1
|
Giacaglia MB, Pires V, Santana MFM, Passarelli M. Unraveling the Pleiotropic Role of High-Density Lipoproteins (HDLs) in Autoimmune Rheumatic Diseases. Int J Rheumatol 2024; 2024:1896817. [PMID: 39574464 PMCID: PMC11581784 DOI: 10.1155/2024/1896817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 10/23/2024] [Indexed: 11/24/2024] Open
Abstract
Autoimmune rheumatic diseases (ARDs) exhibit an elevated incidence of cardiovascular disease (CVD). The elevation of inflammatory and immune stress accompanying ARDs contributes to atherosclerosis development and alterations in lipid metabolism and lipoprotein profile add to cardiovascular (CV) risk. The plasma concentration of high-density lipoprotein cholesterol (HDLc) is inversely related to CVD and serves as a discriminator of CV risk. However, this association is not unequivocal, and changes in HDL functionality appear to emerge as a better indicator of CV risk, albeit difficult to measure and monitor clinically. The modulation of HDLc itself can bring benefits in controlling autoimmunity and reducing ARD activity. Understanding HDL function and each peculiarity involved in ARDs enables to seek means to prevent ischemic outcomes associated with CVD, in the face of the residual CV risk persisting even with controlled disease activity and classic risk factors. By comprehending HDL's structural and functional nuances, it will be possible to develop more effective strategies to manage the evolution and outcomes of ARDs. It is also necessary to standardize diagnostic methods and establish different markers for each specific disease allowing the design of intervention strategies to restore HDL functionality, reduce residual CV, and prevent, alleviate, or even suppress ARD activity.
Collapse
Affiliation(s)
- Marcia B. Giacaglia
- Programa de Pós-Graduação em Medicina, Universidade Nove de Julho (UNINOVE) 01525-000, São Paulo, Brazil
| | - Vitória Pires
- Laboratório de Lípides (LIM10), Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo 01246-000, São Paulo, Brazil
| | - Monique F. M. Santana
- Laboratório de Lípides (LIM10), Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo 01246-000, São Paulo, Brazil
| | - Marisa Passarelli
- Programa de Pós-Graduação em Medicina, Universidade Nove de Julho (UNINOVE) 01525-000, São Paulo, Brazil
- Laboratório de Lípides (LIM10), Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo 01246-000, São Paulo, Brazil
| |
Collapse
|
2
|
Zhao X, Lian X, Xie J, Liu G. Accumulated cholesterol protects tumours from elevated lipid peroxidation in the microenvironment. Redox Biol 2023; 62:102678. [PMID: 36940607 PMCID: PMC10036943 DOI: 10.1016/j.redox.2023.102678] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 02/24/2023] [Accepted: 03/14/2023] [Indexed: 03/17/2023] Open
Abstract
Elevated lipid peroxidation (LPO), usually present in the tumour microenvironment (TME), is profoundly implicated in antitumour immunity and may be targeted for the development of new antitumour therapies. However, tumour cells may also rewire their metabolism to survive elevated LPO. Here, we report a novel and nonantioxidant mechanism by which tumour cells benefit from accumulated cholesterol to restrain LPO and ferroptosis, a nonapoptotic form of cell death characterized by accumulated LPO. Modulating cholesterol metabolism, especially LDLR-mediated cholesterol uptake, shifted the susceptibility of tumour cells to ferroptosis. Elevation of cellular cholesterol content specifically restrained LPO triggered by GSH-GPX4 inhibition or oxidizing factors in the TME. Furthermore, depletion of TME cholesterol by MβCD efficiently enhanced the antitumour efficacy of ferroptosis in a mouse xenograft model. Distinct from the antioxidant effect of its metabolic intermediates, the protective role of cholesterol was ascribed to its ability to decrease membrane fluidity and promote lipid raft formation, which affects the diffusion of LPO substrates. A correlation between LPO and lipid rafts was also found in tumour tissues from renal cancer patients. Together, our findings have identified a general and nonsacrificial mechanism by which cholesterol suppresses LPO, which can be exploited to enhance the efficacy of ferroptosis-based antitumour strategies.
Collapse
Affiliation(s)
- Xi Zhao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China; Department of Pharmaceutical Analysis, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Xinyu Lian
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China; Department of Pharmaceutical Analysis, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Jianlan Xie
- Department of Pathology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Guoquan Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China; Department of Pharmaceutical Analysis, School of Pharmaceutical Sciences, Peking University, Beijing, China.
| |
Collapse
|
3
|
Yuan X, Bhat OM, Zou Y, Li X, Zhang Y, Li PL. Endothelial Acid Sphingomyelinase Promotes NLRP3 Inflammasome and Neointima Formation During Hypercholesterolemia. J Lipid Res 2022; 63:100298. [PMID: 36252682 PMCID: PMC9672920 DOI: 10.1016/j.jlr.2022.100298] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 09/27/2022] [Accepted: 09/29/2022] [Indexed: 11/18/2022] Open
Abstract
The NOD-like receptor pyrin domain 3 (NLRP3) inflammasome is activated during atherogenesis, but how this occurs is unclear. Here, we explored the mechanisms activating and regulating NLRP3 inflammasomes via the acid sphingomyelinase (ASM)-ceramide signaling pathway. As a neointima formation model, partial left carotid ligations were performed on endothelial cell (EC)-specific ASM transgene mice (Smpd1trg/ECcre) and their control littermates (Smpd1trg/WT and WT/WT) fed on the Western diet (WD). We found neointima formation remarkably increased in Smpd1trg/ECcre mice over their control littermates. Next, we observed enhanced colocalization of NLRP3 versus adaptor protein ASC (the adaptor molecule apoptosis-associated speck-like protein containing a CARD) or caspase-1 in the carotid ECs of WD-treated Smpd1trg/ECcre mice but not in their control littermates. In addition, we used membrane raft (MR) marker flotillin-1 and found more aggregation of ASM and ceramide in the intima of Smpd1trg/ECcre mice than their control littermates. Moreover, we demonstrated by in situ dihydroethidium staining, carotid intimal superoxide levels were much higher in WD-treated Smpd1trg/ECcre mice than in their control littermates. Using ECs from Smpd1trg/ECcre and WT/WT mice, we showed ASM overexpression markedly enhanced 7-ketocholesterol (7-Ket)-induced increases in NLRP3 inflammasome formation, accompanied by enhanced caspase-1 activity and elevated interleukin-1β levels. These 7-Ket-induced increases were significantly attenuated by ASM inhibitor amitriptyline. Furthermore, we determined that increased MR clustering with NADPH oxidase subunits to produce superoxide contributes to 7-Ket-induced NLRP3 inflammasome activation via a thioredoxin-interacting protein-mediated controlling mechanism. We conclude that ceramide from ASM plays a critical role in NLRP3 inflammasome activation during hypercholesterolemia via MR redox signaling platforms to produce superoxide, which leads to TXNIP dissociation.
Collapse
Affiliation(s)
- Xinxu Yuan
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Owais M Bhat
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Yao Zou
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Xiang Li
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, USA
| | - Yang Zhang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, USA
| | - Pin-Lan Li
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
4
|
Chen Y, Li G, Bhat OM, Li X, Zhang Y, Li PL. Impairment of Ceramide-Mediated Endothelial Instant Membrane Resealing During Diabetes Mellitus. Front Physiol 2022; 13:910339. [PMID: 35874544 PMCID: PMC9298829 DOI: 10.3389/fphys.2022.910339] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 06/14/2022] [Indexed: 01/01/2023] Open
Abstract
Recent studies have indicated that instant cell membrane resealing (ICMR) controls the activation of NOD-like receptor pyrin domain containing 3 (Nlrp3) inflammasomes in endothelial cells, thereby initiating and promoting vascular inflammation. It remains unknown whether this impaired ICMR occurs under diabetic condition or hyperglycemia contributing to endothelial dysfunction leading to vascular inflammation, a hallmark of diabetic vascular injury. The present study aims to examine whether ICMR occurs during in control and diabetic mice and to explore related molecular mechanisms associated with acid sphingomyelinase (ASM)-mediated ceramide production. Using confocal microscopy, we demonstrated that mouse aortic endothelial cells (MAECs) exposed to high glucose levels exhibited much more retarded ICMR after laser-induced membrane injury, compared to that in control cells. The high glucose-induced impairment of membrane resealing in MAECs was prevented when these cells were pretreated with sphingomyelin or C24-ceramide. Mechanistically, high glucose treatment decreased association of membrane ceramide with annexin A5, an essential element of membrane repair machinery. Consistently, the association of ceramide with annexin A5 was significantly reduced in the coronary arterial endothelium of mice with streptozotocin-induced diabetes mellitus compared to that in non-diabetic control mice. Moreover, a marked reduction of the association of ceramide with annexin A5 was observed in coronary arterial endothelium of ASM knockout mice regardless of their diabetic status. Lastly, high glucose treatment or ASM gene deletion substantially impaired ICMR in coronary arterial endothelium of mice receiving membrane puncturing agents. Collectively, our data suggest that ceramide-mediated ICMR in vascular endothelial cells is impaired during diabetes mellitus due to dissociation of ceramide with annexin A5 and ASM play a critical role in this ICMR.
Collapse
Affiliation(s)
- Yang Chen
- School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Guangbi Li
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Owais M. Bhat
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Xiang Li
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, United States
| | - Yang Zhang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, United States
| | - Pin-Lan Li
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
5
|
Statins: Neurobiological underpinnings and mechanisms in mood disorders. Neurosci Biobehav Rev 2021; 128:693-708. [PMID: 34265321 DOI: 10.1016/j.neubiorev.2021.07.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 06/28/2021] [Accepted: 07/10/2021] [Indexed: 12/26/2022]
Abstract
Statins (3-hydroxy-3-methylglutaryl-coenzyme A reductase inhibitors) treat dyslipidaemia and cardiovascular disease by inhibiting cholesterol biosynthesis. They also have immunomodulatory and anti-inflammatory properties. Beyond cardiovascular disease, cholesterol and inflammation appear to be components of the pathogenesis and pathophysiology of neuropsychiatric disorders. Statins may therefore afford some therapeutic benefit in mood disorders. In this paper, we review the pathophysiology of mood disorders with a focus on pharmacologically relevant pathways, using major depressive disorder and bipolar disorder as exemplars. Statins are discussed in the context of these disorders, with particular focus on the putative mechanisms involved in their anti-inflammatory and immunomodulatory effects. Recent clinical data suggest that statins may have antidepressant properties, however given their interactions with many known biological pathways, it has not been fully elucidated which of these are the major determinants of clinical outcomes in mood disorders. Moreover, it remains unclear what the appropriate dose, or appropriate patient phenotype for adjunctive treatment may be. High quality randomised control trials in concert with complementary biological investigations are needed if the potential clinical effects of statins on mood disorders, as well as their biological correlates, are to be better understood.
Collapse
|
6
|
Orlowski S, Mourad JJ, Gallo A, Bruckert E. Coronaviruses, cholesterol and statins: Involvement and application for Covid-19. Biochimie 2021; 189:51-64. [PMID: 34153377 PMCID: PMC8213520 DOI: 10.1016/j.biochi.2021.06.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 06/01/2021] [Accepted: 06/14/2021] [Indexed: 12/17/2022]
Abstract
The infectious power of coronaviruses is dependent on cholesterol present in the membranes of their target cells. Indeed, the virus enters the infected cell either by fusion or by endocytosis, in both cases involving cholesterol-enriched membrane microdomains. These membrane domains can be disorganized in-vitro by various cholesterol-altering agents, including statins that inhibit cell cholesterol biosynthesis. As a consequence, numerous cell physiology processes, such as signaling cascades, can be compromised. Also, some examples of anti-bacterial and anti-viral effects of statins have been observed for infectious agents known to be cholesterol dependent. In-vivo, besides their widely-reported hypocholesterolemic effect, statins display various pleiotropic effects mediated, at least partially, by perturbation of membrane microdomains as a consequence of the alteration of endogenous cholesterol synthesis. It should thus be worth considering a high, but clinically well-tolerated, dose of statin to treat Covid-19 patients, in the early phase of infection, to inhibit virus entry into the target cells, in order to control the viral charge and hence avoid severe clinical complications. Based on its efficacy and favorable biodisposition, an option would be considering Atorvastatin, but randomized controlled clinical trials are required to test this hypothesis. This new therapeutic proposal takes benefit from being a drug repurposing, applied to a widely-used drug presenting a high efficiency-to-toxicity ratio. Additionally, this therapeutic strategy avoids any risk of drug resistance by viral mutation since it is host-targeted. Noteworthy, the same pharmacological approach could also be proposed to address different animal coronavirus endemic infections that are responsible for heavy economic losses.
Collapse
Affiliation(s)
- Stéphane Orlowski
- Institute for Integrative Biology of the Cell (I2BC), CNRS UMR 9198, and CEA / DRF / Institut des Sciences du Vivant Frédéric-Joliot / SB2SM, and Université Paris-Saclay, 91191, Gif-sur-Yvette, Cedex, France.
| | - Jean-Jacques Mourad
- Department of Internal Medicine and ESH Excellence Centre, Groupe Hospitalier Paris Saint-Joseph, Paris, France.
| | - Antonio Gallo
- Department of Endocrinology and Prevention of Cardiovascular Diseases, Institute of Cardiometabolism and Nutrition (ICAN), La Pitié-Salpêtrière Hospital, AP-HP, Paris, France.
| | - Eric Bruckert
- Department of Endocrinology and Prevention of Cardiovascular Diseases, Institute of Cardiometabolism and Nutrition (ICAN), La Pitié-Salpêtrière Hospital, AP-HP, Paris, France.
| |
Collapse
|
7
|
McGurk KA, Keavney BD, Nicolaou A. Circulating ceramides as biomarkers of cardiovascular disease: Evidence from phenotypic and genomic studies. Atherosclerosis 2021; 327:18-30. [PMID: 34004484 DOI: 10.1016/j.atherosclerosis.2021.04.021] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/25/2021] [Accepted: 04/30/2021] [Indexed: 12/20/2022]
Abstract
There is a need for new biomarkers of atherosclerotic cardiovascular disease (ACVD), the main cause of death globally. Ceramides, a class of potent bioactive lipid mediators, have signalling roles in apoptosis, cellular stress and inflammation. Recent studies have highlighted circulating ceramides as novel biomarkers of coronary artery disease, type-2 diabetes and insulin resistance. Ceramides are highly regulated by enzymatic reactions throughout the body in terms of their activity and metabolism, including production, degradation and transport. The genetic studies that have been completed to date on the main ceramide species found in circulation are described, highlighting the importance of DNA variants in genes involved in ceramide biosynthesis as key influencers of heritable, circulating ceramide levels. We also review studies of disease associations with ceramides and discuss mechanistic insights deriving from recent genomic studies. The signalling activities of ceramides in vascular inflammation and apoptosis, associations between circulating ceramides and coronary artery disease risk, type-2 diabetes and insulin resistance, and the potential importance of ceramides with regard to ACVD risk factors, such as blood pressure, lipoproteins and lifestyle factors, are also discussed.
Collapse
Affiliation(s)
- Kathryn A McGurk
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, UK; Laboratory for Lipidomics and Lipid Research, Division of Pharmacy and Optometry, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, UK
| | - Bernard D Keavney
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, UK; Manchester Heart Centre, Manchester University NHS Foundation Trust, UK
| | - Anna Nicolaou
- Laboratory for Lipidomics and Lipid Research, Division of Pharmacy and Optometry, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, UK; Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK.
| |
Collapse
|
8
|
McGurk KA, Williams SG, Guo H, Watkins H, Farrall M, Cordell HJ, Nicolaou A, Keavney BD. Heritability and family-based GWAS analyses of the N-acyl ethanolamine and ceramide plasma lipidome. Hum Mol Genet 2021; 30:500-513. [PMID: 33437986 PMCID: PMC8101358 DOI: 10.1093/hmg/ddab002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 11/25/2020] [Accepted: 12/23/2020] [Indexed: 12/11/2022] Open
Abstract
Signalling lipids of the N-acyl ethanolamine (NAE) and ceramide (CER) classes have emerged as potential biomarkers of cardiovascular disease (CVD). We sought to establish the heritability of plasma NAEs (including the endocannabinoid anandamide) and CERs, to identify common DNA variants influencing the circulating concentrations of the heritable lipids, and assess causality of these lipids in CVD using 2-sample Mendelian randomization (2SMR). Nine NAEs and 16 CERs were analyzed in plasma samples from 999 members of 196 British Caucasian families, using targeted ultra-performance liquid chromatography with tandem mass spectrometry. All lipids were significantly heritable (h2 = 36-62%). A missense variant (rs324420) in the gene encoding the enzyme fatty acid amide hydrolase (FAAH), which degrades NAEs, associated at genome-wide association study (GWAS) significance (P < 5 × 10-8) with four NAEs (DHEA, PEA, LEA and VEA). For CERs, rs680379 in the SPTLC3 gene, which encodes a subunit of the rate-limiting enzyme in CER biosynthesis, associated with a range of species (e.g. CER[N(24)S(19)]; P = 4.82 × 10-27). We observed three novel associations between SNPs at the CD83, SGPP1 and DEGS1 loci, and plasma CER traits (P < 5 × 10-8). 2SMR in the CARDIoGRAMplusC4D cohorts (60 801 cases; 123 504 controls) and in the DIAGRAM cohort (26 488 cases; 83 964 controls), using the genetic instruments from our family-based GWAS, did not reveal association between genetically determined differences in CER levels and CVD or diabetes. Two of the novel GWAS loci, SGPP1 and DEGS1, suggested a casual association between CERs and a range of haematological phenotypes, through 2SMR in the UK Biobank, INTERVAL and UKBiLEVE cohorts (n = 110 000-350 000).
Collapse
Affiliation(s)
- Kathryn A McGurk
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9NT, UK
- Laboratory for Lipidomics and Lipid Biology, Division of Pharmacy and Optometry, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PG, UK
| | - Simon G Williams
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9NT, UK
| | - Hui Guo
- Division of Population Health, Health Services Research & Primary Care, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PL, UK
| | - Hugh Watkins
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Martin Farrall
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Heather J Cordell
- Population Health Sciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE1 7RU, UK
| | - Anna Nicolaou
- Laboratory for Lipidomics and Lipid Biology, Division of Pharmacy and Optometry, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PG, UK
| | - Bernard D Keavney
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9NT, UK
- Manchester Heart Centre, Manchester University NHS Foundation Trust, Manchester M13 9WL, UK
| |
Collapse
|
9
|
Haimovitz-Friedman A, Mizrachi A, Jaimes EA. Manipulating Oxidative Stress Following Ionizing Radiation. JOURNAL OF CELLULAR SIGNALING 2020; 1:8-13. [PMID: 32550605 PMCID: PMC7299208 DOI: 10.33696/signaling.1.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
| | - Aviram Mizrachi
- Department of Otorhinolaryngology Head and Neck Surgery and Center for Translational Research in Head and Neck Cancer, Rabin Medical Center, Petah Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Edgar A Jaimes
- Department of Medicine, Renal Service, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
10
|
Zhang Y, Chen Y, Zhang Y, Li PL, Li X. Contribution of cathepsin B-dependent Nlrp3 inflammasome activation to nicotine-induced endothelial barrier dysfunction. Eur J Pharmacol 2019; 865:172795. [PMID: 31733211 PMCID: PMC6925381 DOI: 10.1016/j.ejphar.2019.172795] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 11/04/2019] [Accepted: 11/12/2019] [Indexed: 12/22/2022]
Abstract
Recent studies indicate that endothelial Nlrp3 inflammasome is critically involved in the development of cardiovascular complications. However, it remains unknown whether endothelial inflammasome is involved in endothelial barrier dysfunction associated with smoking. This study aims to investigate the role of endothelial Nlrp3 inflammasome in nicotine-induced disruption of inter-endothelial tight junctions and consequent endothelial barrier dysfunction. The confocal microscopic analysis demonstrated that mice treated with nicotine exhibited disrupted inter-endothelial tight junctions as shown by decreased ZO-1 and ZO-2 expression in the coronary arterial endothelium, whereas the decreases in ZO-1/2 were prevented by Nlrp3 gene deficiency. In cultured endothelial cells, nicotine caused Nlrp3 inflammasome complex formation and enhances the inflammasome activity as shown by increased cleavage of pro-caspase-1, and interleukin-1β (IL-1β) production. Further, nicotine disrupted tight junction and increased permeability in an endothelial cell monolayer, and this nicotine-induced effect was prevented by silencing of Nlrp3 gene, inhibition of caspase-1, or blockade of high mobility group box 1 (HMGB1). Nicotine increased endothelial cell lysosomal membrane permeability and triggered the lysosomal release of cathepsin B, whereas these events were prevented by pretreating cells with a lysosome stabilizing agent, dexamethasone. Collectively, our data suggest that nicotine enhances cathepsin B-dependent Nlrp3 inflammasome activation and the consequent production of a novel permeability factor HMGB1, which causes disruption of inter-endothelial tight junctions leading to endothelial hyperpermeability. Instigation of endothelial inflammasomes may serve as an important pathogenic mechanism contributing to the early onset of vasculopathy associated with smoking.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Pharmacology & Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, 23298, USA; Department of Pharmacological & Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, 77204, USA
| | - Yang Chen
- Department of Pharmacology & Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Youzhi Zhang
- Department of Pharmacology & Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, 23298, USA; School of Pharmacy, Hubei University of Science and Technology, Xianning, China
| | - Pin-Lan Li
- Department of Pharmacology & Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Xiang Li
- Department of Pharmacology & Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, 23298, USA; Department of Pharmacological & Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, 77204, USA.
| |
Collapse
|
11
|
NADPH oxidases and oxidase crosstalk in cardiovascular diseases: novel therapeutic targets. Nat Rev Cardiol 2019; 17:170-194. [PMID: 31591535 DOI: 10.1038/s41569-019-0260-8] [Citation(s) in RCA: 358] [Impact Index Per Article: 59.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/19/2019] [Indexed: 02/07/2023]
Abstract
Reactive oxygen species (ROS)-dependent production of ROS underlies sustained oxidative stress, which has been implicated in the pathogenesis of cardiovascular diseases such as hypertension, aortic aneurysm, hypercholesterolaemia, atherosclerosis, diabetic vascular complications, cardiac ischaemia-reperfusion injury, myocardial infarction, heart failure and cardiac arrhythmias. Interactions between different oxidases or oxidase systems have been intensively investigated for their roles in inducing sustained oxidative stress. In this Review, we discuss the latest data on the pathobiology of each oxidase component, the complex crosstalk between different oxidase components and the consequences of this crosstalk in mediating cardiovascular disease processes, focusing on the central role of particular NADPH oxidase (NOX) isoforms that are activated in specific cardiovascular diseases. An improved understanding of these mechanisms might facilitate the development of novel therapeutic agents targeting these oxidase systems and their interactions, which could be effective in the prevention and treatment of cardiovascular disorders.
Collapse
|
12
|
Ames PRJ, Bucci T, Merashli M, Amaral M, Arcaro A, Gentile F, Nourooz-Zadeh J, DelgadoAlves J. Oxidative/nitrative stress in the pathogenesis of systemic sclerosis: are antioxidants beneficial? Free Radic Res 2018; 52:1063-1082. [PMID: 30226391 DOI: 10.1080/10715762.2018.1525712] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Systemic sclerosis (SSc) is a multisystem autoimmune disease: characterised from the clinical side by progressive vasculopathy and fibrosis of the skin and different organs and from the biochemical side by fibroblast deregulation with excessive production of collagen and increased expression of nicotinamide adenine dinucleotide phosphate oxidase 4 (NOX4). The latter contributes to an overproduction of reactive oxygen species that through an autocrine loop maintains NOX4 in a state of activation. Reactive oxygen and nitrogen species are implicated in the origin and perpetuation of several clinical manifestations of SSc having vascular damage in common; attempts to dampen oxidative and nitrative stress through different agents with antioxidant properties have not translated into a sustained clinical benefit. Objective of this narrative review is to describe the origin and clinical implications of oxidative and nitrative stress in SSc, with particular focus on the central role of NOX4 and its interactions, to re-evaluate the antioxidant approaches so far used to limit disease progression, to appraise the complexity of antioxidant treatment and to touch on novel pathways elements of which may represent specific treatment targets in the not so distant future.
Collapse
Affiliation(s)
- Paul R J Ames
- a Immune Response and Vascular Disease Unit , CEDOC, Nova University , Lisboa , Portugal.,b Department of Haematology , Dumfries Royal Infirmary , Dumfries , UK
| | - Tommaso Bucci
- c Division of Allergy and Clinical Immunology, Department of Internal Medicine , University of Salerno , Baronissi , Italy
| | - Mira Merashli
- d Department of Rheumatology , American University of Beirut , Beirut , Lebanon
| | - Marta Amaral
- a Immune Response and Vascular Disease Unit , CEDOC, Nova University , Lisboa , Portugal
| | - Alessia Arcaro
- e Department of Medicine & Health Sciences , Universita' del Molise , Campobasso , Italy
| | - Fabrizio Gentile
- e Department of Medicine & Health Sciences , Universita' del Molise , Campobasso , Italy
| | - Jaffar Nourooz-Zadeh
- f Nephrology & Kidney Transplantation Research Center , Urmia University of Medical Sciences , Urmia , Iran
| | - Jose DelgadoAlves
- a Immune Response and Vascular Disease Unit , CEDOC, Nova University , Lisboa , Portugal.,g Immunomediated Systemic Diseases Unit, Medicine 4 , Hospital Fernando Fonseca , Amadora , Portugal
| |
Collapse
|
13
|
Imran M, Sergent O, Tête A, Gallais I, Chevanne M, Lagadic-Gossmann D, Podechard N. Membrane Remodeling as a Key Player of the Hepatotoxicity Induced by Co-Exposure to Benzo[a]pyrene and Ethanol of Obese Zebrafish Larvae. Biomolecules 2018; 8:biom8020026. [PMID: 29757947 PMCID: PMC6023014 DOI: 10.3390/biom8020026] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 05/04/2018] [Accepted: 05/04/2018] [Indexed: 12/11/2022] Open
Abstract
The rise in prevalence of non-alcoholic fatty liver disease (NAFLD) constitutes an important public health concern worldwide. Including obesity, numerous risk factors of NAFLD such as benzo[a]pyrene (B[a]P) and ethanol have been identified as modifying the physicochemical properties of the plasma membrane in vitro thus causing membrane remodeling—changes in membrane fluidity and lipid-raft characteristics. In this study, the possible involvement of membrane remodeling in the in vivo progression of steatosis to a steatohepatitis-like state upon co-exposure to B[a]P and ethanol was tested in obese zebrafish larvae. Larvae bearing steatosis as the result of a high-fat diet were exposed to ethanol and/or B[a]P for seven days at low concentrations coherent with human exposure in order to elicit hepatotoxicity. In this condition, the toxicant co-exposure raised global membrane order with higher lipid-raft clustering in the plasma membrane of liver cells, as evaluated by staining with the fluoroprobe di-4-ANEPPDHQ. Involvement of this membrane’s remodeling was finally explored by using the lipid-raft disruptor pravastatin that counteracted the effects of toxicant co-exposure both on membrane remodeling and toxicity. Overall, it can be concluded that B[a]P/ethanol co-exposure can induce in vivo hepatotoxicity via membrane remodeling which could be considered as a good target mechanism for developing combination therapy to deal with steatohepatitis.
Collapse
Affiliation(s)
- Muhammad Imran
- Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail)-UMR_S 1085, University of Rennes, F-35000 Rennes, France.
| | - Odile Sergent
- Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail)-UMR_S 1085, University of Rennes, F-35000 Rennes, France.
| | - Arnaud Tête
- Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail)-UMR_S 1085, University of Rennes, F-35000 Rennes, France.
| | - Isabelle Gallais
- Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail)-UMR_S 1085, University of Rennes, F-35000 Rennes, France.
| | - Martine Chevanne
- Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail)-UMR_S 1085, University of Rennes, F-35000 Rennes, France.
| | - Dominique Lagadic-Gossmann
- Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail)-UMR_S 1085, University of Rennes, F-35000 Rennes, France.
| | - Normand Podechard
- Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail)-UMR_S 1085, University of Rennes, F-35000 Rennes, France.
| |
Collapse
|
14
|
Dinoff A, Saleem M, Herrmann N, Mielke MM, Oh PI, Venkata SLV, Haughey NJ, Lanctôt KL. Plasma sphingolipids and depressive symptoms in coronary artery disease. Brain Behav 2017; 7:e00836. [PMID: 29201542 PMCID: PMC5698859 DOI: 10.1002/brb3.836] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 08/18/2017] [Accepted: 09/01/2017] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Depression is highly prevalent in individuals with coronary artery disease (CAD) and increases the risk of future cardiac events and mortality. Sphingolipids have been implicated in the pathophysiology of both CAD and depression. This study assessed the association between plasma sphingolipid concentrations and depressive symptoms in CAD subjects. METHODS Depressive symptoms were measured using the depression subscale of the self-reported Hospital Anxiety and Depression Scale (HADS). Sphingolipid concentrations were measured from fasting plasma samples using high-performance liquid chromatography-coupled electrospray ionization tandem mass spectrometry (LC/MS/MS). Linear regression models were used to assess associations between log-transformed concentrations of plasma sphingolipids and depressive symptoms. RESULTS A total of 111 CAD patients (mean (SD) age = 63.6 ± 6.4, 84.7% male) were included. In linear regression analyses, higher plasma concentrations of ceramides C16:0 (β = 0.204, p = .026) and C18:0 (β = 0.209, p = .023) and sphingomyelin SM18:1 (β = 0.210, p = .024) were significantly associated with higher HADS depression subscale score after adjusting for covariates. CONCLUSION Sphingolipids, in particular the ceramide species C16:0 and C18:0 and the sphingomyelin species SM18:1, may be implicated in the pathophysiology of depression in CAD. The association between plasma sphingolipid concentrations and depression should be further examined in CAD patients and in other populations.
Collapse
Affiliation(s)
- Adam Dinoff
- Neuropsychopharmacology Research Group Hurvitz Brain Sciences Program Sunnybrook Research Institute Toronto ON Canada.,Department of Pharmacology and Toxicology Faculty of Medicine University of Toronto Toronto ON Canada
| | - Mahwesh Saleem
- Neuropsychopharmacology Research Group Hurvitz Brain Sciences Program Sunnybrook Research Institute Toronto ON Canada.,Department of Pharmacology and Toxicology Faculty of Medicine University of Toronto Toronto ON Canada
| | - Nathan Herrmann
- Neuropsychopharmacology Research Group Hurvitz Brain Sciences Program Sunnybrook Research Institute Toronto ON Canada.,Department of Psychiatry Faculty of Medicine University of Toronto Toronto ON Canada
| | - Michelle M Mielke
- Department of Neurology Mayo Clinic Rochester MN USA.,Department of Health Sciences Research Mayo Clinic Rochester MN USA
| | - Paul I Oh
- Division of Clinical Pharmacology Sunnybrook Health Sciences Centre Toronto ON Canada.,Toronto Rehabilitation Institute Rumsey Centre Cardiac Rehabilitation Toronto ON Canada
| | | | - Norman J Haughey
- Department of Neurology Johns Hopkins University School of Medicine Baltimore MD USA.,Department of Psychiatry Johns Hopkins University School of Medicine Baltimore MD USA
| | - Krista L Lanctôt
- Neuropsychopharmacology Research Group Hurvitz Brain Sciences Program Sunnybrook Research Institute Toronto ON Canada.,Department of Pharmacology and Toxicology Faculty of Medicine University of Toronto Toronto ON Canada.,Department of Psychiatry Faculty of Medicine University of Toronto Toronto ON Canada.,Toronto Rehabilitation Institute Rumsey Centre Cardiac Rehabilitation Toronto ON Canada
| |
Collapse
|
15
|
Koka S, Xia M, Chen Y, Bhat OM, Yuan X, Boini KM, Li PL. Endothelial NLRP3 inflammasome activation and arterial neointima formation associated with acid sphingomyelinase during hypercholesterolemia. Redox Biol 2017; 13:336-344. [PMID: 28633109 PMCID: PMC5479959 DOI: 10.1016/j.redox.2017.06.004] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 06/12/2017] [Accepted: 06/13/2017] [Indexed: 12/17/2022] Open
Abstract
The NLRP3 inflammasome has been reported to be activated by atherogenic factors, whereby endothelial injury and consequent atherosclerotic lesions are triggered in the arterial wall. However, the mechanisms activating and regulating NLRP3 inflammasomes remain poorly understood. The present study tested whether acid sphingomyelinase (ASM) and ceramide associated membrane raft (MR) signaling platforms contribute to the activation of NLRP3 inflammasomes and atherosclerotic lesions during hypercholesterolemia. We found that 7-ketocholesterol (7-Keto) or cholesterol crystal (ChC) markedly increased the formation and activation of NLRP3 inflammasomes in mouse carotid arterial endothelial cells (CAECs), as shown by increased colocalization of NLRP3 with ASC or caspase-1, enhanced caspase-1 activity and elevated IL-1β levels, which were markedly attenuated by mouse Asm siRNA, ASM inhibitor- amitriptyline, and deletion of mouse Asm gene. In CAECs with NLRP3 inflammasome formation, membrane raft (MR) clustering with NADPH oxidase subunits was found remarkably increased as shown by CTXB (MR marker) and gp91phox aggregation indicating the formation of MR redox signaling platforms. This MR clustering was blocked by MR disruptor (MCD), ROS scavenger (Tempol) and TXNIP inhibitor (verapamil), accompanied by attenuation of 7-Keto or ChC-induced increase in caspase-1 activity. In animal experiments, Western diet fed mice with partially ligated left carotid artery (PLCA) were found to have significantly increased neointimal formation, which was associated with increased NLRP3 inflammasome formation and IL-1β production in the intima of Asm+/+ mice but not in Asm-/- mice. These results suggest that Asm gene and ceramide associated MR clustering are essential to endothelial inflammasome activation and dysfunction in the carotid arteries, ultimately determining the extent of atherosclerotic lesions.
Collapse
Affiliation(s)
- Saisudha Koka
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA 23298, USA; Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA.
| | - Min Xia
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Yang Chen
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Owais M Bhat
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Xinxu Yuan
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Krishna M Boini
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA
| | - Pin-Lan Li
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA 23298, USA.
| |
Collapse
|
16
|
Lv ZH, Phuong TA, Jin SJ, Li XX, Xu M. Protection by simvastatin on hyperglycemia-induced endothelial dysfunction through inhibiting NLRP3 inflammasomes. Oncotarget 2017; 8:91291-91305. [PMID: 29207644 PMCID: PMC5710924 DOI: 10.18632/oncotarget.20443] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2017] [Accepted: 07/25/2017] [Indexed: 12/24/2022] Open
Abstract
Recent studies have demonstrated that NLRP3 inflammasome complex acts as pivotal elements to initiate inflammatory responses and plays an important role in the dysfunction of cardiovascular complications. Meanwhile, simvastatin prevents vascular endothelial dysfunction from inflammasome invasion contributing to reduce cardiovascular risk. However, Whether or not the simvastatin improves vascular endothelial barrier function through inhibiting the activation of NLRP3 inflammasome pathway remains unknown. Here, we explored the role and mechanisms of simvastatin in the activation of NLRP3 inflammasome which are involved in vascular endothelial hyperpermeability causing by the disruption of tight junction protein ZO-1 and adherens junction protein VE-Cadherin, an early initiation of cardiovascular complication. Our results found that high glucose significantly induced the formation and activation of NLRP3 inflammasome through NADPH oxidase-dependent reactive oxygen species (ROS) formation, associated with vascular endothelial hyperpermeability causing by ZO-1 and VE-Cadherin disruption in the rat aortic endothelial cells (RAECs). Simvastatin treatment remarkably abolished vascular endothelial hyperpermeability and enhanced the protein expression of ZO-1 and VE-Cadherin through NLRP3 inflammasome. Mechanistically, the inhibitory role of simvastatin endothelial hyperpermeability is attributed to the decreased release of cytoplasmic high mobility group box protein-1 (HMGB1) derived from endothelial NLRP3 inflammasome activation. We further confirm the protective role of simvastatin on vascular leakage in the heart of diabetic rats injected with Evans blue dye, which was associated with HMGB1 release in the serum. Collectively, the mechanism of simvastatin treatment alleviating vascular endothelial permeability dysfunction may be through inhibiting the NLRP3 inflammasome-dependent HMGB1 release in RAECs.
Collapse
Affiliation(s)
- Zhen-Huan Lv
- Department of Clinical Pharmacy, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Trinh Anh Phuong
- Department of Clinical Pharmacy, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Shi-Jie Jin
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou 311400, China
| | - Xiao-Xue Li
- Department of Pathology, Medical School of Southeast University, Nanjing 210009, China
| | - Ming Xu
- Department of Clinical Pharmacy, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
17
|
Chubinskiy-Nadezhdin VI, Negulyaev YA, Morachevskaya EA. Simvastatin induced actin cytoskeleton disassembly in normal and transformed fibroblasts without affecting lipid raft integrity. Cell Biol Int 2017; 41:1020-1029. [PMID: 28656734 DOI: 10.1002/cbin.10812] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 06/24/2017] [Indexed: 02/02/2023]
Abstract
Statins are the most commonly prescribed agents used to modulate cholesterol levels in course of hypercholesterolemia treatment because of their relative tolerability and LDL-C lowering effect. Recently, there are emerging interests in the perspectives of statin drugs as anticancer agents based on preclinical evidence of their antiproliferative, proapoptotic, and anti-invasive properties. Functional impact of statin application on transformed cells still remains obscure that requires systematic study on adequate cellular models to provide correct comparison with their non-transformed counterparts. Cholesterol is the major lipid component of mammalian cells and it plays a crucial role in organization, lateral heterogeneity, and dynamics of plasma membrane as well as in membrane-cytoskeleton interrelations. To date, it is uncertain whether cellular effects of statins involve lipid-dependent alteration of plasma membrane. Here, the effects of simvastatin on lipid rafts, F-actin network and cellular viability were determined in comparative experiments on transformed fibroblasts and their non-transformed counterpart. GM1 lipid raft marker staining indicated no change of lipid raft integrity after short- or long-term simvastatin treatments. In the same time, simvastatin induced cytoskeleton rearrangement including partial F-actin disruption in cholesterol- and lipid raft-independent manner. Simvastatin dose-dependently affected viability of BALB/3T3 and 3T3B-SV40 cell lines: transformed fibroblasts were noticeably more sensitive to simvastatin comparing to non-transformed cells.
Collapse
Affiliation(s)
| | - Yuri A Negulyaev
- Institute of Cytology RAS, 4 Tikhoretsky Ave., St. Petersburg, 194064, Russia.,Department of Medical Physics, Peter the Great St. Petersburg Polytechnic University, 29, Polytechnicheskaya st., St.Petersburg, 195251, Russia
| | | |
Collapse
|
18
|
Niaudet C, Bonnaud S, Guillonneau M, Gouard S, Gaugler MH, Dutoit S, Ripoche N, Dubois N, Trichet V, Corre I, Paris F. Plasma membrane reorganization links acid sphingomyelinase/ceramide to p38 MAPK pathways in endothelial cells apoptosis. Cell Signal 2017; 33:10-21. [PMID: 28179144 DOI: 10.1016/j.cellsig.2017.02.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 02/01/2017] [Accepted: 02/01/2017] [Indexed: 12/11/2022]
Abstract
The p38 MAPK signaling pathway is essential in the cellular response to stress stimuli, in particular in the endothelial cells that are major target of external stress. The importance of the bioactive sphingolipid ceramide generated by acid sphingomyelinase is also firmly established in stress-induced endothelial apoptotic cell death. Despite a suggested link between the p38 MAPK and ceramide pathways, the exact molecular events of this connection remain elusive. In the present study, by using two different activators of p38 MAPK, namely anisomycin and ionizing radiation, we depicted how ceramide generated by acid sphingomyelinase was involved in p38 MAPK-dependent apoptosis of endothelial cells. We first proved that both anisomycin and ionizing radiation conducted to apoptosis through activation of p38 MAPK in human microvascular endothelial cells HMEC-1. We then found that both treatments induced activation of acid sphingomyelinase and the generation of ceramide. This step was required for p38 MAPK activation and apoptosis. We finally showed that irradiation, as well as treatment with exogenous C16-ceramide or bacterial sphingomyelinase, induced in endothelial cells a deep reorganization of the plasma membrane with formation of large lipid platforms at the cell surface, leading to p38 MAPK activation and apoptosis in endothelial cells. Altogether, our results proved that the plasma membrane reorganization leading to ceramide production is essential for stress-induced activation of p38 MAPK and apoptosis in endothelial cells and established the link between the acid sphingomyelinase/ceramide and p38 MAPK pathways.
Collapse
Affiliation(s)
- Colin Niaudet
- CRCNA, INSERM, CNRS, Université de Nantes, Nantes, France
| | | | | | | | - Marie-Hélène Gaugler
- CRCNA, INSERM, CNRS, Université de Nantes, Nantes, France; IRSN, Fontenay-aux-roses, France
| | - Soizic Dutoit
- CRCNA, INSERM, CNRS, Université de Nantes, Nantes, France
| | | | - Nolwenn Dubois
- CRCNA, INSERM, CNRS, Université de Nantes, Nantes, France
| | | | - Isabelle Corre
- CRCNA, INSERM, CNRS, Université de Nantes, Nantes, France
| | - François Paris
- CRCNA, INSERM, CNRS, Université de Nantes, Nantes, France; Laboratoire de Biologie du Cancer et Théranostique, ICO, Saint-Herblain, France.
| |
Collapse
|
19
|
Amiya E. Interaction of hyperlipidemia and reactive oxygen species: Insights from the lipid-raft platform. World J Cardiol 2016; 8:689-694. [PMID: 28070236 PMCID: PMC5183968 DOI: 10.4330/wjc.v8.i12.689] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 09/07/2016] [Accepted: 10/09/2016] [Indexed: 02/06/2023] Open
Abstract
Reactive oxygen species (ROS) and oxidative stress are closely associated with the development of atherosclerosis, and the most important regulator of ROS production in endothelial cells is NADPH oxidase. Activation of NADPH oxidase requires the assembly of multiple subunits into lipid rafts, which include specific lipid components, including free cholesterol and specific proteins. Disorders of lipid metabolism such as hyperlipidemia affect the cellular lipid components included in rafts, resulting in modification of cellular reactions that produce ROS. In the similar manner, several pathways associating ROS production are affected by the presence of lipid disorder through raft compartments. In this manuscript, we review the pathophysiological implications of hyperlipidemia and lipid rafts in the production of ROS.
Collapse
|
20
|
Podechard N, Chevanne M, Fernier M, Tête A, Collin A, Cassio D, Kah O, Lagadic-Gossmann D, Sergent O. Zebrafish larva as a reliable model for in vivo assessment of membrane remodeling involvement in the hepatotoxicity of chemical agents. J Appl Toxicol 2016; 37:732-746. [PMID: 27896850 DOI: 10.1002/jat.3421] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 10/20/2016] [Accepted: 10/28/2016] [Indexed: 12/13/2022]
Abstract
The easy-to-use in vivo model, zebrafish larva, is being increasingly used to screen chemical-induced hepatotoxicity, with a good predictivity for various mechanisms of liver injury. However, nothing is known about its applicability in exploring the mechanism called membrane remodeling, depicted as changes in membrane fluidity or lipid raft properties. The aim of this study was, therefore, to substantiate the zebrafish larva as a suitable in vivo model in this context. Ethanol was chosen as a prototype toxicant because it is largely described, both in hepatocyte cultures and in rodents, as capable of inducing a membrane remodeling leading to hepatocyte death and liver injury. The zebrafish larva model was demonstrated to be fully relevant as membrane remodeling was maintained even after a 1-week exposure without any adaptation as usually reported in rodents and hepatocyte cultures. It was also proven to exhibit a high sensitivity as it discriminated various levels of cytotoxicity depending on the extent of changes in membrane remodeling. In this context, its sensitivity appeared higher than that of WIF-B9 hepatic cells, which is suited for analyzing this kind of hepatotoxicity. Finally, the protection afforded by a membrane stabilizer, ursodeoxycholic acid (UDCA), or by a lipid raft disrupter, pravastatin, definitely validated zebrafish larva as a reliable model to quickly assess membrane remodeling involvement in chemical-induced hepatotoxicity. In conclusion, this model, compatible with a high throughput screening, might be adapted to seek hepatotoxicants via membrane remodeling, and also drugs targeting membrane features to propose new preventive or therapeutic strategies in chemical-induced liver diseases. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Normand Podechard
- UMR Inserm 1085, IRSET, UFR des Sciences Pharmaceutiques et Biologiques, bâtiment 5, 35043, Rennes Cédex, France.,Biosit UMS3480, Université de Rennes 1, 35043, Rennes Cédex, France
| | - Martine Chevanne
- UMR Inserm 1085, IRSET, UFR des Sciences Pharmaceutiques et Biologiques, bâtiment 5, 35043, Rennes Cédex, France.,Biosit UMS3480, Université de Rennes 1, 35043, Rennes Cédex, France
| | - Morgane Fernier
- UMR Inserm 1085, IRSET, UFR des Sciences Pharmaceutiques et Biologiques, bâtiment 5, 35043, Rennes Cédex, France.,Biosit UMS3480, Université de Rennes 1, 35043, Rennes Cédex, France
| | - Arnaud Tête
- UMR Inserm 1085, IRSET, UFR des Sciences Pharmaceutiques et Biologiques, bâtiment 5, 35043, Rennes Cédex, France.,Biosit UMS3480, Université de Rennes 1, 35043, Rennes Cédex, France
| | - Aurore Collin
- UMR Inserm 1085, IRSET, UFR des Sciences Pharmaceutiques et Biologiques, bâtiment 5, 35043, Rennes Cédex, France.,Biosit UMS3480, Université de Rennes 1, 35043, Rennes Cédex, France
| | - Doris Cassio
- Inserm, UMR-S 757; Orsay, France; Université Paris-Sud, Orsay, France
| | - Olivier Kah
- Biosit UMS3480, Université de Rennes 1, 35043, Rennes Cédex, France.,UMR Inserm 1085, IRSET, Université de Rennes 1, bâtiment 9, 35000, Rennes, France
| | - Dominique Lagadic-Gossmann
- UMR Inserm 1085, IRSET, UFR des Sciences Pharmaceutiques et Biologiques, bâtiment 5, 35043, Rennes Cédex, France.,Biosit UMS3480, Université de Rennes 1, 35043, Rennes Cédex, France
| | - Odile Sergent
- UMR Inserm 1085, IRSET, UFR des Sciences Pharmaceutiques et Biologiques, bâtiment 5, 35043, Rennes Cédex, France.,Biosit UMS3480, Université de Rennes 1, 35043, Rennes Cédex, France
| |
Collapse
|
21
|
Song J, Ping LY, Duong DM, Gao XY, He CY, Wei L, Wu JZ. Native low density lipoprotein promotes lipid raft formation in macrophages. Mol Med Rep 2016; 13:2087-93. [PMID: 26781977 PMCID: PMC4768993 DOI: 10.3892/mmr.2016.4781] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 12/07/2015] [Indexed: 01/09/2023] Open
Abstract
Oxidized low-density lipoprotein (LDL) has an important role in atherogenesis; however, the mechanisms underlying cell-mediated LDL oxidation remain to be elucidated. The present study investigated whether native-LDL induced lipid raft formation, in order to gain further insight into LDL oxidation. Confocal microscopic analysis revealed that lipid rafts were aggregated or clustered in the membrane, which were colocalized with myeloperoxidase (MPO) upon native LDL stimulation; however, in the presence of methyl-β-cyclodextrin (MβCD), LDL-stimulated aggregation, translocation, and colocalization of lipid rafts components was abolished.. In addition, lipid raft disruptors MβCD and filipin decreased malondialdehyde expression levels. Density gradient centrifugation coupled to label-free quantitative proteomic analysis identified 1,449 individual proteins, of which 203 were significantly upregulated following native-LDL stimulation. Functional classification of the proteins identified in the lipid rafts revealed that the expression levels of translocation proteins were upregulated. In conclusion, the results of the present study indicated that native-LDL induced lipid raft clustering in macrophages, and the expression levels of several proteins were altered in the stimulated macrophages, which provided novel insights into the mechanism underlying LDL oxidation.
Collapse
Affiliation(s)
- Jian Song
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Ling-Yan Ping
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Duc M Duong
- Department of Biochemistry, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Xiao-Yan Gao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Chun-Yan He
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Lei Wei
- Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Jun-Zhu Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei 430071, P.R. China
| |
Collapse
|
22
|
Wang HJ, Park JY, Kwon O, Choe EY, Kim CH, Hur KY, Lee MS, Yun M, Cha BS, Kim YB, Lee H, Kang ES. Chronic HMGCR/HMG-CoA reductase inhibitor treatment contributes to dysglycemia by upregulating hepatic gluconeogenesis through autophagy induction. Autophagy 2015; 11:2089-2101. [PMID: 26389569 DOI: 10.1080/15548627.2015.1091139] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Statins (HMGCR/HMG-CoA reductase [3-hydroxy-3-methylglutaryl-CoA reductase] inhibitors) are widely used to lower blood cholesterol levels but have been shown to increase the risk of type 2 diabetes mellitus. However, the molecular mechanism underlying diabetogenic effects remains to be elucidated. Here we show that statins significantly increase the expression of key gluconeogenic enzymes (such as G6PC [glucose-6-phosphatase] and PCK1 (phosphoenolpyruvate carboxykinase 1 [soluble]) in vitro and in vivo and promote hepatic glucose output. Statin treatment activates autophagic flux in HepG2 cells. Acute suppression of autophagy with lysosome inhibitors in statin treated HepG2 cells reduced gluconeogenic enzymes expression and glucose output. Importantly, the ability of statins to increase gluconeogenesis was impaired when ATG7 was deficient and BECN1 was absent, suggesting that autophagy plays a critical role in the diabetogenic effects of statins. Moreover autophagic vacuoles and gluconeogenic genes expression in the liver of diet-induced obese mice were increased by statins, ultimately leading to elevated hepatic glucose production, hyperglycemia, and insulin resistance. Together, these data demonstrate that chronic statin therapy results in insulin resistance through the activation of hepatic gluconeogenesis, which is tightly coupled to hepatic autophagy. These data further contribute to a better understanding of the diabetogenic effects of stains in the context of insulin resistance.
Collapse
Affiliation(s)
- Hye Jin Wang
- a Division of Endocrinology and Metabolism ; Department of Internal Medicine ; Yonsei University College of Medicine ; Seoul , Korea.,b Institute of Endocrine Research; Yonsei University College of Medicine ; Seoul , Korea.,c Brain Korea 21 PLUS Project for Medical Science; Yonsei University College of Medicine ; Seoul , Korea
| | - Jae Yeo Park
- d Department of Clinical Nursing Science ; Yonsei University College of Nursing ; Seoul , Korea.,e Nursing Policy and Research Institute; Biobehavioral Research Center; Yonsei University ; Seoul , Korea
| | - Obin Kwon
- c Brain Korea 21 PLUS Project for Medical Science; Yonsei University College of Medicine ; Seoul , Korea.,f Department of Pharmacology ; Yonsei University College of Medicine ; Seoul , Korea
| | - Eun Yeong Choe
- a Division of Endocrinology and Metabolism ; Department of Internal Medicine ; Yonsei University College of Medicine ; Seoul , Korea.,b Institute of Endocrine Research; Yonsei University College of Medicine ; Seoul , Korea
| | - Chul Hoon Kim
- c Brain Korea 21 PLUS Project for Medical Science; Yonsei University College of Medicine ; Seoul , Korea.,f Department of Pharmacology ; Yonsei University College of Medicine ; Seoul , Korea
| | - Kyu Yeon Hur
- g Department of Medicine ; Samsung Medical Center; Sungkyunkwan University School of Medicine ; Seoul , Korea
| | - Myung-Shik Lee
- g Department of Medicine ; Samsung Medical Center; Sungkyunkwan University School of Medicine ; Seoul , Korea.,h Samsung Advanced Institute for Health Sciences and Technology; Sungkyunkwan University School of Medicine ; Seoul , Korea
| | - Mijin Yun
- i Department of Nuclear Medicine ; Yonsei University College of Medicine ; Seoul , Korea
| | - Bong Soo Cha
- a Division of Endocrinology and Metabolism ; Department of Internal Medicine ; Yonsei University College of Medicine ; Seoul , Korea.,b Institute of Endocrine Research; Yonsei University College of Medicine ; Seoul , Korea.,c Brain Korea 21 PLUS Project for Medical Science; Yonsei University College of Medicine ; Seoul , Korea
| | - Young-Bum Kim
- j Division of Endocrinology , Diabetes, and Metabolism ; Department of Medicine ; Beth Israel Deaconess Medical Center and Harvard Medical School ; Boston , MA USA
| | - Hyangkyu Lee
- d Department of Clinical Nursing Science ; Yonsei University College of Nursing ; Seoul , Korea.,e Nursing Policy and Research Institute; Biobehavioral Research Center; Yonsei University ; Seoul , Korea
| | - Eun Seok Kang
- a Division of Endocrinology and Metabolism ; Department of Internal Medicine ; Yonsei University College of Medicine ; Seoul , Korea.,b Institute of Endocrine Research; Yonsei University College of Medicine ; Seoul , Korea.,c Brain Korea 21 PLUS Project for Medical Science; Yonsei University College of Medicine ; Seoul , Korea
| |
Collapse
|
23
|
Chen Y, Pitzer AL, Li X, Li PL, Wang L, Zhang Y. Instigation of endothelial Nlrp3 inflammasome by adipokine visfatin promotes inter-endothelial junction disruption: role of HMGB1. J Cell Mol Med 2015; 19:2715-27. [PMID: 26293846 PMCID: PMC4687695 DOI: 10.1111/jcmm.12657] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 06/23/2015] [Indexed: 01/24/2023] Open
Abstract
Recent studies have indicated that the inflammasome plays a critical role in the pathogenesis of vascular diseases. However, the pathological relevance of this inflammasome activation, particularly in vascular cells, remains largely unknown. Here, we investigated the role of endothelial (Nucleotide‐binding Oligomerization Domain) NOD‐like receptor family pyrin domain containing three (Nlrp3) inflammasomes in modulating inter‐endothelial junction proteins, which are associated with endothelial barrier dysfunction, an early onset of obesity‐associated endothelial injury. Our findings demonstrate that the activation of Nlrp3 inflammasome by visfatin markedly decreased the expression of inter‐endothelial junction proteins including tight junction proteins ZO‐1, ZO‐2 and occludin, and adherens junction protein VE‐cadherin in cultured mouse vascular endothelial (VE) cell monolayers. Such visfatin‐induced down‐regulation of junction proteins in endothelial cells was attributed to high mobility group box protein 1 (HMGB1) release derived from endothelial inflammasome‐dependent caspase‐1 activity. Similarly, in the coronary arteries of wild‐type mice, high‐fat diet (HFD) treatment caused a down‐regulation of inter‐endothelial junction proteins ZO‐1, ZO‐2, occludin and VE‐cadherin, which was accompanied with enhanced inflammasome activation and HMGB1 expression in the endothelium as well as transmigration of CD43+ T cells into the coronary arterial wall. In contrast, all these HFD‐induced alterations in coronary arteries were prevented in mice with Nlrp3 gene deletion. Taken together, these data strongly suggest that the activation of endothelial Nlrp3 inflammasomes as a result of the increased actions of injurious adipokines such as visfatin produces HMGB1, which act in paracrine or autocrine fashion to disrupt inter‐endothelial junctions and increase paracellular permeability of the endothelium contributing to the early onset of endothelial injury during metabolic disorders such as obesity or high‐fat/cholesterol diet.
Collapse
Affiliation(s)
- Yang Chen
- Department of Pharmacology & Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Ashley L Pitzer
- Department of Pharmacology & Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Xiang Li
- Department of Pharmacology & Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA.,Department of Pharmacological & Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, USA
| | - Pin-Lan Li
- Department of Pharmacology & Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Lei Wang
- Department of Pharmacology & Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Yang Zhang
- Department of Pharmacology & Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA.,Department of Pharmacological & Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, USA
| |
Collapse
|
24
|
Ong WY, Herr DR, Farooqui T, Ling EA, Farooqui AA. Role of sphingomyelinases in neurological disorders. Expert Opin Ther Targets 2015; 19:1725-42. [DOI: 10.1517/14728222.2015.1071794] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
25
|
Abstract
Endothelial progenitor cells (EPCs) play a critical role in maintenance of the endothelial integrity and vascular homeostasis, as well as in neovascularization. Dysfunctional EPCs are believed to contribute to the endothelial dysfunction and are closely related to the development of various cardiovascular diseases, such as hypertension, hyperlipidemia, and stroke. However, the underlying mechanisms of EPC dysfunction are complicated and remain largely elusive. Recent studies have demonstrated that reactive oxygen species (ROS) are key factors that involve in modulation of stem and progenitor cell function under various physiologic and pathologic conditions. It has been shown that NADPH oxidase (NOX)-derived ROS are the major sources of ROS in cardiovascular system. Accumulating evidence suggests that NOX-mediated oxidative stress can modulate EPC bioactivities, such as mobilization, migration, and neovascularization, and that inhibition of NOX has been shown to improve EPC functions. This review summarized recent progress in the studies on the correlation between NOX-mediated EPC dysfunction and cardiovascular diseases.
Collapse
|
26
|
Zhang Y, Li X, Pitzer AL, Chen Y, Wang L, Li PL. Coronary endothelial dysfunction induced by nucleotide oligomerization domain-like receptor protein with pyrin domain containing 3 inflammasome activation during hypercholesterolemia: beyond inflammation. Antioxid Redox Signal 2015; 22:1084-96. [PMID: 25739025 PMCID: PMC4403230 DOI: 10.1089/ars.2014.5978] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
AIMS This study hypothesized that activation of endothelial nucleotide oligomerization domain-like receptor protein with pyrin domain containing 3 (Nlrp3) inflammasomes directly produces endothelial dysfunction during hypercholesterolemia, which is distinct from its canonical roles in inflammation. RESULTS Acute hypercholesterolemia in mice was induced by intraperitoneal administration of poloxamer 407 (0.5 g/kg) for 24 h. Endothelial dysfunction was assessed by evaluating endothelium-dependent vasodilation in isolated, perfused, and pressurized coronary arteries in response to bradykinin (10(-10)-10(-6) M) and acetylcholine (10(-9)-10(-5) M). Impaired endothelium-dependent vasodilation was observed in Nlrp3(+/+) mice with acute hypercholesterolemia, which was markedly ameliorated in Nlrp3(-/-) mice. Treatment of mice with inhibitors for caspase-1 or high mobility group box 1 (HMGB1) significantly restored endothelium-dependent vasodilation in Nlrp3(+/+) mice with acute hypercholesterolemia. Confocal microscopic analysis demonstrated that hypercholesterolemia markedly increased caspase-1 activity and HMGB1 expression in coronary arterial endothelium of Nlrp3(+/+) mice, which was absent in Nlrp3-deficient mice. Further, recombinant HMGB1 directly induced endothelial dysfunction in normal Nlrp3(+/+) coronary arteries. In vitro, Nlrp3 inflammasome formation and its activity were instigated in cultured endothelial cells by cholesterol crystal, a danger factor associated with hypercholesterolemia. Moreover, cholesterol crystals directly induced endothelial dysfunction in coronary arteries from Nlrp3(+/+) mice, which was attenuated in Nlrp3(-/-) arteries. Such cholesterol crystal-induced impairment was associated with enhanced superoxide production, downregulation of endothelial nitric oxide synthase activity, and pyroptosis. INNOVATION AND CONCLUSION Our data provide the first evidence that activation of endothelial Nlrp3 inflammasome directly impairs endothelial function beyond its canonical inflammatory actions. This novel non-canonical action of Nlrp3 inflammasomes may initiate or exacerbate vascular injury during hypercholesterolemia.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University , Richmond, Virginia
| | | | | | | | | | | |
Collapse
|
27
|
Drummond GR, Sobey CG. Endothelial NADPH oxidases: which NOX to target in vascular disease? Trends Endocrinol Metab 2014; 25:452-63. [PMID: 25066192 DOI: 10.1016/j.tem.2014.06.012] [Citation(s) in RCA: 248] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Revised: 06/29/2014] [Accepted: 06/30/2014] [Indexed: 02/03/2023]
Abstract
NADPH oxidases (NOXs) are reactive oxygen species (ROS)-generating enzymes implicated in the pathophysiology of vascular diseases such as hypertension and stroke. Endothelial cells express four NOX isoforms including the superoxide-generating enzymes NOX1, NOX2, and NOX5 and the hydrogen peroxide-generating enzyme NOX4. Studies on arteries from patients with coronary artery disease, and in animals with experimentally induced hypertension, diabetes, or atherosclerosis, suggest that NOX1, NOX2, and NOX5 promote endothelial dysfunction, inflammation, and apoptosis in the vessel wall, whereas NOX4 is by contrast vasoprotective in increasing nitric oxide bioavailability and suppressing cell death pathways. Based on these findings and promising preclinical studies with the NOX1/NOX2 antagonist, apocynin, we suggest that the field is poised for clinical evaluation of NOX inhibitors as therapeutics for cardiovascular disease.
Collapse
Affiliation(s)
- Grant R Drummond
- Vascular Biology and Immunopharmacology Group, Department of Pharmacology, Monash University, Clayton, Victoria, Australia; Department of Surgery, Monash Medical Centre, Southern Clinical School, Monash University, Clayton, Victoria, Australia.
| | - Christopher G Sobey
- Vascular Biology and Immunopharmacology Group, Department of Pharmacology, Monash University, Clayton, Victoria, Australia; Department of Surgery, Monash Medical Centre, Southern Clinical School, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
28
|
Li X, Xu M, Pitzer AL, Xia M, Boini KM, Li PL, Zhang Y. Control of autophagy maturation by acid sphingomyelinase in mouse coronary arterial smooth muscle cells: protective role in atherosclerosis. J Mol Med (Berl) 2014; 92:473-85. [PMID: 24463558 PMCID: PMC4211081 DOI: 10.1007/s00109-014-1120-y] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Revised: 12/06/2013] [Accepted: 01/02/2014] [Indexed: 12/20/2022]
Abstract
UNLABELLED Recent studies have indicated a protective role of autophagy in regulating vascular smooth muscle cells homeostasis in atherogenesis, but the mechanisms controlling autophagy, particularly autophagy maturation, are poorly understood. Here, we investigated whether acid sphingomyelinase (ASM)-regulated lysosome function is involved in autophagy maturation in coronary arterial smooth muscle cells (CASMCs) in the pathogenesis of atherosclerosis. In coronary arterial wall of ASM-deficient (Smpd1⁻/⁻) mice on Western diet, there were high expression levels of both LC3B, a robust marker of autophagosomes (APs), and p62, a selective autophagy substrate, compared with those in wild-type (Smpd1⁺/⁺) mice. By Western blotting and flow cytometry, atherogenic stimulation of Smpd1⁺/⁺ CASMCs with 7-ketocholesterol was found to significantly enhance LC3B expression and increase the content of both APs and autophagolysosomes (APLs). In Smpd1⁻/⁻ CASMCs, such 7-ketocholesterol-induced increases in LC3B and p62 expression and APs were further augmented, but APLs formation was abolished. Analysis of fluorescence resonance energy transfer between fluorescence-labeled LC3B and Lamp1 (lysosome marker) showed that 7-ketocholesterol markedly induced fusion of APs with lysosomes in Smpd1⁺/⁺ CASMCs, which was abolished in Smpd1⁻/⁻ CASMCs. Moreover, 7-ketocholesterol-induced expression of cell dedifferentiation marker vimentin and proliferation was enhanced in Smpd1⁻/⁻ CASMCs compared with those in Smpd1⁺/⁺ CASMCs. Lastly, overexpression of ASM further increased APLs formation in Smpd1⁺/⁺ CASMCs and restored APLs formation in Smpd1⁻/⁻ CASMCs indicating that increased ASM expression is highly correlated with enhanced APLs formation. Taken together, our data suggest that the control of lysosome trafficking and fusion by ASM is essential to a normal autophagic flux in CASMCs, which implicates that the deficiency of ASM-mediated regulation of autophagy maturation may result in imbalance of arterial smooth muscle cell homeostasis and thus serve as an important atherogenic mechanism in coronary arteries. KEY MESSAGES Acid sphingomyelinase (ASM) controls autophagy maturation in smooth muscle cells. ASM maintains smooth muscle cell homeostasis and its contractile phenotype. ASM plays a protective role in smooth muscle dysfunction and atherosclerosis.
Collapse
Affiliation(s)
- Xiang Li
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Medical College of Virginia Campus, Richmond, VA, 23298, USA
| | | | | | | | | | | | | |
Collapse
|
29
|
Endogenous ceramide contributes to the transcytosis of oxLDL across endothelial cells and promotes its subendothelial retention in vascular wall. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014; 2014:823071. [PMID: 24817993 PMCID: PMC4003761 DOI: 10.1155/2014/823071] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Revised: 03/17/2014] [Accepted: 03/21/2014] [Indexed: 11/24/2022]
Abstract
Oxidized low density of lipoprotein (oxLDL) is the major lipid found in atherosclerotic lesion and elevated plasma oxLDL is recognized to be a risk factor of atherosclerosis. Whether plasma oxLDL could be transported across endothelial cells and initiate atherosclerotic changes remains unknown. In an established in vitro cellular transcytosis model, the present study found that oxLDL could traffic across vascular endothelial cells and further that the regulation of endogenous ceramide production by ceramide metabolizing enzyme inhibitors significantly altered the transcytosis of oxLDL across endothelial cells. It was found that acid sphingomyelinase inhibitor, desipramine (DES), and de novo ceramide synthesis inhibitor, myriocin (MYR), both decreasing the endogenous ceramide production, significantly inhibited the transcytosis of oxLDL. Ceramidase inhibitor, N-oleoylethanolamine (NOE), and sphingomyelin synthase inhibitor, O-Tricyclo[5.2.1.02,6]dec-9-yl dithiocarbonate potassium salt (D609), both increasing the endogenous ceramide production, significantly upregulated the transcytosis of oxLDL. In vivo, injection of fluorescence labeled oxLDL into mice body also predisposed to the subendothelial retention of these oxidized lipids. The observations provided in the present study demonstrate that endogenous ceramide contributes to the transcytosis of oxLDL across endothelial cells and promotes the initiating step of atherosclerosis—the subendothelial retention of lipids in vascular wall.
Collapse
|
30
|
Margaritis M, Channon KM, Antoniades C. Statins as regulators of redox state in the vascular endothelium: beyond lipid lowering. Antioxid Redox Signal 2014; 20:1198-215. [PMID: 24111702 PMCID: PMC3934595 DOI: 10.1089/ars.2013.5430] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
SIGNIFICANCE Endothelial dysfunction and the imbalance between nitric oxide (NO) and reactive oxygen species production in the vascular endothelium are important early steps in atherogenesis, a major socioeconomic health problem. Statins have well-established roles in primary and secondary prevention of cardiovascular disease (CVD), due to both their lipid-lowering capacity and their pleiotropic properties. It is therefore important to understand the mechanisms by which statins can modify endothelial function and affect atherogenesis. RECENT ADVANCES In the last decade, the concept of statin pleiotropy has been reinforced by a large number of cell culture, animal, and translational studies. Statins have been shown to suppress the activity of pro-oxidant enzymes (such as NADPH oxidase) and pro-inflammatory transcriptional pathways in the endothelium. At the same time, they enhance endothelial NO synthase expression and activity while they also improve its enzymatic coupling. This leads to increased NO bioavailability and improved endothelial function. CRITICAL ISSUES Despite significant recent advances, the exact mechanisms of statin pleitropy are still only partially understood. The vast majority of the published literature relies on animal studies, while the actual mechanistic studies in humans are limited. FUTURE DIRECTIONS The success of statins as endothelium redox-modifying agents with a direct impact on clinical outcome highlights the importance of the endothelium as a therapeutic target in CVD. Better understanding of the mechanisms that underlie endothelial dysfunction could lead to the design of novel therapeutic strategies that target the vascular endothelium for the prevention and treatment of CVD.
Collapse
Affiliation(s)
- Marios Margaritis
- Division of Cardiovascular Medicine, University of Oxford , Oxford, United Kingdom
| | | | | |
Collapse
|
31
|
Activation of Nlrp3 inflammasomes enhances macrophage lipid-deposition and migration: implication of a novel role of inflammasome in atherogenesis. PLoS One 2014; 9:e87552. [PMID: 24475307 PMCID: PMC3903678 DOI: 10.1371/journal.pone.0087552] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Accepted: 12/21/2013] [Indexed: 12/30/2022] Open
Abstract
Although Nlrp3 inflammasome activation in macrophages has been shown to be critical for the development of atherosclerosis upon atherogenic stimuli, it remains unknown whether activated Nlrp3 inflammasomes by other non-atherogenic stimuli induce alterations in macrophages that may contribute in the concert with other factors to atherogenesis. Thus, the present study tested the hypothesis that activation of Nlrp3 inflammasomes by ATP, which is a classical non-lipid danger stimulus, enhances the migration of macrophage and increases lipids deposition in macrophages accelerating foam cell formation. We first demonstrated that extracellular ATP (2.5 mM) markedly increased the formation and activation of Nlrp3 inflammasomes in bone marrow macrophages (BMMs) from wild type (Asc+/+) mice resulting in activation of caspase-1 and IL-1β production. In these Asc+/+ macrophages, such stimulation of inflammasomes by non-lipid ATP was similar to those induced by atherogenic stimuli such as cholesterol crystals or 7-ketocholesterol. Both non-lipid and lipid forms of stimuli induced formation and activation of Nlrp3 inflammasomes, which were prevented by Asc gene deletion. Interestingly, Asc+/+ BMMs had dramatic lipids accumulation after stimulation with ATP. Further, we demonstrated that large amount of cholesterol was accumulated in lysosomes of Asc+/+ BMMs when inflammasomes were activated by ATP. Such intracellular and lysosomal lipids deposition was not observed in Asc−/− BMMs and also prevented by caspase-1 inhibitor WEHD. In addition, in vitro and in vivo experiments revealed that migration of Asc+/+ BMMs increased due to stimulation of Nlrp3 inflammasomes, which was markedly attenuated in Asc−/− BMMs. Together, these results suggest that activation of Nlrp3 inflammasomes remarkably increases the susceptibility of macrophages to lipid deposition and their migration ability. Such novel action of inflammasomes may facilitate entry or retention of macrophages into the arterial wall, where they form foam cells and ultimately induce atherosclerosis.
Collapse
|
32
|
Corre I, Guillonneau M, Paris F. Membrane signaling induced by high doses of ionizing radiation in the endothelial compartment. Relevance in radiation toxicity. Int J Mol Sci 2013; 14:22678-96. [PMID: 24252908 PMCID: PMC3856084 DOI: 10.3390/ijms141122678] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Revised: 11/01/2013] [Accepted: 11/06/2013] [Indexed: 01/30/2023] Open
Abstract
Tumor areas can now be very precisely delimited thanks to technical progress in imaging and ballistics. This has also led to the development of novel radiotherapy protocols, delivering higher doses of ionizing radiation directly to cancer cells. Despite this, radiation toxicity in healthy tissue remains a major issue, particularly with dose-escalation in these new protocols. Acute and late tissue damage following irradiation have both been linked to the endothelium irrigating normal tissues. The molecular mechanisms involved in the endothelial response to high doses of radiation are associated with signaling from the plasma membrane, mainly via the acid sphingomyelinase/ceramide pathway. This review describes this signaling pathway and discusses the relevance of targeting endothelial signaling to protect healthy tissues from the deleterious effects of high doses of radiation.
Collapse
Affiliation(s)
- Isabelle Corre
- CRCNA-UMR Inserm U892-CNRS 6299-Institut de Recherche en Santé de l'Université de Nantes, Nantes 44007, France.
| | | | | |
Collapse
|
33
|
Wei YM, Li X, Xu M, Abais JM, Chen Y, Riebling CR, Boini KM, Li PL, Zhang Y. Enhancement of autophagy by simvastatin through inhibition of Rac1-mTOR signaling pathway in coronary arterial myocytes. Cell Physiol Biochem 2013; 31:925-37. [PMID: 23817226 PMCID: PMC3753088 DOI: 10.1159/000350111] [Citation(s) in RCA: 103] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/20/2013] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND/AIMS In addition to their action of lowering blood cholesterol levels, statins modulate biological characteristics and functions of arterial myocytes such as viability, proliferation, apoptosis, survival and contraction. The present study tested whether simvastatin, as a prototype statin, enhances autophagy in coronary arterial myocytes (CAMs) to thereby exert their beneficial effects in atherosclerosis. METHODS AND RESULTS Using flow cytometry, we demonstrated that simvastatin significantly increased the autophagsome formation in CAMs. Western blot analysis confirmed that simvastatin significantly increased protein expression of typical autophagy markers LC3B and Beclin1 in these CAMs. Confocal microscopy further demonstrated that simvastatin increased fusion of autophagosomes with lysosomes, which was blocked by autophagy inhibitor 3-methyladenine or silencing of Atg7 genes. Simvastatin reduced mammalian target of rapamycin (mTOR) activity, which was reversed by Rac1-GTPase overexpression and the mTOR agonist phosphatidic acid. Moreover, both Rac1-GTPase overexpression and activation of mTOR by phosphatidic acid drastically blocked simvastatin-induced autophagosome formation in CAMs. Interestingly, simvastatin increased protein expression of a contractile phenotype marker calponin in CAMs, which was blocked by autophagy inhibitor 3-methyladenine. Simvastatin markedly reduced proliferation of CAMs under both control and proatherogenic stimulation. However, this inhibitory effect of simvastatin on CAM proliferation was blocked by by autophagy inhibitor 3-methyladenine or silencing of Atg7 genes. Lastly, animal experiments demonstrated that simvastatin increased protein expression of LC3B and calponin in mouse coronary arteries. CONCLUSION Our results indicate that simvastatin inhibits the Rac1-mTOR pathway and thereby increases autophagy in CAMs which may stabilize CAMs in the contractile phenotype to prevent proliferation and growth of these cells.
Collapse
Affiliation(s)
- Yu-Miao Wei
- Department of Pharmacology & Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23298, USA
| | | | | | | | | | | | | | | | | |
Collapse
|