Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Ann Clin Transl Neurol ; 9(10): 1643-1659, 2022 10.
Article in English | MEDLINE | ID: mdl-36165097

ABSTRACT

OBJECTIVE: To compare "hybrid immunity" (prior COVID-19 infection plus vaccination) and post-vaccination immunity to SARS CoV-2 in MS patients on different disease-modifying therapies (DMTs) and to assess the impact of vaccine product and race/ethnicity on post-vaccination immune responses. METHODS: Consecutive MS patients from NYU MS Care Center (New York, NY), aged 18-60, who completed primary COVID-19 vaccination series ≥6 weeks previously were evaluated for SARS CoV-2-specific antibody responses with electro-chemiluminescence and multiepitope bead-based immunoassays and, in a subset, live virus immunofluorescence-based microneutralization assay. SARS CoV-2-specific cellular responses were assessed with cellular stimulation TruCulture IFNγ and IL-2 assay and, in a subset, with IFNγ and IL-2 ELISpot assays. Multivariate analyses examined associations between immunologic responses and prior COVID-19 infection while controlling for age, sex, DMT at vaccination, time-to-vaccine, and vaccine product. RESULTS: Between 6/01/2021 and 11/11/2021, 370 MS patients were recruited (mean age 40.6 years; 76% female; 53% non-White; 22% with prior infection; common DMT classes: ocrelizumab 40%; natalizumab 15%, sphingosine-1-phosphate receptor modulators 13%; and no DMT 8%). Vaccine-to-collection time was 18.7 (±7.7) weeks and 95% of patients received mRNA vaccines. In multivariate analyses, patients with laboratory-confirmed prior COVID-19 infection had significantly increased antibody and cellular post-vaccination responses compared to those without prior infection. Vaccine product and DMT class were independent predictors of antibody and cellular responses, while race/ethnicity was not. INTERPRETATION: Prior COVID-19 infection is associated with enhanced antibody and cellular post-vaccine responses independent of DMT class and vaccine type. There were no differences in immune responses across race/ethnic groups.


Subject(s)
COVID-19 , Viral Vaccines , Adult , Antibodies, Viral , COVID-19 Vaccines , Female , Humans , Interleukin-2 , Male , Natalizumab , SARS-CoV-2 , Sphingosine-1-Phosphate Receptors , Viral Vaccines/genetics
2.
J Tradit Complement Med ; 12(5): 447-454, 2022 Sep.
Article in English | MEDLINE | ID: mdl-36081818

ABSTRACT

Background and aim: Resveratrol is a bioactive molecule used in dietary supplements and herbal medicines and consumed worldwide. Prior work showed that resveratrol's anti-atherogenic properties are mediated in part through the adenosine A2A receptor. The present study explores the potential contribution of adenosine A2A receptor activation to neuroprotective action of resveratrol on cognitive deficits in a model of atherosclerosis-prone systemic lupus erythematosus. Experimental procedure: Using behavioral analysis (open field, static rod, novel object recognition) and QRT-PCR, this study measured working memory, anxiety, motor coordination, and expression of mRNA in the brain. Results and conclusion: Data indicate that resveratrol increases working memory, on average but not statistically, and shows a trend towards improved motor coordination (p = 0.07) in atherosclerosis-prone lupus mice. Additionally, resveratrol tends to increase mRNA levels of SIRT1, decrease vascular endothelial growth factor and CX3CL1 mRNA in the hippocampus. Istradefylline, an adenosine A2A receptor antagonist, antagonizes the effects of resveratrol on working memory (p = 0.04) and the expression of SIRT1 (p = 0.03), vascular endothelial growth factor (p = 0.04), and CX3CL1 (p = 0.03) in the hippocampus.This study demonstrates that resveratrol could potentially be a therapeutic candidate in the modulation of cognitive dysfunction in neuropsychiatric lupus, especially motor incoordination. Further human studies, as well as optimization of resveratrol administration, could confirm whether resveratrol may be an additional resource available to reduce the burden of cognitive impairment associated with lupus. Additionally, further studies need to address the role of A2A blockade in cognitive function among the autoimmune population. Section: 3. Dietary therapy/nutrients supplements. Taxonomy classification by EVISE: autoimmunity, inflammation, neurology.

3.
Ann Neurol ; 91(6): 782-795, 2022 06.
Article in English | MEDLINE | ID: mdl-35289960

ABSTRACT

OBJECTIVE: The objective of this study was to determine the impact of multiple sclerosis (MS) disease-modifying therapies (DMTs) on the development of cellular and humoral immunity to severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2) infection. METHODS: Patients with MS aged 18 to 60 years were evaluated for anti-nucleocapsid and anti-Spike receptor-binding domain (RBD) antibody with electro-chemiluminescence immunoassay; antibody responses to Spike protein, RBD, N-terminal domain with multiepitope bead-based immunoassays (MBI); live virus immunofluorescence-based microneutralization assay; T-cell responses to SARS-CoV-2 Spike using TruCulture enzyme-linked immunosorbent assay (ELISA); and IL-2 and IFNγ ELISpot assays. Assay results were compared by DMT class. Spearman correlation and multivariate analyses were performed to examine associations between immunologic responses and infection severity. RESULTS: Between January 6, 2021, and July 21, 2021, 389 patients with MS were recruited (mean age 40.3 years; 74% women; 62% non-White). Most common DMTs were ocrelizumab (OCR)-40%; natalizumab -17%, Sphingosine 1-phosphate receptor (S1P) modulators -12%; and 15% untreated. One hundred seventy-seven patients (46%) had laboratory evidence of SARS-CoV-2 infection; 130 had symptomatic infection, and 47 were asymptomatic. Antibody responses were markedly attenuated in OCR compared with other groups (p ≤0.0001). T-cell responses (IFNγ) were decreased in S1P (p = 0.03), increased in natalizumab (p <0.001), and similar in other DMTs, including OCR. Cellular and humoral responses were moderately correlated in both OCR (r = 0.45, p = 0.0002) and non-OCR (r = 0.64, p <0.0001). Immune responses did not differ by race/ethnicity. Coronavirus disease 2019 (COVID-19) clinical course was mostly non-severe and similar across DMTs; 7% (9/130) were hospitalized. INTERPRETATION: DMTs had differential effects on humoral and cellular immune responses to SARS-CoV-2 infection. Immune responses did not correlate with COVID-19 clinical severity in this relatively young and nondisabled group of patients with MS. ANN NEUROL 2022;91:782-795.


Subject(s)
COVID-19 , Multiple Sclerosis , Adult , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Viral , Ethnicity , Female , Humans , Immunity, Cellular , Immunity, Humoral , Male , Natalizumab/therapeutic use , SARS-CoV-2
4.
Inflamm Res ; 69(10): 975-988, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32699989

ABSTRACT

PURPOSE: Cardiovascular disease (CVD) is the leading cause of death, globally, and its prevalence is only expected to rise due to the increasing incidence of co-morbidities such as obesity and diabetes. Medical treatment of CVD is directed primarily at slowing or reversing the underlying atherosclerotic process by managing circulating lipids with an emphasis on control of low-density lipoprotein (LDL) cholesterol. However, over the past several decades, there has been increasing recognition that chronic inflammation and immune system activation are important contributors to atherosclerosis. This shift in focus has led to the elucidation of the complex interplay between cholesterol and cellular secretion of cytokines involved in CVD pathogenesis. Of the vast array of cytokine promoting atherosclerosis, interferon (IFN)-γ is highly implicated and, therefore, of great interest. METHODS: Literature review was performed to further understand the effect of IFN-γ on the development of atherosclerotic CVD. RESULTS: IFN-γ, the sole member of the type II IFN family, is produced by T cells and macrophages, and has been found to induce production of other cytokines and to have multiple effects on all stages of atherogenesis. IFN-γ activates a variety of signaling pathways, most commonly the Janus kinase (JAK)/signal transducer and activator of transcription (STAT) pathway, to induce oxidative stress, promote foam cell accumulation, stimulate smooth muscle cell proliferation and migration into the arterial intima, enhance platelet-derived growth factor expression, and destabilize plaque. These are just a few of the contributions of IFN-γ to the initiation and progression of atherosclerotic CVD. CONCLUSION: Given the pivotal role of IFN-γ in the advancement of CVD, activation of its signaling pathways is being explored as a driver of atherosclerosis. Manipulation of this key cytokine may lead to novel therapeutic avenues for CVD prevention and treatment. A number of therapies are being explored with IFN-γ as the potential target.


Subject(s)
Cardiovascular Diseases/immunology , Interferon-gamma/immunology , Animals , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/metabolism , Cholesterol/metabolism , Humans , Interferon-gamma/chemistry , Rheumatic Diseases/immunology
5.
Medicina (Kaunas) ; 55(9)2019 Aug 21.
Article in English | MEDLINE | ID: mdl-31438615

ABSTRACT

Background and Objectives: Atherosclerotic cardiovascular disease (CVD) remains a major cause of morbidity and mortality in persons with systemic lupus erythematosus (SLE, lupus). Atherosclerosis, which involves interplay between cholesterol metabolism and cellular inflammatory pathways, is primarily treated with statins since statins have lipid-lowering and anti-inflammatory properties. The Lupus Atherosclerosis Prevention Study (LAPS) was designed to investigate the efficacy of statins against CVD in SLE patients. LAPS demonstrated that 2 years of atorvastatin administration did not reduce atherosclerosis progression in lupus patients. In this LAPs substudy, we use cultured macrophages to explore the atherogenic properties of plasma from LAPS subjects to explain the mechanistic rationale for the inability of statins to reduce CVD in lupus. Materials and Methods: THP-1 differentiated macrophages were treated for 18 h with 10% SLE patient plasma obtained pre- and post-atorvastatin therapy or placebo. Gene expression of the following cholesterol transport genes was measured by qRT-PCR. For efflux-ATP binding cassette transporter (ABC)A1 and ABCG1, 27-hydroxylase, peroxisome proliferator-activated receptor (PPAR)γ, and liver X receptor (LXR)α; and for influx-cluster of differentiation 36 (CD36) and scavenger receptor (ScR)A1. Results: Macrophages exposed to plasma from both statin-treated and placebo-treated groups showed a significant decrease in cholesterol efflux proteins ATP binding cassette (ABC) transporters A1 and ABCG1, an increase in 27-hydroxylase, an increase in the LDL receptor and a decrease in intracellular free cholesterol. No change in influx receptors ScRA1 and CD36, nor nuclear proteins LXRα and PPARγ was observed. Conclusions: Statins do not normalize pro-atherogenic changes induced by lupus and these changes continue to worsen over time. This study provides mechanistic insight into LAPS findings by demonstrating that statins are overall ineffective in altering the balance of cholesterol transport gene expression in human macrophages. Furthermore, our study suggests that statins as a CVD treatment may not be useful in attenuating lipid overload in the SLE environment.


Subject(s)
Atherosclerosis/drug therapy , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Lupus Erythematosus, Systemic/complications , Macrophages/drug effects , ATP Binding Cassette Transporter 1/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 1/genetics , ATP-Binding Cassette Transporters/genetics , Adult , Atherosclerosis/complications , Atorvastatin/therapeutic use , Cholesterol/metabolism , Disease Progression , Female , Gene Expression , Humans , Lipid Metabolism , Liver X Receptors/genetics , Lupus Erythematosus, Systemic/blood , Macrophages/metabolism , Male , Mixed Function Oxygenases/genetics , PPAR gamma/genetics , Plasma , RNA, Messenger/metabolism
6.
J Transl Med ; 17(1): 232, 2019 07 22.
Article in English | MEDLINE | ID: mdl-31331347

ABSTRACT

BACKGROUND: Macrophage cholesterol efflux capacity has been identified as a predictor for cardiovascular disease. We assessed the relationship between adipocyte-derived extracellular vesicle microRNAs and macrophage cholesterol efflux capacity. METHODS: We assessed an adolescent cohort (n = 93, Age, median (IQR) = 17 (3) year, Female = 71, Male = 22) throughout the BMI continuum (BMI = 45.2 (13.2) kg/m2) for: (1) cholesterol efflux capacity and lipoprotein profiles; (2) adipocyte-derived extracellular vesicle microRNAs in serum; (3) the role of visceral adipose tissue extracellular vesicle in regulation of cholesterol efflux and cholesterol efflux gene expression in THP-1 macrophages in vitro. RESULTS: Efflux capacity was significantly associated with HDL (r = 0.30, p = 0.01) and LDL (r = 0.33, p = 0.005) particle size. Multivariate-analysis identified six microRNAs associated (p < 0.05) with cholesterol efflux capacity: miR-3129-5p (Beta = 0.695), miR-20b (0.430), miR9-5p (0.111), miR-320d (- 0.190), miR301a-5p (0.042), miR-155-5p (0.004). In response to increasing concentrations (1 µg/mL vs. 3 µg/mL) of VAT extracellular vesicle, cholesterol efflux (66% ± 10% vs. 49% ± 2%; p < 0.01) and expression of ABCA1 (FC = 1.9 ± 0.8 vs 0.5 ± 0.2; p < 0.001), CD36 (0.7 ± 0.4 vs. 2.1 ± 0.8, p = 0.02), CYP27A1 (1.4 ± 0.4 vs. 0.9 ± 0.5; p < 0.05), and LXRA (1.8 ± 1.1 vs. 0.5 ± 0.2; p < 0.05) was altered in THP-1 cells in vitro. CONCLUSION: Adipocyte-derived extracellular vesicle microRNAs may, in part, be involved macrophage cholesterol efflux regulation.


Subject(s)
Adipose Tissue/metabolism , Cholesterol/metabolism , Extracellular Vesicles/genetics , MicroRNAs/metabolism , Pediatric Obesity/genetics , Adolescent , Biological Transport , Circulating MicroRNA/blood , Circulating MicroRNA/genetics , Female , Humans , Lipoproteins/blood , Macrophages/metabolism , Male , MicroRNAs/genetics , Pediatric Obesity/blood , THP-1 Cells
7.
Inflamm Res ; 68(9): 787-799, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31227843

ABSTRACT

OBJECTIVE: Mycophenolate (MPA) and cyclosporin A (CsA) are two immunosuppressive agents currently used for the treatment of autoimmune diseases. However, reports regarding their effects on inflammation and lipid handling are controversial. Here, we compare the effect of these two drugs on the expression of proteins involved in cholesterol handling and lipid accumulation in a macrophage cell system utilizing M0, M1 and M2 human macrophages and in murine bone marrow-derived macrophages (BMDM). METHODS: Differentiated M0, M1 and M2 subsets of THP-1 human macrophages were subjected to various concentrations of either MPA or CsA. Expression of proteins involved in reverse cholesterol transport (ABCA1 and 27-hydroxylase) and scavenger receptors, responsible for uptake of modified lipids (CD36, ScR-A1, CXCL16 and LOX-1), were evaluated by real-time PCR and confirmed with Western blot. DiI-oxidized LDL internalization assay was used to assess foam cell formation. The influence of MPA was also evaluated in BMDM obtained from atherosclerosis-prone transgenic mice, ApoE-/- and ApoE-/-Fas-/-. RESULTS: In M0 macrophages, MPA increased expression of ABCA1 and CXCL16 in a concentration-dependent manner. In M1 THP-1 macrophages, MPA caused a significant increase of 27-hydroxylase mRNA and CD36 and SR-A1 receptor mRNAs. Exposure of M2 macrophages to MPA also stimulated expression of 27-hydroxylase, while downregulating all evaluated scavenger receptors. In contrast, CsA had no impact on cholesterol efflux in M0 and M1 macrophages, but significantly augmented expression of ABCA1 and 27-hydroxylase in M2 macrophages. CsA significantly increased expression of the LOX1 receptor in naïve macrophages, downregulated expression of CD36 and SR-A1 in the M1 subpopulation and upregulated expression of all evaluated scavenger receptors. However, CsA enhanced foam cell transformation in M0 and M2 macrophages, while MPA had no effect on foam cell formation unless used at a high concentration in the M2 subtype. CONCLUSIONS: Our results clearly underline the importance of further evaluation of the effects of these drugs when used in atherosclerosis-prone patients with autoimmune or renal disease.


Subject(s)
Cyclosporine/pharmacology , Immunosuppressive Agents/pharmacology , Lipid Metabolism/drug effects , Macrophages/drug effects , Mycophenolic Acid/pharmacology , Animals , Atherosclerosis/metabolism , Bone Marrow Cells/cytology , Cell Differentiation , Cholesterol/metabolism , Foam Cells , Humans , Immunosuppression Therapy , Macrophages/metabolism , Mice , Monocytes/cytology , THP-1 Cells
8.
Atherosclerosis ; 278: 49-59, 2018 11.
Article in English | MEDLINE | ID: mdl-30253289

ABSTRACT

Mineral bone disease (MBD) is a common complication of chronic kidney disease (CKD) characterized by disruption of normal mineral homeostasis within the body. One or more of the following may occur: hypocalcemia, hyperphosphatemia, secondary hyperparathyroidism (SHPT), decreased vitamin D and vascular calcification (VC). The greater the decrease in renal function, the worse the progression of CKD-MBD. These abnormalities may lead to bone loss, osteoporosis and fractures. CKD-MBD is a major contributor to the high morbidity and mortality among patients with CKD. Another well-known complication of CKD is cardiovascular disease (CVD) caused by increased atherosclerosis and VC. CVD is the leading cause of morbidity and mortality in CKD patients. VC is linked to reduced arterial compliance that may lead to widened pulse pressure and impaired cardiovascular function. VC is a strong predicator of cardiovascular mortality among patients with CKD. Elevated phosphorus levels and increased calcium-phosphorus product promote VC. Controlling mineral disturbances such as hyperphosphatemia and SHPT is still considered among the current strategies for treatment of VC in CKD through restriction of calcium based phosphate binders in hyperphosphatemic patients across all severities of CKD along with dietary phosphate restriction and use of calciminetics. Additionally, Vitamin D insufficiency is common in CKD and dialysis patients. The causes are multifactorial and a serious consequence is SHPT. Vitamin D compounds remain the first-line therapy for prevention and treatment of SHPT in CKD. Vitamin D may also have atheroprotective effects on the arterial wall, but clinical studies do not show clear evidence of reduced cardiovascular mortality with vitamin D administration. This review discusses the issues surrounding CKD-MBD, cardiovascular disease and approaches to treatment.


Subject(s)
Atherosclerosis/complications , Bone Density , Bone Diseases/complications , Calcium/metabolism , Renal Insufficiency, Chronic/complications , Vascular Calcification/complications , Animals , Atherosclerosis/pathology , Bone Diseases/pathology , Bone and Bones/pathology , Cardiotonic Agents/pharmacology , Clinical Trials as Topic , Homeostasis , Humans , Hyperparathyroidism, Secondary/complications , Hyperparathyroidism, Secondary/pathology , Hyperphosphatemia/complications , Hyperphosphatemia/pathology , Kidney/physiopathology , Phosphates/metabolism , Renal Dialysis , Renal Insufficiency, Chronic/pathology , Sevelamer/pharmacology , Vascular Calcification/pathology , Vitamin D/pharmacology
9.
J Investig Med ; 65(3): 694-704, 2017 03.
Article in English | MEDLINE | ID: mdl-27940550

ABSTRACT

It is the second decade of controversy regarding the cardiovascular effects of cyclo-oxygenase-2 (COX-2) inhibitors. At this time, celecoxib is the only available COX-2-specific inhibitor for treatment of pain and inflammation. Therefore, the present study was designed primarily to determine the impact of celecoxib on cholesterol handling (uptake via scavenger receptors and efflux from the cells) and foam cell formation in human THP-1 macrophages, followed by comparison to rofecoxib and other non-steroidal anti-inflammatory drugs (NSAIDs). THP-1 human macrophages and peripheral blood mononuclear cells were incubated with: celecoxib, rofecoxib, naproxen (at 5, 10, 25 µM) and acetaminophen (0.5 mM, 1 mM)±oxidized low-density lipoprotein (oxLDL, 25 µg/mL). Scavenger receptors: CD36, LOX-1, SR-A1, and CXCL16 and cholesterol efflux proteins: ATP-binding cassette transporter (ABC) A1 and G1, and 27-hydroxylase were detected. The adhesion of monocytes to cultured endothelial cells with/ without COX-2 inhibitors/NSAIDs was also analyzed. The presence of celecoxib and rofecoxib (at high concentrations) significantly decreased expression of 27-hydroxylase and ABCA1, interfering with normal cholesterol outflow from macrophages. Acetaminophen and the non-specific COX inhibitor naproxen had no significant effect on these proteins. Only celecoxib had a profound effect on the class B scavenger receptor CD36 and the class E receptor LOX1. We demonstrate that in contrast to celecoxib, rofecoxib and naproxen increased adhesive properties of monocytes to endothelial cells. This work might contribute to our understanding of multiple mechanisms underlying elevated cardiovascular risk upon the use of COX-2 inhibitors and uncover new possibilities to enhance the safety profile of existing COX-2 inhibitors.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Atherosclerosis/enzymology , Atherosclerosis/pathology , Cyclooxygenase 2 Inhibitors/pharmacology , Cyclooxygenase 2/metabolism , Macrophages/pathology , Monocytes/pathology , Atherosclerosis/genetics , Biological Transport/drug effects , Celecoxib/pharmacology , Cell Adhesion/drug effects , Cell Line , Cholesterol/metabolism , Disease Progression , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Foam Cells/drug effects , Foam Cells/enzymology , Foam Cells/pathology , Gene Expression Regulation/drug effects , Gene Silencing/drug effects , Humans , Inflammation/pathology , Lactones/pharmacology , Lipoproteins, LDL/metabolism , Macrophages/drug effects , Macrophages/enzymology , Models, Biological , Monocytes/drug effects , Monocytes/enzymology , Receptors, Scavenger/metabolism , Sulfones/pharmacology
10.
Exp Biol Med (Maywood) ; 241(14): 1611-9, 2016 08.
Article in English | MEDLINE | ID: mdl-27190277

ABSTRACT

Resveratrol is a bioactive molecule used in dietary supplements and herbal medicines and consumed worldwide. Numerous investigations by our group and others have indicated cardioprotective and anti-inflammatory properties of resveratrol. The present study explored potential atheroprotective actions of resveratrol on cholesterol efflux in cultured human macrophages exposed to plasma from systemic lupus erythematosus (SLE) patients. These results were confirmed in ApoE(-/-)Fas(-/-) double knockout mice, displaying a lupus profile with accelerated atherosclerosis. Resveratrol treatment attenuated atherosclerosis in these mice. THP-1 human macrophages were exposed to 10% pooled or individual plasma from patients who met diagnostic criteria for SLE. Expression of multiple proteins involved in reverse cholesterol transport (ABCA1, ABCG1, SR-B1, and cytochrome P450 27-hydroxylase) was assessed using QRT-PCR and Western blotting techniques. Ten-week-old ApoE(-/-)Fas(-/-) double knockout mice (n = 30) were randomly divided into two equal groups of 15, one of which received 0.01% resveratrol for 10 consecutive weeks. Atherosclerosis progression was evaluated in murine aortas. Bone marrow-derived macrophages (BMDM) were cultured and expression of cholesterol efflux proteins was analyzed in each group of mice. Our data indicate that inhibition of cholesterol efflux by lupus plasma in THP-1 human macrophages is rescued by resveratrol. Similarly, administration of resveratrol in a lupus-like murine model reduces plaque formation in vivo and augments cholesterol efflux in BMDM. This study presents evidence for a beneficial role of resveratrol in atherosclerosis in the specific setting of SLE. Therefore, resveratrol may merit investigation as an additional resource available to reduce lipid deposition and atherosclerosis in humans, especially in such vulnerable populations as lupus patients.


Subject(s)
Cholesterol/metabolism , Lupus Erythematosus, Systemic/complications , Stilbenes/therapeutic use , Animals , Aorta/drug effects , Aorta/metabolism , Aorta/pathology , Apolipoproteins E/genetics , Atherosclerosis/complications , Atherosclerosis/metabolism , Atherosclerosis/pathology , Humans , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Resveratrol , fas Receptor/genetics
11.
Am J Kidney Dis ; 66(6): 1071-82, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26337134

ABSTRACT

Patients with chronic kidney disease (CKD) have a substantial risk of developing coronary artery disease. Traditional cardiovascular disease (CVD) risk factors such as hypertension and hyperlipidemia do not adequately explain the high prevalence of CVD in CKD. Both CVD and CKD are inflammatory states and inflammation adversely affects lipid balance. Dyslipidemia in CKD is characterized by elevated triglyceride levels and high-density lipoprotein levels that are both decreased and dysfunctional. This dysfunctional high-density lipoprotein becomes proinflammatory and loses its atheroprotective ability to promote cholesterol efflux from cells, including lipid-overloaded macrophages in the arterial wall. Elevated triglyceride levels result primarily from defective clearance. The weak association between low-density lipoprotein cholesterol level and coronary risk in CKD has led to controversy over the usefulness of statin therapy. This review examines disrupted cholesterol transport in CKD, presenting both clinical and preclinical evidence of the effect of the uremic environment on vascular lipid accumulation. Preventative and treatment strategies are explored.


Subject(s)
Cardiovascular Diseases/metabolism , Cholesterol, HDL/metabolism , Renal Insufficiency, Chronic/metabolism , Animals , Biological Transport/physiology , Cardiovascular Diseases/diagnosis , Cardiovascular Diseases/epidemiology , Humans , Renal Insufficiency, Chronic/diagnosis , Renal Insufficiency, Chronic/epidemiology , Risk Factors
12.
Biomed Res Int ; 2014: 312647, 2014.
Article in English | MEDLINE | ID: mdl-24587984

ABSTRACT

Tumor necrosis factor- (TNF-) α is a proinflammatory proatherogenic cytokine. Infliximab, an anti-TNF-α monoclonal antibody, is effective in treating rheumatoid arthritis. However, its impact on cardiovascular burden and lipid transport is unclear. The present study investigates the effect of TNF-α and infliximab on reverse cholesterol transport (RCT) proteins. Uptake of modified lipoproteins by macrophages in the vasculature leads to atherogenic foam cell formation. RCT is mediated by proteins including ATP binding cassette transporters A1 (ABCA1), G1 (ABCG1), liver X receptor- (LXR-) α, and 27-hydroxylase. RCT counteracts lipid overload by ridding cells of excess cholesterol. THP-1 human monocytes were incubated with either TNF-α alone or TNF-α with infliximab. Expression of proteins involved in cholesterol efflux was analyzed. TNF-α significantly reduced both ABCA1 and LXR-α mRNA (to 68.5 ± 1.59%, P < 0.05, and 41.2 ± 0.25%, P < 0.01, versus control set as 100%, resp.). Infliximab nullified the TNF-α effect. Results were confirmed by Western blot. Infliximab abolished the increase in foam cells induced by TNF-α. TNF-α treatment significantly reduces ABCA1 and LXR-α expression in monocytes, thus bringing about a proatherogenic state. The anti-TNF drug infliximab, commonly used in rheumatology, restored RCT proteins. This is the first report of an atheroprotective effect of infliximab on RCT in monocytes.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Antibodies, Monoclonal/pharmacology , Atherosclerosis/metabolism , Cholestanetriol 26-Monooxygenase/metabolism , Cholesterol/metabolism , Orphan Nuclear Receptors/metabolism , Tumor Necrosis Factor-alpha/pharmacology , ATP-Binding Cassette Transporters/genetics , Biological Transport/drug effects , Cell Line , Humans , Infliximab , Lipoproteins, LDL/metabolism , Liver X Receptors , Macrophages/drug effects , Macrophages/metabolism , Monocytes/drug effects , Monocytes/metabolism , Orphan Nuclear Receptors/genetics
13.
Trends Cardiovasc Med ; 24(1): 45-51, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23916809

ABSTRACT

Atherosclerosis is considered to be a chronic inflammatory disease of the arterial wall. Atherogenesis is accompanied by local production and release of inflammatory mediators, for which the macrophage is a major source. The proinflammatory cytokine, interferon (IFN)-γ derived from T cells, is expressed at high levels in atherosclerotic lesions. IFN-γ is the classic macrophage-activating factor, vital for both innate and adaptive immunity. It primes macrophages to produce chemokines and cytotoxic molecules and induces expression of genes that regulate lipid uptake. IFN-γ is a key trigger for the formation and release of reactive oxygen species. IFN-γ has important effects on endothelial cells, promoting expression of adhesion molecules. Atherogenic effects of IFN-γ have been shown in murine models where exogenous administration enhances atherosclerotic lesion formation while knockout of IFN-γ or its receptor reduces lesion size. IFN-γ signaling is largely mediated by a Janus kinase (JAK) to signal transduction and activator of transcription (STAT)1 cytosolic factor pathway. A clear understanding of IFN-γ effects on atherogenesis should enable development of novel targeted interventions for clinical use in the prevention and treatment of atherosclerosis. This review will discuss the actions of the cytokine IFN-γ and its complex effects on cells involved in atherosclerosis.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Atherosclerosis/drug therapy , Cardiovascular Agents/therapeutic use , Inflammation Mediators/antagonists & inhibitors , Interferon-gamma/antagonists & inhibitors , Animals , Atherosclerosis/immunology , Atherosclerosis/metabolism , Drug Design , Humans , Inflammation Mediators/metabolism , Interferon-gamma/metabolism , Molecular Targeted Therapy , Signal Transduction/drug effects
14.
Exp Biol Med (Maywood) ; 238(10): 1192-7, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24000379

ABSTRACT

Immunologic derangements in rheumatoid arthritis (RA) patients likely contribute to premature atherosclerotic cardiovascular disease (CVD). Traditional CVD risk factors do not reliably identify at-risk RA patients, probably because disease-associated mechanisms are not taken into account. The purpose of this study was to determine whether plasma from subjects with RA exhibits atheroma-promoting properties leading to disruption of cholesterol homeostasis in human monocytes/macrophages. Twenty-one healthy controls (HC) and 22 RA patients were enrolled in an IRB approved study at Winthrop University Hospital. Naïve THP-1 macrophages were exposed to plasma from each HC and RA patient. Following incubation, RNA and protein were isolated. QRT-PCR and Western blotting techniques were then used to measure expression of proteins responsible for cholesterol efflux (ATP binding cassette transporter (ABC)A1, ABCG1, 27-hydroxylase) and cholesterol uptake (CD36, ScR-A1, lectin oxidized low density lipoprotein receptor (LOX)-1, CXCL16). To confirm the pro-atherogenic effects of RA plasma on macrophages, foam cell formation was quantified. Results showed that RA plasma downregulates cholesterol efflux proteins and upregulates scavenger receptors CD36, LOX1 and CXCL16. These pro-atherogenic changes in gene expression in the presence of RA plasma are associated with augmented lipid accumulation and foam cell formation by THP-1 macrophages. RA plasma induces macrophage cholesterol overload. Demonstration of disrupted cholesterol homeostasis mediated by RA plasma provides further evidence of the involvement of the immune system in atherogenesis. Our data suggest that chronic exposure to RA plasma adversely affects the capacity of monocytes/macrophages in the arterial wall to metabolize cholesterol and maintain lipid homeostasis, thereby contributing to the development of premature atherosclerosis.


Subject(s)
Arthritis, Rheumatoid/physiopathology , Atherosclerosis/physiopathology , Cholesterol/metabolism , Macrophages/metabolism , Membrane Transport Proteins/biosynthesis , Plasma/chemistry , Adult , Arthritis, Rheumatoid/complications , Atherosclerosis/etiology , Blotting, Western , Cell Line , Female , Gene Expression Profiling , Homeostasis , Humans , Middle Aged , Proteome/analysis , Real-Time Polymerase Chain Reaction
15.
Eur J Pharmacol ; 698(1-3): 299-309, 2013 Jan 05.
Article in English | MEDLINE | ID: mdl-23041272

ABSTRACT

Resveratrol is a bioactive molecule used in dietary supplements and herbal medicines and consumed worldwide. Known cardioprotective and anti-inflammatory properties of resveratrol have spurred investigation of the mechanisms involved. The present study explored potential atheroprotective actions of resveratrol on cholesterol metabolism in cells of the arterial wall, including human macrophages and arterial endothelium. Using QRT-PCR and Western blotting techniques, we measured expression of the proteins involved in reverse cholesterol transport (ABCA1, ABCG1 and SR-B1) and the scavenger receptors responsible for uptake of modified cholesterol (CD36, SR-A1 and LOX-1). We analyzed the effect of resveratrol on apoA-1-and HDL-mediated cholesterol efflux in human THP-1 macrophages. The effect of resveratrol on oxLDL internalization and foam cell formation were evaluated using confocal and light microscopy. Our data indicate that resveratrol regulates expression of major proteins involved in cholesterol transport, promotes apoA-1 and HDL-mediated efflux, downregulates oxLDL uptake and diminishes foam cell formation. Mechanistically, resveratrol effects were dependent upon PPAR-γ and adenosine 2A receptor pathways. For the first time we demonstrate that resveratrol regulates expression of the cholesterol metabolizing enzyme cytochrome P450 27-hydroxylase, providing efficient cholesterol elimination via formation of oxysterols. This study establishes that resveratrol attenuates lipid accumulation in cultured human macrophages via effects on cholesterol transport. Further in vivo studies are needed to determine whether resveratrol may be an additional resource available to reduce lipid deposition and atherosclerosis in humans.


Subject(s)
Adenosine/metabolism , Atherosclerosis/prevention & control , Cholesterol/metabolism , Macrophages/drug effects , Macrophages/metabolism , PPAR gamma/metabolism , Stilbenes/pharmacology , Animals , Apolipoprotein A-I/metabolism , Atherosclerosis/genetics , Atherosclerosis/metabolism , Biological Transport/drug effects , CD36 Antigens/genetics , Cell Line , Cholesterol/biosynthesis , Endothelium/drug effects , Endothelium/metabolism , Foam Cells/cytology , Foam Cells/drug effects , Foam Cells/metabolism , Gene Expression Regulation/drug effects , Humans , Lipoproteins, HDL/metabolism , Lipoproteins, LDL/metabolism , Monocytes/drug effects , Monocytes/metabolism , Receptors, Adenosine A2/metabolism , Resveratrol
16.
Biochim Biophys Acta ; 1831(2): 407-16, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23168167

ABSTRACT

The adenosine A(2A) receptor (A(2A)R) plays an important role in the regulation of inflammatory and immune responses. Our previous work has demonstrated that A(2A)R agonists exhibit atheroprotective effects by increasing expression of reverse cholesterol transport proteins in cultured human macrophages. This study explores the impact of pharmacologic activation/inhibition and gene silencing of A(2A)R on cholesterol homeostasis in both THP-1 human monocytes/macrophages and primary human aortic endothelial cells (HAEC). THP-1 human monocytes/macrophages and HAEC exposed to the A(2A)R-specific agonist ATL313 exhibited upregulation of proteins responsible for cholesterol efflux: the ABCA1 and G1 transporters. Further, activation of A(2A)R led to upregulation of the cholesterol metabolizing enzyme P450 27-hydroxylase, accompanied by intracellular changes in level of oxysterols. We demonstrate that anti-atherogenic properties of A(2A)R activation are not limited to the regulation of lipid efflux in vasculature, but include protection from lipid overload in macrophages, particularly via suppression of the CD36 scavenger receptor. The reduced lipid accumulation manifests directly as a diminution in foam cell transformation. In THP-1 macrophages, either A(2A)R pharmacological blockade or gene silencing promote lipid accumulation and enhance foam cell transformation. Our pre-clinical data provides evidence suggesting that A(2A)R stimulation by ATL313 has the potential to be a viable therapeutic strategy for cardiovascular disease prevention, particularly in patients with elevated risk due to immune/inflammatory disorders.


Subject(s)
Cholesterol/metabolism , Endothelium, Vascular/metabolism , Macrophages/metabolism , Receptor, Adenosine A2A/metabolism , Base Sequence , Blotting, Western , Cell Line , DNA Primers , Endothelium, Vascular/cytology , Humans , Microscopy, Confocal , Microscopy, Fluorescence , Polymerase Chain Reaction
17.
J Med Food ; 15(9): 763-73, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22856383

ABSTRACT

Resveratrol, a natural polyphenol produced by plants in response to environmental stress, has received great attention during the past few years due to its beneficial roles in longevity and glucose homeostasis. Resveratrol has been found to display antioxidant, anti-inflammatory, antifibrotic, and cardioprotective properties. Resveratrol reduces platelet aggregation, induces vasorelaxation, limits endothelial activation, and modulates lipid and lipoprotein metabolism. Although the mechanisms of action of resveratrol have not been completely defined, there is evidence that some of the effects of resveratrol may be mediated via activation of sirtuin 1 and AMP-activated protein kinase and through inhibition of the pleiotropic transcription factor nuclear factor κB. Pathways proposed to underlie resveratrol-mediated cardioprotection include reduction of oxidative stress and activation of endothelial nitric oxide synthase. Adenosinergic mechanisms may play a role in its atheroprotective activity. The ability of the nutraceutical resveratrol to positively influence the future treatment of cardiovascular disease is discussed.


Subject(s)
Antioxidants/therapeutic use , Atherosclerosis/prevention & control , Cholesterol/metabolism , Dietary Supplements , Stilbenes/therapeutic use , Angiogenesis Modulating Agents/adverse effects , Angiogenesis Modulating Agents/metabolism , Angiogenesis Modulating Agents/therapeutic use , Animals , Antioxidants/adverse effects , Antioxidants/metabolism , Atherosclerosis/blood , Atherosclerosis/metabolism , Cardiotonic Agents/adverse effects , Cardiotonic Agents/metabolism , Cardiotonic Agents/therapeutic use , Cholesterol/blood , Dietary Supplements/adverse effects , Humans , Resveratrol , Stilbenes/adverse effects , Stilbenes/metabolism , Vasodilator Agents/adverse effects , Vasodilator Agents/metabolism , Vasodilator Agents/therapeutic use
18.
J Investig Med ; 60(3): 576-82, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22367100

ABSTRACT

Alzheimer disease (AD) is an age-related neurodegenerative disorder that manifests as a progressive loss of memory and deterioration of higher cognitive functions. Alzheimer disease is characterized by accumulation in the brain of the ß-amyloid peptide generated by ß- and γ-secretase processing of amyloid precursor protein. Epidemiological studies have linked elevated plasma cholesterol and lipoprotein levels in midlife with AD development. Cholesterol-fed animal models exhibit neuropathologic features of AD including accumulation of ß-amyloid peptide. Specific isoforms of the cholesterol transporter apolipoprotein E are associated with susceptibility to AD. Although multiple lines of evidence indicate a role for cholesterol in AD, the exact impact and mechanisms involved remain largely unknown. This review summarizes the current state of our knowledge of the influence of cholesterol and lipid pathways in AD pathogenesis in vitro and in vivo.


Subject(s)
Alzheimer Disease/metabolism , Brain/metabolism , Cholesterol/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Animals , Humans , Lipid Metabolism/physiology , Signal Transduction/physiology
19.
Prog Lipid Res ; 50(3): 213-24, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21352852

ABSTRACT

Atherosclerotic cardiovascular disease is the leading cause of morbidity and mortality in the United States and in many other countries. Dysfunctional lipid homeostasis plays a central role in the initiation and progression of atherosclerotic lesions. The ATP-binding cassette (ABC) transporters are transmembrane proteins that hydrolyze ATP and use the energy to drive the transport of various molecules across cell membranes. Several ABC transporters play a pivotal role in lipid trafficking. They are critically involved in cholesterol and phospholipid efflux and reverse cholesterol transport (RCT), processes that maintain cellular cholesterol homeostasis and protect arteries from atherosclerosis. In this article we provide a review of the current literature on the biogenesis of ABC transporters and highlight their proposed functions in atheroprotection.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Atherosclerosis/metabolism , Cholesterol/metabolism , Lipid Metabolism , Animals , Biliary Tract/physiology , Biological Transport , Cholesterol, HDL/metabolism , Cholesterol, LDL/metabolism , Humans , Structure-Activity Relationship
20.
Lipids ; 46(2): 131-42, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21181286

ABSTRACT

Cardiovascular safety of cyclooxygenase (COX)-2-selective inhibitors and nonselective nonsteroidal anti-inflammatory drugs (NSAIDs) is of worldwide concern. COX-2 inhibitors and NSAIDs act by inhibiting arachidonic acid metabolism to prostaglandins. They confer a cardiovascular hazard manifested as an elevated risk of myocardial infarction. Mechanisms underlying these cardiovascular effects are uncertain. Here we determine whether interference with cytosolic phospholipase A2 (cPLA-2) or COX-2 through pharmacologic blockade or silencing RNA impacts expression of scavenger receptor CD36 and scavenger receptor A, both involved in cholesterol uptake in monocytes and macrophages. THP-1 human monocytes and human peripheral blood mononuclear cells were exposed to celecoxib, a COX-2 selective inhibitor currently in clinical use, and to arachidonyl trifluoromethyl ketone (AACOCF3), an arachidonic acid analog that selectively inhibits cPLA-2. Celecoxib and AACOCF3 each upregulated expression of CD36, but not scavenger receptor A, as determined by quantitative PCR and immunoblotting. Silencing of cPLA-2 or COX-2 had comparable effects to pharmacologic treatments. Oil red O staining revealed a profound increase in foam cell transformation of THP-1 macrophages exposed to either celecoxib or AACOCF3 (both 25 µM), supporting a role for the COX pathway in maintaining macrophage cholesterol homeostasis. Demonstration of disrupted cholesterol balance by AACOCF3 and celecoxib provides further evidence of the possible mechanism by which COX inhibition may promote lipid overload leading to atheromatous lesion formation and increased cardiovascular events.


Subject(s)
Arachidonic Acids/adverse effects , CD36 Antigens/biosynthesis , Cyclooxygenase 2 Inhibitors/adverse effects , Cyclooxygenase 2/metabolism , Enzyme Inhibitors/adverse effects , Foam Cells/enzymology , Phospholipases A2, Cytosolic/antagonists & inhibitors , Pyrazoles/adverse effects , Sulfonamides/adverse effects , CD36 Antigens/genetics , Celecoxib , Cell Culture Techniques , Cells, Cultured , Cholesterol/metabolism , Cyclooxygenase 2/genetics , Gene Expression , Gene Silencing , Humans , Monocytes/drug effects , Monocytes/metabolism , Phospholipases A2, Cytosolic/genetics , Phospholipases A2, Cytosolic/metabolism , Plaque, Atherosclerotic/chemically induced , Plaque, Atherosclerotic/enzymology , Scavenger Receptors, Class A/biosynthesis , Scavenger Receptors, Class A/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...