for patients initiating empagliflozin versus DPP-4i, respectively. Empagliflozin was associated with significant reduction in rates of HHF (hazard ratio [HR] = 0.67; 95% confidence interval: 0.49-0.91), ACM (HR = 0.53; 0.41-0.68), HHF + ACM (HR = 0.59; 0.48-0.73), MI + stroke + ACM (HR = 0.68; 0.57-0.81), CVM (HR = 0.46; 0.29-0.73), HHF + CVM (HR = 0.61; 0.47-0.79) and MI + stroke + CVM (HR = 0.79; 0.63-0.98) versus DPP-4i. Empagliflozin also reduced the rates of ESRD (HR = 0.13; 0.03-0.57) and eGFR decline (HR = 0.83; 0.70-0.99). Regarding HCRU, empagliflozin was associated with lower risk of first inpatient stay (HR = 0.87; 0.81-0.93), and lower rate of inpatient and outpatient visits (rate ratio [RR] = 0.85; 0.80-0.89 and RR = 0.96; 0.94-0.98) than DPP-4i. CONCLUSIONS: Empagliflozin treatment compared to DPP-4i reduced cardiorenal events and overall mortality, which may explain lower HCRU among empagliflozin users in Sweden.


Asunto(s)
Diabetes Mellitus Tipo 2 , Inhibidores de la Dipeptidil-Peptidasa IV , Accidente Cerebrovascular , Humanos , Inhibidores de la Dipeptidil-Peptidasa IV/uso terapéutico , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/epidemiología , Estudios de Cohortes , Estudios Retrospectivos , Accidente Cerebrovascular/epidemiología , Accidente Cerebrovascular/prevención & control , Atención a la Salud , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas
2.
Diabetes Obes Metab ; 25(3): 726-734, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36371525

RESUMEN

AIMS: To assess hospital-based care, work absence, associated costs, and mortality in patients with type 2 diabetes with and without established cardiovascular disease (eCVD) compared to matched controls. MATERIALS AND METHODS: In a population-based cohort study, we analysed individual-level data from national health, social insurance and socio-economic registers for people diagnosed with type 2 diabetes before age 70 years and controls (5:1) in Sweden. Regression analysis was used to attribute costs and days absent due to eCVD. Mortality was analysed using Cox proportional hazard regression, stratified by birth year and adjusted for sex and education. RESULTS: Thirty percent (n = 136 135 of 454 983) of people with type 2 diabetes had ≥1 person-year with eCVD (women 24%; men 34%). The mean annual costs of hospital-based care for diabetes complications were EUR 2629 (95% confidence interval [CI] 2601-2657) of which EUR 2337 (95% CI 2309-2365) were attributed to eCVD (89%). The most costly person-years (10th percentile) were observed in a broad subgroup, 42% of people with type 2 diabetes and eCVD. People with type 2 diabetes had on average 146 days absent (95% CI 145-147) per year, of which 68 days (47%; 95% CI 67-70) were attributed to eCVD. The mortality hazard ratio for type 2 diabetes with eCVD was 4.63 (95%CI 4.58-4.68) and without eCVD was 1.86 (95% CI 1.84-1.88) compared to controls without eCVD. CONCLUSION: The sizable burden of eCVD on both the individual with type 2 diabetes and society calls for efficient management in order to reduce the risks for those living with eCVD and to postpone its onset.


Asunto(s)
Enfermedades Cardiovasculares , Diabetes Mellitus Tipo 2 , Masculino , Humanos , Femenino , Anciano , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/epidemiología , Enfermedades Cardiovasculares/complicaciones , Enfermedades Cardiovasculares/epidemiología , Estudios de Cohortes , Centros de Día , Hospitales
3.
Diabetes Obes Metab ; 25(3): 748-757, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36371543

RESUMEN

AIM: To perform a model-based analysis of the short- and long-term health benefits and costs of further increased implementation of empagliflozin for people with type 2 diabetes and established cardiovascular disease (eCVD) in Sweden. MATERIALS AND METHODS: The validated Institute for Health Economics Diabetes Cohort Model (IHE-DCM) was used to estimate health benefits and a 3-year budget impact, and lifetime costs per quality-adjusted life years (QALY) gained of increased implementation of adding empagliflozin to standard of care (SoC) for people with type 2 diabetes and eCVD in a Swedish setting. Scenarios with 100%/75%/50% implementation were explored. Analyses were based on 30 model cohorts with type 2 diabetes and eCVD (n = 131 412 at baseline) from national health data registers. Sensitivity analyses explored the robustness of results. RESULTS: Over 3 years, SoC with empagliflozin (100% implementation) versus SoC before empagliflozin resulted in 7700 total life years gained and reductions in cumulative incidence of cardiovascular deaths by 30% and heart failures by 28%. Annual costs increased by 15% from higher treatment costs and increased survival. Half of these benefits and costs are not yet reached with current implementation below 50%. SoC with empagliflozin yielded 0.37 QALYs per person, with an incremental cost-effectiveness ratio of 16 000 EUR per QALY versus SoC before empagliflozin. CONCLUSIONS: Model simulations using real-world data and trial treatment effects indicated that a broader implementation of empagliflozin, in line with current guidelines for treatment of people with type 2 diabetes and eCVD, would lead to further benefits even from a short-term perspective.


Asunto(s)
Enfermedades Cardiovasculares , Diabetes Mellitus Tipo 2 , Humanos , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/epidemiología , Enfermedades Cardiovasculares/complicaciones , Enfermedades Cardiovasculares/tratamiento farmacológico , Enfermedades Cardiovasculares/epidemiología , Compuestos de Bencidrilo/uso terapéutico , Costos de la Atención en Salud , Análisis Costo-Beneficio , Años de Vida Ajustados por Calidad de Vida
4.
Arterioscler Thromb Vasc Biol ; 42(5): 659-676, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35321563

RESUMEN

BACKGROUND: Understanding the processes behind carotid plaque instability is necessary to develop methods for identification of patients and lesions with stroke risk. Here, we investigated molecular signatures in human plaques stratified by echogenicity as assessed by duplex ultrasound. METHODS: Lesion echogenicity was correlated to microarray gene expression profiles from carotid endarterectomies (n=96). The findings were extended into studies of human and mouse atherosclerotic lesions in situ, followed by functional investigations in vitro in human carotid smooth muscle cells (SMCs). RESULTS: Pathway analyses highlighted muscle differentiation, iron homeostasis, calcification, matrix organization, cell survival balance, and BCLAF1 (BCL2 [B-cell lymphoma 2]-associated transcription factor 1) as the most significant signatures. BCLAF1 was downregulated in echolucent plaques, positively correlated to proliferation and negatively to apoptosis. By immunohistochemistry, BCLAF1 was found in normal medial SMCs. It was repressed early during atherogenesis but reappeared in CD68+ cells in advanced plaques and interacted with BCL2 by proximity ligation assay. In cultured SMCs, BCLAF1 was induced by differentiation factors and mitogens and suppressed by macrophage-conditioned medium. BCLAF1 silencing led to downregulation of BCL2 and SMC markers, reduced proliferation, and increased apoptosis. Transdifferentiation of SMCs by oxLDL (oxidized low-denisty lipoprotein) was accompanied by upregulation of BCLAF1, CD36, and CD68, while oxLDL exposure with BCLAF1 silencing preserved MYH (myosin heavy chain) 11 expression and prevented transdifferentiation. BCLAF1 was associated with expression of cell differentiation, contractility, viability, and inflammatory genes, as well as the scavenger receptors CD36 and CD68. BCLAF1 expression in CD68+/BCL2+ cells of SMC origin was verified in plaques from MYH11 lineage-tracing atherosclerotic mice. Moreover, BCLAF1 downregulation associated with vulnerability parameters and cardiovascular risk in patients with carotid atherosclerosis. CONCLUSIONS: Plaque echogenicity correlated with enrichment of distinct molecular pathways and identified BCLAF1, previously not described in atherosclerosis, as the most significant gene. Functionally, BCLAF1 seems necessary for survival and transdifferentiation of SMCs into a macrophage-like phenotype. The role of BCLAF1 in plaque vulnerability should be further evaluated.


Asunto(s)
Aterosclerosis , Placa Aterosclerótica , Proteínas Represoras/metabolismo , Animales , Aterosclerosis/diagnóstico por imagen , Aterosclerosis/genética , Aterosclerosis/metabolismo , Transdiferenciación Celular , Humanos , Lípidos , Ratones , Miocitos del Músculo Liso/metabolismo , Placa Aterosclerótica/patología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Represoras/genética , Transcriptoma , Proteínas Supresoras de Tumor/genética , Ultrasonografía
5.
EMBO Rep ; 21(7): e49343, 2020 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-32449307

RESUMEN

Regulation of endothelial nutrient transport is poorly understood. Vascular endothelial growth factor B (VEGF-B) signaling in endothelial cells promotes uptake and transcytosis of fatty acids from the bloodstream to the underlying tissue, advancing pathological lipid accumulation and lipotoxicity in diabetic complications. Here, we demonstrate that VEGF-B limits endothelial glucose transport independent of fatty acid uptake. Specifically, VEGF-B signaling impairs recycling of low-density lipoprotein receptor (LDLR) to the plasma membrane, leading to reduced cholesterol uptake and membrane cholesterol loading. Reduced cholesterol levels in the membrane leads to a decrease in glucose transporter 1 (GLUT1)-dependent endothelial glucose uptake. Inhibiting VEGF-B in vivo reconstitutes membrane cholesterol levels and restores glucose uptake, which is of particular relevance for conditions involving insulin resistance and diabetic complications. In summary, our study reveals a mechanism whereby VEGF-B regulates endothelial nutrient uptake and highlights the impact of membrane cholesterol for regulation of endothelial glucose transport.


Asunto(s)
Glucosa , Factor B de Crecimiento Endotelial Vascular , Colesterol , Células Endoteliales/metabolismo , Transcitosis , Factor B de Crecimiento Endotelial Vascular/metabolismo
6.
J Infect Dis ; 222(12): 2041-2051, 2020 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-32852032

RESUMEN

Multiple viruses are implicated in atherosclerosis, but the mechanisms by which they infect cells and contribute to plaque formation in arterial walls are not well understood. Based on reports showing the presence of enterovirus in atherosclerotic plaques we hypothesized that the coxsackievirus and adenovirus receptor (CXADR/CAR), although absent in normal arteries, could be induced during plaque formation. Large-scale microarray and mass spectrometric analyses revealed significant up-regulation of CXADR messenger RNA and protein levels in plaque-invested carotid arteries compared with control arteries. Macrophages were identified as a previously unknown cellular source of CXADR in human plaques and plaques from Ldr-/-Apob100/100 mice. CXADR was specifically associated with M1-polarized macrophages and foam cells and was experimentally induced during macrophage differentiation. Furthermore, it was significantly correlated with receptors for other viruses linked to atherosclerosis. The results show that CXADR is induced in macrophages during plaque formation, suggesting a mechanism by which enterovirus infect cells in atherosclerotic plaques.


Asunto(s)
Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus/metabolismo , Macrófagos/metabolismo , Placa Aterosclerótica/metabolismo , Animales , Arterias Carótidas/virología , Modelos Animales de Enfermedad , Enterovirus/patogenicidad , Humanos , Macrófagos/virología , Ratones , Ratones Noqueados , Placa Aterosclerótica/virología , ARN Mensajero/metabolismo
7.
Arterioscler Thromb Vasc Biol ; 37(3): 534-542, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28062492

RESUMEN

OBJECTIVE: Recently, poliovirus receptor-related 2 (Pvrl2) emerged as a top gene in a global gene expression study aiming to detect plasma cholesterol-responsive genes causally related to atherosclerosis regression in hypercholesterolemic mice. PVRL2 is an adherens junction protein implied to play a role in transendothelial migration of leukocytes, a key feature in atherosclerosis development. In this study, we investigated the effect of Pvrl2 deficiency on atherosclerosis development and transendothelial migration of leukocytes activity. APPROACH AND RESULTS: Pvrl2-deficient mice bred onto an atherosclerosis-prone background (Pvrl2-/-Ldlr-/-Apob100/100) had less atherosclerotic lesions and more stable plaques compared with littermate controls (Pvrl2+/+Ldlr-/-Apob100/100). Pvrl2-/-Ldlr-/-Apob100/100 mice also showed a 49% decrease in transendothelial migration of leukocytes activity observed using the in vivo air pouch model. In accordance, augmented arterial wall expression of Pvrl2 during atherosclerosis progression coincided with an increased gene expression of migrating leukocytes into the vessel wall. Both in human and mice, gene and protein expression of PVRL2 was predominantly observed in the vascular endothelium according to the immunohistochemical and gene expression data. In addition, the cholesterol responsiveness of PVRL2 was also observed in humans. CONCLUSIONS: PVRL2 is a plasma cholesterol-responsive gene acting at endothelial sites of vascular inflammation that could potentially be a new therapeutic target for atherosclerosis prevention through its suggested transendothelial migration of leukocytes modulating activity.


Asunto(s)
Aorta Torácica/metabolismo , Enfermedades de la Aorta/metabolismo , Aterosclerosis/metabolismo , Moléculas de Adhesión Celular/metabolismo , Colesterol/sangre , Endotelio Vascular/metabolismo , Leucocitos/metabolismo , Migración Transendotelial y Transepitelial , Animales , Aorta Torácica/patología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/patología , Apolipoproteína B-100 , Apolipoproteínas B/deficiencia , Apolipoproteínas B/genética , Aterosclerosis/genética , Aterosclerosis/patología , Adhesión Celular , Moléculas de Adhesión Celular/deficiencia , Moléculas de Adhesión Celular/genética , Línea Celular Tumoral , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Endotelio Vascular/patología , Predisposición Genética a la Enfermedad , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Nectinas , Fenotipo , Interferencia de ARN , Receptores de LDL/deficiencia , Receptores de LDL/genética , Transducción de Señal , Factores de Tiempo , Transfección
8.
PLoS Genet ; 10(2): e1004201, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24586211

RESUMEN

Plasma cholesterol lowering (PCL) slows and sometimes prevents progression of atherosclerosis and may even lead to regression. Little is known about how molecular processes in the atherosclerotic arterial wall respond to PCL and modify responses to atherosclerosis regression. We studied atherosclerosis regression and global gene expression responses to PCL (≥80%) and to atherosclerosis regression itself in early, mature, and advanced lesions. In atherosclerotic aortic wall from Ldlr(-/-)Apob (100/100) Mttp (flox/flox)Mx1-Cre mice, atherosclerosis regressed after PCL regardless of lesion stage. However, near-complete regression was observed only in mice with early lesions; mice with mature and advanced lesions were left with regression-resistant, relatively unstable plaque remnants. Atherosclerosis genes responding to PCL before regression, unlike those responding to the regression itself, were enriched in inherited risk for coronary artery disease and myocardial infarction, indicating causality. Inference of transcription factor (TF) regulatory networks of these PCL-responsive gene sets revealed largely different networks in early, mature, and advanced lesions. In early lesions, PPARG was identified as a specific master regulator of the PCL-responsive atherosclerosis TF-regulatory network, whereas in mature and advanced lesions, the specific master regulators were MLL5 and SRSF10/XRN2, respectively. In a THP-1 foam cell model of atherosclerosis regression, siRNA targeting of these master regulators activated the time-point-specific TF-regulatory networks and altered the accumulation of cholesterol esters. We conclude that PCL leads to complete atherosclerosis regression only in mice with early lesions. Identified master regulators and related PCL-responsive TF-regulatory networks will be interesting targets to enhance PCL-mediated regression of mature and advanced atherosclerotic lesions.


Asunto(s)
Aorta/metabolismo , Aterosclerosis/sangre , Colesterol/sangre , Receptores de LDL/genética , Animales , Aorta/efectos de los fármacos , Apolipoproteínas B/genética , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/patología , Regulación de la Expresión Génica/efectos de los fármacos , N-Metiltransferasa de Histona-Lisina/biosíntesis , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/administración & dosificación , Ratones , Ratones Transgénicos , Proteínas Nucleares/biosíntesis , Ribonucleoproteínas/biosíntesis , Factores de Empalme Serina-Arginina
9.
Arterioscler Thromb Vasc Biol ; 34(9): 2068-77, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24925974

RESUMEN

OBJECTIVE: Using a multi-tissue, genome-wide gene expression approach, we recently identified a gene module linked to the extent of human atherosclerosis. This atherosclerosis module was enriched with inherited risk for coronary and carotid artery disease (CAD) and overlapped with genes in the transendothelial migration of leukocyte (TEML) pathway. Among the atherosclerosis module genes, the transcription cofactor Lim domain binding 2 (LDB2) was the most connected in a CAD vascular wall regulatory gene network. Here, we used human genomics and atherosclerosis-prone mice to evaluate the possible role of LDB2 in TEML and atherosclerosis. APPROACH AND RESULTS: mRNA profiles generated from blood macrophages in patients with CAD were used to infer transcription factor regulatory gene networks; Ldlr(-/-)Apob(100/100) mice were used to study the effects of Ldb2 deficiency on TEML activity and atherogenesis. LDB2 was the most connected gene in a transcription factor regulatory network inferred from TEML and atherosclerosis module genes in CAD macrophages. In Ldlr(-/-)Apob(100/100) mice, loss of Ldb2 increased atherosclerotic lesion size ≈2-fold and decreased plaque stability. The exacerbated atherosclerosis was caused by increased TEML activity, as demonstrated in air-pouch and retinal vasculature models in vivo, by ex vivo perfusion of primary leukocytes, and by leukocyte migration in vitro. In THP1 cells, migration was increased by overexpression and decreased by small interfering RNA inhibition of LDB2. A functional LDB2 variant (rs10939673) was associated with the risk and extent of CAD across several cohorts. CONCLUSIONS: As a key driver of the TEML pathway in CAD macrophages, LDB2 is a novel candidate to target CAD by inhibiting the overall activity of TEML.


Asunto(s)
Aterosclerosis/fisiopatología , Enfermedades de las Arterias Carótidas/patología , Quimiotaxis de Leucocito/fisiología , Enfermedad de la Arteria Coronaria/patología , Proteínas con Dominio LIM/fisiología , Factores de Transcripción/fisiología , Migración Transendotelial y Transepitelial/fisiología , Animales , Apolipoproteína B-100/genética , Enfermedades de las Arterias Carótidas/genética , Línea Celular Tumoral , Quimiocina CCL2/farmacología , Enfermedad de la Arteria Coronaria/genética , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Humanos , Proteínas con Dominio LIM/deficiencia , Proteínas con Dominio LIM/genética , Macrófagos/metabolismo , Ratones , Ratones Noqueados , ARN Mensajero/biosíntesis , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Migración Transendotelial y Transepitelial/genética
10.
PLoS Genet ; 5(12): e1000754, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19997623

RESUMEN

Environmental exposures filtered through the genetic make-up of each individual alter the transcriptional repertoire in organs central to metabolic homeostasis, thereby affecting arterial lipid accumulation, inflammation, and the development of coronary artery disease (CAD). The primary aim of the Stockholm Atherosclerosis Gene Expression (STAGE) study was to determine whether there are functionally associated genes (rather than individual genes) important for CAD development. To this end, two-way clustering was used on 278 transcriptional profiles of liver, skeletal muscle, and visceral fat (n = 66/tissue) and atherosclerotic and unaffected arterial wall (n = 40/tissue) isolated from CAD patients during coronary artery bypass surgery. The first step, across all mRNA signals (n = 15,042/12,621 RefSeqs/genes) in each tissue, resulted in a total of 60 tissue clusters (n = 3958 genes). In the second step (performed within tissue clusters), one atherosclerotic lesion (n = 49/48) and one visceral fat (n = 59) cluster segregated the patients into two groups that differed in the extent of coronary stenosis (P = 0.008 and P = 0.00015). The associations of these clusters with coronary atherosclerosis were validated by analyzing carotid atherosclerosis expression profiles. Remarkably, in one cluster (n = 55/54) relating to carotid stenosis (P = 0.04), 27 genes in the two clusters relating to coronary stenosis were confirmed (n = 16/17, P<10(-27 and-30)). Genes in the transendothelial migration of leukocytes (TEML) pathway were overrepresented in all three clusters, referred to as the atherosclerosis module (A-module). In a second validation step, using three independent cohorts, the A-module was found to be genetically enriched with CAD risk by 1.8-fold (P<0.004). The transcription co-factor LIM domain binding 2 (LDB2) was identified as a potential high-hierarchy regulator of the A-module, a notion supported by subnetwork analysis, by cellular and lesion expression of LDB2, and by the expression of 13 TEML genes in Ldb2-deficient arterial wall. Thus, the A-module appears to be important for atherosclerosis development and, together with LDB2, merits further attention in CAD research.


Asunto(s)
Movimiento Celular/genética , Enfermedad de la Arteria Coronaria/genética , Células Endoteliales/patología , Perfilación de la Expresión Génica , Redes Reguladoras de Genes/genética , Leucocitos/patología , Factores de Transcripción/metabolismo , Anciano , Animales , Aterosclerosis/genética , Arterias Carótidas/patología , Análisis por Conglomerados , Estudios de Cohortes , Biología Computacional , Células Endoteliales/metabolismo , Femenino , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Humanos , Proteínas con Dominio LIM , Leucocitos/metabolismo , Masculino , Ratones , Especificidad de Órganos/genética , Reproducibilidad de los Resultados , Suecia , Factores de Transcripción/genética
11.
PLoS Genet ; 4(3): e1000036, 2008 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-18369455

RESUMEN

Despite the well-documented effects of plasma lipid lowering regimes halting atherosclerosis lesion development and reducing morbidity and mortality of coronary artery disease and stroke, the transcriptional response in the atherosclerotic lesion mediating these beneficial effects has not yet been carefully investigated. We performed transcriptional profiling at 10-week intervals in atherosclerosis-prone mice with human-like hypercholesterolemia and a genetic switch to lower plasma lipoproteins (Ldlr(-/-)Apo(100/100)Mttp(flox/flox) Mx1-Cre). Atherosclerotic lesions progressed slowly at first, then expanded rapidly, and plateaued after advanced lesions formed. Analysis of lesion expression profiles indicated that accumulation of lipid-poor macrophages reached a point that led to the rapid expansion phase with accelerated foam-cell formation and inflammation, an interpretation supported by lesion histology. Genetic lowering of plasma cholesterol (e.g., lipoproteins) at this point all together prevented the formation of advanced plaques and parallel transcriptional profiling of the atherosclerotic arterial wall identified 37 cholesterol-responsive genes mediating this effect. Validation by siRNA-inhibition in macrophages incubated with acetylated-LDL revealed a network of eight cholesterol-responsive atherosclerosis genes regulating cholesterol-ester accumulation. Taken together, we have identified a network of atherosclerosis genes that in response to plasma cholesterol-lowering prevents the formation of advanced plaques. This network should be of interest for the development of novel atherosclerosis therapies.


Asunto(s)
Aterosclerosis/genética , Aterosclerosis/metabolismo , Colesterol/metabolismo , Animales , Apolipoproteína B-100/genética , Aterosclerosis/etiología , Aterosclerosis/patología , Proteínas Portadoras/genética , Células Espumosas/metabolismo , Perfilación de la Expresión Génica , Humanos , Macrófagos/metabolismo , Ratones , Ratones Noqueados , Ratones Mutantes , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , Receptores de LDL/deficiencia , Receptores de LDL/genética
12.
Neurobiol Aging ; 89: 12-23, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32143981

RESUMEN

The nigrostriatal dopaminergic system (NDS) controls motor activity, and its impairment during type 2 diabetes (T2D) progression could increase Parkinson's disease risk in diabetics. If so, whether glycemia regulation prevents this impairment needs to be addressed. We investigated whether T2D impairs the NDS and whether dipeptidyl peptidase-4 inhibition (DPP-4i; a clinical strategy against T2D but also neuroprotective in animal models) prevents this effect, in middle-aged mice. Neither T2D (induced by 12 months of high-fat diet) nor aging (14 months) changed striatal dopamine content assessed by high-performance liquid chromatography. However, T2D reduced basal and amphetamine-stimulated striatal extracellular dopamine, assessed by microdialysis. Both the DPP-4i linagliptin and the sulfonylurea glimepiride (an antidiabetic comparator unrelated to DPP-4i) counteracted these effects. The functional T2D-induced effects did not correlate with NDS neuronal/glial alterations. However, aging itself affected striatal neurons/glia, and the glia effects were counteracted mainly by DPP-4i. These findings show NDS functional pathophysiology in T2D and suggest the preventive use of two unrelated anti-T2D drugs. Moreover, DPP-4i counteracted striatal age-related glial alterations suggesting striatal rejuvenation properties.


Asunto(s)
Envejecimiento/metabolismo , Cuerpo Estriado/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Inhibidores de la Dipeptidil-Peptidasa IV/farmacología , Dopamina/metabolismo , Linagliptina/farmacología , Sustancia Negra/metabolismo , Compuestos de Sulfonilurea/farmacología , Animales , Diabetes Mellitus Tipo 2/complicaciones , Progresión de la Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/prevención & control , Riesgo
14.
BMC Med Genomics ; 12(Suppl 6): 108, 2019 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-31345219

RESUMEN

BACKGROUND: Genetic loss-of-function variants (LoFs) associated with disease traits are increasingly recognized as critical evidence for the selection of therapeutic targets. We integrated the analysis of genetic and clinical data from 10,511 individuals in the Mount Sinai BioMe Biobank to identify genes with loss-of-function variants (LoFs) significantly associated with cardiovascular disease (CVD) traits, and used RNA-sequence data of seven metabolic and vascular tissues isolated from 600 CVD patients in the Stockholm-Tartu Atherosclerosis Reverse Network Engineering Task (STARNET) study for validation. We also carried out in vitro functional studies of several candidate genes, and in vivo studies of one gene. RESULTS: We identified LoFs in 433 genes significantly associated with at least one of 10 major CVD traits. Next, we used RNA-sequence data from the STARNET study to validate 115 of the 433 LoF harboring-genes in that their expression levels were concordantly associated with corresponding CVD traits. Together with the documented hepatic lipid-lowering gene, APOC3, the expression levels of six additional liver LoF-genes were positively associated with levels of plasma lipids in STARNET. Candidate LoF-genes were subjected to gene silencing in HepG2 cells with marked overall effects on cellular LDLR, levels of triglycerides and on secreted APOB100 and PCSK9. In addition, we identified novel LoFs in DGAT2 associated with lower plasma cholesterol and glucose levels in BioMe that were also confirmed in STARNET, and showed a selective DGAT2-inhibitor in C57BL/6 mice not only significantly lowered fasting glucose levels but also affected body weight. CONCLUSION: In sum, by integrating genetic and electronic medical record data, and leveraging one of the world's largest human RNA-sequence datasets (STARNET), we identified known and novel CVD-trait related genes that may serve as targets for CVD therapeutics and as such merit further investigation.


Asunto(s)
Enfermedades Cardiovasculares/genética , Genómica , Mutación , Enfermedades Cardiovasculares/sangre , Colesterol/sangre , Genotipo , Humanos , Triglicéridos/sangre
15.
Pediatr Diabetes ; 9(5): 472-9, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18721168

RESUMEN

OBJECTIVE: The aim of this study was to compare safety, metabolic control, and treatment satisfaction in children/adolescents at onset of type 1 diabetes mellitus who were treated with either continuous subcutaneous insulin infusion (CSII) or multiple daily injections (MDI). RESEARCH DESIGN AND METHODS: Seventy-two children/adolescents (7-17 yr of age) were enrolled in this open, randomized, parallel, multicenter study. Approximately half of the patients were treated with MDI (natural protamine hagedorn [NPH] insulin twice daily and rapid-acting insulin three to -four times daily, n = 38) by pen, and the other half received CSII (n = 34). The patients were followed for 24 months with clinical visits at the entry of the study and after 1, 6, 12, and 24 months. During these visits, hemoglobin A1c, insulin doses, weight, and height were registered. Severe episodes of hypoglycemia and ketoacidosis as well as technical problems were recorded. In addition, the patients/parents answered the Diabetes Treatment Satisfaction Questionnaire. RESULTS: There was no significant difference in metabolic control between the treatment groups. Treatment satisfaction was significantly higher in the group treated with CSII compared with the MDI group (p

Asunto(s)
Diabetes Mellitus Tipo 1/tratamiento farmacológico , Sistemas de Infusión de Insulina , Insulina/administración & dosificación , Satisfacción del Paciente , Adolescente , Glucemia/metabolismo , Niño , Femenino , Hemoglobina Glucada/metabolismo , Humanos , Hiperglucemia/sangre , Hipoglucemia/sangre , Hipoglucemiantes/administración & dosificación , Inyecciones Subcutáneas , Masculino
16.
Int J Mol Med ; 21(6): 819-24, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18506377

RESUMEN

The peroxisome proliferator-activated receptor delta (PPARdelta) is a transcription factor that regulates genes of importance in lipid and glucose metabolism. ApoA-II is one of the major proteins of the HDL-particle. The aim of this study was to investigate the regulation of apoA-II gene expression by PPARdelta. Treatment of HepG2 cells with the PPARdelta specific agonist GW501516 increased apoA-II mRNA expression. Likewise, reporter gene assays using a construct containing 2.7 kb of the proximal apoA-II promoter showed increased activity after treatment with GW501516, both in HepG2 and in HuH-7 cells. Mutation of two putative PPAR response elements (PPREs) in this region showed that the PPRE at position -737/-717 is the functional site. Binding of PPARdelta to this site was confirmed by chromatin immunoprecipitation and gel retardation analyses. In conclusion, PPARdelta increases the expression of the human apoA-II gene in liver cells via a PPRE in the proximal promoter.


Asunto(s)
Apolipoproteína A-II/genética , Expresión Génica/efectos de los fármacos , PPAR delta/agonistas , Tiazoles/farmacología , Apolipoproteína A-II/metabolismo , Secuencia de Bases , Sitios de Unión/genética , Metabolismo de los Hidratos de Carbono/efectos de los fármacos , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Inmunoprecipitación de Cromatina/métodos , Ensayo de Cambio de Movilidad Electroforética/métodos , Humanos , Metabolismo de los Lípidos/efectos de los fármacos , Mutación , PPAR delta/genética , PPAR delta/metabolismo , Regiones Promotoras Genéticas/genética , Unión Proteica/efectos de los fármacos , Elementos de Respuesta/genética , Homología de Secuencia de Ácido Nucleico
17.
PeerJ ; 6: e4466, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29527417

RESUMEN

RNA editing modifies transcripts and may alter their regulation or function. In humans, the most common modification is adenosine to inosine (A-to-I). We examined the global characteristics of RNA editing in 4,301 human tissue samples. More than 1.6 million A-to-I edits were identified in 62% of all protein-coding transcripts. mRNA recoding was extremely rare; only 11 novel recoding sites were uncovered. Thirty single nucleotide polymorphisms from genome-wide association studies were associated with RNA editing; one that influences type 2 diabetes (rs2028299) was associated with editing in ARPIN. Twenty-five genes, including LRP11 and PLIN5, had editing sites that were associated with plasma lipid levels. Our findings provide new insights into the genetic regulation of RNA editing and establish a rich catalogue for further exploration of this process.

18.
Acta Neuropathol Commun ; 6(1): 14, 2018 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-29471869

RESUMEN

Recent data suggest that olfactory deficits could represent an early marker and a pathogenic mechanism at the basis of cognitive decline in type 2 diabetes (T2D). However, research is needed to further characterize olfactory deficits in diabetes, their relation to cognitive decline and underlying mechanisms.The aim of this study was to determine whether T2D impairs odour detection, olfactory memory as well as neuroplasticity in two major brain areas responsible for olfaction and odour coding: the main olfactory bulb (MOB) and the piriform cortex (PC), respectively. Dipeptidyl peptidase-4 inhibitors (DPP-4i) are clinically used T2D drugs exerting also beneficial effects in the brain. Therefore, we aimed to determine whether DPP-4i could reverse the potentially detrimental effects of T2D on the olfactory system.Non-diabetic Wistar and T2D Goto-Kakizaki rats, untreated or treated for 16 weeks with the DPP-4i linagliptin, were employed. Odour detection and olfactory memory were assessed by using the block, the habituation-dishabituation and the buried pellet tests. We assessed neuroplasticity in the MOB by quantifying adult neurogenesis and GABAergic inhibitory interneurons positive for calbindin, parvalbumin and carletinin. In the PC, neuroplasticity was assessed by quantifying the same populations of interneurons and a newly identified form of olfactory neuroplasticity mediated by post-mitotic doublecortin (DCX) + immature neurons.We show that T2D dramatically reduced odour detection and olfactory memory. Moreover, T2D decreased neurogenesis in the MOB, impaired the differentiation of DCX+ immature neurons in the PC and altered GABAergic interneurons protein expression in both olfactory areas. DPP-4i did not improve odour detection and olfactory memory. However, it normalized T2D-induced effects on neuroplasticity.The results provide new knowledge on the detrimental effects of T2D on the olfactory system. This knowledge could constitute essentials for understanding the interplay between T2D and cognitive decline and for designing effective preventive therapies.


Asunto(s)
Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Inhibidores de la Dipeptidil-Peptidasa IV/farmacología , Linagliptina/farmacología , Nootrópicos/farmacología , Percepción Olfatoria/efectos de los fármacos , Animales , Diabetes Mellitus Experimental/fisiopatología , Diabetes Mellitus Experimental/psicología , Diabetes Mellitus Tipo 2/patología , Diabetes Mellitus Tipo 2/fisiopatología , Diabetes Mellitus Tipo 2/psicología , Dipeptidil Peptidasa 4/metabolismo , Proteína Doblecortina , Neuronas GABAérgicas/efectos de los fármacos , Neuronas GABAérgicas/patología , Neuronas GABAérgicas/fisiología , Interneuronas/efectos de los fármacos , Interneuronas/patología , Interneuronas/fisiología , Masculino , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/patología , Trastornos de la Memoria/fisiopatología , Neurogénesis/efectos de los fármacos , Neurogénesis/fisiología , Plasticidad Neuronal/efectos de los fármacos , Plasticidad Neuronal/fisiología , Bulbo Olfatorio/efectos de los fármacos , Bulbo Olfatorio/patología , Bulbo Olfatorio/fisiopatología , Percepción Olfatoria/fisiología , Corteza Piriforme/efectos de los fármacos , Corteza Piriforme/patología , Corteza Piriforme/fisiopatología , Ratas Wistar
19.
Atherosclerosis ; 267: 39-48, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29100060

RESUMEN

BACKGROUND AND AIMS: Mitochondrial damage and augmented production of reactive oxygen species (ROS) may represent an intermediate step by which hypercholesterolemia exacerbates atherosclerotic lesion formation. METHODS: To test this hypothesis, in mice with severe but genetically reversible hypercholesterolemia (i.e. the so called Reversa mouse model), we performed time-resolved analyses of mitochondrial transcriptome in the aortic arch employing a systems-level network approach. RESULTS: During hypercholesterolemia, we observed a massive down-regulation (>28%) of mitochondrial genes, specifically at the time of rapid atherosclerotic lesion expansion and foam cell formation, i.e. between 30 and 40 weeks of age. Both phenomena - down-regulation of mitochondrial genes and lesion expansion - were largely reversible by genetically lowering plasma cholesterol (by >80%, from 427 to 54 ± 31 mg/L) at 30 weeks. Co-expression network analysis revealed that both mitochondrial signature genes were highly connected in two modules, negatively correlating with lesion size and supported as causal for coronary artery disease (CAD) in humans, as expression-associated single nucleotide polymorphisms (eSNPs) representing their genes overlapped markedly with established disease risk loci. Within these modules, we identified the transcription factor estrogen related receptor (ERR)-α and its co-factors PGC1-α and -ß, i.e. two members of the peroxisome proliferator-activated receptor γ co-activator 1 family of transcription regulators, as key regulatory genes. Together, these factors are known as major orchestrators of mitochondrial biogenesis and antioxidant responses. CONCLUSIONS: Using a network approach, we demonstrate how hypercholesterolemia could hamper mitochondrial activity during atherosclerosis progression and pinpoint potential therapeutic targets to counteract these processes.


Asunto(s)
Aterosclerosis/metabolismo , Enfermedad de la Arteria Coronaria/metabolismo , Regulación de la Expresión Génica , Genes Mitocondriales , Hipercolesterolemia/metabolismo , Animales , Antioxidantes/metabolismo , Aorta Torácica/metabolismo , Sitios de Unión , Proteínas Portadoras/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Perfilación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Humanos , Ratones , Mitocondrias/metabolismo , Proteínas Nucleares/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Polimorfismo de Nucleótido Simple , Proteínas de Unión al ARN , Especies Reactivas de Oxígeno/metabolismo , Receptores de Estrógenos/metabolismo , Factores de Riesgo , Biología de Sistemas , Factores de Transcripción/metabolismo , Transcriptoma , Receptor Relacionado con Estrógeno ERRalfa
20.
Diabetes ; 54(4): 1157-63, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15793256

RESUMEN

Activators of peroxisome proliferator-activated receptor (PPAR)gamma have been studied intensively for their insulin-sensitizing properties and antidiabetic effects. Recently, a specific PPARdelta activator (GW501516) was reported to attenuate plasma glucose and insulin levels when administered to genetically obese ob/ob mice. This study was performed to determine whether specific activation of PPARdelta has direct effects on insulin action in skeletal muscle. Specific activation of PPARdelta using two pharmacological agonists (GW501516 and GW0742) increased glucose uptake independently of insulin in differentiated C2C12 myotubes. In cultured primary human skeletal myotubes, GW501516 increased glucose uptake independently of insulin and enhanced subsequent insulin stimulation. PPARdelta agonists increased the respective phosphorylation and expression of AMP-activated protein kinase 1.9-fold (P < 0.05) and 1.8-fold (P < 0.05), of extracellular signal-regulated kinase 1/2 mitogen-activated protein kinase (MAPK) 2.2-fold (P < 0.05) and 1.7-fold (P < 0.05), and of p38 MAPK 1.2-fold (P < 0.05) and 1.4-fold (P < 0.05). Basal and insulin-stimulated protein kinase B/Akt was unaltered in cells preexposed to PPARdelta agonists. Preincubation of myotubes with the p38 MAPK inhibitor SB203580 reduced insulin- and PPARdelta-mediated increase in glucose uptake, whereas the mitogen-activated protein kinase kinase inhibitor PD98059 was without effect. PPARdelta agonists reduced mRNA expression of PPARdelta, sterol regulatory element binding protein (SREBP)-1a, and SREBP-1c (P < 0.05). In contrast, mRNA expression of PPARgamma, PPARgamma coactivator 1, GLUT1, and GLUT4 was unaltered. Our results provide evidence to suggest that PPARdelta agonists increase glucose metabolism and promote gene regulatory responses in cultured human skeletal muscle. Moreover, we provide biological validation of PPARdelta as a potential target for antidiabetic therapy.


Asunto(s)
Glucosa/metabolismo , Fibras Musculares Esqueléticas/metabolismo , PPAR delta/fisiología , Adipocitos , Animales , Transporte Biológico/efectos de los fármacos , Transporte Biológico/fisiología , Línea Celular , Células Cultivadas , Fibroblastos , Flavonoides/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Imidazoles/farmacología , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Fibras Musculares Esqueléticas/efectos de los fármacos , Músculo Esquelético/citología , PPAR delta/agonistas , Piridinas/farmacología , Tiazoles/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA