ABSTRACT
The incidence of atherosclerosis is higher among patients with systemic lupus erythematosus (SLE); however, the mechanism by which an atherogenic environment affects autoimmunity remains unclear. We found that reconstitution of atherosclerosis-prone Apoe-/- and Ldlr-/- mice with bone marrow from lupus-prone BXD2 mice resulted in increased autoantibody production and glomerulonephritis. This enhanced disease was associated with an increase in CXCR3+ follicular helper T cells (TFH cells). TFH cells isolated from Apoe-/- mice had higher expression of genes associated with inflammatory responses and SLE and were more potent in inducing production of the immunoglobulin IgG2c. Mechanistically, the atherogenic environment induced the cytokine IL-27 from dendritic cells in a Toll-like receptor 4 (TLR4)-dependent manner, which in turn triggered the differentiation of CXCR3+ TFH cells while inhibiting the differentiation of follicular regulatory T cells. Blockade of IL-27 signals diminished the increased TFH cell responses in atherogenic mice. Thus, atherogenic dyslipidemia augments autoimmune TFH cell responses and subsequent IgG2c production in a TLR4- and IL-27-dependent manner.
Subject(s)
Atherosclerosis/immunology , Dyslipidemias/immunology , Interleukins/immunology , Lupus Erythematosus, Systemic/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Autoimmunity/immunology , Cell Differentiation/immunology , Dendritic Cells/immunology , Mice , Mice, Knockout , Toll-Like Receptor 4/immunologyABSTRACT
Atherosclerosis is a lipid-driven chronic inflammatory disease that is modulated by innate and adaptive immunity including humoral immunity. Importantly, antibody alterations achieved by genetic means or active and passive immunization strategies in preclinical studies can improve or aggravate atherosclerosis. Additionally, a wide range of epidemiological data demonstrate not only an association between the total levels of different antibody isotypes but also levels of antibodies targeting specific antigens with atherosclerotic cardiovascular disease. Here, we discuss the potential role of atherogenic dyslipidemia on the antibody repertoire and review potential antibody-mediated effector mechanisms involved in atherosclerosis development highlighting the major atherosclerosis-associated antigens that trigger antibody responses.
Subject(s)
Atherosclerosis , Humans , Atherosclerosis/immunology , Atherosclerosis/blood , Animals , Antibody Specificity , Immunity, Humoral , Dyslipidemias/immunology , Dyslipidemias/blood , Dyslipidemias/epidemiology , Adaptive Immunity , Antibodies/immunologyABSTRACT
BACKGROUND: Allergic diseases begin early in life and are often chronic, thus creating an inflammatory environment that may precede or exacerbate other pathologies. In this regard, allergy has been associated to metabolic disorders and with a higher risk of cardiovascular disease, but the underlying mechanisms remain incompletely understood. METHODS: We used a murine model of allergy and atherosclerosis, different diets and sensitization methods, and cell-depleting strategies to ascertain the contribution of acute and late phase inflammation to dyslipidemia. Untargeted lipidomic analyses were applied to define the lipid fingerprint of allergic inflammation at different phases of allergic pathology. Expression of genes related to lipid metabolism was assessed in liver and adipose tissue at different times post-allergen challenge. Also, changes in serum triglycerides (TGs) were evaluated in a group of 59 patients ≥14 days after the onset of an allergic reaction. RESULTS: We found that allergic inflammation induces a unique lipid signature that is characterized by increased serum TGs and changes in the expression of genes related to lipid metabolism in liver and adipose tissue. Alterations in blood TGs following an allergic reaction are independent of T-cell-driven late phase inflammation. On the contrary, the IgG-mediated alternative pathway of anaphylaxis is sufficient to induce a TG increase and a unique lipid profile. Lastly, we demonstrated an increase in serum TGs in 59 patients after undergoing an allergic reaction. CONCLUSION: Overall, this study reveals that IgG-mediated allergic inflammation regulates lipid metabolism.
Subject(s)
Disease Models, Animal , Dyslipidemias , Hypersensitivity , Immunoglobulin G , Inflammation , Lipid Metabolism , Signal Transduction , Animals , Dyslipidemias/metabolism , Dyslipidemias/immunology , Dyslipidemias/etiology , Mice , Inflammation/immunology , Inflammation/metabolism , Humans , Immunoglobulin G/blood , Immunoglobulin G/immunology , Hypersensitivity/immunology , Hypersensitivity/metabolism , Hypersensitivity/etiology , Male , Female , Triglycerides/blood , Triglycerides/metabolism , Adipose Tissue/metabolism , Adipose Tissue/immunology , Liver/metabolism , Liver/immunology , Liver/pathologyABSTRACT
Evidence to support the use of steroids in coronavirus disease 2019 (COVID-19) pneumonia is lacking. We aim to determine the impact of steroid use for COVID-19 pneumonia on hospital mortality. We performed a single-center retrospective cohort study in a university hospital in Madrid, Spain, during March of 2020. To determine the role of steroids in in-hospital mortality, patients admitted with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pneumonia and treated with steroids were compared to patients not treated with steroids, and we adjusted with a propensity score for patients on steroid treatment. Survival times were compared using the log rank test. Different steroid regimens were compared and adjusted with a second propensity score. During the study period, 463 out of 848 hospitalized patients with COVID-19 pneumonia fulfilled inclusion criteria. Among them, 396 (46.7%) patients were treated with steroids and 67 patients were not. Global mortality was 15.1%. The median time to steroid treatment from symptom onset was 10 days (interquartile range [IQR], 8 to 13 days). In-hospital mortality was lower in patients treated with steroids than in controls (13.9% [55/396] versus 23.9% [16/67]; hazard ratio [HR], 0.51 [95% confidence interval, 0.27 to 0.96]; P = 0.044). Steroid treatment reduced mortality by 41.8% relative to the mortality with no steroid treatment (relative risk reduction, 0.42 [95% confidence interval, 0.048 to 0.65]). Initial treatment with 1 mg/kg of body weight/day of methylprednisolone versus steroid pulses was not associated with in-hospital mortality (13.5% [42/310] versus 15.1% [13/86]; odds ratio [OR], 0.880 [95% confidence interval, 0.449 to 1.726]; P = 0.710). Our results show that the survival of patients with SARS-CoV-2 pneumonia is higher in patients treated with glucocorticoids than in those not treated. Rates of in-hospital mortality were not different between initial regimens of 1 mg/kg/day of methylprednisolone and glucocorticoid pulses.
Subject(s)
Antiviral Agents/therapeutic use , Azithromycin/therapeutic use , Betacoronavirus/drug effects , Coronavirus Infections/drug therapy , Hydroxychloroquine/therapeutic use , Interferons/therapeutic use , Lopinavir/therapeutic use , Methylprednisolone/therapeutic use , Pneumonia, Viral/drug therapy , Ritonavir/therapeutic use , Aged , Betacoronavirus/immunology , Betacoronavirus/pathogenicity , COVID-19 , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/immunology , Cardiovascular Diseases/mortality , Cardiovascular Diseases/virology , Comorbidity , Coronavirus Infections/immunology , Coronavirus Infections/mortality , Coronavirus Infections/virology , Diabetes Mellitus/drug therapy , Diabetes Mellitus/immunology , Diabetes Mellitus/mortality , Diabetes Mellitus/virology , Drug Administration Schedule , Drug Combinations , Drug Therapy, Combination , Dyslipidemias/drug therapy , Dyslipidemias/immunology , Dyslipidemias/mortality , Dyslipidemias/virology , Female , Hospitals, University , Humans , Intensive Care Units , Length of Stay/statistics & numerical data , Male , Middle Aged , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/mortality , Neoplasms/virology , Pandemics , Pneumonia, Viral/immunology , Pneumonia, Viral/mortality , Pneumonia, Viral/virology , Retrospective Studies , SARS-CoV-2 , Survival AnalysisABSTRACT
To the best of our knowledge, there is no published study on the use of interferon ß-1a (IFN ß-1a) in the treatment of severe COVID-19. In this randomized clinical trial, the efficacy and safety of IFN ß-1a were evaluated in patients with severe COVID-19. Forty-two patients in the interferon group received IFN ß-1a in addition to the national protocol medications (hydroxychloroquine plus lopinavir-ritonavir or atazanavir-ritonavir). Each 44-µg/ml (12 million IU/ml) dose of interferon ß-1a was subcutaneously injected three times weekly for two consecutive weeks. The control group consisted of 39 patients who received only the national protocol medications. The primary outcome of the study was time to reach clinical response. Secondary outcomes were duration of hospital stay, length of intensive care unit stay, 28-day mortality, effect of early or late administration of IFN on mortality, adverse effects, and complications during the hospitalization. Between 29 February and 3 April 2020, 92 patients were recruited, and a total of 42 patients in the IFN group and 39 patients in the control group completed the study. As the primary outcome, time to the clinical response was not significantly different between the IFN and the control groups (9.7 ± 5.8 versus 8.3 ± 4.9 days, respectively, P = 0.95). On day 14, 66.7% versus 43.6% of patients in the IFN group and the control group, respectively, were discharged (odds ratio [OR], 2.5; 95% confidence interval [CI], 1.05 to 6.37). The 28-day overall mortality was significantly lower in the IFN than the control group (19% versus 43.6%, respectively, P = 0.015). Early administration significantly reduced mortality (OR, 13.5; 95% CI, 1.5 to 118). Although IFN did not change the time to reach the clinical response, adding it to the national protocol significantly increased discharge rate on day 14 and decreased 28-day mortality. (This study is in the Iranian Registry of Clinical Trials under identifier IRCT20100228003449N28.).
Subject(s)
Antiviral Agents/therapeutic use , Atazanavir Sulfate/therapeutic use , Betacoronavirus/drug effects , Coronavirus Infections/drug therapy , Interferon beta-1a/therapeutic use , Lopinavir/therapeutic use , Pneumonia, Viral/drug therapy , Ritonavir/therapeutic use , Adult , Aged , Betacoronavirus/immunology , Betacoronavirus/pathogenicity , COVID-19 , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/immunology , Cardiovascular Diseases/mortality , Cardiovascular Diseases/virology , Comorbidity , Coronavirus Infections/immunology , Coronavirus Infections/mortality , Coronavirus Infections/virology , Diabetes Mellitus/drug therapy , Diabetes Mellitus/immunology , Diabetes Mellitus/mortality , Diabetes Mellitus/virology , Drug Administration Schedule , Drug Combinations , Drug Therapy, Combination , Dyslipidemias/drug therapy , Dyslipidemias/immunology , Dyslipidemias/mortality , Dyslipidemias/virology , Female , Humans , Hydroxychloroquine/therapeutic use , Intensive Care Units , Length of Stay , Male , Middle Aged , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/mortality , Neoplasms/virology , Pandemics , Patient Safety , Pneumonia, Viral/immunology , Pneumonia, Viral/mortality , Pneumonia, Viral/virology , SARS-CoV-2 , Survival Analysis , Treatment OutcomeABSTRACT
BACKGROUND: Psoriasis is associated with elevated risk of heart attack and increased accumulation of subclinical noncalcified coronary burden by coronary computed tomography angiography (CCTA). Machine learning algorithms have been shown to effectively analyze well-characterized data sets. OBJECTIVE: In this study, we used machine learning algorithms to determine the top predictors of noncalcified coronary burden by CCTA in psoriasis. METHODS: The analysis included 263 consecutive patients with 63 available variables from the Psoriasis Atherosclerosis Cardiometabolic Initiative. The random forest algorithm was used to determine the top predictors of noncalcified coronary burden by CCTA. We evaluated our results using linear regression models. RESULTS: Using the random forest algorithm, we found that the top 10 predictors of noncalcified coronary burden were body mass index, visceral adiposity, total adiposity, apolipoprotein A1, high-density lipoprotein, erythrocyte sedimentation rate, subcutaneous adiposity, small low-density lipoprotein particle, cholesterol efflux capacity and the absolute granulocyte count. Linear regression of noncalcified coronary burden yielded results consistent with our machine learning output. LIMITATION: We were unable to provide external validation and did not study cardiovascular events. CONCLUSION: Machine learning methods identified the top predictors of noncalcified coronary burden in psoriasis. These factors were related to obesity, dyslipidemia, and inflammation, showing that these are important targets when treating comorbidities in psoriasis.
Subject(s)
Coronary Artery Disease/epidemiology , Machine Learning , Psoriasis/complications , Adult , Comorbidity , Coronary Artery Disease/blood , Coronary Artery Disease/diagnosis , Coronary Artery Disease/immunology , Coronary Vessels/diagnostic imaging , Dyslipidemias/blood , Dyslipidemias/epidemiology , Dyslipidemias/immunology , Female , Humans , Inflammation/blood , Inflammation/epidemiology , Inflammation/immunology , Male , Middle Aged , Obesity/blood , Obesity/epidemiology , Obesity/immunology , Prospective Studies , Psoriasis/blood , Psoriasis/epidemiology , Psoriasis/immunology , Risk Assessment/methods , Risk Factors , Tomography, X-Ray ComputedABSTRACT
Diabetic peripheral neuropathy (DPN) is one of the most widespread and disabling neurological conditions, accounting for half of all neuropathy cases worldwide. Despite its high prevalence, no approved disease modifying therapies exist. There is now a growing body of evidence that DPN secondary to type 1 diabetes mellitus (T1DM) and type 2 diabetes mellitus (T2DM) represents different disease processes, with T2DM DPN best understood within the context of metabolic syndrome rather than hyperglycemia. In this review, we highlight currently understood mechanisms of DPN, along with their corresponding potential therapeutic targets. We frame this discussion within a practical overview of how the field evolved from initial human observations to murine pathomechanistic and therapeutic models into ongoing and human clinical trials, with particular emphasis on T2DM DPN and metabolic syndrome.
Subject(s)
Diabetic Neuropathies , Dyslipidemias , Energy Metabolism , Inflammation , Metabolic Syndrome , Animals , Diabetic Neuropathies/drug therapy , Diabetic Neuropathies/immunology , Diabetic Neuropathies/metabolism , Dyslipidemias/drug therapy , Dyslipidemias/immunology , Dyslipidemias/metabolism , Energy Metabolism/drug effects , Energy Metabolism/immunology , Humans , Inflammation/drug therapy , Inflammation/immunology , Inflammation/metabolism , Metabolic Syndrome/drug therapy , Metabolic Syndrome/immunology , Metabolic Syndrome/metabolism , MiceABSTRACT
Objective- Dyslipidemia is a component of the metabolic syndrome, an established risk factor for atherosclerotic cardiovascular disease, and is also observed in various autoimmune and chronic inflammatory conditions. However, there are limited opportunities to study the impact of acquired dyslipidemia on cardiovascular and immune pathology. Approach and Results- We designed a model system that allows for the conversion to a state of acute hyperlipidemia in adult life, so that the consequences of such a transition could be observed, through conditionally deleting APOE (apolipoprotein E) in the adult mouse. The transition to hypercholesterolemia was accompanied by adaptive immune responses, including the expansion of T lymphocyte helper cell 1, T follicular helper cell, and T regulatory subsets and the formation of germinal centers. Unlike steady-state Apoe-/- mice, abrupt loss of APOE induced rapid production of antibodies recognizing rheumatoid disease autoantigens. Genetic ablation of the germinal center reduced both autoimmunity and atherosclerosis, indicating that the immune response that follows loss of APOE is independent of atherosclerosis but nevertheless promotes plaque development. Conclusions- Our findings suggest that immune activation in response to hyperlipidemia could contribute to a wide range of inflammatory autoimmune diseases, including atherosclerosis.
Subject(s)
Adaptive Immunity , Aorta/immunology , Aortic Diseases/immunology , Apolipoproteins E/immunology , Atherosclerosis/immunology , Autoimmunity , Dyslipidemias/immunology , Inflammation/immunology , Animals , Aorta/metabolism , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/metabolism , Aortic Diseases/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cells, Cultured , Disease Models, Animal , Disease Progression , Dyslipidemias/genetics , Dyslipidemias/metabolism , Dyslipidemias/pathology , Germinal Center/immunology , Germinal Center/metabolism , Immunity, Humoral , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Mice, Inbred C57BL , Mice, Knockout, ApoE , Plaque, Atherosclerotic , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Time FactorsABSTRACT
PURPOSE: Serum immunoglobulins (Igs) play a critical role in modulating the immune response by neutralizing pathogens, although little is known about the effect of Igs in development of atherosclerotic cardiovascular disease (ASCVD). Elevated serum Immunoglobulin A (IgA) concentrations have been identified in previous studies in populations with obesity and hypertriglyceridemia, whereas variable concentrations of Immunoglobulin M (IgM) have been observed in the setting of dyslipidemia. METHODS: In this cross-sectional study, investigators examined the association of serum Ig concentrations with components of metabolic syndrome, including obesity, diabetes, and dyslipidemia. All consecutive adult patients aged 18 years or older discharged from two academic teaching hospitals with serum Immunoglobulin G (IgG) concentration measured during their admission were evaluated, with a total of 1809 individuals included and stratified into two groups: those with and those without dyslipidemia. RESULTS: Mean IgG concentration in individuals with and without dyslipidemia was 997 ± 485 mg/dL and 1144 ± 677 mg/dL, respectively (P < 0.0001). After controlling for confounders in the generalized linear model (GLM), the least square mean IgG concentration in individuals with and without dyslipidemia was 1095 and 1239 mg/dL, respectively (P < 0.0001). The mean IgA and IgM concentrations were not significantly different in individuals with and without dyslipidemia both before and after adjusting covariates. After controlling for confounding variables, all three serum Ig concentrations were not significantly different in individuals with and without diabetes. CONCLUSION: Dyslipidemia was associated with a lower mean serum IgG concentration. No association with any serum Ig was indentified in individuals with diabetes. Exploration of the association between alterations in serum Igs and metabolic syndrome and the role of alterations of Ig concentrations in disease progression represents an important step in identification of appropriate targeted treatment options for reducing cardiovascular risk.
Subject(s)
Diabetes Mellitus/blood , Dyslipidemias/blood , Immunoglobulin G/blood , Metabolic Syndrome/blood , Obesity/blood , Adult , Aged , Aged, 80 and over , Biomarkers/blood , Cross-Sectional Studies , Diabetes Mellitus/diagnosis , Diabetes Mellitus/immunology , Dyslipidemias/diagnosis , Dyslipidemias/immunology , Female , Humans , Immunoglobulin A/blood , Immunoglobulin M/blood , Male , Metabolic Syndrome/diagnosis , Metabolic Syndrome/immunology , Middle Aged , New Jersey , Obesity/diagnosis , Obesity/immunologyABSTRACT
The effect of sulfated polysaccharide from brown alga Fucus evanescens (fucoidan) administered via different routes (peroral and parenteral) on the dynamic of some lipid metabolism parameters and markers of systemic inflammation in mice with experimental dyslipidemia induced by prolonged administration of poloxamer P-407. It was found that fucoidan corrected the main parameters of lipid metabolism, reduced the level of endothelial dysfunction marker endothelin-1 and proinflammatory cytokines TNFα, IFNγ in blood serum in animals with experimental dyslipidemia. These findings open prospects for using fucoidan in the complex treatment of metabolic disorders and atherosclerotic inflammation.
Subject(s)
Anti-Inflammatory Agents/pharmacology , Dyslipidemias/drug therapy , Fucus/chemistry , Gene Expression Regulation/drug effects , Polysaccharides/pharmacology , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/isolation & purification , Cholesterol, HDL/blood , Cholesterol, LDL/blood , Cholesterol, VLDL/blood , Dyslipidemias/chemically induced , Dyslipidemias/genetics , Dyslipidemias/immunology , Endothelin-1/genetics , Endothelin-1/immunology , Inflammation/prevention & control , Interferon-gamma/genetics , Interferon-gamma/immunology , Lipid Metabolism/drug effects , Lipid Metabolism/genetics , Male , Mice , Mice, Inbred BALB C , Poloxamer/administration & dosage , Polysaccharides/chemistry , Polysaccharides/isolation & purification , Triglycerides/blood , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunologyABSTRACT
Non-alcoholic fatty liver disease (NAFLD) is characterized by hepatic steatosis, insulin resistance, dyslipidemia and a systemic pro-inflammatory response, a leading cause of cirrhosis and hepatocellular carcinoma. Here, we showed that PDZ-LIM domain-containing protein 2 (PDLIM2) was an effective suppressor of steatohepatitis. After 16 weeks on a high fat diet (HFD), obesity, insulin resistance, hepatic dyslipidemia and inflammation were markedly aggravated in PDLIM2-knockout (KO) mice. PDLIM2 deletion resulted in lipid accumulation in liver tissue samples of HFD-induced mice, as evidenced by the significant increase of hepatic TG and TC through reducing the expression of lipogenesis- and transcriptional regulators of lipid metabolism-related genes and enhancing fatty acid oxidation-associated molecules. In addition, PDLIM2-ablation promoted the expression of pro-inflammatory cytokines by activating nuclear factor kappa-B (NF-κB) signaling pathway, as supported by the remarkable increase of phosphorylated IKKß, IκBα and NF-κB expressions in liver of HFD-fed mice. Of note, the in vitro study demonstrated that PDLIM2 ablation-enhanced inflammatory response and disorder of lipid metabolism were abrogated by suppressing NF-κB activity. Collectively, the findings could lead to the development of potential therapeutic strategy to prevent NAFLD and associated metabolic disorders by targeting PDLIM2.
Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Diet, High-Fat/adverse effects , Inflammation/immunology , LIM Domain Proteins/immunology , Lipogenesis , Liver/immunology , NF-kappa B/immunology , Adaptor Proteins, Signal Transducing/genetics , Animals , Cells, Cultured , Dyslipidemias/etiology , Dyslipidemias/genetics , Dyslipidemias/immunology , Inflammation/etiology , Inflammation/genetics , Insulin Resistance , LIM Domain Proteins/genetics , Liver/metabolism , Mice , Mice, Knockout , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/immunologyABSTRACT
BACKGROUND: Alternative N-glycosylation has significant structural and functional consequences on immunoglobulin G (IgG) and can affect immune responses, acting as a switch between pro- and anti-inflammatory IgG functionality. Studies have demonstrated that IgG N-glycosylation is associated with ageing, body mass index, type 2 diabetes and hypertension. METHODS: Herein, we have demonstrated patterns of IgG glycosylation that are associated with blood lipids in a cross-sectional study including 598 Han Chinese aged 20-68 years. The IgG glycome composition was analysed by ultra-performance liquid chromatography. RESULTS: Blood lipids were positively correlated with glycan peak GP6, whereas they were negatively correlated with GP18 (P < 0.05/57). The canonical correlation analysis indicated that initial N-glycan structures, including GP4, GP6, GP9-12, GP14, GP17, GP18 and GP23, were significantly correlated with blood lipids, including total cholesterol, total triglycerides (TG) and low-density lipoprotein (r = 0.390, P < 0.001). IgG glycans patterns were able to distinguish patients with dyslipidaemia from the controls, with an area under the curve of 0.692 (95% confidence interval 0.644-0.740). CONCLUSIONS: Our findings indicated that a possible association between blood lipids and the observed loss of galactose and sialic acid, as well as the addition of bisecting GlcNAcs, which might be related to the chronic inflammation accompanying with the development and procession of dyslipidaemia.
Subject(s)
Dyslipidemias/blood , Dyslipidemias/immunology , Glycosylation , Immunoglobulin G/chemistry , Lipids/blood , Adult , Aged , Anthropometry , Body Mass Index , China , Chromatography, High Pressure Liquid , Cross-Sectional Studies , Female , Humans , Male , Middle Aged , Odds Ratio , Polysaccharides/chemistry , Risk Factors , Sensitivity and Specificity , Young AdultABSTRACT
BACKGROUND: The energy homeostasis-associated gene (ENHO), retinoid X receptor alpha gene (RXRA), and liver X receptor alpha gene (LXRA) are involved in adipogenic/lipogenic regulation. We investigated whether single-nucleotide polymorphisms in these genes (ENHO rs2281997, rs72735260; RXRA rs749759, rs10776909, rs10881578; LXRA rs2279238, rs7120118, rs11039155) are associated with dyslipidaemia, related comorbidities and survival of haemodialysis (HD) patients also tested for T-helper (Th) cell interleukin genes (IL). METHODS: The study was carried out in 873 HD patients. Dyslipidaemia was diagnosed by the recommendations of the Kidney Disease Outcomes Quality Initiative (K/DOQI) guidelines (2003); atherogenic dyslipidaemia was referred to if the TG/HDL cholesterol ratio was equal to or higher than 3.8. Genotyping of ENHO SNPs, LXRA SNPs, and IL12A rs568408 was carried out using HRM analysis. RXRA SNPs, IL12B rs3212227, and IL18 rs360719 were genotyped using PCR-RFLP analysis. The circulating adropin concentration was determined in 126 patients by enzyme-linked immunosorbent assay. Survival probability was analysed using the Kaplan-Meier method in 440 patients followed through 7.5 years. RESULTS: Dyslipidaemia by K/DOQI was diagnosed in 459 patients (91% revealed hyper-LDL- cholesterolaemia), atherogenic dyslipidaemia was diagnosed in 454 patients, and 231 patients were free of dyslipidaemia by both criteria. The variant allele (T) of ENHO rs2281997 was associated with the hyper-LDL cholesterolaemic pattern of dyslipidaemia by K/DOQI. The frequency of atherogenic dyslipidaemia was lower in T-allele bearers than in CC-genotype patients. The rs2281997 T allele was associated with lower cardiovascular mortality in HD patients showing atherogenic dyslipidaemia. ENHO, RXRA, and LXRA showed epistatic interactions in dyslipidaemia. Circulating adropin was lower in atherogenic dyslipidaemia than in non-atherogenic conditions. RXRA rs10776909 was associated with myocardial infarction. Bearers of LXRA rs2279238, rs7120118 or rs11039155 minor alleles showed higher mortality. ENHO SNP positions fell within the same DNase 1 hypersensitivity site expressed in the Th1 cell line. Epistatic interactions occurred between rs2281997 and Th1 IL SNPs (rs360719, rs568408). CONCLUSIONS: Atherogenic dyslipidaemia occurs in HD patients in whom ENHO encodes less adropin. ENHO, RXRA, and LXRA SNPs, separately or jointly, are associated with dyslipidaemia, myocardial infarction, and survival in HD patients. Differences in the availability of transcription binding sites may contribute to these associations.
Subject(s)
Blood Proteins/genetics , Dyslipidemias/genetics , Liver X Receptors/genetics , Myocardial Infarction/genetics , Peptides/genetics , Polymorphism, Single Nucleotide , Renal Dialysis , Renal Insufficiency, Chronic/genetics , Retinoid X Receptor alpha/genetics , Adipogenesis/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Alleles , Blood Proteins/immunology , Cholesterol, HDL/blood , Cholesterol, LDL/blood , Comorbidity , Cross-Sectional Studies , Dyslipidemias/immunology , Dyslipidemias/mortality , Dyslipidemias/therapy , Epistasis, Genetic , Female , Genotype , Humans , Intercellular Signaling Peptides and Proteins , Interleukin-12 Subunit p40/genetics , Interleukin-12 Subunit p40/immunology , Interleukin-18/genetics , Interleukin-18/immunology , Kaplan-Meier Estimate , Liver X Receptors/immunology , Male , Middle Aged , Myocardial Infarction/immunology , Myocardial Infarction/mortality , Myocardial Infarction/therapy , Peptides/immunology , Renal Insufficiency, Chronic/immunology , Renal Insufficiency, Chronic/mortality , Renal Insufficiency, Chronic/therapy , Retinoid X Receptor alpha/immunology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/pathology , Triglycerides/bloodABSTRACT
Peroxisome proliferator-activated receptor-delta (PPAR-δ), one of three members of the PPAR group in the nuclear receptor superfamily, is a ligand-activated transcription factor. PPAR-δ regulates important cellular metabolic functions that contribute to maintaining energy balance. PPAR-δ is especially important in regulating fatty acid uptake, transport, and ß-oxidation as well as insulin secretion and sensitivity. These salutary PPAR-δ functions in normal cells are thought to protect against metabolic-syndrome-related diseases, such as obesity, dyslipidemia, insulin resistance/type 2 diabetes, hepatosteatosis, and atherosclerosis. Given the high clinical burden these diseases pose, highly selective synthetic activating ligands of PPAR-δ were developed as potential preventive/therapeutic agents. Some of these compounds showed some efficacy in clinical trials focused on metabolic-syndrome-related conditions. However, the clinical development of PPAR-δ agonists was halted because various lines of evidence demonstrated that cancer cells upregulated PPAR-δ expression/activity as a defense mechanism against nutritional deprivation and energy stresses, improving their survival and promoting cancer progression. This review discusses the complex relationship between PPAR-δ in health and disease and highlights our current knowledge regarding the different roles that PPAR-δ plays in metabolism, inflammation, and cancer.
Subject(s)
Inflammation/metabolism , Neoplasms/metabolism , PPAR delta/metabolism , Animals , Diabetes Mellitus, Type 2/immunology , Diabetes Mellitus, Type 2/metabolism , Dyslipidemias/immunology , Dyslipidemias/metabolism , Fatty Liver/immunology , Fatty Liver/metabolism , Humans , Inflammation/immunology , Insulin Resistance , Metabolic Syndrome/immunology , Metabolic Syndrome/metabolism , Neoplasms/immunology , PPAR delta/immunologyABSTRACT
Predictive models of comorbidity, dyslipidemic disorders and essential arterial hypertension, in Russian adolescents aged 12 to 18 years (mean 15.48±1.53) were formulated with consideration for biochemical (lipid profiles) and genetic parameters (carrier state of gene polymorphic variants of apolipoprotein genes ApoA1 (-75G/A and +83C/T), ApoB (Ins/Del), ApoC3 (S1/S2), and ApoE (ε2/ε3/ε4). Significant prognostic risk factors for the mentioned comorbid pathologies were lipid metabolism parameters HDL-Ch, LDL-Ch, VLDL-Ch and carrier state of the +83T allele of the ApoA1 gene and Del allele of the ApoB gene. The obtained mathematical model is characterized by high predictive accuracy: the percentage of correct classification or the rate of correct assignment of each participant to the proper group was 96.33%.
Subject(s)
Apolipoprotein A-I/genetics , Apolipoprotein B-100/genetics , Dyslipidemias/diagnosis , Essential Hypertension/diagnosis , Genetic Predisposition to Disease , Models, Statistical , Polymorphism, Genetic , Adolescent , Alleles , Apolipoprotein A-I/immunology , Apolipoprotein B-100/immunology , Apolipoprotein C-III/genetics , Apolipoprotein C-III/immunology , Apolipoproteins E/genetics , Apolipoproteins E/immunology , Carrier State , Child , Cholesterol, HDL/blood , Cholesterol, LDL/blood , Cholesterol, VLDL/blood , Discriminant Analysis , Dyslipidemias/blood , Dyslipidemias/genetics , Dyslipidemias/immunology , Essential Hypertension/blood , Essential Hypertension/genetics , Essential Hypertension/immunology , Female , Gene Expression , Gene Frequency , Humans , Male , Prognosis , Risk Factors , Russia , Triglycerides/bloodABSTRACT
Drug discovery opportunities where loss-of-function alleles of a target gene link to a disease-relevant phenotype often require an agonism approach to up-regulate or re-establish the activity of the target gene. Antibody therapy is increasingly recognized as a favored drug modality due to multiple desirable pharmacological properties. However, agonistic antibodies that enhance the activities of the target enzymes are rarely developed because the discovery of agonistic antibodies remains elusive. Here we report an innovative scheme of discovery and characterization of human antibodies capable of binding to and agonizing a circulating enzyme lecithin cholesterol acyltransferase (LCAT). Utilizing a modified human LCAT protein with enhanced enzymatic activity as an immunogen, we generated fully human monoclonal antibodies using the XenoMouse(TM) platform. One of the resultant agonistic antibodies, 27C3, binds to and substantially enhances the activity of LCAT from humans and cynomolgus macaques. X-ray crystallographic analysis of the 2.45 Å LCAT-27C3 complex shows that 27C3 binding does not induce notable structural changes in LCAT. A single administration of 27C3 to cynomolgus monkeys led to a rapid increase of plasma LCAT enzymatic activity and a 35% increase of the high density lipoprotein cholesterol that was observed up to 32 days after 27C3 administration. Thus, this novel scheme of immunization in conjunction with high throughput screening may represent an effective strategy for discovering agonistic antibodies against other enzyme targets. 27C3 and other agonistic human anti-human LCAT monoclonal antibodies described herein hold potential for therapeutic development for the treatment of dyslipidemia and cardiovascular disease.
Subject(s)
Antibodies, Monoclonal , Cardiovascular Diseases , Cholesterol, HDL , Dyslipidemias , Phosphatidylcholine-Sterol O-Acyltransferase , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antigen-Antibody Complex/blood , Antigen-Antibody Complex/chemistry , Antigen-Antibody Complex/immunology , Binding Sites, Antibody , CHO Cells , Cardiovascular Diseases/blood , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/immunology , Cholesterol, HDL/blood , Cholesterol, HDL/immunology , Cricetinae , Cricetulus , Dyslipidemias/blood , Dyslipidemias/drug therapy , Dyslipidemias/immunology , Humans , Macaca fascicularis , Mice , Phosphatidylcholine-Sterol O-Acyltransferase/antagonists & inhibitors , Phosphatidylcholine-Sterol O-Acyltransferase/blood , Phosphatidylcholine-Sterol O-Acyltransferase/chemistry , Phosphatidylcholine-Sterol O-Acyltransferase/immunology , Protein Structure, QuaternaryABSTRACT
PURPOSE OF REVIEW: Interest in the metabolic syndrome-associated osteoarthritis phenotype is increasing. Here, we summarize recently published significant findings. RECENT FINDINGS: Meta-analyses confirmed an association between type 2 diabetes and osteoarthritis and between cardiovascular diseases and osteoarthritis. Recent advances in the study of metabolic syndrome-associated osteoarthritis have focused on a better understanding of the role of metabolic diseases in inducing or aggravating joint damage. In-vivo models of obesity, diabetes, or dyslipidemia have helped to better decipher this association. They give emerging evidence that, beyond the role of common pathogenic mechanisms for metabolic diseases and osteoarthritis (i.e., low-grade inflammation and oxidative stress), metabolic diseases have a direct systemic effect on joints. In addition to the impact of weight, obesity-associated inflammation is associated with osteoarthritis severity and may modulate osteoarthritis progression in mouse models. As well, osteoarthritis synovium from type 2 diabetic patients shows insulin-resistant features, which may participate in joint catabolism. Finally, exciting data are emerging on the association of gut microbiota and circadian rhythm and metabolic syndrome-associated osteoarthritis. SUMMARY: The systemic role of metabolic syndrome in osteoarthritis pathophysiology is now better understood, but new avenues of research are being pursued to better decipher the metabolic syndrome-associated osteoarthritis phenotype.
Subject(s)
Cardiovascular Diseases/epidemiology , Diabetes Mellitus, Type 2/epidemiology , Metabolic Syndrome/epidemiology , Obesity/epidemiology , Osteoarthritis/epidemiology , Cardiovascular Diseases/immunology , Diabetes Mellitus, Type 2/immunology , Diabetes Mellitus, Type 2/metabolism , Dyslipidemias/epidemiology , Dyslipidemias/immunology , Humans , Inflammation/immunology , Insulin Resistance , Metabolic Syndrome/immunology , Obesity/immunology , Osteoarthritis/immunology , Oxidative Stress/immunology , Synovial Membrane/metabolismABSTRACT
BACKGROUND: The CD4+ T-lymphocytes and their subtype CD4 + CD25(high)FoxP3+ regulatory T cells are receiving growing interest as major regulators of atherogenesis. We sought to investigate 1) whether the CD4 + cell subsets were expressed differently in dyslipidemic patients (Pts) and healthy subjects (HS) and 2) whether atorvastatin treatment could be associated in-vivo and in-vitro with cell changes in expression and functional response. METHODS: CD4+ subsets frequency (CD4 + CD25(high)FoxP3+, CD4 + CD25-FoxP3+) and mRNA expression for FoxP3, IL-10 and TGF-ß were evaluated in 30 consecutive Pts at baseline and after a 3-month atorvastatin therapy, and in 17 HS. RESULTS: The % of CD4 + cells did not differ between HS and Pts. The % of CD4 + CD25(high)FoxP3+ was higher in Pts than HS and did not change during treatment. The CD4 + CD25-FoxP3+ cells were similar between the two groups and were lower in Pts at visit 2. Cytokine expression and FoxP3 did not differ in HS and Pts and no substantial change was observed during treatment. At visit 1, CD4 + CD25(high)FoxP3+ cells were significantly correlated with both total-cholesterol (r = 0.570, P = 0.0002), LDL-cholesterol (r = 0.715, P = 0.0001), Apolipoprotein B (r = 0.590, P = 0.0001). In-vitro atorvastatin (up to 5 µM) failed to induce any significant modulation of cell functions. CONCLUSION: CD4 + CD25(high)FoxP3+ regulatory cells seem to be over-stimulated in the early pre-clinical phase of atherosclerosis and a relationship exists between their frequency and circulating lipids. A potential immuno-modulation by statin treatment is not achieved through a normalization in peripheral CD4 + cell subsets.
Subject(s)
Atorvastatin/therapeutic use , Dyslipidemias/drug therapy , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , RNA, Messenger/metabolism , T-Lymphocytes, Regulatory/immunology , Adult , Apolipoproteins A/metabolism , Apolipoproteins B/metabolism , C-Reactive Protein/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Case-Control Studies , Cell Proliferation , Cell Survival/immunology , Cholesterol, HDL/metabolism , Cholesterol, LDL/metabolism , Dyslipidemias/immunology , Dyslipidemias/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Humans , In Vitro Techniques , Interleukin-10/genetics , Interleukin-10/immunology , Longitudinal Studies , Male , Middle Aged , Real-Time Polymerase Chain Reaction , T-Lymphocyte Subsets/immunology , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/immunology , Triglycerides/metabolismABSTRACT
RATIONALE: Insulin resistance and low high-density lipoprotein (HDL) are associated with pulmonary morbidity, including asthma, but the underlying mechanisms are not well elucidated. OBJECTIVES: To investigate whether systemic inflammation underlies the association of metabolic abnormalities with pulmonary function among urban adolescents. METHODS: Th-cell responses and monocyte subsets, and their association with serum homeostatic model assessment of insulin resistance (HOMA-IR) and HDL, and pulmonary function were quantified in 168 adolescents, including 42 obese subjects with asthma, 42 normal-weight subjects with asthma, 40 obese subjects without asthma, and 44 healthy control subjects. Th-cell responses (Th1 [CD4(+)IFNγ(+)] and Th2 [CD4(+)IL4(+)] cells) to stimulation with phytohemagglutinin, leptin, and dust mite, and classical (CD14(+)CD16(-)), resident (CD14(+)CD16(+)), and patrolling (CD14dimCD16(+)) monocytes, and their C-C chemokine receptor type-2 (CCR2) expression were quantified by flow cytometry. MEASUREMENTS AND MAIN RESULTS: Th1/Th2 ratio to all three stimuli was higher in obese subjects with asthma than normal-weight subjects with asthma and directly correlated with HOMA-IR. Classical monocytes inversely associated with Th1/Th2 ratio to phytohemagglutinin (r = -0.43; P = 0.01) and directly with Asthma Control Test score (ß = 1.09; P = 0.04), while patrolling monocytes correlated with Composite Asthma Severity Index score (ß = 1.11; P = 0.04) only among obese subjects with asthma. HDL was inversely associated with patrolling monocytes and directly associated with CCR2 expression on resident monocytes. CCR2 expression on patrolling monocytes predicted residual volume (RV), RV/TLC ratio, and FRC, after adjusting for HDL, but not after adjusting for body mass index. Association of Th1/Th2 ratio with RV, FRC, and inspiratory capacity was attenuated after adjusting for HOMA-IR. CONCLUSIONS: Th1 polarization and monocyte activation among obese subjects with asthma correlates with metabolic abnormalities. Association of monocyte activation with pulmonary function is mediated by body mass index, whereas that of Th1 polarization is mediated by insulin resistance.
Subject(s)
Asthma , Dyslipidemias , Inflammation , Insulin Resistance/physiology , Lung/physiopathology , Obesity , Adolescent , Black or African American , Asthma/ethnology , Asthma/immunology , Asthma/metabolism , Body Mass Index , Case-Control Studies , Comorbidity , Cytokines/blood , Dyslipidemias/ethnology , Dyslipidemias/immunology , Dyslipidemias/metabolism , Female , Hispanic or Latino , Humans , Immunity, Cellular , Incidence , Linear Models , Male , Obesity/ethnology , Obesity/immunology , Obesity/metabolism , Risk Factors , Severity of Illness Index , Urban HealthABSTRACT
During the past few decades, there have been numerous studies related to free radical chemistry. Free radicals including reactive oxygen species (ROS) and reactive nitrogen species are generated by the human body by various endogenous systems, exposure to different physiochemical conditions, or pathological states, and have been implicated in the pathogenesis of many diseases. These free radicals are also the common by-products of many oxidative biochemical reactions in cells. When free radicals overwhelm the body's ability to regulate them, a condition known as oxidative stress ensues. They adversely alter lipids, proteins, and DNA, which trigger a number of human diseases. In a number of pathophysiological conditions, the delicate equilibrium between free radical production and antioxidant capability is distorted, leading to oxidative stress and increased tissue injury. ROS which are mainly produced by vascular cells are implicated as possible underlying pathogenic mechanisms in a progression of cardiovascular diseases including ischemic heart disease, atherosclerosis, cardiac arrhythmia, hypertension, and diabetes. This review summarizes the key roles played by free radicals in the pathogenesis of atherosclerosis, diabetes, and dyslipidaemia. Although not comprehensive, this review also provides a brief perspective on some of the current research being conducted in this area for a better understanding of the role free radicals play in the pathogenesis of atherosclerosis, diabetes, and dyslipidaemia.