101
|
McKay JT, Haro MA, Daly CA, Yammani RD, Pang B, Swords WE, Haas KM. PD-L2 Regulates B-1 Cell Antibody Production against Phosphorylcholine through an IL-5-Dependent Mechanism. THE JOURNAL OF IMMUNOLOGY 2017; 199:2020-2029. [PMID: 28768724 DOI: 10.4049/jimmunol.1700555] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 07/11/2017] [Indexed: 11/19/2022]
Abstract
B-1 cells produce natural Abs which provide an integral first line of defense against pathogens while also performing important homeostatic housekeeping functions. In this study, we demonstrate that programmed cell death 1 ligand 2 (PD-L2) regulates the production of natural Abs against phosphorylcholine (PC). Naive PD-L2-deficient (PD-L2-/-) mice produced significantly more PC-reactive IgM and IgA. This afforded PD-L2-/- mice with selectively enhanced protection against PC-expressing nontypeable Haemophilus influenzae, but not PC-negative nontypeable Haemophilus influenzae, relative to wild-type mice. PD-L2-/- mice had significantly increased PC-specific CD138+ splenic plasmablasts bearing a B-1a phenotype, and produced PC-reactive Abs largely of the T15 Id. Importantly, PC-reactive B-1 cells expressed PD-L2 and irradiated chimeras demonstrated that B cell-intrinsic PD-L2 expression regulated PC-specific Ab production. In addition to increased PC-specific IgM, naive PD-L2-/- mice and irradiated chimeras reconstituted with PD-L2-/- B cells had significantly higher levels of IL-5, a potent stimulator of B-1 cell Ab production. PD-L2 mAb blockade of wild-type B-1 cells in culture significantly increased CD138 and Blimp1 expression and PC-specific IgM, but did not affect proliferation. PD-L2 mAb blockade significantly increased IL-5+ T cells in culture. Both IL-5 neutralization and STAT5 inhibition blunted the effects of PD-L2 mAb blockade on B-1 cells. Thus, B-1 cell-intrinsic PD-L2 expression inhibits IL-5 production by T cells and thereby limits natural Ab production by B-1 cells. These findings have broad implications for the development of therapeutic strategies aimed at altering natural Ab levels critical for protection against infectious disease, autoimmunity, allergy, cancer, and atherosclerosis.
Collapse
Affiliation(s)
- Jerome T McKay
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | - Marcela A Haro
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | - Christina A Daly
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | - Rama D Yammani
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | - Bing Pang
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | - W Edward Swords
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | - Karen M Haas
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC 27157
| |
Collapse
|
102
|
Akhi R, Wang C, Kyrklund M, Kummu O, Turunen SP, Hyvärinen K, Kullaa A, Salo T, Pussinen PJ, Hörkkö S. Cross-reactive saliva IgA antibodies to oxidized LDL and periodontal pathogens in humans. J Clin Periodontol 2017; 44:682-691. [PMID: 28548243 DOI: 10.1111/jcpe.12748] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/20/2017] [Indexed: 02/02/2023]
Abstract
AIM Oxidized low-density lipoproteins (oxLDL) are formed as a result of lipid peroxidation and are highly immunogenic and proatherogenic. In this study, saliva antibodies binding to oxLDL, Porphyromonas gingivalis (Pg) and Aggregatibacter actinomycetemcomitans (Aa) were characterized and their cross-reactivity was evaluated. MATERIALS AND METHODS Resting and stimulated saliva samples were collected from 36 healthy adults (mean age 26 years). Saliva IgA, IgG and IgM autoantibody levels to copper oxidized LDL (CuOx-LDL) and malondialdehyde acetaldehyde-modified LDL (MAA-LDL) were determined with chemiluminescence immunoassay. RESULTS Saliva IgA and IgG antibodies binding to MAA-LDL and CuOx-LDL were detected in all samples and they were associated with the saliva levels of IgA and IgG to P. gingivalis and A. actinomycetemcomitans. Competitive immunoassay showed that saliva antibodies to MAA-LDL cross-reacted specifically with P. gingivalis. The autoantibody levels to oxLDL in saliva were not associated with the autoantibody levels to oxLDL in plasma or with saliva apolipoprotein B 100 levels. CONCLUSIONS Saliva contains IgA and IgG binding to oxLDL, which showed cross-reactive properties with the periodontal pathogens Porphyromonas gingivalis (P.g). The data suggest that secretory IgA to P.g may participate in immune reactions involved in LDL oxidation through molecular mimicry.
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.,Nordlab, Oulu University Hospital, Oulu, Finland.,Research Unit of Oral Health Sciences, 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.,Nordlab, Oulu University Hospital, 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.,Nordlab, Oulu University Hospital, Oulu, Finland
| | - Outi Kummu
- 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.,Nordlab, Oulu University Hospital, Oulu, Finland
| | - Sini Pauliina Turunen
- 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.,Research Programs Unit, Genome-Scale Biology, University of Helsinki, Helsinki, Finland
| | - Kati Hyvärinen
- Oral and Maxillofacial Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Arja Kullaa
- Research Unit of Oral Health Sciences, University of Oulu, Oulu, Finland
| | - Tuula Salo
- Medical Research Center, Oulu University Hospital and University of Oulu, Oulu, Finland.,Cancer Research and Translational Medicine Research Unit, University of Oulu, Oulu, 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.,Nordlab, Oulu University Hospital, Oulu, Finland
| |
Collapse
|
103
|
Srikakulapu P, McNamara CA. B cells and atherosclerosis. Am J Physiol Heart Circ Physiol 2017; 312:H1060-H1067. [PMID: 28314764 DOI: 10.1152/ajpheart.00859.2016] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 03/08/2017] [Accepted: 03/13/2017] [Indexed: 12/14/2022]
Abstract
B cells have emerged as important immune cells in cardiovascular disease. Initial studies have suggested that B cells protect against atherosclerosis development. However, subsequent studies demonstrating aggravation of atherosclerosis by B-2 cells have shed light on the subset-dependent effects of B cells. Here, we review the literature that has led to our current understanding of B cell regulation of atherosclerosis, touching on the importance of subsets, local regulation, human translation, and therapeutic potential.
Collapse
Affiliation(s)
| | - Coleen A McNamara
- Cardiovascular Research Center, Charlottesville, Virginia; and.,Division of Cardiovascular Medicine, Department of Medicine, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
104
|
Förstermann U, Xia N, Li H. Roles of Vascular Oxidative Stress and Nitric Oxide in the Pathogenesis of Atherosclerosis. Circ Res 2017; 120:713-735. [DOI: 10.1161/circresaha.116.309326] [Citation(s) in RCA: 692] [Impact Index Per Article: 86.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 12/19/2016] [Accepted: 12/26/2016] [Indexed: 12/13/2022]
Abstract
Major reactive oxygen species (ROS)–producing systems in vascular wall include NADPH (reduced form of nicotinamide adenine dinucleotide phosphate) oxidase, xanthine oxidase, the mitochondrial electron transport chain, and uncoupled endothelial nitric oxide (NO) synthase. ROS at moderate concentrations have important signaling roles under physiological conditions. Excessive or sustained ROS production, however, when exceeding the available antioxidant defense systems, leads to oxidative stress. Animal studies have provided compelling evidence demonstrating the roles of vascular oxidative stress and NO in atherosclerosis. All established cardiovascular risk factors such as hypercholesterolemia, hypertension, diabetes mellitus, and smoking enhance ROS generation and decrease endothelial NO production. Key molecular events in atherogenesis such as oxidative modification of lipoproteins and phospholipids, endothelial cell activation, and macrophage infiltration/activation are facilitated by vascular oxidative stress and inhibited by endothelial NO. Atherosclerosis develops preferentially in vascular regions with disturbed blood flow (arches, branches, and bifurcations). The fact that these sites are associated with enhanced oxidative stress and reduced endothelial NO production is a further indication for the roles of ROS and NO in atherosclerosis. Therefore, prevention of vascular oxidative stress and improvement of endothelial NO production represent reasonable therapeutic strategies in addition to the treatment of established risk factors (hypercholesterolemia, hypertension, and diabetes mellitus).
Collapse
Affiliation(s)
- Ulrich Förstermann
- From the Department of Pharmacology, Johannes Gutenberg University Medical Center, Mainz, Germany (U.F., N.X., H.L.); Center for Translational Vascular Biology (CTVB), Johannes Gutenberg University Medical Center, Mainz, Germany (H.L.); and German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany (H.L.)
| | - Ning Xia
- From the Department of Pharmacology, Johannes Gutenberg University Medical Center, Mainz, Germany (U.F., N.X., H.L.); Center for Translational Vascular Biology (CTVB), Johannes Gutenberg University Medical Center, Mainz, Germany (H.L.); and German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany (H.L.)
| | - Huige Li
- From the Department of Pharmacology, Johannes Gutenberg University Medical Center, Mainz, Germany (U.F., N.X., H.L.); Center for Translational Vascular Biology (CTVB), Johannes Gutenberg University Medical Center, Mainz, Germany (H.L.); and German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany (H.L.)
| |
Collapse
|
105
|
Miller YI, Shyy JYJ. Context-Dependent Role of Oxidized Lipids and Lipoproteins in Inflammation. Trends Endocrinol Metab 2017; 28:143-152. [PMID: 27931771 PMCID: PMC5253098 DOI: 10.1016/j.tem.2016.11.002] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 10/26/2016] [Accepted: 11/02/2016] [Indexed: 01/13/2023]
Abstract
Oxidized low-density lipoprotein (OxLDL), which contains hundreds of different oxidized lipid molecules, is a hallmark of hyperlipidemia and atherosclerosis. The same oxidized lipids found in OxLDL are also formed in apoptotic cells, and are present in tissues as well as in the circulation under pathological conditions. In many disease contexts, oxidized lipids constitute damage signals, or patterns, that activate pattern-recognition receptors (PRRs) and significantly contribute to inflammation. Here, we review recent discoveries and emerging trends in the field of oxidized lipids and the regulation of inflammation, focusing on oxidation products of polyunsaturated fatty acids esterified into cholesteryl esters (CEs) and phospholipids (PLs). We also highlight context-dependent activation and biased agonism of Toll-like receptor-4 (TLR4) and the NLRP3 inflammasome, among other signaling pathways activated by oxidized lipids.
Collapse
Affiliation(s)
- Yury I Miller
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA.
| | - John Y-J Shyy
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
106
|
Chyu KY, Dimayuga PC, Shah PK. Vaccine against arteriosclerosis: an update. THERAPEUTIC ADVANCES IN VACCINES 2017; 5:39-47. [PMID: 28515939 DOI: 10.1177/2051013617693753] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 01/18/2017] [Indexed: 12/13/2022]
Abstract
Substantial data from experimental and clinical investigation support the role of immune-mediated mechanisms in atherogenesis, with immune systems responding to many endogenous and exogenous antigens that play either proatherogenic or atheroprotective roles. An active immunization strategy against many of these antigens could potentially alter the natural history of atherosclerosis. This review mainly focuses on the important studies on the search for antigens that have been tested in vaccine formulations to reduce atherosclerosis in preclinical models. It will also address the opportunities and challenges associated with potential clinical application of this novel therapeutic paradigm.
Collapse
Affiliation(s)
- Kuang-Yuh Chyu
- Oppenheimer Atherosclerosis Research Center, Division of Cardiology, Cedars-Sinai Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Paul C Dimayuga
- Oppenheimer Atherosclerosis Research Center, Division of Cardiology, Cedars-Sinai Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Prediman K Shah
- Cedars-Sinai Medical Center, 127 South San Vicente Blvd., Suite A-3307, Los Angeles, CA 90048, USA
| |
Collapse
|
107
|
Popat RJ, Hakki S, Thakker A, Coughlan AM, Watson J, Little MA, Spickett CM, Lavender P, Afzali B, Kemper C, Robson MG. Anti-myeloperoxidase antibodies attenuate the monocyte response to LPS and shape macrophage development. JCI Insight 2017; 2:e87379. [PMID: 28138552 PMCID: PMC5256146 DOI: 10.1172/jci.insight.87379] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Anti-neutrophil cytoplasmic antibody (ANCA) vasculitis is characterized by the presence of autoantibodies to myeloperoxidase and proteinase-3, which bind monocytes in addition to neutrophils. While a pathological effect on neutrophils is acknowledged, the impact of ANCA on monocyte function is less well understood. Using IgG from patients we investigated the effect of these autoantibodies on monocytes and found that anti-myeloperoxidase antibodies (MPO-ANCA) reduced both IL-10 and IL-6 secretion in response to LPS. This reduction in IL-10 and IL-6 depended on Fc receptors and enzymatic myeloperoxidase and was accompanied by a significant reduction in TLR-driven signaling pathways. Aligning with changes in TLR signals, oxidized phospholipids, which function as TLR4 antagonists, were increased in monocytes in the presence of MPO-ANCA. We further observed that MPO-ANCA increased monocyte survival and differentiation to macrophages by stimulating CSF-1 production. However, this was independent of myeloperoxidase enzymatic activity and TLR signaling. Macrophages differentiated in the presence of MPO-ANCA secreted more TGF-β and further promoted the development of IL-10– and TGF-β–secreting CD4+ T cells. Thus, MPO-ANCA may promote inflammation by reducing the secretion of antiinflammatory IL-10 from monocytes, and MPO-ANCA can alter the development of macrophages and T cells to potentially promote fibrosis. Anti-myeloperoxidase antibodies from patients with anti-neutrophil cytoplasmic antibody (ANCA) vasculitis alter monocyte function in addition to previously described effects on neutrophils.
Collapse
Affiliation(s)
- Reena J Popat
- Division of Transplant Immunology and Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, Great Maze Pond, London, United Kingdom
| | - Seran Hakki
- Division of Transplant Immunology and Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, Great Maze Pond, London, United Kingdom
| | - Alpesh Thakker
- School of Life & Health Sciences, Aston University, Aston Triangle, Birmingham, United Kingdom
| | - Alice M Coughlan
- Trinity Health Kidney Centre, Department of Clinical Medicine, Trinity College Dublin, St. James' Hospital Campus, Dublin, Ireland
| | - Julie Watson
- MRC and Asthma UK Centre in Allergic Mechanisms of Asthma, King's College London, Guy's Hospital, Great Maze Pond, London, United Kingdom
| | - Mark A Little
- Trinity Health Kidney Centre, Department of Clinical Medicine, Trinity College Dublin, St. James' Hospital Campus, Dublin, Ireland
| | - Corinne M Spickett
- School of Life & Health Sciences, Aston University, Aston Triangle, Birmingham, United Kingdom
| | - Paul Lavender
- MRC and Asthma UK Centre in Allergic Mechanisms of Asthma, King's College London, Guy's Hospital, Great Maze Pond, London, United Kingdom
| | - Behdad Afzali
- Division of Transplant Immunology and Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, Great Maze Pond, London, United Kingdom
| | - Claudia Kemper
- Division of Transplant Immunology and Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, Great Maze Pond, London, United Kingdom
| | - Michael G Robson
- Division of Transplant Immunology and Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, Great Maze Pond, London, United Kingdom
| |
Collapse
|
108
|
Liu B, Tai Y, Caceres AI, Achanta S, Balakrishna S, Shao X, Fang J, Jordt SE. Oxidized Phospholipid OxPAPC Activates TRPA1 and Contributes to Chronic Inflammatory Pain in Mice. PLoS One 2016; 11:e0165200. [PMID: 27812120 PMCID: PMC5094666 DOI: 10.1371/journal.pone.0165200] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 10/07/2016] [Indexed: 01/13/2023] Open
Abstract
Oxidation products of the naturally occurring phospholipid 1-palmitoyl-2-arachidonoyl-sn-glycerol-3-phosphatidylcholine (PAPC), which are known as OxPAPC, accumulate in atherosclerotic lesions and at other sites of inflammation in conditions such as septic inflammation and acute lung injury to exert pro- or anti-inflammatory effects. It is currently unknown whether OxPAPC also contributes to inflammatory pain and peripheral neuronal excitability in these conditions. Here, we observed that OxPAPC dose-dependently and selectively activated human TRPA1 nociceptive ion channels expressed in HEK293 cells in vitro, without any effect on other TRP channels, including TRPV1, TRPV4 and TRPM8. OxPAPC agonist activity was dependent on essential cysteine and lysine residues within the N-terminus of the TRPA1 channel protein. OxPAPC activated calcium influx into a subset of mouse sensory neurons which were also sensitive to the TRPA1 agonist mustard oil. Neuronal OxPAPC responses were largely abolished in neurons isolated from TRPA1-deficient mice. Intraplantar injection of OxPAPC into the mouse hind paw induced acute pain and persistent mechanical hyperalgesia and this effect was attenuated by the TRPA1 inhibitor, HC-030031. More importantly, we found levels of OxPAPC to be significantly increased in inflamed tissue in a mouse model of chronic inflammatory pain, identified by the binding of an OxPAPC-specific antibody. These findings suggest that TRPA1 is a molecular target for OxPAPC and OxPAPC may contribute to chronic inflammatory pain through TRPA1 activation. Targeting against OxPAPC and TRPA1 signaling pathway may be promising in inflammatory pain treatment.
Collapse
Affiliation(s)
- Boyi Liu
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, P.R. China
| | - Yan Tai
- Department of Laboratory and Equipment Administration, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, P.R. China
- Department of Anesthesiology, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Ana I. Caceres
- Department of Anesthesiology, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Satyanarayana Achanta
- Department of Anesthesiology, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Shrilatha Balakrishna
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Xiaomei Shao
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, P.R. China
| | - Junfan Fang
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, P.R. China
| | - Sven-Eric Jordt
- Department of Anesthesiology, Duke University School of Medicine, Durham, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
109
|
Jeurissen MLJ, Walenbergh SMA, Houben T, Gijbels MJJ, Li J, Hendrikx T, Oligschlaeger Y, van Gorp PJ, Binder CJ, Donners MMPC, Shiri-Sverdlov R. Prevention of oxLDL uptake leads to decreased atherosclerosis in hematopoietic NPC1-deficient Ldlr -/- mice. Atherosclerosis 2016; 255:59-65. [PMID: 27816810 DOI: 10.1016/j.atherosclerosis.2016.10.038] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 10/03/2016] [Accepted: 10/19/2016] [Indexed: 01/07/2023]
Abstract
BACKGROUND AND AIMS Atherosclerosis is a chronic inflammatory disease of medium and large vessels and is typically characterized by the predominant accumulation of low-density lipoprotein (LDL)-cholesterol inside macrophages that reside in the vessel walls. Previous studies clearly demonstrated an association specifically between the oxidized type of LDL (oxLDL) and atherosclerotic lesion formation. Further observations revealed that these atherosclerotic lesions displayed enlarged, lipid-loaded lysosomes. By increasing natural antibodies against oxLDL, pneumococcal vaccination has been shown to reduce atherosclerosis in LDL receptor knockout (Ldlr-/-) mice. Relevantly, loss of the lysosomal membrane protein Niemann-Pick Type C1 (NPC1) led to lysosomal accumulation of various lipids and promoted atherosclerosis. Yet, the importance of lysosomal oxLDL accumulation inside macrophages, compared to non-modified LDL, in atherosclerosis has never been established. METHODS By transplanting NPC1 bone marrow into lethally irradiated Ldlr-/- mice, a hematopoietic mouse model for lysosomal cholesterol accumulation was created. Through injections with heat-inactivated pneumococci, we aimed to demonstrate the specific contribution of lysosomal oxLDL accumulation inside macrophages in atherosclerosis development. RESULTS While there were no differences in plaque morphology, a reduction in plaque size and plaque inflammation was found in immunized NPC1mut-transplanted mice, compared to non-immunized NPC1mut-transplanted mice. CONCLUSIONS Lysosomal oxLDL accumulation within macrophages contributes to murine atherosclerosis. Future intervention strategies should focus specifically on preventing oxLDL, unlike non-modified LDL, from being internalized into lysosomes. Such an intervention can have an additive effect to current existing treatments against atherosclerosis.
Collapse
Affiliation(s)
- Mike L J Jeurissen
- Departments of Molecular Genetics and Pathology, School of Nutrition and Translational Research in Metabolism (NUTRIM) and School for Cardiovascular Diseases (CARIM), Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Sofie M A Walenbergh
- Departments of Molecular Genetics and Pathology, School of Nutrition and Translational Research in Metabolism (NUTRIM) and School for Cardiovascular Diseases (CARIM), Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Tom Houben
- Departments of Molecular Genetics and Pathology, School of Nutrition and Translational Research in Metabolism (NUTRIM) and School for Cardiovascular Diseases (CARIM), Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Marion J J Gijbels
- Departments of Molecular Genetics and Pathology, School of Nutrition and Translational Research in Metabolism (NUTRIM) and School for Cardiovascular Diseases (CARIM), Maastricht University Medical Centre+, Maastricht, The Netherlands; Experimental Vascular Biology, Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Jieyi Li
- Departments of Molecular Genetics and Pathology, School of Nutrition and Translational Research in Metabolism (NUTRIM) and School for Cardiovascular Diseases (CARIM), Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Tim Hendrikx
- Departments of Molecular Genetics and Pathology, School of Nutrition and Translational Research in Metabolism (NUTRIM) and School for Cardiovascular Diseases (CARIM), Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Yvonne Oligschlaeger
- Departments of Molecular Genetics and Pathology, School of Nutrition and Translational Research in Metabolism (NUTRIM) and School for Cardiovascular Diseases (CARIM), Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Patrick J van Gorp
- Departments of Molecular Genetics and Pathology, School of Nutrition and Translational Research in Metabolism (NUTRIM) and School for Cardiovascular Diseases (CARIM), Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria; Center for Molecular Medicine (CeMM), Austrian Academy of Sciences, Vienna, Austria
| | - Marjo M P C Donners
- Departments of Molecular Genetics and Pathology, School of Nutrition and Translational Research in Metabolism (NUTRIM) and School for Cardiovascular Diseases (CARIM), Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Ronit Shiri-Sverdlov
- Departments of Molecular Genetics and Pathology, School of Nutrition and Translational Research in Metabolism (NUTRIM) and School for Cardiovascular Diseases (CARIM), Maastricht University Medical Centre+, Maastricht, The Netherlands.
| |
Collapse
|
110
|
Zhu L, Fang L. AIBP: A Novel Molecule at the Interface of Cholesterol Transport, Angiogenesis, and Atherosclerosis. Methodist Debakey Cardiovasc J 2016; 11:160-5. [PMID: 26634023 DOI: 10.14797/mdcj-11-3-160] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Cardiovascular disease, which is often driven by hypercholesterolemia and subsequent coronary atherosclerosis, is the number-one cause of morbidity and mortality in the United States. Based on long-term epidemiological studies, high-density lipoprotein cholesterol (HDL-C) levels are inversely correlated with risk for coronary artery disease (CAD). HDL-mediated reverse cholesterol transport (RCT) is responsible for cholesterol removal from the peripheral tissues and return to the liver for final elimination.1 In atherosclerosis, intraplaque angiogenesis promotes plaque growth and increases plaque vulnerability. Conceivably, the acceleration of RCT and disruption of intraplaque angiogenesis would inhibit atherosclerosis and reduce CAD. We have identified a protein called apoA-I binding protein (AIBP) that augments HDL functionality by accelerating cholesterol efflux. Furthermore, AIBP inhibits vascular endothelial growth factor receptor 2 activation in endothelial cells and limits angiogenesis.2 The following discusses the prospect of using AIBP as a novel therapeutic approach for the treatment of CAD.
Collapse
Affiliation(s)
- Laurence Zhu
- Houston Methodist Research Institute, Houston Methodist Hospital, Houston, Texas
| | - Longhou Fang
- Houston Methodist Research Institute, Houston Methodist Hospital, Houston, Texas
| |
Collapse
|
111
|
Ampuero J, Ranchal I, Gallego-Durán R, Pareja MJ, Del Campo JA, Pastor-Ramírez H, Rico MC, Picón R, Pastor L, García-Monzón C, Andrade R, Romero-Gómez M. Oxidized low-density lipoprotein antibodies/high-density lipoprotein cholesterol ratio is linked to advanced non-alcoholic fatty liver disease lean patients. J Gastroenterol Hepatol 2016; 31:1611-8. [PMID: 26946071 DOI: 10.1111/jgh.13335] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Revised: 02/26/2016] [Accepted: 02/26/2016] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIM A small but significant proportion of patients with normal body mass index show non-alcoholic fatty liver disease (NAFLD). Oxidized low-density lipoprotein (LDL) is a powerful immunogenic molecule, which causes oxidative stress and produces antibodies (oxLDL-ab). We aimed to analyze the role of oxLDL-ab on histological features in lean-NAFLD patients. METHODS Seventy-two biopsy-proven NAFLD patients were included. Lean patients showed body index mass of <30 kg/m(2) . Liver biopsies were assessed by one pathologist blinded to clinical data. Histological features were non-alcoholic steatohepatitis (NASH), steatosis, hepatocellular ballooning, and liver fibrosis. Metabolic and hepatic profiles were analyzed, and lipid-lowering medication was recorded. OxLDL-ab levels were measured by ELISA. OxLDL-ab-based lipid indexes analyzed: oxLDL-ab/total cholesterol ratio; oxLDL-ab/LDL-c ratio; oxLDL-ab/high-density lipoprotein cholesterol (HDL-c) ratio; and oxLDL-ab/oxLDL ratio. RESULTS Lean-NAFLD patients presented 26.5% (9/34) of NASH. OxLDL-ab/HDL-c ratio (r = 0.570; n = 34; P = 0.001) correlated with NAS score and was the only variable associated with NASH in the multivariate analysis [odds ratio, OR, 1.10 (95% confidence interval, CI: 1.01-1.21); P = 0.039]. Severe steatosis was present in 41.2% (14/34) of lean-NAFLD patients. OxLDL-ab/HDL-c ratio was higher in patients with grade-III steatosis (54.9 (37.3-124.6)) than those with grade II (37.1 (20.2-71.1)) and grade I (17.7 (13.1-22.8)) (P = 0.018). Hepatocellular ballooning was present in 20.6% (7/34) of lean-NAFLD patients, and OxLDL-ab/HDL-c ratio (OR 1.03 [95% CI: 1.01-1.05]; P = 0.050) was independently associated with histological features. OxLDL-ab/HDL-c ratio was higher in patients with advanced fibrosis (39.8 (22.9-121.6) vs 17.7 (13.9-30.9); P = 0.025), increasing gradually with the fibrosis stage (P = 0.042) and remained in the final multivariate model [OR 1.05 (95% CI: 1.00-1.11); P = 0.05]. However, in obese-NAFLD patients, oxLDL/HDL-c ratio was not associated with histological features. CONCLUSIONS Oxidized low-density lipoprotein antibodies/high-density lipoprotein cholesterol ratio could represent an interesting biomarker associated with NASH, hepatocellular ballooning, and liver fibrosis, in lean patients. OxLDL-ab/HDL-c could play an important role for distinguishing patients with and without NAFLD complications.
Collapse
Affiliation(s)
- Javier Ampuero
- Inter-Centre Unit of Digestive Diseases & CIBERehd. Virgen Macarena - Virgen del Rocío University Hospitals, University of Sevilla, Sevilla, Spain.,Instituto de Biomedicina de Sevilla, Sevilla, Spain
| | | | - Rocío Gallego-Durán
- Inter-Centre Unit of Digestive Diseases & CIBERehd. Virgen Macarena - Virgen del Rocío University Hospitals, University of Sevilla, Sevilla, Spain.,Instituto de Biomedicina de Sevilla, Sevilla, Spain
| | | | - Jose Antonio Del Campo
- Instituto de Biomedicina de Sevilla, Sevilla, Spain.,Valme University Hospital, Sevilla, Spain
| | - Helena Pastor-Ramírez
- Inter-Centre Unit of Digestive Diseases & CIBERehd. Virgen Macarena - Virgen del Rocío University Hospitals, University of Sevilla, Sevilla, Spain.,Instituto de Biomedicina de Sevilla, Sevilla, Spain
| | - María Carmen Rico
- Inter-Centre Unit of Digestive Diseases & CIBERehd. Virgen Macarena - Virgen del Rocío University Hospitals, University of Sevilla, Sevilla, Spain.,Instituto de Biomedicina de Sevilla, Sevilla, Spain
| | - Rocío Picón
- Cardiology Unit, Valme University Hospital, Sevilla, Spain
| | - Luis Pastor
- Cardiology Unit, Valme University Hospital, Sevilla, Spain
| | - Carmelo García-Monzón
- Liver Research Unit, Department of Gastroenterology, Santa Cristina University Hospital, Madrid, Spain
| | - Raúl Andrade
- Unit for the Medical Management of Digestive Diseases & CIBERehd, Virgen de la Victoria University Hospital, Biomedical Research Institute of Málaga - IBIMA, Málaga, Spain
| | - Manuel Romero-Gómez
- Inter-Centre Unit of Digestive Diseases & CIBERehd. Virgen Macarena - Virgen del Rocío University Hospitals, University of Sevilla, Sevilla, Spain. .,Instituto de Biomedicina de Sevilla, Sevilla, Spain.
| |
Collapse
|
112
|
Dennis EA. Liberating Chiral Lipid Mediators, Inflammatory Enzymes, and LIPID MAPS from Biological Grease. J Biol Chem 2016; 291:24431-24448. [PMID: 27555328 DOI: 10.1074/jbc.x116.723791] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
In 1970, it was well accepted that the central role of lipids was in energy storage and metabolism, and it was assumed that amphipathic lipids simply served a passive structural role as the backbone of biological membranes. As a result, the scientific community was focused on nucleic acids, proteins, and carbohydrates as information-containing molecules. It took considerable effort until scientists accepted that lipids also "encode" specific and unique biological information and play a central role in cell signaling. Along with this realization came the recognition that the enzymes that act on lipid substrates residing in or on membranes and micelles must also have important signaling roles, spurring curiosity into their potentially unique modes of action differing from those acting on water-soluble substrates. This led to the creation of the concept of "surface dilution kinetics" for describing the mechanism of enzymes acting on lipid substrates, as well as the demonstration that lipid enzymes such as phospholipase A2 (PLA2) contain allosteric activator sites for specific phospholipids as well as for membranes. As our understanding of phospholipases advanced, so did the understanding that many of the lipids released by these enzymes are chiral information-containing signaling molecules; for example, PLA2 regulates the generation of precursors for the biosynthesis of eicosanoids and other bioactive lipid mediators of inflammation and resolution underlying disease progression. The creation of the LIPID MAPS initiative in 2003 and the ensuing development of the lipidomics field have revealed that lipid metabolites are central to human metabolism. Today lipids are recognized as key mediators of health and disease as we enter a new era of biomarkers and personalized medicine. This article is my personal "reflection" on these scientific advances.
Collapse
Affiliation(s)
- Edward A Dennis
- From the Department of Chemistry and Biochemistry and Department of Pharmacology, School of Medicine, University of California at San Diego, La Jolla, California 92093-0601.
| |
Collapse
|
113
|
Bochkov V, Schoenenberger AW, Oskolkova O, Toth U, Stöckl J, Majdic O, Daci A, Resink TJ, Erne P, Philippova M. Novel immune assay for quantification of plasma protective capacity against oxidized phospholipids. Biomark Med 2016; 10:797-810. [DOI: 10.2217/bmm-2016-0096] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Aim: Oxidized phospholipids (OxPL) are the major pathogenic component of oxidized low-density lipoproteins (OxLDL). Endogenous anti-OxPL activity, defined as the ability to neutralize adverse effects of oxidized lipids, may have biomarker potential. Methods & results: Using two anti-OxPL monoclonal antibodies (commercial mAB-E06 and custom mAB-509) we developed a novel ELISA that measures the global capacity of plasma to inactivate OxPL. Preincubation of OxLDL with plasma inhibits its binding of anti-OxPL mABs. This phenomenon (‘masking’) reflects anti-OxPL plasma activity. A pilot clinical application of the assay revealed reduced anti-OxPL activity in hypertension, coronary artery disease, acute coronary syndrome and diabetes. Conclusion: Inadequate anti-OxPL protection may contribute to cardiovascular disease and have biomarker potential in conditions associated with abnormal lipid peroxidation.
Collapse
Affiliation(s)
- Valery Bochkov
- Institute of Pharmaceutical Sciences, University of Graz, Austria
| | - Andreas W Schoenenberger
- Division of Geriatrics, Department of General Internal Medicine, Inselspital, Bern University Hospital & University of Bern, Bern, Switzerland
| | - Olga Oskolkova
- Institute of Pharmaceutical Sciences, University of Graz, Austria
| | - Ursula Toth
- Department of Vascular Biology & Thombosis Research, Medical University of Vienna, Vienna, Austria
| | - Johannes Stöckl
- Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Otto Majdic
- Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Armond Daci
- Department of Vascular Biology & Thombosis Research, Medical University of Vienna, Vienna, Austria
- Department of Pharmacy, Faculty of Medicine, University of Prishtina, Kosovo
| | - Thérèse J Resink
- Signaling Laboratory, Department of Biomedicine, Basel University Hospital, Basel, Switzerland
| | - Paul Erne
- Signaling Laboratory, Department of Biomedicine, Basel University Hospital, Basel, Switzerland
| | - Maria Philippova
- Signaling Laboratory, Department of Biomedicine, Basel University Hospital, Basel, Switzerland
| |
Collapse
|
114
|
Matsuura E, Kobayashi K, Inoue K, Lopez LR, Shoenfeld Y. Oxidized LDL/β2-glycoprotein I complexes: new aspects in atherosclerosis. Lupus 2016; 14:736-41. [PMID: 16218478 DOI: 10.1191/0961203305lu2211oa] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
β2-glycoprotein I (β2GPI) is a major antigenic target for antiphospholipid antibodies. Oxidized low-density lipoprotein (oxLDL) is the principal lipoprotein found in atherosclerotic lesions, and it colocalizes with β2GPI and immunoreactive lymphocytes. oxLDL/β2GPI complexes appeared in the blood circulation of patients with diseases, such as systemic lupus erythematosus (SLE), antiphospholipid syndrome (APS), systemic sclerosis, diabetes mellitus and chronic renal diseases. Thus, the complexes may be associated with systemic and chronic inflammation of the vasculature. IgG anti-oxLDL/β2GPI complexes autoantibodies and their immune complexes were detected only in SLE/APS patients and in its animal model and were strongly associated with arterial thrombosis. The oxLDL/β2GPI complexes were internalized by macrophages via IgG anti-β2GPI antibody-mediated phagocytosis. In contrast, IgM anti-oxLDL antibodies derived from hyperlipidemic mice reduced the incidence of atherosclerosis. The distribution patterns of IgG and IgM anti-oxLDL antibodies in patients suggest the different roles of these antibodies.
Collapse
Affiliation(s)
- E Matsuura
- Department of Cell Chemistry, Okayama University Graduate School of Medicine and Dentistry, Okayama, Japan.
| | | | | | | | | |
Collapse
|
115
|
Abstract
Ageing, infections and inflammation result in oxidative stress that can irreversibly damage cellular structures. The oxidative damage of lipids in membranes or lipoproteins is one of these deleterious consequences that not only alters lipid function but also leads to the formation of neo-self epitopes - oxidation-specific epitopes (OSEs) - which are present on dying cells and damaged proteins. OSEs represent endogenous damage-associated molecular patterns that are recognized by pattern recognition receptors and the proteins of the innate immune system, and thereby enable the host to sense and remove dangerous biological waste and to maintain homeostasis. If this system is dysfunctional or overwhelmed, the accumulation of OSEs can trigger chronic inflammation and the development of diseases, such as atherosclerosis and age-related macular degeneration. Understanding the molecular components and mechanisms that are involved in this process will help to identify individuals with an increased risk of developing chronic inflammation, and will also help to indicate novel modes of therapeutic intervention.
Collapse
|
116
|
Lobo PI. Role of Natural Autoantibodies and Natural IgM Anti-Leucocyte Autoantibodies in Health and Disease. Front Immunol 2016; 7:198. [PMID: 27375614 PMCID: PMC4893492 DOI: 10.3389/fimmu.2016.00198] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 05/06/2016] [Indexed: 11/13/2022] Open
Abstract
We review how polyreactive natural IgM autoantibodies (IgM-NAA) protect the host from invading micro-organisms and host neo-antigens that are constantly being produced by oxidation mechanisms and cell apoptosis. Second, we discuss how IgM-NAA and IgM anti-leukocyte antibodies (IgM-ALA) inhibits autoimmune inflammation by anti-idiotypic mechanisms, enhancing removal of apoptotic cells, masking neo-antigens, and regulating the function of dendritic cells (DC) and effector cells. Third, we review how natural IgM prevents autoimmune disorders arising from pathogenic IgG autoantibodies, triggered by genetic mechanisms (e.g., SLE) or micro-organisms, as well as by autoreactive B and T cells that have escaped tolerance mechanisms. Studies in IgM knockout mice have clearly demonstrated that regulatory B and T cells require IgM to effectively regulate inflammation mediated by innate, adaptive, and autoimmune mechanisms. It is, therefore, not surprising why the host positively selects such autoreactive B1 cells that generate IgM-NAA, which are also evolutionarily conserved. Fourth, we show that IgM-ALA levels and their repertoire can vary in normal humans and disease states and this variation may partly explain the observed differences in the inflammatory response after infection, ischemic injury, or after a transplant. We also show how protective IgM-NAA can be rendered pathogenic under non-physiological conditions. We also review IgG-NAA that are more abundant than IgM-NAA in plasma. However, we need to understand if the (Fab)(2) region of IgG-NAA has physiological relevance in non-disease states, as in plasma, their functional activity is blocked by IgM-NAA having anti-idiotypic activity. Some IgG-NAA are produced by B2 cells that have escaped tolerance mechanisms and we show how such pathogenic IgG-NAA are regulated to prevent autoimmune disease. The Fc region of IgG-NAA can influence inflammation and B cell function in vivo by binding to activating and inhibitory FcγR. IgM-NAA has therapeutic potential. Polyclonal IgM infusions can be used to abrogate on-going inflammation. Additionally, inflammation arising after ischemic kidney injury, e.g., during high-risk elective cardiac surgery or after allograft transplantation, can be prevented by pre-emptively infusing polyclonal IgM or DC pretreated ex vivo with IgM or by increasing in vivo IgM with a vaccine approach. Cell therapy is appealing as less IgM will be required.
Collapse
Affiliation(s)
- Peter Isaac Lobo
- Department of Internal Medicine, Division of Nephrology, Center of Immunology, Inflammation and Regenerative Medicine, University of Virginia Health Center, Charlottesville, VA, USA
| |
Collapse
|
117
|
Guo X, Meng G, Liu F, Zhang Q, Liu L, Wu H, Du H, Shi H, Xia Y, Liu X, Li C, Bao X, Su Q, Gu Y, Fang L, Yu F, Yang H, Yu B, Sun S, Wang X, Zhou M, Jia Q, Chen X, Huang G, Song K, Niu K. Serum levels of immunoglobulins in an adult population and their relationship with type 2 diabetes. Diabetes Res Clin Pract 2016; 115:76-82. [PMID: 27242126 DOI: 10.1016/j.diabres.2016.03.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 01/29/2016] [Accepted: 03/04/2016] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Some studies have found patients with diabetes had an impaired humoral immune response. Immunoglobulins provide key information on the humoral immune status. But few population-based studies comprehensively estimated the serum immunoglobulins concentration in type 2 diabetes (T2D). So we design a cross-sectional study to investigate the relationships between immunoglobulin levels and prevalence of T2D in a large-scale adult population. METHODS A cross-sectional assessment was performed in 10,691 participants living in Tianjin, China. Type 2 diabetes was defined in accordance with the criteria of the world health organization, and serum levels of immunoglobulins were determined by the immunonephelometric technique. Adjusted logistic models were used to assess relationships between the quintiles of immunoglobulins concentration and the prevalence of T2D. RESULTS In this study, the prevalence of T2D was 11.7%, and the means (standard deviation) of immunoglobulins (IgG, IgE, IgM, IgA) were 1192.3 (241.1)mg/dL, 92.3 (234.6)IU/mL, 104.8 (55.8)mg/dL, 234.1 (96.2)mg/dL, respectively. The adjusted odds ratio (95% confidence interval) of T2D for the highest immunoglobulins (IgG, IgE, IgM, IgA) quintile, when compared to the lowest quintile were 0.64 (0.52, 0.78), 1.00 (0.81, 1.22), 0.77 (0.62, 0.95) and 1.57 (1.29, 1.92), respectively. CONCLUSIONS Decreased IgG and IgM, and increased IgA levels were independently related to the prevalence of T2D among the adult population. Our findings indicate that the immunoglobulins might useful predictive factors for T2D in the general adult population. Further studies are needed to explore the causality and exact mechanisms of immunoglobulins in T2D.
Collapse
Affiliation(s)
- Xiaoyan Guo
- Nutritional Epidemiology Institute and School of Public Health, Tianjin Medical University, Tianjin, China
| | - Ge Meng
- Nutritional Epidemiology Institute and School of Public Health, Tianjin Medical University, Tianjin, China
| | - Fangfang Liu
- Nutritional Epidemiology Institute and School of Public Health, Tianjin Medical University, Tianjin, China
| | - Qing Zhang
- Health Management Centre, Tianjin Medical University General Hospital, Tianjin, China
| | - Li Liu
- Health Management Centre, Tianjin Medical University General Hospital, Tianjin, China
| | - Hongmei Wu
- Nutritional Epidemiology Institute and School of Public Health, Tianjin Medical University, Tianjin, China
| | - Huanmin Du
- Nutritional Epidemiology Institute and School of Public Health, Tianjin Medical University, Tianjin, China
| | - Hongbin Shi
- Health Management Centre, Tianjin Medical University General Hospital, Tianjin, China
| | - Yang Xia
- Nutritional Epidemiology Institute and School of Public Health, Tianjin Medical University, Tianjin, China
| | - Xing Liu
- Nutritional Epidemiology Institute and School of Public Health, Tianjin Medical University, Tianjin, China
| | - Chunlei Li
- Nutritional Epidemiology Institute and School of Public Health, Tianjin Medical University, Tianjin, China
| | - Xue Bao
- Nutritional Epidemiology Institute and School of Public Health, Tianjin Medical University, Tianjin, China
| | - Qian Su
- Nutritional Epidemiology Institute and School of Public Health, Tianjin Medical University, Tianjin, China
| | - Yeqing Gu
- Nutritional Epidemiology Institute and School of Public Health, Tianjin Medical University, Tianjin, China
| | - Liyun Fang
- Nutritional Epidemiology Institute and School of Public Health, Tianjin Medical University, Tianjin, China
| | - Fei Yu
- Nutritional Epidemiology Institute and School of Public Health, Tianjin Medical University, Tianjin, China
| | - Huijun Yang
- Nutritional Epidemiology Institute and School of Public Health, Tianjin Medical University, Tianjin, China
| | - Bin Yu
- Nutritional Epidemiology Institute and School of Public Health, Tianjin Medical University, Tianjin, China
| | - Shaomei Sun
- Health Management Centre, Tianjin Medical University General Hospital, Tianjin, China
| | - Xing Wang
- Health Management Centre, Tianjin Medical University General Hospital, Tianjin, China
| | - Ming Zhou
- Health Management Centre, Tianjin Medical University General Hospital, Tianjin, China
| | - Qiyu Jia
- Health Management Centre, Tianjin Medical University General Hospital, Tianjin, China
| | - Xin Chen
- Nutritional Epidemiology Institute and School of Public Health, Tianjin Medical University, Tianjin, China
| | - Guowei Huang
- Nutritional Epidemiology Institute and School of Public Health, Tianjin Medical University, Tianjin, China
| | - Kun Song
- Health Management Centre, Tianjin Medical University General Hospital, Tianjin, China
| | - Kaijun Niu
- Nutritional Epidemiology Institute and School of Public Health, Tianjin Medical University, Tianjin, China; Health Management Centre, Tianjin Medical University General Hospital, Tianjin, China.
| |
Collapse
|
118
|
Effect of therapeutic interventions on oxidized phospholipids on apolipoprotein B100 and lipoprotein(a). J Clin Lipidol 2016; 10:594-603. [DOI: 10.1016/j.jacl.2016.01.005] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 12/31/2015] [Accepted: 01/26/2016] [Indexed: 11/20/2022]
|
119
|
Rothstein TL. Natural Antibodies as Rheostats for Susceptibility to Chronic Diseases in the Aged. Front Immunol 2016; 7:127. [PMID: 27092140 PMCID: PMC4823301 DOI: 10.3389/fimmu.2016.00127] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 03/21/2016] [Indexed: 02/06/2023] Open
Abstract
Natural antibodies are spontaneously produced in the absence of infection or immunization, and are both anti-microbial and autoreactive. Autoreactive natural antibodies can bind noxious molecules, such as those involved in clinical situations of atherosclerosis (oxLDL), malignancy (NGcGM3), and neurodegeneration (amyloid, tau) and can affect the fate of their targets or the cells bearing them to maintain homeostasis. Clinically relevant natural antibodies have been shown to decline with advancing age in those few situations where measurements have been made. Consistent with this, human B-1 cells that are thought to be responsible for generating natural antibodies also decline with advancing age. These findings together suggest that an age-related decline in amount or efficacy of homeostatic natural antibodies is associated with relative loss of protection against molecules involved in several diseases whose incidence rises in the older age population, and that those individuals experiencing greatest loss are at greatest risk. In this view, natural antibodies act as rheostats for susceptibility to several age-related diseases. These considerations suggest that administration of natural antibodies, or of factors that maintain B-1 cells and/or enhance production of natural antibodies by B-1 cells, may serve to counteract the onset or progression of age-related chronic illness.
Collapse
Affiliation(s)
- Thomas L Rothstein
- Center for Oncology and Cell Biology, The Feinstein Institute for Medical Research , Manhasset, NY , USA
| |
Collapse
|
120
|
Li X, Cao X, Zhang X, Kang Y, Zhang W, Yu M, Ma C, Han J, Duan Y, Chen Y. MEK1/2 inhibitors induce interleukin-5 expression in mouse macrophages and lymphocytes. Biochem Biophys Res Commun 2016; 473:939-946. [PMID: 27045084 DOI: 10.1016/j.bbrc.2016.03.156] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 03/31/2016] [Indexed: 01/27/2023]
Abstract
Uptake of oxidized low-density lipoprotein (oxLDL) by macrophages facilitates the formation of foam cells, the prominent part of atherosclerotic lesions. Interleukin-5 (IL-5) is a cytokine regulating interactions between immune cells. It also activates the production of T15/EO6 IgM antibodies in B-1 cells, which can bind oxLDL thereby demonstrating anti-atherogenic properties. We previously reported that inhibition of extracellular signal-regulated kinases 1 and 2 (ERK1/2) by mitogen-activated protein kinase kinases 1/2 (MEK1/2) inhibitors can reduce atherosclerosis. In this study, we determined the effects of MEK1/2 inhibitors on IL-5 production both in vitro and in vivo. In vitro, MEK1/2 inhibitors (PD98059 and U0126) substantially inhibited phosphorylation of MEK1/2 and ERK1/2. Associated with inhibition of ERK1/2 phosphorylation both in vitro and in vivo, MEK1/2 inhibitors induced IL-5 protein expression in macrophages (RAW macrophages and peritoneal macrophages) and lymphocytes (EL-4 cells). In vivo, administration of mice with MEK1/2 inhibitors increased serum IL-5 levels, and IL-5 protein expression in mouse spleen and liver. At the mechanistic level, we determined that MEK1/2 inhibitors activated IL-5 mRNA expression and IL-5 promoter activity in the liver X receptor (LXR) dependent manner indicating the induction of IL-5 transcription. In addition, we determined that MEK1/2 inhibitors enhanced IL-5 protein stability. Taken together, our study demonstrates that MEK1/2 inhibitors induce IL-5 production which suggests another anti-atherogenic mechanism of MEK1/2 inhibitors.
Collapse
Affiliation(s)
- Xiaoju Li
- College of Life Sciences, Nankai University, Tianjin, China
| | - Xingyue Cao
- College of Life Sciences, Nankai University, Tianjin, China
| | - Xiaomeng Zhang
- College of Life Sciences, Nankai University, Tianjin, China
| | - Yanhua Kang
- School of Medicine, Hangzhou Normal University, Hangzhou, China
| | - Wenwen Zhang
- College of Life Sciences, Nankai University, Tianjin, China
| | - Miao Yu
- College of Life Sciences, Nankai University, Tianjin, China
| | - Chuanrui Ma
- College of Life Sciences, Nankai University, Tianjin, China
| | - Jihong Han
- College of Medical Engineering, Hefei University of Technology, Hefei, China; College of Life Sciences, Nankai University, Tianjin, China
| | - Yajun Duan
- College of Medical Engineering, Hefei University of Technology, Hefei, China; College of Life Sciences, Nankai University, Tianjin, China.
| | - Yuanli Chen
- College of Medical Engineering, Hefei University of Technology, Hefei, China; School of Medicine, Nankai University, Tianjin, China.
| |
Collapse
|
121
|
Yang H, Yan L, Qian P, Duan H, Wu J, Li B, Wang S. Icariin inhibits foam cell formation by down-regulating the expression of CD36 and up-regulating the expression of SR-BI. J Cell Biochem 2016; 116:580-8. [PMID: 25389062 DOI: 10.1002/jcb.25009] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 11/06/2014] [Indexed: 12/21/2022]
Abstract
Icariin is an important pharmacologically active flavonol diglycoside that can inhibit inflammation in lipopolysaccharide (LPS)-stimulated macrophages. However, little is known about the molecular mechanisms underlying the inhibitory effect of Icariin in the formation of foam cells. In this study, macrophages were cultured with LPS and oxidized low-density lipoprotein (oxLDL) in the presence or absence of Icariin. RT-PCR and western blot were used to detect the levels of mRNA and protein expression of CD36, scavenger receptor class B type I (SR-BI) and the phosphorylation of p38MAPK. It was demonstrated that 4 µM or 20 µM Icariin treatment significantly inhibited the cholesterol ester (CE)/total cholesterol (TC) and oxLDL-mediated foam cell formation (P < 0.05). The binding of oxLDL to LPS-activated macrophages was also significantly hindered by Icariin (P < 0.05). Furthermore, Icariin down-regulated the expression of CD36 in LPS-activated macrophages in a dose-dependent manner and CD36 over-expression restored the inhibitory effect of Icariin on foam cell formation. The phosphorylation of p38MAPK was reduced by Icariin, indicating that Icariin reduced the expression of CD36 through the p38MAPK pathway. In addition, Icariin up-regulated SR-BI protein expression in a dose-dependent manner, and SR-BI gene silencing restored the inhibitory effect of Icariin on foam cell formation. These data demonstrate that Icariin inhibited foam cell formation by down-regulating the expression of CD36 and up-regulating the expression of SR-BI. Therefore, our findings provide a new explanation as to why Icariin could inhibit atherosclerosis.
Collapse
Affiliation(s)
- Haitao Yang
- Department of Cardiology, Henan Provincial People's Hospital, Zhengzhou, 450003, China
| | | | | | | | | | | | | |
Collapse
|
122
|
Ley K. 2015 Russell Ross Memorial Lecture in Vascular Biology: Protective Autoimmunity in Atherosclerosis. Arterioscler Thromb Vasc Biol 2016; 36:429-38. [PMID: 26821946 PMCID: PMC4970520 DOI: 10.1161/atvbaha.115.306009] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 01/18/2016] [Indexed: 01/18/2023]
Abstract
Atherosclerosis is an inflammatory disease of the arterial wall. It is accompanied by an autoimmune response against apolipoprotein B-100, the core protein of low-density lipoprotein, which manifests as CD4 T cell and antibody responses. To assess the role of the autoimmune response in atherosclerosis, the nature of the CD4 T cell response against apolipoprotein B-100 was studied with and without vaccination with major histocompatibility complex-II-restricted apolipoprotein B-100 peptides. The immunologic basis of autoimmunity in atherosclerosis is discussed in the framework of theories of adaptive immunity. Older vaccination approaches are also discussed. Vaccinating Apoe(-/-) mice with major histocompatibility complex-II-restricted apolipoprotein B-100 peptides reduces atheroma burden in the aorta by ≈40%. The protective mechanism likely includes secretion of interleukin-10. Protective autoimmunity limits atherosclerosis in mice and suggests potential for developing preventative and therapeutic vaccines for humans.
Collapse
Affiliation(s)
- Klaus Ley
- From the La Jolla Institute for Allergy & Immunology and Department of Bioengineering, UCSD, La Jolla, CA
| |
Collapse
|
123
|
Harmon DB, Srikakulapu P, Kaplan JL, Oldham SN, McSkimming C, Garmey JC, Perry HM, Kirby JL, Prohaska TA, Gonen A, Hallowell P, Schirmer B, Tsimikas S, Taylor AM, Witztum JL, McNamara CA. Protective Role for B-1b B Cells and IgM in Obesity-Associated Inflammation, Glucose Intolerance, and Insulin Resistance. Arterioscler Thromb Vasc Biol 2016; 36:682-91. [PMID: 26868208 DOI: 10.1161/atvbaha.116.307166] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2014] [Accepted: 02/01/2016] [Indexed: 02/04/2023]
Abstract
OBJECTIVE Little is known about the role(s) B cells play in obesity-induced metabolic dysfunction. This study used a mouse with B-cell-specific deletion of Id3 (Id3(Bcell KO)) to identify B-cell functions involved in the metabolic consequences of obesity. APPROACH AND RESULTS Diet-induced obese Id3(Bcell KO) mice demonstrated attenuated inflammation and insulin resistance in visceral adipose tissue (VAT), and improved systemic glucose tolerance. VAT in Id3(Bcell KO) mice had increased B-1b B cells and elevated IgM natural antibodies to oxidation-specific epitopes. B-1b B cells reduced cytokine production in VAT M1 macrophages, and adoptively transferred B-1b B cells trafficked to VAT and produced natural antibodies for the duration of 13-week studies. B-1b B cells null for Id3 demonstrated increased proliferation, established larger populations in Rag1(-/-) VAT, and attenuated diet-induced glucose intolerance and VAT insulin resistance in Rag1(-/-) hosts. However, transfer of B-1b B cells unable to secrete IgM had no effect on glucose tolerance. In an obese human population, results provided the first evidence that B-1 cells are enriched in human VAT and IgM antibodies to oxidation-specific epitopes inversely correlated with inflammation and insulin resistance. CONCLUSIONS NAb-producing B-1b B cells are increased in Id3(Bcell KO) mice and attenuate adipose tissue inflammation and glucose intolerance in diet-induced obese mice. Additional findings are the first to identify VAT as a reservoir for human B-1 cells and to link anti-inflammatory IgM antibodies with reduced inflammation and improved metabolic phenotype in obese humans.
Collapse
Affiliation(s)
- Daniel B Harmon
- From the Cardiovascular Research Center (D.B.H., P.S., J.L.K., S.N.O., C.M.S., J.C.G., H.M.P., C.A.M.), Department of Biochemistry, Molecular Biology, and Genetics (D.B.H.), Division of Cardiovascular Medicine, Department of Medicine (P.S., A.M.T., C.A.M.), Department of Pathology (J.L.K., H.M.P.), Division of Endocrinology and Metabolism, Department of Medicine (J.L.K.), Department of Surgery (P.H., B.S.), Beirne B. Carter Center for Immunology Research (C.A.M.), Department of Molecular Physiology and Biological Physics (C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, University of California San Diego, La Jolla (T.A.P., A.G., S.T., J.L.W.)
| | - Prasad Srikakulapu
- From the Cardiovascular Research Center (D.B.H., P.S., J.L.K., S.N.O., C.M.S., J.C.G., H.M.P., C.A.M.), Department of Biochemistry, Molecular Biology, and Genetics (D.B.H.), Division of Cardiovascular Medicine, Department of Medicine (P.S., A.M.T., C.A.M.), Department of Pathology (J.L.K., H.M.P.), Division of Endocrinology and Metabolism, Department of Medicine (J.L.K.), Department of Surgery (P.H., B.S.), Beirne B. Carter Center for Immunology Research (C.A.M.), Department of Molecular Physiology and Biological Physics (C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, University of California San Diego, La Jolla (T.A.P., A.G., S.T., J.L.W.)
| | - Jennifer L Kaplan
- From the Cardiovascular Research Center (D.B.H., P.S., J.L.K., S.N.O., C.M.S., J.C.G., H.M.P., C.A.M.), Department of Biochemistry, Molecular Biology, and Genetics (D.B.H.), Division of Cardiovascular Medicine, Department of Medicine (P.S., A.M.T., C.A.M.), Department of Pathology (J.L.K., H.M.P.), Division of Endocrinology and Metabolism, Department of Medicine (J.L.K.), Department of Surgery (P.H., B.S.), Beirne B. Carter Center for Immunology Research (C.A.M.), Department of Molecular Physiology and Biological Physics (C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, University of California San Diego, La Jolla (T.A.P., A.G., S.T., J.L.W.)
| | - Stephanie N Oldham
- From the Cardiovascular Research Center (D.B.H., P.S., J.L.K., S.N.O., C.M.S., J.C.G., H.M.P., C.A.M.), Department of Biochemistry, Molecular Biology, and Genetics (D.B.H.), Division of Cardiovascular Medicine, Department of Medicine (P.S., A.M.T., C.A.M.), Department of Pathology (J.L.K., H.M.P.), Division of Endocrinology and Metabolism, Department of Medicine (J.L.K.), Department of Surgery (P.H., B.S.), Beirne B. Carter Center for Immunology Research (C.A.M.), Department of Molecular Physiology and Biological Physics (C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, University of California San Diego, La Jolla (T.A.P., A.G., S.T., J.L.W.)
| | - Chantel McSkimming
- From the Cardiovascular Research Center (D.B.H., P.S., J.L.K., S.N.O., C.M.S., J.C.G., H.M.P., C.A.M.), Department of Biochemistry, Molecular Biology, and Genetics (D.B.H.), Division of Cardiovascular Medicine, Department of Medicine (P.S., A.M.T., C.A.M.), Department of Pathology (J.L.K., H.M.P.), Division of Endocrinology and Metabolism, Department of Medicine (J.L.K.), Department of Surgery (P.H., B.S.), Beirne B. Carter Center for Immunology Research (C.A.M.), Department of Molecular Physiology and Biological Physics (C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, University of California San Diego, La Jolla (T.A.P., A.G., S.T., J.L.W.)
| | - James C Garmey
- From the Cardiovascular Research Center (D.B.H., P.S., J.L.K., S.N.O., C.M.S., J.C.G., H.M.P., C.A.M.), Department of Biochemistry, Molecular Biology, and Genetics (D.B.H.), Division of Cardiovascular Medicine, Department of Medicine (P.S., A.M.T., C.A.M.), Department of Pathology (J.L.K., H.M.P.), Division of Endocrinology and Metabolism, Department of Medicine (J.L.K.), Department of Surgery (P.H., B.S.), Beirne B. Carter Center for Immunology Research (C.A.M.), Department of Molecular Physiology and Biological Physics (C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, University of California San Diego, La Jolla (T.A.P., A.G., S.T., J.L.W.)
| | - Heather M Perry
- From the Cardiovascular Research Center (D.B.H., P.S., J.L.K., S.N.O., C.M.S., J.C.G., H.M.P., C.A.M.), Department of Biochemistry, Molecular Biology, and Genetics (D.B.H.), Division of Cardiovascular Medicine, Department of Medicine (P.S., A.M.T., C.A.M.), Department of Pathology (J.L.K., H.M.P.), Division of Endocrinology and Metabolism, Department of Medicine (J.L.K.), Department of Surgery (P.H., B.S.), Beirne B. Carter Center for Immunology Research (C.A.M.), Department of Molecular Physiology and Biological Physics (C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, University of California San Diego, La Jolla (T.A.P., A.G., S.T., J.L.W.)
| | - Jennifer L Kirby
- From the Cardiovascular Research Center (D.B.H., P.S., J.L.K., S.N.O., C.M.S., J.C.G., H.M.P., C.A.M.), Department of Biochemistry, Molecular Biology, and Genetics (D.B.H.), Division of Cardiovascular Medicine, Department of Medicine (P.S., A.M.T., C.A.M.), Department of Pathology (J.L.K., H.M.P.), Division of Endocrinology and Metabolism, Department of Medicine (J.L.K.), Department of Surgery (P.H., B.S.), Beirne B. Carter Center for Immunology Research (C.A.M.), Department of Molecular Physiology and Biological Physics (C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, University of California San Diego, La Jolla (T.A.P., A.G., S.T., J.L.W.)
| | - Thomas A Prohaska
- From the Cardiovascular Research Center (D.B.H., P.S., J.L.K., S.N.O., C.M.S., J.C.G., H.M.P., C.A.M.), Department of Biochemistry, Molecular Biology, and Genetics (D.B.H.), Division of Cardiovascular Medicine, Department of Medicine (P.S., A.M.T., C.A.M.), Department of Pathology (J.L.K., H.M.P.), Division of Endocrinology and Metabolism, Department of Medicine (J.L.K.), Department of Surgery (P.H., B.S.), Beirne B. Carter Center for Immunology Research (C.A.M.), Department of Molecular Physiology and Biological Physics (C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, University of California San Diego, La Jolla (T.A.P., A.G., S.T., J.L.W.)
| | - Ayelet Gonen
- From the Cardiovascular Research Center (D.B.H., P.S., J.L.K., S.N.O., C.M.S., J.C.G., H.M.P., C.A.M.), Department of Biochemistry, Molecular Biology, and Genetics (D.B.H.), Division of Cardiovascular Medicine, Department of Medicine (P.S., A.M.T., C.A.M.), Department of Pathology (J.L.K., H.M.P.), Division of Endocrinology and Metabolism, Department of Medicine (J.L.K.), Department of Surgery (P.H., B.S.), Beirne B. Carter Center for Immunology Research (C.A.M.), Department of Molecular Physiology and Biological Physics (C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, University of California San Diego, La Jolla (T.A.P., A.G., S.T., J.L.W.)
| | - Peter Hallowell
- From the Cardiovascular Research Center (D.B.H., P.S., J.L.K., S.N.O., C.M.S., J.C.G., H.M.P., C.A.M.), Department of Biochemistry, Molecular Biology, and Genetics (D.B.H.), Division of Cardiovascular Medicine, Department of Medicine (P.S., A.M.T., C.A.M.), Department of Pathology (J.L.K., H.M.P.), Division of Endocrinology and Metabolism, Department of Medicine (J.L.K.), Department of Surgery (P.H., B.S.), Beirne B. Carter Center for Immunology Research (C.A.M.), Department of Molecular Physiology and Biological Physics (C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, University of California San Diego, La Jolla (T.A.P., A.G., S.T., J.L.W.)
| | - Bruce Schirmer
- From the Cardiovascular Research Center (D.B.H., P.S., J.L.K., S.N.O., C.M.S., J.C.G., H.M.P., C.A.M.), Department of Biochemistry, Molecular Biology, and Genetics (D.B.H.), Division of Cardiovascular Medicine, Department of Medicine (P.S., A.M.T., C.A.M.), Department of Pathology (J.L.K., H.M.P.), Division of Endocrinology and Metabolism, Department of Medicine (J.L.K.), Department of Surgery (P.H., B.S.), Beirne B. Carter Center for Immunology Research (C.A.M.), Department of Molecular Physiology and Biological Physics (C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, University of California San Diego, La Jolla (T.A.P., A.G., S.T., J.L.W.)
| | - Sotirios Tsimikas
- From the Cardiovascular Research Center (D.B.H., P.S., J.L.K., S.N.O., C.M.S., J.C.G., H.M.P., C.A.M.), Department of Biochemistry, Molecular Biology, and Genetics (D.B.H.), Division of Cardiovascular Medicine, Department of Medicine (P.S., A.M.T., C.A.M.), Department of Pathology (J.L.K., H.M.P.), Division of Endocrinology and Metabolism, Department of Medicine (J.L.K.), Department of Surgery (P.H., B.S.), Beirne B. Carter Center for Immunology Research (C.A.M.), Department of Molecular Physiology and Biological Physics (C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, University of California San Diego, La Jolla (T.A.P., A.G., S.T., J.L.W.)
| | - Angela M Taylor
- From the Cardiovascular Research Center (D.B.H., P.S., J.L.K., S.N.O., C.M.S., J.C.G., H.M.P., C.A.M.), Department of Biochemistry, Molecular Biology, and Genetics (D.B.H.), Division of Cardiovascular Medicine, Department of Medicine (P.S., A.M.T., C.A.M.), Department of Pathology (J.L.K., H.M.P.), Division of Endocrinology and Metabolism, Department of Medicine (J.L.K.), Department of Surgery (P.H., B.S.), Beirne B. Carter Center for Immunology Research (C.A.M.), Department of Molecular Physiology and Biological Physics (C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, University of California San Diego, La Jolla (T.A.P., A.G., S.T., J.L.W.)
| | - Joseph L Witztum
- From the Cardiovascular Research Center (D.B.H., P.S., J.L.K., S.N.O., C.M.S., J.C.G., H.M.P., C.A.M.), Department of Biochemistry, Molecular Biology, and Genetics (D.B.H.), Division of Cardiovascular Medicine, Department of Medicine (P.S., A.M.T., C.A.M.), Department of Pathology (J.L.K., H.M.P.), Division of Endocrinology and Metabolism, Department of Medicine (J.L.K.), Department of Surgery (P.H., B.S.), Beirne B. Carter Center for Immunology Research (C.A.M.), Department of Molecular Physiology and Biological Physics (C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, University of California San Diego, La Jolla (T.A.P., A.G., S.T., J.L.W.)
| | - Coleen A McNamara
- From the Cardiovascular Research Center (D.B.H., P.S., J.L.K., S.N.O., C.M.S., J.C.G., H.M.P., C.A.M.), Department of Biochemistry, Molecular Biology, and Genetics (D.B.H.), Division of Cardiovascular Medicine, Department of Medicine (P.S., A.M.T., C.A.M.), Department of Pathology (J.L.K., H.M.P.), Division of Endocrinology and Metabolism, Department of Medicine (J.L.K.), Department of Surgery (P.H., B.S.), Beirne B. Carter Center for Immunology Research (C.A.M.), Department of Molecular Physiology and Biological Physics (C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, University of California San Diego, La Jolla (T.A.P., A.G., S.T., J.L.W.).
| |
Collapse
|
124
|
Lu C, Zhou F, Wu S, Liu L, Xing D. Phototherapy-Induced Antitumor Immunity: Long-Term Tumor Suppression Effects via Photoinactivation of Respiratory Chain Oxidase-Triggered Superoxide Anion Burst. Antioxid Redox Signal 2016; 24:249-62. [PMID: 26413929 DOI: 10.1089/ars.2015.6334] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
AIMS Our previous studies have demonstrated that as a mitochondria-targeting cancer phototherapy, high-fluence, low-power laser irradiation (HF-LPLI) results in oxidative damage that induces tumor cell apoptosis. In this study, we focused on the immunological effects of HF-LPLI phototherapy and explored its antitumor immune regulatory mechanism. RESULTS We found not only that HF-LPLI treatment induced tumor cell apoptosis but also that HF-LPLI-treated apoptotic tumor cells activated macrophages. Due to mitochondrial superoxide anion burst after HF-LPLI treatment, tumor cells displayed a high level of phosphatidylserine oxidation, which mediated the recognition and uptake by macrophages with the subsequent secretion of cytokines and generation of cytotoxic T lymphocytes. In addition, in vivo results showed that HF-LPLI treatment caused leukocyte infiltration into the tumor and efficaciously inhibited tumor growth in an EMT6 tumor model. These phenomena were absent in the respiration-deficient EMT6 tumor model, implying that the HF-LPLI-elicited immunological effects were dependent on the mitochondrial superoxide anion burst. INNOVATION In this study, for the first time, we show that HF-LPLI mediates tumor-killing effects via targeting photoinactivation of respiratory chain oxidase to trigger a superoxide anion burst, leading to a high level of oxidatively modified moieties, which contributes to the phenotypic changes in macrophages and mediates the antitumor immune response. CONCLUSION Our results suggest that HF-LPLI may be an effective cancer treatment modality that both eradicates the treated primary tumors and induces an antitumor immune response via photoinactivation of respiratory chain oxidase to trigger superoxide anion burst.
Collapse
Affiliation(s)
- Cuixia Lu
- 1 MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University , Guangzhou, China
| | - Feifan Zhou
- 1 MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University , Guangzhou, China .,2 Joint Laboratory of Laser Oncology with Cancer Center of Sun Yat-sen University, South China Normal University , Guangzhou, China
| | - Shengnan Wu
- 1 MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University , Guangzhou, China .,2 Joint Laboratory of Laser Oncology with Cancer Center of Sun Yat-sen University, South China Normal University , Guangzhou, China
| | - Lei Liu
- 1 MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University , Guangzhou, China
| | - Da Xing
- 1 MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University , Guangzhou, China .,2 Joint Laboratory of Laser Oncology with Cancer Center of Sun Yat-sen University, South China Normal University , Guangzhou, China
| |
Collapse
|
125
|
Yeang C, Cotter B, Tsimikas S. Experimental Animal Models Evaluating the Causal Role of Lipoprotein(a) in Atherosclerosis and Aortic Stenosis. Cardiovasc Drugs Ther 2016; 30:75-85. [DOI: 10.1007/s10557-015-6634-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
126
|
Vavuli S, Salonurmi T, Loukovaara S, Nissinen AE, Savolainen MJ, Liinamaa MJ. Elevated Levels of Plasma IgA Autoantibodies against Oxidized LDL Found in Proliferative Diabetic Retinopathy but Not in Nonproliferative Retinopathy. J Diabetes Res 2016; 2016:2614153. [PMID: 28090539 PMCID: PMC5206457 DOI: 10.1155/2016/2614153] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 11/10/2016] [Accepted: 11/28/2016] [Indexed: 01/08/2023] Open
Abstract
Aims. This study investigated the association of autoantibodies binding to oxidized low-density lipoproteins (oxLDL) in diabetic retinopathy (DR). Methods. Plasma from 229 types 1 and 2 patients with DR including diabetic macular edema (DME) and proliferative diabetic retinopathy (PDR) was analysed with ELISA-based assay to determine IgA, IgG, and IgM autoantibody levels binding to oxLDL. The controls were 106 diabetic patients without retinopathy (NoDR) and 139 nondiabetic controls (C). Results. PDR group had significantly higher IgA autoantibody levels than DME or NoDR: mean 94.9 (SD 54.7) for PDR, 75.5 (41.8) for DME (p = 0.001), and 76.1 (48.2) for NoDR (p = 0.008). There were no differences in IgG, IgM, or IgA that would be specific for DR or for DME. Type 2 diabetic patients had higher levels of IgA autoantibodies than type 1 diabetic patients (86.0 and 65.5, resp., p = 0.004) and the highest levels in IgA were found in type 2 diabetic patients with PDR (119.1, p > 0.001). Conclusions. IgA autoantibodies were increased in PDR, especially in type 2 diabetes. The high levels of IgA in PDR, and especially in type 2 PDR patients, reflect the inflammatory process and enlighten the role of oxLDL and its autoantibodies in PDR.
Collapse
Affiliation(s)
- Satu Vavuli
- PEDEGO Research Unit, Department of Ophthalmology, Medical Research Center (MRC Oulu), Oulu University Hospital and University of Oulu, Oulu, Finland
- Research Unit of Internal Medicine, Medical Research Center Oulu (MRC Oulu), Oulu University Hospital and University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Tuire Salonurmi
- Research Unit of Internal Medicine, Medical Research Center Oulu (MRC Oulu), Oulu University Hospital and University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Sirpa Loukovaara
- Department of Ophthalmology, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Antti E. Nissinen
- Research Unit of Internal Medicine, Medical Research Center Oulu (MRC Oulu), Oulu University Hospital and University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
- Research Unit of Biomedicine, Medical Research Center Oulu (MRC Oulu), Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Markku J. Savolainen
- Research Unit of Internal Medicine, Medical Research Center Oulu (MRC Oulu), Oulu University Hospital and University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
| | - M. Johanna Liinamaa
- PEDEGO Research Unit, Department of Ophthalmology, Medical Research Center (MRC Oulu), Oulu University Hospital and University of Oulu, Oulu, Finland
- Research Unit of Internal Medicine, Medical Research Center Oulu (MRC Oulu), Oulu University Hospital and University of Oulu, Oulu, Finland
- *M. Johanna Liinamaa:
| |
Collapse
|
127
|
Centa M, Gruber S, Nilsson D, Polyzos KA, Johansson DK, Hansson GK, Ketelhuth DFJ, Binder CJ, Malin S. Atherosclerosis Susceptibility in Mice Is Independent of the V1 Immunoglobulin Heavy Chain Gene. Arterioscler Thromb Vasc Biol 2015; 36:25-36. [PMID: 26564818 PMCID: PMC4684249 DOI: 10.1161/atvbaha.115.305990] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 11/04/2015] [Indexed: 12/02/2022]
Abstract
Supplemental Digital Content is available in the text. The V1 (VHS107.1.42) immunoglobulin heavy chain gene is thought to be critical in producing IgM natural antibodies of the T15-idiotype that protect against both atherosclerosis and infection from Streptococcus pneumoniae. Our aim was to determine whether genetic loss of the V1 gene increased atherosclerotic plaque burden in vivo because of a reduction in the T15-idiotype or other atheroprotective antibodies.
Collapse
Affiliation(s)
- Monica Centa
- From the Department of Medicine and Center for Molecular Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden (M.C., D.N., K.A.P., D.K.J., G.K.H., D.F.J.K., S.M.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria (S.G., C.J.B.); and Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria (S.G., C.J.B.)
| | - Sabrina Gruber
- From the Department of Medicine and Center for Molecular Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden (M.C., D.N., K.A.P., D.K.J., G.K.H., D.F.J.K., S.M.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria (S.G., C.J.B.); and Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria (S.G., C.J.B.)
| | - Daniel Nilsson
- From the Department of Medicine and Center for Molecular Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden (M.C., D.N., K.A.P., D.K.J., G.K.H., D.F.J.K., S.M.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria (S.G., C.J.B.); and Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria (S.G., C.J.B.)
| | - Konstantinos A Polyzos
- From the Department of Medicine and Center for Molecular Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden (M.C., D.N., K.A.P., D.K.J., G.K.H., D.F.J.K., S.M.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria (S.G., C.J.B.); and Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria (S.G., C.J.B.)
| | - Daniel K Johansson
- From the Department of Medicine and Center for Molecular Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden (M.C., D.N., K.A.P., D.K.J., G.K.H., D.F.J.K., S.M.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria (S.G., C.J.B.); and Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria (S.G., C.J.B.)
| | - Göran K Hansson
- From the Department of Medicine and Center for Molecular Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden (M.C., D.N., K.A.P., D.K.J., G.K.H., D.F.J.K., S.M.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria (S.G., C.J.B.); and Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria (S.G., C.J.B.)
| | - Daniel F J Ketelhuth
- From the Department of Medicine and Center for Molecular Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden (M.C., D.N., K.A.P., D.K.J., G.K.H., D.F.J.K., S.M.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria (S.G., C.J.B.); and Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria (S.G., C.J.B.)
| | - Christoph J Binder
- From the Department of Medicine and Center for Molecular Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden (M.C., D.N., K.A.P., D.K.J., G.K.H., D.F.J.K., S.M.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria (S.G., C.J.B.); and Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria (S.G., C.J.B.)
| | - Stephen Malin
- From the Department of Medicine and Center for Molecular Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden (M.C., D.N., K.A.P., D.K.J., G.K.H., D.F.J.K., S.M.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria (S.G., C.J.B.); and Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria (S.G., C.J.B.).
| |
Collapse
|
128
|
Plasma PAF-AH (PLA2G7): Biochemical Properties, Association with LDLs and HDLs, and Regulation of Expression. Enzymes 2015; 38:71-93. [PMID: 26612648 DOI: 10.1016/bs.enz.2015.09.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
This chapter is focused on the plasma form of PAF-acetylhydrolase (PAF-AH), a lipoprotein-bound, calcium-independent phospholipase A2 activity also referred to as lipoprotein-associated phospholipase A2 and PLA2G7. PAF-AH catalyzes the removal of the acyl group at the sn-2 position of PAF and truncated phospholipids generated in settings of inflammation and oxidant stress. Here, I discuss current knowledge related to the structural features of this enzyme, including the molecular basis for association with lipoproteins and susceptibility to oxidative inactivation. The circulating form of PAF-AH is constitutively active and its expression is upregulated by mediators of inflammation at the transcriptional level. Several new mechanisms of regulation have been identified in recent years, including effects mediated by PPARs, VEGFR, and the state of cellular differentiation. Moreover, I discuss recent studies describing significant variations in the structure and regulation of PAF-AH from diverse species, which is likely to have important implications for the function of this enzyme in vivo.
Collapse
|
129
|
Wolf D, Zirlik A, Ley K. Beyond vascular inflammation--recent advances in understanding atherosclerosis. Cell Mol Life Sci 2015; 72:3853-69. [PMID: 26100516 PMCID: PMC4577451 DOI: 10.1007/s00018-015-1971-6] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2015] [Revised: 06/10/2015] [Accepted: 06/15/2015] [Indexed: 12/23/2022]
Abstract
Atherosclerosis is the most life-threatening pathology worldwide. Its major clinical complications, stroke, myocardial infarction, and heart failure, are on the rise in many regions of the world--despite considerable progress in understanding cause, progression, and consequences of atherosclerosis. Originally perceived as a lipid-storage disease of the arterial wall (Die cellularpathologie in ihrer begründung auf physiologische und pathologische gewebelehre. August Hirschwald Verlag Berlin, [1871]), atherosclerosis was recognized as a chronic inflammatory disease in 1986 (New Engl J Med 314:488-500, 1986). The presence of lymphocytes in atherosclerotic lesions suggested autoimmune processes in the vessel wall (Clin Exp Immunol 64:261-268, 1986). Since the advent of suitable mouse models of atherosclerosis (Science 258:468-471, 1992; Cell 71:343-353, 1992; J Clin Invest 92:883-893, 1993) and the development of flow cytometry to define the cellular infiltrate in atherosclerotic lesions (J Exp Med 203:1273-1282, 2006), the origin, lineage, phenotype, and function of distinct inflammatory cells that trigger or inhibit the inflammatory response in the atherosclerotic plaque have been studied. Multiphoton microscopy recently enabled direct visualization of antigen-specific interactions between T cells and antigen-presenting cells in the vessel wall (J Clin Invest 122:3114-3126, 2012). Vascular immunology is now emerging as a new field, providing evidence for protective as well as damaging autoimmune responses (Int Immunol 25:615-622, 2013). Manipulating inflammation and autoimmunity both hold promise for new therapeutic strategies in cardiovascular disease. Ongoing work (J Clin Invest 123:27-36, 2013; Front Immunol 2013; Semin Immunol 31:95-101, 2009) suggests that it may be possible to develop antigen-specific immunomodulatory prevention and therapy-a vaccine against atherosclerosis.
Collapse
Affiliation(s)
- Dennis Wolf
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle Drive, La Jolla, CA, 92037, USA
| | - Andreas Zirlik
- Atherogenesis Research Group, Cardiology and Angiology I, Heart Center, University of Freiburg, Freiburg, Germany
| | - Klaus Ley
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle Drive, La Jolla, CA, 92037, USA.
| |
Collapse
|
130
|
Improved intervention of atherosclerosis and cardiac hypertrophy through biodegradable polymer-encapsulated delivery of glycosphingolipid inhibitor. Biomaterials 2015; 64:125-135. [PMID: 26111596 DOI: 10.1016/j.biomaterials.2015.06.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Revised: 05/28/2015] [Accepted: 06/01/2015] [Indexed: 01/22/2023]
Abstract
D-Threo-1-phenyl-2-decanoylamino-3-morpholino-1-propanol (D-PDMP), a glycosphingolipid synthesis inhibitor, holds promise for the treatment of atherosclerosis and cardiac hypertrophy but rapid in vivo clearance has severely hindered translation to the clinic. To overcome this impediment, we used a materials-based delivery strategy wherein D-PDMP was encapsulated within a biodegradable polymer composed of poly ethylene glycol (PEG) and sebacic acid (SA). PEG-SA was formulated into nanoparticles that were doped with (125)I-labeled PEG to allow in vivo bio-distribution and release kinetics of D-PDMP to be determined by using γ-scintigraphy and subsequently, by mass spectrometry. Polymer-encapsulation increased the residence time of D-PDMP in the body of a treated mouse from less than one hour to at least four hours (and up to 48 h or longer). This substantially increased in vivo longevity provided by polymer encapsulation resulted in an order of magnitude gain in efficacy for interfering with atherosclerosis and cardiac hypertrophy in apoE-/- mice fed a high fat and high cholesterol (HFHC) diet. These results establish that D-PDMP encapsulated in a biodegradable polymer provides a superior mode of delivery compared to unconjugated D-PDMP by way of increased gastrointestinal absorption and increased residence time thus providing this otherwise rapidly cleared compound with therapeutic relevance in interfering with atherosclerosis, cardiac hypertrophy, and probably other diseases associated with the deleterious effects of abnormally high glycosphingolipid biosynthesis or deficient catabolism.
Collapse
|
131
|
Yeang C, Tsimikas S. HDL-C, ABCA1-mediated cholesterol efflux, and lipoprotein(a): insights into a potential novel physiologic role of lipoprotein(a). J Lipid Res 2015; 56:1241-4. [PMID: 26014961 DOI: 10.1194/jlr.e060947] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Calvin Yeang
- Division of Cardiovascular Medicine, University of California San Diego, Sulpizio Cardiovascular Center, La Jolla, CA
| | - Sotirios Tsimikas
- Division of Cardiovascular Medicine, University of California San Diego, Sulpizio Cardiovascular Center, La Jolla, CA
| |
Collapse
|
132
|
Inflammatory mediators in vascular disease: identifying promising targets for intracranial aneurysm research. Mediators Inflamm 2015; 2015:896283. [PMID: 25922566 PMCID: PMC4397479 DOI: 10.1155/2015/896283] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Revised: 03/18/2015] [Accepted: 03/19/2015] [Indexed: 12/21/2022] Open
Abstract
Inflammatory processes are implicated in many diseases of the vasculature and have been shown to play a key role in the formation of intracranial aneurysms (IAs). Although the specific mechanisms underlying these processes have been thoroughly investigated in related pathologies, such as atherosclerosis, there remains a paucity of information regarding the immunopathology of IA. Cells such as macrophages and lymphocytes and their effector molecules have been suggested to be players in IA, but their specific interactions and the role of other components of the inflammatory response have yet to be determined. Drawing parallels between the pathogenesis of IA and other vascular disorders could provide a roadmap for developing a mechanistic understanding of the immunopathology of IA and uncovering useful targets for therapeutic intervention. Future research should address the presence and function of leukocyte subsets, mechanisms of leukocyte recruitment and activation, and the role of damage-associated molecular patterns in IA.
Collapse
|
133
|
Khoo LHB, Thiam CH, Soh SY, Angeli V. Splenic extrafollicular reactions and BM plasma cells sustain IgM response associated with hypercholesterolemia. Eur J Immunol 2015; 45:1300-12. [DOI: 10.1002/eji.201344347] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Revised: 01/14/2015] [Accepted: 01/29/2015] [Indexed: 11/06/2022]
Affiliation(s)
- Lawrence Han Boon Khoo
- Department of Microbiology; Immunology Programme; National University of Singapore; Singapore
- Singapore Immunology Network; Agency for Science; Technology and Research; Biopolis Singapore
| | - Chung Hwee Thiam
- Department of Microbiology; Immunology Programme; National University of Singapore; Singapore
| | - Serena Ying Soh
- Department of Microbiology; Immunology Programme; National University of Singapore; Singapore
| | - Véronique Angeli
- Department of Microbiology; Immunology Programme; National University of Singapore; Singapore
| |
Collapse
|
134
|
Geller BJ, Mega JL, Morrow DA, Guo J, Hoffman EB, Gibson CM, Ruff CT. Autoantibodies to phosphorylcholine and cardiovascular outcomes in patients with acute coronary syndromes in the ATLAS ACS-TIMI 46 trial. J Thromb Thrombolysis 2015; 37:310-6. [PMID: 23860881 DOI: 10.1007/s11239-013-0968-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Atherogenesis is a complex inflammatory process stemming from the accumulation and oxidation of low density lipoproteins (LDL). IgM autoantibodies against phosphorylcholine (anti-PC) bind to the PC epitope on oxidized LDL (OxLDL), inhibiting the uptake of oxLDL by macrophages in atherosclerotic lesions. Anti-PC autoantibodies have been reported to be protective against atherothrombosis. We investigated the relationship of anti-PC concentrations with cardiovascular outcomes in patients with acute coronary syndromes (ACS). We measured anti-PC levels within 7 days of an ACS in 3,356 patients enrolled in the ATLAS ACS-TIMI 46 trial, a randomized dose ranging study of rivaroxaban versus placebo. The primary endpoint was death, myocardial infarction (MI), stroke, or severe recurrent ischemia (SRI) requiring revascularization during 6 months. The median baseline anti-PC concentration was 40.9 U/mL (25th, 75th percentiles: 25.4, 67.4). There was no significant association between anti-PC levels and the primary endpoint (Q1: 6.8 %, Q2: 4.2 %, Q3: 7.8 %, Q4: 5.4 %, p-trend = 0.87), all-cause mortality (Q1: 1.4 %, Q2: 0.7 %, Q3: 2.4 %, Q4: 0.9 %, p-trend = 0. 96), or any of the other individual endpoint components (MI: p-trend = 0.87, Stroke: p-trend = 0.43, SRI: p-trend = 0.66). Using the previously reported anti-PC cutpoint of 17 U/mL did not reveal a significant relationship between anti-PC concentrations and cardiovascular outcomes (<17 U/mL: 8.1 % vs. ≥17 U/mL: 5.8 %; p = 0.11). Similarly, evaluation of anti-PC as a continuous variable did not reveal a significant association (p = 0.30). In this study of patients early after ACS undergoing intensive secondary preventive therapy, IgM anti-PC titers did not exhibit a significant relationship with cardiovascular outcomes.
Collapse
Affiliation(s)
- Bram J Geller
- TIMI Study Group, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 350 Longwood Avenue, First Floor, Boston, MA, USA,
| | | | | | | | | | | | | |
Collapse
|
135
|
Tsiantoulas D, Perkmann T, Afonyushkin T, Mangold A, Prohaska TA, Papac-Milicevic N, Millischer V, Bartel C, Hörkkö S, Boulanger CM, Tsimikas S, Fischer MB, Witztum JL, Lang IM, Binder CJ. Circulating microparticles carry oxidation-specific epitopes and are recognized by natural IgM antibodies. J Lipid Res 2014; 56:440-8. [PMID: 25525116 PMCID: PMC4306697 DOI: 10.1194/jlr.p054569] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Oxidation-specific epitopes (OSEs) present on apoptotic cells and oxidized low density lipoprotein (OxLDL) represent danger-associated molecular patterns that are recognized by different arcs of innate immunity, including natural IgM antibodies. Here, we investigated whether circulating microparticles (MPs), which are small membrane vesicles released by apoptotic or activated cells, are physiological carriers of OSEs. OSEs on circulating MPs isolated from healthy donors and patients with ST-segment elevation myocardial infarction (STE-MI) were characterized by flow cytometry using a panel of OSE-specific monoclonal antibodies. We found that a subset of MPs carry OSEs on their surface, predominantly malondialdehyde (MDA) epitopes. Consistent with this, a majority of IgM antibodies bound on the surface of circulating MPs were found to have specificity for MDA-modified LDL. Moreover, we show that MPs can stimulate THP-1 (human acute monocytic leukemia cell line) and human primary monocytes to produce interleukin 8, which can be inhibited by a monoclonal IgM with specificity for MDA epitopes. Finally, we show that MDA+ MPs are elevated at the culprit lesion site of patients with STE-MI. Our results identify a subset of OSE+ MPs that are bound by OxLDL-specific IgM. These findings demonstrate a novel mechanism by which anti-OxLDL IgM antibodies could mediate protective functions in CVD.
Collapse
Affiliation(s)
- Dimitrios Tsiantoulas
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences, Vienna, Austria Christian Doppler Laboratory for Innovative Therapy Approaches in Sepsis, Krems, Austria
| | - Thomas Perkmann
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Taras Afonyushkin
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences, Vienna, Austria
| | - Andreas Mangold
- Department of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Thomas A Prohaska
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences, Vienna, Austria
| | - Nikolina Papac-Milicevic
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences, Vienna, Austria
| | - Vincent Millischer
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences, Vienna, Austria
| | - Caroline Bartel
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Sohvi Hörkkö
- Medical Research Center and Department of Medical Microbiology and Immunology, Institute of Diagnostics, University of Oulu, Oulu, Finland
| | | | - Sotirios Tsimikas
- Department of Medicine, University of California San Diego, La Jolla, CA
| | - Michael B Fischer
- Christian Doppler Laboratory for Innovative Therapy Approaches in Sepsis, Krems, Austria Department of Blood Group Serology Transfusion Medicine, Medical University of Vienna, Vienna, Austria
| | - Joseph L Witztum
- Department of Medicine, University of California San Diego, La Jolla, CA
| | - Irene M Lang
- Department of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences, Vienna, Austria Christian Doppler Laboratory for Innovative Therapy Approaches in Sepsis, Krems, Austria
| |
Collapse
|
136
|
Shah PK, Chyu KY, Dimayuga PC, Nilsson J. Vaccine for Atherosclerosis. J Am Coll Cardiol 2014; 64:2779-91. [DOI: 10.1016/j.jacc.2014.10.018] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 10/08/2014] [Accepted: 10/10/2014] [Indexed: 11/25/2022]
|
137
|
Abstract
Atherosclerosis is a chronic inflammatory disease of the artery wall. Atherosclerotic lesions contain monocytes, macrophages, smooth muscle cells and T lymphocytes. Here, we review the role of T-lymphocyte subsets in atherosclerosis. Among CD4⁺T cells, T(h)1 cells are pro-atherogenic, T(reg) cells are athero-protective and the role of T(h)2 and T(h)17 cells remains unclear. The role of follicular helper T cells in atherosclerosis remains unknown, as is the role of CD8⁺T cells. NKT cells bind glycolipid antigens and exert a pro-atherogenic role. The antigen specificity of T-cell responses in atherosclerosis is poorly understood. In order to enable antigen-specific prevention or therapy, a better understanding of these mechanisms is needed.
Collapse
Affiliation(s)
- Kevin Tse
- Department of Internal Medicine, Division of Rheumatology, Allergy and Immunology, University of California at San Diego Medical Center, La Jolla, CA 92093, USA
| | | | | | | | | |
Collapse
|
138
|
Sreckovic I, Birner-Gruenberger R, Besenboeck C, Miljkovic M, Stojakovic T, Scharnagl H, Marsche G, Lang U, Kotur-Stevuljevic J, Jelic-Ivanovic Z, Desoye G, Wadsack C. Gestational diabetes mellitus modulates neonatal high-density lipoprotein composition and its functional heterogeneity. Biochim Biophys Acta Mol Cell Biol Lipids 2014; 1841:1619-27. [DOI: 10.1016/j.bbalip.2014.07.021] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Revised: 07/14/2014] [Accepted: 07/29/2014] [Indexed: 01/26/2023]
|
139
|
Ganini D, Mason RP. Absence of an effect of vitamin E on protein and lipid radical formation during lipoperoxidation of LDL by lipoxygenase. Free Radic Biol Med 2014; 76:61-8. [PMID: 25091900 PMCID: PMC4252844 DOI: 10.1016/j.freeradbiomed.2014.07.031] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Revised: 07/09/2014] [Accepted: 07/24/2014] [Indexed: 02/07/2023]
Abstract
Low-density lipoprotein (LDL) oxidation is the primary event in atherosclerosis, and LDL lipoperoxidation leads to modifications in apolipoprotein B-100 (apo B-100) and lipids. Intermediate species of lipoperoxidation are known to be able to generate amino acid-centered radicals. Thus, we hypothesized that lipoperoxidation intermediates induce protein-derived free radical formation during LDL oxidation. Using DMPO and immuno-spin trapping, we detected the formation of protein free radicals on LDL incubated with Cu(2+) or the soybean lipoxidase (LPOx)/phospholipase A2 (PLA2). With low concentrations of DMPO (1mM), Cu(2+) dose-dependently induced oxidation of LDL and easily detected apo B-100 radicals. Protein radical formation in LDL incubated with Cu(2+) showed maximum yields after 30 min. In contrast, the yields of apo B-100 radicals formed by LPOx/PLA2 followed a typical enzyme-catalyzed kinetics that was unaffected by DMPO concentrations of up to 50mM. Furthermore, when we analyzed the effect of antioxidants on protein radical formation during LDL oxidation, we found that ascorbate, urate, and Trolox dose-dependently reduced apo B-100 free radical formation in LDL exposed to Cu(2+). In contrast, Trolox was the only antioxidant that even partially protected LDL from LPOx/PLA2. We also examined the kinetics of lipid radical formation and protein radical formation induced by Cu(2+) or LPOx/PLA2 for LDL supplemented with α-tocopherol. In contrast to the potent antioxidant effect of α-tocopherol on the delay of LDL oxidation induced by Cu(2+), when we used the oxidizing system LPOx/PLA2, no significant protection was detected. The lack of protection of α-tocopherol on the apo B-100 and lipid free radical formation by LPOx may explain the failure of vitamin E as a cardiovascular protective agent for humans.
Collapse
Affiliation(s)
- Douglas Ganini
- Free Radical Metabolism Section, Laboratory of Toxicology and Pharmacology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA.
| | - Ronald P Mason
- Free Radical Metabolism Section, Laboratory of Toxicology and Pharmacology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| |
Collapse
|
140
|
Baldan A, Gonen A, Choung C, Que X, Marquart TJ, Hernandez I, Bjorkhem I, Ford DA, Witztum JL, Tarling EJ. ABCG1 is required for pulmonary B-1 B cell and natural antibody homeostasis. THE JOURNAL OF IMMUNOLOGY 2014; 193:5637-48. [PMID: 25339664 DOI: 10.4049/jimmunol.1400606] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Many metabolic diseases, including atherosclerosis, type 2 diabetes, pulmonary alveolar proteinosis, and obesity, have a chronic inflammatory component involving both innate and adaptive immunity. Mice lacking the ATP-binding cassette transporter G1 (ABCG1) develop chronic inflammation in the lungs, which is associated with the lipid accumulation (cholesterol, cholesterol ester, and phospholipid) and cholesterol crystal deposition that are characteristic of atherosclerotic lesions and pulmonary alveolar proteinosis. In this article, we demonstrate that specific lipids, likely oxidized phospholipids and/or sterols, elicit a lung-specific immune response in Abcg1(-/-) mice. Loss of ABCG1 results in increased levels of specific oxysterols, phosphatidylcholines, and oxidized phospholipids, including 1-palmitoyl-2-(5'-oxovaleroyl)-sn-glycero-3-phosphocholine, in the lungs. Further, we identify a niche-specific increase in natural Ab (NAb)-secreting B-1 B cells in response to this lipid accumulation that is paralleled by increased titers of IgM, IgA, and IgG against oxidation-specific epitopes, such as those on oxidized low-density lipoprotein and malondialdehyde-modified low-density lipoprotein. Finally, we identify a cytokine/chemokine signature that is reflective of increased B cell activation, Ab secretion, and homing. Collectively, these data demonstrate that the accumulation of lipids in Abcg1(-/-) mice induces the specific expansion and localization of B-1 B cells, which secrete NAbs that may help to protect against the development of atherosclerosis. Indeed, despite chronic lipid accumulation and inflammation, hyperlipidemic mice lacking ABCG1 develop smaller atherosclerotic lesions compared with controls. These data also suggest that Abcg1(-/-) mice may represent a new model in which to study the protective functions of B-1 B cells/NAbs and suggest novel targets for pharmacologic intervention and treatment of disease.
Collapse
Affiliation(s)
- Angel Baldan
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095; Edward A. Doisy Department of Biochemistry and Molecular Biology, St. Louis University, St. Louis, MO 63104
| | - Ayelet Gonen
- Department of Medicine, University of California San Diego, La Jolla, CA 92093
| | - Christina Choung
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095
| | - Xuchu Que
- Department of Medicine, University of California San Diego, La Jolla, CA 92093
| | - Tyler J Marquart
- Edward A. Doisy Department of Biochemistry and Molecular Biology, St. Louis University, St. Louis, MO 63104
| | - Irene Hernandez
- Instituto de Investigaciones Biomédicas "Alberto Sols" Consejo Superior de Investigaciones Cientificas - Universidad Autonoma de Madrid, Madrid 28006; Unidad Asociada de Biomedicina IIBM-Universidad de Las Palmas de Gran Canaria, Las Palmas 35016, Spain; and
| | | | - David A Ford
- Edward A. Doisy Department of Biochemistry and Molecular Biology, St. Louis University, St. Louis, MO 63104
| | - Joseph L Witztum
- Department of Medicine, University of California San Diego, La Jolla, CA 92093
| | - Elizabeth J Tarling
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095;
| |
Collapse
|
141
|
Abstract
Adaptive immunity is involved in the pathogenesis of atherosclerosis, but the recruitment of T and B lymphocytes to atherosclerotic lesions is not as well studied as that of monocytes. In this review, we summarize the current understanding of the role of lymphocyte subsets in the pathogenesis of atherosclerosis and discuss chemokines and chemokine receptors involved in lymphocyte homing to atherosclerotic lesions. We review evidence for involvement of the chemokines CCL5, CCL19, CCL21, CXCL10, and CXCL16 and macrophage migration inhibitory factor in lymphocyte homing in atherosclerosis. Also, we review the role of their receptors CCR5, CCR6, CCR7, CXCR3, CXCR6, and CXCR2/CXCR4 and the role of the L-selectin in mouse models of atherosclerosis.
Collapse
Affiliation(s)
- Jie Li
- From the Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, CA
| | - Klaus Ley
- From the Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, CA.
| |
Collapse
|
142
|
Imaging of oxidation-specific epitopes with targeted nanoparticles to detect high-risk atherosclerotic lesions: progress and future directions. J Cardiovasc Transl Res 2014; 7:719-36. [PMID: 25297940 DOI: 10.1007/s12265-014-9590-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Accepted: 09/12/2014] [Indexed: 12/17/2022]
Abstract
Oxidation-specific epitopes (OSE) within developing atherosclerotic lesions are key antigens that drive innate and adaptive immune responses in atherosclerosis, leading to chronic inflammation. Oxidized phospholipids and malondialdehyde-lysine epitopes are well-characterized OSE present in human atherosclerotic lesions, particularly in pathologically defined vulnerable plaques. Using murine and human OSE-specific antibodies as targeting agents, we have developed radionuclide and magnetic resonance based nanoparticles, containing gadolinium, manganese or lipid-coated ultrasmall superparamagnetic iron oxide, to non-invasively image OSE within experimental atherosclerotic lesions. These methods quantitate plaque burden, allow detection of lesion progression and regression, plaque stabilization, and accumulation of OSE within macrophage-rich areas of the artery wall, suggesting they detect the most active lesions. Future studies will focus on using "natural" antibodies, lipopeptides, and mimotopes for imaging applications. These approaches should enhance the clinical translation of this technique to image, monitor, evaluate efficacy of novel therapeutic agents, and guide optimal therapy of high-risk atherosclerotic lesions.
Collapse
|
143
|
Sims KH, Tytler EM, Tipton J, Hill KL, Burgess SW, Shaw WA. Avanti lipid tools: connecting lipids, technology, and cell biology. Biochim Biophys Acta Mol Cell Biol Lipids 2014; 1841:1038-48. [PMID: 24954118 DOI: 10.1016/j.bbalip.2014.05.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Revised: 05/21/2014] [Accepted: 05/23/2014] [Indexed: 11/15/2022]
Abstract
Lipid research is challenging owing to the complexity and diversity of the lipidome. Here we review a set of experimental tools developed for the seasoned lipid researcher, as well as, those who are new to the field of lipid research. Novel tools for probing protein-lipid interactions, applications for lipid binding antibodies, enhanced systems for the cellular delivery of lipids, improved visualization of lipid membranes using gold-labeled lipids, and advances in mass spectrometric analysis techniques will be discussed. Because lipid mediators are known to participate in a host of signal transduction and trafficking pathways within the cell, a comprehensive lipid toolbox that aids the science of lipidomics research is essential to better understand the molecular mechanisms of interactions between cellular components. This article is part of a Special Issue entitled Tools to study lipid functions.
Collapse
Affiliation(s)
- Kacee H Sims
- Avanti Polar Lipids, Inc., 700 Industrial Park Drive, Alabaster, Al 35007, USA.
| | - Ewan M Tytler
- Avanti Polar Lipids, Inc., 700 Industrial Park Drive, Alabaster, Al 35007, USA.
| | - John Tipton
- Avanti Polar Lipids, Inc., 700 Industrial Park Drive, Alabaster, Al 35007, USA.
| | - Kasey L Hill
- Avanti Polar Lipids, Inc., 700 Industrial Park Drive, Alabaster, Al 35007, USA.
| | - Stephen W Burgess
- Avanti Polar Lipids, Inc., 700 Industrial Park Drive, Alabaster, Al 35007, USA.
| | - Walter A Shaw
- Avanti Polar Lipids, Inc., 700 Industrial Park Drive, Alabaster, Al 35007, USA.
| |
Collapse
|
144
|
Abstract
The development of atherosclerosis is the major etiological factor causing cardiovascular disease and constitutes a lipid-induced, chronic inflammatory and autoimmune disease of the large arteries. A long-standing view of the protective role of B cells in atherosclerosis has been challenged by recent studies using B cell depletion in animal models. Whereas complete B cell deficiency increases atherosclerosis, depletion of B2 but not B1 cells reduces atherosclerosis. This has led to a re-evaluation of the multiple potential pathways by which B cells can regulate atherosclerosis, and the apparent opposing roles of B1 and B2 cells. B cells, in addition to having the unique ability to produce antibodies, are now recognized to play a number of important roles in the immune system, including cytokine production and direct regulation of T cell responses. This review summarizes current knowledge on B cell subsets and functions, and how these could distinctly influence atherosclerosis development.
Collapse
Affiliation(s)
- Andrew P Sage
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge , Cambridge , UK
| | | |
Collapse
|
145
|
Abstract
Insights into the important contribution of inflammation and immune functions in the development and progression of atherosclerosis have greatly improved our understanding of this disease. Although the role of T cells has been extensively studied for decades, only recently has the role of B cells gained more attention. Recent studies have identified differential effects of different B-cell subsets and helped to clarify the still poorly understood mechanisms by which these act. B1 cells have been shown to prevent lesion formation, whereas B2 cells have been suggested to promote it. Natural IgM antibodies, mainly derived from B1 cells, have been shown to mediate atheroprotective effects, but the functional role of other immunoglobulin classes, particularly IgG, still remains elusive. In this review, we will focus on recent insights on the role of B cells and various immunoglobulin classes and how these may mediate their effects in atherosclerotic lesion formation. Moreover, we will highlight potential therapeutic approaches focusing on B-cell depletion that could be used to translate experimental evidence to human disease.
Collapse
Affiliation(s)
- Dimitrios Tsiantoulas
- From the Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences, Vienna, Austria (D.T., C.J.B.); Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria (D.T., C.J.B.); and Department of Medicine, University of California San Diego, La Jolla (C.J.D., J.L.W.)
| | - Cody J Diehl
- From the Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences, Vienna, Austria (D.T., C.J.B.); Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria (D.T., C.J.B.); and Department of Medicine, University of California San Diego, La Jolla (C.J.D., J.L.W.)
| | - Joseph L Witztum
- From the Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences, Vienna, Austria (D.T., C.J.B.); Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria (D.T., C.J.B.); and Department of Medicine, University of California San Diego, La Jolla (C.J.D., J.L.W.)
| | - Christoph J Binder
- From the Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences, Vienna, Austria (D.T., C.J.B.); Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria (D.T., C.J.B.); and Department of Medicine, University of California San Diego, La Jolla (C.J.D., J.L.W.).
| |
Collapse
|
146
|
Ait-Oufella H, Sage AP, Mallat Z, Tedgui A. Adaptive (T and B cells) immunity and control by dendritic cells in atherosclerosis. Circ Res 2014; 114:1640-60. [PMID: 24812352 DOI: 10.1161/circresaha.114.302761] [Citation(s) in RCA: 152] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Chronic inflammation in response to lipoprotein accumulation in the arterial wall is central in the development of atherosclerosis. Both innate and adaptive immunity are involved in this process. Adaptive immune responses develop against an array of potential antigens presented to effector T lymphocytes by antigen-presenting cells, especially dendritic cells. Functional analysis of the role of different T-cell subsets identified the Th1 responses as proatherogenic, whereas regulatory T-cell responses exert antiatherogenic activities. The effect of Th2 and Th17 responses is still debated. Atherosclerosis is also associated with B-cell activation. Recent evidence established that conventional B-2 cells promote atherosclerosis. In contrast, innate B-1 B cells offer protection through secretion of natural IgM antibodies. This review discusses the recent development in our understanding of the role of T- and B-cell subsets in atherosclerosis and addresses the role of dendritic cell subpopulations in the control of adaptive immunity.
Collapse
Affiliation(s)
- Hafid Ait-Oufella
- From INSERM UMR-S 970, Paris Cardiovascular Research Center (PARCC), Université Paris Descartes, Sorbonne Paris Cité, Paris, France (H.A.-O., Z.M., A.T.); Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint-Antoine, Paris, France (H.A.-O.); and Department of Medicine, University of Cambridge, Cambridge, United Kingdom (A.P.S., Z.M.)
| | | | | | | |
Collapse
|
147
|
Ryan BJ, Nissim A, Winyard PG. Oxidative post-translational modifications and their involvement in the pathogenesis of autoimmune diseases. Redox Biol 2014; 2:715-24. [PMID: 24955328 PMCID: PMC4062766 DOI: 10.1016/j.redox.2014.05.004] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2014] [Revised: 05/22/2014] [Accepted: 05/23/2014] [Indexed: 02/07/2023] Open
Abstract
Tissue inflammation results in the production of numerous reactive oxygen, nitrogen and chlorine species, in addition to the products of lipid and sugar oxidation. Some of these products are capable of chemically modifying amino acids. This in turn results in changes to the structure and function of proteins. Increasing evidence demonstrates that such oxidative post-translational modifications result in the generation of neo-epitopes capable of eliciting both innate and adaptive immune responses. In this paper, we focus on how free radicals and related chemical species generated in inflammatory environments modulate the antigenicity of self-proteins, resulting in immune responses which involve the generation of autoantibodies against key autoantigens in autoimmune diseases. As examples, we will focus on Ro-60 and C1q in systemic lupus erythematosus, along with type-II collagen in rheumatoid arthritis. This review also covers some of the emerging literature which demonstrates that neo-epitopes generated by oxidation are conserved, as exemplified by the evolutionarily conserved pathogen-associated molecular patterns (PAMPs). We discuss how these observations relate to the pathogenesis of both human autoimmune diseases and inflammatory disease, such as atherosclerosis. The potential for these neo-epitopes and the immune responses against them to act as biomarkers or therapeutic targets is also discussed. Oxidants can generate stable post-translational modifications (PTMs) on proteins. Oxidative PTMs are recognised in evolutionarily-conserved innate immune responses. These PTMs can represent neo-epitopes that break tolerance in autoimmune disease. Antibodies targeting these PTMs in diseases e.g. RA and SLE, can be biomarkers.
Collapse
Affiliation(s)
- Brent J. Ryan
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
| | - Ahuva Nissim
- Centre for Biochemical Pharmacology, William Harvey Research Institute, Queen Mary, University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Paul G. Winyard
- University of Exeter Medical School, St Luke's Campus, Exeter, Devon EX1 2LU, UK
- Corresponding author.
| |
Collapse
|
148
|
Kummu O, Turunen SP, Prus P, Lehtimäki J, Veneskoski M, Wang C, Hörkkö S. Human monoclonal Fab and human plasma antibodies to carbamyl-epitopes cross-react with malondialdehyde-adducts. Immunology 2014; 141:416-30. [PMID: 24168430 DOI: 10.1111/imm.12204] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Revised: 10/23/2013] [Accepted: 10/23/2013] [Indexed: 12/30/2022] Open
Abstract
Oxidized low-density lipoprotein (OxLDL) plays a crucial role in the development of atherosclerosis. Carbamylated LDL has been suggested to promote atherogenesis in patients with chronic kidney disease. Here we observed that plasma IgG and IgM antibodies to carbamylated epitopes were associated with IgG and IgM antibodies to oxidation-specific epitopes (ρ = 0·65-0·86, P < 0·001) in healthy adults, suggesting a cross-reaction between antibodies recognizing carbamyl-epitopes and malondialdehyde (MDA)/malondialdehyde acetaldehyde (MAA) -adducts. We used a phage display technique to clone a human Fab antibody that bound to carbamylated LDL and other carbamylated proteins. Anti-carbamyl-Fab (Fab106) cross-reacted with oxidation-specific epitopes, especially with MDA-LDL and MAA-LDL. We showed that Fab106 bound to apoptotic Jurkat cells known to contain these oxidation-specific epitopes, and the binding was competed with soluble carbamylated and MDA-/MAA-modified LDL and BSA. In addition, Fab106 was able to block the uptake of carbamyl-LDL and MDA-LDL by macrophages and stained mouse atherosclerotic lesions. The observed cross-reaction between carbamylated and MDA-/MAA-modified LDL and its contribution to enhanced atherogenesis in uraemic patients require further investigation.
Collapse
Affiliation(s)
- Outi Kummu
- Department of Medical Microbiology and Immunology, Institute of Diagnostics, University of Oulu, Oulu, Finland; Medical Research Center Oulu, Oulu, Finland; NordLab Oulu, Oulu University Hospital, Oulu, Finland
| | | | | | | | | | | | | |
Collapse
|
149
|
Birukova AA, Singleton PA, Gawlak G, Tian X, Mirzapoiazova T, Mambetsariev B, Dubrovskyi O, Oskolkova OV, Bochkov VN, Birukov KG. GRP78 is a novel receptor initiating a vascular barrier protective response to oxidized phospholipids. Mol Biol Cell 2014; 25:2006-16. [PMID: 24829380 PMCID: PMC4072574 DOI: 10.1091/mbc.e13-12-0743] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Vascular integrity and the maintenance of blood vessel continuity are fundamental features of the circulatory system maintained through endothelial cell-cell junctions. Defects in the endothelial barrier become an initiating factor in several pathologies, including ischemia/reperfusion, tumor angiogenesis, pulmonary edema, sepsis, and acute lung injury. Better understanding of mechanisms stimulating endothelial barrier enhancement may provide novel therapeutic strategies. We previously reported that oxidized phospholipids (oxidized 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphocholine [OxPAPC]) promote endothelial cell (EC) barrier enhancement both in vitro and in vivo. This study examines the initiating mechanistic events triggered by OxPAPC to increase vascular integrity. Our data demonstrate that OxPAPC directly binds the cell membrane-localized chaperone protein, GRP78, associated with its cofactor, HTJ-1. OxPAPC binding to plasma membrane-localized GRP78 leads to GRP78 trafficking to caveolin-enriched microdomains (CEMs) on the cell surface and consequent activation of sphingosine 1-phosphate receptor 1, Src and Fyn tyrosine kinases, and Rac1 GTPase, processes essential for cytoskeletal reorganization and EC barrier enhancement. Using animal models of acute lung injury with vascular hyperpermeability, we observed that HTJ-1 knockdown blocked OxPAPC protection from interleukin-6 and ventilator-induced lung injury. Our data indicate for the first time an essential role of GRP78 and HTJ-1 in OxPAPC-mediated CEM dynamics and enhancement of vascular integrity.
Collapse
Affiliation(s)
- Anna A Birukova
- Lung Injury Center, Section of Pulmonary and Critical Care, Department of Medicine, Division of Biomedical Sciences, University of Chicago, Chicago, IL 60637
| | - Patrick A Singleton
- Lung Injury Center, Section of Pulmonary and Critical Care, Department of Medicine, Division of Biomedical Sciences, University of Chicago, Chicago, IL 60637
| | - Grzegorz Gawlak
- Lung Injury Center, Section of Pulmonary and Critical Care, Department of Medicine, Division of Biomedical Sciences, University of Chicago, Chicago, IL 60637
| | - Xinyong Tian
- Lung Injury Center, Section of Pulmonary and Critical Care, Department of Medicine, Division of Biomedical Sciences, University of Chicago, Chicago, IL 60637
| | - Tamara Mirzapoiazova
- Lung Injury Center, Section of Pulmonary and Critical Care, Department of Medicine, Division of Biomedical Sciences, University of Chicago, Chicago, IL 60637
| | - Bolot Mambetsariev
- Lung Injury Center, Section of Pulmonary and Critical Care, Department of Medicine, Division of Biomedical Sciences, University of Chicago, Chicago, IL 60637
| | - Oleksii Dubrovskyi
- Lung Injury Center, Section of Pulmonary and Critical Care, Department of Medicine, Division of Biomedical Sciences, University of Chicago, Chicago, IL 60637
| | - Olga V Oskolkova
- Lung Injury Center, Section of Pulmonary and Critical Care, Department of Medicine, Division of Biomedical Sciences, University of Chicago, Chicago, IL 60637Department of Vascular Biology and Thrombosis Research, Medical University of Vienna, 1090 Vienna, Austria
| | - Valery N Bochkov
- Department of Vascular Biology and Thrombosis Research, Medical University of Vienna, 1090 Vienna, Austria
| | - Konstantin G Birukov
- Department of Vascular Biology and Thrombosis Research, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
150
|
Grönwall C, Silverman GJ. Natural IgM: beneficial autoantibodies for the control of inflammatory and autoimmune disease. J Clin Immunol 2014; 34 Suppl 1:S12-21. [PMID: 24691998 DOI: 10.1007/s10875-014-0025-4] [Citation(s) in RCA: 119] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Accepted: 03/19/2014] [Indexed: 12/13/2022]
Abstract
Natural IgM are highly represented in the circulation at birth, and these often autoreactive antibodies have been postulated to have innate-like properties and play crucial roles in apoptotic cell clearance, tissue homeostasis, and immune modulation. This review summarizes the known properties of these IgM autoantibodies, and the evidence that these anti-apoptotic cell IgM natural antibodies can regulate inflammatory responses through ancient pathways of the innate immune system that first arose long before the initial emergence of the adaptive immune system. While the regulatory contributions of these natural IgM autoantibodies are certainly not an essential and fundamental component of host defenses, these provide an additional layer to further protect the host. More importantly, these IgM antibody responses are highly inducible and their up-regulation can be a powerful means for the host to survive in a setting of chronic inflammation. The observed beneficial clinical associations for cardiovascular disease and autoimmunity, as well as opportunities for potential therapeutic implications are discussed.
Collapse
Affiliation(s)
- Caroline Grönwall
- Department of Medicine, New York University School of Medicine, New York, NY, 10016, USA,
| | | |
Collapse
|