Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Curr Opin Clin Nutr Metab Care ; 27(5): 434-442, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-39083430

RESUMEN

PURPOSE OF REVIEW: Contemporary guidelines for the prevention of cardio-metabolic diseases focus on the control of dietary fat intake, because of their adverse metabolic effects. Moreover, fats alter innate immune defenses, by eliciting pro-inflammatory epigenetic mechanisms on the long-living hematopoietic cell progenitors which, in the bone marrow, mainly give rise to short-living neutrophils. Nevertheless, the heterogenicity of fats and the complexity of the biology of neutrophils pose challenges in the understanding on how this class of nutrients could contribute to the development of cardio-metabolic diseases via specific molecular mechanisms activating the inflammatory response. RECENT FINDINGS: The knowledge on the biology of neutrophils is expanding and there are now different cellular networks orchestrating site-specific reprogramming of these cells to optimize the responses against pathogens. The innate immune competence of neutrophil is altered in response to high fat diet and contributes to the development of metabolic alterations, although the precise mechanisms are still poorly understood. SUMMARY: Defining the different molecular mechanisms involved in the fat-neutrophil crosstalk will help to reconcile the sparse data about the interaction of dietary fats with neutrophils and to tailor strategies to target neutrophils in the context of cardio-metabolic diseases.


Asunto(s)
Grasas de la Dieta , Neutrófilos , Humanos , Neutrófilos/metabolismo , Neutrófilos/inmunología , Inmunidad Innata , Inflamación , Dieta Alta en Grasa/efectos adversos , Animales , Enfermedades Cardiovasculares/prevención & control
2.
J Cell Immunol ; 5(5): 168-173, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38327649

RESUMEN

Metabolic alterations induced by unhealthy lifestyles, including obesity and insulin resistance are often associated with increased innate immune response and chronic inflammation. Cholesterol has been identified as a key metabolite driving the activation of the inflammasome and the "epigenetic memory" in long-term living hematopoietic stem cells. In addition to these mechanisms, the physiological aging of short-living neutrophils is a relevant modifier of their immune competency, as while they egress from medullary niches as "fresh", fully competent, cells, they turn into "aged", disarmed cells, when they extravasate into peripheral tissues to fight against pathogens or they reach the spleen for disposal. We recently observed that cardio-metabolic alterations induced by a lipid enriched unhealthy diet critically accelerate this process. Indeed, the chronic feeding with a high fat diet (HFD) results in the increase of aged neutrophils in the circulation and their accumulation in liver. This profile is associated with a deteriorated insulin response and obesity. The HFD primes aged, but not fresh neutrophils, to infiltrate in the liver and promotes inflammation coupled to altered cell immune architecture in visceral adipose tissue. Preventing the aging of neutrophils via selective ablation of CXCR2, reduces the development of obesity and improves the sensitivity to insulin. In humans, plasma levels of CXCL1, one of the cytokines binding CXCR2 and promoting neutrophil aging, are directly associated with abdominal adiposity and fatty liver independently of other risk factors. Together these findings point to a direct role of aged neutrophils in the development of metabolic disorders.

4.
Eur J Prev Cardiol ; 29(5): 721-729, 2022 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-33624064

RESUMEN

AIMS: Leucocyte telomere length (LTL) shortening is a marker of cellular senescence and associates with increased risk of cardiovascular disease (CVD). A number of cardiovascular risk factors affect LTL, but the correlation between elevated LDL cholesterol (LDL-C) and shorter LTL is debated: in small cohorts including subjects with a clinical diagnosis of familial hypercholesterolaemia (FH). We assessed the relationship between LDL-C and LTL in subjects with genetic familial hypercholesterolaemia (HeFH) compared to those with clinically diagnosed, but not genetically confirmed FH (CD-FH), and normocholesterolaemic subjects. METHODS AND RESULTS: LTL was measured in mononuclear cells-derived genomic DNA from 206 hypercholesterolaemic subjects (135 HeFH and 71 CD-FH) and 272 controls. HeFH presented shorter LTL vs. controls (1.27 ± 0.07 vs. 1.59 ± 0.04, P = 0.045). In particular, we found shorter LTL in young HeFH as compared to young controls (<35 y) (1.34 ± 0.08 vs. 1.64 ± 0.08, P = 0.019); moreover, LTL was shorter in statin-naïve HeFH subjects as compared to controls (1.23 ± 0.08 vs. 1.58 ± 0.04, P = 0.001). HeFH subjects presented shorter LTL compared to LDL-C matched CD-FH (1.33 ± 0.05 vs. 1.55 ± 0.08, P = 0.029). Shorter LTL was confirmed in leucocytes of LDLR-KO vs. wild-type mice and associated with lower abundance of long-term haematopoietic stem and progenitor cells (LT-HSPCs) in the bone marrow. Accordingly, HeFH subjects presented lower circulating haematopoietic precursors (CD34 + CD45dim cells) vs. CD-FH and controls. CONCLUSIONS: We found (i) shorter LTL in genetically determined hypercholesterolaemia, (ii) lower circulating haematopoietic precursors in HeFH subjects, and reduced bone marrow resident LT-HSPCs in LDLR-KO mice. We support early cellular senescence and haematopoietic alterations in subjects with FH.


Asunto(s)
Hipercolesterolemia , Hiperlipoproteinemia Tipo II , Animales , LDL-Colesterol , Humanos , Hipercolesterolemia/genética , Hiperlipoproteinemia Tipo II/diagnóstico , Hiperlipoproteinemia Tipo II/genética , Leucocitos , Ratones , Telómero/genética
5.
Immunometabolism ; 3(2): e210014, 2021 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-33859831

RESUMEN

Hematopoiesis is the process that leads to multiple leukocyte lineage generation within the bone marrow. This process is maintained throughout life thanks to a nonstochastic division of hematopoietic stem cells (HSCs), where during each division, one daughter cell retains pluripotency while the other differentiates into a restricted multipotent progenitor (MPP) that converts into mature, committed circulating cell. This process is tightly regulated at the level of cellular metabolism and the shift from anaerobic glycolysis, typical of quiescent HSC, to oxidative metabolism fosters HSCs proliferation and commitment. Systemic and local factors influencing metabolism alter HSCs balance under pathological conditions, with chronic metabolic and inflammatory diseases driving HSCs commitment toward activated blood immune cell subsets. This is the case of atherosclerosis, where impaired systemic lipid metabolism affects HSCs epigenetics that reflects into increased differentiation toward activated circulating subsets. Aim of this review is to discuss the impact of lipids and lipoproteins on HSCs pathophysiology, with a focus on the molecular mechanisms influencing cellular metabolism. A better understanding of these aspects will shed light on innovative strategies to target atherosclerosis-associated inflammation.

6.
Cancer Res ; 80(18): 3906-3919, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32699136

RESUMEN

Extracellular ATP (eATP) is a signaling molecule that variably affects all cells of the immune system either directly or after hydrolysis to adenosine. Although eATP is virtually absent in the interstitium of normal tissues, it can be present in the hundreds of micromolar range in tumors, a concentration compatible with activation of the ATP-gated ionotropic P2X7 receptor. Here, we show that P2X7 activity in tumor-infiltrating lymphocytes (TIL) induces cellular senescence and limits tumor suppression. P2X7 stimulation affected cell cycling of effector T cells and resulted in generation of mitochondrial reactive oxygen species and p38 MAPK-dependent upregulation of cyclin-dependent kinase inhibitor 1A (Cdkn1a, encoding for p21Waf1/Cip1). Lack of P2X7 promoted a transcriptional signature that correlated with enhanced cytotoxic T-cell response in human solid tumors. In mice, transfer of tumor-specific T cells with deletion of P2rx7 significantly reduced tumor growth and extended survival. Collectively, these findings uncover a purinergic checkpoint that can be targeted to improve the efficacy of cancer immunotherapy strategies. SIGNIFICANCE: These findings suggest that the purinergic checkpoint P2X7 may be targeted to enhance T-cell-mediated cancer immunotherapy and improve T effector cell accumulation in the tumor microenvironment. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/18/3906/F1.large.jpg.


Asunto(s)
Inhibición de Migración Celular , Senescencia Celular/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Melanoma Experimental/inmunología , Receptores Purinérgicos P2X7/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Ciclo Celular , Línea Celular Tumoral , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Citometría de Flujo/métodos , Perfilación de la Expresión Génica , Humanos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inmunoterapia Adoptiva/métodos , Melanoma Experimental/patología , Melanoma Experimental/terapia , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Trasplante de Neoplasias , Antagonistas del Receptor Purinérgico P2X , Especies Reactivas de Oxígeno/metabolismo , Receptores Purinérgicos P2X7/deficiencia , Linfocitos T Citotóxicos/inmunología , Transcripción Genética , Microambiente Tumoral/inmunología , Regulación hacia Arriba
7.
Atheroscler Suppl ; 42: e41-e48, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33589223

RESUMEN

BACKGROUND AND AIM: The metabolic syndrome (MetS) has become one of the most important clinical issues in the cardiovascular field for this decade because of the marked increase in cardiovascular (CV) risk associated with a clustering of risk factors. The aim of the current study was to evaluate the relationship between MetS and its components and cardiovascular disease (CVD). METHODS: This population-based cross-sectional study was based on data from two studies carried out in Russia (ESSE-RF) and Italy (PLIC). One sample from each cohort was selected, matching individuals by sex and age. A comparison between samples of MetS components distribution and CV risk, according to SCORE chart, has been conducted. RESULTS: A total of 609 individuals (mean [SD] age 55 [8] years, about 39% males) for each cohort were selected. Almost half of PLIC cohort participants belonged to the moderate CV risk group (47% vs 27%), while in ESSE-RF cohort a relatively higher prevalence of individuals classified in the high and very high risk group was observed (19% vs 11%, 21% vs 6%, respectively). Overall, 43% of ESSE-RF participants were diagnosed with MetS, compared with the 27% of PLIC members (the difference in prevalence becomes 37% vs 21%, considering a more conservative cut-off for waist circumference). Both cohorts showed a trend towards the increase of MetS components moving from the lowest to the highest CV risk class, with a high prevalence of patients with four or five MetS determinants allocated in the high/very high CV risk group. CONCLUSIONS: Developing effective public health strategies for the prevention, detection and treatment of MetS should be an urgent priority to reduce the burden of CVD, not only in subjects at high/very high CV risk, but also in those characterized by a lower risk, as even rare CV events that come from low risk group bring a tangible burden to healthcare systems.


Asunto(s)
Enfermedades Cardiovasculares/epidemiología , Síndrome Metabólico/epidemiología , Colesterol/sangre , LDL-Colesterol/sangre , Estudios Transversales , Diabetes Mellitus/epidemiología , Femenino , Humanos , Hipertensión/epidemiología , Italia/epidemiología , Masculino , Persona de Mediana Edad , Prevalencia , Medición de Riesgo , Federación de Rusia/epidemiología , Fumar/epidemiología , Circunferencia de la Cintura
8.
Eur Heart J ; 41(26): 2487-2497, 2020 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-31289820

RESUMEN

AIMS: Here, we aimed to determine the therapeutic effect of longevity-associated variant (LAV)-BPIFB4 gene therapy on atherosclerosis. METHODS AND RESULTS: ApoE knockout mice (ApoE-/-) fed a high-fat diet were randomly allocated to receive LAV-BPIFB4, wild-type (WT)-BPIFB4, or empty vector via adeno-associated viral vector injection. The primary endpoints of the study were to assess (i) vascular reactivity and (ii) atherosclerotic disease severity, by Echo-Doppler imaging, histology and ultrastructural analysis. Moreover, we assessed the capacity of the LAV-BPIFB4 protein to shift monocyte-derived macrophages of atherosclerotic mice and patients towards an anti-inflammatory phenotype. LAV-BPIFB4 gene therapy rescued endothelial function of mesenteric and femoral arteries from ApoE-/- mice; this effect was blunted by AMD3100, a CXC chemokine receptor type 4 (CXCR4) inhibitor. LAV-BPIFB4-treated mice showed a CXCR4-mediated shift in the balance between Ly6Chigh/Ly6Clow monocytes and M2/M1 macrophages, along with decreased T cell proliferation and elevated circulating levels of interleukins IL-23 and IL-27. In vitro conditioning with LAV-BPIFB4 protein of macrophages from atherosclerotic patients resulted in a CXCR4-dependent M2 polarization phenotype. Furthermore, LAV-BPIFB4 treatment of arteries explanted from atherosclerotic patients increased the release of atheroprotective IL-33, while inhibiting the release of pro-inflammatory IL-1ß, inducing endothelial nitric oxide synthase phosphorylation and restoring endothelial function. Finally, significantly lower plasma BPIFB4 was detected in patients with pathological carotid stenosis (>25%) and intima media thickness >2 mm. CONCLUSION: Transfer of the LAV of BPIFB4 reduces the atherogenic process and skews macrophages towards an M2-resolving phenotype through modulation of CXCR4, thus opening up novel therapeutic possibilities in cardiovascular disease.


Asunto(s)
Aterosclerosis , Placa Aterosclerótica , Anciano , Animales , Apolipoproteínas E , Aterosclerosis/genética , Grosor Intima-Media Carotídeo , Femenino , Humanos , Inflamación , Péptidos y Proteínas de Señalización Intercelular , Longevidad , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Noqueados para ApoE , Persona de Mediana Edad , Fosfoproteínas , Receptores CXCR4
9.
Cardiovasc Res ; 115(13): 1861-1872, 2019 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-30859179

RESUMEN

AIMS: Low-grade chronic inflammation characterizes obesity and metabolic syndrome. Here, we aim at investigating the impact of the acute-phase protein long pentraxin 3 (PTX3) on the immune-inflammatory response occurring during diet-induced obesity. METHODS AND RESULTS: PTX3 deficiency in mice fed a high-fat diet for 20 weeks protects from weight gain and adipose tissue deposition in visceral and subcutaneous depots. This effect is not related to changes in glucose homeostasis and lipid metabolism but is associated with an improved immune cell phenotype in the adipose tissue of Ptx3 deficient animals, which is characterized by M2-macrophages polarization and increased angiogenesis. These findings are recapitulated in humans where carriers of a PTX3 haplotype (PTX3 h2/h2 haplotype), resulting in lower PTX3 plasma levels, presented with a reduced prevalence of obesity and decreased abdominal adiposity compared with non-carriers. CONCLUSION: Our results support a critical role for PTX3 in the onset of obesity by promoting inflammation and limiting adipose tissue vascularization and delineate PTX3 targeting as a valuable strategy for the treatment of adipose tissue-associated inflammatory response.


Asunto(s)
Proteína C-Reactiva/deficiencia , Dieta Alta en Grasa , Metabolismo Energético , Inmunidad Innata , Mediadores de Inflamación/metabolismo , Inflamación/prevención & control , Grasa Intraabdominal/irrigación sanguínea , Grasa Intraabdominal/metabolismo , Proteínas del Tejido Nervioso/deficiencia , Obesidad/metabolismo , Grasa Subcutánea/irrigación sanguínea , Grasa Subcutánea/metabolismo , Adipogénesis , Adiposidad , Anciano , Animales , Proteína C-Reactiva/genética , Plasticidad de la Célula , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Haplotipos , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/fisiopatología , Mediadores de Inflamación/inmunología , Grasa Intraabdominal/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Neovascularización Fisiológica , Proteínas del Tejido Nervioso/genética , Obesidad/inmunología , Obesidad/fisiopatología , Obesidad Abdominal/epidemiología , Obesidad Abdominal/genética , Obesidad Abdominal/fisiopatología , Fenotipo , Componente Amiloide P Sérico/genética , Transducción de Señal , Grasa Subcutánea/inmunología , Aumento de Peso
10.
Nat Commun ; 9(1): 3083, 2018 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-30082772

RESUMEN

Cholesterol homeostasis has a pivotal function in regulating immune cells. Here we show that apolipoprotein E (apoE) deficiency leads to the accumulation of cholesterol in the cell membrane of dendritic cells (DC), resulting in enhanced MHC-II-dependent antigen presentation and CD4+ T-cell activation. Results from WT and apoE KO bone marrow chimera suggest that apoE from cells of hematopoietic origin has immunomodulatory functions, regardless of the onset of hypercholesterolemia. Humans expressing apoE4 isoform (ε4/3-ε4/4) have increased circulating levels of activated T cells compared to those expressing WT apoE3 (ε3/3) or apoE2 isoform (ε2/3-ε2/2). This increase is caused by enhanced antigen-presentation by apoE4-expressing DCs, and is reversed when these DCs are incubated with serum containing WT apoE3. In summary, our study identifies myeloid-produced apoE as a key physiological modulator of DC antigen presentation function, paving the way for further explorations of apoE as a tool to improve the management of immune diseases.


Asunto(s)
Presentación de Antígeno , Apolipoproteínas E/genética , Células Dendríticas/metabolismo , Activación de Linfocitos , Células Mieloides/metabolismo , Linfocitos T/metabolismo , Animales , Apolipoproteína E4/genética , Células de la Médula Ósea/citología , Diferenciación Celular , Movimiento Celular , Colesterol/metabolismo , Células Dendríticas/citología , Ácidos Grasos/metabolismo , Femenino , Células Madre Hematopoyéticas/citología , Antígenos de Histocompatibilidad Clase II , Humanos , Hipercolesterolemia/metabolismo , Complejo Mayor de Histocompatibilidad , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Oxiesteroles/química , Oxiesteroles/metabolismo , Fosfolípidos/química
11.
Immunity ; 47(5): 875-889.e10, 2017 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-29166588

RESUMEN

Migration of activated regulatory T (Treg) cells to inflamed tissue is crucial for their immune-modulatory function. While metabolic reprogramming during Treg cell differentiation has been extensively studied, the bioenergetics of Treg cell trafficking remains undefined. We have investigated the metabolic demands of migrating Treg cells in vitro and in vivo. We show that glycolysis was instrumental for their migration and was initiated by pro-migratory stimuli via a PI3K-mTORC2-mediated pathway culminating in induction of the enzyme glucokinase (GCK). Subsequently, GCK promoted cytoskeletal rearrangements by associating with actin. Treg cells lacking this pathway were functionally suppressive but failed to migrate to skin allografts and inhibit rejection. Similarly, human carriers of a loss-of-function GCK regulatory protein gene-leading to increased GCK activity-had reduced numbers of circulating Treg cells. These cells displayed enhanced migratory activity but similar suppressive function, while conventional T cells were unaffected. Thus, GCK-dependent glycolysis regulates Treg cell migration.


Asunto(s)
Glucoquinasa/fisiología , Glucólisis , Linfocitos T Reguladores/fisiología , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Antígenos CD28/fisiología , Antígeno CTLA-4/fisiología , Células Cultivadas , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina/fisiología , Diana Mecanicista del Complejo 2 de la Rapamicina/fisiología , Ratones , Ratones Endogámicos , Fosfatidilinositol 3-Quinasas/fisiología , Proteínas Proto-Oncogénicas c-akt/fisiología
12.
Cardiovasc Res ; 113(13): 1627-1638, 2017 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-29016810

RESUMEN

AIMS: Atherosclerosis is an inflammatory disease wherein cholesterol-loaded macrophages play a major role. MicroRNAs and microparticles propagate inflammatory pathways and are involved in cardiovascular disease. We aimed to screen and validate circulating microRNAs correlated with atherosclerosis development in humans, and to dissect the molecular mechanisms associated with atherogenesis using in vitro and in vivo approaches. METHODS AND RESULTS: A panel of 179 secreted microRNAs was screened in plasma samples of patients with and without atherosclerosis, and validated cross-sectionally and prospectively in patients followed for up to 11 years. miR-30c-5p was inversely correlated with total and LDL cholesterol, carotid intimal media thickness (CIMT), presence and future development of plaques. Using a human macrophage line and in vitro gene silencing strategies, we found that miR-30c-5p was downregulated by oxidized LDL (oxLDL) via the scavenger receptor CD36 and inhibition miR processing by Dicer. In turn, miR-30c-5p downregulation was responsible for the effects of oxLDL on macrophage IL-1ß release, caspase-3 expression, and apoptosis. miR-30c-5p loaded into microparticles was uptaken by macrophages and regulated target genes, like caspase-3, at transcriptional level. To establish the relevance of this pathway on endothelial damage as the earliest step of atherogenesis, we show that systemic miR-30c-5p knockdown induced caspase-3 and impaired endothelial healing after carotid injury in C57Bl/6 J mice. CONCLUSIONS: With an unbiased screening of secreted microRNAs, we identify reduction of miR-30c-5p in microparticles as a promoter of early atherosclerosis, by conveying pro-inflammatory pro-apoptotic signals and impairing endothelial healing. Therefore, stimulation of miR-30c-5p is a candidate direct anti-atherosclerotic therapy.


Asunto(s)
Enfermedades de las Arterias Carótidas/metabolismo , MicroARN Circulante/metabolismo , Inflamación/metabolismo , Macrófagos/metabolismo , MicroARNs/metabolismo , Placa Aterosclerótica , Animales , Apoptosis , Antígenos CD36/metabolismo , Enfermedades de las Arterias Carótidas/diagnóstico por imagen , Enfermedades de las Arterias Carótidas/genética , Enfermedades de las Arterias Carótidas/patología , Traumatismos de las Arterias Carótidas/genética , Traumatismos de las Arterias Carótidas/metabolismo , Traumatismos de las Arterias Carótidas/patología , Grosor Intima-Media Carotídeo , Estudios de Casos y Controles , Caspasa 3/metabolismo , LDL-Colesterol/sangre , MicroARN Circulante/sangre , MicroARN Circulante/genética , Estudios Transversales , ARN Helicasas DEAD-box/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Células Endoteliales/patología , Humanos , Inflamación/genética , Interleucina-1beta/metabolismo , Lipoproteínas LDL/metabolismo , Macrófagos/patología , Ratones Endogámicos C57BL , MicroARNs/sangre , MicroARNs/genética , Estudios Prospectivos , Ribonucleasa III/metabolismo , Células THP-1 , Factores de Tiempo
13.
Cell Metab ; 25(3): 593-609, 2017 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-28190771

RESUMEN

Low-grade systemic inflammation associated to obesity leads to cardiovascular complications, caused partly by infiltration of adipose and vascular tissue by effector T cells. The signals leading to T cell differentiation and tissue infiltration during obesity are poorly understood. We tested whether saturated fatty acid-induced metabolic stress affects differentiation and trafficking patterns of CD4+ T cells. Memory CD4+ T cells primed in high-fat diet-fed donors preferentially migrated to non-lymphoid, inflammatory sites, independent of the metabolic status of the hosts. This was due to biased CD4+ T cell differentiation into CD44hi-CCR7lo-CD62Llo-CXCR3+-LFA1+ effector memory-like T cells upon priming in high-fat diet-fed animals. Similar phenotype was observed in obese subjects in a cohort of free-living people. This developmental bias was independent of any crosstalk between CD4+ T cells and dendritic cells and was mediated via direct exposure of CD4+ T cells to palmitate, leading to increased activation of a PI3K p110δ-Akt-dependent pathway upon priming.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Diferenciación Celular , Memoria Inmunológica , Obesidad/inmunología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Estrés Fisiológico , Adiposidad , Animales , Presentación de Antígeno/inmunología , Movimiento Celular , Células Dendríticas/inmunología , Dieta Alta en Grasa , Ácidos Grasos/metabolismo , Femenino , Humanos , Inflamación/patología , Activación de Linfocitos/inmunología , Tejido Linfoide/patología , Masculino , Ratones Endogámicos C57BL , Obesidad/enzimología , Obesidad/patología , Oxidación-Reducción , Fenotipo , Receptores CXCR3/metabolismo
14.
PLoS One ; 11(9): e0162473, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27627804

RESUMEN

BACKGROUND AND AIMS: Epicardial adipose tissue (EAT) has been proposed as a cardiometabolic and hepatic fibrosis risk factor in patients with non alcoholic fatty liver disease (NAFLD). Aim of this study was to evaluate the role of EAT in NAFLD by analyzing 1) the association between EAT, the other metabolic parameters and the severity of steatosis 2) the relationship between cardiovascular (cIMT, cplaques, E/A), liver (presence of NASH and significant fibrosis) damage and metabolic risk factors including EAT 3) the relationship between EAT and genetic factors strongly influencing liver steatosis. METHODS: In a cross-sectional study, we considered 512 consecutive patients with NAFLD (confirmed by biopsy in 100). EAT, severity of steatosis, carotid intima-media thickness (cIMT) and plaques were evaluated by ultrasonography and results analysed by multiple linear and logistic regression models. Variables independently associated with EAT (mm) were female gender (p = 0.003), age (p = 0.001), BMI (p = 0.01), diastolic blood pressure (p = 0.009), steatosis grade 2 (p = 0.01) and 3 (p = 0.04), fatty liver index (p = 0.001) and statin use (p = 0.03). Variables independently associated with carotid IMT were age (p = 0.0001), hypertension (p = 0.009), diabetes (p = 0.04), smoking habits (p = 0.04) and fatty liver index (p = 0.02), with carotid plaques age (p = 0.0001), BMI (p = 0.03), EAT (p = 0.02),) and hypertension (p = 0.02), and with E/A age (p = 0.0001), diabetes (p = 0.005), hypertension (p = 0.04) and fatty liver index (p = 0.004). In the 100 patients with available liver histology non alcoholic steatohepatitis (NASH) was independently associated with EAT (p = 0.04) and diabetes (p = 0.054) while significant fibrosis with EAT (p = 0.02), diabetes (p = 0.01) and waist circumference (p = 0.05). No association between EAT and PNPLA3 and TM6SF2 polymorphisms was found. CONCLUSION: In patients with NAFLD, EAT is associated with the severity of liver and vascular damage besides with the known metabolic risk factors.


Asunto(s)
Tejido Adiposo/patología , Enfermedades Cardiovasculares/patología , Hígado/patología , Enfermedad del Hígado Graso no Alcohólico/patología , Pericardio/patología , Anciano , Estudios Transversales , Femenino , Humanos , Masculino , Persona de Mediana Edad
15.
Circ Cardiovasc Genet ; 8(5): 696-706, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26276885

RESUMEN

BACKGROUND: Carotid intima-media thickness (IMT) is a marker of subclinical atherosclerosis that can predict cardiovascular disease events over traditional risk factors. This study examined the BCAR1-CFDP1-TMEM170A locus on chromosome 16, associated with carotid IMT and coronary artery disease in the IMT and IMT-Progression as Predictors of Vascular Events (IMPROVE) cohort, to identify the functional variant. METHODS AND RESULTS: In analysis of the locus lead single nucleotide polymorphism (SNP; rs4888378, intronic in CFDP1) in Progressione della Lesione Intimale Carotidea (PLIC), the protective AA genotype was associated with slower IMT progression in women (P=0.04) but not in men. Meta-analysis of 5 cohort studies also supported a protective effect of the A allele on common carotid IMT in women only (women: ß=-0.0047, P=1.63 × 10(-4); men: ß=-0.0029, P=0.0678). Two hundred fourteen noncoding variants in strong linkage disequilibrium (r(2) ≥ 0.8) with rs4888378 were identified from 1000 Genome Project. ENCODE regulatory chromatin marks were used to create a shortlist of 6 possible regulatory variants. Electrophoretic mobility shift assays on the shortlist detected allele-specific protein binding to the lead SNP rs4888378; multiplexed competitor electrophoretic mobility shift assays implicated FOXA as the protein. Luciferase reporter assays on rs4888378 showed a significant 35% to 92% (P=0.0057; P=4.0 × 10(-22)) decrease in gene expression with the A allele. Expression quantitative trait loci analysis confirmed previously reported associations of rs4888378 with BCAR1 in vascular tissues. CONCLUSIONS: Molecular studies suggest the lead SNP as a potentially causal SNP at the BCAR1-CFDP1-TMEM170A locus, and expression quantitative trait loci studies implicate BCAR1 as the causal gene. This variant showed stronger effects on common carotid IMT in women, raising questions about the mechanism of the causal SNP on atherosclerosis.


Asunto(s)
Aterosclerosis/genética , Grosor Intima-Media Carotídeo , Enfermedad de la Arteria Coronaria/genética , Proteína Sustrato Asociada a CrK/genética , Anciano , Línea Celular Tumoral , Cromosomas Humanos Par 16 , Estudios de Cohortes , Biología Computacional , ADN/metabolismo , Proteínas de Unión al ADN/metabolismo , Progresión de la Enfermedad , Femenino , Expresión Génica , Genes Reporteros , Humanos , Masculino , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , Estudios Prospectivos , Sitios de Carácter Cuantitativo , Factores de Riesgo , Factores Sexuales
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA