Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 203
Filter
1.
Int J Mol Sci ; 25(4)2024 Feb 17.
Article in English | MEDLINE | ID: mdl-38397063

ABSTRACT

Persistent immune activation is linked to an increased risk of cardiovascular disease (CVD) in people with HIV (PWH) on antiretroviral therapy (ART). The NLRP3 inflammasome may contribute to elevated CVD risk in PWH. This study utilized peripheral blood mononuclear cells (PBMCs) from 25 PWH and 25 HIV-negative controls, as well as HIV in vitro infections. Transcriptional changes were analyzed using RNAseq and pathway analysis. Our results showed that in vitro HIV infection of macrophages and PBMCs from PWH had increased foam cell formation and expression of the NLRP3 inflammasome components and downstream cytokines (caspase-1, IL-1ß, and IL-18), which was reduced with inhibition of NLRP3 activity using MCC950. Transcriptomic analysis revealed an increased expression of multiple genes involved in lipid metabolism, cholesterol storage, coronary microcirculation disorders, ischemic events, and monocyte/macrophage differentiation and function with HIV infection and oxLDL treatment. HIV infection and NLRP3 activation increased foam cell formation and expression of proinflammatory cytokines, providing insights into the mechanisms underlying HIV-associated atherogenesis. This study suggests that HIV itself may contribute to increased CVD risk in PWH. Understanding the involvement of the inflammasome pathway in HIV atherosclerosis can help identify potential therapeutic targets to mitigate cardiovascular risks in PWH.


Subject(s)
Atherosclerosis , Foam Cells , HIV Infections , Humans , Atherosclerosis/immunology , Cytokines , Foam Cells/immunology , HIV Infections/complications , HIV Infections/immunology , Inflammasomes/metabolism , Interleukin-1beta/metabolism , Leukocytes, Mononuclear/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
2.
Mol Immunol ; 163: 196-206, 2023 11.
Article in English | MEDLINE | ID: mdl-37837955

ABSTRACT

Macrophages play an important role in the pathogenesis of atherosclerosis (AS) by mediating oxidative stress, inflammation and lipid metabolism, which can lead to the formation of vascular plaque. The Rac family isoforms of small molecules GTPase are active by binding to GTPase, but are inactivated by binding to GDP, and play a role in the switch of cell information conduction. This experiment adopts shRNA interference THP-1 cells respectively each subtype expression and inhibiting Rac1, Rac2, Rac3 activity, each subtype of Rac family on lipid metabolism, inflammatory reaction and oxidative stress. THP-1 cells were stimulated with Ox-LDL to establish AS cell models including lipid loading, adhesion, migration and chemotaxis. Oil Red O staining, cell immunofluorescence, scratching test, transwell, Western blot and other experiments were performed. To observe the different effects of three subtypes of Rac family on multiple links in the foaming process of THP-1 cells. ApoE-/- mice on a high-fat diet were used as animal models to examine the effects of Rac subtypes in vivo. The results showed that the activation of immune cells induced by ox-LDL was inhibited when Rac1, Rac2 and Rac3 in THP-1 were decreased, respectively. Thus, Rac1 and Rac3 act in combination with ox-LDL and are associated with cellular oxidative stress and inflammation. This study provides new means and ideas for finding potential intervention targets that have important regulatory effects on atherosclerosis, and provides a new direction for the development of clinical drugs.


Subject(s)
Atherosclerosis , Foam Cells , Plaque, Atherosclerotic , Animals , Mice , Atherosclerosis/metabolism , Foam Cells/immunology , Immunity , Inflammation/metabolism , Lipoproteins, LDL/pharmacology , Lipoproteins, LDL/metabolism , Plaque, Atherosclerotic/immunology
3.
Front Immunol ; 13: 1050984, 2022.
Article in English | MEDLINE | ID: mdl-36591228

ABSTRACT

Nonalcoholic steatohepatitis (NASH) is the progressive form of nonalcoholic fatty liver disease (NAFLD), and the dysregulation of lipid metabolism and oxidative stress are the typical features. Subsequent dyslipidemia and oxygen radical production may render the formation of modified lipids. Macrophage scavenger receptor 1 (MSR1) is responsible for the uptake of modified lipoprotein and is one of the key molecules in atherosclerosis. However, the unrestricted uptake of modified lipoproteins by MSR1 and the formation of cholesterol-rich foamy macrophages also can be observed in NASH patients and mouse models. In this review, we highlight the dysregulation of lipid metabolism and oxidative stress in NASH, the alteration of MSR1 expression in physiological and pathological conditions, the formation of modified lipoproteins, and the role of MSR1 on macrophage foaming and NASH development and progression.


Subject(s)
Foam Cells , Macrophages , Non-alcoholic Fatty Liver Disease , Scavenger Receptors, Class A , Animals , Mice , Foam Cells/immunology , Foam Cells/pathology , Lipoproteins/immunology , Macrophages/immunology , Macrophages/pathology , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/immunology , Non-alcoholic Fatty Liver Disease/pathology , Scavenger Receptors, Class A/genetics , Scavenger Receptors, Class A/immunology , Disease Progression , Humans
4.
Exp Cell Res ; 409(2): 112922, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34780785

ABSTRACT

Atherosclerosis is generally accepted as a chronic inflammatory disease and is the most important pathological process underlying the cardiovascular diseases. MiR-22 exerts an important role in tumorgenesis, obesity and NAFLD development, as well as cardiovascular diseases. However, a certain role of miR-22 in the pathogenesis of atherosclerosis remains undetermined. Here, we showed that miR-22 exhibited a negative association with the deteriorated atherosclerotic plaque and showed significant downregulated expression in macrophages. Next, treatment of ApoE deficiency (ApoE-/-) mice with miR-22 inhibitors which were then subjected to high fat diet (HFD) for 12 weeks were performed to investigate the function of miR-22 on atherogenesis. The results exhibited that miR-22 inhibition dramatically promoted atherosclerotic plaques but attenuated plaque stabilization which were accompanied by decreased smooth muscle cell and collagen content, but increased macrophage infiltration and lipid accumulation. More importantly, the in vivo and in vitro experiments suggested that miR-22 inhibition accelerated inflammatory response and foam cell formation. Mechanistically, we demonstrated interferon regulator factor 5 (IRF5) was an important target of miR-22 and it was required for the regulation of inflammation mediated by miR-22 inhibition. Collectively, these evidences revealed that miR-22 inhibition promoted the atherosclerosis progression through activation of IRF5.


Subject(s)
Atherosclerosis/pathology , Foam Cells/immunology , Gene Expression Regulation , Inflammation/pathology , Interferon Regulatory Factors/metabolism , MicroRNAs/antagonists & inhibitors , Plaque, Atherosclerotic/pathology , Animals , Apoptosis , Atherosclerosis/etiology , Atherosclerosis/metabolism , Cell Proliferation , Cells, Cultured , Diet, High-Fat , Inflammation/etiology , Inflammation/metabolism , Interferon Regulatory Factors/genetics , Mice , Mice, Knockout, ApoE , MicroRNAs/genetics , Plaque, Atherosclerotic/etiology , Plaque, Atherosclerotic/metabolism
5.
Front Immunol ; 12: 611280, 2021.
Article in English | MEDLINE | ID: mdl-34539620

ABSTRACT

Introduction: Lung diseases are an increasing global health burden affecting millions of people worldwide. Only a few new inhaled medicines have reached the market in the last 30 years, in part due to foamy alveolar macrophage (FAM) responses observed in pre-clinical rat studies. The induction mechanism and signaling pathways involved in the development of highly vacuolated 'foamy' phenotype is not known. Furthermore, it has not been determined if these observations are adaptive or adverse responses. Aim: To determine if high content image analysis techniques can distinguish between alveolar macrophage activation (LPS/IFN-γ activated and IL-4 activated macrophages) and if this could be applied to understanding the generation of 'foamy' macrophage phenotypes. Methods: NR8383 rat alveolar macrophages were stimulated with a mix of cytokines (LPS/IFN-γ or IL-4) for 24 h. The cells were further exposed to FAM inducing-compounds amiodarone and staurosporine. Following 24 h incubation, phagocytosis and lipid accumulation were measured using flow cytometry and high content image analysis techniques. The alveolar macrophages responses after exposure to cytokines were assessed by evaluation: (i) cell surface and biochemical markers such as: nitric oxide production, arginase-1 activity and MRC-1 receptor expression (ii) cellular morphology (iii) cellular functionality (phagocytic activity and lipids accumulation). Results: Macrophages activated with LPS/IFN-γ showed distinct morphological (increased vacuolation) features and functionality (increased lipidosis, decreased phagocytic activity). Foamy macrophage phenotypes induced by amiodarone also displayed characteristics of proinflammatory macrophages (significantly increased nitric oxide production, increased vacuolation and lipidosis and decreased phagocytosis). In contrast, staurosporine treatment resulted in increased NO production, as well as arginase-1 activity. Conclusion: High content image analysis was able to determine distinct differences in morphology between non-activated and LPS/IFN-γ activated macrophages, characterized by increased vacuolation and lipidosis. When exposed to compounds that induce a FAM phenotype, healthy non-activated macrophages displayed proinflammatory (amiodarone) or pro-apoptotic (staurosporine) characteristics but these responses were independent of a change in activation status. This technique could be applied in early drug discovery safety assessment to identify immune responses earlier and increase the understanding of alveolar macrophage responses to new molecules challenge in development of new inhalation therapies, which in turn will enhance decision-making in an early safety assessment of novel drug candidates.


Subject(s)
Foam Cells/metabolism , Foam Cells/pathology , Macrophage Activation/immunology , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/pathology , Macrophages/cytology , Macrophages/metabolism , Molecular Imaging , Biomarkers , Cells, Cultured , Cytokines/metabolism , Foam Cells/immunology , Immunophenotyping , Lipid Metabolism , Macrophages/immunology , Macrophages, Alveolar/immunology , Molecular Imaging/methods , Nitric Oxide/metabolism , Phagocytosis/immunology
6.
Biochem Biophys Res Commun ; 556: 65-71, 2021 06 04.
Article in English | MEDLINE | ID: mdl-33839416

ABSTRACT

Ethyl gallate (EG) is a well-known constituent of medicinal plants, but its effects on atherosclerosis development are not clear. In the present study, the anti-atherosclerosis effects of EG and the underlying mechanisms were explored using macrophage cultures, zebrafish and apolipoprotein (apo) E deficient mice. Treatment of macrophages with EG (20 µM) enhanced cellular cholesterol efflux to HDL, and reduced net lipid accumulation in response to oxidized LDL. Secretion of monocyte chemotactic protein-1 (MCP-1) and interleukin-6 (IL-6) from activated macrophages was also blunted by EG. Fluorescence imaging techniques revealed EG feeding of zebrafish reduced vascular lipid accumulation and inflammatory responses in vivo. Similar results were obtained in apoE-/- mice 6.5 months of age, where plaque lesions and monocyte infiltration into the artery wall were reduced by 70% and 42%, respectively, after just 6 weeks of injections with EG (20 mg/kg). HDL-cholesterol increased 2-fold, serum cholesterol efflux capacity increased by ∼30%, and the levels of MCP-1 and IL-6 were reduced with EG treatment of mice. These results suggest EG impedes early atherosclerosis development by reducing the lipid and macrophage-content of plaque. Underlying mechanisms appeared to involve HDL cholesterol efflux mechanisms and suppression of pro-inflammatory cytokine secretion.


Subject(s)
Atherosclerosis/drug therapy , Atherosclerosis/metabolism , Benzoates/metabolism , Gallic Acid/analogs & derivatives , Lipid Metabolism/drug effects , Plants, Medicinal/metabolism , ATP-Binding Cassette Transporters/biosynthesis , ATP-Binding Cassette Transporters/genetics , Animals , Apolipoproteins E/deficiency , Atherosclerosis/pathology , Atherosclerosis/prevention & control , Cholesterol, HDL/blood , Cholesterol, HDL/metabolism , Cytokines/metabolism , Disease Models, Animal , Female , Foam Cells/cytology , Foam Cells/drug effects , Foam Cells/immunology , Foam Cells/metabolism , Gallic Acid/administration & dosage , Gallic Acid/metabolism , Gallic Acid/pharmacology , Gallic Acid/therapeutic use , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/prevention & control , Inflammation Mediators/metabolism , Mice , Mice, Inbred C57BL , Plaque, Atherosclerotic/drug therapy , Plaque, Atherosclerotic/metabolism , Plaque, Atherosclerotic/pathology , Plaque, Atherosclerotic/prevention & control , RAW 264.7 Cells , Up-Regulation/drug effects , Zebrafish/metabolism
7.
Molecules ; 26(1)2021 Jan 01.
Article in English | MEDLINE | ID: mdl-33401401

ABSTRACT

There is a high level of interest in identifying metabolites of endogenously produced or dietary compounds generated by the gastrointestinal (GI) tract microbiota, and determining the functions of these metabolites in health and disease. There is a wealth of compelling evidence that the microbiota is linked with many complex chronic inflammatory diseases, including atherosclerosis. Macrophages are key target immune cells in atherosclerosis. A hallmark of atherosclerosis is the accumulation of pro-inflammatory macrophages in coronary arteries that respond to pro-atherogenic stimuli and failure of digesting lipids that contribute to foam cell formation in atherosclerotic plaques. This review illustrates the role of tryptophan-derived microbiota metabolites as an aryl hydrocarbon receptor (AhR) ligand that has immunomodulatory properties. Also, microbiota-dependent trimethylamine-N-oxide (TMAO) metabolite production is associated with a deleterious effect that promotes atherosclerosis, and metabolite indoxyl sulfate has been shown to exacerbate atherosclerosis. Our objective in this review is to discuss the role of microbiota-derived metabolites in atherosclerosis, specifically the consequences of microbiota-induced effects of innate immunity in response to atherogenic stimuli, and how specific beneficial/detrimental metabolites impact the development of atherosclerosis by regulating chronic endotoxemic and lipotoxic inflammation.


Subject(s)
Atherosclerosis , Foam Cells , Gastrointestinal Microbiome/immunology , Indican , Methylamines , Animals , Atherosclerosis/immunology , Atherosclerosis/metabolism , Atherosclerosis/microbiology , Atherosclerosis/pathology , Basic Helix-Loop-Helix Transcription Factors/immunology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Foam Cells/immunology , Foam Cells/metabolism , Foam Cells/pathology , Humans , Indican/immunology , Indican/metabolism , Inflammation/immunology , Inflammation/metabolism , Inflammation/microbiology , Inflammation/pathology , Methylamines/immunology , Methylamines/metabolism , Receptors, Aryl Hydrocarbon/immunology , Receptors, Aryl Hydrocarbon/metabolism
8.
Front Immunol ; 12: 775326, 2021.
Article in English | MEDLINE | ID: mdl-34975863

ABSTRACT

Mycobacterium tuberculosis infects primarily macrophages in the lungs. Infected macrophages are surrounded by other immune cells in well organised structures called granulomata. As part of the response to TB, a type of macrophage loaded with lipid droplets arises which we call Foam cell macrophages. They are macrophages filled with lipid laden droplets, which are synthesised in response to increased uptake of extracellular lipids, metabolic changes and infection itself. They share the appearance with atherosclerosis foam cells, but their lipid contents and roles are different. In fact, lipid droplets are immune and metabolic organelles with emerging roles in Tuberculosis. Here we discuss lipid droplet and foam cell formation, evidence regarding the inflammatory and immune properties of foam cells in TB, and address gaps in our knowledge to guide further research.


Subject(s)
Foam Cells/physiology , Lipid Droplets/physiology , Tuberculosis/immunology , Foam Cells/immunology , Humans , Triglycerides/biosynthesis
9.
Inflammation ; 44(2): 758-768, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33151398

ABSTRACT

Foam cell formation and inflammation in macrophages contribute to the development of atherosclerosis (AS). Clematichinenoside AR (AR) is a major active ingredient extracted from the traditional Chinese herb Clematis chinensis and has potent pharmacological effects on various diseases, including AS. However, little is known about the exact role and mechanism of AR in AS. RAW264.7 macrophages were exposed to oxidized low-density lipoprotein (ox-LDL) to induce AS in vitro. Cell viability was assessed by the CCK-8 assay. Foam cell formation was detected by Oil Red staining. Cholesterol levels were determined by corresponding commercial kits. The expression of inflammatory cytokines was detected by ELISA. Western blot and immunofluorescence assays were employed to detect the expression of corresponding genes. The results indicated that AR treatment inhibited the formation of foam cells and cholesterol accumulation but promoted cholesterol efflux by upregulating ABCA1/ABCG1 in ox-LDL-induced RAW264.7 macrophages. In addition, AR decreased the production of inflammatory cytokines by blunting the activation of the NLRP3 inflammasome and inducing autophagy. However, these effects of AR were weakened by the autophagy inhibitor bafilomycin A1 but were similar to those produced by the autophagy activator rapamycin. Collectively, our study provides novel insights into the beneficial effects of AR on promoting cholesterol efflux as well as inhibiting foam cell formation and inflammation by regulating autophagy, thus identifying AR as a promising therapeutic agent for the treatment of AS.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Atherosclerosis/drug therapy , Autophagy/drug effects , Foam Cells/drug effects , Macrophages/drug effects , Saponins/pharmacology , Triterpenes/pharmacology , Animals , Anti-Inflammatory Agents/therapeutic use , Atherosclerosis/chemically induced , Atherosclerosis/immunology , Atherosclerosis/metabolism , Autophagy/immunology , Biomarkers/metabolism , Blotting, Western , Cell Survival/drug effects , Cell Survival/immunology , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Foam Cells/immunology , Foam Cells/metabolism , Lipoproteins, LDL , Macrophages/immunology , Macrophages/metabolism , Mice , RAW 264.7 Cells , Saponins/therapeutic use , Triterpenes/therapeutic use
10.
Cardiovasc Res ; 117(9): 2054-2068, 2021 07 27.
Article in English | MEDLINE | ID: mdl-32991689

ABSTRACT

AIMS: Atherosclerosis, the leading cause of cardiovascular diseases, is driven by high blood cholesterol levels and chronic inflammation. Low-density lipoprotein receptors (LDLR) play a critical role in regulating blood cholesterol levels by binding to and clearing LDLs from the circulation. The disruption of the interaction between proprotein convertase subtilisin/kexin 9 (PCSK9) and LDLR reduces blood cholesterol levels. It is not well known whether other members of the LDLR superfamily may be targets of PCSK9. The aim of this work was to determine if LDLR-related protein 5 (LRP5) is a PCSK9 target and to study the role of PCSK9 and LRP5 in foam cell formation and lipid accumulation. METHODS AND RESULTS: Primary cultures of human inflammatory cells (monocytes and macrophages) were silenced for LRP5 or PCSK9 and challenged with LDLs. We first show that LRP5 is needed for macrophage lipid uptake since LRP5-silenced macrophages show less intracellular CE accumulation. In macrophages, internalization of LRP5-bound LDL is already highly evident after 5 h of LDL incubation and lasts up to 24 h; however, in the absence of both LRP5 and PCSK9, there is a strong reduction of CE accumulation indicating a role for both proteins in lipid uptake. Immunoprecipitation experiments show that LRP5 forms a complex with PCSK9 in lipid-loaded macrophages. Finally, PCSK9 participates in TLR4/NFkB signalling; a decreased TLR4 protein expression levels and a decreased nuclear translocation of NFκB were detected in PCSK9 silenced cells after lipid loading, indicating a downregulation of the TLR4/NFκB pathway. CONCLUSION: Our results show that both LRP5 and PCSK9 participate in lipid uptake in macrophages. In the absence of LRP5, there is a reduced release of PCSK9 indicating that LRP5 also participates in the mechanism of release of soluble PCSK9. Furthermore, PCSK9 up-regulates TLR4/NFκB favouring inflammation.


Subject(s)
Atherosclerosis/enzymology , Inflammation/enzymology , Lipid Metabolism , Low Density Lipoprotein Receptor-Related Protein-5/metabolism , Macrophages/enzymology , Monocytes/enzymology , Proprotein Convertase 9/metabolism , Atherosclerosis/genetics , Atherosclerosis/immunology , Biological Transport , Cells, Cultured , Cholesterol/metabolism , Foam Cells/enzymology , Foam Cells/immunology , Humans , Immunity, Innate , Inflammation/genetics , Inflammation/immunology , Lipoproteins, LDL/metabolism , Low Density Lipoprotein Receptor-Related Protein-5/genetics , Macrophages/immunology , Monocytes/immunology , NF-kappa B/metabolism , Proprotein Convertase 9/genetics , Signal Transduction , Sterol Regulatory Element Binding Protein 2/metabolism , Toll-Like Receptor 4/metabolism , Wnt3A Protein
11.
EBioMedicine ; 60: 102982, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32919101

ABSTRACT

BACKGROUND: An outbreak of E-cigarette or Vaping Product Use-Associated Lung Injury (EVALI) with significant morbidity and mortality was reported in 2019. While most patients with EVALI report vaping tetrahydrocannabinol (THC) oils contaminated with vitamin E acetate, a subset report only vaping with nicotine-containing electronic cigarettes (e-cigs). Whether or not e-cigs cause EVALI, the outbreak highlights the need for identifying long term health effects of e-cigs. EVALI pathology includes alveolar damage, pneumonitis and/or organizing pneumonia, often with lipid-laden macrophages (LLM). We assessed LLM in the lungs of healthy smokers, e-cig users, and never-smokers as a potential marker of e-cig toxicity and EVALI. METHODS: A cross-sectional study using bronchoscopy was conducted in healthy smokers, e-cig users, and never-smokers (n = 64). LLM, inflammatory cell counts, and cytokines were determined in bronchial alveolar fluids (BAL). E-cig users included both never-smokers and former light smokers. FINDINGS: High LLM was found in the lungs of almost all smokers and half of the e-cig users, but not those of never-smokers. LLM were not related to THC exposure or smoking history. LLM were significantly associated with inflammatory cytokines IL-4 and IL-10 in e-cig users, but not smoking-related cytokines. INTERPRETATION: This is the first report of lung LLM comparing apparently healthy smokers, e-cig users, and never-smokers. LLM are not a specific marker for EVALI given the frequent positivity in smokers; whether LLMs are a marker of lung inflammation in some e-cig users requires further study. FUNDING: The National Cancer Institute, the National Heart, Lung, and Blood Institute, the Food and Drug Administration Center for Tobacco Products, the National Center For Advancing Translational Sciences, and Pelotonia Intramural Research Funds.


Subject(s)
Electronic Nicotine Delivery Systems , Foam Cells/pathology , Lung Injury/epidemiology , Lung Injury/etiology , Vaping/adverse effects , Adult , Biomarkers , Cross-Sectional Studies , Cytokines/metabolism , Environmental Exposure/adverse effects , Female , Foam Cells/immunology , Foam Cells/metabolism , Healthy Volunteers , Humans , Immunohistochemistry , Inflammation Mediators , Lung Injury/metabolism , Male , Non-Smokers , Public Health Surveillance , Smoking , Young Adult
13.
Front Immunol ; 11: 1592, 2020.
Article in English | MEDLINE | ID: mdl-32849539

ABSTRACT

Cholesterol-laden, foam macrophages constitute the most characteristic component of human atherosclerotic plaques. Persistent uptake of oxLDLs results in accumulation of lipid bodies inside the cells and determines their phenotype and subsequent functions. In this work, we describe the phenotype of human monocyte-derived foam cells obtained by differentiation in the constant presence of oxLDLs for 30 days (prolonged-hMDFCs). Although neither the total cellular nor the cell surface expression of Toll-like receptors (TLR) was regulated by oxLDLs, the prolonged-hMDFCs changed dramatically their responsiveness to TLR ligands and inactivated bacteria. Using multiplex technology, we observed an acute decline in cytokine and chemokine production after surface and endosomal TLR stimulation with the exception of TLR2/6 triggering with agonists Pam2CSK4 and MALP-2. We also noted significant reduction of some surface receptors which can have accessory function in recognition of particulate antigens (CD47, CD81, and CD11b). In contrast, the prolonged-hMDFCs responded to inflammasome activation by LPS/nigericin with extensive, necrotic type cell death, which was partially independent of caspase-1. This pyroptosis-like cell death was aggravated by necrostatin-1 and rapamycin. These findings identify a potential contribution of mature foam cells to inflammatory status by increasing the immunogenic cell death burden. The observed cross-talk between foam cell death pathways may lead to recognition of a potential new marker for atherosclerosis disease severity. Overall, our study demonstrates that, in contrast to other cellular models of foam cells, the prolonged-hMDFCs acquire a functional phenotype which may help understanding the role of foam cells in the pathogenesis of atherosclerosis.


Subject(s)
Foam Cells/immunology , Foam Cells/metabolism , Host-Pathogen Interactions , Lipoproteins, LDL/metabolism , Pathogen-Associated Molecular Pattern Molecules/metabolism , Phenotype , Biomarkers , Cells, Cultured , Cytokines/metabolism , Host-Pathogen Interactions/immunology , Humans , Inflammasomes/metabolism , Inflammation Mediators/metabolism , Monocytes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pyroptosis/immunology
14.
Int Immunopharmacol ; 83: 106382, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32193098

ABSTRACT

Vascular inflammation plays a decisive role in the formation of foam cells and in the pathophysiology of atherosclerosis. However, the underlying mechanisms of these processes are not clearly understood. Macrophages engulf oxidized low-density lipoproteins (OxLDLs) via a scavenger receptor (SR), an event that mediates the elaboration of proinflammatory cytokines to initiate necrotic core formation in atherogenic plaques. In this study, we demonstrate that Nitric oxide synthase 1 (NOS1)-derived nitric oxide (NO) promotes OxLDL uptake and enhances the release of proinflammatory cytokines by macrophages. Conversely, we show that NOS1 inhibition by N(G)-nitro-L-arginine methyl ester (L-NAME) suppresses OxLDL uptake and proinflammatory cytokine expression. Current studies indicate that NOS1 plays a crucial role in vascular inflammation and in the progression of atherosclerosis. Therefore, interference with NOS1 enzymatic activity should serve as an effective strategy to reduce foam cell formation and limit the extent of atherosclerotic plaque expansion.


Subject(s)
Atherosclerosis/immunology , Foam Cells/immunology , Inflammation/immunology , Nitric Oxide Synthase Type I/metabolism , Animals , Cell Differentiation , Cells, Cultured , Disease Models, Animal , Disease Progression , Humans , Lipoproteins, LDL/metabolism , Macrophages/metabolism , Mice , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/metabolism , Nitric Oxide Synthase Type I/antagonists & inhibitors
15.
Cells ; 9(3)2020 03 01.
Article in English | MEDLINE | ID: mdl-32121535

ABSTRACT

Atherosclerosis is associated with acute cardiovascular conditions, such as ischemic heart disease, myocardial infarction, and stroke, and is the leading cause of morbidity and mortality worldwide. Our understanding of atherosclerosis and the processes triggering its initiation is constantly improving, and, during the last few decades, many pathological processes related to this disease have been investigated in detail. For example, atherosclerosis has been considered to be a chronic inflammation triggered by the injury of the arterial wall. However, recent works showed that atherogenesis is a more complex process involving not only the immune system, but also resident cells of the vessel wall, genetic factors, altered hemodynamics, and changes in lipid metabolism. In this review, we focus on foam cells that are crucial for atherosclerosis lesion formation. It has been demonstrated that the formation of foam cells is induced by modified low-density lipoprotein (LDL). The beneficial effects of the majority of therapeutic strategies with generalized action, such as the use of anti-inflammatory drugs or antioxidants, were not confirmed by clinical studies. However, the experimental therapies targeting certain stages of atherosclerosis, among which are lipid accumulation, were shown to be more effective. This emphasizes the relevance of future detailed investigation of atherogenesis and the importance of new therapies development.


Subject(s)
Atherosclerosis/immunology , Cardiovascular Diseases/immunology , Foam Cells/immunology , Cardiovascular Diseases/pathology , Humans , Lipoproteins, LDL/metabolism , Signal Transduction
16.
J Immunol Res ; 2020: 5284728, 2020.
Article in English | MEDLINE | ID: mdl-32149158

ABSTRACT

Atherosclerosis is a multifactorial chronic inflammatory arterial disease forming the pathological basis of many cardiovascular diseases such as coronary heart disease, heart failure, and stroke. Numerous studies have implicated inflammation as a key player in the initiation and progression of atherosclerosis. Galectin-3 (Gal-3) is a 30 kDa ß-galactose, highly conserved and widely distributed intracellularly and extracellularly. Gal-3 has been demonstrated in recent years to be a novel inflammatory factor participating in the process of intravascular inflammation, lipid endocytosis, macrophage activation, cellular proliferation, monocyte chemotaxis, and cell adhesion. This review focuses on the role of Gal-3 in atherosclerosis and the mechanism involved and several classical Gal-3 agonists and antagonists in the current studies.


Subject(s)
Atherosclerosis/etiology , Atherosclerosis/metabolism , Disease Susceptibility , Galectin 3/metabolism , Animals , Atherosclerosis/epidemiology , Atherosclerosis/pathology , Biomarkers , Blood Proteins , Disease Models, Animal , Endothelium/metabolism , Foam Cells/immunology , Foam Cells/metabolism , Foam Cells/pathology , Galectin 3/chemistry , Galectin 3/genetics , Galectins , Humans , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Mice , Oxidative Stress
17.
AIDS ; 34(4): 513-518, 2020 03 15.
Article in English | MEDLINE | ID: mdl-32108672

ABSTRACT

OBJECTIVE: People living with HIV have an increased risk of cardiovascular disease (CVD) despite effective antiretroviral therapy (ART). Monocytes play a key role in the early stages of atherosclerosis-driven CVD by forming lipid-laden foam cells within artery walls. HIV infection potentiates foam cell formation ex vivo, but the mechanisms contributing to this are not known. METHODS: We investigated the atherosclerosis-promoting potential of monocytes from 39 virologically suppressed men living with HIV (MLHIV) on ART and no evidence of CVD, and 25 HIV-uninfected controls of comparable age, sex, smoking status and CVD risk. RESULTS: Despite absence of clinical atherosclerosis in both MLHIV and uninfected cohorts (evidenced by a carotid intima-media thickness of 0.6 mm for both groups; P = 0.254), monocytes from MLHIV showed increased potential to form atherosclerosis-promoting foam cells compared with controls in an ex-vivo assay (36.6% vs. 27.6%, respectively, P = 0.003). Consistent with observations of persistent inflammation and immune/endothelial activation in ART-treated HIV infection, levels of soluble tumour necrosis factor receptor II, CXCL10 and soluble VCAM-1 were elevated in MLHIV (P ≤ 0.005 for all), but were not significantly associated with foam cell formation. Foam cell formation was associated with an impaired ability of monocytes to undergo reverse transmigration, and a reduced ability to efflux cholesterol ex vivo (P < 0.05 for both). Importantly, foam cell formation declined significantly with duration of viral suppression (P = 0.004). CONCLUSION: These findings highlight the persistence of HIV-related changes to the atherogenic potential of monocytes despite long-term viral suppression, and provide insights into mechanisms potentially driving increased CVD in ART-treated HIV infection.


Subject(s)
Atherosclerosis/etiology , Atherosclerosis/metabolism , HIV Infections/complications , HIV Infections/virology , Monocytes/immunology , Monocytes/metabolism , Adult , Aged , Antiretroviral Therapy, Highly Active , Atherosclerosis/pathology , Cardiovascular Diseases/etiology , Cardiovascular Diseases/metabolism , Carotid Intima-Media Thickness , Cross-Sectional Studies , Foam Cells/immunology , Foam Cells/metabolism , Foam Cells/pathology , HIV Infections/drug therapy , Humans , Inflammation/pathology , Linear Models , Male , Middle Aged , Risk Factors
18.
Front Immunol ; 11: 24, 2020.
Article in English | MEDLINE | ID: mdl-32082313

ABSTRACT

Atherosclerosis is characterized as a chronic inflammatory response to cholesterol deposition in arteries. Low-density lipoprotein (LDL), especially the oxidized form (ox-LDL), plays a crucial role in the occurrence and development of atherosclerosis by inducing endothelial cell (EC) dysfunction, attracting monocyte-derived macrophages, and promoting chronic inflammation. However, the mechanisms linking cholesterol accumulation with inflammation in macrophage foam cells are poorly understood. Long non-coding RNAs (lncRNAs) are a group of non-protein-coding RNAs longer than 200 nucleotides and are found to regulate the progress of atherosclerosis. Recently, many lncRNAs interfering with cholesterol deposition or inflammation were identified, which might help elucidate their underlying molecular mechanism or be used as novel therapeutic targets. In this review, we summarize and highlight the role of lncRNAs linking cholesterol (mainly ox-LDL) accumulation with inflammation in macrophages during the process of atherosclerosis.


Subject(s)
Atherosclerosis/immunology , Atherosclerosis/metabolism , Foam Cells/immunology , Lipoproteins, LDL/metabolism , RNA, Long Noncoding/metabolism , Animals , Anticholesteremic Agents/pharmacology , Anticholesteremic Agents/therapeutic use , Atherosclerosis/drug therapy , Cholesterol/metabolism , Disease Models, Animal , Humans , Inflammation/immunology , Inflammation/metabolism , Mice , Molecular Targeted Therapy/methods , RNA, Long Noncoding/antagonists & inhibitors
19.
Int J Mol Sci ; 21(3)2020 Jan 27.
Article in English | MEDLINE | ID: mdl-32012706

ABSTRACT

Excessive accumulation of lipid inclusions in the arterial wall cells (foam cell formation) caused by modified low-density lipoprotein (LDL) is the earliest and most noticeable manifestation of atherosclerosis. The mechanisms of foam cell formation are not fully understood and can involve altered lipid uptake, impaired lipid metabolism, or both. Recently, we have identified the top 10 master regulators that were involved in the accumulation of cholesterol in cultured macrophages induced by the incubation with modified LDL. It was found that most of the identified master regulators were related to the regulation of the inflammatory immune response, but not to lipid metabolism. A possible explanation for this unexpected result is a stimulation of the phagocytic activity of macrophages by modified LDL particle associates that have a relatively large size. In the current study, we investigated gene regulation in macrophages using transcriptome analysis to test the hypothesis that the primary event occurring upon the interaction of modified LDL and macrophages is the stimulation of phagocytosis, which subsequently triggers the pro-inflammatory immune response. We identified genes that were up- or downregulated following the exposure of cultured cells to modified LDL or latex beads (inert phagocytosis stimulators). Most of the identified master regulators were involved in the innate immune response, and some of them were encoding major pro-inflammatory proteins. The obtained results indicated that pro-inflammatory response to phagocytosis stimulation precedes the accumulation of intracellular lipids and possibly contributes to the formation of foam cells. In this way, the currently recognized hypothesis that the accumulation of lipids triggers the pro-inflammatory response was not confirmed. Comparative analysis of master regulators revealed similarities in the genetic regulation of the interaction of macrophages with naturally occurring LDL and desialylated LDL. Oxidized and desialylated LDL affected a different spectrum of genes than naturally occurring LDL. These observations suggest that desialylation is the most important modification of LDL occurring in vivo. Thus, modified LDL caused the gene regulation characteristic of the stimulation of phagocytosis. Additionally, the knock-down effect of five master regulators, such as IL15, EIF2AK3, F2RL1, TSPYL2, and ANXA1, on intracellular lipid accumulation was tested. We knocked down these genes in primary macrophages derived from human monocytes. The addition of atherogenic naturally occurring LDL caused a significant accumulation of cholesterol in the control cells. The knock-down of the EIF2AK3 and IL15 genes completely prevented cholesterol accumulation in cultured macrophages. The knock-down of the ANXA1 gene caused a further decrease in cholesterol content in cultured macrophages. At the same time, knock-down of F2RL1 and TSPYL2 did not cause an effect. The results obtained allowed us to explain in which way the inflammatory response and the accumulation of cholesterol are related confirming our hypothesis of atherogenesis development based on the following viewpoints: LDL particles undergo atherogenic modifications that, in turn, accompanied by the formation of self-associates; large LDL associates stimulate phagocytosis; as a result of phagocytosis stimulation, pro-inflammatory molecules are secreted; these molecules cause or at least contribute to the accumulation of intracellular cholesterol. Therefore, it became obvious that the primary event in this sequence is not the accumulation of cholesterol but an inflammatory response.


Subject(s)
Foam Cells/metabolism , Foam Cells/pathology , Lipoproteins, LDL/metabolism , Phagocytosis , Biomarkers , Foam Cells/immunology , Gene Expression Profiling , Gene Knockdown Techniques , Humans , Immunity, Innate , Lipid Metabolism , Macrophages/immunology , Macrophages/metabolism , Monocytes/immunology , Monocytes/metabolism , Oxidation-Reduction , Phagocytosis/genetics , Phagocytosis/immunology , Signal Transduction , Transcriptome
20.
Int Immunopharmacol ; 80: 106173, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31945610

ABSTRACT

Oxidative stress, inflammation, and foam cell formation in vascular smooth muscle cells (VSMCs) are considered to play crucial roles in the pathogenesis of atherosclerosis. Poria cocos polysaccharides (PCP) has been shown to possess anti-inflammatory, antitumor and anti-oxidative properties. In this study we explored the effects of PCP on ox-LDL-induced inflammation, oxidative stress and foam cell formation in VSMCs. PCP significantly attenuated ox-LDL-induced oxidative stress, as evidenced by the decreased reactive oxygen species (ROS) and MDA levels, and the increased SOD activity in VSMCs. PCP suppressed the induction effect of ox-LDL on inflammatory cytokines and inflammatory mediators. PCP also substantially inhibited VSMCs foam cell formation and intracellular lipids accumulation. Mechanistically, PCP suppressed ox-LDL-induced up-regulation of LOX-1, which is responsible for ox-LDL uptake. Western blotting suggested that PCP activated ERK1/2 signaling pathway, increased Nrf2 translocated from cytoplasm to nucleus and heme oxygenase-1 (HO-1) expression. Up-regulation of PCP on Nrf2/HO-1 signaling was reversed by pretreatment with ERK inhibitor PD98059, indicating the involvement of ERK in PCP activation of Nrf2/HO-1 signaling. In conclusion, these results demonstrated that PCP exerted its protection against oxidative stress and inflammation via the ERK/Nrf2/HO-1 signaling pathway and that PCP may be a promising candidate for the therapy of atherosclerosis.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Atherosclerosis/drug therapy , Fungal Polysaccharides/pharmacology , Myocytes, Smooth Muscle/drug effects , Signal Transduction/drug effects , Wolfiporia/chemistry , Atherosclerosis/immunology , Extracellular Signal-Regulated MAP Kinases/metabolism , Foam Cells/drug effects , Foam Cells/immunology , Heme Oxygenase-1/metabolism , Humans , Lipid Metabolism/drug effects , Lipid Metabolism/immunology , Lipoproteins, LDL/immunology , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/immunology , Myocytes, Smooth Muscle/metabolism , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Oxidative Stress/immunology , Reactive Oxygen Species/metabolism , Signal Transduction/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...