Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Sci Rep ; 14(1): 8196, 2024 04 08.
Article in English | MEDLINE | ID: mdl-38589444

ABSTRACT

In atherosclerotic lesions, monocyte-derived macrophages are major source of interferon gamma (IFN-γ), a pleotropic cytokine known to regulate the expression of numerous genes, including the antiviral gene RSAD2. While RSAD2 was reported to be expressed in endothelial cells of human carotid lesions, its significance for the development of atherosclerosis remains utterly unknown. Here, we harnessed publicly available human carotid atherosclerotic data to explore RSAD2 in lesions and employed siRNA-mediated gene-knockdown to investigate its function in IFN-γ-stimulated human aortic smooth muscle cells (hAoSMCs). Silencing RSAD2 in IFN-γ-stimulated hAoSMCs resulted in reduced expression and secretion of key CXCR3-chemokines, CXCL9, CXCL10, and CXCL11. Conditioned medium from RSAD2-deficient hAoSMCs exhibited diminished monocyte attraction in vitro compared to conditioned medium from control cells. Furthermore, RSAD2 transcript was elevated in carotid lesions where it was expressed by several different cell types, including endothelial cells, macrophages and smooth muscle cells. Interestingly, RSAD2 displayed significant correlations with CXCL10 (r = 0.45, p = 0.010) and CXCL11 (r = 0.53, p = 0.002) in human carotid lesions. Combining our findings, we uncover a novel role for RSAD2 in hAoSMCs, which could potentially contribute to monocyte recruitment in the context of atherosclerosis.


Subject(s)
Atherosclerosis , Plaque, Atherosclerotic , Humans , Plaque, Atherosclerotic/genetics , Interferons , Endothelial Cells/metabolism , Culture Media, Conditioned/pharmacology , Chemokines/genetics , Chemokines/metabolism , Chemokine CXCL11/genetics , Chemokine CXCL11/metabolism , Chemokine CXCL9/metabolism , Interferon-gamma/pharmacology , Interferon-gamma/metabolism , Atherosclerosis/genetics , Myocytes, Smooth Muscle/metabolism , Chemokine CXCL10/genetics , Chemokine CXCL10/metabolism , Receptors, CXCR3/genetics , Receptors, CXCR3/metabolism , Viperin Protein
2.
J Trop Pediatr ; 68(6)2022 10 06.
Article in English | MEDLINE | ID: mdl-36375036

ABSTRACT

BACKGROUND: Atherosclerosis is a cardiovascular disease, highly predictable, and associated with different atherogenic indices (AI) in adults. However, such indexes in the pediatric population are far less explored. The objective of this study was to evaluate the AI and the cardiovascular factors in the pediatric population in the South-Southeast of México. METHODS: A total of 481 children between 2 and 17 years old were recruited. Anthropometric evaluation, blood pressure (BP), lipid profile, apolipoprotein A-I (ApoA-I) and apolipoprotein B (ApoB) were measured, and AI were calculated. The population was grouped by age, binary logistic regression analysis was performed to analysis for associations of AI and cardiovascular risk factors. Sensibility and specificity of AI to detect metabolic alteration were evaluated for curve ROC. RESULTS: The atherogenic risk presented a high prevalence in the pediatric population, such as LDL-c/ApoB (86.9%), AIP (78%) and AC (36.6%). Preschoolers showed a higher risk of ApoB/ApoA-I and ApoB/LDL-c, while adolescents have a high risk of AIP. CRI-I and AC were associated with elements of lipid profile and body mass index (BMI). ROC curves analysis shows that AIP is the best index evaluating metabolic syndrome (MS) (0.87) and dyslipidemia (0.91). CONCLUSION: Such pediatric population showed a high risk of AI, mainly by LDL-c/ApoB and AIP. The BMI was the cardiovascular risk factors most frequently related to AI, AIP is the best index for detecting cases of MS and dyslipidemia. This is the first study carried out in the pediatric population from the South-Southeast of Mexico that evaluated the AI.


Subject(s)
Atherosclerosis , Dyslipidemias , Adolescent , Child , Child, Preschool , Humans , Apolipoprotein A-I , Apolipoproteins B , Atherosclerosis/epidemiology , Cholesterol, LDL , Dyslipidemias/epidemiology , Mexico/epidemiology , Risk Factors
3.
Clin Exp Rheumatol ; 40(1): 173-182, 2022 01.
Article in English | MEDLINE | ID: mdl-34128791

ABSTRACT

Osteopontin (OPN) is a phosphoglycoprotein involved in bone remodelling, wound healing, cell adhesion, tissue remodelling, and immune response that is distributed widely in normal adult tissues. OPN biological activity is regulated by thrombin and matrix metalloproteinases (MMPs) cleavage, where the full-length (OPN-FL) protein and the cleaved OPN-N are associated with autoimmune diseases such as systemic lupus erythematosus (SLE). OPN overexpression has been associated with a predisposition to SLE and bad prognosis since OPN could mediate a sustained polyclonal B cell activation that besides to intracellular OPN (iOPN) form, promote the T follicular helper (TFH) cells and enhance anti-nuclear antibody production. Currently, the role of OPN in lupus nephritis (LN) has been reported and extensively studied; however, no data are available about the potential mechanism of OPN in neuropsychiatric SLE (NPSLE). In this review, we highlighted the contribution of OPN and iOPN in LN and NPSLE immunopathology.


Subject(s)
Lupus Erythematosus, Systemic , Lupus Nephritis , Lupus Vasculitis, Central Nervous System , Humans , Osteopontin , Prognosis
5.
Vascul Pharmacol ; 141: 106927, 2021 12.
Article in English | MEDLINE | ID: mdl-34715373

ABSTRACT

Abdominal aortic aneurysm (AAA) is associated with increased plasma levels of microRNA (miR) -10b. 5 nmols of miR-10b or miR control was administrated to Apolipoprotein E-deficient mice three days prior implantation of osmotic mini-pumps containing angiotensin II, and for three additional times once a week, which increased expression of miR-10b in plasma. Animals receiving miR-10b had a mortality rate due to aortic rupture of 61% compared to 11% in the miR controls (p < 0.05). Further, miR- 10b resulted in an increased aneurysm formation and growth (p < 0.05), which was accompanied by increased elastin degradation, neutrophil and mast cell markers (p < 0.05). In conclusion, miR-10b is functionally affecting aneurysm development and rupture and not only a marker of AAA. More mechanistic studies are required to better understand miR-10b's role in AAA formation.


Subject(s)
Aortic Aneurysm, Abdominal , Aortic Rupture , MicroRNAs , Angiotensin II/metabolism , Animals , Aorta, Abdominal/metabolism , Aortic Aneurysm, Abdominal/chemically induced , Aortic Aneurysm, Abdominal/genetics , Aortic Rupture/genetics , Aortic Rupture/metabolism , Disease Models, Animal , Mice , Mice, Inbred C57BL , Mice, Knockout, ApoE , MicroRNAs/genetics , MicroRNAs/metabolism
6.
PLoS One ; 16(10): e0259114, 2021.
Article in English | MEDLINE | ID: mdl-34705865

ABSTRACT

INTRODUCTION: Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by the production of autoantibodies such as anti-Sm. Studies in patients with SLE and murine models of lupus reveal that the most critical anti-Sm autoantibodies are predominantly direct against D1(83-119), D2, and B´/B epitopes. OBJECTIVES: The present study aimed to analyze the induction of antigen-specific tolerance after prophylactic immunization with a DNA vaccine encoding the epitopes: D183-119, D2, B´/B, and B´/BCOOH in co-vaccination with IFN-γ or IL-10 in a murine model of lupus induced by pristane. MATERIAL AND METHODS: To obtain endotoxin-free DNA vaccines, direct cloning techniques using pcDNA were performed: D183-119, D2, B´/B, B´/BCOOH, IFN-γ, or IL-10. Lupus was induced by 0.5 mL of pristane via intraperitoneal in BALB/c female mice. Immunoprecipitation with K562 cells was metabolically labeled with 35S and ELISA to detect serum antibodies or mice IgG1, IgG2a isotypes. ELISA determined IL-10 and IFN-γ from splenocytes supernatants. Proteinuria was assessed monthly, and lupus nephritis was evaluated by immunofluorescence, and electron microscopy. RESULTS: The prophylactic co-vaccination with D2/IL-10 reduced the expression of kidney damage observed by electron microscopy, direct immunofluorescence, and H & E, along with reduced level of anti-nRNP/Sm antibodies (P = 0.048). CONCLUSION: The prophylactic co-vaccination of IL-10 with D2 in pristane-induced lupus ameliorates the renal damage maybe by acting as prophylactic DNA tolerizing therapy.


Subject(s)
Interleukin-10 , Lupus Erythematosus, Systemic/prevention & control , Vaccines, DNA , Animals , Autoantibodies/immunology , Autoantigens/immunology , Female , Interleukin-10/administration & dosage , Interleukin-10/pharmacology , Mice , Mice, Inbred BALB C , Therapies, Investigational , Vaccination , Vaccines, DNA/administration & dosage , Vaccines, DNA/pharmacology
7.
J Am Heart Assoc ; 10(17): e020231, 2021 09 07.
Article in English | MEDLINE | ID: mdl-34420357

ABSTRACT

Background While numerous interventions effectively interfered with abdominal aortic aneurysm (AAA) formation/progression in preclinical models, none of the successes translated into clinical success. Hence, a systematic exploration of parallel and divergent processes in clinical AAA disease and its 2 primary models (the porcine pancreatic elastase and angiotensin-II infusion [AngII] murine model) was performed to identify mechanisms relevant for aneurysm disease. Methods and Results This study combines Movat staining and pathway analysis for histological and genomic comparisons between clinical disease and its models. The impact of a notable genomic signal for metabolic reprogramming was tested in a rescue trial (AngII model) evaluating the impact of 1-(4-pyridinyl)-3-(2-quinolinyl)-2-propen-1-one (PFK15)-mediated interference with main glycolytic switch PFKFB3. Histological evaluation characterized the AngII model as a dissection model that is accompanied by adventitial fibrosis. The porcine pancreatic elastase model showed a transient inflammatory response and aortic dilatation, followed by stabilization and fibrosis. Normalization of the genomic responses at day 14 confirmed the self-limiting nature of the porcine pancreatic elastase model. Clear parallel genomic responses with activated adaptive immune responses, and particularly strong signals for metabolic switching were observed in human AAA and the AngII model. Rescue intervention with the glycolysis inhibitor PFK15 in the AngII model showed that interference with the glycolytic switching quenches aneurysm formation. Conclusions Despite clear morphological contrasts, remarkable genomic parallels exist for clinical AAA disease and the AngII model. The metabolic response appears causatively involved in AAA progression and provides a novel therapeutic target. The clear transient genomic response classifies the porcine pancreatic elastase model as a disease initiation model.


Subject(s)
Aortic Aneurysm, Abdominal , Angiotensin II , Animals , Aorta, Abdominal/pathology , Aortic Aneurysm, Abdominal/chemically induced , Aortic Aneurysm, Abdominal/genetics , Aortic Aneurysm, Abdominal/pathology , Disease Models, Animal , Fibrosis , Genomics , Humans , Mice , Mice, Inbred C57BL , Pancreatic Elastase , Swine
8.
J Endovasc Ther ; 27(3): 468-472, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32193989

ABSTRACT

Purpose: To validate a new 2D-3D registration method of fusion imaging during aortic repair in a system prepared only for 3D-3D registration and to compare radiation doses and accuracy. Materials and Methods: The study involved 189 patients, including 94 patients (median age 70 years; 85 men) who underwent abdominal endovascular aneurysm repair (EVAR) with 2D-3D fusion on an Artis zee imaging system and 95 EVAR patients (median age 70 years; 81 men) from a prior study who had 3D-3D registration done using cone beam computed tomography (CBCT). For the 2D-3D registration, an offline CBCT of the empty operating table was imported into the intraoperative dataset and superimposed on the preoperative computed tomography angiogram (CTA). Then 2 intraoperative single-frame 2D images of the skeleton were aligned with the patient's skeleton on the preoperative CTA to complete the registration process. A digital subtraction angiogram was done to correct any misalignment of the aortic CTA volume. Values are given as the median [interquartile range (IQR) Q1, Q3]. Results: The 2D-3D registration had an accuracy of 4.0 mm (IQR 3.0, 5.0) after bone matching compared with the final correction with DSA (78% within 5 mm). By applying the 2D-3D protocol the radiation exposure (dose area product) from the registration of the fusion image was significantly reduced compared with the 3D-3D registration [1.12 Gy∙cm2 (IQR 0.41, 2.14) vs 43.4 Gy∙cm2 (IQR 37.1, 49.0), respectively; p<0.001). Conclusion: The new 2D-3D registration protocol based on 2 single-frame images avoids an intraoperative CBCT and can be used for fusion imaging registration in a system originally designed for 3D-3D only. This 2D-3D registration protocol is accurate and leads to a significant reduction in radiation exposure.


Subject(s)
Aorta, Abdominal/diagnostic imaging , Aortic Aneurysm, Abdominal/diagnostic imaging , Aortography , Computed Tomography Angiography , Imaging, Three-Dimensional , Aged , Anatomic Landmarks , Aorta, Abdominal/surgery , Aortic Aneurysm, Abdominal/surgery , Aortography/adverse effects , Blood Vessel Prosthesis Implantation , Computed Tomography Angiography/adverse effects , Endovascular Procedures , Female , Humans , Imaging, Three-Dimensional/adverse effects , Male , Predictive Value of Tests , Radiation Dosage , Radiation Exposure/adverse effects , Radiation Exposure/prevention & control , Reproducibility of Results , Risk Factors
9.
Circulation ; 138(16): 1693-1705, 2018 10 16.
Article in English | MEDLINE | ID: mdl-29739755

ABSTRACT

BACKGROUND: In addition to enhanced proinflammatory signaling, impaired resolution of vascular inflammation plays a key role in atherosclerosis. Proresolving lipid mediators formed through the 12/15 lipoxygenase pathways exert protective effects against murine atherosclerosis. n-3 Polyunsaturated fatty acids, including eicosapentaenoic acid (EPA), serve as the substrate for the formation of lipid mediators, which transduce potent anti-inflammatory and proresolving actions through their cognate G-protein-coupled receptors. The aim of this study was to identify signaling pathways associated with EPA supplementation and lipid mediator formation that mediate atherosclerotic disease progression. METHODS: Lipidomic plasma analysis were performed after EPA supplementation in Apoe-/- mice. Erv1/Chemr23-/- xApoe-/- mice were generated for the evaluation of atherosclerosis, phagocytosis, and oxidized low-density lipoprotein uptake. Histological and mRNA analyses were done on human atherosclerotic lesions. RESULTS: Here, we show that EPA supplementation significantly attenuated atherosclerotic lesion growth induced by Western diet in Apoe-/- mice and was associated with local cardiovascular n-3 enrichment and altered lipoprotein metabolism. Our systematic plasma lipidomic analysis identified the resolvin E1 precursor 18-monohydroxy EPA as a central molecule formed during EPA supplementation. Targeted deletion of the resolvin E1 receptor Erv1/Chemr23 in 2 independent hyperlipidemic murine models was associated with proatherogenic signaling in macrophages, increased oxidized low-density lipoprotein uptake, reduced phagocytosis, and increased atherosclerotic plaque size and necrotic core formation. We also demonstrate that in macrophages the resolvin E1-mediated effects in oxidized low-density lipoprotein uptake and phagocytosis were dependent on Erv1/Chemr23. When analyzing human atherosclerotic specimens, we identified ERV1/ChemR23 expression in a population of macrophages located in the proximity of the necrotic core and demonstrated augmented ERV1/ChemR23 mRNA levels in plaques derived from statin users. CONCLUSIONS: This study identifies 18-monohydroxy EPA as a major plasma marker after EPA supplementation and demonstrates that the ERV1/ChemR23 receptor for its downstream mediator resolvin E1 transduces protective effects in atherosclerosis. ERV1/ChemR23 signaling may represent a previously unrecognized therapeutic pathway to reduce atherosclerotic cardiovascular disease.


Subject(s)
Aorta/drug effects , Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , Eicosapentaenoic Acid/pharmacology , Lipoproteins, LDL/metabolism , Macrophages/drug effects , Phagocytosis/drug effects , Plaque, Atherosclerotic , Receptors, G-Protein-Coupled/agonists , Animals , Aorta/metabolism , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/metabolism , Aortic Diseases/pathology , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cytochrome Reductases/genetics , Cytochrome Reductases/metabolism , Diet, Western , Disease Models, Animal , Eicosapentaenoic Acid/analogs & derivatives , Eicosapentaenoic Acid/blood , Eicosapentaenoic Acid/metabolism , Genetic Predisposition to Disease , Humans , Macrophages/metabolism , Macrophages/pathology , Male , Mice, Inbred C57BL , Mice, Knockout, ApoE , Necrosis , Oxidoreductases Acting on Sulfur Group Donors , Phenotype , Receptors, Chemokine , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, LDL/genetics , Receptors, LDL/metabolism , Signal Transduction/drug effects
10.
JACC Basic Transl Sci ; 3(6): 719-727, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30623131

ABSTRACT

An abdominal aortic aneurysm (AAA) is a progressive aortic dilation that may lead to rupture, which is usually lethal. This study identifies the state of failure in the resolution of inflammation by means of decreased expression of the pro-resolving receptor A lipoxin/formyl peptide receptor 2 (ALX/FPR2) in the adventitia of human AAA lesions. Mimicking this condition by genetic deletion of the murine ALX/FPR2 ortholog in hyperlipidemic mice exacerbated the aortic dilation induced by angiotensin II infusion, associated with decreased vascular collagen and increased inflammation. The authors also identified key roles of lipoxin formation through 12/15-lipoxygenase and neutrophil p38 mitogen-activated protein kinase. In conclusion, this study established pro-resolving signaling by means of the ALX/FPR2 receptor in aneurysms and vascular inflammation.

11.
Medicine (Baltimore) ; 96(33): e7862, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28816989

ABSTRACT

The aim of this study was to analyze the impact of disease duration on carotid to femoral pulse wave velocity (cfPWV) in rheumatoid arthritis (RA) patients without either known traditional cardiovascular risk factors or previous comorbidities.Patients with RA diagnosis attending the rheumatology outpatient clinic of Hospital Civil Juan I. Menchaca, Guadalajara, Mexico, were analyzed. A total of 106 RA patients without known traditional cardiovascular risk factors were selected. All subjects were evaluated for RA disease duration, RA disease activity score on 28 joints (DAS28), serum lipids, rheumatoid factor and anti-cyclic citrullinated peptide (anti-CCP) antibodies. Arterial stiffness was measured as cfPWV by noninvasive tonometry. A multivariate regression model was used to analyze the contribution of RA disease duration and age on cfPWV. cfPWV was positively correlated with age (r = 0.450, P < .001), RA disease duration (r = 0.340, P < .001), total cholesterol (r = 0.312, P = .002), and low density lipoprotein (LDL-c) cholesterol (r = 0.268, P = .012). Patients with a RA disease duration ≥10 years exhibited significantly increased cfPWV compared with patients with disease duration <2 years (8.4 ±â€Š1.8 vs 7.0 ±â€Š0.8) and ≥2 to <10 years (8.4 ±â€Š1.8 vs 7.8 ±â€Š1.3), respectively. Age, RA disease duration, and triglycerides were predictors of cfPWV in multivariate analyses. According to the ß-coefficients, each year of disease duration (ß = 0.072) had a greater impact on cfPWV than age (ß = 0.054).Each year of life with RA contributes to a higher rate of vascular aging or stiffening than a year of life without RA. The cumulative damage provided by RA was most pronounced in patients with disease duration ≥10 years.


Subject(s)
Arthritis, Rheumatoid/physiopathology , Carotid Arteries/pathology , Femoral Artery/pathology , Vascular Stiffness/physiology , Adult , Age Factors , Age of Onset , Cross-Sectional Studies , Humans , Lipids/blood , Manometry , Middle Aged , Peptides, Cyclic/immunology , Pulse Wave Analysis , Rheumatoid Factor/blood , Risk Factors , Severity of Illness Index
12.
Eur J Immunol ; 47(11): 1918-1924, 2017 11.
Article in English | MEDLINE | ID: mdl-28742217

ABSTRACT

Sortilin-1, a receptor of the VPS10p family, has been associated with cardiovascular disease in genome-wide association studies. It is implicated in lipoprotein metabolism, secretion of proprotein convertase subtilisin/kexin type 9 (PCSK9) and secretion of inflammatory cytokines. However, its own regulation remains unclear. Chronic inflammation is a hallmark of atherosclerosis and the absence of regulatory T (Treg) cells is associated with reduced protein expression of sortilin-1 in the liver. Therefore, we postulated that mediator(s) of inflammation known to be downregulated by Treg cells may modulate sortilin-1 expression. In this study, we identify interferon-gamma (IFN-γ) as the key inflammatory mediator controlling sortilin-1 levels. In vitro cultures of murine hepatocytes cell line and in silico experiments showed that the transcription factor Signal transducer and activator of transcription 1 was activated and bound to the Sort-1 gene upon IFN-γ treatment. This reduced the expression of sortilin-1, while disrupting the IFN-γ signaling pathway prevented the effect. These data unravel an intricate mechanism by which inflammation modulates receptors involved in lipoprotein turnover.


Subject(s)
Adaptor Proteins, Vesicular Transport/biosynthesis , Hepatocytes/metabolism , Interferon-gamma/metabolism , Janus Kinases/metabolism , STAT Transcription Factors/metabolism , Adaptor Proteins, Vesicular Transport/immunology , Animals , Gene Expression Regulation/immunology , Hepatocytes/immunology , Interferon-gamma/immunology , Janus Kinases/immunology , Mice , Mice, Inbred C57BL , STAT Transcription Factors/immunology , Signal Transduction/immunology
13.
Br J Pharmacol ; 174(22): 4043-4054, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28071789

ABSTRACT

BACKGROUND AND PURPOSE: Atherosclerosis is characterized by a chronic non-resolving inflammation in the arterial wall. Aspirin-triggered lipoxin A4 (ATL) is a potent anti-inflammatory mediator, involved in the resolution of inflammation. However, the therapeutic potential of immune targeting by means of ATL in atherosclerosis has not previously been explored. The aim of the present study was to determine the effects of ATL and its receptor Fpr2 on atherosclerosis development and progression in apolipoprotein E deficient (ApoE-/- ) mice. EXPERIMENTAL APPROACH: ApoE-/-  × Fpr2+/+ and ApoE-/-  × Fpr2-/- mice were generated. Four-week-old mice fed a high-fat diet for 4 weeks and 16-week-old mice fed chow diet received osmotic pumps containing either vehicle or ATL for 4 weeks. Atherosclerotic lesion size and cellular composition were measured in the aortic root and thoracic aorta. Lipid levels and leukocyte counts were measured in blood and mRNA was isolated from abdominal aorta and spleen. KEY RESULTS: ATL blocked atherosclerosis progression in the aortic root and thoracic aorta of ApoE-/- mice. In addition, ATL reduced macrophage infiltration and apoptotic cells in atherosclerotic lesions. The mRNA levels of several cytokines and chemokines in the spleen and aorta were reduced by ATL, whereas circulating leukocyte levels were unchanged. The ATL-induced athero-protection was absent in ApoE-/- mice lacking the Fpr2 receptor. CONCLUSION AND IMPLICATIONS: ATL blocked atherosclerosis progression by means of an Fpr2-mediated reduced local and systemic inflammation. These results suggest this anti-inflammatory and pro-resolving agent has therapeutic potential for the treatment of atherosclerosis. LINKED ARTICLES: This article is part of a themed section on Targeting Inflammation to Reduce Cardiovascular Disease Risk. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.22/issuetoc and http://onlinelibrary.wiley.com/doi/10.1111/bcp.v82.4/issuetoc.


Subject(s)
Apolipoproteins E/genetics , Aspirin/therapeutic use , Atherosclerosis/drug therapy , Lipoxins/therapeutic use , Receptors, Formyl Peptide/genetics , Animals , Aorta/drug effects , Aorta/metabolism , Aorta/pathology , Aspirin/pharmacology , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cytokines/genetics , Female , Lipoxins/pharmacology , Mice, Knockout
14.
Mediators Inflamm ; 2016: 3085390, 2016.
Article in English | MEDLINE | ID: mdl-27239101

ABSTRACT

BACKGROUND: In obesity there is a subclinical chronic low-grade inflammatory response where insulin resistance (IR) may develop. Chemerin is secreted in white adipose tissue and promotes low-grade inflammatory process, where it expressed CMKLR1 receptor. The role of chemerin and CMKLR1 in inflammatory process secondary to obesity is not defined yet. METHODS: Cross-sectional study with 134 individuals classified as with and without obesity by body mass index (BMI) and IR. Body fat storage measurements and metabolic and inflammatory markers were measured by routine methods. Soluble chemerin and basal levels of insulin by ELISA and relative expression of CMKLR1 were evaluated with qPCR and 2(-ΔΔCT) method. RESULTS: Differences (P < 0.05) were observed between obesity and lean individuals in body fat storage measurements and metabolic-inflammatory markers. Both CMKLR1 expression and chemerin levels were increased in obesity without IR. Soluble chemerin levels correlate with adiposity and metabolic markers (r = 8.8% to 38.5%), P < 0.05. CONCLUSION: The increment of CMKLR1 expression was associated with insulin production. Increased serum levels of chemerin in obesity were observed, favoring a dysmetabolic response. The results observed in this study suggest that both chemerin and CMKLR1 have opposite expression in the context of low-grade inflammatory response manifested in the development of IR.


Subject(s)
Chemokines/blood , Insulin Resistance/physiology , Intercellular Signaling Peptides and Proteins/blood , Obesity/blood , Obesity/immunology , Receptors, Chemokine/blood , Adiposity/physiology , Adult , Cross-Sectional Studies , Dyslipidemias/blood , Female , Humans , Male , Middle Aged , Young Adult
15.
J Diabetes Res ; 2016: 5675739, 2016.
Article in English | MEDLINE | ID: mdl-26839895

ABSTRACT

Genetic susceptibility has been described in insulin resistance (IR). Chemokine (C-C motif) ligand-2 (CCL2) is overexpressed in white adipose tissue and is the ligand of C-C motif receptor-2 (CCR2). The CCL2 G-2518A polymorphism is known to regulate gene expression, whereas the physiological effects of the CCR2Val64Ile polymorphism are unknown. The aim of the study is to investigate the relationship between these polymorphisms with soluble CCL2 levels (sCCL2), metabolic markers, and adiposity. In a cross-sectional study we included 380 Mexican-Mestizo individuals, classified with IR according to Stern criteria. Polymorphism was identified using PCR-RFLP/sequence-specific primers. Anthropometrics and metabolic markers were measured by routine methods and adipokines and sCCL2 by ELISA. The CCL2 polymorphism was associated with IR (polymorphic A+ phenotype frequencies were 70.9%, 82.6%, in individuals with and without IR, resp.). Phenotype carriers CCL2 (A+) displayed lower body mass and fat indexes, insulin and HOMA-IR, and higher adiponectin levels. Individuals with IR presented higher sCCL2 compared to individuals without IR and was associated with CCR2 (Ile+) phenotype. The double-polymorphic phenotype carriers (A+/Ile+) exhibited higher sCCL2 than double-wild-type phenotype carriers (A-/Ile-). The present findings suggest that sCCL2 production possibly will be associated with the adiposity and polymorphic phenotypes of CCL2 and CCR2, in Mexican-Mestizos with IR.


Subject(s)
Adiposity , Chemokine CCL2/blood , Insulin Resistance/ethnology , Polymorphism, Genetic , Receptors, CCR2/blood , Adult , Aged , Anthropometry , Chemokine CCL2/genetics , Cross-Sectional Studies , Enzyme-Linked Immunosorbent Assay , Female , Genetic Association Studies , Humans , Insulin Resistance/genetics , Male , Mexico , Middle Aged , Obesity/ethnology , Obesity/genetics , Phenotype , Polymorphism, Restriction Fragment Length , Polymorphism, Single Nucleotide , Receptors, CCR2/genetics , Young Adult
16.
Prostaglandins Other Lipid Mediat ; 121(Pt A): 138-43, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26248046

ABSTRACT

INTRODUCTION: Lipoxygenase pathway yields both pro-inflammatory leukotrienes and pro-resolving lipoxins. The aim of the present study was to determine the effects of T-lymphocytes and pro-inflammatory stimuli on the expression levels of the lipoxin FPR2/ALX receptor, and the leukotriene BLT1 receptor in monocytes and macrophages, and to characterize LXA4-induced effects on pro-inflammatory mediators. METHODS: Human macrophages were co-cultured with activated CD4(+) cells. THP-1 cells were stimulated with different cytokines, LXA4 and supernatant from activated CD4(+) cells. mRNA was extracted for qPCR experiments and protein was analyzed by flow cytometry. RESULTS: Co-culture of macrophages with activated CD4(+) cells or their supernatants up-regulated macrophage FPR2/ALX expression but did not alter BLT1 receptor expression. Monocyte stimulation with IFN-γ up-regulated FPR2/ALX mRNA and protein levels, whereas BLT1 mRNA was down-regulated. Finally, LXA4 decreased mRNA levels of MMP-9, CXCL16, IL-1ß, and IL-8 in THP-1 cells. CONCLUSION: The present study shows that pro-inflammatory stimuli lead to FPR2/ALX expression. LXA4 induces an anti-inflammatory response, which could participate in the resolution of inflammation.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Down-Regulation , Monocytes/metabolism , Receptors, Formyl Peptide/genetics , Receptors, Leukotriene B4/genetics , Receptors, Lipoxin/genetics , Up-Regulation , Down-Regulation/drug effects , Humans , Interferon-gamma/pharmacology , Intracellular Space/drug effects , Intracellular Space/metabolism , Monocytes/cytology , Monocytes/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Up-Regulation/drug effects
18.
Cardiovasc Res ; 105(1): 65-74, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25341894

ABSTRACT

AIMS: The formyl peptide receptor (FPR) subtype FPR2/ALX transduces pro-inflammatory responses and participates in the resolution of inflammation depending on activation. The aim of the present study was to unravel the role of FPR2/ALX signalling in atherosclerosis. METHODS AND RESULTS: Expression of FPR2/ALX was analysed in 127 human carotid atherosclerotic lesions and revealed that this receptor was expressed on macrophages, smooth muscle cells (SMCs), and endothelial cells. Furthermore, FPR2/ALX mRNA levels were significantly up-regulated in atherosclerotic lesions compared with healthy vessels. In multiple regression, age, creatinine, and clinical signs of increased cerebral ischaemia were independent predictors of FPR2/ALX expression. To provide mechanistic insights into these observations, we generated Ldlr(-/-)xFpr2(-/-) mice, which exhibited delayed atherosclerosis development and less macrophage infiltration compared with Ldlr(-/-)xFpr2(+/+) mice. These findings were reproduced by transplantation of Fpr2(-/-) bone marrow into Ldlr(-/-) mice and further extended by in vitro experiments, demonstrating a lower inflammatory state in Fpr2(-/-) macrophages. FPR2/ALX expression correlated with chemo- and cytokines in human atherosclerotic lesions and leucocytes. Finally, atherosclerotic lesions in Ldlr(-/-)xFpr2(-/-) mice exhibited decreased collagen content, and Fpr2(-/-) SMCs exhibited a profile of increased collagenase and decreased collagen production pathways. CONCLUSION: FPR2/ALX is proatherogenic due to effects on bone marrow-derived cells, but promoted a more stable plaque phenotype through effects on SMCs. Taken together, these results suggest a dual role of FPR2/ALX signalling in atherosclerosis by way of promoting disease progression and but increasing plaque stability.


Subject(s)
Atherosclerosis/etiology , Atherosclerosis/metabolism , Receptors, Formyl Peptide/metabolism , Receptors, Lipoxin/metabolism , Aged , Aged, 80 and over , Animals , Atherosclerosis/genetics , Bone Marrow Transplantation , Carotid Artery Diseases/etiology , Carotid Artery Diseases/genetics , Carotid Artery Diseases/metabolism , Carotid Stenosis/pathology , Collagen/metabolism , Female , Humans , Macrophages/metabolism , Male , Mice , Mice, Knockout , Middle Aged , Myocytes, Smooth Muscle/metabolism , Plaque, Atherosclerotic/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Formyl Peptide/deficiency , Receptors, Formyl Peptide/genetics , Receptors, LDL/deficiency , Receptors, LDL/genetics , Receptors, LDL/metabolism , Receptors, Lipoxin/genetics , Signal Transduction , Up-Regulation
19.
Article in English | MEDLINE | ID: mdl-25139400

ABSTRACT

The n-3 polyunsaturated fatty acid, docosahexaenoic acid (DHA), displays anti-inflammatory properties that may prevent atherosclerosis progression. Exposure of apolipoprotein-E deficient (ApoE(-/-)) mice to chronic intermittent hypoxia (CIH) accelerates atherosclerosis progression. Our aim was to assess DHA-supplementation influence on fatty acid incorporation in different tissues/organs and on atherosclerosis progression in ApoE(-/-) mice exposed to CIH. ApoE(-/-) mice were exposed to CIH or normoxia (N) and randomized to four groups (N control, CIH control, N+DHA, and CIH+DHA). DHA-supplementation enhanced DHA and reduced arachidonic acid (AA) contents in tissues/organs. CIH control mice exhibited increased atherosclerosis lesion sizes compared to N control mice. DHA prevented CIH induced atherosclerosis but did not improve atherosclerosis burden in N mice. Aortic matrix metalloproteinase-2 (MMP-2) expression was decreased in CIH+DHA mice (p=0.007). DHA-supplementation prevented CIH-induced atherosclerosis acceleration. This was associated with a decrease of AA incorporation and of aortic MMP-2 gene expression.


Subject(s)
Apolipoproteins E/deficiency , Atherosclerosis/drug therapy , Atherosclerosis/metabolism , Docosahexaenoic Acids/therapeutic use , Fatty Acids/metabolism , Hypoxia/physiopathology , Animals , Apolipoproteins E/genetics , Male , Mice , Mice, Knockout
20.
Biochem Biophys Res Commun ; 441(2): 393-8, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24161392

ABSTRACT

The prothrombotic mediator thromboxane A2 is derived from arachidonic acid metabolism through the cyclooxygenase and thromboxane synthase pathways, and transduces its effect through the thromboxane prostanoid (TP) receptor. The aim of this study was to determine the effect of the TP receptor antagonist and thromboxane synthase inhibitor EV-077 on inflammatory markers in human umbilical vein endothelial cells and on human coronary artery smooth muscle cell proliferation. To this end, mRNA levels of different proinflammatory mediators were studied by real time quantitative PCR, supernatants were analyzed by enzyme immune assay, and cell proliferation was assessed using WST-1. EV-077 significantly decreased mRNA levels of ICAM-1 and PTX3 after TNFα incubation, whereas concentrations of 6-keto PGF1α in supernatants of endothelial cells incubated with TNFα were significantly increased after EV-077 treatment. Although U46619 did not alter coronary artery smooth muscle cell proliferation, this thromboxane mimetic enhanced the proliferation induced by serum, insulin and growth factors, which was significantly inhibited by EV-077. In conclusion, EV-077 inhibited TNFα-induced endothelial inflammation and reduced the enhancement of smooth muscle cell proliferation induced by a thromboxane mimetic, supporting that the thromboxane pathway may be associated with early atherosclerosis in terms of endothelial dysfunction and vascular hypertrophy.


Subject(s)
Endothelium, Vascular/drug effects , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Receptors, Thromboxane/antagonists & inhibitors , Thromboxane-A Synthase/antagonists & inhibitors , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , C-Reactive Protein/antagonists & inhibitors , C-Reactive Protein/biosynthesis , Cell Proliferation/drug effects , Cells, Cultured , Coronary Vessels/cytology , Coronary Vessels/drug effects , Coronary Vessels/metabolism , Endothelium, Vascular/metabolism , Gene Expression/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Intercellular Adhesion Molecule-1/biosynthesis , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Prostaglandins F/metabolism , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/biosynthesis , Serum Amyloid P-Component/antagonists & inhibitors , Serum Amyloid P-Component/biosynthesis , Thromboxane A2/physiology , Tumor Necrosis Factor-alpha/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL