Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 87
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Yale J Biol Med ; 95(1): 129-152, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35370493

RESUMEN

High oxidative stress, Th1/Th17 immune response, M1 macrophage inflammation, and cell death are associated with cardiovascular diseases. Controlled oxidative stress, Th2/Treg anti-tumor immune response, M2 macrophage inflammation, and survival are associated with cancer. MiR-21 protects against cardiovascular diseases but may induce tumor growth by retaining the anti-inflammatory M2 macrophage and Treg phenotypes and inhibiting apoptosis. Down-regulation of let-7, miR-1, miR-9, miR-16, miR-20a, miR-22a, miR-23a, miR-24a, miR-26a, miR-29, miR-30a, miR-34a, miR-124, miR-128, miR-130a, miR-133, miR-140, miR-143-145, miR-150, miR-153, miR-181a, miR-378, and miR-383 may aid cancer cells to escape from stresses. Upregulation of miR-146 and miR-223 may reduce anti-tumor immune response together with miR-21 that also protects against apoptosis. MiR-155 and silencing of let-7e, miR-125, and miR-126 increase anti-tumor immune response. MiR expression depends on oxidative stress, cytokines, MYC, and TGF-ß, and expression of silencing lncRNAs and circ-RNAs. However, one lncRNA or circ-RNA may have opposite effects by targeting several miRs. For example, PVT1 induces apoptosis by targeting miR-16a and miR-30a but inhibits apoptosis by silencing miR-17. In addition, levels of a non-coding RNA in a cell type depend not only on expression in that cell type but also on an exchange of microvesicles between cell types and tumors. Although we got more insight into the function of a growing number of individual non-coding RNAs, overall, we do not know enough how several of them interact in functional networks and how their expression changes at different stages of disease progression.


Asunto(s)
Enfermedades Cardiovasculares , MicroARNs , Neoplasias , ARN Largo no Codificante , Enfermedades Cardiovasculares/genética , Humanos , Inflamación/genética , Inflamación/metabolismo , MicroARNs/genética , Neoplasias/genética , Estrés Oxidativo , ARN Largo no Codificante/genética
2.
Adv Exp Med Biol ; 998: 45-58, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28936731

RESUMEN

Extracellular vesicles are now widely recognized as key players in the prevention, repair or progression of cardiovascular disease. Here we first focus on the functional roles of extracellular vesicles in the cross-talk between cardiomyocytes and endothelial cells, important for maintaining normal development and function of the heart. Second, we discuss the role of extracellular vesicles secreted by embryonic and non-embryonic stem cells in repairing cardiomyocyte function and in restoring angiogenic potential after myocardial ischemia-reperfusion injury. Third, we focus on the role of extracellular vesicles in Endothelial to Mesenchymal Transition (EndMT), leading to conversion of endothelial cells to fibroblasts, secretion of extracellular proteins collagen and fibronectin, and fibrosis. Finally, we discuss the role of extracellular vesicles secreted under stress by endothelial cells, macrophages and vascular smooth muscle cells in atherosclerosis. On aggregate, the reviewed preclinical studies present evidence that extracellular vesicles secreted by cardiomyocytes, fibroblasts, endothelial cells, immune-system-related cells, vascular smooth muscle cells and stem cells play an important role in the pathogenesis of cardiovascular disease. However, further studies are needed to gain better insight into the mechanisms used to select specific content to transfer to specific target cells, and to induce or repress signaling pathways in their target cells.


Asunto(s)
Enfermedad de la Arteria Coronaria/metabolismo , Vasos Coronarios/metabolismo , Exosomas/metabolismo , Cardiopatías/metabolismo , Miocardio/metabolismo , Transducción de Señal , Animales , Comunicación Celular , Enfermedad de la Arteria Coronaria/patología , Vasos Coronarios/patología , Células Endoteliales/metabolismo , Células Endoteliales/patología , Transición Epitelial-Mesenquimal , Exosomas/patología , Cardiopatías/patología , Humanos , Miocardio/patología , Placa Aterosclerótica , Células Madre/metabolismo , Células Madre/patología
3.
Curr Opin Lipidol ; 26(5): 412-9, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26309202

RESUMEN

PURPOSE OF REVIEW: Microvesicles, in general, and exosomes together with their delivered content in particular, are now being widely recognized as key players in atherosclerosis. We have previously reviewed the role of microvesicles in atherosclerosis pathogenesis, diagnosis and therapy. Here, we focus on the roles of exosomes and discuss their emergent role in mediating activation and response to inflammation, vessel infiltration and induction of coagulation. We will finally give an outlook to discuss novel detection techniques and systems biology based data analyses to investigate exosome-mediated cell-to-cell communication. RECENT FINDINGS: Recent research points to a role of exosomes in delivering apoptotic and inflammatory content between blood cells and vascular cells, with a potential contribution of exosomes secreted by adipose tissue. An atheroprotective role of exosomes in response to coagulation that may contrast with the procoagulatory role of platelet-derived larger microvesicles is envisaged. New detection and separation methods and systems biology techniques are emerging. CONCLUSION: We project that the development of novel detection, separation and analysis mechanism and systems-based analysis methods will further unravel the paracrine and endocrine 'communication protocol' between cellular players in atherosclerosis, mediating inflammation, oxidative stress and apoptosis.


Asunto(s)
Aterosclerosis/metabolismo , Exosomas/fisiología , Microvasos/metabolismo , Animales , Apoptosis , Células Endoteliales/fisiología , Transferencia de Gen Horizontal , Humanos , Microvasos/patología , Estrés Oxidativo , Comunicación Paracrina , Transducción de Señal
4.
Eur Heart J ; 35(43): 3013-20, 3020a-3020d, 2014 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-25230814

RESUMEN

Blood vessels are exposed to multiple mechanical forces that are exerted on the vessel wall (radial, circumferential and longitudinal forces) or on the endothelial surface (shear stress). The stresses and strains experienced by arteries influence the initiation of atherosclerotic lesions, which develop at regions of arteries that are exposed to complex blood flow. In addition, plaque progression and eventually plaque rupture is influenced by a complex interaction between biological and mechanical factors-mechanical forces regulate the cellular and molecular composition of plaques and, conversely, the composition of plaques determines their ability to withstand mechanical load. A deeper understanding of these interactions is essential for designing new therapeutic strategies to prevent lesion development and promote plaque stabilization. Moreover, integrating clinical imaging techniques with finite element modelling techniques allows for detailed examination of local morphological and biomechanical characteristics of atherosclerotic lesions that may be of help in prediction of future events. In this ESC Position Paper on biomechanical factors in atherosclerosis, we summarize the current 'state of the art' on the interface between mechanical forces and atherosclerotic plaque biology and identify potential clinical applications and key questions for future research.


Asunto(s)
Arterias/fisiología , Aterosclerosis/fisiopatología , Apoptosis/fisiología , Biomarcadores/metabolismo , Fenómenos Biomecánicos/fisiología , Proliferación Celular/fisiología , Senescencia Celular/fisiología , Progresión de la Enfermedad , Células Endoteliales/fisiología , Endotelio Vascular/fisiología , Homeostasis/fisiología , Humanos , Mecanorreceptores/fisiología , Placa Aterosclerótica/fisiopatología , Rotura Espontánea/fisiopatología , Transducción de Señal/fisiología , Estrés Mecánico , Remodelación Vascular/fisiología
5.
J Proteome Res ; 13(3): 1731-43, 2014 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-24479691

RESUMEN

Peroxisome proliferator-activated receptors (PPAR) play an important role in the regulation of lipid and glucose metabolism, inflammatory, and vascular responses. We show the effect of treatment with two PPAR agonists, fenofibrate (FF) and rosiglitazone (RSG), on ob/ob and LDLR-double deficient mice, by combined gene-expression and metabolomic analyses. Male mice were daily treated for 12 weeks with RSG (10 mg·kg(1-)·day(-1) per os (p.o.), n = 8) and FF (50 mg·kg(1-)·day(-1) p.o., n = 8). Twelve untreated ob/ob and LDLR-double deficient mice were used as controls. To integrate the transcriptomic and metabolomic results, we designed a hierarchical algorithm, based on the average linkage method in clustering. Data were also interpreted with the Ingenuity Pathway Analysis program. FF and RSG treatments significantly increased the hepatic triglyceride content in the liver when compared with the control group, and the treatments induced an increase in the number and size of hepatic lipid droplets. Both drugs simultaneously activate pro-steatotic and antisteatotic metabolic pathways with a well-ordered result of aggravation of the hepatic lipid accumulation. The present study is a cautionary note not only to researchers on the basic mechanism of the action of PPAR activators but also to the use of these compounds in clinical practice.


Asunto(s)
Hígado Graso/metabolismo , Fenofibrato/efectos adversos , Hiperlipidemias/metabolismo , Hipoglucemiantes/efectos adversos , Hipolipemiantes/efectos adversos , Obesidad/metabolismo , Tiazolidinedionas/efectos adversos , Animales , Modelos Animales de Enfermedad , Hígado Graso/inducido químicamente , Hígado Graso/tratamiento farmacológico , Hígado Graso/patología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Hiperlipidemias/tratamiento farmacológico , Hiperlipidemias/patología , Hígado/química , Hígado/metabolismo , Hígado/patología , Masculino , Metaboloma , Ratones , Ratones Transgénicos , Obesidad/tratamiento farmacológico , Obesidad/patología , Receptores Activados del Proliferador del Peroxisoma/agonistas , Receptores Activados del Proliferador del Peroxisoma/genética , Receptores Activados del Proliferador del Peroxisoma/metabolismo , Mapeo de Interacción de Proteínas , Rosiglitazona , Transducción de Señal , Transcriptoma
6.
Cancers (Basel) ; 15(4)2023 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-36831498

RESUMEN

Mitochondria in cancer cells tend to overproduce reactive oxygen species (ROS), inducing a vicious cycle between mitochondria, ROS, genomic instability, and cancer development. The first part of this review deals with the role of noncoding RNAs in regulating mitochondrial ROS production and the expression of antioxidants in cancer cells, preventing the increase of ROS in the tumor microenvironment. In addition, cytotoxic T and natural killer cells release high levels of ROS, inducing cell death, while anti-immune regulatory T cells, tumor-associated M2 macrophages, and myeloid-derived suppressor cells, at least at the initial stage of tumor growth, release low levels of ROS supporting tumor growth. Therefore, this review's second part deals with noncoding RNAs' role in regulating the metabolic reprogramming of immune cells about ROS release. Furthermore, the enrichment of noncoding RNAs in microvesicles allows communication between cell types in a tumor and between a tumor and tumor-adjacent tissues. Therefore, the third part illustrates how noncoding RNA-containing microvesicles secreted by mesenchymal stem cells and primary tumor cells may primarily aid the shift of immune cells to a pro-oncogenic phenotype. Conversely, microvesicles released by tumor-adjacent tissues may have the opposite effect. Our review reveals that a specific noncoding RNA may affect oxidative stress by several mechanisms, which may have opposite effects on tumor growth. Furthermore, they may be involved in mechanisms other than regulating oxidative stress, which may level out their effects on oxidative stress and tumor growth. In addition, several noncoding RNAs might share a specific function, making it very unlikely that intervening with only one of these noncoding RNAs will block this particular mechanism. Overall, further validation of the interaction between noncoding RNAs about cancer types and stages of tumor development is warranted.

7.
Cells ; 12(24)2023 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-38132104

RESUMEN

We performed a systematic search of the PubMed database for English-language articles related to the function of adipose-derived stem cells in the pathogenesis of cardiovascular diseases. In preclinical models, adipose-derived stem cells protected arteries and the heart from oxidative stress and inflammation and preserved angiogenesis. However, clinical trials did not reiterate successful treatments with these cells in preclinical models. The low success in patients may be due to aging and metabolic reprogramming associated with the loss of proliferation capacity and increased senescence of stem cells, loss of mitochondrial function, increased oxidative stress and inflammation, and adipogenesis with increased lipid deposition associated with the low potential to induce endothelial cell function and angiogenesis, cardiomyocyte survival, and restore heart function. Then, we identify noncoding RNAs that may be mechanistically related to these dysfunctions of human adipose-derived stem cells. In particular, a decrease in let-7, miR-17-92, miR-21, miR-145, and miR-221 led to the loss of their function with obesity, type 2 diabetes, oxidative stress, and inflammation. An increase in miR-34a, miR-486-5p, and mir-24-3p contributed to the loss of function, with a noteworthy increase in miR-34a with age. In contrast, miR-146a and miR-210 may protect stem cells. However, a systematic analysis of other noncoding RNAs in human adipose-derived stem cells is warranted. Overall, this review gives insight into modes to improve the functionality of human adipose-derived stem cells.


Asunto(s)
Enfermedades Cardiovasculares , Diabetes Mellitus Tipo 2 , Células Madre Mesenquimatosas , MicroARNs , Humanos , MicroARNs/metabolismo , Enfermedades Cardiovasculares/terapia , Enfermedades Cardiovasculares/metabolismo , Reprogramación Metabólica , Diabetes Mellitus Tipo 2/metabolismo , Células Madre Mesenquimatosas/metabolismo , Envejecimiento , Células Madre/metabolismo , Inflamación/patología
8.
Curr Atheroscler Rep ; 14(3): 264-76, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22350585

RESUMEN

High levels of reactive oxygen species (ROS) are observed in chronic human diseases such as obesity, type 2 diabetes, atherosclerosis, and cardiovascular diseases. In addition to the presence of oxidative stress, these diseases are also characterized by deregulated inflammatory responses. Our first aim is to discuss distinct molecular pathways that determine the rate of mitochondrial ROS (mtROS) production and identify agents and enzymes that disrupt the balance between ROS generation and ROS elimination. Recent studies exploring the mechanisms linking ROS and inflammation found that ROS derived from mitochondria act as signal-transducing molecules that provoke endothelial dysfunction associated with uncoupling of nitric oxide synthase, induce the infiltration and activation of inflammatory cells, and increase apoptosis of endothelial and vascular smooth muscle cells. Therefore, our second aim is to give a comprehensive overview of the role of mtROS in all these processes contributing to atherosclerotic lesion progression and causing plaque erosion and rupture. Our third aim is to emphasize the role of the inflammatory toll-like receptor 2/NF-κB signaling pathway in the induction of pro-inflammatory cytokines and mtROS production in relation to insulin resistance, type 2 diabetes, and atherosclerosis. Because mtROS play an active role in several pathogenic mechanisms there is need for mitochondria-targeted antioxidants. Preliminary experiments in cell and animal models of cardiovascular diseases showed that some mitochondria-targeted antioxidants indeed reduce ROS production. However, wide-spread use in humans requires the development of specific and sensitive assays to evaluate mitochondrial oxidative stress and the development of orally active compounds.


Asunto(s)
Aterosclerosis/metabolismo , Mitocondrias/metabolismo , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Animales , Humanos , Factores de Riesgo , Transducción de Señal
9.
FASEB J ; 25(8): 2515-27, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21507901

RESUMEN

A primary event in atherogenesis is the infiltration of activated inflammatory cells into the arterial wall. There they secrete reactive oxygen species and oxidize lipoproteins, inducing foam cell formation and endothelial cell apoptosis, which in turn lead to plaque growth, erosion, and rupture. In addition, there is evidence that this vicious circle between oxidative stress and inflammation occurs not only in the diseased arterial wall but also in adipose tissues in obesity. In this condition, oxidative stress and inflammation impair adipocyte maturation, resulting in defective insulin action and adipocytokine signaling. This observation raises questions regarding what molecules are probably common regulators of these pathogenic processes in adipose and vascular tissues. Candidates are small, noncoding, microRNAs (miRs) that control gene expression by inducing mRNA degradation or blocking translation. This review summarizes recent insights into the roles of miRs in regulation of oxidative stress and inflammation in vascular and adipose tissues. It emphasizes the role of miR-containing microvesicles in the interaction between inflammatory cells and endothelial cells within these tissues and in communication between these tissues, possibly explaining the similarity and the simultaneity of molecular changes and interactions in adipose and vascular tissues.


Asunto(s)
Aterosclerosis/genética , Inflamación/genética , MicroARNs/genética , Obesidad/genética , Estrés Oxidativo/genética , Adipocitos/metabolismo , Adipocitos/patología , Animales , Apoptosis/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Diferenciación Celular/genética , Humanos , Microvasos/metabolismo , Microvasos/patología , Modelos Biológicos , Neovascularización Patológica/genética , Obesidad/metabolismo , Obesidad/patología , Transducción de Señal
10.
Redox Biol ; 44: 102011, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34049219

RESUMEN

Iron deficiency is known to aggravate the prognosis of patients with heart failure. Iron has functions in the mitochondrial respiratory chain. In patients with reduced mitochondrial respiration, the mitochondrial ratio between the level of nicotinamide adenine dinucleotide and its reduced form decreases. Due to the mitochondrial-lysosomal interplay, decreased mitochondrial respiration also leads to inhibition of lysosomal hydrolysis. As a result, cobalamin and iron will be trapped in lysosomes. This will, even if iron and cobalamin have been consumed and absorbed in sufficient amounts, lead to their functional deficiencies.1 Functional iron deficiency can further impede mitochondrial respiration. Increased plasma levels of methylmalonic acid were shown to predict all-cause and cardiovascular mortality in the general population. Treatments targeting mitochondrial and lysosomal function may correct the functional deficiencies and improve prognosis in a subgroup of patients with heart failure, notably those with skeletal muscle wasting. Methylmalonic acid levels may be used for monitoring response to treatment, thereby identifying patients of the subgroup in which disease outcome may improve.


Asunto(s)
Insuficiencia Cardíaca , Ácido Metilmalónico , Homocisteína , Humanos , Hierro , Lisosomas , Vitamina B 12
11.
Artículo en Inglés | MEDLINE | ID: mdl-33557426

RESUMEN

Previously, miR-1, miR-122, miR-126, miR-132, miR-133, and miR-370 were found to be related to coronary artery disease (CAD) progression. However, their relationship with subclinical atherosclerosis, especially in subjects with metabolic syndrome, is unknown. Therefore, our aim was to determine their relationship with arterial markers of subclinical atherosclerosis. Metabolic syndrome subjects (n = 182) with high cardiovascular risk but without overt cardiovascular disease (CVD) were recruited from the Lithuanian High Cardiovascular Risk (LitHiR) primary prevention program. The ardio-ankle vascular index (CAVI), augmentation index normalized to a heart rate of 75 bpm (AIxHR75), aortic pulse wave velocity (AoPWV), and carotid artery stiffness were assessed. MicroRNAs (miRs) were analyzed in serum. Pearson correlation and a univariate linear regression t-test showed that miR-1, miR-133b, and miR-133a were negatively associated with CAVI mean, whereas miR-122 was positively associated. MiR-1, miR-133b and miR-133a, and miR-145 were negatively associated with AIxHR75. MiR-122 correlated negatively with AoPWV. In multivariate linear regression models, miR-133b and miR-122 predicted CAVImean, miR-133 predicted AIxHR75, and miR-122 predicted AoPWV. MiR-132 predicted right carotid artery stiffness, and miR-1 predicted left carotid artery stiffness. The addition of smoking to miR-133b and miR-122 enhanced the prediction of CAVI. Age and triglycerides enhanced the prediction of AoPWV by miR-122. A cluster of four miRs are related to subclinical atherosclerosis in subjects with metabolic syndrome. Combined, they may have a more substantial diagnostic or prognostic value than any single miR. Future follow-up studies are needed to establish their clinical relevance.


Asunto(s)
Aterosclerosis , Síndrome Metabólico , MicroARNs , Rigidez Vascular , Aterosclerosis/genética , Arterias Carótidas , Humanos , Síndrome Metabólico/genética , MicroARNs/genética , Análisis de la Onda del Pulso
12.
J Cell Mol Med ; 14(1-2): 70-8, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19968738

RESUMEN

The initial event in atherogenesis is the increased transcytosis of low density lipoprotein, and its subsequent deposition, retention and modification in the subendothelium. It is followed by the infiltration of activated inflammatory cells from the coronary circulation into the arterial wall. There they secrete reactive oxygen species (ROS) and produce oxidized lipoproteins capable of inducing endothelial cell apoptosis, and thereby plaque erosion. Activated T lymphocytes, macrophages and mast cells, accumulate in the eroded plaque where they secrete a variety of proteases capable of inducing degradation of extracellular proteins, thereby rendering the plaques more prone to rupture. This review summarizes the recent advancements in the understanding of the roles of ROS and oxidized lipoproteins in the activation of inflammatory cells and inducing signalling pathways related to cell death and apoptosis. In addition, it presents evidence that this vicious circle between oxidative stress and inflammation does not only occur in the diseased arterial wall, but also in adipose tissues. There, oxidative stress and inflammation impair adipocyte maturation resulting in defective insulin action and adipocytokine signalling. The latter is associated with increased infiltration of inflammatory cells, loss of anti-oxidant protection and cell death in the arterial wall.


Asunto(s)
Aterosclerosis , Inflamación , Estrés Oxidativo/fisiología , Tejido Adiposo/citología , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Tejido Adiposo/fisiopatología , Aterosclerosis/patología , Aterosclerosis/fisiopatología , Humanos , Inflamación/patología , Inflamación/fisiopatología , Lipoproteínas/química , Lipoproteínas/metabolismo , Macrófagos/citología , Macrófagos/metabolismo , Oxidación-Reducción , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/fisiología
13.
Am J Epidemiol ; 172(2): 180-9, 2010 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-20519263

RESUMEN

Change and fluctuation in body mass index (BMI; weight (kg)/height (m)(2)) may be associated differently with coronary artery calcification (CAC) than with carotid artery intima-media thickness (IMT). The authors analyzed data on 2,243 participants in the Coronary Artery Risk Development in Young Adults (CARDIA) Study, initially aged 18-30 years, who were examined every 2-5 years over a 20-year period (1985-2006). BMI at year 0 was associated positively and linearly with CAC at year 20; however, the association of BMI with year 20 CAC became progressively U-shaped in subsequent examinations (years 10, 15, and 20). To understand the deepening U shape, the authors modeled year 20 BMI and its history using 3 indices: year 0 BMI, linear slope of BMI during 20 years, and BMI fluctuation during 20 years. In models including these 3 terms, year 0 BMI was associated positively with CAC, as was BMI fluctuation. However, adjusted odds ratios across quintiles of BMI slope (vs. the lowest quintile) were 0.7, 0.4, 0.5, and 0.4 (P(trend) < 0.01), suggesting higher risk of CAC with weight loss, plateauing after moderate weight gain. In contrast, IMT was associated positively with BMI at all examinations and with 20-year BMI slope and was unassociated with BMI fluctuation. Surprisingly, CAC risk was higher with BMI loss and lower with BMI gain, whereas associations with IMT were as expected.


Asunto(s)
Peso Corporal , Arteria Carótida Común/patología , Vasos Coronarios/patología , Túnica Íntima/patología , Túnica Media/patología , Adolescente , Adulto , Factores de Edad , Índice de Masa Corporal , Conductas Relacionadas con la Salud , Humanos , Estudios Longitudinales , Persona de Mediana Edad , Factores Sexuales
14.
Circ Res ; 103(10): 1181-9, 2008 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-18818405

RESUMEN

Thrombospondin (TSP)1 is implicated in various inflammatory processes, but its role in atherosclerotic plaque formation and progression is unclear. Therefore, the development of atherosclerosis was compared in ApoE(-/-) and Tsp1(-/-)ApoE(-/-) mice kept on a normocholesterolemic diet. At 6 months, morphometric analysis of the aortic root of both mouse genotypes showed comparable lesion areas. Even when plaque burden increased approximately 5-fold in ApoE(-/-) and 10-fold in Tsp1(-/-)ApoE(-/-) mice, during the subsequent 3 months, total plaque areas were comparable at 9 months. In contrast, plaque composition differed substantially between genotypes: smooth muscle cell areas, mostly located in the fibrous cap of ApoE(-/-) plaques, both at 6 and 9 months, were 3-fold smaller in Tsp1(-/-)ApoE(-/-) plaques, which, in addition, were also more fibrotic. Moreover, inflammation by macrophages was twice as high in Tsp1(-/-)ApoE(-/-) plaques. This correlated with a 30-fold elevated incidence of elastic lamina degradation, with matrix metalloproteinase-9 accumulation, underneath plaques and manifestation of ectasia, exclusively in Tsp1(-/-)ApoE(-/-) mice. At 9 months, the necrotic core was 1.4-fold larger and 4-fold higher numbers of undigested disintegrated apoptotic cells were found in Tsp1(-/-)ApoE(-/-) plaques. Phagocytosis of platelets by cultured Tsp1(-/-) macrophages revealed the instrumental role of TSP1 in phagocytosis, corroborating the defective intraplaque phagocytosis of apoptotic cells. Hence, the altered smooth muscle cell phenotype in Tsp1(-/-)ApoE(-/-) mice has limited quantitative impact on atherosclerosis, but defective TSP1-mediated phagocytosis enhanced plaque necrotic core formation, accelerating inflammation and macrophage-induced elastin degradation by metalloproteinases, speeding up plaque maturation and vessel wall degeneration.


Asunto(s)
Apolipoproteínas E , Aterosclerosis/metabolismo , Macrófagos Peritoneales/metabolismo , Miocitos del Músculo Liso/metabolismo , Fagocitosis , Trombospondina 1 , Animales , Apolipoproteínas E/genética , Apoptosis/genética , Aterosclerosis/genética , Aterosclerosis/patología , Plaquetas/metabolismo , Plaquetas/patología , Elastina/genética , Elastina/metabolismo , Genotipo , Macrófagos Peritoneales/patología , Ratones , Ratones Noqueados , Miocitos del Músculo Liso/patología , Necrosis/metabolismo , Necrosis/patología , Fagocitosis/genética , Trombospondina 1/genética
15.
Arterioscler Thromb Vasc Biol ; 29(12): 2041-6, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19797705

RESUMEN

OBJECTIVE: To study the association of PAF-acetyl hydrolase (PAFAH) activity with inflammation, oxidative stress, and atherosclerosis in hypercholesterolemic swine. METHODS AND RESULTS: Cholesterol-rich diet feeding of miniature pigs was associated with an increase in PAFAH activity and an increase of the PAFAH to PON1 ratio. PLA2G7 RNA (coding for PAFAH) expression was increased in blood monocytes and plaque macrophages. Increased PAFAH activity was associated with higher plasma lysophosphatidylcholine and correlated with oxidized LDL. In THP1 monocytes and macrophages and in human blood-derived macrophages, oxidized LDL induced PLA2G7 RNA expression. Atherogenic diet feeding induced the accumulation of macrophages and oxidized LDL in the arterial wall leading to atherosclerosis. PAFAH activity correlated positively with plaque size and TNFalpha expression in plaque macrophages. CONCLUSIONS: We demonstrated that an increase in PAFAH activity was associated with increased levels of lysophosphatidylcholine, oxidized LDL, and inflammation, resulting in accelerated atherosclerosis in hypercholesterolemic minipigs. The significant correlation between PLA2G7 RNA expression in plaque macrophages and plasma PAFAH activity suggests that the latter is a consequence, rather than a cause of macrophage accumulation. Our cell experiments suggest that oxidized LDL can induce PAFAH, resulting in accumulation of lysophosphatidylcholine that increases the inflammatory action of oxidized LDL.


Asunto(s)
1-Alquil-2-acetilglicerofosfocolina Esterasa/metabolismo , Aterosclerosis/etiología , Aterosclerosis/metabolismo , Hipercolesterolemia/complicaciones , Hipercolesterolemia/metabolismo , Inflamación/etiología , Inflamación/metabolismo , Metabolismo de los Lípidos , 1-Alquil-2-acetilglicerofosfocolina Esterasa/genética , Secuencia de Aminoácidos , Animales , Arildialquilfosfatasa/metabolismo , Aterosclerosis/genética , Aterosclerosis/patología , Vasos Coronarios/patología , Dieta Aterogénica , Expresión Génica , Humanos , Hipercolesterolemia/genética , Inflamación/genética , Macrófagos/metabolismo , Macrófagos/patología , Datos de Secuencia Molecular , Oxidación-Reducción , Estrés Oxidativo , ARN/genética , ARN/metabolismo , Porcinos , Porcinos Enanos
16.
Nat Commun ; 11(1): 5872, 2020 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-33208733

RESUMEN

Substantial evidence implicates crosstalk between metabolic tissues and the immune system in the inception and progression of obesity. However, molecular regulators that orchestrate metaflammation both centrally and peripherally remains incompletely understood. Here, we identify myeloid Krüppel-like factor 2 (KLF2) as an essential regulator of obesity and its sequelae. In mice and humans, consumption of a fatty diet downregulates myeloid KLF2 levels. Under basal conditions, myeloid-specific KLF2 knockout mice (K2KO) exhibit increased feeding and weight gain. High-fat diet (HFD) feeding further exacerbates the K2KO metabolic disease phenotype. Mechanistically, loss of myeloid KLF2 increases metaflammation in peripheral and central tissues. A combination of pair-feeding, bone marrow-transplant, and microglial ablation implicate central and peripheral contributions to K2KO-induced metabolic dysfunction observed. Finally, overexpression of myeloid KLF2 protects mice from HFD-induced obesity and insulin resistance. Together, these data establish myeloid KLF2 as a nodal regulator of central and peripheral metabolic inflammation in homeostasis and disease.


Asunto(s)
Factores de Transcripción de Tipo Kruppel/inmunología , Enfermedades Metabólicas/inmunología , Células Mieloides/inmunología , Obesidad/inmunología , Animales , Sistema Nervioso Central/inmunología , Dieta Alta en Grasa/efectos adversos , Ingestión de Alimentos , Humanos , Inflamación , Resistencia a la Insulina , Factores de Transcripción de Tipo Kruppel/genética , Masculino , Enfermedades Metabólicas/genética , Enfermedades Metabólicas/metabolismo , Enfermedades Metabólicas/fisiopatología , Ratones , Ratones Noqueados , Obesidad/etiología , Obesidad/genética , Obesidad/fisiopatología , Sistema Nervioso Periférico/inmunología
17.
J Lipid Res ; 50(9): 1735-43, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19359705

RESUMEN

Plasma levels of lipoprotein-associated phospholipase A(2) (Lp-PLA(2)) and oxidized low density lipoprotein (oxLDL) have been identified as risk factors for cardiovascular disease. Lp-PLA(2) is the sole enzyme responsible for the hydrolysis of oxidized phospholipids on LDL particles in atherosclerotic plaques. We have studied the relationship between Lp-PLA(2) and oxLDL in carotid endarterectomy (CEA) tissues and in matched plasmas. In extracts from CEA anatomical segments, the levels of oxLDL were significantly associated with the levels of Lp-PLA(2) protein (r = 0.497) and activity (r = 0.615). OxLDL and Lp-PLA(2) mass/activity were most abundant in the carotid bifurcation and internal segments where plaque was most abundant. In extracts from CEA atheroma, the levels of oxLDL and Lp-PLA(2) were significantly correlated (r = 0.634). In matched plasma and atheroma extracts, the levels of Lp-PLA(2) were negatively correlated (r = - 0.578). The ratio of Lp-PLA(2) to oxLDL was higher in atheromatous tissue (277:1) than in normal tissue (135:1) and plasma (13:1). Immunohistochemical experiments indicated that in plaques, oxLDL and Lp-PLA(2) existed in overlapping but distinctly different distribution. Fluorescence microscopy showed both oxLDL and Lp-PLA(2) epitopes on the same LDL particle in plasma but not in plaque. These results suggest that the relationship between Lp-PLA(2) and oxLDL in the atherosclerotic plaque is different from that in the plasma compartment.


Asunto(s)
1-Alquil-2-acetilglicerofosfocolina Esterasa/metabolismo , Enfermedades de las Arterias Carótidas/metabolismo , Lipoproteínas LDL/metabolismo , 1-Alquil-2-acetilglicerofosfocolina Esterasa/sangre , Anciano , Aterosclerosis/metabolismo , Aterosclerosis/patología , Aterosclerosis/cirugía , Transporte Biológico , Arterias Carótidas/citología , Arterias Carótidas/metabolismo , Arterias Carótidas/patología , Arterias Carótidas/cirugía , Enfermedades de las Arterias Carótidas/sangre , Enfermedades de las Arterias Carótidas/patología , Endarterectomía Carotidea , Femenino , Humanos , Lipoproteínas LDL/sangre , Masculino
18.
Diabetes Metab Res Rev ; 25(8): 733-9, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19780064

RESUMEN

BACKGROUND: Accumulating evidence suggests a cross-sectional association between oxidative stress and type 2 diabetes (T2D). Systemic oxidative stress, as measured by oxidized LDL (oxLDL), has been correlated with visceral fat. We examined the relationship between oxLDL, and T2D- and obesity-related traits in a bi-racial sample of 2985 subjects at baseline and after 7 years of follow-up. METHODS: We examined six T2D-related traits (T2D status, HbA(1c), fasting glucose, insulin, adiponectin and HOMA-IR) as well as six obesity-related traits (obesity status, BMI, leptin, % body fat, visceral and subcutaneous fat mass) using logistic and linear regression models. RESULTS: In all subjects at baseline, oxLDL was positively associated with T2D (OR = 1.3, 95% CI:1.1-1.5), fasting glucose (ss = 0.03 +/- 0.006), HbA(1c) (ss = 0.02 +/- 0.004), fasting insulin (ss = 0.12 +/- 0.02), HOMA-IR (ss = 0.13 +/- 0.02) and negatively with adiponectin (ss = -0.16 +/- 0.03), (all p < 0.001). The strength and magnitude of these associations did not differ much between blacks and whites. In both blacks and whites, oxLDL was also associated with obesity (OR = 1.3, 95% CI:1.1-1.4) and three of its related traits (ss = 0.60 +/- 0.14 for BMI, ss = 0.74 +/- 0.17 for % body fat, ss = 0.29 +/- 0.06 for visceral fat; all p < 0.001). Furthermore, of four traits measured after 7 years of follow-up (fasting glucose, HbA1c, BMI and % fat), their relationship with oxLDL was similar to baseline observations. No significant association was found between oxLDL and incident T2D. Interestingly, oxLDL was significantly associated with % change in T2D- and obesity-related traits in whites but not in blacks. CONCLUSION/INTERPRETATION: Our data suggest that systemic oxidative stress may be a novel risk factor for T2D and obesity.


Asunto(s)
Diabetes Mellitus Tipo 2/complicaciones , Lipoproteínas LDL/sangre , Obesidad/complicaciones , Adiponectina/sangre , Negro o Afroamericano/estadística & datos numéricos , Anciano , Glucemia/análisis , Composición Corporal , Índice de Masa Corporal , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/fisiopatología , Femenino , Hemoglobina Glucada/análisis , Humanos , Insulina/sangre , Resistencia a la Insulina , Leptina/sangre , Masculino , Obesidad/sangre , Obesidad/fisiopatología , Oxidación-Reducción , Valor Predictivo de las Pruebas , Estudios Prospectivos , Estadística como Asunto , Encuestas y Cuestionarios , Estados Unidos , Población Blanca/estadística & datos numéricos
19.
Cardiovasc Res ; 77(2): 371-9, 2008 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-18006491

RESUMEN

AIMS: Type II diabetes, often associated with abdominal obesity, frequently leads to heart failure. Clinical and epidemiological evidence suggests that supplemental dyslipidaemia and hypertension, as clustered in the metabolic syndrome, aggravate the cardiovascular outcome. The differential impact of type II diabetes and the metabolic syndrome on left ventricular function, however, remains incompletely defined. METHODS AND RESULTS: We studied left ventricular function in vivo using pressure-volume analysis in obese diabetic mice with leptin deficiency (ob/ob) and obese diabetic dyslipidemic mice with combined leptin and low-density lipoprotein-receptor deficiency (DKO). ob/ob and DKO mice developed a diabetic cardiomyopathy, characterized by impaired contractility and relaxation, from the age of 24 weeks onwards. This was-at least partially-explained by increased apoptosis and disturbed Ca(2+) reuptake in the sarcoplasmic reticulum (SR) in both mouse models. DKO, but not ob/ob, developed increased end-diastolic ventricular stiffness, paralleled by increased left ventricular myocardial fibrosis. Cardiac output was preserved in ob/ob mice by favourable loading conditions, whereas it decreased in DKO mice. CONCLUSIONS: Type II diabetes in mice leads to impaired contractility and relaxation due to disturbed Ca(2+) reuptake in the SR, but only when dyslipidaemia and hypertension are superimposed does vascular-ventricular stiffening increase and left ventricular myocardial fibrosis develop.


Asunto(s)
Diabetes Mellitus Tipo 2/complicaciones , Dislipidemias/fisiopatología , Contracción Miocárdica , Función Ventricular Izquierda , Animales , Presión Sanguínea , Dislipidemias/complicaciones , Femenino , Leptina/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocardio/patología , Receptores de LDL/fisiología , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/fisiología
20.
JCI Insight ; 52019 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-30990466

RESUMEN

Adipose tissue macrophages (ATM) are crucial for maintaining adipose tissue homeostasis and mediating obesity-induced metabolic abnormalities, including prediabetic conditions and type 2 diabetes mellitus. Despite their key functions in regulating adipose tissue metabolic and immunologic homeostasis under normal and obese conditions, a high-resolution transcriptome annotation system that can capture ATM multifaceted activation profiles has not yet been developed. This is primarily attributed to the complexity of their differentiation/activation process in adipose tissue and their diverse activation profiles in response to microenvironmental cues. Although the concept of multifaceted macrophage action is well-accepted, no current model precisely depicts their dynamically regulated in vivo features. To address this knowledge gap, we generated single-cell transcriptome data from primary bone marrow-derived macrophages under polarizing and non-polarizing conditions to develop new high-resolution algorithms. The outcome was creation of a two-index platform, MacSpectrum (https://macspectrum.uconn.edu), that enables comprehensive high-resolution mapping of macrophage activation states from diverse mixed cell populations. MacSpectrum captured dynamic transitions of macrophage subpopulations under both in vitro and in vivo conditions. Importantly, MacSpectrum revealed unique "signature" gene sets in ATMs and circulating monocytes that displayed significant correlation with BMI and homeostasis model assessment of insulin resistance (HOMA-IR) in obese human patients. Thus, MacSpectrum provides unprecedented resolution to decode macrophage heterogeneity and will open new areas of clinical translation.


Asunto(s)
Tejido Adiposo/inmunología , Tejido Adiposo/metabolismo , Activación de Macrófagos/inmunología , Macrófagos/metabolismo , Transcriptoma , Animales , Diferenciación Celular , Diabetes Mellitus Tipo 2/metabolismo , Homeostasis , Humanos , Inflamación , Activación de Macrófagos/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Obesidad/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA