1
|
Webster SE, Les SM, Deleon N, Heck DM, Tsuj NL, Clemente MJ, Jones P, Holodick NE. Secreted IgM deficiency alters the retinal landscape enhancing neurodegeneration associated with aging. Immun Ageing 2025; 22:9. [PMID: 39994686 DOI: 10.1186/s12979-025-00502-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 02/08/2025] [Indexed: 02/26/2025]
Abstract
BACKGROUND Maintenance of the retina, part of the central nervous system, and other structures in the eye is critical for vision preservation. Aging increases the prevalence of vision impairment, including glaucoma, macular degeneration, and diabetic retinopathy. The retina is primarily maintained by glial cells; however, recent literature suggests that lymphocytes may play a role in the homeostasis of central nervous system tissues. Natural antibodies are produced by B cells without infection or immunization and maintain tissue homeostasis. Here, we explored the potential role of natural immunoglobulin M (IgM) produced by B lymphocytes in maintaining retinal health during aging in mice. RESULTS Our results indicate that the vitreous humor of both mice and humans contains IgM and IgG, suggesting that these immunoglobulins may play a role in ocular function. Furthermore, we observed that aged mice lacking secreted IgM (µs-/-) exhibited pronounced retinal degeneration, accompanied by reactive gliosis, and a proinflammatory cytokine environment. This contrasts with the aged wild-type counterparts, which retain their ability to secrete IgM and maintain a better retinal structure and anti-inflammatory environment. In addition to these findings, the absence of secreted IgM was associated with significant alterations in the retinal pigment epithelium, including disruptions to its morphology and signs of increased stress. This was further observed in changes to the blood-retinal-barrier, which is critical for regulation of retinal homeostasis. CONCLUSIONS These data suggest a previously unrecognized association between a lack of secreted IgM and alterations in the retinal microenvironment, leading to enhanced retinal degeneration during aging. Although the precise mechanism remains unclear, these findings highlight the potential importance of secreted IgM in processes that support retinal health over time. By increasing our understanding of ocular aging, these results show that there is a broader role for the immune system in retinal function and integrity in advanced age, opening new areas for the exploration of immune-related interventions in age-associated retinal conditions.
Collapse
Affiliation(s)
- Sarah E Webster
- Center for Immunobiology, Department of Investigative Medicine, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, 49007, USA.
| | - Sydney M Les
- Center for Immunobiology, Department of Investigative Medicine, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, 49007, USA
- Department of Medicine, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, 49007, United States of America
| | - Nico Deleon
- Center for Immunobiology, Department of Investigative Medicine, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, 49007, USA
| | - Daken M Heck
- Center for Immunobiology, Department of Investigative Medicine, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, 49007, USA
| | - Naomi L Tsuj
- Center for Immunobiology, Department of Investigative Medicine, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, 49007, USA
| | - Michael J Clemente
- Center for Immunobiology, Department of Investigative Medicine, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, 49007, USA
- Flow Cytometry and Imaging Core, Department of Investigative Medicine, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, 49007, USA
| | - Prentiss Jones
- Department of Pathology, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, 49007, USA
| | - Nichol E Holodick
- Center for Immunobiology, Department of Investigative Medicine, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, 49007, USA
- Flow Cytometry and Imaging Core, Department of Investigative Medicine, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, 49007, USA
| |
Collapse
|
2
|
Fujisaki K, Okazaki S, Ogawa S, Takeda M, Sugihara E, Imai K, Mizuno S, Takahashi S, Goitsuka R. B Cells of Early-life Origin Defined by RAG2-based Lymphoid Cell Tracking under Native Hematopoietic Conditions. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:296-305. [PMID: 38874543 DOI: 10.4049/jimmunol.2400072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 05/22/2024] [Indexed: 06/15/2024]
Abstract
During the perinatal period, the immune system sets the threshold to select either response or tolerance to environmental Ags, which leads to the potential to provide a lifetime of protection and health. B-1a B cells have been demonstrated to develop during this perinatal time window, showing a unique and restricted BCR repertoire, and these cells play a major role in natural Ab secretion and immune regulation. In the current study, we developed a highly efficient temporally controllable RAG2-based lymphoid lineage cell labeling and tracking system and applied this system to understand the biological properties and contribution of B-1a cells generated at distinct developmental periods to the adult B-1a compartments. This approach revealed that B-1a cells with a history of RAG2 expression during the embryonic and neonatal periods dominate the adult B-1a compartment, including those in the bone marrow (BM), peritoneal cavity, and spleen. Moreover, the BCR repertoire of B-1a cells with a history of RAG2 expression during the embryonic period was restricted, becoming gradually more diverse during the neonatal period, and then heterogeneous at the adult stage. Furthermore, more than half of plasmablasts/plasma cells in the adult BM had embryonic and neonatal RAG2 expression histories. Moreover, BCR analysis revealed a high relatedness between BM plasmablasts/plasma cells and B-1a cells derived from embryonic and neonatal periods, suggesting that these cell types have a common origin. Taken together, these findings define, under native hematopoietic conditions, the importance in adulthood of B-1a cells generated during the perinatal period.
Collapse
Affiliation(s)
- Keiko Fujisaki
- Division of Cell Fate Regulation, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan
| | - Shogo Okazaki
- Department of Microbiology and Immunology, Nihon University School of Dentistry, Tokyo, Japan
| | - Shuhei Ogawa
- Division of Integrated Research, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan
| | - Miyama Takeda
- Division of Cell Fate Regulation, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan
| | - Eiji Sugihara
- Open Facility Center and Cancer Center, Fujita Health University, Aichi, Japan
| | - Kenichi Imai
- Department of Microbiology and Immunology, Nihon University School of Dentistry, Tokyo, Japan
| | - Seiya Mizuno
- Laboratory Animal Resource Center, Transborder Medical Research Center, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Satoru Takahashi
- Laboratory Animal Resource Center, Transborder Medical Research Center, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
- Department of Anatomy and Embryology, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Ryo Goitsuka
- Division of Cell Fate Regulation, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan
| |
Collapse
|
3
|
Raposo-Gutiérrez I, Rodríguez-Ronchel A, Ramiro AR. Atherosclerosis antigens as targets for immunotherapy. NATURE CARDIOVASCULAR RESEARCH 2023; 2:1129-1147. [PMID: 39196152 DOI: 10.1038/s44161-023-00376-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 10/18/2023] [Indexed: 08/29/2024]
Abstract
Atherosclerosis is a chronic inflammatory disease of the arteries that can lead to thrombosis, infarction and stroke, underlying the first cause of mortality worldwide. Adaptive immunity plays critical roles in atherosclerosis, and numerous studies have ascribed both atheroprotective and atherogenic functions to specific subsets of T and B cells. However, less is known on how antigen specificity determines the protective or adverse outcome of such adaptive responses. Understanding antigen triggers in atherosclerosis is crucial to delve deeper into mechanisms of disease initiation and progression and to implement specific immunotherapeutic approaches, including vaccination strategies. Here we review the role of adaptive immunity in atherosclerosis and the insights that single-cell technology has provided into the function of distinct immune cell subsets. We outline the most relevant atherosclerosis antigens and antibodies reported to date and examine their immunotherapeutic potential. Finally, we review the most promising vaccination-based clinical trials targeting the adaptive immune system.
Collapse
Affiliation(s)
- Irene Raposo-Gutiérrez
- B Lymphocyte Lab, Novel Mechanisms of Atherosclerosis Program, Spanish National Center for Cardiovascular Research, Madrid, Spain
| | - Ana Rodríguez-Ronchel
- B Lymphocyte Lab, Novel Mechanisms of Atherosclerosis Program, Spanish National Center for Cardiovascular Research, Madrid, Spain
| | - Almudena R Ramiro
- B Lymphocyte Lab, Novel Mechanisms of Atherosclerosis Program, Spanish National Center for Cardiovascular Research, Madrid, Spain.
| |
Collapse
|
4
|
Xue S, Su Z, Liu D. Immunometabolism and immune response regulate macrophage function in atherosclerosis. Ageing Res Rev 2023; 90:101993. [PMID: 37379970 DOI: 10.1016/j.arr.2023.101993] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 06/23/2023] [Indexed: 06/30/2023]
Abstract
Macrophages are crucial in the progression of atherosclerotic cardiovascular disease (ASCVD). In the atherosclerotic lesions, macrophages play a central role in maintaining inflammatory response, promoting plaque development, and facilitating thrombosis. Increasing studies indicate that metabolic reprogramming and immune response mediate macrophage functional changes in all stages of atherosclerosis. In this review article, we explain how metabolic changes in glycolysis, oxidative phosphorylation, the tricarboxylic acid cycle, fatty acid synthesis, fatty acid oxidation, and cholesterol metabolism regulate macrophage function in atherosclerosis. We discuss how immune response to oxidized lipids regulate macrophage function in atherosclerosis. Additionally, we explore how abnormal metabolism leads to macrophage mitochondrial dysfunction in atherosclerosis.
Collapse
Affiliation(s)
- Sheng Xue
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266003 China.
| | - Zhe Su
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266003 China
| | - Dacheng Liu
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266003 China
| |
Collapse
|
5
|
Dumić J, Cvetko A, Abramović I, Šupraha Goreta S, Perović A, Njire Bratičević M, Kifer D, Sinčić N, Gornik O, Žarak M. Changes in Specific Biomarkers Indicate Cardiac Adaptive and Anti-inflammatory Response of Repeated Recreational SCUBA Diving. Front Cardiovasc Med 2022; 9:855682. [PMID: 35360010 PMCID: PMC8964121 DOI: 10.3389/fcvm.2022.855682] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 02/21/2022] [Indexed: 11/13/2022] Open
Abstract
ObjectiveRecreational SCUBA (rSCUBA) diving has become a highly popular and widespread sport. Yet, information on molecular events underlying (patho)physiological events that follow exposure to the specific environmental conditions (hyperbaric conditions, coldness, immersion, and elevated breathing pressure), in which rSCUBA diving is performed, remain largely unknown. Our previous study suggested that repeated rSCUBA diving triggers an adaptive response of cardiovascular and immune system. To elucidate further molecular events underlying cardiac and immune system adaptation and to exclude possible adverse effects we measured blood levels of specific cardiac and inflammation markers.MethodsThis longitudinal intervention study included fourteen recreational divers who performed five dives, one per week, on the depth 20–30 m that lasted 30 min, after the non-dive period of 5 months. Blood samples were taken immediately before and after the first, third, and fifth dives. Copeptin, immunoglobulins A, G and M, complement components C3 and C4, and differential blood count parameters, including neutrophil-to-lymphocyte ratio (NLR) were determined using standard laboratory methods. Cell-free DNA was measured by qPCR analysis and N-glycans released from IgG and total plasma proteins (TPP), were analyzed by hydrophilic interaction ultra-performance liquid chromatography.ResultsCopeptin level increased after the first dive but decreased after the third and fifth dive. Increases in immunoglobulins level after every dive and during whole studied period were observed, but no changes in C3, C4, and cfDNA level were detected. NLR increased only after the first dive. IgG and TPP N-glycosylation alterations toward anti-inflammatory status over whole studied period were manifested as an increase in monogalyctosylated and core-fucosylated IgG N-glycans and decrease in agalactosylated TPP N-glycans.ConclusionrSCUBA diving practiced on a regular basis promotes anti-inflammatory status thus contributing cardioprotection and conferring multiple health benefits.
Collapse
Affiliation(s)
- Jerka Dumić
- Department of Biochemistry and Molecular Biology, University of Zagreb Faculty of Pharmacy and Biochemistry, Zagreb, Croatia
| | - Ana Cvetko
- Department of Biochemistry and Molecular Biology, University of Zagreb Faculty of Pharmacy and Biochemistry, Zagreb, Croatia
| | - Irena Abramović
- Department of Medical Biology, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Sandra Šupraha Goreta
- Department of Biochemistry and Molecular Biology, University of Zagreb Faculty of Pharmacy and Biochemistry, Zagreb, Croatia
| | - Antonija Perović
- Department of Laboratory Diagnostics, Dubrovnik General Hospital, Dubrovnik, Croatia
| | | | - Domagoj Kifer
- Department of Biophysics, University of Zagreb Faculty of Pharmacy and Biochemistry, Zagreb, Croatia
| | - Nino Sinčić
- Department of Medical Biology, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Olga Gornik
- Department of Biochemistry and Molecular Biology, University of Zagreb Faculty of Pharmacy and Biochemistry, Zagreb, Croatia
| | - Marko Žarak
- Clinical Department of Laboratory Diagnostics, Dubrava University Hospital, Zagreb, Croatia
- *Correspondence: Marko Žarak,
| |
Collapse
|
6
|
Afzal M, Park J, Jeon JS, Akmal M, Yoon TS, Sung HJ. Acoustofluidic Separation of Proteins Using Aptamer-Functionalized Microparticles. Anal Chem 2021; 93:8309-8317. [PMID: 34075739 DOI: 10.1021/acs.analchem.1c01198] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
We propose an acoustofluidic method for the triseparation of proteins conjugated with aptamer-coated microparticles inside a microchannel. Traveling surface acoustic waves (TSAWs) produced from a slanted-finger interdigital transducer (SFIT) are used to separate the protein-loaded microparticles of different sizes via the TSAW-driven acoustic radiation force (ARF). The acoustofluidic device consists of an SFIT deposited onto a piezoelectric lithium niobate substrate and a polydimethylsiloxane (PDMS) microfluidic channel on top of the substrate. The TSAWs propagating on the substrate penetrate into the sample fluid flow, where the human protein-conjugated microparticles are suspended, inside the PDMS microchannel. The microparticles are subjected to the TSAW-driven ARF with varying magnitude depending on their size and thus flow along different streamlines, leading to triseparation of the proteins. In this work, we used two different-sized streptavidin-functionalized polystyrene (PS) microparticles to capture two kinds of aptamers (apt15 and aptD17.4), which were labeled with a respective biotin molecule at one end. The biotin ends of the aptamers were attached to the microparticles through streptavidin-biotin linkage, whereas the free ends of the aptamers were used to capture their target proteins of thrombin (th) and immunoglobulin E (IgE). The resultant PS-apt15-th and PS-aptD17.4-IgE complexes, as well as mCardinal2, were used for experimental demonstration of acoustofluidic triseparation of the human proteins. We achieved simultaneous separation of proteins of three kinds (th, IgE, and mCardinal2) for the first time via the TSAW-driven ARF in the proposed acoustofluidic device.
Collapse
Affiliation(s)
- Muhammad Afzal
- Department of Mechanical Engineering, KAIST, 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Korea
| | - Jinsoo Park
- School of Mechanical Engineering, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 61186, Korea
| | - Jessie S Jeon
- Department of Mechanical Engineering, KAIST, 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Korea
| | - Muhammad Akmal
- Department of Materials Engineering, KAIST, 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Korea
| | - Tae-Sung Yoon
- Department of Proteome Structural Biology, KRIBB, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea
| | - Hyung Jin Sung
- Department of Mechanical Engineering, KAIST, 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Korea
| |
Collapse
|
7
|
Hong LZ, Xue Q, Shao H. Inflammatory Markers Related to Innate and Adaptive Immunity in Atherosclerosis: Implications for Disease Prediction and Prospective Therapeutics. J Inflamm Res 2021; 14:379-392. [PMID: 33628042 PMCID: PMC7897977 DOI: 10.2147/jir.s294809] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 01/21/2021] [Indexed: 12/20/2022] Open
Abstract
Several lines of evidence have linked a dysregulated inflammatory setting to the pathogenesis of atherosclerosis, which is a form of chronic vascular inflammation. Various inflammatory biomarkers have been associated with inflammation and are recognized as potential tools to monitor the progression of atherosclerosis. A well-studied inflammatory marker in the context of cardiovascular diseases is C-reactive protein (CRP) or, more accurately, highly sensitive-CRP (hs-CRP), which has been established as an inflammatory biomarker for atherosclerotic events. In addition, a growing body of investigations has attempted to disclose the potential of inflammatory cytokines, enzymes, and genetic polymorphisms related to innate and adaptive immunity as biomarkers for predicting the development of atherosclerosis. In this review article, we clarify both traditional and novel inflammatory biomarkers related to components of the innate and adaptive immune system that may mirror the progression or phases of atherosclerotic inflammation/lesions. Furthermore, the contribution of the inflammatory biomarkers in developing potential therapeutics against atherosclerotic treatment will be discussed.
Collapse
Affiliation(s)
- Ling-Zhi Hong
- Emergency Department, Chun’an First People’s Hospital (Zhejiang Provincial People’s Hospital Chun’an Branch), Hangzhou, 311700, Zhejiang Province, People’s Republic of China
| | - Qi Xue
- Department of Cardiology, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, 310014, Zhejiang Province, People’s Republic of China
| | - Hong Shao
- Department of Cardiology, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, 310014, Zhejiang Province, People’s Republic of China
| |
Collapse
|
8
|
Zbesko JC, Frye JB, Becktel DA, Gerardo DK, Stokes J, Calderon K, Nguyen TVV, Bhattacharya D, Doyle KP. IgA natural antibodies are produced following T-cell independent B-cell activation following stroke. Brain Behav Immun 2021; 91:578-586. [PMID: 32956832 PMCID: PMC8279117 DOI: 10.1016/j.bbi.2020.09.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 09/08/2020] [Accepted: 09/15/2020] [Indexed: 12/22/2022] Open
Abstract
Up to 30% of stroke patients experience cognitive decline within one year of their stroke. There are currently no FDA-approved drugs that can prevent post-stroke cognitive decline, in part due to a poor understanding of the mechanisms involved. We have previously demonstrated that a B-lymphocyte response to stroke, marked by IgA + cells, can cause delayed cognitive dysfunction in mice and that a similar adaptive immune response occurs in the brains of some human stroke patients that suffer from vascular dementia. The stimuli which trigger B-lymphocyte activation following stroke, and their target antigens, are still unknown. Therefore, to learn more about the mechanisms by which B-lymphocytes become activated following stroke we first characterized the temporal kinetics of the B-lymphocyte, T-lymphocyte, and plasma cell (PC) response to stroke in the brain by immunohistochemistry (IHC). We discovered that B-lymphocyte, T-lymphocyte, and plasma cell infiltration within the infarct progressively increases between 2 and 7 weeks after stroke. We then compared the B-lymphocyte response to stroke in WT, MHCII-/-, CD4-/-, and MyD88-/- mice to determine if B-lymphocytes mature into IgA + PCs through a T-lymphocyte and MyD88 dependent mechanism. Our data from a combination of IHC and flow cytometry indicate that following stroke, a population of IgA + PCs develops independently of CD4 + helper T-lymphocytes and MyD88 signaling. Subsequent sequencing of immunoglobulin genes of individual IgA + PCs present within the infarct identified a novel population of natural antibodies with few somatic mutations in complementarity-determining regions. These findings indicate that a population of IgA + PCs develops in the infarct following stroke by B-lymphocytes interacting with one or more thymus independent type 2 (TI-2) antigens, and that they produce IgA natural antibodies.
Collapse
Affiliation(s)
- Jacob C. Zbesko
- Department of Immunobiology, University of Arizona, Tucson, AZ 85719, USA
| | | | | | - Diana K. Gerardo
- Department of Immunobiology, University of Arizona, Tucson, AZ 85719, USA
| | - Jessica Stokes
- Department of Pediatrics, University of Arizona, Tucson, AZ 85719, USA
| | - Kylie Calderon
- Department of Immunobiology, University of Arizona, Tucson, AZ 85719, USA
| | - Thuy-Vi V. Nguyen
- Department of Immunobiology, University of Arizona, Tucson, AZ 85719, USA,Department of Neurology, University of Arizona, Tucson, AZ 85719, USA
| | | | - Kristian P. Doyle
- Department of Immunobiology, University of Arizona, Tucson, AZ 85719, USA,Department of Neurology, University of Arizona, Tucson, AZ 85719, USA,Arizona Center on Aging, University of Arizona, Tucson, AZ 85719, USA,Corresponding author at: Department of Immunobiology, University of Arizona, Tucson, 1656 E. Mabel Street, Tucson, Arizona 85719, USA. (K.P. Doyle)
| |
Collapse
|
9
|
Zhang TY, Zhao Q, Liu ZS, Zhang CY, Yang J, Meng K. Relationship between monocyte/lymphocyte ratio and non-culprit plaque vulnerability in patients with acute coronary syndrome: An optical coherence tomography study. Medicine (Baltimore) 2020; 99:e21562. [PMID: 33031255 PMCID: PMC7544167 DOI: 10.1097/md.0000000000021562] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The importance of monocyte/lymphocyte ratio (MLR) has been indicated in the initiation and progression of coronary artery disease. However, few previous researches demonstrated the relationship between MLR and plaque vulnerability. We aimed to investigate coronary non-culprit plaque vulnerability in patients with acute coronary syndrome (ACS) by optical coherence tomography (OCT).A total of 72 ACS patients who underwent coronary angiography and OCT test in Beijing Anzhen Hospital were included in this retrospective study. The plaque vulnerability and plaque morphology were assessed by OCT.The non-culprit plaque in high MLR group exhibited more vulnerable features, characterizing as thinner thickness of fibrous cap (P = .013), greater maximum lipid core angle (P = .010) and longer lipid plaque length (P = .041). A prominently negative liner relation was found between MLR and thickness of fibrous cap (R = -0.225, P = .005). Meanwhile, the proportion of OCT-detected thin cap fibro-atheroma (TCFA) (P = .014) and plaque rupture (P = .017) were higher in high MLR group. Most importantly, multivariable logistic regression analysis showed MLR level was identified as an independent contributor to the presence of TCFA (OR:3.316, 95%: 1.448-7.593, P = .005). MLR could differentiate TCFA with a sensitivity of 60.0% and a specificity of 85.1%.Circulating MLR level has potential value in identifying the presence of vulnerable plaque in patients with ACS. MLR, as a non- invasive biomarker of inflammation, may be valuable in revealing plaque vulnerability.
Collapse
Affiliation(s)
- Ting-yu Zhang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University
| | - Qi Zhao
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University
| | - Ze-sen Liu
- Department of Cardiology, Beijing Luhe Hospital, Capital Medical University
| | - Chao-yi Zhang
- Department of Cardiology, Beijing Zhongguanchun Hospital, Beijing, China
| | - Jie Yang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University
| | - Kang Meng
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University
| |
Collapse
|
10
|
Reyneveld GIJ, Savelkoul HFJ, Parmentier HK. Current Understanding of Natural Antibodies and Exploring the Possibilities of Modulation Using Veterinary Models. A Review. Front Immunol 2020; 11:2139. [PMID: 33013904 PMCID: PMC7511776 DOI: 10.3389/fimmu.2020.02139] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 08/06/2020] [Indexed: 12/20/2022] Open
Abstract
Natural antibodies (NAb) are defined as germline encoded immunoglobulins found in individuals without (known) prior antigenic experience. NAb bind exogenous (e.g., bacterial) and self-components and have been found in every vertebrate species tested. NAb likely act as a first-line immune defense against infections. A large part of NAb, so called natural autoantibodies (NAAb) bind to and clear (self) neo-epitopes, apoptotic, and necrotic cells. Such self-binding antibodies cannot, however, be considered as pathogenic autoantibodies in the classical sense. IgM and IgG NAb and NAAb and their implications in health and disease are relatively well-described in humans and mice. NAb are present in veterinary (and wildlife) species, but their relation with diseases and disorders in veterinary species are much less known. Also, there is little known of IgA NAb. IgA is the most abundant immunoglobulin with essential pro-inflammatory and homeostatic properties urging for more research on the importance of IgA NAb. Since NAb in humans were indicated to fulfill important functions in health and disease, their role in health of veterinary species should be investigated more often. Furthermore, it is unknown whether levels of NAb-isotypes and/or idiotypes can and should be modulated. Veterinary species as models of choice fill in a niche between mice and (non-human) primates, and the study of NAb in veterinary species may provide valuable new insights that will likely improve health management. Below, examples of the involvement of NAb in several diseases in mostly humans are shown. Possibilities of intravenous immunoglobulin administration, targeted immunotherapy, immunization, diet, and genetic modulation are discussed, all of which could be well-studied using animal models. Arguments are given why veterinary immunology should obtain inspiration from human studies and why human immunology would benefit from veterinary models. Within the One Health concept, findings from veterinary (and wildlife) studies can be related to human studies and vice versa so that both fields will mutually benefit. This will lead to a better understanding of NAb: their origin, activation mechanisms, and their implications in health and disease, and will lead to novel health management strategies for both human and veterinary species.
Collapse
Affiliation(s)
- G. IJsbrand Reyneveld
- Faculty of Science, VU University, Amsterdam, Netherlands
- Adaptation Physiology Group, Department of Animal Sciences, Wageningen University, Wageningen, Netherlands
| | - Huub F. J. Savelkoul
- Cell Biology and Immunology Group, Department of Animal Sciences, Wageningen University, Wageningen, Netherlands
| | - Henk K. Parmentier
- Adaptation Physiology Group, Department of Animal Sciences, Wageningen University, Wageningen, Netherlands
| |
Collapse
|
11
|
Abstract
PURPOSE OF REVIEW Atherosclerosis is a complex disease process with lipid as a traditional modifiable risk factor and therapeutic target in treating atherosclerotic cardiovascular disease (ACVD). Recent evidence indicates that genetic influence and host immune response also are vital in this process. How these elements interact and modify each other and if immune response may emerge as a novel modifiable target remain poorly understood. RECENT FINDINGS Numerous preclinical studies have clearly demonstrated that hypercholesterolemia is essential for atherogenesis, but genetic variations and host immune-inflammatory responses can modulate the pro-atherogenic effect of elevated LDL-C. Clinical studies also suggest that a similar paradigm may also be operational in atherogenesis in humans. More importantly each element modifies the biological behavior of the other two elements, forming a triangular relationship among the three. Modulating any one of them will have downstream impact on atherosclerosis. This brief review summarizes the relationship among lipids, genes, and immunity in atherogenesis and presents evidence to show how these elements affect each other. Modulation of immune response, though in its infancy, has a potential to emerge as a novel clinical strategy in treating ACVD.
Collapse
|
12
|
Mestre AP, Amavet PS, van der Sloot IS, Carletti JV, Poletta GL, Siroski PA. Effects of glyphosate, cypermethrin, and chlorpyrifos on hematological parameters of the tegu lizard (Salvator merianae) in different embryo stages. CHEMOSPHERE 2020; 252:126433. [PMID: 32182507 DOI: 10.1016/j.chemosphere.2020.126433] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 03/04/2020] [Accepted: 03/05/2020] [Indexed: 06/10/2023]
Abstract
Different studies have demonstrated effects of pesticides during embryo development in vertebrates and stage-dependent effects, but there is no information concerning this for Salvator merianae. We evaluated the effects of the herbicides Glyphosate Roundup (GLY-RU) and Glyphosate Panzer Gold (GLY-PZ); and the insecticides Chlorpyrifos (CPF) and Cypermethrin (CYP), and their complex mixtures, at different concentrations in hematological parameters of S. merianae embryos at two different development stages. The analyzed parameters were Total and Differential White Blood Cells Count, Heterophils/Lymphocytes index (H/L), Lobularity index, and Natural Antibodies (Nabs titres), as well as growth, embryo mortality and birth delay. Heterophils decreased in the intermediate concentrations tested of CYP and GLY-RU, in animals exposed at 33-days development. Lymphocytes increased in the intermediate concentration tested of GLY-RU, and the H/L index decreased in the maximum concentration tested of GLY-RU. NAbs titres increased in those animals exposed to the maximum CYP concentration tested. However, animals exposed at 3/5-days development showed no differences among treatments in most of the analyzed parameters, suggesting a stage-dependent response. Nevertheless, those animals exposed to GLY-PZ showed lower Nabs titres in relation to negative control. These results suggest effects on different hematological parameters related to the immune system of S. merianae, according to the used pesticide (herbicide or insecticide), its concentration and commercial formulation (GLY-RU or GLY-PZ), and the stages of development of the exposed animals. Our results reveal the importance of carrying out studies that evaluate the effects of permanent exposure of living beings and their environments to these toxics.
Collapse
Affiliation(s)
- Ana Paula Mestre
- Laboratorio de Ecología Molecular Aplicada, Instituto de Ciencias Veterinarias del Litoral (ICiVet Litoral) UNL - CONICET, Esperanza, Argentina; Laboratorio de Genética, Departamento de Ciencias Naturales (FHUC - UNL), CONICET, Santa Fe, Argentina; Laboratorio de Zoología Aplicada: Anexo Vertebrados, Facultad de Humanidades y Ciencias, Universidad Nacional del Litoral, (FHUC - UNL /MMA), Santa Fe, Argentina.
| | - Patricia Susana Amavet
- Laboratorio de Genética, Departamento de Ciencias Naturales (FHUC - UNL), CONICET, Santa Fe, Argentina; Laboratorio de Zoología Aplicada: Anexo Vertebrados, Facultad de Humanidades y Ciencias, Universidad Nacional del Litoral, (FHUC - UNL /MMA), Santa Fe, Argentina.
| | | | - Julieta Verónica Carletti
- Laboratorio de Ecología Molecular Aplicada, Instituto de Ciencias Veterinarias del Litoral (ICiVet Litoral) UNL - CONICET, Esperanza, Argentina; Laboratorio de Genética, Departamento de Ciencias Naturales (FHUC - UNL), CONICET, Santa Fe, Argentina; Laboratorio de Zoología Aplicada: Anexo Vertebrados, Facultad de Humanidades y Ciencias, Universidad Nacional del Litoral, (FHUC - UNL /MMA), Santa Fe, Argentina.
| | - Gisela Laura Poletta
- Laboratorio de Ecología Molecular Aplicada, Instituto de Ciencias Veterinarias del Litoral (ICiVet Litoral) UNL - CONICET, Esperanza, Argentina; Cátedra de Toxicología, Farmacología y Bioquímica Legal (FBCB-UNL), CONICET, Santa Fe, Argentina.
| | - Pablo Ariel Siroski
- Laboratorio de Ecología Molecular Aplicada, Instituto de Ciencias Veterinarias del Litoral (ICiVet Litoral) UNL - CONICET, Esperanza, Argentina; Laboratorio de Genética, Departamento de Ciencias Naturales (FHUC - UNL), CONICET, Santa Fe, Argentina.
| |
Collapse
|
13
|
Nazarian-Samani Z, Sewell RDE, Rafieian-Kopaei M. Inflammasome Signaling and Other Factors Implicated in Atherosclerosis Development and Progression. Curr Pharm Des 2020; 26:2583-2590. [DOI: 10.2174/1381612826666200504115045] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 03/17/2020] [Indexed: 12/20/2022]
Abstract
Chronic inflammation plays an extensive role in the onset and progression of metabolic disorders such
as atherosclerosis, type 2 diabetes, gout and obesity. Atherosclerosis accounts for up to 70% mortality in patients
with type 2 diabetes and is also a chronic condition that causes atrial stenosis due to a lipometabolism imbalance.
The purpose of this article is to consider the inflammatory factors implicated in atherosclerosis and their role in
the development and progression of this vascular disease. The inflammasome signaling pathway is an important
inflammatory mechanism involved in the development of atherosclerosis. The most important inflammasome
pathway in this respect is the NLRP3 inflammasome (Nucleotide-binding oligomerization domain (NOD)-like
receptor with a pyrin domain 3), whose activation leads to the generation of important inflammatory cytokines
including interleukins 1β and 18 (IL-1β and 18). The activities of these mature cytokines and inflammatory factors
produced by other inflammatory pathways lead to arterial inflammation and eventually arterial occlusion,
which can result in life-threatening complications such as myocardial infarction and stroke. Therefore, it is essential
to seek out more precise mechanisms for the activation of inflammasomes and other inflammatory pathways
for the development of therapeutic strategies of atherosclerosis.
Collapse
Affiliation(s)
- Zeinab Nazarian-Samani
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Robert D. E. Sewell
- Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, CF10 3NB. Wales, United Kingdom
| | - Mahmoud Rafieian-Kopaei
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
14
|
Chen WY, Chen YF, Chan HC, Chung CH, Peng HY, Ho YC, Chen CH, Chang KC, Tang CH, Lee AS. Role of apolipoprotein E in electronegative low-density lipoprotein-induced mitochondrial dysfunction in cardiomyocytes. Metabolism 2020; 107:154227. [PMID: 32275974 DOI: 10.1016/j.metabol.2020.154227] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 03/18/2020] [Accepted: 04/07/2020] [Indexed: 12/23/2022]
Abstract
OBJECTIVE L5, a highly electronegative subtype of low-density lipoprotein (LDL), is likely associated with the development of atherosclerosis and cardiovascular diseases. Normal LDL is composed mainly of apolipoprotein (Apo) B, but L5 has additional proteins such as ApoE. We previously demonstrated that L5 induces endothelial cell senescence by increasing mitochondrial reactive oxygen species. In the present study, we examined the effect of L5 on mitochondrial function in cardiomyocytes. METHODS We used the Seahorse XF24 extracellular flux analyzer to examine the effect of L5 and its components on mitochondrial energy production. The effects of L5 on mitochondrial morphology were examined by immunofluorescence using MitoTracker Green FM and the corresponding probes in H9c2 cardiomyoblasts. Mitochondrial permeability was assessed by using a calcium-induced swelling assay with a voltage-dependent anion-selective channel (VDAC) inhibitor to determine VDAC-dependence both in vitro and in vivo. L5 without ApoE, referred to as △L5, was used to clarify the role of ApoE in L5-induced mitochondrial dysfunction. RESULTS L5 not only significantly decreased basal (P < 0.05) and maximal respiration (P < 0.01) but also reduced spare respiratory capacity (P < 0.01) in H9c2 cells. Additionally, L5 caused phosphorylation of Drp1 and mitochondrial fission. Recombinant ApoE mimicked the mitochondrial effects of L5, but △L5 did not cause similar effects. After entering cells, ApoE on L5 colocalized with mitochondrial VDAC and caused mitochondria swelling both in vitro and in vivo. This effect was also seen with recombinant ApoE but not △L5. CONCLUSIONS ApoE may play an important role in electronegative LDL-induced mitochondrial dysfunction through the opening of the mitochondrial permeability transition pore via the interaction of ApoE and VDAC.
Collapse
Affiliation(s)
- Wei-Yu Chen
- Graduate Institute of Basic Medical Science, China Medical University, Taichung 40402, Taiwan; Department of Medicine, Mackay Medical College, New Taipei City 25245, Taiwan; Cardiovascular Research Laboratory, China Medical University Hospital, Taichung 40447, Taiwan
| | - Yun-Fang Chen
- Department of Medicine, Mackay Medical College, New Taipei City 25245, Taiwan; Cardiovascular Research Laboratory, China Medical University Hospital, Taichung 40447, Taiwan; Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung 40402, Taiwan
| | - Hua-Cheng Chan
- Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan
| | - Ching-Hu Chung
- Department of Medicine, Mackay Medical College, New Taipei City 25245, Taiwan
| | - Hsien-Yu Peng
- Department of Medicine, Mackay Medical College, New Taipei City 25245, Taiwan
| | - Yu-Cheng Ho
- Department of Medicine, Mackay Medical College, New Taipei City 25245, Taiwan.
| | - Chu-Huang Chen
- Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan; Vascular and Medicinal Research, Texas Heart Institute, Houston, TX 77030, USA; New York Heart Research Foundation, Mineola, New York 11501, USA.
| | - Kuan-Cheng Chang
- Cardiovascular Research Laboratory, China Medical University Hospital, Taichung 40447, Taiwan
| | - Chih-Hsin Tang
- Department of Pharmacology, School of Medicine, China Medical University, Taichung 40402, Taiwan.
| | - An-Sheng Lee
- Department of Medicine, Mackay Medical College, New Taipei City 25245, Taiwan; Cardiovascular Research Laboratory, China Medical University Hospital, Taichung 40447, Taiwan.
| |
Collapse
|
15
|
Artyukov AA, Zelepuga EA, Bogdanovich LN, Lupach NM, Novikov VL, Rutckova TA, Kozlovskaya EP. Marine Polyhydroxynaphthoquinone, Echinochrome A: Prevention of Atherosclerotic Inflammation and Probable Molecular Targets. J Clin Med 2020; 9:E1494. [PMID: 32429179 PMCID: PMC7291202 DOI: 10.3390/jcm9051494] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 05/06/2020] [Accepted: 05/13/2020] [Indexed: 12/16/2022] Open
Abstract
The effect of low doses of echinochrome A (EchA), a natural polyhydroxy-1,4-naphthoquinone pigment from the sea urchin Scaphechinus mirabilis, has been studied in clinical trials, when it was used as an active substance of the drug Histochrome® and biologically active supplement Thymarin. Several parameters of lipid metabolism, antioxidant status, and the state of the immune system were analyzed in patients with cardiovascular diseases (CVD), including contaminating atherosclerosis. It has been shown that EchA effectively normalizes lipid metabolism, recovers antioxidant status and reduces atherosclerotic inflammation, regardless of the method of these preparations' administrations. Treatment of EchA has led to the stabilization of patients, improved function of the intracellular matrix and decreased epithelial dysfunction. The increased expression of surface human leukocyte antigen DR isotype (HLA-DR) receptors reflects the intensification of intercellular cooperation of immune cells, as well as an increase in the efficiency of processing and presentation of antigens, while the regulation of CD95 + expression levels suggests the stimulation of cell renewal processes. The immune system goes to a different level of functioning. Computer simulations suggest that EchA, with its aromatic structure of the naphthoquinone nucleus, may be a suitable ligand of the cytosolic aryl cell receptor, which affects the response of the immune system and causes the rapid expression of detoxification enzymes such as CYP and DT diaphorase, which play a protective role with CVD. Therefore, EchA possesses not only an antiradical effect and antioxidant activity, but is also a SOD3 mimetic, producing hydrogen peroxide and controlling the expression of cell enzymes through hypoxia-inducible factors (HIF), peroxisome proliferator-activated receptors (PPARs) and aryl hydrocarbon receptor (AhR).
Collapse
Affiliation(s)
- Aleksandr A. Artyukov
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Science, 159 Prospect 100-letiya Vladivostoka, Vladivostok 690022, Russia; (A.A.A.); (E.A.Z.); (V.L.N.); (T.A.R.)
| | - Elena A. Zelepuga
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Science, 159 Prospect 100-letiya Vladivostoka, Vladivostok 690022, Russia; (A.A.A.); (E.A.Z.); (V.L.N.); (T.A.R.)
| | - Larisa N. Bogdanovich
- Medical Association of the Far Eastern Branch of the Russian Academy of Sciences (FEB RAS MO), Kirov Str., 95, Vladivostok 690022, Russia;
| | - Natalia M. Lupach
- Primorye Regional Clinical Hospital No. One (SHI), Aleutskaya Str., 57, Vladivostok, Primorsky Krai 690091, Russia;
| | - Vyacheslav L. Novikov
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Science, 159 Prospect 100-letiya Vladivostoka, Vladivostok 690022, Russia; (A.A.A.); (E.A.Z.); (V.L.N.); (T.A.R.)
| | - Tatyana A. Rutckova
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Science, 159 Prospect 100-letiya Vladivostoka, Vladivostok 690022, Russia; (A.A.A.); (E.A.Z.); (V.L.N.); (T.A.R.)
| | - Emma P. Kozlovskaya
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Science, 159 Prospect 100-letiya Vladivostoka, Vladivostok 690022, Russia; (A.A.A.); (E.A.Z.); (V.L.N.); (T.A.R.)
| |
Collapse
|
16
|
Abstract
BACKGROUND AND AIMS The long-term effect of immune tolerance has not been explored so far in atherosclerosis. In the present study, we assessed the effect of mucosal tolerance to a multi antigenic construct expressing three peptides from ApoB, HSP60, and outer membrane protein from Chlamydia pneumonia (AHC) for 30 weeks at every 6-week interval to understand the kinetics of immune modulation in disease progression. The safety profile of the molecule was also evaluated in mice. METHODS Apobtm2SgyLdlrtm1Her/J mice (5-6 weeks) were orally dosed with multi antigenic construct (AHC) molecule on alternate days, followed by high-fat diet feeding to initiate atherosclerosis. RESULTS Treated animals showed an efficient reduction in plaque growth and lipid accumulation at 6 weeks (49%, p < 0.01) and 12 weeks (42.3%, p < 0.01) which decreased to 29% (p = 0.0001) at 18 weeks and at later time points. Macrophage accumulation was significantly lower at all time points (53% at 12 weeks to 27% at 30 weeks). Regulatory T cells increased in the spleen following treatment until 12 weeks (week 0 (2.57 ± 0.18 vs. 6.36 ± 0.03, p = 0.02), week 6 (4.52 ± 0.2 vs. 8.87 ± 0.32, p = 0.02), and week 12 (8.74 ± 0.37 vs. 15.4 ± 0.27, p = 0.02)) but showed a decline later. A similar trend was observed with tolerogenic dendritic cells. We observed an increase in antibody levels to low-density lipoprotein and oxidized LDL at later stages. AHC molecule was found to be safe in acute and repeated dose toxicity studies. CONCLUSIONS Our results suggest that immune tolerance to AHC protein by oral administration is able to provide efficient atheroprotection up to 18 weeks and moderately at later stages. Apart from immune regulatory cells, protective antibodies may also have a role in controlling atherosclerosis.
Collapse
|
17
|
Knutsson A, Björkbacka H, Dunér P, Engström G, Binder CJ, Nilsson AH, Nilsson J. Associations of Interleukin-5 With Plaque Development and Cardiovascular Events. JACC Basic Transl Sci 2019; 4:891-902. [PMID: 31909299 PMCID: PMC6939009 DOI: 10.1016/j.jacbts.2019.07.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 07/09/2019] [Accepted: 07/10/2019] [Indexed: 12/13/2022]
Abstract
Experimental studies have suggested an atheroprotective role of interleukin (IL)-5 through the stimulation of natural immunoglobulin M antibody expression. In the present study we show that there are no associations between baseline levels of IL-5 and risk for development of coronary events or stroke during a 15.7 ± 6.3 years follow-up of 696 subjects randomly sampled from the Malmö Diet and Cancer study. However, presence of a plaque at the carotid bifurcation was associated with lower IL-5 and IL-5 deficiency resulted in increased plaque development at sites of oscillatory blood flow in Apoe -/- mice suggesting a protective role for IL-5 in plaque development.
Collapse
Affiliation(s)
- Anki Knutsson
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Harry Björkbacka
- Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Pontus Dunér
- Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Gunnar Engström
- Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | | | - Jan Nilsson
- Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| |
Collapse
|
18
|
Mestre AP, Amavet PS, Vanzetti AI, Moleón MS, Parachú Marcó MV, Poletta GL, Siroski PA. Effects of cypermethrin (pyrethroid), glyphosate and chlorpyrifos (organophosphorus) on the endocrine and immune system of Salvator merianae (Argentine tegu). ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2019; 169:61-67. [PMID: 30419507 DOI: 10.1016/j.ecoenv.2018.10.057] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 10/11/2018] [Accepted: 10/13/2018] [Indexed: 05/27/2023]
Abstract
Several geographical areas where Salvator merianae is distributed in Argentina are included in regions with agricultural activity and exposed to pesticide formulations. Some pesticides could affect defense mechanisms being able alter structures of some components of immune and endocrine systems. To assess the potential effects of pesticides in this reptile under seminatural conditions, on the immune system and endocrine responses in S. merianae we analyzed several blood parameters. Total (TWBCC), differential (DWBCC) white blood cells count, heterophils/lymphocytes index (H/L), lobularity index (LI), natural antibodies (NAbs) titres, complement system (CS), and corticosterone concentration were analyzed in animals exposed to a mixture of cypermethrin (25%), glyphosate (66.2%) and chlorpyrifos (48%) formulations. In addition, body size was considered in these analyzes. TWBCC and NAbs revealed lower values in organisms exposed to pesticides respect to a control indicating a possible immunosuppression effect. Besides, the LI showed a greater number of lobes in organism exposed demonstrating symptoms of chronic infection. In addition, we observed a reduced growth in these animals possibly related to a less energy investment in body mass to maintain an active defense against pesticides. Finally, we found high levels of plasma corticosterone in animals exposed to mix formulation that could demonstrate neuroendocrine axis activation. Other parameters like DWBCC, H/L index and activity of CS showed no differences in treated animals respect to control group, which could indicate low sensibility of these parameters to the concentration of pesticides used. Our results provide evidence of the toxic effects of pesticides on different immune system parameters, but also a trade-off among these parameters, corticosterone levels and growth. In this way, we can conclude that the formulated pesticides applied widely and constantly in the areas occupied by S. merianae, would be affecting its immune and endocrine systems and therefore its ability to defend against external agents. This kind of studies is of great interest to know the possible responses of wild species to anthropogenic disturbances such as pesticide contamination.
Collapse
Affiliation(s)
- Ana P Mestre
- Laboratorio de Zoología Aplicada: Anexo Vertebrados, Facultad de Humanidades y Ciencias, Universidad Nacional del Litoral, (FHUC - UNL /MMA) Santa Fe, Argentina; Laboratorio de Genética, Departamento de Ciencias Naturales (FHUC - UNL), CONICET, Santa Fe, Argentina.
| | - Patricia S Amavet
- Laboratorio de Genética, Departamento de Ciencias Naturales (FHUC - UNL), CONICET, Santa Fe, Argentina.
| | - Agustín I Vanzetti
- Laboratorio de Genética, Departamento de Ciencias Naturales (FHUC - UNL), CONICET, Santa Fe, Argentina.
| | - Ma Soledad Moleón
- Laboratorio de Zoología Aplicada: Anexo Vertebrados, Facultad de Humanidades y Ciencias, Universidad Nacional del Litoral, (FHUC - UNL /MMA) Santa Fe, Argentina; Laboratorio de Ecología Molecular Aplicada, Instituto de Ciencias Veterinarias del Litoral (ICiVet Litoral) UNL - CONICET, Esperanza, Argentina.
| | - Ma Virginia Parachú Marcó
- Laboratorio de Zoología Aplicada: Anexo Vertebrados, Facultad de Humanidades y Ciencias, Universidad Nacional del Litoral, (FHUC - UNL /MMA) Santa Fe, Argentina; Laboratorio de Ecología Molecular Aplicada, Instituto de Ciencias Veterinarias del Litoral (ICiVet Litoral) UNL - CONICET, Esperanza, Argentina.
| | - Gisela L Poletta
- Laboratorio de Zoología Aplicada: Anexo Vertebrados, Facultad de Humanidades y Ciencias, Universidad Nacional del Litoral, (FHUC - UNL /MMA) Santa Fe, Argentina; Cátedra de Toxicología, Farmacología y Bioquímica Legal (FBCB-UNL), CONICET, Santa Fe, Argentina.
| | - Pablo A Siroski
- Laboratorio de Zoología Aplicada: Anexo Vertebrados, Facultad de Humanidades y Ciencias, Universidad Nacional del Litoral, (FHUC - UNL /MMA) Santa Fe, Argentina; Laboratorio de Ecología Molecular Aplicada, Instituto de Ciencias Veterinarias del Litoral (ICiVet Litoral) UNL - CONICET, Esperanza, Argentina.
| |
Collapse
|
19
|
Jakubowski H. Homocysteine Modification in Protein Structure/Function and Human Disease. Physiol Rev 2019; 99:555-604. [PMID: 30427275 DOI: 10.1152/physrev.00003.2018] [Citation(s) in RCA: 169] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Epidemiological studies established that elevated homocysteine, an important intermediate in folate, vitamin B12, and one carbon metabolism, is associated with poor health, including heart and brain diseases. Earlier studies show that patients with severe hyperhomocysteinemia, first identified in the 1960s, exhibit neurological and cardiovascular abnormalities and premature death due to vascular complications. Although homocysteine is considered to be a nonprotein amino acid, studies over the past 2 decades have led to discoveries of protein-related homocysteine metabolism and mechanisms by which homocysteine can become a component of proteins. Homocysteine-containing proteins lose their biological function and acquire cytotoxic, proinflammatory, proatherothrombotic, and proneuropathic properties, which can account for the various disease phenotypes associated with hyperhomocysteinemia. This review describes mechanisms by which hyperhomocysteinemia affects cellular proteostasis, provides a comprehensive account of the biological chemistry of homocysteine-containing proteins, and discusses pathophysiological consequences and clinical implications of their formation.
Collapse
Affiliation(s)
- Hieronim Jakubowski
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers-New Jersey Medical School, International Center for Public Health , Newark, New Jersey ; and Department of Biochemistry and Biotechnology, Poznań University of Life Sciences , Poznań , Poland
| |
Collapse
|
20
|
Crane ED, Al-Hashimi AA, Chen J, Lynn EG, Won KD, Lhoták Š, Naeim M, Platko K, Lebeau P, Byun JH, Shayegan B, Krepinsky JC, Rayner KJ, Marchiò S, Pasqualini R, Arap W, Austin RC. Anti-GRP78 autoantibodies induce endothelial cell activation and accelerate the development of atherosclerotic lesions. JCI Insight 2018; 3:99363. [PMID: 30568038 DOI: 10.1172/jci.insight.99363] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 11/09/2018] [Indexed: 12/31/2022] Open
Abstract
The 78-kDa glucose-regulated protein (GRP78) is an ER molecular chaperone that aids in protein folding and secretion. However, pathological conditions that cause ER stress can promote the relocalization of GRP78 to the cell surface (csGRP78), where it acts as a signaling receptor to promote cancer progression. csGRP78 also possesses antigenic properties, leading to the production of anti-GRP78 autoantibodies, which contribute to tumor growth. In contrast, the presence and role of anti-GRP78 autoantibodies in atherosclerosis is unknown. Here, we show that atherosclerotic-prone ApoE-/- mice develop circulating anti-GRP78 autoantibodies that bind to csGRP78 on lesion-resident endothelial cells. Moreover, GRP78-immunized ApoE-/- mice exhibit a marked increase in circulating anti-GRP78 autoantibody titers that correlated with accelerated lesion growth. Mechanistically, engagement of anti-GRP78 autoantibodies with csGRP78 on human endothelial cells activated NF-κB, thereby inducing the expression of ICAM-1 and VCAM-1, a process blocked by NF-κB inhibitors. Disrupting the autoantibody/csGRP78 complex with enoxaparin, a low-molecular-weight heparin, reduced the expression of adhesion molecules and attenuated lesion growth. In conclusion, anti-GRP78 autoantibodies play a crucial role in atherosclerosis development, and disruption of the interaction between anti-GRP78 autoantibodies and csGRP78 represents a therapeutic strategy.
Collapse
Affiliation(s)
| | - Ali A Al-Hashimi
- Department of Medicine, Division of Nephrology, and.,Division of Urology, Department of Surgery, McMaster University and The Research Institute of St. Joe's Hamilton, Hamilton, Ontario, Canada
| | - Jack Chen
- Department of Medicine, Division of Nephrology, and
| | | | | | - Šárka Lhoták
- Department of Medicine, Division of Nephrology, and
| | - Magda Naeim
- Department of Medicine, Division of Nephrology, and
| | | | - Paul Lebeau
- Department of Medicine, Division of Nephrology, and
| | | | - Bobby Shayegan
- Division of Urology, Department of Surgery, McMaster University and The Research Institute of St. Joe's Hamilton, Hamilton, Ontario, Canada
| | | | - Katey J Rayner
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa and University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Serena Marchiò
- Department of Oncology, University of Turin, Candiolo, Italy.,Candiolo Cancer Institute - Fondazione del Piemonte per l'Oncologia, Istituto di Ricerca e Cura a Carattere Scientifico, Candiolo, Italy
| | - Renata Pasqualini
- Rutgers Cancer Institute of New Jersey, Newark, New Jersey, USA.,Division of Cancer Biology, Department of Radiation Oncology, and
| | - Wadih Arap
- Rutgers Cancer Institute of New Jersey, Newark, New Jersey, USA.,Division of Hematology/Oncology, Department of Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Richard C Austin
- Department of Biochemistry and Biomedical Sciences.,Department of Medicine, Division of Nephrology, and
| |
Collapse
|
21
|
Reustle A, Torzewski M. Role of p38 MAPK in Atherosclerosis and Aortic Valve Sclerosis. Int J Mol Sci 2018; 19:ijms19123761. [PMID: 30486366 PMCID: PMC6321637 DOI: 10.3390/ijms19123761] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 11/16/2018] [Accepted: 11/22/2018] [Indexed: 12/16/2022] Open
Abstract
Atherosclerosis and aortic valve sclerosis are cardiovascular diseases with an increasing prevalence in western societies. Statins are widely applied in atherosclerosis therapy, whereas no pharmacological interventions are available for the treatment of aortic valve sclerosis. Therefore, valve replacement surgery to prevent acute heart failure is the only option for patients with severe aortic stenosis. Both atherosclerosis and aortic valve sclerosis are not simply the consequence of degenerative processes, but rather diseases driven by inflammatory processes in response to lipid-deposition in the blood vessel wall and the aortic valve, respectively. The p38 mitogen-activated protein kinase (MAPK) is involved in inflammatory signaling and activated in response to various intracellular and extracellular stimuli, including oxidative stress, cytokines, and growth factors, all of which are abundantly present in atherosclerotic and aortic valve sclerotic lesions. The responses generated by p38 MAPK signaling in different cell types present in the lesions are diverse and might support the progression of the diseases. This review summarizes experimental findings relating to p38 MAPK in atherosclerosis and aortic valve sclerosis and discusses potential functions of p38 MAPK in the diseases with the aim of clarifying its eligibility as a pharmacological target.
Collapse
Affiliation(s)
- Anna Reustle
- Dr. Margarete-Fischer-Bosch-Institute of Clinical Pharmacology, 70376 Stuttgart, Germany.
- University of Tuebingen, 72074 Tuebingen, Germany.
| | - Michael Torzewski
- Department of Laboratory Medicine and Hospital Hygiene, Robert Bosch-Hospital, 70376 Stuttgart, Germany.
| |
Collapse
|
22
|
Ponnusamy T, Venkatachala SK, Ramanjappa M, Kakkar VV, Mundkur LA. Inverse association of ApoB and HSP60 antibodies with coronary artery disease in Indian population. HEART ASIA 2018; 10:e011018. [PMID: 30018661 DOI: 10.1136/heartasia-2018-011018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 05/31/2018] [Accepted: 06/01/2018] [Indexed: 12/12/2022]
Abstract
Objective Atherosclerosis is an autoimmune condition and the underlying cause of coronary artery disease (CAD). Circulating antibodies to self-antigens can have a pathogenic or protective function in atherosclerosis. The objective of the study was to understand the association of autoantibody levels with CAD and its correlation with circulating immune cells. Methods We assessed antigen concentration and antibodies to apolipoprotein B (ApoB) and heat shock protein (HSP)60 by ELISA in 252 acute coronary syndromes (ACS), 112 patients with stable angina (SA) and 203 healthy controls from Indian population. T cells in peripheral blood mononuclear cells (PBMC) were enumerated by flow cytometry. Cytokine concentrations were measured by multiplex assay. Results IgG and IgM antibodies to ApoB and HSP60 proteins were significantly lower in patients with ACS while only IgG levels to ApoB were lower in patients with SA, compared with control. Subjects in the highest tertile of antibodies showed significantly lower OR for ACS (IgG 0.52, 95% CI 0.31 to 0.88, p=0.02 and IgM 0.58, 95% CI 0.34 to 0.98, p=0.04), ApoB100 (IgG 0.52, 95% CI 0.31 to 0.88, p=0.02 and IgM 0.58, 95% CI 0.34 to 0.99, p=0.04) and HSP60, respectively. Interestingly, T helper 17 (TH17) cells showed an inverse relationship with ApoB and HSP60 IgG antibodies (r2=-0.17, p<0.001 and r2=-0.20, p<0.001, respectively), while interleukin 17 concentrations were negatively correlated with IgM antibodies to the proteins. Conclusion This study shows that higher antibodies to ApoB and HSP60 proteins are less often associated with ACS and that these antibodies are inversely associated with inflammatory Th17 cells.
Collapse
Affiliation(s)
- Thiruvelselvan Ponnusamy
- Mary and Gary Western and Tata Molecular Immunology Unit, Thrombosis Research Institute, Manipal University, Bangalore, India
| | | | | | - Vijay V Kakkar
- Mary and Gary Western and Tata Molecular Immunology Unit, Thrombosis Research Institute, Manipal University, Bangalore, India
| | - Lakshmi A Mundkur
- Mary and Gary Western and Tata Molecular Immunology Unit, Thrombosis Research Institute, Manipal University, Bangalore, India
| |
Collapse
|
23
|
Rahman K, Fisher EA. Insights From Pre-Clinical and Clinical Studies on the Role of Innate Inflammation in Atherosclerosis Regression. Front Cardiovasc Med 2018; 5:32. [PMID: 29868610 PMCID: PMC5958627 DOI: 10.3389/fcvm.2018.00032] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Accepted: 03/20/2018] [Indexed: 12/11/2022] Open
Abstract
Atherosclerosis, the underlying cause of coronary artery (CAD) and other cardiovascular diseases, is initiated by macrophage-mediated immune responses to lipoprotein and cholesterol accumulation in artery walls, which result in the formation of plaques. Unlike at other sites of inflammation, the immune response becomes maladaptive and inflammation fails to resolve. The most common treatment for reducing the risk from atherosclerosis is low density lipoprotein cholesterol (LDL-C) lowering. Studies have shown, however, that while significant lowering of LDL-C reduces the risk of heart attacks to some degree, there is still residual risk for the majority of the population. We and others have observed “residual inflammatory risk” of atherosclerosis after plasma cholesterol lowering in pre-clinical studies, and that this phenomenon is clinically relevant has been dramatically reinforced by the recent Canakinumab Anti-inflammatory Thrombosis Outcomes Study (CANTOS) trial. This review will summarize the role of the innate immune system, specifically macrophages, in atherosclerosis progression and regression, as well as the pre-clinical and clinical models that have provided significant insights into molecular pathways involved in the resolution of plaque inflammation and plaque regression. Partnered with clinical studies that can be envisioned in the post-CANTOS period, including progress in developing targeted plaque therapies, we expect that pre-clinical studies advancing on the path summarized in this review, already revealing key mechanisms, will continue to be essential contributors to achieve the goals of dampening plaque inflammation and inducing its resolution in order to maximize the therapeutic benefits of conventional risk factor modifications, such as LDL-C lowering.
Collapse
Affiliation(s)
- Karishma Rahman
- Department of Medicine, Division of Cardiology, New York University School of Medicine, New York, NY, United States
| | - Edward A Fisher
- Department of Medicine, Division of Cardiology, New York University School of Medicine, New York, NY, United States
| |
Collapse
|
24
|
Bayersdorf R, Fruscalzo A, Catania F. Linking autoimmunity to the origin of the adaptive immune system. EVOLUTION MEDICINE AND PUBLIC HEALTH 2018; 2018:2-12. [PMID: 29423226 PMCID: PMC5793817 DOI: 10.1093/emph/eoy001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
In jawed vertebrates, the adaptive immune system (AIS) cooperates with the innate immune system (IIS) to protect hosts from infections. Although targeting non-self-components, the AIS also generates self-reactive antibodies which, when inadequately counter-selected, can give rise to autoimmune diseases (ADs). ADs are on the rise in western countries. Why haven’t ADs been eliminated during the evolution of a ∼500 million-year old system? And why have they become more frequent in recent decades? Self-recognition is an attribute of the phylogenetically more ancient IIS and empirical data compellingly show that some self-reactive antibodies, which are classifiable as elements of the IIS rather then the AIS, may protect from (rather than cause) ADs. Here, we propose that the IIS’s self-recognition system originally fathered the AIS and, as a consequence of this relationship, its activity is dampened in hygienic environments. Rather than a mere breakdown or failure of the mechanisms of self-tolerance, ADs might thus arise from architectural constraints.
Collapse
Affiliation(s)
- Robert Bayersdorf
- Institute for Genome Stability in Aging and Disease, Medical Faculty, University of Cologne, 50931 Cologne, Germany.,Institute for Evolution and Biodiversity, University of Münster, 48149 Münster, Germany
| | - Arrigo Fruscalzo
- Clinic of Obstetrics and Gynecology, St Franziskus Hospital, 59227 Ahlen, Germany.,Department of Obstetrics and Gynecology, University Hospital of Münster, 48149 Münster, Germany
| | - Francesco Catania
- Institute for Evolution and Biodiversity, University of Münster, 48149 Münster, Germany
| |
Collapse
|
25
|
Immunization with gingipain A hemagglutinin domain of Porphyromonas gingivalis induces IgM antibodies binding to malondialdehyde-acetaldehyde modified low-density lipoprotein. PLoS One 2018; 13:e0191216. [PMID: 29329335 PMCID: PMC5766137 DOI: 10.1371/journal.pone.0191216] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 12/29/2017] [Indexed: 12/12/2022] Open
Abstract
Treatment of periodontitis has beneficial effects on systemic inflammation markers that relate to progression of atherosclerosis. We aimed to investigate whether immunization with A hemagglutinin domain (Rgp44) of Porphyromonas gingivalis (Pg), a major etiologic agent of periodontitis, would lead to an antibody response cross-reacting with oxidized low-density lipoprotein (OxLDL) and how it would affect the progression of atherosclerosis in low-density lipoprotein receptor-deficient (LDLR-/-) mice. The data revealed a prominent IgM but not IgG response to malondialdehyde-acetaldehyde modified LDL (MAA-LDL) after Rgp44 and Pg immunizations, implying that Rgp44/Pg and MAA adducts may share cross-reactive epitopes that prompt IgM antibody production and consequently confer atheroprotection. A significant negative association was observed between atherosclerotic lesion and plasma IgA to Rgp44 in Rgp44 immunized mice, supporting further the anti-atherogenic effect of Rgp44 immunization. Plasma IgA levels to Rgp44 and to Pg in both Rgp44- and Pg-immunized mice were significantly higher than those in saline control, suggesting that IgA to Rgp44 could be a surrogate marker of immunization in Pg-immunized mice. Distinct antibody responses in plasma IgA levels to MAA-LDL, to Pg lipopolysaccharides (Pg-LPS), and to phosphocholine (PCho) were observed after Rgp44 and Pg immunizations, indicating that different immunogenic components between Rpg44 and Pg may behave differently in regard of their roles in the development of atherosclerosis. Immunization with Rgp44 also displayed atheroprotective features in modulation of plaque size through association with plasma levels of IL-1α whereas whole Pg bacteria achieved through regulation of anti-inflammatory cytokine levels of IL-5 and IL-10. The present study may contribute to refining therapeutic approaches aiming to modulate immune responses and inflammatory/anti-inflammatory processes in atherosclerosis.
Collapse
|
26
|
Hernandez AM, Holodick NE. Editorial: Natural Antibodies in Health and Disease. Front Immunol 2017; 8:1795. [PMID: 29326696 PMCID: PMC5733483 DOI: 10.3389/fimmu.2017.01795] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 11/30/2017] [Indexed: 11/13/2022] Open
Affiliation(s)
- Ana Maria Hernandez
- Natural Antibodies Group, Tumor Immunology Division, Center of Molecular Immunology, Havana, Cuba
| | - Nichol E Holodick
- Department of Biomedical Sciences, Center for Immunobiology, Western Michigan University Homer Stryker MD School of Medicine, Kalamazoo, MI, United States
| |
Collapse
|
27
|
Serbulea V, DeWeese D, Leitinger N. The effect of oxidized phospholipids on phenotypic polarization and function of macrophages. Free Radic Biol Med 2017; 111:156-168. [PMID: 28232205 PMCID: PMC5511074 DOI: 10.1016/j.freeradbiomed.2017.02.035] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 02/09/2017] [Accepted: 02/15/2017] [Indexed: 12/26/2022]
Abstract
Oxidized phospholipids are products of lipid oxidation that are found on oxidized low-density lipoproteins and apoptotic cell membranes. These biologically active lipids were shown to affect a variety of cell types and attributed pro-as well as anti-inflammatory effects. In particular, macrophages exposed to oxidized phospholipids drastically change their gene expression pattern and function. These 'Mox,'macrophages were identified in atherosclerotic lesions, however, it remains unclear how lipid oxidation products are sensed by macrophages and how they influence their biological function. Here, we review recent developments in the field that provide insight into the structure, recognition, and downstream signaling of oxidized phospholipids in macrophages.
Collapse
Affiliation(s)
- Vlad Serbulea
- Robert M. Berne Cardiovascular Research Center and Department of Pharmacology, University of Virginia, USA
| | - Dory DeWeese
- Robert M. Berne Cardiovascular Research Center and Department of Pharmacology, University of Virginia, USA
| | - Norbert Leitinger
- Robert M. Berne Cardiovascular Research Center and Department of Pharmacology, University of Virginia, USA
| |
Collapse
|
28
|
Wu MY, Li CJ, Hou MF, Chu PY. New Insights into the Role of Inflammation in the Pathogenesis of Atherosclerosis. Int J Mol Sci 2017; 18:ijms18102034. [PMID: 28937652 PMCID: PMC5666716 DOI: 10.3390/ijms18102034] [Citation(s) in RCA: 268] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 09/19/2017] [Accepted: 09/19/2017] [Indexed: 02/07/2023] Open
Abstract
Atherosclerosis is a chronic inflammatory disease characterized by the accumulation of lipids, smooth muscle cell proliferation, cell apoptosis, necrosis, fibrosis, and local inflammation. Immune and inflammatory responses have significant effects on every phase of atherosclerosis, and increasing evidence shows that immunity plays a more important role in atherosclerosis by tightly regulating its progression. Therefore, understanding the relationship between immune responses and the atherosclerotic microenvironment is extremely important. This article reviews existing knowledge regarding the pathogenesis of immune responses in the atherosclerotic microenvironment, and the immune mechanisms involved in atherosclerosis formation and activation.
Collapse
Affiliation(s)
- Meng-Yu Wu
- Department of Emergency Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 231, Taiwan.
- Department of Emergency Medicine, School of Medicine, Tzu Chi University, Hualien 970, Taiwan.
| | - Chia-Jung Li
- Research Assistant Center, Show Chwan Memorial Hospital, Changhua 500, Taiwan.
| | - Ming-Feng Hou
- Department of Surgery, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Department of Surgery, Kaohsiung Municipal Hsiao Kang Hospital, Kaohsiung 807, Taiwan.
- Division of Breast Surgery, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan.
| | - Pei-Yi Chu
- Department of Pathology, Show Chwan Memorial Hospital, Changhua 500, Taiwan.
- School of Medicine, College of Medicine, Fu-Jen Catholic University, New Taipei 242, Taiwan.
- National Institute of Cancer Research, National Health Research Institutes, Tainan 704, Taiwan.
| |
Collapse
|
29
|
Meeuwsen JAL, van Duijvenvoorde A, Gohar A, Kozma MO, van de Weg SM, Gijsberts CM, Haitjema S, Björkbacka H, Fredrikson GN, de Borst GJ, den Ruijter HM, Pasterkamp G, Binder CJ, Hoefer IE, de Jager SCA. High Levels of (Un)Switched Memory B Cells Are Associated With Better Outcome in Patients With Advanced Atherosclerotic Disease. J Am Heart Assoc 2017; 6:e005747. [PMID: 28882820 PMCID: PMC5634255 DOI: 10.1161/jaha.117.005747] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 07/07/2017] [Indexed: 12/31/2022]
Abstract
BACKGROUND Atherosclerosis is an inflammatory lipid disorder and the main underlying pathology of acute ischemic events. Despite a vast amount of data from murine atherosclerosis models, evidence of B-cell involvement in human atherosclerotic disease is limited. We therefore investigated the association of circulating B-cell subtypes with the occurrence of secondary cardiovascular events in advanced atherosclerotic disease. METHODS AND RESULTS This cohort study consists of 168 patients who were included in the Athero-Express biobank between 2009 and 2011. Before surgery, peripheral blood mononuclear cells were isolated and stored in liquid nitrogen. After gentle thawing of the peripheral blood mononuclear cells, different B-cell subtypes including naïve, (un)switched memory, and CD27+CD43+ B1-like B cells, were analyzed by flow cytometry. Univariable and multivariable Cox proportional hazard models were used to analyze associations between B-cell subtypes, circulating antibodies and secondary cardiovascular manifestations during the 3-year follow-up period. Mean age was 70.1±9.6 years, males represented 62.8% of the population, and 54 patients had secondary manifestations during follow-up. High numbers of unswitched memory cells were protective against secondary outcome (hazard ratio, 0.30 [95% CI, 0.13-0.69]; P<0.01). Similar results were obtained for the switched memory cells that also showed to be protective against secondary outcome (hazard ratio, 0.33 [95% CI, 0.14-0.77]; P=0.01). CONCLUSIONS A high number of (un)switched memory B cells is associated with better outcome following carotid artery endarterectomy. These findings suggest a potential role for B-cell subsets in prediction and prevention of secondary cardiovascular events in patients with atherosclerosis.
Collapse
Affiliation(s)
- John A L Meeuwsen
- Laboratory for Experimental Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Amerik van Duijvenvoorde
- Laboratory for Experimental Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Aisha Gohar
- Laboratory for Experimental Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Maria O Kozma
- Department of Laboratory Medicine, Medical University of Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Sander M van de Weg
- Laboratory for Experimental Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Crystel M Gijsberts
- Laboratory for Experimental Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Saskia Haitjema
- Laboratory for Experimental Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Harry Björkbacka
- Department of Clinical Sciences, Skåne University Hospital Malmö, Lund University, Malmö, Sweden
| | - Gunilla N Fredrikson
- Department of Clinical Sciences, Skåne University Hospital Malmö, Lund University, Malmö, Sweden
| | - Gert J de Borst
- Department of Vascular Surgery, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Hester M den Ruijter
- Laboratory for Experimental Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Gerard Pasterkamp
- Laboratory for Experimental Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
- Laboratory for Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Imo E Hoefer
- Laboratory for Experimental Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
- Laboratory for Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Saskia C A de Jager
- Laboratory for Experimental Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| |
Collapse
|
30
|
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
|
31
|
Holodick NE, Rodríguez-Zhurbenko N, Hernández AM. Defining Natural Antibodies. Front Immunol 2017; 8:872. [PMID: 28798747 PMCID: PMC5526850 DOI: 10.3389/fimmu.2017.00872] [Citation(s) in RCA: 142] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 07/10/2017] [Indexed: 01/05/2023] Open
Abstract
The traditional definition of natural antibodies (NAbs) states that these antibodies are present prior to the body encountering cognate antigen, providing a first line of defense against infection thereby, allowing time for a specific antibody response to be mounted. The literature has a seemingly common definition of NAbs; however, as our knowledge of antibodies and B cells is refined, re-evaluation of the common definition of Nabs may be required. Defining Nabs becomes important as the function of NAb production is used to define B cell subsets (1) and as these important molecules are shown to play numerous roles in the immune system (Figure 1). Herein, we aim to briefly summarize our current knowledge of NAbs in the context of initiating a discussion within the field of how such an important and multifaceted group of molecules should be defined.
Collapse
Affiliation(s)
- Nichol E Holodick
- Department of Biomedical Sciences, Center for Immunobiology, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, United States
| | - Nely Rodríguez-Zhurbenko
- Natural Antibodies Group, Tumor Immunology Division, Center of Molecular Immunology, Havana, Cuba
| | - Ana María Hernández
- Natural Antibodies Group, Tumor Immunology Division, Center of Molecular Immunology, Havana, Cuba
| |
Collapse
|
32
|
Oehler B, Kistner K, Martin C, Schiller J, Mayer R, Mohammadi M, Sauer RS, Filipovic MR, Nieto FR, Kloka J, Pflücke D, Hill K, Schaefer M, Malcangio M, Reeh PW, Brack A, Blum R, Rittner HL. Inflammatory pain control by blocking oxidized phospholipid-mediated TRP channel activation. Sci Rep 2017; 7:5447. [PMID: 28710476 PMCID: PMC5511297 DOI: 10.1038/s41598-017-05348-3] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 06/01/2017] [Indexed: 12/31/2022] Open
Abstract
Phospholipids occurring in cell membranes and lipoproteins are converted into oxidized phospholipids (OxPL) by oxidative stress promoting atherosclerotic plaque formation. Here, OxPL were characterized as novel targets in acute and chronic inflammatory pain. Oxidized 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphocholine (OxPAPC) and its derivatives were identified in inflamed tissue by mass spectrometry and binding assays. They elicited calcium influx, hyperalgesia and induced pro-nociceptive peptide release. Genetic, pharmacological and mass spectrometric evidence in vivo as well as in vitro confirmed the role of transient receptor potential channels (TRPA1 and TRPV1) as OxPAPC targets. Treatment with the monoclonal antibody E06 or with apolipoprotein A-I mimetic peptide D-4F, capturing OxPAPC in atherosclerosis, prevented inflammatory hyperalgesia, and in vitro TRPA1 activation. Administration of D-4F or E06 to rats profoundly ameliorated mechanical hyperalgesia and inflammation in collagen-induced arthritis. These data reveal a clinically relevant role for OxPAPC in inflammation offering therapy for acute and chronic inflammatory pain treatment by scavenging OxPAPC.
Collapse
Affiliation(s)
- Beatrice Oehler
- Department of Anesthesiology, University Hospital of Wuerzburg, Wuerzburg, Germany.,Institute of Clinical Neurobiology, University Hospital of Wuerzburg, Wuerzburg, Germany
| | - Katrin Kistner
- Institute for Physiology and Pathophysiology, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - Corinna Martin
- Department of Anesthesiology, University Hospital of Wuerzburg, Wuerzburg, Germany.,Institute of Clinical Neurobiology, University Hospital of Wuerzburg, Wuerzburg, Germany
| | - Jürgen Schiller
- Institute for Medical Physics and Biophysics, University of Leipzig, Leipzig, Germany
| | - Rafaela Mayer
- Department of Anesthesiology, University Hospital of Wuerzburg, Wuerzburg, Germany.,Institute of Clinical Neurobiology, University Hospital of Wuerzburg, Wuerzburg, Germany
| | - Milad Mohammadi
- Department of Anesthesiology, University Hospital of Wuerzburg, Wuerzburg, Germany.,Institute of Clinical Neurobiology, University Hospital of Wuerzburg, Wuerzburg, Germany
| | - Reine-Solange Sauer
- Department of Anesthesiology, University Hospital of Wuerzburg, Wuerzburg, Germany
| | - Milos R Filipovic
- Department of Chemistry and Pharmacy, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany.,University of Bordeaux, IBGC, UMR 5095, Bordeaux, France
| | - Francisco R Nieto
- Wolfson CARD, King's College London, Guys' Campus, London, United Kingdom.,University of Granada, Department of Pharmacology, Granada, Spain
| | - Jan Kloka
- Department of Anesthesiology, University Hospital of Wuerzburg, Wuerzburg, Germany.,Institute of Clinical Neurobiology, University Hospital of Wuerzburg, Wuerzburg, Germany
| | - Diana Pflücke
- Department of Anesthesiology, University Hospital of Wuerzburg, Wuerzburg, Germany
| | - Kerstin Hill
- Rudolf-Boehm-Institute for Pharmacology and Toxicology, University of Leipzig, Leipzig, Germany
| | - Michael Schaefer
- Rudolf-Boehm-Institute for Pharmacology and Toxicology, University of Leipzig, Leipzig, Germany
| | - Marzia Malcangio
- Wolfson CARD, King's College London, Guys' Campus, London, United Kingdom
| | - Peter W Reeh
- Institute for Physiology and Pathophysiology, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - Alexander Brack
- Department of Anesthesiology, University Hospital of Wuerzburg, Wuerzburg, Germany
| | - Robert Blum
- Institute of Clinical Neurobiology, University Hospital of Wuerzburg, Wuerzburg, Germany
| | - Heike L Rittner
- Department of Anesthesiology, University Hospital of Wuerzburg, Wuerzburg, Germany.
| |
Collapse
|
33
|
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
|
34
|
Ji H, Li Y, Fan Z, Zuo B, Jian X, Li L, Liu T. Monocyte/lymphocyte ratio predicts the severity of coronary artery disease: a syntax score assessment. BMC Cardiovasc Disord 2017; 17:90. [PMID: 28359298 PMCID: PMC5374608 DOI: 10.1186/s12872-017-0507-4] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Accepted: 03/01/2017] [Indexed: 12/21/2022] Open
Abstract
Background We aimed to explore whether monocyte to lymphocyte ratio (MLR) provides predictive value of the lesion severity in patients with coronary artery disease (CAD). Methods Five hundred forty-three patients undergoing coronary angiography were analyzed in this retrospective study. Patients with coronary stenosis were divided into three groups on the basis of Syntax score. The control group consisted of patients with normal coronary arteries. MLR was calculated by dividing monocytes count by lymphocytes count obtained from routine blood examination. Multivariate logistic analysis was used to assess risk factors of CAD. Ordinal logistic regression analysis was used to assess the relationship between MLR and the lesion severity of coronary arteries. Results MLR was found to be an independent risk factor of the presence of CAD (OR: 3.94, 95% CI: 1.20–12.95) and a predictor of the lesion severity (OR: 2.05, 95% CI: 1.15–3.66). Besides, MLR was positively correlated with Syntax score(r = 0.437, p < 0.001). In the receiver-operating characteristic (ROC) curve analysis, MLR, with an optimal cut-off value of 0.25, predicted the severe coronary lesion with a sensitivity of 60.26% and specificity of 78.49%. Conclusions MLR was an independent risk factor of the presence of CAD, and a predictor of the lesion severity. Compared to neutrophil to lymphocyte ratio (NLR), MLR has better performance to reflect the severity of coronary lesion.
Collapse
Affiliation(s)
- Hanhua Ji
- Department of Cardiology, Civil Aviation General Hospital, Civil Aviation Clinical Medical College of Peking University, No.1, Gaojingjia, Chaoyang District, Beijing, 100100, China.
| | - Yang Li
- Department of Cardiology, Civil Aviation General Hospital, Civil Aviation Clinical Medical College of Peking University, No.1, Gaojingjia, Chaoyang District, Beijing, 100100, China
| | - Zeyuan Fan
- Department of Cardiology, Civil Aviation General Hospital, Civil Aviation Clinical Medical College of Peking University, No.1, Gaojingjia, Chaoyang District, Beijing, 100100, China
| | - Bo Zuo
- Department of Cardiology, Peking University Third Hospital, Beijing, 100100, China
| | - Xinwen Jian
- Department of Cardiology, Civil Aviation General Hospital, Civil Aviation Clinical Medical College of Peking University, No.1, Gaojingjia, Chaoyang District, Beijing, 100100, China
| | - Li Li
- Department of Cardiology, Civil Aviation General Hospital, Civil Aviation Clinical Medical College of Peking University, No.1, Gaojingjia, Chaoyang District, Beijing, 100100, China
| | - Tao Liu
- Department of Cardiology, Civil Aviation General Hospital, Civil Aviation Clinical Medical College of Peking University, No.1, Gaojingjia, Chaoyang District, Beijing, 100100, China
| |
Collapse
|
35
|
Kimura T, Tse K, McArdle S, Gerhardt T, Miller J, Mikulski Z, Sidney J, Sette A, Wolf D, Ley K. Atheroprotective vaccination with MHC-II-restricted ApoB peptides induces peritoneal IL-10-producing CD4 T cells. Am J Physiol Heart Circ Physiol 2017; 312:H781-H790. [PMID: 28087520 DOI: 10.1152/ajpheart.00798.2016] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 01/09/2017] [Accepted: 01/09/2017] [Indexed: 12/14/2022]
Abstract
Although immunization with major histocompatibility complex (MHC) class II-restricted apolipoprotein B (ApoB) peptides has been shown to be atheroprotective, the mechanism is unclear. Here, we investigated CD4+ T cell populations in immunized atherosclerotic mice. Peptides (16-mers) from mouse ApoB, the core protein of low-density lipoprotein (LDL), were screened for binding to I-Ab by computer prediction and confirmed by radiolabeled peptide competition. Three new peptides, P101 (FGKQGFFPDSVNKALY, 5.5 nM IC50), P102 (TLYALSHAVNSYFDVD, 6.8 nM), and P103 (LYYKEDKTSLSASAAS, 95 nM), were tested in an atherosclerosis model (Apoe-/- mice on Western diet). Immunization with each of the three peptides (1 time in complete Freund's adjuvant subcuntaneously and 4 time in incomplete Freund's adjuvant intraperitoneally) but not with adjuvant alone showed significantly reduced atherosclerotic plaques in the aortic root by serial sections and in the whole aorta by en face staining. There were no differences in body weight, LDL cholesterol, or triglycerides. Peritoneal leukocytes from ApoB peptide-immunized mice, but not control mice, secreted significant amounts of IL-10 (150 pg/ml). Flow cytometry showed that peptide immunization induced IL-10 in 10% of peritoneal CD4+ T cells, some of which also expressed chemokine (C-C motif) receptor 5 (CCR5). Vaccination with ApoB peptides expanded peritoneal FoxP3+ regulatory CD4+ T cells and more than tripled the number of CCR5+FoxP3+ cells. Similar trends were also seen in the draining mediastinal lymph nodes but not in the nondraining inguinal lymph nodes. We conclude that vaccination with MHC class II-restricted autologous ApoB peptides induces regulatory T cells (Tregs) and IL-10, suggesting a plausible mechanism for atheroprotection.NEW & NOTEWORTHY Vaccination against apolipoprotein B (ApoB), the protein of LDL, attracts attention as a novel approach to prevent atherosclerosis. We discovered major histocompatibility complex class II-restricted ApoB peptides, which reduce atherosclerosis and induce IL-10-producing CD4+ T cells and chemokine (C-C motif) receptor 5 expression on regulatory T cells, suggesting that immunization with ApoB peptides inhibits atherosclerosis by inducing anti-inflammatory cytokines.
Collapse
Affiliation(s)
- Takayuki Kimura
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California; and
| | - Kevin Tse
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California; and
| | - Sara McArdle
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California; and
| | - Teresa Gerhardt
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California; and
| | - Jacqueline Miller
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California; and
| | - Zbigniew Mikulski
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California; and
| | - John Sidney
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, California
| | - Alessandro Sette
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, California
| | - Dennis Wolf
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California; and
| | - Klaus Ley
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California; and
| |
Collapse
|
36
|
Fatkhullina AR, Peshkova IO, Koltsova EK. The Role of Cytokines in the Development of Atherosclerosis. BIOCHEMISTRY (MOSCOW) 2017; 81:1358-1370. [PMID: 27914461 DOI: 10.1134/s0006297916110134] [Citation(s) in RCA: 169] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Atherosclerosis contributes to the development of many cardiovascular diseases, which remain the leading cause of death in developed countries. Atherosclerosis is a chronic inflammatory disease of large and medium-sized arteries. It is caused by dyslipidemia and mediated by both innate and adaptive immune responses. Inflammation is a key factor at all stages of atherosclerosis progression. Cells involved in pathogenesis of atherosclerosis were shown to be activated by soluble factors, cytokines, that strongly influence the disease development. Pro-inflammatory cytokines accelerate atherosclerosis progression, while anti-inflammatory cytokines ameliorate the disease. In this review, we discuss the latest findings on the role of cytokines in the development and progression of atherosclerosis.
Collapse
|
37
|
Hosseini H, Li Y, Kanellakis P, Tay C, Cao A, Liu E, Peter K, Tipping P, Toh BH, Bobik A, Kyaw T. Toll-Like Receptor (TLR)4 and MyD88 are Essential for Atheroprotection by Peritoneal B1a B Cells. J Am Heart Assoc 2016; 5:e002947. [PMID: 27930350 PMCID: PMC5210362 DOI: 10.1161/jaha.115.002947] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Accepted: 10/05/2016] [Indexed: 01/16/2023]
Abstract
BACKGROUND We previously identified peritoneal B1a cells that secrete natural IgM as a key atheroprotective B cell subset. However, the molecules that activate atheroprotective B1a cells are unknown. Here, we investigated whether Toll-like receptors (TLRs) TLR2, TLR4, and TLR9 expressed by B1a cells are required for IgM-mediated atheroprotection. METHODS AND RESULTS We adoptively transferred B1a cells from wild-type mice or from mice deficient in TLR2, TLR4, TLR9, or myeloid differentiation primary response 88 (MyD88) into ApoE-/- mice depleted of peritoneal B1a cells by splenectomy and fed a high-fat diet for 8 weeks. Elevations in plasma total, anti-oxLDL (oxidized low-density lipoprotein), anti-leukocyte, anti-CD3, anti-CD8, and anti-CD4 IgMs in atherosclerotic mice required B1a cells expressing TLR4 and MyD88, indicating a critical role for TLR4-MyD88 signaling for IgM secretion. Suppression of atherosclerosis was also critically dependent on B1a cells expressing TLR4-MyD88. Atherosclerosis suppression was associated not only with reductions in lesion apoptotic cells, necrotic cores, and oxLDL, but also with reduced lesion CD4+ and CD8+ T cells. Transforming growth factor beta 1 (TGF-β1) expression, including macrophages expressing TGF-β1, was increased, consistent with increased IgM-mediated phagocytosis of apoptotic cells by macrophages. Reductions in lesion inflammatory cytokines tumor necrosis factor alpha (TNF-α), interleukin (IL) 1β, and IL-18 were consistent with augmented TGF-β1 expression. CONCLUSIONS TLR4-MyD88 expression on B1a cells is critical for their IgM-dependent atheroprotection that not only reduced lesion apoptotic cells and necrotic cores, but also decreased CD4 and CD8 T-cell infiltrates and augmented TGF-β1 expression accompanied by reduced lesion inflammatory cytokines TNF-α, IL-1β, and IL-18.
Collapse
Affiliation(s)
- Hamid Hosseini
- BakerIDI heart and Diabetes Institute, Melbourne, Australia
- Department of Medicine, Centre for Inflammatory Diseases, Southern Clinical School, Clayton, Australia
| | - Yi Li
- BakerIDI heart and Diabetes Institute, Melbourne, Australia
- Department of Medicine, Centre for Inflammatory Diseases, Southern Clinical School, Clayton, Australia
| | | | - Christopher Tay
- BakerIDI heart and Diabetes Institute, Melbourne, Australia
- Department of Medicine, Centre for Inflammatory Diseases, Southern Clinical School, Clayton, Australia
| | - Anh Cao
- BakerIDI heart and Diabetes Institute, Melbourne, Australia
| | - Edgar Liu
- BakerIDI heart and Diabetes Institute, Melbourne, Australia
| | - Karlheinz Peter
- BakerIDI heart and Diabetes Institute, Melbourne, Australia
- Department of Immunology, Faculty of Medicine, Nursing and Health Sciences Monash University, Clayton, Australia
| | - Peter Tipping
- Department of Medicine, Centre for Inflammatory Diseases, Southern Clinical School, Clayton, Australia
| | - Ban-Hock Toh
- Department of Medicine, Centre for Inflammatory Diseases, Southern Clinical School, Clayton, Australia
| | - Alex Bobik
- BakerIDI heart and Diabetes Institute, Melbourne, Australia
- Department of Immunology, Faculty of Medicine, Nursing and Health Sciences Monash University, Clayton, Australia
| | - Tin Kyaw
- BakerIDI heart and Diabetes Institute, Melbourne, Australia
- Department of Medicine, Centre for Inflammatory Diseases, Southern Clinical School, Clayton, Australia
| |
Collapse
|
38
|
Bernelot Moens SJ, van Leuven SI, Zheng KH, Havik SR, Versloot MV, van Duivenvoorde LM, Hahne M, Stroes ESG, Baeten DL, Hamers AAJ. Impact of the B Cell Growth Factor APRIL on the Qualitative and Immunological Characteristics of Atherosclerotic Plaques. PLoS One 2016; 11:e0164690. [PMID: 27820817 PMCID: PMC5098816 DOI: 10.1371/journal.pone.0164690] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 09/29/2016] [Indexed: 11/18/2022] Open
Abstract
Studies on the role of B lymphocytes in atherosclerosis development, have yielded contradictory results. Whereas B lymphocyte-deficiency aggravates atherosclerosis in mice; depletion of mature B lymphocytes reduces atherosclerosis. These observations led to the notion that distinct B lymphocyte subsets have different roles. B1a lymphocytes exert an atheroprotective effect, which has been attributed to secretion of IgM, which can be deposited in atherosclerotic lesions thereby reducing necrotic core formation. Tumor necrosis factor (TNF)-family member 'A Proliferation-Inducing Ligand' (APRIL, also known as TNFSF13) was previously shown to increase serum IgM levels in a murine model. In this study, we investigated the effect of APRIL overexpression on advanced lesion formation and composition, IgM production and B cell phenotype. We crossed APRIL transgenic (APRIL-Tg) mice with ApoE knockout (ApoE-/-) mice. After a 12-week Western Type Diet, ApoE-/-APRIL-Tg mice and ApoE-/- littermates showed similar increases in body weight and lipid levels. Histologic evaluation showed no differences in lesion size, stage or necrotic area. However, smooth muscle cell (α-actin stain) content was increased in ApoE-/-APRIL-Tg mice, implying more stable lesions. In addition, increases in both plaque IgM deposition and plasma IgM levels were found in ApoE-/-APRIL-Tg mice compared with ApoE-/- mice. Flow cytometry revealed a concomitant increase in peritoneal B1a lymphocytes in ApoE-/-APRIL-Tg mice. This study shows that ApoE-/-APRIL-Tg mice have increased oxLDL-specific serum IgM levels, potentially mediated via an increase in B1a lymphocytes. Although no differences in lesion size were found, transgenic ApoE-/-APRIL-Tg mice do show potential plaque stabilizing features in advanced atherosclerotic lesions.
Collapse
Affiliation(s)
| | - Sander I. van Leuven
- Amsterdam Rheumatology and Immunology Center, Department of Clinical Immunology and Rheumatology, Academic Medical Center, Amsterdam, The Netherlands
| | - Kang H. Zheng
- Department of Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Stefan R. Havik
- Department of Experimental Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Miranda V. Versloot
- Department of Experimental Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Leonie M. van Duivenvoorde
- Amsterdam Rheumatology and Immunology Center, Department of Clinical Immunology and Rheumatology, Academic Medical Center, Amsterdam, The Netherlands
- Department of Experimental Immunology, Academic Medical Center, Amsterdam, The Netherlands
| | - Michael Hahne
- Institut de Génétique Moléculaire de Montpellier, Centre National de la Recherche Scientifique, Université de Montpellier, Montpellier, France
| | - Erik S. G. Stroes
- Department of Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Dominique L. Baeten
- Amsterdam Rheumatology and Immunology Center, Department of Clinical Immunology and Rheumatology, Academic Medical Center, Amsterdam, The Netherlands
- Department of Experimental Immunology, Academic Medical Center, Amsterdam, The Netherlands
| | - Anouk A. J. Hamers
- Department of Experimental Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
39
|
Yin C, Mohanta SK, Srikakulapu P, Weber C, Habenicht AJR. Artery Tertiary Lymphoid Organs: Powerhouses of Atherosclerosis Immunity. Front Immunol 2016; 7:387. [PMID: 27777573 PMCID: PMC5056324 DOI: 10.3389/fimmu.2016.00387] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 09/14/2016] [Indexed: 11/15/2022] Open
Abstract
Artery tertiary lymphoid organs (ATLOs) are atherosclerosis-associated lymphoid aggregates with varying degrees of complexity ranging from small T/B-cell clusters to well-structured lymph node-like though unencapsulated lymphoid tissues. ATLOs arise in the connective tissue that surrounds diseased arteries, i.e., the adventitia. ATLOs have been identified in aged atherosclerosis-prone hyperlipidemic apolipoprotein E-deficient (ApoE-/-) mice: they are organized into distinct immune cell compartments, including separate T-cell areas, activated B-cell follicles, and plasma cell niches. Analyses of ATLO immune cell subsets indicate antigen-specific T- and B-cell immune reactions within the atherosclerotic arterial wall adventitia. Moreover, ATLOs harbor innate immune cells, including a large component of inflammatory macrophages, B-1 cells, and an aberrant set of antigen-presenting cells. There is marked neoangiogenesis, irregular lymphangiogenesis, neoformation of high endothelial venules, and de novo synthesis of lymph node-like conduits. Molecular mechanisms of ATLO formation remain to be identified though media vascular smooth muscle cells may adopt features of lymphoid tissue organizer-like cells by expressing lymphorganogenic chemokines, i.e., CXCL13 and CCL21. Although these data are consistent with the view that ATLOs participate in primary T- and B-cell responses against elusive atherosclerosis-specific autoantigens, their specific protective or disease-promoting roles remain to be identified. In this review, we discuss what is currently known about ATLOs and their potential impact on atherosclerosis and make attempts to define challenges ahead.
Collapse
Affiliation(s)
- Changjun Yin
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Sarajo Kumar Mohanta
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany
| | - Prasad Srikakulapu
- Cardiovascular Research Center (CVRC), University of Virginia, Charlottesville, VA, USA
| | - Christian Weber
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | | |
Collapse
|
40
|
Quách TD, Hopkins TJ, Holodick NE, Vuyyuru R, Manser T, Bayer RL, Rothstein TL. Human B-1 and B-2 B Cells Develop from Lin-CD34+CD38lo Stem Cells. THE JOURNAL OF IMMUNOLOGY 2016; 197:3950-3958. [PMID: 27815443 DOI: 10.4049/jimmunol.1600630] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 09/14/2016] [Indexed: 12/24/2022]
Abstract
The B-1 B cell population is an important bridge between innate and adaptive immunity primarily because B-1 cells produce natural Ab. Murine B-1 and B-2 cells arise from distinct progenitors; however, in humans, in part because it has been difficult to discriminate between them phenotypically, efforts to pinpoint the developmental origins of human B-1 and B-2 cells have lagged. To characterize progenitors of human B-1 and B-2 cells, we separated cord blood and bone marrow Lin-CD34+ hematopoietic stem cells into Lin-CD34+CD38lo and Lin-CD34+CD38hi populations. We found that transplanted Lin-CD34+CD38lo cells, but not Lin-CD34+CD38hi cells, generated a CD19+ B cell population after transfer into immunodeficient NOD.Cg-Prkdcscid Il2rgtm1wjl/SxJ neonates. The emergent CD19+ B cell population was found in spleen, bone marrow, and peritoneal cavity of humanized mice and included distinct populations displaying the B-1 or the B-2 cell phenotype. Engrafted splenic B-1 cells exhibited a mature phenotype, as evidenced by low-to-intermediate expression levels of CD24 and CD38. The engrafted B-1 cell population expressed a VH-DH-JH composition similar to cord blood B-1 cells, including frequent use of VH4-34 (8 versus 10%, respectively). Among patients with hematologic malignancies who underwent hematopoietic stem cell transplantation, B-1 cells were found in the circulation as early as 8 wk posttransplantation. Altogether, our data demonstrate that human B-1 and B-2 cells develop from a Lin-CD34+CD38lo stem cell population, and engrafted B-1 cells in humanized mice exhibit an Ig-usage pattern comparable to B-1 cells in cord blood.
Collapse
Affiliation(s)
- Tâm D Quách
- Center for Oncology and Cell Biology, The Feinstein Institute for Medical Research, Manhasset, NY 11030
| | - Thomas J Hopkins
- Center for Oncology and Cell Biology, The Feinstein Institute for Medical Research, Manhasset, NY 11030
| | - Nichol E Holodick
- Center for Oncology and Cell Biology, The Feinstein Institute for Medical Research, Manhasset, NY 11030
| | - Raja Vuyyuru
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA 19107
| | - Tim Manser
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA 19107
| | - Ruthee-Lu Bayer
- Monter Cancer Center, North Shore University Hospital, Northwell Health, Lake Success, NY 11042; and
| | - Thomas L Rothstein
- Center for Oncology and Cell Biology, The Feinstein Institute for Medical Research, Manhasset, NY 11030; .,Hofstra-Northwell Health School of Medicine, Hempstead, NY 11549
| |
Collapse
|
41
|
Kuneš P, Holubcová Z, Koláčková M, Krejsek J. The Counter-Regulation of Atherogenesis: a Role for Interleukin-33. ACTA MEDICA (HRADEC KRÁLOVÉ) 2016. [DOI: 10.14712/18059694.2016.71] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The recently recognized cytokine interleukin-33 and its receptor ST2 play a favorable role during atherogenesis by inducing a Th1→Th2 shift of the immune response. IL-33 also protects the failing human heart from harmful biomechanical forces which lead to cardiomyocyte hypertrophy and exaggerated interstitial fibrosis. IL-33 inevitably displays side effects common to other Th2 cytokines, the most grave of which is a predisposition to allergic reactions. IL-33 is a nuclear transcription factor of endothelial cells. As such, it is abundant in nonproliferating vessels. Its down-regulation is required for angiogenesis, which may be profitable in wound healing or deleterious in tumor growth.
Collapse
|
42
|
Khasbiullina NR, Bovin NV. Hypotheses of the origin of natural antibodies: a glycobiologist's opinion. BIOCHEMISTRY (MOSCOW) 2016; 80:820-35. [PMID: 26541997 DOI: 10.1134/s0006297915070032] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
It is generally accepted that the generation of antibodies proceeds due to immunization of an organism by alien antigens, and the level and affinity of antibodies are directly correlated to the presence of immunogen. At the same time, vast experimental material has been obtained providing evidence of antibodies whose level remains unchanged and affinity is constant during a lifetime. In contrast to the first, adaptive immunoglobulins, the latter are named natural antibodies (nAbs). The nAbs are produced by B1 cells, whereas adaptive Abs are produced by B2. This review summarizes general data on nAbs and presents in more detail data on antigens of carbohydrate origin. Hypotheses on the origin of nAbs and their activation mechanisms are discussed. We present our thoughts on this matter supported by our experimental data on nAbs to glycans.
Collapse
Affiliation(s)
- N R Khasbiullina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia.
| | | |
Collapse
|
43
|
Nilsson J. Can Antibodies Protect Us Against Cardiovascular Disease? EBioMedicine 2016; 9:29-30. [PMID: 27381475 PMCID: PMC4972560 DOI: 10.1016/j.ebiom.2016.06.039] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 06/27/2016] [Indexed: 12/02/2022] Open
Affiliation(s)
- Jan Nilsson
- Department of Clinical Sciences, Jan Waldenströms gata 35, 20502 Malmö, Sweden.
| |
Collapse
|
44
|
Holodick NE, Vizconde T, Hopkins TJ, Rothstein TL. Age-Related Decline in Natural IgM Function: Diversification and Selection of the B-1a Cell Pool with Age. THE JOURNAL OF IMMUNOLOGY 2016; 196:4348-57. [PMID: 27183643 DOI: 10.4049/jimmunol.1600073] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 03/18/2016] [Indexed: 01/21/2023]
Abstract
Streptococcus pneumoniae is the most common cause of pneumonia, which claims the lives of people over the age of 65 y seven times more frequently than those aged 5-49 y. B-1a cells provide immediate and essential protection from S. pneumoniae through production of natural Ig, which has minimal insertion of N-region additions added by the enzyme TdT. In experiments with SCID mice infected with S. pneumoniae, we found passive transfer of IgG-depleted serum from aged (18-24 mo old) mice had no effect whereas IgG-depleted serum from young (3 mo old) mice was protective. This suggests protective natural IgM changes with age. Using single cell PCR we found N-region addition, which is initially low in fetal-derived B-1a cell IgM developing in the absence of TdT, increased in 7- to 24-mo-old mice as compared with 3-mo-old mice. To determine the mechanism responsible for the age related change in B-1a cell IgM, we established a mixed chimera system in which mice were reconstituted with allotype-marked mature peritoneal B-1a cells and adult bone marrow cells. We demonstrated even in the presence of mature peritoneal B-1a cells, adult bone marrow contributed to the mature B-1a cell pool. More importantly, using this system we found over a 10-mo-period peritoneal B-1a cell IgM changed, showing the number of cells lacking N-region additions at both junctions fell from 49 to 29% of sequences. These results strongly suggest selection-induced skewing alters B-1a cell-derived natural Ab, which may in turn be responsible for the loss of natural IgM-mediated protection against pneumococcal infection.
Collapse
Affiliation(s)
- Nichol E Holodick
- Center for Oncology and Cell Biology, Feinstein Institute for Medical Research, Hofstra Northwell School of Medicine, Manhasset, NY 11030;
| | - Teresa Vizconde
- Center for Oncology and Cell Biology, Feinstein Institute for Medical Research, Hofstra Northwell School of Medicine, Manhasset, NY 11030
| | - Thomas J Hopkins
- Center for Oncology and Cell Biology, Feinstein Institute for Medical Research, Hofstra Northwell School of Medicine, Manhasset, NY 11030
| | - Thomas L Rothstein
- Center for Oncology and Cell Biology, Feinstein Institute for Medical Research, Hofstra Northwell School of Medicine, Manhasset, NY 11030; Department of Medicine, Hofstra Northwell School of Medicine, Manhasset, NY 11030; and Department of Molecular Medicine, Hofstra Northwell School of Medicine, Manhasset, NY 11030
| |
Collapse
|
45
|
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
|
46
|
Foks AC, Engelbertsen D, Kuperwaser F, Alberts-Grill N, Gonen A, Witztum JL, Lederer J, Jarolim P, DeKruyff RH, Freeman GJ, Lichtman AH. Blockade of Tim-1 and Tim-4 Enhances Atherosclerosis in Low-Density Lipoprotein Receptor-Deficient Mice. Arterioscler Thromb Vasc Biol 2016; 36:456-65. [PMID: 26821944 PMCID: PMC4853762 DOI: 10.1161/atvbaha.115.306860] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 01/14/2016] [Indexed: 11/16/2022]
Abstract
OBJECTIVE T cell immunoglobulin and mucin domain (Tim) proteins are expressed by numerous immune cells, recognize phosphatidylserine on apoptotic cells, and function as costimulators or coinhibitors. Tim-1 is expressed by activated T cells but is also found on dendritic cells and B cells. Tim-4, present on macrophages and dendritic cells, plays a critical role in apoptotic cell clearance, regulates the number of phosphatidylserine-expressing activated T cells, and is genetically associated with low low-density lipoprotein and triglyceride levels. Because these functions of Tim-1 and Tim-4 could affect atherosclerosis, their modulation has potential therapeutic value in cardiovascular disease. APPROACH AND RESULTS ldlr(-/-) mice were fed a high-fat diet for 4 weeks while being treated with control (rat immunoglobulin G1) or anti-Tim-1 (3D10) or -Tim-4 (21H12) monoclonal antibodies that block phosphatidylserine recognition and phagocytosis. Both anti-Tim-1 and anti-Tim-4 treatments enhanced atherosclerosis by 45% compared with controls by impairment of efferocytosis and increasing aortic CD4(+)T cells. Consistently, anti-Tim-4-treated mice showed increased percentages of activated T cells and late apoptotic cells in the circulation. Moreover, in vitro blockade of Tim-4 inhibited efferocytosis of oxidized low-density lipoprotein-induced apoptotic macrophages. Although anti-Tim-4 treatment increased T helper cell (Th)1 and Th2 responses, anti-Tim-1 induced Th2 responses but dramatically reduced the percentage of regulatory T cells. Finally, combined blockade of Tim-1 and Tim-4 increased atherosclerotic lesion size by 59%. CONCLUSIONS Blockade of Tim-4 aggravates atherosclerosis likely by prevention of phagocytosis of phosphatidylserine-expressing apoptotic cells and activated T cells by Tim-4-expressing cells, whereas Tim-1-associated effects on atherosclerosis are related to changes in Th1/Th2 balance and reduced circulating regulatory T cells.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/toxicity
- Aortic Diseases/chemically induced
- Aortic Diseases/genetics
- Aortic Diseases/immunology
- Aortic Diseases/metabolism
- Aortic Diseases/pathology
- Apoptosis/drug effects
- Atherosclerosis/chemically induced
- Atherosclerosis/genetics
- Atherosclerosis/immunology
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- CD4-Positive T-Lymphocytes/drug effects
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/pathology
- Cell Proliferation/drug effects
- Cells, Cultured
- Coculture Techniques
- Diet, High-Fat
- Disease Models, Animal
- Female
- Hepatitis A Virus Cellular Receptor 1
- Lipoproteins, LDL/metabolism
- Lymphocyte Activation/drug effects
- Macrophages/drug effects
- Macrophages/immunology
- Macrophages/pathology
- Male
- Membrane Proteins/antagonists & inhibitors
- Membrane Proteins/immunology
- Membrane Proteins/metabolism
- Mice, Knockout
- Phagocytosis/drug effects
- Plaque, Atherosclerotic
- Receptors, LDL/deficiency
- Receptors, LDL/genetics
- Signal Transduction/drug effects
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- Th1 Cells/drug effects
- Th1 Cells/immunology
- Th2 Cells/drug effects
- Th2 Cells/immunology
Collapse
Affiliation(s)
- Amanda C Foks
- From the Department of Pathology (A.C.F., D.E., F.K., N.A.-G., P.J., A.H.L.) and Department of Surgery (J.L.), Harvard Medical School, Brigham and Women's Hospital, Boston, MA; Department of Medicine, University of California, San Diego, La Jolla (A.G., J.L.W.); Department of Medicine, Stanford University, Stanford, CA (R.H.D.); and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA (G.J.F.)
| | - Daniel Engelbertsen
- From the Department of Pathology (A.C.F., D.E., F.K., N.A.-G., P.J., A.H.L.) and Department of Surgery (J.L.), Harvard Medical School, Brigham and Women's Hospital, Boston, MA; Department of Medicine, University of California, San Diego, La Jolla (A.G., J.L.W.); Department of Medicine, Stanford University, Stanford, CA (R.H.D.); and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA (G.J.F.)
| | - Felicia Kuperwaser
- From the Department of Pathology (A.C.F., D.E., F.K., N.A.-G., P.J., A.H.L.) and Department of Surgery (J.L.), Harvard Medical School, Brigham and Women's Hospital, Boston, MA; Department of Medicine, University of California, San Diego, La Jolla (A.G., J.L.W.); Department of Medicine, Stanford University, Stanford, CA (R.H.D.); and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA (G.J.F.)
| | - Noah Alberts-Grill
- From the Department of Pathology (A.C.F., D.E., F.K., N.A.-G., P.J., A.H.L.) and Department of Surgery (J.L.), Harvard Medical School, Brigham and Women's Hospital, Boston, MA; Department of Medicine, University of California, San Diego, La Jolla (A.G., J.L.W.); Department of Medicine, Stanford University, Stanford, CA (R.H.D.); and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA (G.J.F.)
| | - Ayelet Gonen
- From the Department of Pathology (A.C.F., D.E., F.K., N.A.-G., P.J., A.H.L.) and Department of Surgery (J.L.), Harvard Medical School, Brigham and Women's Hospital, Boston, MA; Department of Medicine, University of California, San Diego, La Jolla (A.G., J.L.W.); Department of Medicine, Stanford University, Stanford, CA (R.H.D.); and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA (G.J.F.)
| | - Joseph L Witztum
- From the Department of Pathology (A.C.F., D.E., F.K., N.A.-G., P.J., A.H.L.) and Department of Surgery (J.L.), Harvard Medical School, Brigham and Women's Hospital, Boston, MA; Department of Medicine, University of California, San Diego, La Jolla (A.G., J.L.W.); Department of Medicine, Stanford University, Stanford, CA (R.H.D.); and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA (G.J.F.)
| | - James Lederer
- From the Department of Pathology (A.C.F., D.E., F.K., N.A.-G., P.J., A.H.L.) and Department of Surgery (J.L.), Harvard Medical School, Brigham and Women's Hospital, Boston, MA; Department of Medicine, University of California, San Diego, La Jolla (A.G., J.L.W.); Department of Medicine, Stanford University, Stanford, CA (R.H.D.); and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA (G.J.F.)
| | - Petr Jarolim
- From the Department of Pathology (A.C.F., D.E., F.K., N.A.-G., P.J., A.H.L.) and Department of Surgery (J.L.), Harvard Medical School, Brigham and Women's Hospital, Boston, MA; Department of Medicine, University of California, San Diego, La Jolla (A.G., J.L.W.); Department of Medicine, Stanford University, Stanford, CA (R.H.D.); and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA (G.J.F.)
| | - Rosemarie H DeKruyff
- From the Department of Pathology (A.C.F., D.E., F.K., N.A.-G., P.J., A.H.L.) and Department of Surgery (J.L.), Harvard Medical School, Brigham and Women's Hospital, Boston, MA; Department of Medicine, University of California, San Diego, La Jolla (A.G., J.L.W.); Department of Medicine, Stanford University, Stanford, CA (R.H.D.); and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA (G.J.F.)
| | - Gordon J Freeman
- From the Department of Pathology (A.C.F., D.E., F.K., N.A.-G., P.J., A.H.L.) and Department of Surgery (J.L.), Harvard Medical School, Brigham and Women's Hospital, Boston, MA; Department of Medicine, University of California, San Diego, La Jolla (A.G., J.L.W.); Department of Medicine, Stanford University, Stanford, CA (R.H.D.); and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA (G.J.F.)
| | - Andrew H Lichtman
- From the Department of Pathology (A.C.F., D.E., F.K., N.A.-G., P.J., A.H.L.) and Department of Surgery (J.L.), Harvard Medical School, Brigham and Women's Hospital, Boston, MA; Department of Medicine, University of California, San Diego, La Jolla (A.G., J.L.W.); Department of Medicine, Stanford University, Stanford, CA (R.H.D.); and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA (G.J.F.).
| |
Collapse
|
47
|
Wu J, Li L. Autoantibodies in Alzheimer's disease: potential biomarkers, pathogenic roles, and therapeutic implications. J Biomed Res 2016; 30:361-372. [PMID: 27476881 PMCID: PMC5044708 DOI: 10.7555/jbr.30.20150131] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 10/26/2015] [Indexed: 11/29/2022] Open
Abstract
Alzheimer’s disease (AD) is a prevalent and debilitating neurodegenerative disorder in the elderly. The etiology of AD has not been fully defined and currently there is no cure for this devastating disease. Compelling evidence suggests that the immune system plays a critical role in the pathophysiology of AD. Autoantibodies against a variety of molecules have been associated with AD. The roles of these autoantibodies in AD, however, are not well understood. This review attempts to summarize recent findings on these autoantibodies and explore their potential as diagnostic/ prognostic biomarkers for AD, their roles in the pathogenesis of AD, and their implications in the development of effective immunotherapies for AD.
Collapse
Affiliation(s)
- Jianming Wu
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Saint Paul, MN 55108, USA;
| | - Ling Li
- Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, MN 55455, USA;
| |
Collapse
|
48
|
Grimm M, Tischner D, Troidl K, Albarrán Juárez J, Sivaraj KK, Ferreirós Bouzas N, Geisslinger G, Binder CJ, Wettschureck N. S1P2/G12/13 Signaling Negatively Regulates Macrophage Activation and Indirectly Shapes the Atheroprotective B1-Cell Population. Arterioscler Thromb Vasc Biol 2015; 36:37-48. [PMID: 26603156 DOI: 10.1161/atvbaha.115.306066] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 11/11/2015] [Indexed: 12/31/2022]
Abstract
OBJECTIVES Monocyte/macrophage recruitment and activation at vascular predilection sites plays a central role in the pathogenesis of atherosclerosis. Heterotrimeric G proteins of the G12/13 family have been implicated in the control of migration and inflammatory gene expression, but their function in myeloid cells, especially during atherogenesis, is unknown. APPROACH AND RESULTS Mice with myeloid-specific deficiency for G12/13 show reduced atherosclerosis with a clear shift to anti-inflammatory gene expression in aortal macrophages. These changes are because of neither altered monocyte/macrophage migration nor reduced activation of inflammatory gene expression; on the contrary, G12/13-deficient macrophages show an increased nuclear factor-κB-dependent gene expression in the resting state. Chronically increased inflammatory gene expression in resident peritoneal macrophages results in myeloid-specific G12/13-deficient mice in an altered peritoneal micromilieu with secondary expansion of peritoneal B1 cells. Titers of B1-derived atheroprotective antibodies are increased, and adoptive transfer of peritoneal cells from mutant mice conveys atheroprotection to wild-type mice. With respect to the mechanism of G12/13-mediated transcriptional control, we identify an autocrine feedback loop that suppresses nuclear factor-κB-dependent gene expression through a signaling cascade involving sphingosine 1-phosphate receptor subtype 2, G12/13, and RhoA. CONCLUSIONS Together, these data show that selective inhibition of G12/13 signaling in macrophages can augment atheroprotective B-cell populations and ameliorate atherosclerosis.
Collapse
Affiliation(s)
- Myriam Grimm
- From the Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (M.G., D.T., K.T., J.A.J., K.K.S., N.W.); Pharmazentrum Frankfurt/ZAFES, Clinical Pharmacology (N.F.B., G.G.) and Centre for Molecular Medicine, Medical Faculty (N.W.), J.W. Goethe University Frankfurt, Frankfurt, Germany; and Department of Laboratory Medicine, Medical University of Vienna and Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria (C.J.B.)
| | - Denise Tischner
- From the Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (M.G., D.T., K.T., J.A.J., K.K.S., N.W.); Pharmazentrum Frankfurt/ZAFES, Clinical Pharmacology (N.F.B., G.G.) and Centre for Molecular Medicine, Medical Faculty (N.W.), J.W. Goethe University Frankfurt, Frankfurt, Germany; and Department of Laboratory Medicine, Medical University of Vienna and Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria (C.J.B.)
| | - Kerstin Troidl
- From the Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (M.G., D.T., K.T., J.A.J., K.K.S., N.W.); Pharmazentrum Frankfurt/ZAFES, Clinical Pharmacology (N.F.B., G.G.) and Centre for Molecular Medicine, Medical Faculty (N.W.), J.W. Goethe University Frankfurt, Frankfurt, Germany; and Department of Laboratory Medicine, Medical University of Vienna and Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria (C.J.B.)
| | - Julián Albarrán Juárez
- From the Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (M.G., D.T., K.T., J.A.J., K.K.S., N.W.); Pharmazentrum Frankfurt/ZAFES, Clinical Pharmacology (N.F.B., G.G.) and Centre for Molecular Medicine, Medical Faculty (N.W.), J.W. Goethe University Frankfurt, Frankfurt, Germany; and Department of Laboratory Medicine, Medical University of Vienna and Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria (C.J.B.)
| | - Kishor K Sivaraj
- From the Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (M.G., D.T., K.T., J.A.J., K.K.S., N.W.); Pharmazentrum Frankfurt/ZAFES, Clinical Pharmacology (N.F.B., G.G.) and Centre for Molecular Medicine, Medical Faculty (N.W.), J.W. Goethe University Frankfurt, Frankfurt, Germany; and Department of Laboratory Medicine, Medical University of Vienna and Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria (C.J.B.)
| | - Nerea Ferreirós Bouzas
- From the Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (M.G., D.T., K.T., J.A.J., K.K.S., N.W.); Pharmazentrum Frankfurt/ZAFES, Clinical Pharmacology (N.F.B., G.G.) and Centre for Molecular Medicine, Medical Faculty (N.W.), J.W. Goethe University Frankfurt, Frankfurt, Germany; and Department of Laboratory Medicine, Medical University of Vienna and Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria (C.J.B.)
| | - Gerd Geisslinger
- From the Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (M.G., D.T., K.T., J.A.J., K.K.S., N.W.); Pharmazentrum Frankfurt/ZAFES, Clinical Pharmacology (N.F.B., G.G.) and Centre for Molecular Medicine, Medical Faculty (N.W.), J.W. Goethe University Frankfurt, Frankfurt, Germany; and Department of Laboratory Medicine, Medical University of Vienna and Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria (C.J.B.)
| | - Christoph J Binder
- From the Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (M.G., D.T., K.T., J.A.J., K.K.S., N.W.); Pharmazentrum Frankfurt/ZAFES, Clinical Pharmacology (N.F.B., G.G.) and Centre for Molecular Medicine, Medical Faculty (N.W.), J.W. Goethe University Frankfurt, Frankfurt, Germany; and Department of Laboratory Medicine, Medical University of Vienna and Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria (C.J.B.)
| | - Nina Wettschureck
- From the Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (M.G., D.T., K.T., J.A.J., K.K.S., N.W.); Pharmazentrum Frankfurt/ZAFES, Clinical Pharmacology (N.F.B., G.G.) and Centre for Molecular Medicine, Medical Faculty (N.W.), J.W. Goethe University Frankfurt, Frankfurt, Germany; and Department of Laboratory Medicine, Medical University of Vienna and Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria (C.J.B.).
| |
Collapse
|
49
|
Toller-Kawahisa JE, Canicoba NC, Venancio VP, Kawahisa R, Antunes LMG, Cunha TM, Marzocchi-Machado CM. Systemic lupus erythematosus onset in lupus-prone B6.MRL/lpr mice Is influenced by weight gain and Is preceded by an increase in neutrophil oxidative burst activity. Free Radic Biol Med 2015; 86:362-73. [PMID: 26117329 DOI: 10.1016/j.freeradbiomed.2015.06.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 06/08/2015] [Accepted: 06/11/2015] [Indexed: 12/23/2022]
Abstract
In this study, we assessed whether weight gain influenced the systemic lupus erythematosus (SLE) onset and/or outcome, and examined the role that reactive oxygen species (ROS) production by neutrophils played in the SLE onset and/or outcome. Female control (C57BL/6) and lupus-prone B6.MRL/lpr mice (CM and LPM, respectively) at 4 weeks old were fed standard diet or standard diet plus cafeteria diet during 12 weeks. SLE diagnosis relied on the presence of both antinuclear antibodies (ANA) and renal abnormalities. We found that the percentage of weight gain in CM and LPM increased as a function of the length of cafeteria diet feeding period, but it was not associated with energy intake. Cafeteria diet-fed CM and LPM at 8 and 12 weeks old were overweight, while CM and LPM at 16 weeks old were obese. Compared with standard diet-fed CM and LPM, cafeteria diet-fed CM and LPM exhibited elevated glucose and total cholesterol levels, and diminished triglycerides levels. Standard diet-fed 16-week-old LPM and cafeteria diet-fed 12-week-old LPM had nephritis, characterized by the increased interstitial infiltration of leukocytes. Cafeteria diet-induced weight gain rose the frequency of homogeneous and speckled ANA staining patterns in the 12- and 16-week-old LPM groups. Together, these results indicated that weight gain anticipated the SLE onset. In addition, neutrophils from cafeteria diet-fed 8-week-old LPM exhibited augmented ROS production capacity; in standard diet-fed LPM, such rise occurred only in the 16-week-old group. Thus, the neutrophil ROS production capacity was increased before the SLE onset and during its outcome. Overweight and obese CM and LPM displayed elevated levels of kidney, liver, heart, and spleen lipid peroxidation. In conclusion, cafeteria diet-induced weight gain is associated with the increased production of ANA and neutrophil-derived ROS, which may contribute to accelerate the SLE onset.
Collapse
Affiliation(s)
- Juliana Escher Toller-Kawahisa
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Avenida Bandeirantes 3900, Ribeirão Preto, SP 14049-900, Brazil
| | - Nathália Cristina Canicoba
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Avenida do Café s/n, Ribeirão Preto, SP 14040-903, Brazil
| | - Vinicius Paula Venancio
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Avenida do Café s/n, Ribeirão Preto, SP 14040-903, Brazil
| | - Rogério Kawahisa
- Graduação em Fisioterapia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Avenida Bandeirantes 3900, Ribeirão Preto, SP 14049-900, Brazil
| | - Lusânia Maria Greggi Antunes
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Avenida do Café s/n, Ribeirão Preto, SP 14040-903, Brazil
| | - Thiago Mattar Cunha
- Departamento de Farmacologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Avenida Bandeirantes 3900, Ribeirão Preto, SP 14049-900, Brazil
| | - Cleni Mara Marzocchi-Machado
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Avenida do Café s/n, Ribeirão Preto, SP 14040-903, Brazil.
| |
Collapse
|
50
|
Gray M, Gray D. Regulatory B cells mediate tolerance to apoptotic self in health: implications for disease. Int Immunol 2015; 27:505-11. [PMID: 26306497 DOI: 10.1093/intimm/dxv045] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 07/27/2015] [Indexed: 12/17/2022] Open
Abstract
B cells are able to regulate immune responses through the secretion of IL-10 and other inhibitory cytokines, though no transcription factor that can define 'regulatory B cells' as a separate lineage has yet been found. Instead it is likely that this function arises as a result of the immune context in which B cells find themselves and the stimuli they perceive. However, some B cells found within the B1a and the marginal zone subsets have a greater propensity to produce IL-10 than others. What are the natural stimuli for these cells to induce immune regulation? We discuss the role that the recognition of autoantigens exposed by apoptotic cells plays in stimulating IL-10 production in mouse and human studies. This mechanism involves the recognition and uptake of self-antigens by autoreactive BCRs, for delivery to endocytic compartments, where apoptosis-derived DNA binds to TLR9, driving IL-10 production. These 'natural' regulatory B cells represent a way of maintaining tolerance to self. We discuss how this may operate in inflammatory lesions where there is an excess of apoptotic leukocytes and how this impacts on our understanding of autoimmune disease.
Collapse
Affiliation(s)
- Mohini Gray
- Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - David Gray
- Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Ashworth Laboratories, King's Buildings, West Mains Road, Edinburgh EH9 3JT, UK
| |
Collapse
|