1
|
Jin M, Chen X, Zheng L, Peng Y, Lin M, Liang K, Liu X, Xu Z, Yang Y, Wei B, Wan J. Astaxanthin-loaded polylactic acid-glycolic acid nanoparticles alleviates atherosclerosis by suppressing macrophage ferroptosis via the NRF2/SLC7A11/GPX4 pathway. Arch Biochem Biophys 2025; 765:110316. [PMID: 39848420 DOI: 10.1016/j.abb.2025.110316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 11/11/2024] [Accepted: 01/19/2025] [Indexed: 01/25/2025]
Abstract
BACKGROUND Astaxanthin (ASX), a fat-soluble carotenoid mainly sourced from Haematococcus pluvialis, shows promise for clinical applications in chronic inflammatory diseases. This study investigates whether ASX can mitigate atherosclerosis (AS) by modulating macrophage ferroptosis and provides astaxanthin-loaded polylactic acid-glycolic acid nanoparticles (ASX-PLGA NPs) as comparison. METHOD ApoE-/- mice were fed a high-fat diet with ASX or statin intervention. Plaque area, lipid aggregation, collagen content, and ferroptosis-related indicators were assessed. Moreover, ASX-PLGA NPs were synthesized and characterized and were used to pretreat macrophages induced with oxidized low-density lipoprotein (ox-LDL). Indicators linked to ferroptosis and oxidative stress were detected. Finally, the expression of nuclear factor erythroid -related factor 2 (NRF2) was evaluated. RESULTS ASX intervention significantly delayed the progression of AS plaques, characterized by reductions in plaque area and increased collagen fibers. The observed improvements in AS were consistent with statins. ASX-PLGA NPs demonstrate good safety and stability and have better therapeutic effects than ASX alone. Indicators linked to ferroptosis and oxidative stress were significantly improved in groups containing ASX in vivo and vitro. Additionally, ASX facilitated the nuclear translocation of NRF2, which could be attenuated with ML385, a specific inhibitor of NRF2. CONCLUSION ASX-PLGA NPs have better therapeutic effects than ASX alone. The regulation of NRF2/SLC7A11/GPX4 represents a novel mechanism by which ASX can counteract ferroptosis and impede AS progression.
Collapse
Affiliation(s)
- Mengying Jin
- Department of Cardiology, Zhongnan Hospital of Wuhan University, No 169 Donghu Road, Wuchang District, Wuhan, 430071, Hubei Province, China.
| | - Xiao Chen
- Department of Cardiology, Zhongnan Hospital of Wuhan University, No 169 Donghu Road, Wuchang District, Wuhan, 430071, Hubei Province, China.
| | - Lanzhuoying Zheng
- Department of Cardiology, Zhongnan Hospital of Wuhan University, No 169 Donghu Road, Wuchang District, Wuhan, 430071, Hubei Province, China.
| | - Yuanyuan Peng
- Department of Cardiology, Zhongnan Hospital of Wuhan University, No 169 Donghu Road, Wuchang District, Wuhan, 430071, Hubei Province, China.
| | - Mingying Lin
- Department of Cardiology, Hainan General Hospital and Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, China.
| | - Ke Liang
- Department of Cardiology, Zhongnan Hospital of Wuhan University, No 169 Donghu Road, Wuchang District, Wuhan, 430071, Hubei Province, China.
| | - Xinran Liu
- Department of Cardiology, Zhongnan Hospital of Wuhan University, No 169 Donghu Road, Wuchang District, Wuhan, 430071, Hubei Province, China.
| | - Zihan Xu
- Department of Cardiology, Zhongnan Hospital of Wuhan University, No 169 Donghu Road, Wuchang District, Wuhan, 430071, Hubei Province, China.
| | - Yiming Yang
- Department of Cardiology, Zhongnan Hospital of Wuhan University, No 169 Donghu Road, Wuchang District, Wuhan, 430071, Hubei Province, China.
| | - Baozhu Wei
- Department of Cardiology, Zhongnan Hospital of Wuhan University, No 169 Donghu Road, Wuchang District, Wuhan, 430071, Hubei Province, China.
| | - Jing Wan
- Department of Cardiology, Zhongnan Hospital of Wuhan University, No 169 Donghu Road, Wuchang District, Wuhan, 430071, Hubei Province, China.
| |
Collapse
|
2
|
Jokesch P, Holzer L, Jantscher L, Guttzeit S, Übelhart R, Oskolkova O, Bochkov V, Gesslbauer B. Identification of plasma proteins binding oxidized phospholipids using pull-down proteomics and OxLDL masking assay. J Lipid Res 2025; 66:100704. [PMID: 39566852 PMCID: PMC11696850 DOI: 10.1016/j.jlr.2024.100704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/08/2024] [Accepted: 11/05/2024] [Indexed: 11/22/2024] Open
Abstract
Oxidized phospholipids (OxPLs) are increasingly recognized as toxic and proinflammatory mediators, which raises interest in the mechanisms of their detoxification. Circulating OxPLs are bound and neutralized by plasma proteins, including both antibodies and non-immunoglobulin proteins. The latter group of proteins is essentially not investigated because only three OxPC-binding plasma proteins are currently known. The goal of this work was to characterize a broad spectrum of plasma proteins selectively binding OxPLs. Using pull-down-proteomic analysis, we found about 150 non-immunoglobulin proteins preferentially binding oxidized 1-palmitoyl-2-arachidonoyl-sn-glycero-phosphatidylcholine (OxPAPC) as compared to non-oxidized PAPC. To test if candidate proteins indeed can form a barrier isolating OxPLs from recognition by other proteins, we applied an immune masking assay. Oxidized LDL (OxLDL) immobilized in multiwell plates was used as a carrier of OxPLs, while mAbs recognizing OxPC or OxPE were used as "detectors" showing if OxPLs on the surface of OxLDL are physically accessible to external binding partners. Using an orthogonal combination of pull-down and masking assays we confirmed that previously described OxPL-binding proteins (non-fractionated IgM, CFH, and Apo-M) indeed can bind to and mask OxPC and OxPE on liposomes and OxLDL. Furthermore, we identified additional plasma proteins selectively binding and masking OxPC including Apo-D, Apo-H, pulmonary surfactant-associated protein B, and antithrombin-III. We hypothesize that in addition to circulating antibodies, multiple non-immunoglobulin plasma proteins can also bind OxPLs and modulate their recognition by innate and adaptive immunity.
Collapse
Affiliation(s)
- Philipp Jokesch
- Department of Pharmaceutical Chemistry, Institute of Pharmaceutical Sciences, University of Graz, Graz, Austria
| | - Lisa Holzer
- Department of Pharmaceutical Chemistry, Institute of Pharmaceutical Sciences, University of Graz, Graz, Austria
| | - Lydia Jantscher
- Department of Pharmaceutical Chemistry, Institute of Pharmaceutical Sciences, University of Graz, Graz, Austria
| | | | | | - Olga Oskolkova
- Department of Pharmaceutical Chemistry, Institute of Pharmaceutical Sciences, University of Graz, Graz, Austria
| | - Valery Bochkov
- Department of Pharmaceutical Chemistry, Institute of Pharmaceutical Sciences, University of Graz, Graz, Austria; Field of Excellence BioHealth - University of Graz, Graz, Austria.
| | - Bernd Gesslbauer
- Department of Pharmaceutical Chemistry, Institute of Pharmaceutical Sciences, University of Graz, Graz, Austria.
| |
Collapse
|
3
|
Alic L, Dendinovic K, Papac-Milicevic N. The complement system in lipid-mediated pathologies. Front Immunol 2024; 15:1511886. [PMID: 39635529 PMCID: PMC11614835 DOI: 10.3389/fimmu.2024.1511886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 10/31/2024] [Indexed: 12/07/2024] Open
Abstract
The complement system, a coordinator and facilitator of the innate immune response, plays an essential role in maintaining host homeostasis. It promotes clearance of pathogen- and danger-associated molecular patterns, regulates adaptive immunity, and can modify various metabolic processes such as energy expenditure, lipid metabolism, and glucose homeostasis. In this review, we will focus on the intricate interplay between complement components and lipid metabolism. More precisely, we will display how alterations in the activation and regulation of the complement system affect pathological outcome in lipid-associated diseases, such as atherosclerosis, obesity, metabolic syndrome, age-related macular degeneration, and metabolic dysfunction-associated steatotic liver disease. In addition to that, we will present and evaluate underlying complement-mediated physiological mechanisms, observed both in vitro and in vivo. Our manuscript will demonstrate the clinical significance of the complement system as a bridging figure between innate immunity and lipid homeostasis.
Collapse
Affiliation(s)
- Lejla Alic
- Department of Medical Biochemistry, Faculty of Medicine, University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Kristina Dendinovic
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Nikolina Papac-Milicevic
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
4
|
Boffa MB, Koschinsky ML. Lipoprotein(a) and cardiovascular disease. Biochem J 2024; 481:1277-1296. [PMID: 39302109 PMCID: PMC11555715 DOI: 10.1042/bcj20240037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/28/2024] [Accepted: 08/28/2024] [Indexed: 09/22/2024]
Abstract
Elevated plasma levels of lipoprotein(a) (Lp(a)) are a prevalent, independent, and causal risk factor for atherosclerotic cardiovascular disease and calcific aortic valve disease. Lp(a) consists of a lipoprotein particle resembling low density lipoprotein and the covalently-attached glycoprotein apolipoprotein(a) (apo(a)). Novel therapeutics that specifically and potently lower Lp(a) levels are currently in advanced stages of clinical development, including in large, phase 3 cardiovascular outcomes trials. However, fundamental unanswered questions remain concerning some key aspects of Lp(a) biosynthesis and catabolism as well as the true pathogenic mechanisms of the particle. In this review, we describe the salient biochemical features of Lp(a) and apo(a) and how they underlie the disease-causing potential of Lp(a), the factors that determine plasma Lp(a) concentrations, and the mechanism of action of Lp(a)-lowering drugs.
Collapse
Affiliation(s)
- Michael B. Boffa
- Department of Biochemistry, University of Western Ontario, London, Ontario, Canada
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
| | - Marlys L. Koschinsky
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
5
|
Deroissart J, Binder CJ, Porsch F. Role of Antibodies and Their Specificities in Atherosclerotic Cardiovascular Disease. Arterioscler Thromb Vasc Biol 2024; 44:2154-2168. [PMID: 39114917 DOI: 10.1161/atvbaha.124.319843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2024]
Abstract
Atherosclerosis is a lipid-driven chronic inflammatory disease that is modulated by innate and adaptive immunity including humoral immunity. Importantly, antibody alterations achieved by genetic means or active and passive immunization strategies in preclinical studies can improve or aggravate atherosclerosis. Additionally, a wide range of epidemiological data demonstrate not only an association between the total levels of different antibody isotypes but also levels of antibodies targeting specific antigens with atherosclerotic cardiovascular disease. Here, we discuss the potential role of atherogenic dyslipidemia on the antibody repertoire and review potential antibody-mediated effector mechanisms involved in atherosclerosis development highlighting the major atherosclerosis-associated antigens that trigger antibody responses.
Collapse
Affiliation(s)
- Justine Deroissart
- Department of Laboratory Medicine, Medical University of Vienna, Austria
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Austria
| | - Florentina Porsch
- Department of Laboratory Medicine, Medical University of Vienna, Austria
| |
Collapse
|
6
|
Vaghasiya J, Jha A, Basu S, Bagan A, Jengsuksavat SK, Ravandi A, Pascoe CD, Halayko AJ. Neutralizing Oxidized Phosphatidylcholine Reduces Airway Inflammation and Hyperreactivity in a Murine Model of Allergic Asthma. BIOLOGY 2024; 13:627. [PMID: 39194564 DOI: 10.3390/biology13080627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 08/12/2024] [Accepted: 08/15/2024] [Indexed: 08/29/2024]
Abstract
Oxidative stress is associated with asthma pathobiology. We reported that oxidized phosphatidylcholines (OxPCs) are mediators of oxidative stress and accumulate in the lung in response to allergen challenge. The current study begins to unravel mechanisms for OxPC accumulation in the lung, providing the first insights about how OxPCs underpin allergic airway pathophysiology, and pre-clinical testing of selective neutralization of OxPCs in a murine model of allergic asthma. We hypothesized that intranasal delivery of E06, a natural IgM antibody that neutralizes the biological activity of OxPCs, can ameliorate allergen-induced airway inflammation and airway hyperresponsiveness. Adult BALB/c mice were intranasally (i.n.) challenged with house dust mite (HDM) (25 μg/mouse, 2 weeks). Some animals also received E06 monoclonal antibody (mAb) (10 µg) i.n. 1 hr before each HDM challenge. HDM challenge reduced mRNA for anti-oxidant genes (SOD1, SOD2, HO-1, and NFE2L2) in the lung by several orders of magnitude (p < 0.05). Concomitantly, total immune cell number in bronchoalveolar lavage fluid (BALF) increased significantly (p < 0.001). E06 mAb treatment prevented allergen-induced BALF immune cell number by 43% (p < 0.01). This included a significant blockade of eosinophils (by 48%, p < 0.001), neutrophils (by 80%, p < 0.001), macrophages (by 80%, p < 0.05), and CD4 (by 30%, p < 0.05) and CD8 (by 42%, p < 0.01) lymphocytes. E06 effects correlated with a significant reduction in TNF (by 64%, p < 0.001) and IL-1β (by 75%, p < 0.05) and a trend to diminish accumulation of other cytokines (e.g., IL-4, -10, and -33, and IFN-γ). E06 mAb treatment also inhibited HDM exposure-induced increases in total respiratory resistance and small airway resistance by 24% and 26%, respectively. In conclusion, prophylactic treatment with an OxPC-neutralizing antibody significantly limits allergen-induced airway inflammation and airway hyperresponsiveness, suggesting that OxPCs are important mediators of oxidative stress-associated allergic lung pathophysiology.
Collapse
Affiliation(s)
- Jignesh Vaghasiya
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB R3E 3P4, Canada
- Biology of Breathing Group, Children's Research Hospital of Manitoba, Winnipeg, MB R3E 3P4, Canada
| | - Aruni Jha
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB R3E 3P4, Canada
- Biology of Breathing Group, Children's Research Hospital of Manitoba, Winnipeg, MB R3E 3P4, Canada
| | - Sujata Basu
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB R3E 3P4, Canada
- Biology of Breathing Group, Children's Research Hospital of Manitoba, Winnipeg, MB R3E 3P4, Canada
| | - Alaina Bagan
- Biology of Breathing Group, Children's Research Hospital of Manitoba, Winnipeg, MB R3E 3P4, Canada
| | - Siwon K Jengsuksavat
- Biology of Breathing Group, Children's Research Hospital of Manitoba, Winnipeg, MB R3E 3P4, Canada
| | - Amir Ravandi
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB R3E 3P4, Canada
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada
- Department of Internal Medicine, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 3P4, Canada
| | - Christopher D Pascoe
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB R3E 3P4, Canada
- Biology of Breathing Group, Children's Research Hospital of Manitoba, Winnipeg, MB R3E 3P4, Canada
| | - Andrew J Halayko
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB R3E 3P4, Canada
- Biology of Breathing Group, Children's Research Hospital of Manitoba, Winnipeg, MB R3E 3P4, Canada
- Department of Internal Medicine, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 3P4, Canada
| |
Collapse
|
7
|
Mattos MS, Vandendriessche S, Waisman A, Marques PE. The immunology of B-1 cells: from development to aging. Immun Ageing 2024; 21:54. [PMID: 39095816 PMCID: PMC11295433 DOI: 10.1186/s12979-024-00455-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 07/17/2024] [Indexed: 08/04/2024]
Abstract
B-1 cells have intricate biology, with distinct function, phenotype and developmental origin from conventional B cells. They generate a B cell receptor with conserved germline characteristics and biased V(D)J recombination, allowing this innate-like lymphocyte to spontaneously produce self-reactive natural antibodies (NAbs) and become activated by immune stimuli in a T cell-independent manner. NAbs were suggested as "rheostats" for the chronic diseases in advanced age. In fact, age-dependent loss of function of NAbs has been associated with clinically-relevant diseases in the elderly, such as atherosclerosis and neurodegenerative disorders. Here, we analyzed comprehensively the ontogeny, phenotypic characteristics, functional properties and emerging roles of B-1 cells and NAbs in health and disease. Additionally, after navigating through the complexities of B-1 cell biology from development to aging, therapeutic opportunities in the field are discussed.
Collapse
Affiliation(s)
- Matheus Silvério Mattos
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, 3000, Louvain, Belgium
| | - Sofie Vandendriessche
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, 3000, Louvain, Belgium
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Centre of the Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Pedro Elias Marques
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, 3000, Louvain, Belgium.
| |
Collapse
|
8
|
Zehrfeld N, Abelmann M, Benz S, Zippel CL, Beider S, Kramer E, Seeliger T, Sogkas G, Gödecke V, Ahrenstorf G, Armbruster FP, Skripuletz T, Witte T, Derda AA, Sonnenschein K, Ernst D. Primary Sjögren's syndrome independently promotes premature subclinical atherosclerosis. RMD Open 2024; 10:e003559. [PMID: 38663882 PMCID: PMC11043759 DOI: 10.1136/rmdopen-2023-003559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
OBJECTIVES Cardiovascular comorbidities are common in patients with autoimmune diseases. This study investigates the extent of subclinical atherosclerosis in patients with primary Sjögren's syndrome (pSS). Correlations with clinical factors such as organ involvement (OI) or disease activity were analysed and oxLDL antibodies (oxLDL ab) were measured as potential biomarkers of vascular damage. METHODS Patients with pSS were consecutively included from the rheumatology outpatient clinic. Age- and sex-matched controls were recruited (2:1 ratio). Data collection was performed by a standardised questionnaire and Doppler ultrasound to evaluate the plaque extent and carotid intima-media thickness (cIMT). Propensity score matching included all cardiovascular risk (CVR) factors and corresponding laboratory markers. RESULTS Data were available for 299 participants (199 pSS/100 controls), aged 59.4 years (50.6-65.0), 19.1% male. After matching, the pSS cohort had greater cIMT (p<0.001) and plaque extent (OR=1.82; 95% CI 1.14 to 2.95). Subgroup analyses of patients with pSS revealed that OI was associated with increased cIMT (p=0.025) and increased plaque occurrence compared with patients without OI (OR=1.74; 95% CI 1.02 to 3.01). OxLDL ab tended to be lower in patients with plaque (p=0.052). Correlations of higher Oxidized Low Density Lipoprotein (oxLDL) ab with EULAR Sjögren's Syndrome Disease Activity Index (p<0.001) and anti-Sjögren's-syndrome-related antigen A autoantibodies (SSA/Ro antibodies) (p=0.026) were observed. CONCLUSIONS Subclinical atherosclerosis occurs earlier and more severely in patients with pSS. The difference in cIMT between pSS and controls seems mainly driven by patients with OI, suggesting that this subgroup is particularly at risk. OxLDL ab might protect against atherosclerotic progression in patients with pSS. CVR stratification and preventive medications such as Hydroxymethylglutaryl-CoA (HMG-CoA) reductase inhibitors should be discussed and further longitudinal studies are needed.
Collapse
Affiliation(s)
- Nadine Zehrfeld
- Rheumatology & Immunology, Hannover Medical School, Hannover, Lower Saxony, Germany
| | - Malin Abelmann
- Cardiology & Angiology, Hannover Medical School, Hannover, Lower Saxony, Germany
| | - Sabrina Benz
- Faculty of Social Sciences, University of Mannheim, Mannheim, Germany
| | - Clara Luisa Zippel
- Rheumatology & Immunology, Hannover Medical School, Hannover, Lower Saxony, Germany
| | - Sonja Beider
- Rheumatology & Immunology, Hannover Medical School, Hannover, Lower Saxony, Germany
| | - Emelie Kramer
- Rheumatology & Immunology, Hannover Medical School, Hannover, Lower Saxony, Germany
| | - Tabea Seeliger
- Neurology, Hannover Medical School, Hannover, Lower Saxony, Germany
| | - Georgios Sogkas
- Rheumatology & Immunology, Hannover Medical School, Hannover, Lower Saxony, Germany
| | - Vega Gödecke
- Rheumatology & Immunology, Hannover Medical School, Hannover, Lower Saxony, Germany
| | - Gerrit Ahrenstorf
- Rheumatology & Immunology, Hannover Medical School, Hannover, Lower Saxony, Germany
| | | | | | - Torsten Witte
- Rheumatology & Immunology, Hannover Medical School, Hannover, Lower Saxony, Germany
| | - Anselm Arthur Derda
- Cardiology & Angiology, Hannover Medical School, Hannover, Lower Saxony, Germany
| | | | - Diana Ernst
- Rheumatology & Immunology, Hannover Medical School, Hannover, Lower Saxony, Germany
| |
Collapse
|
9
|
Zhang Y, Qi X, Wang S, Zhang W, Yang R, Wang X, Chen W, Ji F, Dong J, Yu X. Serum immunoglobulin M is associated with the severity of coronary artery disease in adults. PeerJ 2024; 12:e17012. [PMID: 38464758 PMCID: PMC10921929 DOI: 10.7717/peerj.17012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 02/05/2024] [Indexed: 03/12/2024] Open
Abstract
Purpose The purpose of this study was to investigate the relationship between serum immunoglobulin M (IgM) and the severity of coronary artery disease in Chinese patients who underwent coronary angiography. Methods A total of 2,045 patients who underwent coronary angiography (CAG) from March 2017 to March 2020 at Beijing Hospital were included in this study. Serum IgM concentration and biochemical indicators were measured before coronary angiography (CAG). The triquartile IgM levels at baseline in the population were analysed. Spearman rank correlation was used to analyse the association between IgM and traditional risk factors for coronary artery disease (CAD). CAD patients were divided into subgroups by affected area, number of affected vessels, and Gensini score to analyse the relationship between IgM and CAD severity. Multivariable logistic regression analysis was used to evaluate the association between IgM and CAD severity. Results Serum IgM levels were significantly lower in the CAD group (63.5 mg/dL) than in the non-coronary artery disease (NCAD) group (72.3 mg/dL) (P < 0.001). Serum IgM levels were significantly associated with sex. Serum IgM levels were positively correlated with traditional CAD risk factors such as TG, TC and LDL-C (P < 0.05), and negatively associated with the number of obstructed vessels, the number of affected areas, and Gensini scores. After adjusting for age, sex, smoking status, hypertension, dyslipidaemia, diabetes, stroke, and statin use history, a high IgM level was independently negatively associated with the severity of CAD expressed by the Gensini score. Conclusion We determined that serum IgM was independently negatively associated with the severity of CAD diagnosed by angiography in Chinese adults.
Collapse
Affiliation(s)
- Yanan Zhang
- The Affiliated Hospital of Qingdao University, Qing Dao, Shan Dong, China
| | - Xi Qi
- Beijing Hospital, Beijing, China
| | | | | | | | | | | | - Fusui Ji
- Beijing Hospital, Beijing, China
| | - Jun Dong
- Beijing Hospital, Beijing, China
| | - Xue Yu
- Beijing Hospital, Beijing, China
| |
Collapse
|
10
|
Happonen N, Härma MA, Akhi R, Nissinen AE, Savolainen MJ, Ruuth M, Öörni K, Adeshara K, Lehto M, Groop PH, Koivukangas V, Hukkanen J, Hörkkö S. Impact of RYGB surgery on plasma immunoglobulins: association between blood pressure and glucose levels six months after surgery. APMIS 2024; 132:187-197. [PMID: 38149431 DOI: 10.1111/apm.13366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 11/23/2023] [Indexed: 12/28/2023]
Abstract
We aimed to study levels of natural antibodies in plasma, and their associations to clinical and fecal biomarkers, before and 6 months after Roux-en-Y gastric bypass (RYGB) surgery. Thirty individuals with obesity [16 type 2 diabetic, 14 non-diabetic (ND)] had RYGB surgery. Total plasma IgA, IgG and IgM antibody levels and specific antibodies to oxidized low-density lipoprotein (oxLDL), malondialdehyde-acetaldehyde adducts, Porphyromonas gingivalis gingipain A hemagglutinin domain (Rgp44), and phosphocholine were measured using chemiluminescence immunoassay. Associations between plasma and fecal antibodies as well as clinical markers were analyzed. RYGB surgery reduced blood pressure, and the glycemic state was improved. A higher level of diastolic blood pressure was associated with lower plasma antibodies to oxLDL after surgery. Also, lower level of glucose markers associated with lower level of plasma antibodies to bacterial virulence factors. Antibodies to oxLDL decreased after surgery, and positive association between active serum lipopolysaccharide and specific oxLDL antibodies was detected. Total IgG levels decreased after surgery, but only in ND individuals. Reduced level of total plasma IgG, improved state of hypertension and hyperglycemia and their associations with decreased levels of specific antibodies in plasma, suggest an improved state of systemic inflammation after RYGB surgery.
Collapse
Affiliation(s)
- Natalie Happonen
- Medical Microbiology and Immunology, Research Unit of Biomedicine and Internal Medicine, University of Oulu, Oulu, Finland
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
- Nordlab, Oulu University Hospital, Oulu, Finland
| | - Mari-Anne Härma
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Ramin Akhi
- Medical Microbiology and Immunology, Research Unit of Biomedicine and Internal Medicine, University of Oulu, Oulu, Finland
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Antti E Nissinen
- Medical Microbiology and Immunology, Research Unit of Biomedicine and Internal Medicine, University of Oulu, Oulu, Finland
| | - Markku J Savolainen
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
- Research Unit of Biomedicine and Internal Medicine and Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Maija Ruuth
- Atherosclerosis Research Laboratory, Wihuri Research Institute, Helsinki, Finland
| | - Katariina Öörni
- Atherosclerosis Research Laboratory, Wihuri Research Institute, Helsinki, Finland
- Molecular and Integrative Biosciences, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Krishna Adeshara
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Markku Lehto
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Per-Henrik Groop
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Vesa Koivukangas
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
- Department of Surgery, Oulu University Hospital, Oulu, Finland
| | - Janne Hukkanen
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
- Research Unit of Biomedicine and Internal Medicine and Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Sohvi Hörkkö
- Medical Microbiology and Immunology, Research Unit of Biomedicine and Internal Medicine, University of Oulu, Oulu, Finland
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| |
Collapse
|
11
|
Zhang T, Pang C, Xu M, Zhao Q, Hu Z, Jiang X, Guo M. The role of immune system in atherosclerosis: Molecular mechanisms, controversies, and future possibilities. Hum Immunol 2024; 85:110765. [PMID: 38369442 DOI: 10.1016/j.humimm.2024.110765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 02/06/2024] [Accepted: 02/08/2024] [Indexed: 02/20/2024]
Abstract
Numerous cardiovascular disorders have atherosclerosis as their pathological underpinning. Numerous studies have demonstrated that, with the aid of pattern recognition receptors, cytokines, and immunoglobulins, innate immunity, represented by monocytes/macrophages, and adaptive immunity, primarily T/B cells, play a critical role in controlling inflammation and abnormal lipid metabolism in atherosclerosis. Additionally, the finding of numerous complement components in atherosclerotic plaques suggests yet again how heavily the immune system controls atherosclerosis. Therefore, it is essential to have a thorough grasp of how the immune system contributes to atherosclerosis. The specific molecular mechanisms involved in the activation of immune cells and immune molecules in atherosclerosis, the controversy surrounding some immune cells in atherosclerosis, and the limitations of extrapolating from relevant animal models to humans were all carefully reviewed in this review from the three perspectives of innate immunity, adaptive immunity, and complement system. This could provide fresh possibilities for atherosclerosis research and treatment in the future.
Collapse
Affiliation(s)
- Tianle Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Chenxu Pang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Mengxin Xu
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Qianqian Zhao
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Zhijie Hu
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Xijuan Jiang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China.
| | - Maojuan Guo
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China.
| |
Collapse
|
12
|
Raposo-Gutiérrez I, Rodríguez-Ronchel A, Ramiro AR. Atherosclerosis antigens as targets for immunotherapy. NATURE CARDIOVASCULAR RESEARCH 2023; 2:1129-1147. [PMID: 39196152 DOI: 10.1038/s44161-023-00376-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 10/18/2023] [Indexed: 08/29/2024]
Abstract
Atherosclerosis is a chronic inflammatory disease of the arteries that can lead to thrombosis, infarction and stroke, underlying the first cause of mortality worldwide. Adaptive immunity plays critical roles in atherosclerosis, and numerous studies have ascribed both atheroprotective and atherogenic functions to specific subsets of T and B cells. However, less is known on how antigen specificity determines the protective or adverse outcome of such adaptive responses. Understanding antigen triggers in atherosclerosis is crucial to delve deeper into mechanisms of disease initiation and progression and to implement specific immunotherapeutic approaches, including vaccination strategies. Here we review the role of adaptive immunity in atherosclerosis and the insights that single-cell technology has provided into the function of distinct immune cell subsets. We outline the most relevant atherosclerosis antigens and antibodies reported to date and examine their immunotherapeutic potential. Finally, we review the most promising vaccination-based clinical trials targeting the adaptive immune system.
Collapse
Affiliation(s)
- Irene Raposo-Gutiérrez
- B Lymphocyte Lab, Novel Mechanisms of Atherosclerosis Program, Spanish National Center for Cardiovascular Research, Madrid, Spain
| | - Ana Rodríguez-Ronchel
- B Lymphocyte Lab, Novel Mechanisms of Atherosclerosis Program, Spanish National Center for Cardiovascular Research, Madrid, Spain
| | - Almudena R Ramiro
- B Lymphocyte Lab, Novel Mechanisms of Atherosclerosis Program, Spanish National Center for Cardiovascular Research, Madrid, Spain.
| |
Collapse
|
13
|
Marcovina SM. Lipoprotein(a): a genetically determined risk factor for Cardiovascular disease. Crit Rev Clin Lab Sci 2023; 60:560-572. [PMID: 37452525 DOI: 10.1080/10408363.2023.2229915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 05/11/2023] [Accepted: 06/22/2023] [Indexed: 07/18/2023]
Abstract
Lipoprotein(a) is a complex lipoprotein with unique characteristics distinguishing it from all the other apolipoprotein B-containing lipoprotein particles. Its lipid composition and the presence of a single molecule of apolipoprotein B per particle, render lipoprotein(a) similar to low-density lipoproteins. However, the presence of a unique, carbohydrate-rich protein termed apolipoprotein(a), linked by a covalent bond to apolipoprotein B imparts unique characteristics to lipoprotein(a) distinguishing it from all the other lipoproteins. Apolipoprotein(a) is highly polymorphic in size ranging in molecular weight from <300 KDa to >800 kDa. Both the size polymorphism and the concentration of lipoprotein(a) in plasma are genetically determined and unlike other lipoproteins, plasma concentration is minimally impacted by lifestyle modifications or lipid-lowering drugs. Many studies involving hundreds of thousands of individuals have provided strong evidence that elevated lipoprotein(a) is genetically determined and a causal risk factor for atherosclerotic cardiovascular disease. The concentration attained in adulthood is already present in children at around 5 years of age and therefore, those with elevated lipoprotein(a) are prematurely exposed to a high risk of cardiovascular disease. Despite the large number of guidelines and consensus statements on the management of lipoprotein(a) in atherosclerotic cardiovascular disease published in the last decade, lipoprotein(a) is still seldom measured in clinical settings. In this review, we provide an overview of the most important features that characterize lipoprotein(a), its role in cardiovascular disease, and the importance of adding the measurement of lipoprotein(a) for screening adults and youths to identify those at increased risk of atherosclerotic cardiovascular disease due to their elevated plasma concentration of lipoprotein(a).
Collapse
|
14
|
Xue S, Su Z, Liu D. Immunometabolism and immune response regulate macrophage function in atherosclerosis. Ageing Res Rev 2023; 90:101993. [PMID: 37379970 DOI: 10.1016/j.arr.2023.101993] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 06/23/2023] [Indexed: 06/30/2023]
Abstract
Macrophages are crucial in the progression of atherosclerotic cardiovascular disease (ASCVD). In the atherosclerotic lesions, macrophages play a central role in maintaining inflammatory response, promoting plaque development, and facilitating thrombosis. Increasing studies indicate that metabolic reprogramming and immune response mediate macrophage functional changes in all stages of atherosclerosis. In this review article, we explain how metabolic changes in glycolysis, oxidative phosphorylation, the tricarboxylic acid cycle, fatty acid synthesis, fatty acid oxidation, and cholesterol metabolism regulate macrophage function in atherosclerosis. We discuss how immune response to oxidized lipids regulate macrophage function in atherosclerosis. Additionally, we explore how abnormal metabolism leads to macrophage mitochondrial dysfunction in atherosclerosis.
Collapse
Affiliation(s)
- Sheng Xue
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266003 China.
| | - Zhe Su
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266003 China
| | - Dacheng Liu
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266003 China
| |
Collapse
|
15
|
Lecis D, Massaro G, Benedetto D, Di Luozzo M, Russo G, Mauriello A, Federici M, Sangiorgi GM. Immunomodulation Therapies for Atherosclerosis: The Past, the Present, and the Future. Int J Mol Sci 2023; 24:10979. [PMID: 37446157 PMCID: PMC10342012 DOI: 10.3390/ijms241310979] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 06/26/2023] [Accepted: 06/30/2023] [Indexed: 07/15/2023] Open
Abstract
Atherosclerotic cardiovascular disease is the most common cause of morbidity and death worldwide. Recent studies have demonstrated that this chronic inflammatory disease of the arterial wall can be controlled through the modulation of immune system activity. Many patients with cardiovascular disease remain at elevated risk of recurrent events despite receiving current, state-of-the-art preventive medical treatment. Much of this residual risk is attributed to inflammation. Therefore, finding new treatment strategies for this category of patients became of common interest. This review will discuss the experimental and clinical data supporting the possibility of developing immune-based therapies for lowering cardiovascular risk, explicitly focusing on vaccination strategies.
Collapse
Affiliation(s)
- Dalgisio Lecis
- Division of Cardiology, “Tor Vergata” University Hospital, Viale Oxford 81, 00133 Rome, Italy; (G.M.); (D.B.); (M.D.L.); (G.R.)
| | - Gianluca Massaro
- Division of Cardiology, “Tor Vergata” University Hospital, Viale Oxford 81, 00133 Rome, Italy; (G.M.); (D.B.); (M.D.L.); (G.R.)
| | - Daniela Benedetto
- Division of Cardiology, “Tor Vergata” University Hospital, Viale Oxford 81, 00133 Rome, Italy; (G.M.); (D.B.); (M.D.L.); (G.R.)
| | - Marco Di Luozzo
- Division of Cardiology, “Tor Vergata” University Hospital, Viale Oxford 81, 00133 Rome, Italy; (G.M.); (D.B.); (M.D.L.); (G.R.)
| | - Giulio Russo
- Division of Cardiology, “Tor Vergata” University Hospital, Viale Oxford 81, 00133 Rome, Italy; (G.M.); (D.B.); (M.D.L.); (G.R.)
| | - Alessandro Mauriello
- Department of Experimental Medicine, University “Tor Vergata”, 00133 Rome, Italy;
| | - Massimo Federici
- Department of Systemic Medicine, University “Tor Vergata”, 00133 Rome, Italy;
| | - Giuseppe Massimo Sangiorgi
- Division of Cardiology, “Tor Vergata” University Hospital, Viale Oxford 81, 00133 Rome, Italy; (G.M.); (D.B.); (M.D.L.); (G.R.)
- Department of Biomedicine and Prevention, “Tor Vergata” University of Rome, 00133 Rome, Italy
| |
Collapse
|
16
|
Mechanism of oxidized phospholipid-related inflammatory response in vascular ageing. Ageing Res Rev 2023; 86:101888. [PMID: 36806379 DOI: 10.1016/j.arr.2023.101888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 02/05/2023] [Accepted: 02/16/2023] [Indexed: 02/20/2023]
Abstract
Vascular ageing is an important factor in the morbidity and mortality of the elderly. Atherosclerosis is a characteristic disease of vascular ageing, which is closely related to the enhancement of vascular inflammation. Phospholipid oxidation products are important factors in inducing cellular inflammation. Through interactions with vascular cells and immune cells, they regulate intracellular signaling pathways, activate the expression of various cytokines, and affect cell behavior, such as metabolic level, proliferation, apoptosis, etc. Intervention in lipid metabolism and anti-inflammation are the two key pathways of drugs for the treatment of atherosclerosis. This review aims to sort out the signaling pathway of oxidized phospholipids-induced inflammatory factors in vascular cells and immune cells and the mechanism leading to changes in cell behavior, and summarize the therapeutic targets in the inflammatory signaling pathway for the development of atherosclerosis drugs.
Collapse
|
17
|
El Hajj S, Canabady-Rochelle L, Gaucher C. Nature-Inspired Bioactive Compounds: A Promising Approach for Ferroptosis-Linked Human Diseases? Molecules 2023; 28:molecules28062636. [PMID: 36985608 PMCID: PMC10059971 DOI: 10.3390/molecules28062636] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 03/02/2023] [Accepted: 03/08/2023] [Indexed: 03/16/2023] Open
Abstract
Ferroptosis is a type of cell death driven by iron overload and lipid peroxidation. It is considered a key mechanism in the development of various diseases such as atherosclerosis, Alzheimer, diabetes, cancer, and renal failure. The redox status of cells, such as the balance between intracellular oxidants (lipid peroxides, reactive oxygen species, free iron ions) and antioxidants (glutathione, glutathione Peroxidase 4), plays a major role in ferroptosis regulation and constitutes its principal biomarkers. Therefore, the induction and inhibition of ferroptosis are promising strategies for disease treatments such as cancer or neurodegenerative and cardiovascular diseases, respectively. Many drugs have been developed to exert ferroptosis-inducing and/or inhibiting reactions, such as erastin and iron-chelating compounds, respectively. In addition, many natural bioactive compounds have significantly contributed to regulating ferroptosis and ferroptosis-induced oxidative stress. Natural bioactive compounds are largely abundant in food and plants and have been for a long time, inspiring the development of various low-toxic therapeutic drugs. Currently, functional bioactive peptides are widely reported for their antioxidant properties and application in human disease treatment. The scientific evidence from biochemical and in vitro tests of these peptides strongly supports the existence of a relationship between their antioxidant properties (such as iron chelation) and ferroptosis regulation. In this review, we answer questions concerning ferroptosis milestones, its importance in physiopathology mechanisms, and its downstream regulatory mechanisms. We also address ferroptosis regulatory natural compounds as well as provide promising thoughts about bioactive peptides.
Collapse
Affiliation(s)
- Sarah El Hajj
- Université de Lorraine, CITHEFOR, F-54505 Vandoeuvre Les Nancy, France
- Université de Lorraine, CNRS, LRGP, F-54000 Nancy, France
| | | | - Caroline Gaucher
- Université de Lorraine, CITHEFOR, F-54505 Vandoeuvre Les Nancy, France
- Université de Lorraine, CNRS, IMoPA, F-54000 Nancy, France
- Correspondence:
| |
Collapse
|
18
|
Taylor JA, Hutchinson MA, Gearhart PJ, Maul RW. Antibodies in action: the role of humoral immunity in the fight against atherosclerosis. Immun Ageing 2022; 19:59. [PMID: 36461105 PMCID: PMC9717479 DOI: 10.1186/s12979-022-00316-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 11/21/2022] [Indexed: 12/03/2022]
Abstract
The sequestering of oxidation-modified low-density lipoprotein by macrophages results in the accumulation of fatty deposits within the walls of arteries. Necrosis of these cells causes a release of intercellular epitopes and the activation of the adaptive immune system, which we predict leads to robust autoantibody production. T cells produce cytokines that act in the plaque environment and further stimulate B cell antibody production. B cells in atherosclerosis meanwhile have a mixed role based on subclass. The current model is that B-1 cells produce protective IgM antibodies in response to oxidation-specific epitopes that work to control plaque formation, while follicular B-2 cells produce class-switched antibodies (IgG, IgA, and IgE) which exacerbate the disease. Over the course of this review, we discuss further the validation of these protective antibodies while evaluating the current dogma regarding class-switched antibodies in atherosclerosis. There are several contradictory findings regarding the involvement of class-switched antibodies in the disease. We hypothesize that this is due to antigen-specificity, and not simply isotype, being important, and that a closer evaluation of these antibodies' targets should be conducted. We propose that specific antibodies may have therapeutical potential in preventing and controlling plaque development within a clinical setting.
Collapse
Affiliation(s)
- Joshua A. Taylor
- grid.419475.a0000 0000 9372 4913Laboratory of Molecular Biology and Immunology, National Institute on Aging, NIH, Baltimore, MD USA ,grid.21107.350000 0001 2171 9311Graduate Program in Immunology, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Mark A. Hutchinson
- grid.419475.a0000 0000 9372 4913Laboratory of Molecular Biology and Immunology, National Institute on Aging, NIH, Baltimore, MD USA
| | - Patricia J. Gearhart
- grid.419475.a0000 0000 9372 4913Laboratory of Molecular Biology and Immunology, National Institute on Aging, NIH, Baltimore, MD USA
| | - Robert W. Maul
- grid.419475.a0000 0000 9372 4913Laboratory of Molecular Biology and Immunology, National Institute on Aging, NIH, Baltimore, MD USA
| |
Collapse
|
19
|
Istomin N, Härma MA, Akhi R, Nissinen AE, Savolainen MJ, Adeshara K, Lehto M, Groop PH, Koivukangas V, Hukkanen J, Hörkkö S. Total fecal IgA levels increase and natural IgM antibodies decrease after gastric bypass surgery. APMIS 2022; 130:637-646. [PMID: 35959517 PMCID: PMC9805076 DOI: 10.1111/apm.13268] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 08/09/2022] [Indexed: 01/10/2023]
Abstract
Obesity is associated with low-grade inflammation and increased systemic oxidative stress. Roux-en-Y gastric bypass (RYGB) surgery is known to ameliorate the obesity-induced metabolic dysfunctions. We aimed to study the levels of natural antibodies in feces, before and 6 months after RYGB surgery in obese individuals with and without type 2 diabetes (T2D). Sixteen individuals with T2D and 14 non-diabetic (ND) individuals were operated. Total IgA, IgG and IgM antibody levels and specific antibodies to oxidized low-density lipoprotein (oxLDL), malondialdehyde-acetaldehyde adducts (MAA adducts), Porphyromonas gingivalis gingipain A hemagglutinin domain (Rgp44) and phosphocholine (PCho) were measured using chemiluminescence immunoassay. Total fecal IgA was elevated, while total IgM and IgG were not affected by the surgery. Fecal natural IgM specific to oxLDL decreased significantly in both T2D and ND individuals, while fecal IgM to Rgp44 and PCho decreased significantly in T2D individuals. A decrease in IgG to MAA-LDL, Rgp44 and PCho was detected. RYGB surgery increases the levels of total fecal IgA and decreases fecal natural IgG and IgM antibodies specific to oxLDL. Natural antibodies and IgA are important in maintaining the normal gut homeostasis and first-line defense against microbes, and their production is markedly altered with RYGB surgery.
Collapse
Affiliation(s)
- Natalie Istomin
- Medical Microbiology and Immunology, Research Unit of Biomedicine, University of Oulu, Oulu, Finland.,Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland.,Nordlab, Oulu University Hospital, Oulu, Finland
| | - Mari-Anne Härma
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland.,Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Clinical and Molecular Metabolism, Faculty of Medicine Research Programs, University of Helsinki, Helsinki, Finland
| | - Ramin Akhi
- Medical Microbiology and Immunology, Research Unit of Biomedicine, University of Oulu, Oulu, Finland.,Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Antti E Nissinen
- Medical Microbiology and Immunology, Research Unit of Biomedicine, University of Oulu, Oulu, Finland
| | - Markku J Savolainen
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland.,Research Unit of Internal Medicine and Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Krishna Adeshara
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland.,Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Clinical and Molecular Metabolism, Faculty of Medicine Research Programs, University of Helsinki, Helsinki, Finland
| | - Markku Lehto
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland.,Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Clinical and Molecular Metabolism, Faculty of Medicine Research Programs, University of Helsinki, Helsinki, Finland
| | - Per-Henrik Groop
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland.,Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Clinical and Molecular Metabolism, Faculty of Medicine Research Programs, University of Helsinki, Helsinki, Finland
| | - Vesa Koivukangas
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland.,Department of Surgery, Oulu University Hospital, Oulu, Finland
| | - Janne Hukkanen
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland.,Research Unit of Internal Medicine and Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Sohvi Hörkkö
- Medical Microbiology and Immunology, Research Unit of Biomedicine, University of Oulu, Oulu, Finland.,Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| |
Collapse
|
20
|
Alic L, Binder CJ, Papac-Milicevic N. The OSE complotype and its clinical potential. Front Immunol 2022; 13:1010893. [PMID: 36248824 PMCID: PMC9561429 DOI: 10.3389/fimmu.2022.1010893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 09/12/2022] [Indexed: 11/17/2022] Open
Abstract
Cellular death, aging, and tissue damage trigger inflammation that leads to enzymatic and non-enzymatic lipid peroxidation of polyunsaturated fatty acids present on cellular membranes and lipoproteins. This results in the generation of highly reactive degradation products, such as malondialdehyde (MDA) and 4-hydroxynonenal (4-HNE), that covalently modify free amino groups of proteins and lipids in their vicinity. These newly generated neoepitopes represent a unique set of damage-associated molecular patterns (DAMPs) associated with oxidative stress termed oxidation-specific epitopes (OSEs). OSEs are enriched on oxidized lipoproteins, microvesicles, and dying cells, and can trigger sterile inflammation. Therefore, prompt recognition and removal of OSEs is required to maintain the homeostatic balance. This is partially achieved by various humoral components of the innate immune system, such as natural IgM antibodies, pentraxins and complement components that not only bind OSEs but in some cases modulate their pro-inflammatory potential. Natural IgM antibodies are potent complement activators, and 30% of them recognize OSEs such as oxidized phosphocholine (OxPC-), 4-HNE-, and MDA-epitopes. Furthermore, OxPC-epitopes can bind the complement-activating pentraxin C-reactive protein, while MDA-epitopes are bound by C1q, C3a, complement factor H (CFH), and complement factor H-related proteins 1, 3, 5 (FHR-1, FHR-3, FHR-5). In addition, CFH and FHR-3 are recruited to 2-(ω-carboxyethyl)pyrrole (CEP), and full-length CFH also possesses the ability to attenuate 4-HNE-induced oxidative stress. Consequently, alterations in the innate humoral defense against OSEs predispose to the development of diseases associated with oxidative stress, as shown for the prototypical OSE, MDA-epitopes. In this mini-review, we focus on the mechanisms of the accumulation of OSEs, the pathophysiological consequences, and the interactions between different OSEs and complement components. Additionally, we will discuss the clinical potential of genetic variants in OSE-recognizing complement proteins – the OSE complotype - in the risk estimation of diseases associated with oxidative stress.
Collapse
Affiliation(s)
- Lejla Alic
- Department of Medical Biochemistry, Faculty of Medicine, University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Christoph J. Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Nikolina Papac-Milicevic
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
- *Correspondence: Nikolina Papac-Milicevic,
| |
Collapse
|
21
|
Srikakulapu P, Pattarabanjird T, Upadhye A, Bontha SV, Osinski V, Marshall MA, Garmey J, Deroissart J, Prohaska TA, Witztum JL, Binder CJ, Holodick NE, Rothstein TL, McNamara CA. B-1b Cells Have Unique Functional Traits Compared to B-1a Cells at Homeostasis and in Aged Hyperlipidemic Mice With Atherosclerosis. Front Immunol 2022; 13:909475. [PMID: 35935999 PMCID: PMC9353528 DOI: 10.3389/fimmu.2022.909475] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 06/02/2022] [Indexed: 11/30/2022] Open
Abstract
Immunoglobulin M (IgM) to oxidation specific epitopes (OSE) are inversely associated with atherosclerosis in mice and humans. The B-1b subtype of B-1 cells secrete IgM to OSE, and unlike B-1a cells, are capable of long-lasting IgM memory. What attributes make B-1b cells different than B-1a cells is unknown. Our objectives were to determine how B-1b cells produce more IgM compared to B-1a cells at homeostatic condition and to see the differences in the B-1a and B-1b cell distribution and IgM CDR-H3 sequences in mice with advanced atherosclerosis. Here, in-vivo studies demonstrated greater migration to spleen, splenic production of IgM and plasma IgM levels in ApoE-/-Rag1-/- mice intraperitoneally injected with equal numbers of B-1b compared to B-1a cells. Bulk RNA seq analysis and flow cytometry of B-1a and B-1b cells identified CCR6 as a chemokine receptor more highly expressed on B-1b cells compared to B-1a. Knockout of CCR6 resulted in reduced B-1b cell migration to the spleen. Moreover, B-1b cell numbers were significantly higher in spleen of aged atherosclerotic ApoE-/- mice compared to young ApoE-/- mice. Single cell sequencing results of IgHM in B-1a and B-1b cells from peritoneal cavity and spleen of atherosclerotic aged ApoE-/- mice revealed significantly more N additions at the V-D and D-J junctions, greater diversity in V region usage and CDR-H3 sequences in B-1b compared to B-1a cells. In summary, B-1b cells demonstrated enhanced CCR6-mediated splenic migration, IgM production, and IgM repertoire diversification compared to B-1a cells. These findings suggest that potential strategies to selectively augment B-1b cell numbers and splenic trafficking could lead to increased and more diverse IgM targeting OSE to limit atherosclerosis.
Collapse
Affiliation(s)
- Prasad Srikakulapu
- Carter Immunology Center, University of Virginia, Charlottesville, VA, United States,*Correspondence: Prasad Srikakulapu, ; Coleen A. McNamara,
| | | | - Aditi Upadhye
- Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States
| | - Sai Vineela Bontha
- Carter Immunology Center, University of Virginia, Charlottesville, VA, United States
| | - Victoria Osinski
- Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States
| | - Melissa A. Marshall
- Carter Immunology Center, University of Virginia, Charlottesville, VA, United States
| | - James Garmey
- Carter Immunology Center, University of Virginia, Charlottesville, VA, United States
| | - Justine Deroissart
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Thomas A. Prohaska
- Department of Medicine, University of California San Diego, La Jolla, CA, United States
| | - Joseph L. Witztum
- Department of Medicine, University of California San Diego, La Jolla, CA, United States
| | - Christoph J. Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Nichol E. Holodick
- Center for Immunobiology and Department of Investigative Medicine, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, United States
| | - Thomas L. Rothstein
- Center for Immunobiology and Department of Investigative Medicine, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, United States
| | - Coleen A. McNamara
- Carter Immunology Center, University of Virginia, Charlottesville, VA, United States,Cardiovascular Division, Department of Medicine, University of Virginia, Charlottesville, VA, United States,*Correspondence: Prasad Srikakulapu, ; Coleen A. McNamara,
| |
Collapse
|
22
|
Upchurch CM, Yeudall S, Pavelec CM, Merk D, Greulich J, Manjegowda M, Raghavan SS, Bochkis IM, Scott MM, Perez-Reyes E, Leitinger N. Targeting oxidized phospholipids by AAV-based gene therapy in mice with established hepatic steatosis prevents progression to fibrosis. SCIENCE ADVANCES 2022; 8:eabn0050. [PMID: 35857497 PMCID: PMC9286512 DOI: 10.1126/sciadv.abn0050] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 06/03/2022] [Indexed: 05/06/2023]
Abstract
Oxidized phosphatidylcholines (OxPCs) are implicated in chronic tissue damage. Hyperlipidemic LDL-R--deficient mice transgenic for an OxPC-recognizing IgM fragment (scFv-E06) are protected against nonalcoholic fatty liver disease (NAFLD). To examine the effect of OxPC elimination at different stages of NAFLD progression, we used cre-dependent, adeno-associated virus serotype 8-mediated expression of the single-chain variable fragment of E06 (AAV8-scFv-E06) in hepatocytes of albumin-cre mice. AAV8-induced expression of scFv-E06 at the start of FPC diet protected mice from developing hepatic steatosis. Independently, expression of scFv-E06 in mice with established steatosis prevented the progression to hepatic fibrosis. Mass spectrometry-based oxophospho-lipidomics identified individual OxPC species that were reduced by scFv-E06 expression. In vitro, identified OxPC species dysregulated mitochondrial metabolism and gene expression in hepatocytes and hepatic stellate cells. We demonstrate that individual OxPC species independently affect disease initiation and progression from hepatic steatosis to steatohepatitis, and that AAV-mediated expression of scFv-E06 is an effective therapeutic intervention.
Collapse
Affiliation(s)
- Clint M. Upchurch
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA 22904, USA
| | - Scott Yeudall
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA 22904, USA
| | - Caitlin M. Pavelec
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA 22904, USA
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22904, USA
| | - Dennis Merk
- Environmentally-Induced Cardiovascular Degeneration, Clinical Chemistry and Laboratory Diagnostics, Medical Faculty, University Hospital and Heinrich-Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Jan Greulich
- Environmentally-Induced Cardiovascular Degeneration, Clinical Chemistry and Laboratory Diagnostics, Medical Faculty, University Hospital and Heinrich-Heine University Düsseldorf, 40225 Düsseldorf, Germany
- IUF-Leibniz Research Institute for Environmental Medicine, 40225 Düsseldorf, Germany
| | - Mohan Manjegowda
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA 22904, USA
| | - Shyam S. Raghavan
- Department of Pathology, University of Virginia, Charlottesville, VA 22904, USA
| | - Irina M. Bochkis
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA 22904, USA
| | - Michael M. Scott
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA 22904, USA
| | - Edward Perez-Reyes
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA 22904, USA
| | - Norbert Leitinger
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA 22904, USA
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22904, USA
| |
Collapse
|
23
|
Nedunchezhiyan U, Varughese I, Sun AR, Wu X, Crawford R, Prasadam I. Obesity, Inflammation, and Immune System in Osteoarthritis. Front Immunol 2022; 13:907750. [PMID: 35860250 PMCID: PMC9289681 DOI: 10.3389/fimmu.2022.907750] [Citation(s) in RCA: 187] [Impact Index Per Article: 62.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 06/10/2022] [Indexed: 12/12/2022] Open
Abstract
Obesity remains the most important risk factor for the incidence and progression of osteoarthritis (OA). The leading cause of OA was believed to be overloading the joints due to excess weight which in turn leads to the destruction of articular cartilage. However, recent studies have proved otherwise, various other factors like adipose deposition, insulin resistance, and especially the improper coordination of innate and adaptive immune responses may lead to the initiation and progression of obesity-associated OA. It is becoming increasingly evident that multiple inflammatory cells are recruited into the synovial joint that serves an important role in pathological changes in the synovial joint. Polarization of macrophages and macrophage-produced mediators are extensively studied and linked to the inflammatory and destructive responses in the OA synovium and cartilage. However, the role of other major innate immune cells such as neutrophils, eosinophils, and dendritic cells in the pathogenesis of OA has not been fully evaluated. Although cells of the adaptive immune system contribute to the pathogenesis of obesity-induced OA is still under exploration, a quantity of literature indicates OA synovium has an enriched population of T cells and B cells compared with healthy control. The interplay between a variety of immune cells and other cells that reside in the articular joints may constitute a vicious cycle, leading to pathological changes of the articular joint in obese individuals. This review addresses obesity and the role of all the immune cells that are involved in OA and summarised animal studies and human trials and knowledge gaps between the studies have been highlighted. The review also touches base on the interventions currently in clinical trials, different stages of the testing, and their shortcomings are also discussed to understand the future direction which could help in understanding the multifactorial aspects of OA where inflammation has a significant function.
Collapse
Affiliation(s)
- Udhaya Nedunchezhiyan
- Centre for Biomedical Technologies, Faculty of Engineering, Queensland University of Technology, Brisbane, QLD, Australia
| | - Ibin Varughese
- Centre for Biomedical Technologies, Faculty of Engineering, Queensland University of Technology, Brisbane, QLD, Australia
| | - Antonia RuJia Sun
- Centre for Biomedical Technologies, Faculty of Engineering, Queensland University of Technology, Brisbane, QLD, Australia
| | - Xiaoxin Wu
- Centre for Biomedical Technologies, Faculty of Engineering, Queensland University of Technology, Brisbane, QLD, Australia
- Department of Orthopedic Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ross Crawford
- Orthopedic Department, The Prince Charles Hospital, Brisbane, QLD, Australia
| | - Indira Prasadam
- Centre for Biomedical Technologies, Faculty of Engineering, Queensland University of Technology, Brisbane, QLD, Australia
- *Correspondence: Indira Prasadam,
| |
Collapse
|
24
|
Jaiswal V, Ang SP, Lnu K, Ishak A, Pokhrel NB, Chia JE, Hajra A, Biswas M, Matetic A, Dhatt R, Mamas MA. Effect of Pneumococcal Vaccine on Mortality and Cardiovascular Outcomes: A Systematic Review and Meta-Analysis. J Clin Med 2022; 11:3799. [PMID: 35807082 PMCID: PMC9267914 DOI: 10.3390/jcm11133799] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/27/2022] [Accepted: 06/27/2022] [Indexed: 11/24/2022] Open
Abstract
Various studies have suggested the possible cardiovascular (CV) protective effects of the pneumococcal vaccine (PV). Therefore, we conducted a meta-analysis to assess the association between recipients of PV with mortality and CV outcomes among patients with and without established cardiovascular disease. We performed a systematic literature search in PubMed, Embase, and Scopus for studies evaluating the effect of PV on mortality and CV outcomes. A total of 15 studies with 347,444 patients were included in the meta-analysis: 111,784 patients received PV (32%) and 235,660 patients were in the unvaccinated group (68%). Recipients of PV were associated with decreased all-cause mortality (HR, 0.76 (95% CI: 0.66 to 0.87), p < 0.001). PV was associated with a decrease in the incidence of myocardial infarction (MI) (HR, 0.73 (95% CI: 0.56−0.96), p = 0.02), without significant reduction in CV mortality (HR, 0.87 (95% CI: 0.72−1.07), p = 0.18) and stroke (HR, 1.01 (95% CI: 0.93−1.10), p = 0.82). Our study found PV was associated with decreased risk of all-cause mortality and MI. Future RCTs will be necessary to confirm benefits associated with receipt of PV.
Collapse
Affiliation(s)
- Vikash Jaiswal
- Department of Medicine, Larkin Community Hospital, South Miami, FL 33143, USA;
| | - Song Peng Ang
- Department of Internal Medicine, Rutgers Health/Community Medical Center, Toms River, NJ 08755, USA;
| | - Kriti Lnu
- Department of Internal Medicine, UPMC Harrisburg, Harrisburg, PA 17105, USA;
| | - Angela Ishak
- School of Medicine, European University Cyprus, Nicosia 2404, Cyprus;
| | | | - Jia Ee Chia
- School of Medicine, International Medical University, Kuala Lumpur 57000, Malaysia;
| | - Adrija Hajra
- Department of Internal Medicine, Jacobi Medical Center/Albert Einstein College of Medicine, The Bronx, NY 10461, USA;
| | - Monodeep Biswas
- Division of Cardiology, Wellspan Cardiology, Lancaster, PA 17602, USA;
| | - Andrija Matetic
- Department of Cardiology, University Hospital of Split, 21000 Split, Croatia;
- Keele Cardiovascular Research Group, Centre for Prognosis Research, Keele University, Keele ST5 5BG, UK
| | - Ravinder Dhatt
- Department of Internal Medicine, UPMC Harrisburg, Harrisburg, PA 17105, USA;
| | - Mamas A. Mamas
- Keele Cardiovascular Research Group, Centre for Prognosis Research, Keele University, Keele ST5 5BG, UK
| |
Collapse
|
25
|
Koschinsky ML, Boffa MB. Oxidized phospholipid modification of lipoprotein(a): Epidemiology, biochemistry and pathophysiology. Atherosclerosis 2022; 349:92-100. [DOI: 10.1016/j.atherosclerosis.2022.04.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/22/2022] [Accepted: 04/01/2022] [Indexed: 02/05/2023]
|
26
|
Dong Y, Yong VW. Oxidized phospholipids as novel mediators of neurodegeneration. Trends Neurosci 2022; 45:419-429. [PMID: 35393134 DOI: 10.1016/j.tins.2022.03.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 02/19/2022] [Accepted: 03/08/2022] [Indexed: 10/18/2022]
Abstract
Neurodegeneration drives the progression of many neurological diseases. Inflammation and oxidative stress occurring in the CNS promote lipid peroxidation, leading to the generation of oxidized phospholipids such as oxidized phosphatidylcholines (OxPCs). OxPCs have been proposed as biomarkers of oxidative stress, where their detection in lesions in multiple sclerosis (MS), frontotemporal lobe dementia, spinal cord injury, and amyotrophic lateral sclerosis (ALS) implies that oxidative insult had occurred. However, recent findings highlight OxPCs as potent neurotoxic species requiring neutralization by microglia. Here, we summarize the science of OxPCs, including lessons from non-CNS diseases. We discuss the potential of OxPCs as common drivers of injury across neurological conditions and encourage investigations of OxPCs as novel neurotoxins.
Collapse
Affiliation(s)
- Yifei Dong
- Hotchkiss Brain Institute and the Department of Clinical Neuroscience, University of Calgary, Calgary, Alberta, Canada
| | - V Wee Yong
- Hotchkiss Brain Institute and the Department of Clinical Neuroscience, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
27
|
Nettersheim FS, Picard FSR, Hoyer FF, Winkels H. Immunotherapeutic Strategies in Cancer and Atherosclerosis-Two Sides of the Same Coin. Front Cardiovasc Med 2022; 8:812702. [PMID: 35097027 PMCID: PMC8792753 DOI: 10.3389/fcvm.2021.812702] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 12/20/2021] [Indexed: 11/16/2022] Open
Abstract
The development and clinical approval of immunotherapies has revolutionized cancer therapy. Although the role of adaptive immunity in atherogenesis is now well-established and several immunomodulatory strategies have proven beneficial in preclinical studies, anti-atherosclerotic immunotherapies available for clinical application are not available. Considering that adaptive immune responses are critically involved in both carcinogenesis and atherogenesis, immunotherapeutic approaches for the treatment of cancer and atherosclerosis may exert undesirable but also desirable side effects on the other condition, respectively. For example, the high antineoplastic efficacy of immune checkpoint inhibitors, which enhance effector immune responses against tumor cells by blocking co-inhibitory molecules, was recently shown to be constrained by substantial proatherogenic properties. In this review, we outline the specific role of immune responses in the development of cancer and atherosclerosis. Furthermore, we delineate how current cancer immunotherapies affect atherogenesis and discuss whether anti-atherosclerotic immunotherapies may similarly have an impact on carcinogenesis.
Collapse
Affiliation(s)
- Felix Sebastian Nettersheim
- Department of Cardiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Felix Simon Ruben Picard
- Department of Cardiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Friedrich Felix Hoyer
- Department of Cardiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Holger Winkels
- Department of Cardiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
28
|
Osinski V, Srikakulapu P, Haider YM, Marshall MA, Ganta VC, Annex BH, McNamara CA. Loss of Id3 (Inhibitor of Differentiation 3) Increases the Number of IgM-Producing B-1b Cells in Ischemic Skeletal Muscle Impairing Blood Flow Recovery During Hindlimb Ischemia. Arterioscler Thromb Vasc Biol 2022; 42:6-18. [PMID: 34809449 PMCID: PMC8702457 DOI: 10.1161/atvbaha.120.315501] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
OBJECTIVE Neovascularization can maintain and even improve tissue perfusion in the setting of limb ischemia during peripheral artery disease. The molecular and cellular mechanisms mediating this process are incompletely understood. We investigate the potential role(s) for Id3 (inhibitor of differentiation 3) in regulating blood flow in a murine model of hindlimb ischemia (HLI). Approach and Results: HLI was modeled through femoral artery ligation and resection and blood flow recovery was quantified by laser Doppler perfusion imaging. Mice with global Id3 deletion had significantly impaired perfusion recovery at 14 and 21 days of HLI. Endothelial- or myeloid cell-specific deletion of Id3 revealed no effect on perfusion recovery while B-cell-specific knockout of Id3 (Id3BKO) revealed a significant attenuation of perfusion recovery. Flow cytometry revealed no differences in ischemia-induced T cells or myeloid cell numbers at 7 days of HLI, yet there was a significant increase in B-1b cells in Id3BKO. Consistent with these findings, ELISA (enzyme-linked immunoassay) demonstrated increases in skeletal muscle and plasma IgM. In vitro experiments demonstrated reduced proliferation and increased cell death when endothelial cells were treated with conditioned media from IgM-producing B-1b cells and tibialis anterior muscles in Id3BKO mice showed reduced density of total CD31+ and αSMA+CD31+ vessels. CONCLUSIONS This study is the first to demonstrate a role for B-cell-specific Id3 in maintaining blood flow recovery during HLI. Results suggest a role for Id3 in promoting blood flow during HLI and limiting IgM-expressing B-1b cell expansion. These findings present new mechanisms to investigate in peripheral artery disease pathogenesis.
Collapse
Affiliation(s)
- Victoria Osinski
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia 22908
- Department of Pathology, University of Virginia, Charlottesville, Virginia 22908
| | - Prasad Srikakulapu
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, Virginia 22908
| | - Young Min Haider
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, Virginia 22908
| | - Melissa A. Marshall
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, Virginia 22908
| | - Vijay C. Ganta
- Vascular Biology Center, Augusta University, Augusta, Georgia 30912
| | - Brian H. Annex
- Vascular Biology Center, Augusta University, Augusta, Georgia 30912
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia 30912
| | - Coleen A. McNamara
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia 22908
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, Virginia 22908
- Department of Medicine, Division of Cardiovascular Medicine, University of Virginia, Charlottesville, Virginia 22908
| |
Collapse
|
29
|
Demasi M, Augusto O, Bechara EJH, Bicev RN, Cerqueira FM, da Cunha FM, Denicola A, Gomes F, Miyamoto S, Netto LES, Randall LM, Stevani CV, Thomson L. Oxidative Modification of Proteins: From Damage to Catalysis, Signaling, and Beyond. Antioxid Redox Signal 2021; 35:1016-1080. [PMID: 33726509 DOI: 10.1089/ars.2020.8176] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Significance: The systematic investigation of oxidative modification of proteins by reactive oxygen species started in 1980. Later, it was shown that reactive nitrogen species could also modify proteins. Some protein oxidative modifications promote loss of protein function, cleavage or aggregation, and some result in proteo-toxicity and cellular homeostasis disruption. Recent Advances: Previously, protein oxidation was associated exclusively to damage. However, not all oxidative modifications are necessarily associated with damage, as with Met and Cys protein residue oxidation. In these cases, redox state changes can alter protein structure, catalytic function, and signaling processes in response to metabolic and/or environmental alterations. This review aims to integrate the present knowledge on redox modifications of proteins with their fate and role in redox signaling and human pathological conditions. Critical Issues: It is hypothesized that protein oxidation participates in the development and progression of many pathological conditions. However, no quantitative data have been correlated with specific oxidized proteins or the progression or severity of pathological conditions. Hence, the comprehension of the mechanisms underlying these modifications, their importance in human pathologies, and the fate of the modified proteins is of clinical relevance. Future Directions: We discuss new tools to cope with protein oxidation and suggest new approaches for integrating knowledge about protein oxidation and redox processes with human pathophysiological conditions. Antioxid. Redox Signal. 35, 1016-1080.
Collapse
Affiliation(s)
- Marilene Demasi
- Laboratório de Bioquímica e Biofísica, Instituto Butantan, São Paulo, Brazil
| | - Ohara Augusto
- Departamento de Bioquímica and Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Etelvino J H Bechara
- Departamento de Química Fundamental, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Renata N Bicev
- Departamento de Bioquímica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Fernanda M Cerqueira
- CENTD, Centre of Excellence in New Target Discovery, Instituto Butantan, São Paulo, Brazil
| | - Fernanda M da Cunha
- Departamento de Bioquímica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Ana Denicola
- Laboratorios Fisicoquímica Biológica-Enzimología, Facultad de Ciencias, Instituto de Química Biológica, Universidad de la República, Montevideo, Uruguay
| | - Fernando Gomes
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | - Sayuri Miyamoto
- Departamento de Bioquímica and Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Luis E S Netto
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | - Lía M Randall
- Laboratorios Fisicoquímica Biológica-Enzimología, Facultad de Ciencias, Instituto de Química Biológica, Universidad de la República, Montevideo, Uruguay
| | - Cassius V Stevani
- Departamento de Química Fundamental, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Leonor Thomson
- Laboratorios Fisicoquímica Biológica-Enzimología, Facultad de Ciencias, Instituto de Química Biológica, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
30
|
Ahmad F, Mitchell RD, Houben T, Palo A, Yadati T, Parnell AJ, Patel K, Shiri-Sverdlov R, Leake DS. Cysteamine Decreases Low-Density Lipoprotein Oxidation, Causes Regression of Atherosclerosis, and Improves Liver and Muscle Function in Low-Density Lipoprotein Receptor-Deficient Mice. J Am Heart Assoc 2021; 10:e017524. [PMID: 34493066 PMCID: PMC8649511 DOI: 10.1161/jaha.120.017524] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Background We have shown previously that low‐density lipoprotein (LDL) can be oxidized in the lysosomes of macrophages, that this oxidation can be inhibited by cysteamine, an antioxidant that accumulates in lysosomes, and that this drug decreases atherosclerosis in LDL receptor–deficient mice fed a high‐fat diet. We have now performed a regression study with cysteamine, which is of more relevance to the treatment of human disease. Methods and Results LDL receptor–deficient mice were fed a high‐fat diet to induce atherosclerotic lesions. They were then reared on chow diet and drinking water containing cysteamine or plain drinking water. Aortic atherosclerosis was assessed, and samples of liver and skeletal muscle were analyzed. There was no regression of atherosclerosis in the control mice, but cysteamine caused regression of between 32% and 56% compared with the control group, depending on the site of the lesions. Cysteamine substantially increased markers of lesion stability, decreased ceroid, and greatly decreased oxidized phospholipids in the lesions. The liver lipid levels and expression of cluster of differentiation 68, acetyl–coenzyme A acetyltransferase 2, cytochromes P450 (CYP)27, and proinflammatory cytokines and chemokines were decreased by cysteamine. Skeletal muscle function and oxidative fibers were increased by cysteamine. There were no changes in the plasma total cholesterol, LDL cholesterol, high‐density lipoprotein cholesterol, or triacylglycerol concentrations attributable to cysteamine. Conclusions Inhibiting the lysosomal oxidation of LDL in atherosclerotic lesions by antioxidants targeted at lysosomes causes the regression of atherosclerosis and improves liver and muscle characteristics in mice and might be a promising novel therapy for atherosclerosis in patients.
Collapse
Affiliation(s)
- Feroz Ahmad
- School of Biological Sciences University of Reading Reading UK
| | | | - Tom Houben
- Department of Molecular Genetics Maastricht University Maastricht the Netherlands
| | - Angela Palo
- Department of Molecular Genetics Maastricht University Maastricht the Netherlands
| | - Tulasi Yadati
- Department of Molecular Genetics Maastricht University Maastricht the Netherlands
| | | | - Ketan Patel
- School of Biological Sciences University of Reading Reading UK
| | - Ronit Shiri-Sverdlov
- Department of Molecular Genetics Maastricht University Maastricht the Netherlands
| | - David S Leake
- School of Biological Sciences University of Reading Reading UK
| |
Collapse
|
31
|
Promoting athero-protective immunity by vaccination with low density lipoprotein-derived antigens. Atherosclerosis 2021; 335:89-97. [PMID: 34462127 DOI: 10.1016/j.atherosclerosis.2021.08.033] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 08/17/2021] [Accepted: 08/19/2021] [Indexed: 11/23/2022]
Abstract
Immune responses activated by LDL particles that have been trapped and oxidized in the arterial wall play an important role in atherosclerosis. Some of these immune responses are protective by facilitating the removal of pro-inflammatory and toxic lipid species formed as result of LDL oxidation. However, should these protective immune responses be insufficient, other more potent pro-inflammatory immune responses instead contributing to disease progression will gradually become dominant. The importance of the balance between protective and pathogenic immunity is particularly apparent when it comes to the adaptive immune system where pro-inflammatory T helper 1 (Th1) type T cells aggravate atherosclerosis, while regulatory T cells (Tregs) have an opposing role. As oxidized LDL is a key autoantigen in atherosclerosis, it has become an interesting possibility that immune-modulatory therapy that favors the activity of apolipoprotein B peptide-specific Tregs could be developed into a novel treatment strategy for prevention/stabilization of atherosclerosis and ischemic cardiovascular events. Indeed, several such oxidized LDL tolerance vaccines have shown promising results in animal models of atherosclerosis. This review will discuss the experimental background for development of atherosclerosis vaccines based on LDL-derived antigens as well as the challenges involved in translating these findings into clinical application.
Collapse
|
32
|
Porsch F, Mallat Z, Binder CJ. Humoral immunity in atherosclerosis and myocardial infarction: from B cells to antibodies. Cardiovasc Res 2021; 117:2544-2562. [PMID: 34450620 DOI: 10.1093/cvr/cvab285] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/30/2021] [Accepted: 08/24/2021] [Indexed: 02/06/2023] Open
Abstract
Immune mechanisms are critically involved in the pathogenesis of atherosclerosis and its clinical manifestations. Associations of specific antibody levels and defined B cell subsets with cardiovascular disease activity in humans as well as mounting evidence from preclinical models demonstrate a role of B cells and humoral immunity in atherosclerotic cardiovascular disease. These include all aspects of B cell immunity, the generation of antigen-specific antibodies, antigen presentation and co-stimulation of T cells, as well as production of cytokines. Through their impact on adaptive and innate immune responses and the regulation of many other immune cells, B cells mediate both protective and detrimental effects in cardiovascular disease. Several antigens derived from (oxidised) lipoproteins, the vascular wall and classical autoantigens have been identified. The unique antibody responses they trigger and their relationship with atherosclerotic cardiovascular disease are reviewed. In particular, we focus on the different effector functions of specific IgM, IgG, and IgE antibodies and the cellular responses they trigger and highlight potential strategies to target B cell functions for therapy.
Collapse
Affiliation(s)
- Florentina Porsch
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Ziad Mallat
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, United Kingdom.,INSERM U970, Paris Cardiovascular Research Centre, Paris, France.,Unversité Paris Descartes, Sorbonne Paris Cité, Paris France
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
33
|
Ferroptosis: the potential value target in atherosclerosis. Cell Death Dis 2021; 12:782. [PMID: 34376636 PMCID: PMC8355346 DOI: 10.1038/s41419-021-04054-3] [Citation(s) in RCA: 112] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 06/08/2021] [Accepted: 06/16/2021] [Indexed: 12/14/2022]
Abstract
In advanced atherosclerosis (AS), defective function-induced cell death leads to the formation of the characteristic necrotic core and vulnerable plaque. The forms and mechanisms of cell death in AS have recently been elucidated. Among them, ferroptosis, an iron-dependent form of necrosis that is characterized by oxidative damage to phospholipids, promotes AS by accelerating endothelial dysfunction in lipid peroxidation. Moreover, disordered intracellular iron causes damage to macrophages, vascular smooth muscle cells (VSMCs), vascular endothelial cells (VECs), and affects many risk factors or pathologic processes of AS such as disturbances in lipid peroxidation, oxidative stress, inflammation, and dyslipidemia. However, the mechanisms through which ferroptosis initiates the development and progression of AS have not been established. This review explains the possible correlations between AS and ferroptosis, and provides a reliable theoretical basis for future studies on its mechanism.
Collapse
|
34
|
Muhammad K, Ayoub MA, Iratni R. Vascular Inflammation in Cardiovascular Disease: Is Immune System Protective or Bystander? Curr Pharm Des 2021; 27:2141-2150. [PMID: 33461451 DOI: 10.2174/1381612827666210118121952] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 12/15/2020] [Indexed: 11/22/2022]
Abstract
Cardiovascular disease (CVD) is one of the leading causes of death worldwide. Chronic atherosclerosis induced vascular inflammation and perturbation of lipid metabolism is believed to be a major cause of CVD. Interplay of innate and adaptive Immune system has been interwined with various risk factors associated with the initiation and progression of atherosclerosis in CVD. A large body of evidence indicates a correlation between immunity and atherosclerosis. Retention of plasma lipoproteins in arterial subendothelial wall triggers the T helper type 1 (Th1) cells and monocyte-derived macrophages to form atherosclerotic plaques. In the present review, we will discuss the pathogenesis of CVD in relation to atherosclerosis with a particular focus on pro-atherogenic role of immune cells. Recent findings have also suggested anti-atherogenic roles of different B cell subsets. Therapeutic approaches to target atherosclerosis risk factors have reduced the mortality, but a need exists for the novel therapies to treat arterial vascular inflammation. These insights into the immune pathogenesis of atherosclerosis can lead to new targeted therapeutics to abate cardiovascular mortality and morbidity.
Collapse
Affiliation(s)
- Khalid Muhammad
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Mohammed A Ayoub
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Rabah Iratni
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
35
|
Abstract
Atherosclerosis is the leading cause of acute cardiovascular events, and vascular calcification is an important pathological phenomenon in atherosclerosis. Recently, many studies have shown that immune cells are closely associated with the development of atherosclerosis and calcification, but there are many conflicting viewpoints because of immune system complications, such as the pro-atherosclerotic and atheroprotective effects of regulatory B cells (Bregs), T helper type 2 (Th2) cells and T helper type 17 (Th17) cells. In this review, we summarize the studies on the roles of immune cells, especially lymphocytes and macrophages, in atherosclerotic calcification. Furthermore, we prepared graphs showing the relationship between T cells, B cells and macrophages and atherosclerotic calcification. Finally, we highlight some potential issues that are closely associated with the function of immune cells in atherosclerotic calcification. Based on current research results, this review summarizes the relationship between immune cells and atherosclerotic calcification, and it will be beneficial to understand the relationship of immune cells and atherosclerotic calcification.
Collapse
Affiliation(s)
- Jingsong Cao
- Clinical Medicine Research Center, 574417The First Affiliated Hospital of University of South China, Hengyang, China.,Department of Endocrinology and Metabolism, 574417The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Xuyu Zu
- Clinical Medicine Research Center, 574417The First Affiliated Hospital of University of South China, Hengyang, China
| | - Jianghua Liu
- Clinical Medicine Research Center, 574417The First Affiliated Hospital of University of South China, Hengyang, China.,Department of Endocrinology and Metabolism, 574417The First Affiliated Hospital of University of South China, Hengyang, Hunan, China.,Department of Metabolism and Endocrinology, 574417The First Affiliated Hospital of University of South China, Hengyang, China
| |
Collapse
|
36
|
de Vries MR, Ewing MM, de Jong RCM, MacArthur MR, Karper JC, Peters EAB, Nordzell M, Karabina SAP, Sexton D, Dahlbom I, Bergman A, Mitchell JR, Frostegård J, Kuiper J, Ninio E, Jukema JW, Pettersson K, Quax PHA. Identification of IgG1 isotype phosphorylcholine antibodies for the treatment of inflammatory cardiovascular diseases. J Intern Med 2021; 290:141-156. [PMID: 33342002 PMCID: PMC8359267 DOI: 10.1111/joim.13234] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 11/23/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND Phosphorylcholine (PC) is an important pro-inflammatory damage-associated molecular pattern. Previous data have shown that natural IgM anti-PC protects against cardiovascular disease. We aimed to develop a monoclonal PC IgG antibody with anti-inflammatory and anti-atherosclerotic properties. METHODS Using various techniques PC antibodies were validated and optimized. In vivo testing was performed in a femoral artery cuff model in ApoE3*Leiden mice. Safety studies are performed in rats and cynomolgus monkeys. RESULTS A chimeric anti-PC (PC-mAb(T15), consisting of a human IgG1 Fc and a mouse T15/E06 Fab) was produced, and this was shown to bind specifically to epitopes in human atherosclerotic tissues. The cuff model results in rapid induction of inflammatory genes and altered expression of genes associated with ER stress and choline metabolism in the lesions. Treatment with PC-mAb(T15) reduced accelerated atherosclerosis via reduced expression of endoplasmic reticulum stress markers and CCL2 production. Recombinant anti-PC Fab fragments were identified by phage display and cloned into fully human IgG1 backbones creating a human monoclonal IgG1 anti-PC (PC-mAbs) that specifically bind PC, apoptotic cells and oxLDL. Based on preventing macrophage oxLDL uptake and CCL2 production, four monoclonal PC-mAbs were selected, which to various extent reduced vascular inflammation and lesion development. Additional optimization and validation of two PC-mAb antibodies resulted in selection of PC-mAb X19-A05, which inhibited accelerated atherosclerosis. Clinical grade production of this antibody (ATH3G10) significantly attenuated vascular inflammation and accelerated atherosclerosis and was tolerated in safety studies in rats and cynomolgus monkeys. CONCLUSIONS Chimeric anti-PCs can prevent accelerated atherosclerosis by inhibiting vascular inflammation directly and through reduced macrophage oxLDL uptake resulting in decreased lesions. PC-mAb represents a novel strategy for cardiovascular disease prevention.
Collapse
Affiliation(s)
- M. R. de Vries
- From theDeptartment of SurgeryLUMCLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLUMCLeidenThe Netherlands
| | - M. M. Ewing
- From theDeptartment of SurgeryLUMCLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLUMCLeidenThe Netherlands
- Deptartment of CardiologyLUMCLeidenThe Netherlands
| | - R. C. M. de Jong
- From theDeptartment of SurgeryLUMCLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLUMCLeidenThe Netherlands
| | - M. R. MacArthur
- Department of Molecular MetabolismHarvard T.H. Chan School of Public HealthBostonMAUSA
| | - J. C. Karper
- From theDeptartment of SurgeryLUMCLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLUMCLeidenThe Netherlands
| | - E. A. B. Peters
- From theDeptartment of SurgeryLUMCLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLUMCLeidenThe Netherlands
| | | | - S. A. P. Karabina
- INSERM UMR_S 933Hôpital Armand‐TrousseauSorbonne UniversitéParisFrance
| | | | - I. Dahlbom
- Dept. of MedicineKarolinska University Hospital Huddinge and Karolinska InstitutetStockholmSweden
| | | | - J. R. Mitchell
- Department of Molecular MetabolismHarvard T.H. Chan School of Public HealthBostonMAUSA
| | - J. Frostegård
- Dept. of MedicineKarolinska University Hospital Huddinge and Karolinska InstitutetStockholmSweden
| | - J. Kuiper
- Division of BioTherapeuticsLACDRLeidenThe Netherlands
| | - E. Ninio
- INSERM UMR_S 1166‐ICANGenomics and Pathophysiology of Cardiovascular DiseasesInstitute of Cardiometabolism and NutritionPitié‐Salpêtrière HôpitalSorbonne UniversitéParisFrance
| | - J. W. Jukema
- Einthoven Laboratory for Experimental Vascular MedicineLUMCLeidenThe Netherlands
- Deptartment of CardiologyLUMCLeidenThe Netherlands
| | | | - P. H. A. Quax
- From theDeptartment of SurgeryLUMCLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLUMCLeidenThe Netherlands
| |
Collapse
|
37
|
Attie AD, Schueler KM, Keller MP, Mitok KA, Simonett SP, Hudkins KL, Mehrotra K, Graham MJ, Lee RG, Alpers CE. Reversal of hypertriglyceridemia in diabetic BTBR ob/ob mice does not prevent nephropathy. J Transl Med 2021; 101:935-941. [PMID: 33911188 PMCID: PMC9093019 DOI: 10.1038/s41374-021-00592-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 03/05/2021] [Accepted: 03/08/2021] [Indexed: 01/11/2023] Open
Abstract
The etiology of diabetic nephropathy in type 2 diabetes is multifactorial. Sustained hyperglycemia is a major contributor, but additional contributions come from the hypertension, obesity, and hyperlipidemia that are also commonly present in patients with type 2 diabetes and nephropathy. The leptin deficient BTBR ob/ob mouse is a model of type 2 diabetic nephropathy in which hyperglycemia, obesity, and hyperlipidemia, but not hypertension, are present. We have shown that reversal of the constellation of these metabolic abnormalities with leptin replacement can reverse the morphologic and functional manifestations of diabetic nephropathy. Here we tested the hypothesis that reversal specifically of the hypertriglyceridemia, using an antisense oligonucleotide directed against ApoC-III, an apolipoprotein that regulates the interactions of VLDL (very low density lipoproteins) with the LDL receptor, is sufficient to ameliorate the nephropathy of Type 2 diabetes. Antisense treatment resulted in reduction of circulating ApoC-III protein levels and resulted in substantial lowering of triglycerides to near-normal levels in diabetic mice versus controls. Antisense treatment did not ameliorate proteinuria or pathologic manifestations of diabetic nephropathy, including podocyte loss. These studies indicate that pathologic manifestations of diabetic nephropathy are unlikely to be reduced by lipid-lowering therapeutics alone, but does not preclude a role for such interventions to be used in conjunction with other therapeutics commonly employed in the treatment of diabetes and its complications.
Collapse
Affiliation(s)
- Alan D Attie
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA.
| | - Kathryn M Schueler
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Mark P Keller
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Kelly A Mitok
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Shane P Simonett
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Kelly L Hudkins
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Kunaal Mehrotra
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | | | | | - Charles E Alpers
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA.
| |
Collapse
|
38
|
Pluijmert NJ, de Jong RCM, de Vries MR, Pettersson K, Atsma DE, Jukema JW, Quax PHA. Phosphorylcholine antibodies restrict infarct size and left ventricular remodelling by attenuating the unreperfused post-ischaemic inflammatory response. J Cell Mol Med 2021; 25:7772-7782. [PMID: 34190404 PMCID: PMC8358891 DOI: 10.1111/jcmm.16662] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 04/30/2021] [Accepted: 05/08/2021] [Indexed: 12/19/2022] Open
Abstract
Phosphorylcholine is a pro‐inflammatory epitope exposed on apoptotic cells, and phosphorylcholine monoclonal immunoglobulin (Ig)G antibodies (PC‐mAb) have anti‐inflammatory properties. In this study, we hypothesize that PC‐mAb treatment reduces adverse cardiac remodelling and infarct size (IS) following unreperfused transmural myocardial infarction (MI). Unreperfused MI was induced by permanent ligation of the left anterior descending (LAD) coronary artery in hypercholesterolaemic APOE*3‐Leiden mice. Three weeks following MI, cardiac magnetic resonance (CMR) imaging showed a reduced LV end‐diastolic volume (EDV) by 21% and IS by 31% upon PC‐mAb treatment as compared to the vehicle control group. In addition, the LV fibrous content was decreased by 27% and LV wall thickness was better preserved by 47% as determined by histological analysis. Two days following MI, CCL2 concentrations, assessed by use of ELISA, were decreased by 81% and circulating monocytes by 64% as assessed by use of FACS analysis. Additionally, local leucocyte infiltration determined by immunohistological analysis showed a 62% decrease after three weeks. In conclusion, the local and systemic inflammatory responses are limited by PC‐mAb treatment resulting in restricted adverse cardiac remodelling and IS following unreperfused MI. This indicates that PC‐mAb holds promise as a therapeutic agent following MI limiting adverse cardiac remodelling.
Collapse
Affiliation(s)
- Niek J Pluijmert
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Rob C M de Jong
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Margreet R de Vries
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Douwe E Atsma
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Paul H A Quax
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
39
|
Akhi R, Nissinen AE, Wang C, Kyrklund M, Paju S, Mäntylä P, Buhlin K, Sinisalo J, Pussinen PJ, Hörkkö S. Salivary IgA antibody to malondialdehyde-acetaldehyde associates with mild periodontal pocket depth. Oral Dis 2021; 28:2285-2293. [PMID: 34124817 DOI: 10.1111/odi.13936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 05/03/2021] [Accepted: 05/30/2021] [Indexed: 10/21/2022]
Abstract
OBJECTIVE Oxidized epitopes such as malondialdehyde-acetaldehyde (MAA) play a crucial role in the progression of atherosclerosis through activation of the humoral immune response. The exact mechanism of the association between atherosclerosis and periodontal diseases is not fully understood. The aim of the current study is to evaluate the association of oral humoral immune response to oxidized epitopes with parameters of periodontal disease. MATERIALS AND METHODS The Parogene cohort consist of patients who have undergone coronary angiography due to cardiac symptoms. In this study, 423 patients were randomly selected for an extensive oral examination. Salivary Immunoglobulin A to oxidized epitopes and bacterial antigens was determined by chemiluminescence immunoassay. RESULTS In a binary logistic regression model adjusted with periodontal disease confounders, periodontal pocket depth (PPD) 4-5 mm associated with salivary IgA antibodies to MAA-LDL (p = 0.034), heat shock protein 60 of Aggregatibacter actinomycetemcomitans (p = 0.045), Porphyromonas gingivalis (p = 0.045), A. actinomycetemcomitans (p = 0.005), P. intermedia (p = 0.020), and total IgA (p = 0.003). CONCLUSIONS The current study shows the association of salivary IgA to MAA-LDL with PPD 4-5 mm in a cohort of patients with chronic coronary artery disease. Humoral immune cross-reactivation to oxidized epitopes such MAA-LDL could partly explain the link of periodontitis with systemic diseases.
Collapse
Affiliation(s)
- Ramin Akhi
- Medical Microbiology and Immunology, Research Unit of Biomedicine, University of Oulu, Oulu, Finland.,Medical Research Center, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Antti E Nissinen
- Medical Microbiology and Immunology, Research Unit of Biomedicine, University of Oulu, Oulu, Finland.,Medical Research Center, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Chunguang Wang
- Medical Microbiology and Immunology, Research Unit of Biomedicine, University of Oulu, Oulu, Finland.,Medical Research Center, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Mikael Kyrklund
- Medical Microbiology and Immunology, Research Unit of Biomedicine, University of Oulu, Oulu, Finland.,Medical Research Center, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Susanna Paju
- Oral and Maxillofacial Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Päivi Mäntylä
- Oral and Maxillofacial Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Institute of Dentistry, University of Eastern Finland, Kuopio, Finland
| | - Kåre Buhlin
- Oral and Maxillofacial Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Division of Periodontology, Department of Dental Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Juha Sinisalo
- HUCH Heart and Lung Center, Helsinki University Hospital, Helsinki, Finland
| | - Pirkko J Pussinen
- Oral and Maxillofacial Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Sohvi Hörkkö
- Medical Microbiology and Immunology, Research Unit of Biomedicine, University of Oulu, Oulu, Finland.,Medical Research Center, Oulu University Hospital and University of Oulu, Oulu, Finland
| |
Collapse
|
40
|
Sveen KA, Bech Holte K, Svanteson M, Hanssen KF, Nilsson J, Bengtsson E, Julsrud Berg T. Autoantibodies Against Methylglyoxal-Modified Apolipoprotein B100 and ApoB100 Peptide Are Associated With Less Coronary Artery Atherosclerosis and Retinopathy in Long-Term Type 1 Diabetes. Diabetes Care 2021; 44:1402-1409. [PMID: 33858856 PMCID: PMC8247486 DOI: 10.2337/dc20-2089] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 02/22/2021] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Methylglyoxal (MGO), a reactive aldehyde forming advanced glycation end products (AGEs), is increased in diabetes and recognized by the immune system, resulting in anti-AGE-specific autoantibodies. The association of these immune responses with macro- and microvascular complications in type 1 diabetes remains unclarified. We investigated associations between MGO-modified apolipoprotein B100 (apoB100) and apoB100 peptide 5 (MGO-p5) autoantibodies and coronary atherosclerosis and retinopathy in type 1 diabetes. RESEARCH DESIGN AND METHODS IgM and IgG against MGO-apoB100 and MGO-p5 were measured by ELISA in plasma from 103 subjects with type 1 diabetes and 63 control subjects (Dialong study) and in a replication cohort of 27 subjects with type 1 diabetes (Oslo study). Coronary atherosclerosis was assessed by computed tomography coronary angiography or intravascular ultrasound. Retinopathy was classified by retinal photos. RESULTS MGO-apoB100 IgM and MGO-p5 IgM levels were higher in subjects with diabetes with no coronary artery stenosis compared with subjects with significant stenosis (median [interquartile range]: 96.2 arbitrary units [AU] [71-126.8] vs. 54 AU [36.1-85.4], P = 0.003 for MGO-apoB100; and 77.4 AU [58-106] vs. 36.9 AU [28.9-57.4], P = 0.005 for MGO-p5). MGO-apoB100 IgM and MGO-p5 IgM were associated with less severe coronary stenosis after adjusting for confounders (odds ratio 0.2 [95% CI 0.05-0.6], P = 0.01; and 0.22 [0.06-0.75], P = 0.02). The inverse association of MGO-p5 IgM and coronary stenosis was confirmed in the replication cohort. Subjects with proliferative retinopathy had significantly lower MGO-apoB100 IgM and MGO-p5 IgM than those with background retinopathy. CONCLUSIONS Autoantibodies against AGE-modified apoB100 are inversely associated with coronary atherosclerosis and proliferative retinopathy, suggesting vascular protective effects of these autoantibodies in type 1 diabetes.
Collapse
Affiliation(s)
- Kari Anne Sveen
- Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, Oslo, Norway .,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Kristine Bech Holte
- Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, Oslo, Norway
| | - Mona Svanteson
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - Kristian F Hanssen
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Jan Nilsson
- Department of Clinical Sciences, Skåne University Hospital, Lund University, Malmø, Sweden
| | - Eva Bengtsson
- Department of Clinical Sciences, Skåne University Hospital, Lund University, Malmø, Sweden
| | - Tore Julsrud Berg
- Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
41
|
Hsu YC, Tsai IJ, Hsu H, Hsu PW, Cheng MH, Huang YL, Chen JH, Lei MH, Lin CY. Using Anti-Malondialdehyde Modified Peptide Autoantibodies to Import Machine Learning for Predicting Coronary Artery Stenosis in Taiwanese Patients with Coronary Artery Disease. Diagnostics (Basel) 2021; 11:diagnostics11060961. [PMID: 34073646 PMCID: PMC8229983 DOI: 10.3390/diagnostics11060961] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/19/2021] [Accepted: 05/24/2021] [Indexed: 02/02/2023] Open
Abstract
Machine learning (ML) algorithms have been applied to predicting coronary artery disease (CAD). Our purpose was to utilize autoantibody isotypes against four different unmodified and malondialdehyde (MDA)-modified peptides among Taiwanese with CAD and healthy controls (HCs) for CAD prediction. In this study, levels of MDA, MDA-modified protein (MDA-protein) adducts, and autoantibody isotypes against unmodified peptides and MDA-modified peptides were measured with enzyme-linked immunosorbent assay (ELISA). To improve the performance of ML, we used decision tree (DT), random forest (RF), and support vector machine (SVM) coupled with five-fold cross validation and parameters optimization. Levels of plasma MDA and MDA-protein adducts were higher in CAD patients than in HCs. IgM anti-IGKC76-99 MDA and IgM anti-A1AT284-298 MDA decreased the most in patients with CAD compared to HCs. In the experimental results of CAD prediction, the decision tree classifier achieved an area under the curve (AUC) of 0.81; the random forest classifier achieved an AUC of 0.94; the support vector machine achieved an AUC of 0.65 for differentiating between CAD patients with stenosis rates of 70% and HCs. In this study, we demonstrated that autoantibody isotypes imported into machine learning algorithms can lead to accurate models for clinical use.
Collapse
Affiliation(s)
- Yu-Cheng Hsu
- Cardiovascular Center, Lo-Hsu Medical Foundation Luodong Poh-Ai Hospital, Yilan 26546, Taiwan;
| | - I-Jung Tsai
- Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan;
| | - Hung Hsu
- Medical Quality Department, Lo-Hsu Medical Foundation Luodong Poh-Ai Hospital, Yilan 26546, Taiwan;
| | - Po-Wen Hsu
- Preventive Medical Center, Lo-Hsu Medical Foundation Luodong Poh-Ai Hospital, Yilan 26546, Taiwan;
| | - Ming-Hui Cheng
- Department of Laboratory Medicine, Lo-Hsu Medical Foundation Luodong Poh-Ai Hospital, Yilan 26546, Taiwan;
| | - Ying-Li Huang
- Section of Laboratory, Lo-Hsu Medical Foundation Lodong Poh-Ai Hospital, Yilan 26546, Taiwan;
| | - Jin-Hua Chen
- Graduate Institute of Data Science, College of Management, Taipei Medical University, Taipei 11031, Taiwan;
- Statistics Center, Institutional Research Center, Office of Data Science, Taipei Medical University, Taipei 11031, Taiwan
| | - Meng-Huan Lei
- Cardiovascular Center, Lo-Hsu Medical Foundation Luodong Poh-Ai Hospital, Yilan 26546, Taiwan;
- Correspondence: (M.-H.L.); (C.-Y.L.); Tel.: +886-3-9543131 (ext. 2162) (M.-H.L.); +886-2-27361661 (ext. 3326) (C.-Y.L.)
| | - Ching-Yu Lin
- Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan;
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- Correspondence: (M.-H.L.); (C.-Y.L.); Tel.: +886-3-9543131 (ext. 2162) (M.-H.L.); +886-2-27361661 (ext. 3326) (C.-Y.L.)
| |
Collapse
|
42
|
Beyond Lipoprotein(a) plasma measurements: Lipoprotein(a) and inflammation. Pharmacol Res 2021; 169:105689. [PMID: 34033878 PMCID: PMC9247870 DOI: 10.1016/j.phrs.2021.105689] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 05/18/2021] [Accepted: 05/19/2021] [Indexed: 12/20/2022]
Abstract
Genome wide association, epidemiological, and clinical studies have established high lipoprotein(a) [Lp(a)] as a causal risk factor for atherosclerotic cardiovascular disease (ASCVD). Lp(a) is an apoB100 containing lipoprotein covalently bound to apolipoprotein(a) [apo(a)], a glycoprotein. Plasma Lp(a) levels are to a large extent determined by genetics. Its link to cardiovascular disease (CVD) may be driven by its pro-inflammatory effects, of which its association with oxidized phospholipids (oxPL) bound to Lp(a) is the most studied. Various inflammatory conditions, such as rheumatoid arthritis (RA), systemic lupus erythematosus, acquired immunodeficiency syndrome, and chronic renal failure are associated with high Lp(a) levels. In cases of RA, high Lp(a) levels are reversed by interleukin-6 receptor (IL-6R) blockade by tocilizumab, suggesting a potential role for IL-6 in regulating Lp(a) plasma levels. Elevated levels of IL-6 and IL-6R polymorphisms are associated with CVD. Therapies aimed at lowering apo(a) and thereby reducing plasma Lp(a) levels are in clinical trials. Their results will determine if reductions in apo(a) and Lp(a) decrease cardiovascular outcomes. As we enter this new arena of available treatments, there is a need to improve our understanding of mechanisms. This review will focus on the role of Lp(a) in inflammation and CVD.
Collapse
|
43
|
Sadras V, Petri MA, Jones SR, Peterlin BL, Chatterjee S. Glycosphingolipid-associated β-1,4 galactosyltransferase is elevated in patients with systemic lupus erythematosus. Lupus Sci Med 2021; 7:7/1/e000368. [PMID: 32665303 PMCID: PMC7359192 DOI: 10.1136/lupus-2019-000368] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 05/30/2020] [Accepted: 06/04/2020] [Indexed: 12/13/2022]
Abstract
OBJECTIVE β-1,4 galactosyltransferase-V (β-1,4 GalT-V) is an enzyme that synthesises a glycosphingolipid known as lactosylceramide, which has been implicated in general inflammation and atherosclerosis. We asked if β-1,4 GalT-V was present at elevated levels in patients with SLE, a disease which is associated with increased risk of atherosclerosis. METHODS In this case-control observational study, serum samples were obtained from patients with SLE who are part of the Johns Hopkins Lupus Cohort. Control serum samples were obtained from healthy adult community members recruited from the Baltimore area. All serum samples (n=50 in the SLE group and n=50 in the healthy control group) were analysed with enzyme-linked immunoassays. These assays used antibodies raised against antigens that enabled us to measure the absorbance of oxidised phosphocholines per apolipoprotein B-100 (ox-PC/apoB) and the concentration of lipoprotein(a) (Lp(a)) and β-1,4 GalT-V. RESULTS Absorbance of ox-PC/apoB and concentrations of Lp(a) and β-1,4 GalT-V were significantly higher in the SLE serum samples as compared with the control serum (p<0.0001). CONCLUSIONS We conclude that patients with SLE have elevated levels of β-1,4 GalT-V and ox-PC, which have previously been recognised as risk factors for atherosclerosis.
Collapse
Affiliation(s)
- Vignesh Sadras
- Department of Pediatrics, Johns Hopkins University, Baltimore, Maryland, USA
| | - Michelle A Petri
- Division of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Steven Richard Jones
- Heart and Vascular Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Barbara Lee Peterlin
- Lancaster General Health Physicians, Penn Medicine, Lancaster, Pennsylvania, USA
| | - Subroto Chatterjee
- Division of Pediatric Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
44
|
Stamenkovic A, O'Hara KA, Nelson DC, Maddaford TG, Edel AL, Maddaford G, Dibrov E, Aghanoori M, Kirshenbaum LA, Fernyhough P, Aliani M, Pierce GN, Ravandi A. Oxidized phosphatidylcholines trigger ferroptosis in cardiomyocytes during ischemia-reperfusion injury. Am J Physiol Heart Circ Physiol 2021; 320:H1170-H1184. [PMID: 33513080 DOI: 10.1152/ajpheart.00237.2020] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 01/22/2021] [Indexed: 12/22/2022]
Abstract
Myocardial ischemia-reperfusion (I/R) injury increases the generation of oxidized phosphatidylcholines (OxPCs), which results in cell death. However, the mechanism by which OxPCs mediate cell death and cardiac dysfunction is largely unknown. The aim of this study was to determine the mechanisms by which OxPC triggers cardiomyocyte cell death during reperfusion injury. Adult rat ventricular cardiomyocytes were treated with increasing concentrations of various purified fragmented OxPCs. Cardiomyocyte viability, bioenergetic response, and calcium transients were determined in the presence of OxPCs. Five different fragmented OxPCs resulted in a decrease in cell viability, with 1-palmitoyl-2-(5'-oxo-valeroyl)-sn-glycero-3-phosphocholine (POVPC) and 1-palmitoyl-2-(9'-oxo-nonanoyl)-sn-glycero-3-phosphocholine (PONPC) having the most potent cardiotoxic effect in both a concentration and time dependent manner (P < 0.05). POVPC and PONPC also caused a significant decrease in Ca2+ transients and net contraction in isolated cardiomyocytes compared to vehicle treated control cells (P < 0.05). PONPC depressed maximal respiration rate (P < 0.01; 54%) and spare respiratory capacity (P < 0.01; 54.5%). Notably, neither caspase 3 activation or TUNEL staining was observed in cells treated with either POVPC or PONPC. Further, cardiac myocytes treated with OxPCs were indistinguishable from vehicle-treated control cells with respect to nuclear high-mobility group box protein 1 (HMGBP1) activity. However, glutathione peroxidase 4 activity was markedly suppressed in cardiomyocytes treated with POVPC and PONPC coincident with increased ferroptosis. Importantly, cell death induced by OxPCs could be suppressed by E06 Ab, directed against OxPCs or by ferrostatin-1, which bound the sn-2 aldehyde of POVPC during I/R. The findings of the present study demonstrate that oxidation of phosphatidylcholines during I/R generate bioactive phospholipid intermediates that disrupt mitochondrial bioenergetics and calcium transients and provoke wide spread cell death through ferroptosis. Neutralization of OxPC with E06 or with ferrostatin-1 prevents cell death during reperfusion. Our study demonstrates a novel signaling pathway that operationally links generation of OxPC during cardiac I/R to ferroptosis. Interventions designed to target OxPCs may prove beneficial in mitigating ferroptosis during I/R injury in individuals with ischemic heart disease.NEW & NOTEWORTHY Oxidized phosphatidylcholines (OxPC) generated during reperfusion injury are potent inducers of cardiomyocyte death. Our studies have shown that OxPCs exert this effect through a ferroptotic process that can be attenuated. A better understanding of the OxPC cell death pathway can prove a novel strategy for prevention of cell death during myocardial reperfusion injury.
Collapse
Affiliation(s)
- Aleksandra Stamenkovic
- Institute of Cardiovascular Sciences, Department of Physiology and Pathophysiology, St. Boniface Hospital, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| | - Kimberley A O'Hara
- Institute of Cardiovascular Sciences, Department of Physiology and Pathophysiology, St. Boniface Hospital, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| | - David C Nelson
- Institute of Cardiovascular Sciences, Department of Physiology and Pathophysiology, St. Boniface Hospital, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| | - Thane G Maddaford
- Institute of Cardiovascular Sciences, Department of Physiology and Pathophysiology, St. Boniface Hospital, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| | - Andrea L Edel
- Institute of Cardiovascular Sciences, Department of Physiology and Pathophysiology, St. Boniface Hospital, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| | - Graham Maddaford
- Institute of Cardiovascular Sciences, Department of Physiology and Pathophysiology, St. Boniface Hospital, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| | - Elena Dibrov
- Institute of Cardiovascular Sciences, Department of Physiology and Pathophysiology, St. Boniface Hospital, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| | - MohamadReza Aghanoori
- Department of Pharmacology and Therapeutics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
- Division of Neurodegenerative Disorders, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, Canada
| | - Lorrie A Kirshenbaum
- Institute of Cardiovascular Sciences, Department of Physiology and Pathophysiology, St. Boniface Hospital, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| | - Paul Fernyhough
- Department of Pharmacology and Therapeutics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
- Division of Neurodegenerative Disorders, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, Canada
| | - Michel Aliani
- Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, Canada
- The Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, Canada
| | - Grant N Pierce
- Institute of Cardiovascular Sciences, Department of Physiology and Pathophysiology, St. Boniface Hospital, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
- The Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, Canada
| | - Amir Ravandi
- Institute of Cardiovascular Sciences, Department of Physiology and Pathophysiology, St. Boniface Hospital, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
- Department of Internal Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| |
Collapse
|
45
|
Srikakulapu P, Upadhye A, Drago F, Perry HM, Bontha SV, McSkimming C, Marshall MA, Taylor AM, McNamara CA. Chemokine Receptor-6 Promotes B-1 Cell Trafficking to Perivascular Adipose Tissue, Local IgM Production and Atheroprotection. Front Immunol 2021; 12:636013. [PMID: 33679793 PMCID: PMC7933012 DOI: 10.3389/fimmu.2021.636013] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/01/2021] [Indexed: 12/21/2022] Open
Abstract
Chemokine receptor-6 (CCR6) mediates immune cell recruitment to inflammatory sites and has cell type-specific effects on diet-induced atherosclerosis in mice. Previously we showed that loss of CCR6 in B cells resulted in loss of B cell-mediated atheroprotection, although the B cell subtype mediating this effect was unknown. Perivascular adipose tissue (PVAT) harbors high numbers of B cells including atheroprotective IgM secreting B-1 cells. Production of IgM antibodies is a major mechanism whereby B-1 cells limit atherosclerosis development. Yet whether CCR6 regulates B-1 cell number and production of IgM in the PVAT is unknown. In this present study, flow cytometry experiments demonstrated that both B-1 and B-2 cells express CCR6, albeit at a higher frequency in B-2 cells in both humans and mice. Nevertheless, B-2 cell numbers in peritoneal cavity (PerC), spleen, bone marrow and PVAT were no different in ApoE -/- CCR6 -/- compared to ApoE -/- CCR6 +/+ mice. In contrast, the numbers of atheroprotective IgM secreting B-1 cells were significantly lower in the PVAT of ApoE -/- CCR6 -/- compared to ApoE -/- CCR6 +/+ mice. Surprisingly, adoptive transfer (AT) of CD43- splenic B cells into B cell-deficient μMT -/- ApoE -/- mice repopulated the PerC with B-1 and B-2 cells and reduced atherosclerosis when transferred into ApoE -/- CCR6 +/+ sIgM -/- mice only when those cells expressed both CCR6 and sIgM. CCR6 expression on circulating human B cells in subjects with a high level of atherosclerosis in their coronary arteries was lower only in the putative human B-1 cells. These results provide evidence that B-1 cell CCR6 expression enhances B-1 cell number and IgM secretion in PVAT to provide atheroprotection in mice and suggest potential human relevance to our murine findings.
Collapse
Affiliation(s)
- Prasad Srikakulapu
- Carter Immunology Center, University of Virginia, Charlottesville, VA, United States
| | - Aditi Upadhye
- Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States
| | - Fabrizio Drago
- Carter Immunology Center, University of Virginia, Charlottesville, VA, United States
| | - Heather M Perry
- Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States
| | - Sai Vineela Bontha
- Carter Immunology Center, University of Virginia, Charlottesville, VA, United States
| | - Chantel McSkimming
- Carter Immunology Center, University of Virginia, Charlottesville, VA, United States
| | - Melissa A Marshall
- Carter Immunology Center, University of Virginia, Charlottesville, VA, United States
| | - Angela M Taylor
- Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States.,Division of Cardiovascular Medicine, Department of Medicine, University of Virginia, Charlottesville, VA, United States
| | - Coleen A McNamara
- Carter Immunology Center, University of Virginia, Charlottesville, VA, United States.,Division of Cardiovascular Medicine, Department of Medicine, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
46
|
Ween MP, White JB, Tran HB, Mukaro V, Jones C, Macowan M, Hodge G, Trim PJ, Snel MF, Hodge SJ. The role of oxidised self-lipids and alveolar macrophage CD1b expression in COPD. Sci Rep 2021; 11:4106. [PMID: 33602992 PMCID: PMC7892841 DOI: 10.1038/s41598-021-82481-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 12/28/2020] [Indexed: 02/08/2023] Open
Abstract
In chronic obstructive pulmonary disease (COPD) apoptotic bronchial epithelial cells are increased, and their phagocytosis by alveolar macrophages (AM) is decreased alongside bacterial phagocytosis. Epithelial cellular lipids, including those exposed on uncleared apoptotic bodies, can become oxidized, and may be recognized and presented as non-self by antigen presenting cells. CD1b is a lipid-presenting protein, previously only described in dendritic cells. We investigated whether CD1b is upregulated in COPD AM, and whether lipid oxidation products are found in the airways of cigarette smoke (CS) exposed mice. We also characterise CD1b for the first time in a range of macrophages and assess CD1b expression and phagocytic function in response to oxidised lipid. Bronchoalveolar lavage and exhaled breath condensate were collected from never-smoker, current-smoker, and COPD patients and AM CD1b expression and airway 8-isoprostane levels assessed. Malondialdehyde was measured in CS-exposed mouse airways by confocal/immunofluorescence. Oxidation of lipids produced from CS-exposed 16HBE14o- (HBE) bronchial epithelial cells was assessed by spectrophotometry and changes in lipid classes assessed by mass spectrometry. 16HBE cell toxicity was measured by flow cytometry as was phagocytosis, CD1b expression, HLA class I/II, and mannose receptor (MR) in monocyte derived macrophages (MDM). AM CD1b was significantly increased in COPD smokers (4.5 fold), COPD ex-smokers (4.3 fold), and smokers (3.9 fold), and AM CD1b significantly correlated with disease severity (FEV1) and smoking pack years. Airway 8-isoprostane also increased in smokers and COPD smokers and ex-smokers. Malondialdehyde was significantly increased in the bronchial epithelium of CS-exposed mice (MFI of 18.18 vs 23.50 for control). Oxidised lipid was produced from CS-exposed bronchial epithelial cells (9.8-fold of control) and showed a different overall lipid makeup to that of control total cellular lipid. This oxidised epithelial lipid significantly upregulated MDM CD1b, caused bronchial epithelial cell toxicity, and reduced MDM phagocytic capacity and MR in a dose dependent manner. Increased levels of oxidised lipids in the airways of COPD patients may be responsible for reduced phagocytosis and may become a self-antigen to be presented by CD1b on macrophages to perpetuate disease progression despite smoking cessation.
Collapse
Affiliation(s)
- Miranda P Ween
- Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, Australia. .,School of Medicine, Faculty of Health Sciences, University of Adelaide, Adelaide, Australia.
| | - Jake B White
- School of Medicine, Faculty of Health Sciences, University of Adelaide, Adelaide, Australia.,Proteomics, Metabolomics and MS Imaging Core Facility, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, Australia.,Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, Australia
| | - Hai B Tran
- Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, Australia.,School of Medicine, Faculty of Health Sciences, University of Adelaide, Adelaide, Australia
| | - Violet Mukaro
- Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, Australia.,Department of Critical Care, Melbourne Medical School, University of Melbourne, Melbourne, Australia
| | - Charles Jones
- School of Medicine, Faculty of Health Sciences, University of Adelaide, Adelaide, Australia
| | - Matthew Macowan
- Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, Australia.,School of Medicine, Faculty of Health Sciences, University of Adelaide, Adelaide, Australia
| | - Gregory Hodge
- Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, Australia.,School of Medicine, Faculty of Health Sciences, University of Adelaide, Adelaide, Australia
| | - Paul J Trim
- School of Medicine, Faculty of Health Sciences, University of Adelaide, Adelaide, Australia.,Proteomics, Metabolomics and MS Imaging Core Facility, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, Australia
| | - Marten F Snel
- School of Medicine, Faculty of Health Sciences, University of Adelaide, Adelaide, Australia.,Proteomics, Metabolomics and MS Imaging Core Facility, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, Australia
| | - Sandra J Hodge
- School of Medicine, Faculty of Health Sciences, University of Adelaide, Adelaide, Australia
| |
Collapse
|
47
|
Functional Role of B Cells in Atherosclerosis. Cells 2021; 10:cells10020270. [PMID: 33572939 PMCID: PMC7911276 DOI: 10.3390/cells10020270] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 01/24/2021] [Accepted: 01/26/2021] [Indexed: 12/30/2022] Open
Abstract
Atherosclerosis is a lipid-driven inflammatory disease of blood vessels, and both innate and adaptive immune responses are involved in its development. The impact of B cells on atherosclerosis has been demonstrated in numerous studies and B cells have been found in close proximity to atherosclerotic plaques in humans and mice. B cells exert both atheroprotective and pro-atherogenic functions, which have been associated with their B cell subset attribution. While B1 cells and marginal zone B cells are considered to protect against atherosclerosis, follicular B cells and innate response activator B cells have been shown to promote atherosclerosis. In this review, we shed light on the role of B cells from a different, functional perspective and focus on the three major B cell functions: antibody production, antigen presentation/T cell interaction, and the release of cytokines. All of these functions have the potential to affect atherosclerosis by multiple ways and are dependent on the cellular milieu and the activation status of the B cell. Moreover, we discuss B cell receptor signaling and the mechanism of B cell activation under atherosclerosis-prone conditions. By summarizing current knowledge of B cells in and beyond atherosclerosis, we are pointing out open questions and enabling new perspectives.
Collapse
|
48
|
Ding WY, Protty MB, Davies IG, Lip GYH. Relationship between lipoproteins, thrombosis and atrial fibrillation. Cardiovasc Res 2021; 118:716-731. [PMID: 33483737 PMCID: PMC8859639 DOI: 10.1093/cvr/cvab017] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 12/14/2020] [Accepted: 01/12/2021] [Indexed: 12/19/2022] Open
Abstract
The prothrombotic state in atrial fibrillation (AF) occurs as a result of multifaceted interactions, known as Virchow’s triad of hypercoagulability, structural abnormalities, and blood stasis. More recently, there is emerging evidence that lipoproteins are implicated in this process, beyond their traditional role in atherosclerosis. In this review, we provide an overview of the various lipoproteins and explore the association between lipoproteins and AF, the effects of lipoproteins on haemostasis, and the potential contribution of lipoproteins to thrombogenesis in AF. There are several types of lipoproteins based on size, lipid composition, and apolipoprotein category, namely: chylomicrons, very low-density lipoprotein, low-density lipoprotein (LDL), intermediate-density lipoprotein, and high-density lipoprotein. Each of these lipoproteins may contain numerous lipid species and proteins with a variety of different functions. Furthermore, the lipoprotein particles may be oxidized causing an alteration in their structure and content. Of note, there is a paradoxical inverse relationship between total cholesterol and LDL cholesterol (LDL-C) levels, and incident AF. The mechanism by which this occurs may be related to the stabilizing effect of cholesterol on myocardial membranes, along with its role in inflammation. Overall, specific lipoproteins may interact with haemostatic pathways to promote excess platelet activation and thrombin generation, as well as inhibiting fibrinolysis. In this regard, LDL-C has been shown to be an independent risk factor for thromboembolic events in AF. The complex relationship between lipoproteins, thrombosis and AF warrants further research with an aim to improve our knowledge base and contribute to our overall understanding of lipoprotein-mediated thrombosis.
Collapse
Affiliation(s)
- Wern Yew Ding
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart & Chest Hospital, Liverpool, United Kingdom
| | - Majd B Protty
- Systems Immunity University Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Ian G Davies
- Research Institute of Sport and Exercise Science, Liverpool John Moores University, Liverpool, United Kingdom
| | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart & Chest Hospital, Liverpool, United Kingdom.,Aalborg Thrombosis Research Unit, Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| |
Collapse
|
49
|
Poznyak AV, Nikiforov NG, Markin AM, Kashirskikh DA, Myasoedova VA, Gerasimova EV, Orekhov AN. Overview of OxLDL and Its Impact on Cardiovascular Health: Focus on Atherosclerosis. Front Pharmacol 2021; 11:613780. [PMID: 33510639 PMCID: PMC7836017 DOI: 10.3389/fphar.2020.613780] [Citation(s) in RCA: 177] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 12/02/2020] [Indexed: 12/19/2022] Open
Abstract
Cardiovascular pathologies maintain the leading position in mortality worldwide. Atherosclerosis is a chronic disease that can result in a variety of serious complications, such as myocardial infarction, stroke, and cardiovascular disease. Inflammation and lipid metabolism alterations play a crucial role in atherogenesis, but the details of relationships and causality of these fundamental processes remain not clear. The oxidation of LDL was considered the main atherogenic modification of LDL within the vascular wall for decades. However, recent investigations provided a growing body of evidence in support of the multiple LDL modification theory. It suggests that LDL particles undergo numerous modifications that change their size, density, and chemical properties within the blood flow and vascular wall. Oxidation is the last stage in this cascade resulting in the atherogenic properties. Moreover, recent investigations have discovered that oxLDL may have both anti-inflammatory and pro-inflammatory properties. Oxidized LDL can trigger inflammation through the activation of macrophages and other cells. After all, oxidized LDL is still a promising object for further investigations that have the potential to clarify the unknown parts of the atherogenic process. In this review, we discuss the role of oxLDL in atherosclerosis development on different levels.
Collapse
Affiliation(s)
- Anastasia V Poznyak
- Institute for Atherosclerosis Research, Skolkovo Innovative Center, Moscow, Russia
| | - Nikita G Nikiforov
- Centre of Collective Usage, Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia.,National Medical Research Center of Cardiology, Institute of Experimental Cardiology, Moscow, Russia.,Laboratory of Cellular and Molecular Pathology of the Cardiovascular System, Institute of Human Morphology, Moscow, Russia.,Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Alexander M Markin
- Laboratory of Cellular and Molecular Pathology of the Cardiovascular System, Institute of Human Morphology, Moscow, Russia
| | - Dmitry A Kashirskikh
- Laboratory of Cellular and Molecular Pathology of the Cardiovascular System, Institute of Human Morphology, Moscow, Russia
| | - Veronika A Myasoedova
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Moscow, Russia.,Centro Cardiologico Monzino, Istituti di Ricovero e Cura a Carattere Scientifico, Milan, Italy
| | - Elena V Gerasimova
- Department of Systemic Rheumatic Diseases, V.A. Nasonova Research Institute of Rheumatology, Moscow, Russia
| | - Alexander N Orekhov
- Institute for Atherosclerosis Research, Skolkovo Innovative Center, Moscow, Russia.,Laboratory of Cellular and Molecular Pathology of the Cardiovascular System, Institute of Human Morphology, Moscow, Russia.,Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Moscow, Russia
| |
Collapse
|
50
|
Zbesko JC, Frye JB, Becktel DA, Gerardo DK, Stokes J, Calderon K, Nguyen TVV, Bhattacharya D, Doyle KP. IgA natural antibodies are produced following T-cell independent B-cell activation following stroke. Brain Behav Immun 2021; 91:578-586. [PMID: 32956832 PMCID: PMC8279117 DOI: 10.1016/j.bbi.2020.09.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 09/08/2020] [Accepted: 09/15/2020] [Indexed: 12/22/2022] Open
Abstract
Up to 30% of stroke patients experience cognitive decline within one year of their stroke. There are currently no FDA-approved drugs that can prevent post-stroke cognitive decline, in part due to a poor understanding of the mechanisms involved. We have previously demonstrated that a B-lymphocyte response to stroke, marked by IgA + cells, can cause delayed cognitive dysfunction in mice and that a similar adaptive immune response occurs in the brains of some human stroke patients that suffer from vascular dementia. The stimuli which trigger B-lymphocyte activation following stroke, and their target antigens, are still unknown. Therefore, to learn more about the mechanisms by which B-lymphocytes become activated following stroke we first characterized the temporal kinetics of the B-lymphocyte, T-lymphocyte, and plasma cell (PC) response to stroke in the brain by immunohistochemistry (IHC). We discovered that B-lymphocyte, T-lymphocyte, and plasma cell infiltration within the infarct progressively increases between 2 and 7 weeks after stroke. We then compared the B-lymphocyte response to stroke in WT, MHCII-/-, CD4-/-, and MyD88-/- mice to determine if B-lymphocytes mature into IgA + PCs through a T-lymphocyte and MyD88 dependent mechanism. Our data from a combination of IHC and flow cytometry indicate that following stroke, a population of IgA + PCs develops independently of CD4 + helper T-lymphocytes and MyD88 signaling. Subsequent sequencing of immunoglobulin genes of individual IgA + PCs present within the infarct identified a novel population of natural antibodies with few somatic mutations in complementarity-determining regions. These findings indicate that a population of IgA + PCs develops in the infarct following stroke by B-lymphocytes interacting with one or more thymus independent type 2 (TI-2) antigens, and that they produce IgA natural antibodies.
Collapse
Affiliation(s)
- Jacob C. Zbesko
- Department of Immunobiology, University of Arizona, Tucson, AZ 85719, USA
| | | | | | - Diana K. Gerardo
- Department of Immunobiology, University of Arizona, Tucson, AZ 85719, USA
| | - Jessica Stokes
- Department of Pediatrics, University of Arizona, Tucson, AZ 85719, USA
| | - Kylie Calderon
- Department of Immunobiology, University of Arizona, Tucson, AZ 85719, USA
| | - Thuy-Vi V. Nguyen
- Department of Immunobiology, University of Arizona, Tucson, AZ 85719, USA,Department of Neurology, University of Arizona, Tucson, AZ 85719, USA
| | | | - Kristian P. Doyle
- Department of Immunobiology, University of Arizona, Tucson, AZ 85719, USA,Department of Neurology, University of Arizona, Tucson, AZ 85719, USA,Arizona Center on Aging, University of Arizona, Tucson, AZ 85719, USA,Corresponding author at: Department of Immunobiology, University of Arizona, Tucson, 1656 E. Mabel Street, Tucson, Arizona 85719, USA. (K.P. Doyle)
| |
Collapse
|