1
|
Giacaglia MB, Pires V, Santana MFM, Passarelli M. Unraveling the Pleiotropic Role of High-Density Lipoproteins (HDLs) in Autoimmune Rheumatic Diseases. Int J Rheumatol 2024; 2024:1896817. [PMID: 39574464 PMCID: PMC11581784 DOI: 10.1155/2024/1896817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 10/23/2024] [Indexed: 11/24/2024] Open
Abstract
Autoimmune rheumatic diseases (ARDs) exhibit an elevated incidence of cardiovascular disease (CVD). The elevation of inflammatory and immune stress accompanying ARDs contributes to atherosclerosis development and alterations in lipid metabolism and lipoprotein profile add to cardiovascular (CV) risk. The plasma concentration of high-density lipoprotein cholesterol (HDLc) is inversely related to CVD and serves as a discriminator of CV risk. However, this association is not unequivocal, and changes in HDL functionality appear to emerge as a better indicator of CV risk, albeit difficult to measure and monitor clinically. The modulation of HDLc itself can bring benefits in controlling autoimmunity and reducing ARD activity. Understanding HDL function and each peculiarity involved in ARDs enables to seek means to prevent ischemic outcomes associated with CVD, in the face of the residual CV risk persisting even with controlled disease activity and classic risk factors. By comprehending HDL's structural and functional nuances, it will be possible to develop more effective strategies to manage the evolution and outcomes of ARDs. It is also necessary to standardize diagnostic methods and establish different markers for each specific disease allowing the design of intervention strategies to restore HDL functionality, reduce residual CV, and prevent, alleviate, or even suppress ARD activity.
Collapse
Affiliation(s)
- Marcia B. Giacaglia
- Programa de Pós-Graduação em Medicina, Universidade Nove de Julho (UNINOVE) 01525-000, São Paulo, Brazil
| | - Vitória Pires
- Laboratório de Lípides (LIM10), Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo 01246-000, São Paulo, Brazil
| | - Monique F. M. Santana
- Laboratório de Lípides (LIM10), Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo 01246-000, São Paulo, Brazil
| | - Marisa Passarelli
- Programa de Pós-Graduação em Medicina, Universidade Nove de Julho (UNINOVE) 01525-000, São Paulo, Brazil
- Laboratório de Lípides (LIM10), Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo 01246-000, São Paulo, Brazil
| |
Collapse
|
2
|
Yan K, Li J, Zhu P, Tang X, Li Y, Yang Y, Gao R, Yuan J, Zhao X. Associations of HDL-C and ApoA-I with Mortality Risk in PCI Patients Across Different hsCRP Levels. J Inflamm Res 2024; 17:4345-4359. [PMID: 38979437 PMCID: PMC11229981 DOI: 10.2147/jir.s465015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 06/25/2024] [Indexed: 07/10/2024] Open
Abstract
Purpose The association between high-density lipoprotein cholesterol (HDL-C) and apolipoprotein A-I (ApoA-I) and cardiovascular risk in patients with coronary artery disease remains inconsistent. Recent investigations indicated potential dysfunctionality of HDL under inflammation. This study endeavors to explore whether the inflammatory status modifies the effects of HDL-C and ApoA-I on cardiovascular risk in individuals with percutaneous coronary intervention (PCI). Patients and Methods Consecutive 10,724 PCI patients at Fuwai hospital in 2013 were enrolled. Inflammation status was defined by high-sensitivity C-reactive proteins (hsCRP) ≥ 2 mg/L. The study endpoint was cardiac mortality. Results Among 9569 PCI patients eventually included, 225 (2.4%) cardiac mortality happened during 5 years. In hsCRP ≥ 2 mg/L group, an U-shaped curve was observed for HDL-C and multivariate Cox regression showed that elevated risks of cardiac mortality correlated to both the lowest quintile (hazard ratio [HR], 2.50; 95% confidence interval [CI], 1.32-4.71) and the highest quintile of HDL-C (HR, 2.28; 95% CI, 1.23-4.25). However, an L-shaped curve existed in ApoA-I, indicating only the lowest quintile level of ApoA-I was associated with an increased cardiac mortality risk (HR, 2.19; 95% CI, 1.28-3.75). Nevertheless, in hsCRP < 2 mg/L group, no significant correlations between HDL-C and ApoA-I and cardiac mortality risk were identified (both P > 0.05). Conclusion In PCI patients with hsCRP ≥ 2 mg/L. both low and high HDL-C levels correlated with higher cardiac mortality risk (U-shaped), while only low ApoA-I levels were linked to elevated risk (L-shaped). However, in patients with hsCRP < 2 mg/L, neither HDL-C nor ApoA-I levels were associated with higher cardiac mortality risk. These findings shed light on the importance of considering inflammation status, particularly hsCRP levels, in managing HDL-C and ApoA-I levels, and suggest targeting elevated ApoA-I levels as a potential therapeutic approach for PCI patients with hsCRP ≥ 2 mg/L.
Collapse
Affiliation(s)
- Kailun Yan
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, People's Republic of China
| | - Jiawen Li
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, People's Republic of China
| | - Pei Zhu
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, People's Republic of China
| | - Xiaofang Tang
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, People's Republic of China
| | - Yulong Li
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, People's Republic of China
| | - Yuejin Yang
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, People's Republic of China
| | - Runlin Gao
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, People's Republic of China
| | - Jinqing Yuan
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, People's Republic of China
| | - Xueyan Zhao
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, People's Republic of China
| |
Collapse
|
3
|
da Silva Siqueira L, Rodrigues FVF, Zanatta Â, Gonçalves JIB, Ghilardi IM, Alcará AM, Becker NB, Pinzetta G, Zanirati G, Becker BMA, Erwig HS, da Costa JC, Marinowic DR. Evaluation of the effects of the Zika Virus-Immunoglobulin G + complex on murine microglial cells. J Neurovirol 2024:10.1007/s13365-024-01218-7. [PMID: 38935226 DOI: 10.1007/s13365-024-01218-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 06/04/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024]
Abstract
After the Zika virus (ZIKV) epidemic in Brazil, ZIKV infections were linked to damage to the central nervous system (CNS) and congenital anomalies. Due to the virus's ability to cross the placenta and reach brain tissue, its effects become severe, leading to Congenital Zika Syndrome (CZS) and resulting in neuroinflammation, microglial activation, and secretion of neurotoxic factors. The presence of ZIKV triggers an inadequate fetal immune response, as the fetus only has the protection of maternal antibodies of the Immunoglobulin G (IgG) class, which are the only antibodies capable of crossing the placenta. Because of limited understanding regarding the long term consequences of ZIKV infection and the involvement of maternal antibodies, this study sought to assess the impact of the ZIKV + IgG⁺complex on murine microglial cells. The cells were exposed to ZIKV, IgG antibodies, and the ZIKV + IgG⁺complex for 24 and 72 h. Treatment-induced cytotoxic effects were evaluated using the cell viability assay, oxidative stress, and mitochondrial membrane potential. The findings indicated that IgG antibodies exhibit cytotoxic effects on microglia, whether alone or in the presence of ZIKV, leading to compromised cell viability, disrupted mitochondrial membrane potential, and heightened oxidative damage. Our conclusion is that IgG antibodies exert detrimental effects on microglia, triggering their activation and potentially disrupting the creation of a neurotoxic environment. Moreover, the presence of antibodies may correlate with an elevated risk of ZIKV-induced neuroinflammation, contributing to long-term CNS damage.
Collapse
Affiliation(s)
- Laura da Silva Siqueira
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga 6690, Porto Alegre, 90610-000, RS, Brazil
- School of Medicine, Graduate Program in Pediatrics and Child Health, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, 90610-000, RS, Brazil
| | - Felipe Valle Fortes Rodrigues
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga 6690, Porto Alegre, 90610-000, RS, Brazil
- School of Medicine, Graduate Program in Pediatrics and Child Health, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, 90610-000, RS, Brazil
| | - Ângela Zanatta
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga 6690, Porto Alegre, 90610-000, RS, Brazil
- School of Medicine, Graduate Program in Pediatrics and Child Health, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, 90610-000, RS, Brazil
| | - João Ismael Budelon Gonçalves
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga 6690, Porto Alegre, 90610-000, RS, Brazil
| | - Isadora Machado Ghilardi
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga 6690, Porto Alegre, 90610-000, RS, Brazil
- School of Medicine, Graduate Program in Pediatrics and Child Health, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, 90610-000, RS, Brazil
| | - Allan Marinho Alcará
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga 6690, Porto Alegre, 90610-000, RS, Brazil
- School of Medicine, Graduate Program in Pediatrics and Child Health, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, 90610-000, RS, Brazil
| | - Nicole Bernd Becker
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga 6690, Porto Alegre, 90610-000, RS, Brazil
| | - Giulia Pinzetta
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga 6690, Porto Alegre, 90610-000, RS, Brazil
| | - Gabriele Zanirati
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga 6690, Porto Alegre, 90610-000, RS, Brazil
- School of Medicine, Graduate Program in Pediatrics and Child Health, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, 90610-000, RS, Brazil
| | - Bruno Maestri Abrianos Becker
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga 6690, Porto Alegre, 90610-000, RS, Brazil
| | - Helena Scartassini Erwig
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga 6690, Porto Alegre, 90610-000, RS, Brazil
| | - Jaderson Costa da Costa
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga 6690, Porto Alegre, 90610-000, RS, Brazil
- School of Medicine, Graduate Program in Pediatrics and Child Health, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, 90610-000, RS, Brazil
| | - Daniel Rodrigo Marinowic
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga 6690, Porto Alegre, 90610-000, RS, Brazil.
- School of Medicine, Graduate Program in Pediatrics and Child Health, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, 90610-000, RS, Brazil.
| |
Collapse
|
4
|
Serum anti-SERPINE1 antibody as a potential biomarker of acute cerebral infarction. Sci Rep 2021; 11:21772. [PMID: 34741085 PMCID: PMC8571331 DOI: 10.1038/s41598-021-01176-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 10/19/2021] [Indexed: 11/11/2022] Open
Abstract
The presence of disease-specific antigens and autoantibodies in the sera of patients with atherosclerosis-related diseases has been widely reported and is considered to result from inflammation of the arterial wall and the involvement of immune factors. The aim of this study was to identify a novel antibody in patients with ischemic stroke by serological identification of antigens using recombinant cDNA expression cloning from patients who had a transient ischemic attack (TIA). We identified the serpin peptidase inhibitor, clade E member 1 (SERPINE1), as a candidate antigen. The serum anti-SERPINE1 antibody levels quantified using amplified luminescent proximity homogeneous assay-linked immunosorbent assay were significantly higher in patients with ischemic stroke, including those with acute cerebral infarction (aCI), TIA, and chronic cerebral infarction, than in healthy donors. The antibody levels were strongly associated with old age, female sex, and presence of hypertension, diabetes mellitus, and cardiovascular disease. Age and intima-media thickness of the carotid artery were positively correlated with antibody levels, which suggests that SERPINE1 may reflect the progression of atherosclerosis. In a multivariate analysis, SERPINE1 antibody level was an independent predictor of aCI. Thus, the serum levels of anti-SERPINE1 antibody could potentially serve as a biomarker of atherothrombotic infarction.
Collapse
|
5
|
Ministrini S, Carbone F. PCSK9 and inflammation. Maybe a role in autoimmune diseases? Focus on rheumatoid arthritis and systemic lupus erythematosus. Curr Med Chem 2021; 29:970-979. [PMID: 34375179 DOI: 10.2174/0929867328666210810150940] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 06/17/2021] [Accepted: 06/28/2021] [Indexed: 11/22/2022]
Abstract
Despite a clear epidemiological link between autoimmune disease and cardiovascular (CV) risk exists, pathophysiological explanations are extremely complex and far from being elucidated. Dysregulation of metabolic pathways and chronic low-grade inflammation represent common pathways, but CV risk still remains underestimated in patients with autoimmune diseas. Among different candidate mediators, pro-protein convertase subtilisin/kexin type 9 (PCSK9) is attracting a growing attention, due to a combined effect on lipid metabolism and inflammatory response. Study on PCSK9 inhibitors have established a clear benefit on CV outcome without an established effect on inflammation. Conversely, evidence from sepsis and HIV infection strongly support a pro-inflammatory role of PCSK9. Still uncertain is instead the role of PCSK9 in autoimmune disease. So far reported clinical findings are controversial and likely reflect the poor knowledge of PCSK9 activity on monocyte/macrophage migration and activation. The complex signaling network around PCSK9 synthesis and metabolism may also have a role, especially concerning the involvement of scavenger receptors such as CD36. Such complexity in PCSK9 signaling seems particularly evident in autoimmune disease model. This would also potentially explain the observed independency between lipid profile and PCSK9 levels, the so-called "lipid paradox". In this narrative review we will summarize the current knowledge about the complex network of PCSK9 signaling. We will focus of upstream and downstream pathways with potential implication in autoimmune disease and potential effects of PCSK9 inhibiting strategies.
Collapse
Affiliation(s)
- Stefano Ministrini
- Internal Medicine, Angiology and Atherosclerosis; Department of Medicine and Surgery, University of Perugia, p.le Gambuli 1, 06132 Perugia, Italy
| | - Federico Carbone
- First Clinic of internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| |
Collapse
|
6
|
Hiwasa T, Wang H, Goto KI, Mine S, Machida T, Kobayashi E, Yoshida Y, Adachi A, Matsutani T, Sata M, Yamagishi K, Iso H, Sawada N, Tsugane S, Kunimatsu M, Kamitsukasa I, Mori M, Sugimoto K, Uzawa A, Muto M, Kuwabara S, Kobayashi Y, Ohno M, Nishi E, Hattori A, Yamamoto M, Maezawa Y, Kobayashi K, Ishibashi R, Takemoto M, Yokote K, Takizawa H, Kishimoto T, Matsushita K, Kobayashi S, Nomura F, Arasawa T, Kagaya A, Maruyama T, Matsubara H, Tomiita M, Hamanaka S, Imai Y, Nakagawa T, Kato N, Terada J, Matsumura T, Katsumata Y, Naito A, Tanabe N, Sakao S, Tatsumi K, Ito M, Shiratori F, Sumazaki M, Yajima S, Shimada H, Shirouzu M, Yokoyama S, Kudo T, Doi H, Iwase K, Ashino H, Li SY, Kubota M, Tomiyoshi G, Shinmen N, Nakamura R, Kuroda H, Iwadate Y. Serum anti-DIDO1, anti-CPSF2, and anti-FOXJ2 antibodies as predictive risk markers for acute ischemic stroke. BMC Med 2021; 19:131. [PMID: 34103026 PMCID: PMC8188684 DOI: 10.1186/s12916-021-02001-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 04/30/2021] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Acute ischemic stroke (AIS) is a serious cause of mortality and disability. AIS is a serious cause of mortality and disability. Early diagnosis of atherosclerosis, which is the major cause of AIS, allows therapeutic intervention before the onset, leading to prevention of AIS. METHODS Serological identification by cDNA expression cDNA libraries and the protein array method were used for the screening of antigens recognized by serum IgG antibodies in patients with atherosclerosis. Recombinant proteins or synthetic peptides derived from candidate antigens were used as antigens to compare serum IgG levels between healthy donors (HDs) and patients with atherosclerosis-related disease using the amplified luminescent proximity homogeneous assay-linked immunosorbent assay. RESULTS The first screening using the protein array method identified death-inducer obliterator 1 (DIDO1), forkhead box J2 (FOXJ2), and cleavage and polyadenylation specificity factor (CPSF2) as the target antigens of serum IgG antibodies in patients with AIS. Then, we prepared various antigens including glutathione S-transferase-fused DIDO1 protein as well as peptides of the amino acids 297-311 of DIDO1, 426-440 of FOXJ2, and 607-621 of CPSF2 to examine serum antibody levels. Compared with HDs, a significant increase in antibody levels of the DIDO1 protein and peptide in patients with AIS, transient ischemic attack (TIA), and chronic kidney disease (CKD) but not in those with acute myocardial infarction and diabetes mellitus (DM). Serum anti-FOXJ2 antibody levels were elevated in most patients with atherosclerosis-related diseases, whereas serum anti-CPSF2 antibody levels were associated with AIS, TIA, and DM. Receiver operating characteristic curves showed that serum DIDO1 antibody levels were highly associated with CKD, and correlation analysis revealed that serum anti-FOXJ2 antibody levels were associated with hypertension. A prospective case-control study on ischemic stroke verified that the serum antibody levels of the DIDO1 protein and DIDO1, FOXJ2, and CPSF2 peptides showed significantly higher odds ratios with a risk of AIS in patients with the highest quartile than in those with the lowest quartile, indicating that these antibody markers are useful as risk factors for AIS. CONCLUSIONS Serum antibody levels of DIDO1, FOXJ2, and CPSF2 are useful in predicting the onset of atherosclerosis-related AIS caused by kidney failure, hypertension, and DM, respectively.
Collapse
Affiliation(s)
- Takaki Hiwasa
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan. .,Department of Biochemistry and Genetics, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan. .,Comprehensive Stroke Center, Chiba University Hospital, Chiba, 260-8677, Japan.
| | - Hao Wang
- Department of Biochemistry and Genetics, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan.,Department of Anesthesia, The First Affiliated Hospital, Jinan University, Guanzhou, 510632, P. R. China
| | - Ken-Ichiro Goto
- Department of Biochemistry and Genetics, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Seiichiro Mine
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan.,Department of Neurological Surgery, Chiba Prefectural Sawara Hospital, Chiba, 287-0003, Japan.,Department of Neurological Surgery, Chiba Cerebral and Cardiovascular Center, Chiba, 290-0512, Japan
| | - Toshio Machida
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan.,Department of Neurological Surgery, Chiba Cerebral and Cardiovascular Center, Chiba, 290-0512, Japan.,Department of Neurosurgery, Eastern Chiba Medical Center, Chiba, 283-8686, Japan
| | - Eiichi Kobayashi
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan.,Comprehensive Stroke Center, Chiba University Hospital, Chiba, 260-8677, Japan
| | - Yoichi Yoshida
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan.,Comprehensive Stroke Center, Chiba University Hospital, Chiba, 260-8677, Japan
| | - Akihiko Adachi
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan.,Comprehensive Stroke Center, Chiba University Hospital, Chiba, 260-8677, Japan
| | - Tomoo Matsutani
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan.,Comprehensive Stroke Center, Chiba University Hospital, Chiba, 260-8677, Japan
| | - Mizuki Sata
- Department of Public Health Medicine, Faculty of Medicine, and Health Services Research and Development Center, University of Tsukuba, Tsukuba, 305-8575, Japan.,Department of Preventive Medicine and Public Health, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Kazumasa Yamagishi
- Department of Public Health Medicine, Faculty of Medicine, and Health Services Research and Development Center, University of Tsukuba, Tsukuba, 305-8575, Japan
| | - Hiroyasu Iso
- Public Health, Department of Social Medicine, Osaka University Graduate School of Medicine, Suita, 565-0871, Japan
| | - Norie Sawada
- Epidemiology and Prevention Group, Center for Public Health Sciences, National Cancer Center, Tokyo, 104-0045, Japan
| | - Shoichiro Tsugane
- Epidemiology and Prevention Group, Center for Public Health Sciences, National Cancer Center, Tokyo, 104-0045, Japan
| | - Mitoshi Kunimatsu
- Department of Home Economics, Nagoya Women's University, Nagoya, 467-8610, Japan
| | - Ikuo Kamitsukasa
- Department of Neurology, Chiba Rosai Hospital, Chiba, 290-0003, Japan.,Department of Neurology, Chibaken Saiseikai Narashino Hospital, Chiba, 275-8580, Japan
| | - Masahiro Mori
- Department of Neurology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Kazuo Sugimoto
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan.,Department of Neurology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Akiyuki Uzawa
- Comprehensive Stroke Center, Chiba University Hospital, Chiba, 260-8677, Japan.,Department of Neurology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Mayumi Muto
- Department of Neurology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Satoshi Kuwabara
- Comprehensive Stroke Center, Chiba University Hospital, Chiba, 260-8677, Japan.,Department of Neurology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Yoshio Kobayashi
- Department of Cardiovascular Medicine, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Mikiko Ohno
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, 606-8507, Japan.,Department of Pharmacology, Shiga University of Medical Science, Shiga, 520-2192, Japan
| | - Eiichiro Nishi
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, 606-8507, Japan.,Department of Pharmacology, Shiga University of Medical Science, Shiga, 520-2192, Japan
| | - Akiko Hattori
- Department of Endocrinology, Hematology and Gerontology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Masashi Yamamoto
- Department of Endocrinology, Hematology and Gerontology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Yoshiro Maezawa
- Department of Endocrinology, Hematology and Gerontology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Kazuki Kobayashi
- Department of Endocrinology, Hematology and Gerontology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Ryoichi Ishibashi
- Department of Endocrinology, Hematology and Gerontology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Minoru Takemoto
- Department of Endocrinology, Hematology and Gerontology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan.,Department of Diabetes, Metabolism and Endocrinology, School of Medicine, International University of Health and Welfare, Chiba, 286-8686, Japan
| | - Koutaro Yokote
- Department of Endocrinology, Hematology and Gerontology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Hirotaka Takizawa
- Port Square Kashiwado Clinic, Kashiwado Memorial Foundation, Chiba, 260-0025, Japan
| | - Takashi Kishimoto
- Department of Molecular Pathology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Kazuyuki Matsushita
- Department of Laboratory Medicine & Division of Clinical Genetics, Chiba University Hospital, Chiba, 260-8677, Japan
| | - Sohei Kobayashi
- Department of Laboratory Medicine & Division of Clinical Genetics, Chiba University Hospital, Chiba, 260-8677, Japan.,Department of Medical Technology and Sciences, School of Health Sciences at Narita, International University of Health and Welfare, Chiba, 286-8686, Japan
| | - Fumio Nomura
- Division of Clinical Genetics, Chiba Foundation for Health Promotion and Disease Prevention, Chiba, 261-0002, Japan
| | - Takahiro Arasawa
- Department of Biochemistry and Genetics, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan.,Department of Frontier Surgery, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Akiko Kagaya
- Department of Frontier Surgery, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Tetsuro Maruyama
- Department of Frontier Surgery, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Hisahiro Matsubara
- Department of Frontier Surgery, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Minako Tomiita
- Department of Allergy and Rheumatology, Chiba Children's Hospital, Chiba, 266-0007, Japan
| | - Shinsaku Hamanaka
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Yushi Imai
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Tomoo Nakagawa
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Naoya Kato
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Jiro Terada
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Takuma Matsumura
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Yusuke Katsumata
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Akira Naito
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Nobuhiro Tanabe
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan.,Department of Advanced Medicine in Pulmonary Hypertension, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Seiichiro Sakao
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Koichiro Tatsumi
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Masaaki Ito
- Department of Gastroenterological Surgery and Clinical Oncology, Toho University Graduate School of Medicine, Tokyo, 143-8541, Japan
| | - Fumiaki Shiratori
- Department of Gastroenterological Surgery and Clinical Oncology, Toho University Graduate School of Medicine, Tokyo, 143-8541, Japan
| | - Makoto Sumazaki
- Department of Gastroenterological Surgery and Clinical Oncology, Toho University Graduate School of Medicine, Tokyo, 143-8541, Japan
| | - Satoshi Yajima
- Department of Gastroenterological Surgery and Clinical Oncology, Toho University Graduate School of Medicine, Tokyo, 143-8541, Japan
| | - Hideaki Shimada
- Department of Gastroenterological Surgery and Clinical Oncology, Toho University Graduate School of Medicine, Tokyo, 143-8541, Japan
| | - Mikako Shirouzu
- Division of Structural and Synthetic Biology, RIKEN Center for Life Science Technologies, Yokohama, Kanagawa, 230-0045, Japan
| | - Shigeyuki Yokoyama
- RIKEN Structural Biology Laboratory, Yokohama, Kanagawa, 230-0045, Japan
| | | | | | - Katsuro Iwase
- Department of Biochemistry and Genetics, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Hiromi Ashino
- Department of Biochemistry and Genetics, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Shu-Yang Li
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan.,Department of Biochemistry and Genetics, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Masaaki Kubota
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Go Tomiyoshi
- Department of Biochemistry and Genetics, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan.,Medical Project Division, Research Development Center, Fujikura Kasei Co., Saitama, 340-0203, Japan
| | - Natsuko Shinmen
- Department of Biochemistry and Genetics, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan.,Medical Project Division, Research Development Center, Fujikura Kasei Co., Saitama, 340-0203, Japan
| | - Rika Nakamura
- Department of Biochemistry and Genetics, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan.,Medical Project Division, Research Development Center, Fujikura Kasei Co., Saitama, 340-0203, Japan
| | - Hideyuki Kuroda
- Medical Project Division, Research Development Center, Fujikura Kasei Co., Saitama, 340-0203, Japan
| | - Yasuo Iwadate
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan.,Comprehensive Stroke Center, Chiba University Hospital, Chiba, 260-8677, Japan
| |
Collapse
|
7
|
Rodríguez-Carrio J, Suárez A. The HDL dysfunction gains momentum: is it time for a new approach in rheumatic diseases? Rheumatology (Oxford) 2020; 59:3121-3123. [PMID: 32830277 DOI: 10.1093/rheumatology/keaa346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/19/2020] [Accepted: 05/21/2020] [Indexed: 11/14/2022] Open
Affiliation(s)
- Javier Rodríguez-Carrio
- Department of Functional Biology, Immunology Area, Faculty of Medicine, University of Oviedo.,Instituto de Investigación Sanitaria del Principado de Asturias (ISPA).,Bone and Mineral Research Unit, Instituto Reina Sofía de Investigación Nefrológica, REDinREN del ISCIII, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Ana Suárez
- Department of Functional Biology, Immunology Area, Faculty of Medicine, University of Oviedo.,Instituto de Investigación Sanitaria del Principado de Asturias (ISPA)
| |
Collapse
|
8
|
Nazir S, Jankowski V, Bender G, Zewinger S, Rye KA, van der Vorst EP. Interaction between high-density lipoproteins and inflammation: Function matters more than concentration! Adv Drug Deliv Rev 2020; 159:94-119. [PMID: 33080259 DOI: 10.1016/j.addr.2020.10.006] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 09/20/2020] [Accepted: 10/13/2020] [Indexed: 02/07/2023]
Abstract
High-density lipoprotein (HDL) plays an important role in lipid metabolism and especially contributes to the reverse cholesterol transport pathway. Over recent years it has become clear that the effect of HDL on immune-modulation is not only dependent on HDL concentration but also and perhaps even more so on HDL function. This review will provide a concise general introduction to HDL followed by an overview of post-translational modifications of HDL and a detailed overview of the role of HDL in inflammatory diseases. The clinical potential of HDL and its main apolipoprotein constituent, apoA-I, is also addressed in this context. Finally, some conclusions and remarks that are important for future HDL-based research and further development of HDL-focused therapies are discussed.
Collapse
|
9
|
Yoshida Y, Zhang XM, Wang H, Machida T, Mine S, Kobayashi E, Adachi A, Matsutani T, Kamitsukasa I, Wada T, Aotsuka A, Iwase K, Tomiyoshi G, Nakamura R, Shinmen N, Kuroda H, Takizawa H, Kashiwado K, Shin H, Akaogi Y, Shimada J, Nishi E, Ohno M, Takemoto M, Yokote K, Kitamura K, Iwadate Y, Hiwasa T. Elevated levels of autoantibodies against DNAJC2 in sera of patients with atherosclerotic diseases. Heliyon 2020; 6:e04661. [PMID: 32904265 PMCID: PMC7452465 DOI: 10.1016/j.heliyon.2020.e04661] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 08/09/2019] [Accepted: 08/06/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Serum antibody markers have been increasingly identified not only for cancer and autoimmune diseases but also for atherosclerosis-related diseases such as acute ischemic stroke (AIS), acute myocardial infarction (AMI), diabetes mellitus (DM), and chronic kidney disease (CKD). Biomarkers for transient ischemic attack (TIA) and non-ST segment elevation acute coronary syndrome (NSTEACS) are potentially useful for detection of early phase of atherosclerotic changes against AIS and AMI, respectively. METHODS We utilized serological identification of antigens by recombinant cDNA expression cloning (SEREX) using a human aortic endothelial cell cDNA phage library and sera from patients with TIA or NSTEACS. Serum antibody levels were measured by amplified luminescent proximity homogeneous assay-linked immunosorbent assay (AlphaLISA) using purified recombinant antigens. RESULTS Screening of sera from patients with TIA identified DnaJ heat shock protein family (Hsp40) member C2 (DNAJC2) as a candidate antigen, which was also isolated by SEREX screening using sera of patients with NSTEACS. The validation cohort revealed significantly higher DNAJC2 antibody (DNAJC2-Ab) levels in the sera of patients with TIA or AIS than those in healthy donors (HDs). Multivariate logistic regression analysis indicated that the predictive odds ratios (OR) of DNAJC2-Ab levels for TIA and AIS were 2.54 (95% confidence interval [CI]: 1.36-4.74, p = 0.0034) and 2.14 (95% CI: 1.39-3.30, p = 0.0005), respectively. Serum DNAJC2-Ab levels were also higher in patients with AMI, DM, and CKD than those in HDs. CONCLUSION Serum DNAJC2-Ab level may be useful for early detection of atherosclerotic lesions, which lead to AIS and AMI.
Collapse
Affiliation(s)
- Yoichi Yoshida
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
- Department of Biochemistry and Genetics, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
- Comprehensive Stroke Center, Chiba University Hospital, Chiba 260-8677, Japan
| | - Xiao-Meng Zhang
- Department of Biochemistry and Genetics, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
| | - Hao Wang
- Department of Biochemistry and Genetics, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
- Department of Anesthesia, The First Affiliated Hospital, Jinan University, Guangzhou 510632, PR China
| | - Toshio Machida
- Department of Neurosurgery, Chiba Cerebral and Cardiovascular Center, Ichihara, 290-0512, Chiba, Japan
- Department of Neurosurgery, Eastern Chiba Medical Center, Chiba 283-8686, Japan
| | - Seiichiro Mine
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
- Department of Neurosurgery, Chiba Cerebral and Cardiovascular Center, Ichihara, 290-0512, Chiba, Japan
- Department of Neurosurgery, Sawara Prefectural Hospital, Chiba 287-0003, Japan
| | - Eiichi Kobayashi
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
- Comprehensive Stroke Center, Chiba University Hospital, Chiba 260-8677, Japan
| | - Akihiko Adachi
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
- Comprehensive Stroke Center, Chiba University Hospital, Chiba 260-8677, Japan
| | - Tomoo Matsutani
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
- Comprehensive Stroke Center, Chiba University Hospital, Chiba 260-8677, Japan
| | - Ikuo Kamitsukasa
- Department of Neurology, Chiba Rosai Hospital, Chiba 290-0003, Japan
- Department of Neurology, Chibaken Saiseikai Narashino Hospital, Chiba 275-8580, Japan
| | - Takeshi Wada
- Department of Internal Medicine, Chiba Aoba Municipal Hospital, Chiba 260-0852, Japan
| | - Akiyo Aotsuka
- Department of Internal Medicine, Chiba Aoba Municipal Hospital, Chiba 260-0852, Japan
| | - Katsuro Iwase
- Department of Biochemistry and Genetics, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
| | - Go Tomiyoshi
- Department of Biochemistry and Genetics, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
- Medical Project Division, Research Development Center, Fujikura Kasei Co., Saitama 340-0203, Japan
| | - Rika Nakamura
- Department of Biochemistry and Genetics, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
- Medical Project Division, Research Development Center, Fujikura Kasei Co., Saitama 340-0203, Japan
| | - Natsuko Shinmen
- Department of Biochemistry and Genetics, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
- Medical Project Division, Research Development Center, Fujikura Kasei Co., Saitama 340-0203, Japan
| | - Hideyuki Kuroda
- Medical Project Division, Research Development Center, Fujikura Kasei Co., Saitama 340-0203, Japan
| | - Hirotaka Takizawa
- Port Square Kashiwado Clinic, Kashiwado Memorial Foundation, Chiba 260-0025, Japan
| | - Koichi Kashiwado
- Department of Neurology, Kashiwado Hospital, Chiba 260-0854, Japan
| | - Hideo Shin
- Department of Neurosurgery, Higashi Funabashi Hospital, Chiba 274-0065, Japan
| | - Yuichi Akaogi
- Department of Neurology, Chiba Cerebral and Cardiovascular Center, Chiba 290-0512, Japan
| | - Junichiro Shimada
- Department of Neurology, Chiba Cerebral and Cardiovascular Center, Chiba 290-0512, Japan
| | - Eiichiro Nishi
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
- Department of Pharmacology, Shiga University of Medical Science, Shiga 520-2192, Japan
| | - Mikiko Ohno
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
- Department of Pharmacology, Shiga University of Medical Science, Shiga 520-2192, Japan
| | - Minoru Takemoto
- Department of Clinical Cell Biology and Medicine, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
- Department of Diabetes, Metabolism and Endocrinology, School of Medicine, International University of Health and Welfare, Chiba 286-8686, Japan
| | - Koutaro Yokote
- Department of Clinical Cell Biology and Medicine, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
| | - Kenichiro Kitamura
- Department of Internal Medicine 3, University of Yamanashi School of Medicine, Yamamashi 409-3898, Japan
| | - Yasuo Iwadate
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
- Comprehensive Stroke Center, Chiba University Hospital, Chiba 260-8677, Japan
| | - Takaki Hiwasa
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
- Department of Biochemistry and Genetics, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
- Comprehensive Stroke Center, Chiba University Hospital, Chiba 260-8677, Japan
| |
Collapse
|
10
|
Aidoud A, Marlet J, Angoulvant D, Debacq C, Gavazzi G, Fougère B. Influenza vaccination as a novel means of preventing coronary heart disease: Effectiveness in older adults. Vaccine 2020; 38:4944-4955. [PMID: 32536551 DOI: 10.1016/j.vaccine.2020.05.070] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 05/03/2020] [Accepted: 05/25/2020] [Indexed: 12/28/2022]
Abstract
Atherosclerosis can have various etiologies, including several newly recognized immunoinflammatory mechanisms. A growing body of evidence suggests that influenza infection is chronologically linked to acute myocardial infarction (AMI), and thus that the virus is a novel cardiovascular disease (CVD) risk factor. Morbidity and mortality rates for both influenza infection and AMI rise markedly with age. Epidemiological studies have demonstrated that influenza vaccination (IV) has a cardioprotective effect, especially in people aged 65 and over; hence, IV may be of value in the management of CVD. These observations justify efforts to better understand the underlying mechanisms and to identify therapeutic targets in older adults. In view of the above, the objective of the present study was to review the literature data on the cellular mechanisms that link IV to the prevention of atherosclerotic complications. Given the greater burden of CVD in older subjects, we also questioned the impact of aging on this association. The most widely recognized benefit of IV is the prevention of influenza infection and the latter's cardiovascular complications. In a new hypothesis, however, an influenza-independent effect is driven by vaccine immunity and modulation of the ongoing immunoinflammatory response in individuals with CVD. Although influenza infection and IV both induce a proinflammatory response, they have opposite effects on the progression of atherosclerosis - suggesting a hormetic phenomenon. Aging is characterized by chronic inflammation (sometimes referred to as "inflammaging") that progresses insidiously during the course of aging-related diseases, including CVD. It remains to be determined whether vaccination has an effect on aging-related diseases other than CVD. Although the studies of this topic had various limitations, the results highlight the potential benefits of vaccination in protecting the health of older adults, and should drive research on the molecular immunology of the response to IV and its correlation with atheroprotective processes.
Collapse
Affiliation(s)
- Amal Aidoud
- Division of Geriatric Medicine, Tours University Hospital, Tours, France.
| | - Julien Marlet
- French National Institute of Health and Medical Research INSERM U1259, University of Tours, Tours, France
| | - Denis Angoulvant
- Cardiology Unit, Trousseau Hospital, CHRU de Tours & EA4245, Loire Valley Cardiovascular Collaboration, Tours University, Tours, France
| | - Camille Debacq
- Division of Geriatric Medicine, Tours University Hospital, Tours, France
| | - Gaëtan Gavazzi
- University Clinics of Geriatrics, University Hospital of Grenoble-Alpes, GREPI EA7408 University of Grenoble Alpes, Grenoble, France
| | - Bertrand Fougère
- Division of Geriatric Medicine, Tours University Hospital, Tours, France; Éducation, éthique, santé (EA 7505), Tours University, Tours, France
| |
Collapse
|
11
|
Zhang Y, Zhao H, Liu B, Li L, Zhang L, Bao M, Ji X, He X, Yi J, Chen P, Lu C, Lu A. Low Level Antibodies Against Alpha-Tropomyosin Are Associated With Increased Risk of Coronary Heart Disease. Front Pharmacol 2020; 11:195. [PMID: 32174839 PMCID: PMC7056748 DOI: 10.3389/fphar.2020.00195] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 02/11/2020] [Indexed: 12/20/2022] Open
Abstract
Objective Natural autoantibodies have been implicated to play a key role in the pathogenesis of coronary heart disease (CHD) because they augment autoimmune activation. The aim of this study was to identify novel specific autoantibodies of CHD, and analyze the relationship between their levels and CHD risk indicators. Approach and Results First, clinical data and sera from CHD patients were collected. Then, one protein microarray containing 37 proteins that represent candidate autoantigens was developed. The arrays were used to profile autoantibodies in randomly selected sera from 35 samples (20 CHD patients, and 15 healthy controls). After that, microarray data were analyzed and autoantibodies for CHD were screened out. Then, ELISA detection was conducted to validate the differentiable autoantibodies using larger numbers of serum samples (131 CHD patients, and 131 healthy controls). Finally, the associations of antibodies with CHD risk indicator parameters were assessed. Inter-group comparison by microarray indicated that three CHD novel autoantibodies, including glucose-6-phosphate isomerase (G6PI), alpha-tropomyosin (TPM1), and heterogeneous nuclear ribonucleoprotein D-like (HnRNPDL), were significantly (P < 0.05) increased when compared with the healthy controls. Moreover, a significant increase of IgG autoantibodies for these three autoantigens was confirmed in CHD patients by ELISA (P < 0.0001). The correction analysis revealed a negative correlation of anti-TPM1 antibody levels and total cholesterol (P = 0.0034), and low-density lipoprotein cholesterol (P = 0.0086), respectively. Conclusion G6PI, TPM1, and HnRNPDL were CHD natural autoantigens, and serum anti-TPM1 antibody could be used as a potential marker to predict the risk for CHD patients.
Collapse
Affiliation(s)
- Yin Zhang
- Key Laboratory for Research on Active Ingredients in Natural Medicine of Jiangxi Province, Yichun University, Yichun, China.,Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Heru Zhao
- Key Laboratory for Research on Active Ingredients in Natural Medicine of Jiangxi Province, Yichun University, Yichun, China.,Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Bin Liu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Li Li
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Lulu Zhang
- Key Laboratory for Research on Active Ingredients in Natural Medicine of Jiangxi Province, Yichun University, Yichun, China.,Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Mei Bao
- Key Laboratory for Research on Active Ingredients in Natural Medicine of Jiangxi Province, Yichun University, Yichun, China.,Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xinyu Ji
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaojuan He
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jianfeng Yi
- Key Laboratory for Research on Active Ingredients in Natural Medicine of Jiangxi Province, Yichun University, Yichun, China
| | - Peng Chen
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Cheng Lu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Aiping Lu
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| |
Collapse
|
12
|
IgG Anti-High Density Lipoprotein Antibodies Are Elevated in Abdominal Aortic Aneurysm and Associated with Lipid Profile and Clinical Features. J Clin Med 2019; 9:jcm9010067. [PMID: 31888089 PMCID: PMC7019833 DOI: 10.3390/jcm9010067] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 12/21/2019] [Accepted: 12/24/2019] [Indexed: 02/06/2023] Open
Abstract
High-density lipoproteins cholesterol (HDLc) levels are decreased in abdominal aortic aneurysm (AAA), which is hallmarked by autoimmunity and lipid aortic deposits. To investigate whether IgG anti-HDL antibodies were present in AAA and their potential association with clinical features, IgG anti-HDL and total IgG along with HDLc plasma levels were measured in 488 AAA patients and 184 controls from the Viborg Vascular (VIVA) study, and in tissue-conditioned media from AAA intraluminal thrombus and media layer samples compared to control aortas. Higher IgG anti-HDL levels were found in AAA compared to controls, even after correcting for total IgG, and after adjusting for potential confounders. IgG anti-HDL levels were correlated with aortic diameter in univariate and adjusted multivariate analyses. IgG anti-HDL antibodies were negatively associated with HDLc levels before and after correcting for potential confounders. Increased anti-HDL antibodies were identified in tissue-conditioned media from AAA samples compared to healthy aortas, with higher levels being observed in the media layer. In conclusion, increased IgG anti-HDL levels (both in plasma and in tissue) are linked to AAA, associated with aortic diameter and HDLc levels. These data suggest a potential immune response against HDL in AAA and support an emerging role of anti-HDL antibodies in AAA.
Collapse
|
13
|
Carbone F, Dallegri F, Montecucco F, Poggi A, Nobili FM, Cacciapaglia F, Afeltra A, Moccetti T, Colombo BM. Serum osteopontin negatively impacts on intima-media thickness in patients with systemic lupus erythematosus. Eur J Clin Invest 2019; 49:e13089. [PMID: 30767212 DOI: 10.1111/eci.13089] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 12/06/2018] [Accepted: 02/12/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Ultrasound evaluation of carotid intima-media thickness (cIMT) has been extensively used for potentially improving cardiovascular (CV) risk stratification in several patients' categories. Subjects with systemic lupus erythematosus (SLE) have been investigated by both imaging and molecular biomarker approaches with contrasting results. Here, we focused on the role of osteopontin (OPN) as biomarker of subclinical atherosclerosis associated with SLE. MATERIALS AND METHODS Eighty females (age 18-65 years) affected by SLE and eighty age-matched healthy female controls without a clinical history of CV disease underwent ultrasound evaluation of cIMT and blood sample assay of high-sensitivity C-reactive protein (hs-CRP) and OPN. RESULTS Healthy controls and SLE patients significantly differed for CV risk factors (ie, waist circumference, hypertension and dyslipidaemia) and the inflammatory status. Noteworthy, an opposite association between cIMT and OPN was observed in the two study groups. Whereas OPN was positively associated with mean cIMT (r = 0.364; P = 0.001) in SLE patients, a negative correlation was found in healthy controls. Furthermore, in SLE patients increased circulating levels of OPN were associated with the use of hydroxychloroquine and the positivity for the anti-dsDNA autoantibodies. At linear regression analysis, only OPN remained independently associated with cIMT also after adjustment for age, smoking pack-year, Heart SCORE, disease length and steroid therapy length. CONCLUSIONS These results indicate that serum OPN levels were strongly associated with subclinical atherosclerosis in patients with LES and it might be a useful CV biomarker that requires additional validation in larger trials.
Collapse
Affiliation(s)
- Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Franco Dallegri
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy.,IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascolar Network, Genoa, Italy
| | - Fabrizio Montecucco
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascolar Network, Genoa, Italy.,First Clinic of Internal Medicine, Department of Internal Medicine and Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy
| | - Alessandro Poggi
- Molecular Oncology and Angiogenesis Unit, Policlinico San Martino, Genoa, Italy
| | - Flavio Mariano Nobili
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascolar Network, Genoa, Italy.,Clinical Neurology, Department of Neuroscience (DINOGMI), University of Genoa, Ospedale Policlinico San Martino, Genoa, Italy
| | - Fabio Cacciapaglia
- Rheumatology Unit, Department of Emergency and Organs Transplantation (DETO), University of Bari, Bari, Italy
| | - Antonella Afeltra
- Unit of Allergology, Immunology, Rheumatology, Department of Medicine, University Campus Bio-Medico, Rome, Italy
| | - Tiziano Moccetti
- Cellular and Molecular Cardiology Laboratory, Cardiocentro Ticino Foundation and Swiss Institute for Regenerative Medicine (SIRM), Lugano, Switzerland
| | - Barbara M Colombo
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascolar Network, Genoa, Italy
| |
Collapse
|
14
|
Ernst D, Widera C, Weiberg D, Derlin T, Ahrenstorf G, Sogkas G, Jablonka A, Schmidt RE, Witte T, Heidecke H, Riemekasten G. Beta-1-Adrenergic Receptor Antibodies in Acute Coronary Syndrome: Is Less Sometimes More? Front Cardiovasc Med 2019; 5:170. [PMID: 30619882 PMCID: PMC6305491 DOI: 10.3389/fcvm.2018.00170] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 11/05/2018] [Indexed: 11/29/2022] Open
Abstract
Background: Anti-beta-1-adrenergic receptor antibodies (anti-β1AR Ab) are associated with ischemic cardiomyopathies (ICM). Evidence continues to emerge supporting an autoimmune component to various cardiac diseases. This study compares anti-β1AR Ab concentrations in patients with different entities of acute coronary syndromes (ACS) to asymptomatic non-ACS patients with positron-emission computed tomography (PET/CT)-proven atherosclerosis, and healthy controls. Methods: Serum anti-β1AR Ab IgG concentrations were measured in 212 ACS patients, 100 atherosclerosis patients, and 62 controls using ELISA. All ACS patients underwent coronary angiography. All 374 patients participating completed a structured questionnaire regarding traditional cardiovascular risk factors. ACS patients were followed up for 6 months. Results: Patients with ACS exhibited lower anti-β1AR Ab levels compared to patients with atherosclerosis or healthy controls (both p < 0.001). No differences in the ab levels were evident between healthy controls and patients with atherosclerosis. In the ACS groups, lower concentrations were found in patients with ST-elevation myocardial infarction (STEMI) (0.67 μg/ml) compared to patients with angina pectoris (AP) and non-ST elevation myocardial infarction (NSTEMI) (both 0.76 μg/ml, p = 0.008). Anti-β1AR Ab levels ≤ 0.772 μg/ml were predictive for death and reinfarction (AUC 0.77, p = 0.006). No significant correlations between anti-β1AR Ab levels and atherosclerotic burden or traditional cardiovascular risk factors were identified. Conclusions: Lower anti-β1AR Ab concentrations appear to characterize ACS phenotypes and could serve as diagnostic and prognostic markers independent from traditional risk factors for atheroscle. The prognostic predictive value of anti-β1AR Ab in ACS remains to be confirmed in larger studies.
Collapse
Affiliation(s)
- Diana Ernst
- Department of Rheumatology and Immunology, Hanover Medical School, Hanover, Germany
| | - Christian Widera
- Department of Cardiology, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Desiree Weiberg
- Department of Nuclear Medicine, Hanover Medical School, Hanover, Germany
| | - Thorsten Derlin
- Department of Nuclear Medicine, Hanover Medical School, Hanover, Germany
| | - Gerrit Ahrenstorf
- Department of Rheumatology and Immunology, Hanover Medical School, Hanover, Germany
| | - Georgios Sogkas
- Department of Rheumatology and Immunology, Hanover Medical School, Hanover, Germany
| | - Alexandra Jablonka
- Department of Rheumatology and Immunology, Hanover Medical School, Hanover, Germany
| | - Reinhold E Schmidt
- Department of Rheumatology and Immunology, Hanover Medical School, Hanover, Germany
| | - Torsten Witte
- Department of Rheumatology and Immunology, Hanover Medical School, Hanover, Germany
| | | | | |
Collapse
|
15
|
Banach M, Mazidi M, Mikhailidis DP, Toth PP, Jozwiak J, Rysz J, Watts GF. Association between phenotypic familial hypercholesterolaemia and telomere length in US adults: results from a multi-ethnic survey. Eur Heart J 2018; 39:3635-3640. [PMID: 30165413 DOI: 10.1093/eurheartj/ehy527] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Accepted: 08/09/2018] [Indexed: 12/24/2022] Open
Abstract
Aims Familial hypercholesterolaemia (FH) accelerates atherosclerotic cardiovascular disease (ASCVD) and accordingly is the most potent hereditary cause of premature coronary heart disease. The association between telomere length (TL), a biological index of ageing, and FH has not been hitherto investigated. We addressed this question using data from the US National Health and Education National Surveys (NHANES, 1999-2002). Methods and results We included individuals, who had TL measurements (with quantitative polymerase chain reaction method) and a phenotypic diagnosis of FH based on the Dutch Lipid Clinic Network (DLCN) criteria. Sample weights were applied for unequal probabilities of selection, non-response bias, and oversampling by complex sample analysis. The adult prevalence of FH in NHANES was 0.43% [95% confidence interval (95% CI) 0.33-0.57]. The frequencies of probable FH (mean DLCN score: 6.2) and definite FH (mean DLCN score: 8.9) were 0.42% (95% CI 0.32-0.48) and 0.03% (95% CI 0.02-0.06), respectively. Subjects with FH had a higher prevalence of non-communicable diseases (hypertension, diabetes 2 type, and obesity) and early atherosclerosis (2.9% in overall population vs. 42.2% in FH). Overall, the mean TL in the non-FH population was 1.09 (95% CI 1.06-1.12) (T/S ratio) and 1.09 (95% CI 1.03-1.12) [(T/S ratio) for total FH]. Telomere length adjusted for age, sex, race, and body mass index was shorter in FH compared with healthy subjects (FH 0.89, 95% CI 0.84-0.93 vs. healthy: 1.05, 95% CI 0.97-1.11 T/S ratio; P < 0.001). Subjects with longer TL (highest quartile) had 12% less chance of having FH compared with those with TL in the lowest quartile (Q1, 95% CI 0.78-0.93). Conclusions These preliminary data suggest an association between TL, an index of biological age, and the presence of FH, the most common inherited cause of premature ASCVD. Given our relatively low sample size, the findings need confirmation in larger studies.
Collapse
Affiliation(s)
- Maciej Banach
- Chair of Nephrology and Hypertension, Medical University of Lodz, Zeromskiego 113, Lodz, Poland
- Polish Mother's Memorial Hospital Research Institute, Rzgowska 281/289, Lodz, Poland
- Cardiovascular Research Centre, University of Zielona Gora, Zyty 28, Zielona Gora, Poland
| | - Mohsen Mazidi
- Department of Biology and Biological Engineering, Food and Nutrition Science, Chalmers University of Technology, Kemigarden 4, Gothenburg, Sweden
| | - Dimitri P Mikhailidis
- Department of Clinical Biochemistry, Royal Free Campus, University College London Medical School, University College London (UCL), Pond Street, London, UK
| | - Peter P Toth
- The Johns Hopkins Ciccarone Center for the Prevention of Heart Disease, 600 N. Wolfe St, Carnegie 565-G, Baltimore, MD, USA
- Preventive Cardiology, CGH Medical Center, 01 East Miller Road, Sterling, IL, USA
| | - Jacek Jozwiak
- Department of Medicine and Public Health, University of Opole, Kopernika 11A, Opole, Poland
| | - Jacek Rysz
- Chair of Nephrology and Hypertension, Medical University of Lodz, Zeromskiego 113, Lodz, Poland
| | - Gerald F Watts
- Cardiometabolic Service, Department of Cardiology, Royal Perth Hospital, School of Medicine, University of Western Australia, 35 Stirling Highway, Crawley, WA, Australia
| |
Collapse
|
16
|
Yoshida Y, Hiwasa T, Machida T, Kobayashi E, Mine S, Matsushima J, Takiguchi M, Iwadate Y. Elevation of Autoantibody in Patients with Ischemic Stroke. Neurol Med Chir (Tokyo) 2018; 58:303-310. [PMID: 29848906 PMCID: PMC6048350 DOI: 10.2176/nmc.ra.2018-0022] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Recent clinical research has revealed a significant correlation between atherosclerosis, one of the primary etiologies of ischemic stroke, and the immune system. Assuming that "disease-specific autoantibodies are produced in the sera of patients with ischemic stroke," we investigated multiple arteriosclerosis-related antibodies using the serological identification of antigens by recombinant cDNA expression cloning (SEREX), an established method for identifying antigenic proteins. We either screened a human aortic endothelial cell cDNA library or conducted protein array screening using the sera from patients with ischemic stroke, such as carotid artery stenosis or transient ischemic attack (TIA). Next, we measured serum antibody levels using amplified luminescent proximity homogeneous assay-linked immunosorbent assay (AlphaLISA) in patient/healthy donor (HD) cohorts and identified several antigens, the antibody levels of which were significantly higher in patients with ischemic stroke than in HDs. This review introduced the method of identifying antigens by the SEREX and protein microarray and summarized antigenic proteins. In particular, it focused on anti-replication protein A2 antibody and anti-programmed cell death 11 antibody, which are significantly related to atherosclerotic plaque and ischemic brain tissue, respectively, and proposed the mechanism of elevated autoantibody levels against them. Furthermore, this review suggests a possibility of clinical application as an atherosclerotic disease diagnostic marker for TIA or cerebral infarction.
Collapse
Affiliation(s)
- Yoichi Yoshida
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University.,Department of Biochemistry and Genetics, Graduate School of Medicine, Chiba University.,Comprehensive Stroke Center, Chiba University Hospital
| | - Takaki Hiwasa
- Department of Biochemistry and Genetics, Graduate School of Medicine, Chiba University
| | - Toshio Machida
- Department of Neurosurgery, Chiba Cerebral and Cardiovascular Center.,Department of Neurosurgery, Eastern Chiba Medical Center
| | - Eiichi Kobayashi
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University.,Comprehensive Stroke Center, Chiba University Hospital
| | - Seiichiro Mine
- Department of Neurosurgery, Chiba Cerebral and Cardiovascular Center.,Department of Neurosurgery, Prefectural Sawara Hospital
| | - Jun Matsushima
- Department of Diagnostic Pathology, Graduate School of Medicine, Chiba University
| | - Masaki Takiguchi
- Department of Biochemistry and Genetics, Graduate School of Medicine, Chiba University
| | - Yasuo Iwadate
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University
| |
Collapse
|
17
|
Mollazadeh H, Carbone F, Montecucco F, Pirro M, Sahebkar A. Oxidative burden in familial hypercholesterolemia. J Cell Physiol 2018; 233:5716-5725. [PMID: 29323716 DOI: 10.1002/jcp.26466] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 01/05/2018] [Indexed: 12/19/2022]
Abstract
Familial hypercholesterolemia (FH) is a genetic disorder characterized by high serum levels of low-density lipoprotein cholesterol (LDL-c). FH is characterized by accelerated development of atherosclerosis and represents the most frequent hereditary cause of premature coronary heart disease. Mutations of the LDL receptor gene are the genetic signature of FH, resulting in abnormal levels of circulating LDLs. Moreover, FH promotes the generation of reactive oxygen species (ROS) which is another key mechanism involved in atherosclerosis development and progression. The aim of this narrative review is to update the current knowledge on the pathophysiological mechanisms linking FH to ROS generation and their detrimental impact on atherosclerotic pathophysiology. With this purpose, we reviewed experimental and clinical data on the association between FH and OS and the functional role of OS as a promoter of inflammation and atherosclerosis. In this regard, oxidant species such as oxidized LDL, malondialdehyde, ROS, and isoprostanes emerged as leading mediators of the oxidative injury in FH. In conclusion, targeting oxidative stress may be a promising therapeutic strategy to reduce atherogenesis in patients with FH.
Collapse
Affiliation(s)
- Hamid Mollazadeh
- Department of Physiology and Pharmacology, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran.,Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy.,Ospedale Policlinico San Martino, Genoa, Italy.,Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy
| | - Matteo Pirro
- Unit of Internal Medicine, Angiology and Arteriosclerosis Diseases, Department of Medicine, University of Perugia, Perugia, Italy
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
18
|
Yoshida Y, Wang H, Hiwasa T, Machida T, Kobayashi E, Mine S, Tomiyoshi G, Nakamura R, Shinmen N, Kuroda H, Takizawa H, Kashiwado K, Kamitsukasa I, Shin H, Wada T, Aotsuka A, Nishi E, Ohno M, Takemoto M, Yokote K, Takahashi S, Matsushima J, Zhang XM, Takiguchi M, Iwadate Y. Elevation of autoantibody level against PDCD11 in patients with transient ischemic attack. Oncotarget 2017; 9:8836-8848. [PMID: 29507658 PMCID: PMC5823671 DOI: 10.18632/oncotarget.23653] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 11/16/2017] [Indexed: 11/25/2022] Open
Abstract
Background Disease specific autoantibodies have been detected in the sera of patients with atherosclerosis-related diseases, such as cerebral infarction, cardiovascular disease. In the present study, we aimed to identify novel autoantibodies responsible for transient ischemic attack (TIA), a prodromal condition for cerebral infarction. Methods To identify candidate antigens, we screened a human aortic endothelial cell cDNA library using sera from 20 patients with TIA. Serum antibody levels were measured using amplified luminescent proximity homogeneous assay-linked immunosorbent assay (AlphaLISA) in 2 independent patient/healthy donor (HD) cohorts (n = 192 and n = 906 in the second screening and validation cohort, respectively). Results First screening identified 3 candidate antigens. Of these, programmed cell death 11 (PDCD11) was determined to be associated with stroke (p < 0.0001), as evidenced from the second screening using AlphaLISA. The validation cohort revealed significantly higher antibody levels against PDCD11 (PDCD11-Ab levels) in patients with TIA than in HDs. Multivariate logistic regression analysis indicated that the predictive value of PDCD11-Ab levels for TIA [Odds ratio (OR): 2.44, 95% confidence interval (CI): 1.33-4.57, p = 0.0039] was not inferior to other known risk factors for ischemic stroke, including age (OR: 4.97, 95% CI: 2.67-9.48, p < 0.0001); hypertension (OR: 3.21, 95% CI: 1.76-5.86, p = 0.0001); and diabetes (OR: 4.31, 95% CI: 1.74-11.2, p = 0.0015). Conclusion Serum PDCD11-Ab level may serve as a potential biomarker for TIA.
Collapse
Affiliation(s)
- Yoichi Yoshida
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan.,Department of Biochemistry and Genetics, Graduate School of Medicine, Chiba University, Chiba, Japan.,Comprehensive Stroke Center, Chiba University Hospital, Chiba, Japan
| | - Hao Wang
- Department of Biochemistry and Genetics, Graduate School of Medicine, Chiba University, Chiba, Japan.,Department of Anesthesia, The First Affiliated Hospital, Jinan University, Guangzhou, P. R. China
| | - Takaki Hiwasa
- Department of Biochemistry and Genetics, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Toshio Machida
- Department of Neurosurgery, Chiba Cerebral and Cardiovascular Center, Ichihara, Chiba, Japan
| | - Eiichi Kobayashi
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan.,Comprehensive Stroke Center, Chiba University Hospital, Chiba, Japan
| | - Seiichiro Mine
- Department of Neurosurgery, Chiba Cerebral and Cardiovascular Center, Ichihara, Chiba, Japan.,Department of Neurosurgery, Sawara Prefectural Hospital, Chiba, Japan
| | - Go Tomiyoshi
- Department of Biochemistry and Genetics, Graduate School of Medicine, Chiba University, Chiba, Japan.,Medical Project Division, Research Development Center, Fujikura Kasei Co., Saitama, Japan
| | - Rika Nakamura
- Department of Biochemistry and Genetics, Graduate School of Medicine, Chiba University, Chiba, Japan.,Medical Project Division, Research Development Center, Fujikura Kasei Co., Saitama, Japan
| | - Natsuko Shinmen
- Department of Biochemistry and Genetics, Graduate School of Medicine, Chiba University, Chiba, Japan.,Medical Project Division, Research Development Center, Fujikura Kasei Co., Saitama, Japan
| | - Hideyuki Kuroda
- Medical Project Division, Research Development Center, Fujikura Kasei Co., Saitama, Japan
| | - Hirotaka Takizawa
- Port Square Kashiwado Clinic, Kashiwado Memorial Foundation, Chiba, Japan
| | | | - Ikuo Kamitsukasa
- Department of Neurology, Chiba Rosai Hospital, Chiba, Japan.,Department of Neurology, Chibaken Saiseikai Narashino Hospital, Chiba, Japan
| | - Hideo Shin
- Department of Neurosurgery, Higashi Funabashi Hospital, Chiba, Japan
| | - Takeshi Wada
- Department of Internal Medicine, Chiba Aoba Municipal Hospital, Chiba, Japan
| | - Akiyo Aotsuka
- Department of Internal Medicine, Chiba Aoba Municipal Hospital, Chiba, Japan
| | - Eiichiro Nishi
- Department of Pharmacology, Shiga University of Medical Science, Shiga, Japan
| | - Mikiko Ohno
- Department of Pharmacology, Shiga University of Medical Science, Shiga, Japan
| | - Minoru Takemoto
- Department of Clinical Cell Biology and Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Koutaro Yokote
- Department of Clinical Cell Biology and Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Sho Takahashi
- Clinical Research Center, Chiba University Hospital, Chiba, Japan
| | - Jun Matsushima
- Department of Diagnostic Pathology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Xiao-Meng Zhang
- Department of Biochemistry and Genetics, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Masaki Takiguchi
- Department of Biochemistry and Genetics, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Yasuo Iwadate
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| |
Collapse
|
19
|
Ernst D, Widera C, Baerlecken NT, Schlumberger W, Daehnrich C, Schmidt RE, Gabrysch K, Wallentin L, Witte T. Antibodies against MYC-Associated Zinc Finger Protein: An Independent Marker in Acute Coronary Syndrome? Front Immunol 2017; 8:1595. [PMID: 29209328 PMCID: PMC5702292 DOI: 10.3389/fimmu.2017.01595] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2017] [Accepted: 11/06/2017] [Indexed: 12/26/2022] Open
Abstract
Introduction Atherosclerosis is considered the pathophysiology underlying cardiovascular (CVD), cerebrovascular, and peripheral vascular diseases. Evidence supporting an autoimmune component is emerging, with imaging studies correlating MYC-associated zinc finger protein antibody (MAZ-Ab) optical density (OD) with plaque activity. This study compares MAZ-Ab OD on ELISA testing among patients presenting with acute coronary syndromes (ACSs) to healthy controls and investigates the association of MAZ-Ab to traditional CVD risk factors. Methods Patients admitted with ACSs between August 2007 and July 2011 were included. Serum samples taken at presentation were retrospectively tested for MAZ-Ab and compared with serum from healthy volunteers with no CVD risk factors. Large-scale assessment of post-ACS prognostic relevance was performed using the established PLATO cohort. Results In total 174 ACS patients and 96 controls were included. Among ACS patients, median MAZ-Ab OD was higher compared with controls (0.46 vs. 0.27; p = 0.001). Although the majority of ACS patients (116/174; 67%) had suffered from a ST-elevation myocardial infarction, no significant differences in MAZ-Ab titers were evident between ACS subtypes (p = 0.682). No associations between MAZ-Ab OD and conventional CVD risk factors were identified. Large-scale testing revealed no prognostic stratification regarding reinfarction (OR 1.04 [95% CI: 0.94–1.16]; p = 0.436). Conclusion MAZ-Ab OD was higher or all ACS phenotypes compared with controls. Given current understanding of MAZ-Ab function, these findings support an autoimmune component to CVD independent of conventional risk factors and indeed the extent of end-organ damage.
Collapse
Affiliation(s)
- Diana Ernst
- Clinic of Rheumatology and Immunology, Hannover Medical School, Hannover, Germany
| | - Christian Widera
- Department of Cardiology, Heart Center Oldenburg, European Medical School Oldenburg-Groningen, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Niklas T Baerlecken
- Clinic of Rheumatology and Immunology, Hannover Medical School, Hannover, Germany
| | | | | | - Reinhold E Schmidt
- Clinic of Rheumatology and Immunology, Hannover Medical School, Hannover, Germany
| | - Katja Gabrysch
- Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
| | - Lars Wallentin
- Department of Medical Sciences, Cardiology, Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
| | - Torsten Witte
- Clinic of Rheumatology and Immunology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
20
|
Ernst D, Weiberg D, Baerlecken NT, Schlumberger W, Daehnrich C, Schmidt RE, Bengel FM, Derlin T, Witte T. Anti-MYC-associated zinc finger protein antibodies are associated with inflammatory atherosclerotic lesions on 18 F-fluorodeoxyglucose positron emission tomography. Atherosclerosis 2017; 259:12-19. [DOI: 10.1016/j.atherosclerosis.2017.02.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2016] [Revised: 02/09/2017] [Accepted: 02/15/2017] [Indexed: 12/29/2022]
|
21
|
Afanasievа OI, Pylaeva EA, Klesareva EA, Potekhina AV, Provatorov SI, Afanasieva MI, Krasnikova TL, Masenko VP, Arefieva TI, Pokrovsky SN. [Lipoprotein(a), its autoantibodies, and circulating T lymphocyte subpopulations as independent risk factors for coronary artery atherosclerosis]. TERAPEVT ARKH 2017; 88:31-38. [PMID: 27735911 DOI: 10.17116/terarkh201688931-38] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
AIM To study the role of lipoprotein(a) [Lp(a)] as a potential autoantigen causing the activation of immunocompetent cells in atherosclerosis. SUBJECTS AND METHODS A total of 104 men with stable coronary artery (CA) disease and different degrees of progressive coronary atherosclerosis were examined. Clinical blood analysis was carried out and lymphocyte subpopulations (CD4+, Th1, Th17, and Treg) were determined using immunofluorescence and flow cytometry. In addition, the indicators of blood lipid composition, Lp(a), autoantibody (autoAb) titer to Lp(a), and low-density lipoproteins (LDL), and the lymphocyte activation marker sCD25 were also measured. RESULTS The Lp(a) level was shown to predict the severity of CA lesions (β=0.28, p<0.05), regardless of age, the level of cholesterol, different T-lymphocyte subpopulations, sCD25, and autoAb. A combination of the concentration of Lp(a) above 11.8 mg/dl, that of Th17 over 11.4∙103 cells/ml and the reduced levels of regulatory T cells and IL-10-producing CD4+ T cells showed a manifold increase in the risk of severe and progressive CA atherosclerosis. There was a direct correlation of the blood level of Th1 with that of IgG autoAb specific to all atherogenic apoB-containing lipoproteins, including Lp(a). There was an inverse correlations of the lymphocyte activation marker sCD25 with IgM anti-Lp(a) autoAb titers (r=-0.36; p<0.005), but this was less significant with autoAbs to native and oxidized LDL (r=-0.21 and r=-0.24; p<0.05, respectively). CONCLUSION The slightly elevated Lp(a) concentration along with changes in the level of T lymphocyte subpopulations was first shown to significantly potentiate the risk of progressive and multiple CA lesion in the examinees. The correlation of IgM anti-Lp(a) autoAb with the lymphocyte activation marker sCD25 and that of IgG anti-Lp(a) autoAb with Th1 have demonstrated that Lp(a) is involved in the autoimmune inflammatory processes in atherosclerosis.
Collapse
Affiliation(s)
- O I Afanasievа
- Russian Cardiology Research and Production Complex, Ministry of Health of Russia, Moscow, Russia
| | - E A Pylaeva
- Russian Cardiology Research and Production Complex, Ministry of Health of Russia, Moscow, Russia
| | - E A Klesareva
- Russian Cardiology Research and Production Complex, Ministry of Health of Russia, Moscow, Russia
| | - A V Potekhina
- Russian Cardiology Research and Production Complex, Ministry of Health of Russia, Moscow, Russia
| | - S I Provatorov
- Russian Cardiology Research and Production Complex, Ministry of Health of Russia, Moscow, Russia
| | - M I Afanasieva
- Russian Cardiology Research and Production Complex, Ministry of Health of Russia, Moscow, Russia
| | - T L Krasnikova
- Russian Cardiology Research and Production Complex, Ministry of Health of Russia, Moscow, Russia
| | - V P Masenko
- Russian Cardiology Research and Production Complex, Ministry of Health of Russia, Moscow, Russia
| | - T I Arefieva
- Russian Cardiology Research and Production Complex, Ministry of Health of Russia, Moscow, Russia
| | - S N Pokrovsky
- Russian Cardiology Research and Production Complex, Ministry of Health of Russia, Moscow, Russia
| |
Collapse
|
22
|
Sage AP, Mallat Z. Readapting the adaptive immune response - therapeutic strategies for atherosclerosis. Br J Pharmacol 2017; 174:3926-3939. [PMID: 28052311 DOI: 10.1111/bph.13700] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 12/13/2016] [Accepted: 12/20/2016] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular diseases remain a major global health issue, with the development of atherosclerosis as a major underlying cause. Our treatment of cardiovascular disease has improved greatly over the past three decades, but much remains to be done reduce disease burden. Current priorities include reducing atherosclerosis advancement to clinically significant stages and preventing plaque rupture or erosion. Inflammation and involvement of the adaptive immune system influences all these aspects and therefore is one focus for future therapeutic development. The atherosclerotic vascular wall is now recognized to be invaded from both sides (arterial lumen and adventitia), for better or worse, by the adaptive immune system. Atherosclerosis is also affected at several stages by adaptive immune responses, overall providing many opportunities to target these responses and to reduce disease progression. Protective influences that may be defective in diseased individuals include humoral responses to modified LDL and regulatory T cell responses. There are many strategies in development to boost these pathways in humans, including vaccine-based therapies. The effects of various existing adaptive immune targeting therapies, such as blocking critical co-stimulatory pathways or B cell depletion, on cardiovascular disease are beginning to emerge with important consequences for both autoimmune disease patients and the potential for wider use of such therapies. Entering the translation phase for adaptive immune targeting therapies is an exciting and promising prospect. LINKED ARTICLES This article is part of a themed section on Targeting Inflammation to Reduce Cardiovascular Disease Risk. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.22/issuetoc and http://onlinelibrary.wiley.com/doi/10.1111/bcp.v82.4/issuetoc.
Collapse
Affiliation(s)
- Andrew P Sage
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Ziad Mallat
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, UK.,INSERM U970, Paris Cardiovascular Research Center, Paris, France, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
23
|
Medin CL, Rothman AL. Zika Virus: The Agent and Its Biology, With Relevance to Pathology. Arch Pathol Lab Med 2016; 141:33-42. [PMID: 27763795 DOI: 10.5858/arpa.2016-0409-ra] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Once obscure, Zika virus (ZIKV) has attracted significant medical and scientific attention in the past year because of large outbreaks associated with the recent introduction of this virus into the Western hemisphere. In particular, the occurrence of severe congenital infections and cases of Guillain-Barré syndrome has placed this virus squarely in the eyes of clinical and anatomic pathologists. This review article provides a basic introduction to ZIKV, its genetics, its structural characteristics, and its biology. A multidisciplinary effort will be essential to establish clinicopathologic correlations of the basic virology of ZIKV in order to advance development of diagnostics, therapeutics, and vaccines.
Collapse
Affiliation(s)
| | - Alan L Rothman
- From the Institute for Immunology and Informatics, Department of Cell and Molecular Biology, University of Rhode Island, Providence. Drs Medin and Rothman both contributed equally to the manuscript
| |
Collapse
|
24
|
Antibodies to paraoxonase 1 are associated with oxidant status and endothelial activation in rheumatoid arthritis. Clin Sci (Lond) 2016; 130:1889-99. [DOI: 10.1042/cs20160374] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 08/12/2016] [Indexed: 02/04/2023]
Abstract
Anti-paraoxonase 1 (PON1) antibodies could be a potential missing link between oxidative status, inflammation and cardiovascular disease (CVD) in rheumatoid arthritis (RA) patients. Therefore, they could represent an emerging clinical biomarker of CV risk in this condition.
Collapse
|
25
|
Carbone F, Satta N, Montecucco F, Virzi J, Burger F, Roth A, Roversi G, Tamborino C, Casetta I, Seraceni S, Trentini A, Padroni M, Dallegri F, Lalive PH, Mach F, Fainardi E, Vuilleumier N. Anti-ApoA-1 IgG serum levels predict worse poststroke outcomes. Eur J Clin Invest 2016; 46:805-17. [PMID: 27490973 DOI: 10.1111/eci.12664] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 08/01/2016] [Indexed: 12/17/2022]
Abstract
BACKGROUND Autoantibodies to apolipoprotein A-1 (anti-ApoA-1 IgG) were shown to predict major adverse cardiovascular events and promote atherogenesis. However, their potential relationship with clinical disability and ischaemic lesion volume after acute ischaemic stroke (AIS) remains unexplored. MATERIALS AND METHODS We included n = 76 patients admitted for AIS and we investigated whether baseline serum anti-ApoA-1 IgG levels could predict (i) AIS-induced clinical disability [assessed by the modified Rankin Scale (mRS)], and (ii) AIS-related ischaemic lesion volume [assessed by Computed Tomography (CT)]. We also evaluated the possible pro-apoptotic and pro-necrotic effects of anti-ApoA-1 IgG on human astrocytoma cell line (U251) using flow cytometry. RESULTS High levels of anti-ApoA-1 IgG were retrieved in 15·8% (12/76) of patients. Increased baseline levels of anti-ApoA-1 IgG were independently correlated with worse mRS [β = 0·364; P = 0·002; adjusted odds ratio (OR): 1·05 (95% CI 1·01-1·09); P = 0·017] and CT-assessed ischaemic lesion volume [β = 0·333; P < 0·001; adjusted OR: 1·06 (95% CI 1·01-1·12); P = 0·048] at 3 months. No difference in baseline clinical, biochemical and radiological characteristics was observed between patients with high vs. low levels of anti-ApoA-1 IgG. Incubating human astrocytoma cells with anti-ApoA-1 IgG dose dependently induced necrosis and apoptosis of U251 cells in vitro. CONCLUSION Anti-ApoA-1 IgG serum levels at AIS onset are associated with poorer clinical recovery and worse brain lesion volume 3 months after AIS. These observations could be partly explained by the deleterious effect of anti-ApoA-1 IgG on human brain cell survival in vitro and may have clinical implication in the prediction of poor outcome in AIS.
Collapse
Affiliation(s)
- Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, School of Medicine, University of Genoa, Genoa, Italy
| | - Nathalie Satta
- Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, School of Medicine, University of Genoa, Genoa, Italy.,IRCCS AOU San Martino - IST, Genoa, Italy.,Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy
| | - Julien Virzi
- Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Fabienne Burger
- Division of Cardiology, Department of Medical Specialties, Foundation for Medical Researches, University of Geneva, Geneva, Switzerland
| | - Aline Roth
- Division of Cardiology, Department of Medical Specialties, Foundation for Medical Researches, University of Geneva, Geneva, Switzerland
| | - Gloria Roversi
- Department of Biological, Psychiatric and Psychological Science, Azienda Ospedaliera-Universitaria, Arcispedale S. Anna, Ferrara, Italy
| | - Carmine Tamborino
- Department of Biological, Psychiatric and Psychological Science, Azienda Ospedaliera-Universitaria, Arcispedale S. Anna, Ferrara, Italy
| | - Ilaria Casetta
- Department of Biological, Psychiatric and Psychological Science, Azienda Ospedaliera-Universitaria, Arcispedale S. Anna, Ferrara, Italy
| | - Silva Seraceni
- Institute for Maternal and Child Health 'IRCCS Burlo Garofolo', Trieste, Italy
| | - Alessandro Trentini
- Section of Medical Biochemistry, Molecular Biology and Genetics, Department of Biomedical and Specialist Surgical Sciences, University of Ferrara, Ferrara, Italy
| | - Marina Padroni
- Section of Medical Biochemistry, Molecular Biology and Genetics, Department of Biomedical and Specialist Surgical Sciences, University of Ferrara, Ferrara, Italy
| | - Franco Dallegri
- First Clinic of Internal Medicine, Department of Internal Medicine, School of Medicine, University of Genoa, Genoa, Italy.,IRCCS AOU San Martino - IST, Genoa, Italy
| | - Patrice H Lalive
- Division of Neurology, Department of Clinical Neurosciences, Geneva University Hospital, Geneva, Switzerland
| | - François Mach
- Division of Cardiology, Department of Medical Specialties, Foundation for Medical Researches, University of Geneva, Geneva, Switzerland
| | - Enrico Fainardi
- Neuroradiology Unit, Department of Neurosciences and Rehabilitation, Azienda Ospedaliera-Universitaria, Arcispedale S. Anna, Ferrara, Italy
| | - Nicolas Vuilleumier
- Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals, Geneva, Switzerland
| |
Collapse
|
26
|
Chistiakov DA, Orekhov AN, Bobryshev YV. ApoA1 and ApoA1-specific self-antibodies in cardiovascular disease. J Transl Med 2016; 96:708-18. [PMID: 27183204 DOI: 10.1038/labinvest.2016.56] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Revised: 03/21/2016] [Accepted: 04/03/2016] [Indexed: 12/15/2022] Open
Abstract
Apolipoprotein A1 (ApoA1) is a main protein moiety in high-density lipoprotein (HDL) particles. Generally, ApoA1 and HDL are considered as atheroprotective. In prooxidant and inflammatory microenvironment in the vicinity to the atherosclerotic lesion, ApoA1/HDL are subjected to modification. The chemical modifications such as oxidation, nitration, etc result in altering native architecture of ApoA1 toward dysfunctionality and abnormality. Neutrophil myeloperoxidase has a prominent role in this mechanism. Neo-epitopes could be formed and then exposed that makes them immunogenic. Indeed, these epitopes may be recognized by immune cells and induce production of proatherogenic ApoA1-specific IgG antibodies. These antibodies are biologically relevant because they are able to react with Toll-like receptor (TLR)-2 and TLR4 in target cells and induce a variety of pro-inflammatory responses. Epidemiological and functional studies underline a prognostic value of ApoA1 self-antibodies for several cardiovascular diseases, including myocardial infarction, acute coronary syndrome, and severe carotid stenosis.
Collapse
Affiliation(s)
- Dimitry A Chistiakov
- Department of Molecular Genetic Diagnostics and Cell Biology, Division of Laboratory Medicine, Institute of Pediatrics, Research Center for Children's Health, Moscow, Russia
| | - Alexander N Orekhov
- Institute of General Pathology and Pathophysiology, Russian Academy of Sciences, Moscow, Russia.,Faculty of Biology, Department of Biophysics, Lomonosov Moscow State University, Moscow, Russia
| | - Yuri V Bobryshev
- Institute of General Pathology and Pathophysiology, Russian Academy of Sciences, Moscow, Russia.,Faculty of Medicine, School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia.,School of Medicine, University of Western Sydney, Campbelltown, NSW, Australia
| |
Collapse
|
27
|
Rodríguez-Carrio J, López-Mejías R, Alperi-López M, López P, Ballina-García FJ, González-Gay MÁ, Suárez A. Paraoxonase 1 Activity Is Modulated by the rs662 Polymorphism and IgG Anti-High-Density Lipoprotein Antibodies in Patients With Rheumatoid Arthritis: Potential Implications for Cardiovascular Disease. Arthritis Rheumatol 2016; 68:1367-76. [DOI: 10.1002/art.39609] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 01/19/2016] [Indexed: 01/06/2023]
Affiliation(s)
| | | | | | | | | | - Miguel Á. González-Gay
- Hospital Universitario Marqués de Valdecilla, Santander, Spain, and Clinical University Hospital of Santiago de Compostela; Santiago de Compostela Spain
| | | |
Collapse
|
28
|
Montecucco F, Carbone F, Schindler TH. Pathophysiology of ST-segment elevation myocardial infarction: novel mechanisms and treatments. Eur Heart J 2016; 37:1268-1283. [PMID: 26543047 DOI: 10.1093/eurheartj/ehv592] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/29/2023] Open
|
29
|
Hiwasa T, Zhang XM, Kimura R, Ohno M, Chen PM, Nishi E, Ono K, Kimura T, Kamitsukasa I, Wada T, Aotsuka A, Mine S, Takizawa H, Kashiwado K, Takemoto M, Kobayashi K, Kawamura H, Ishibashi R, Yokote K, Nakamura R, Tomiyoshi G, Shinmen N, Kuroda H. Elevated Adiponectin Antibody Levels in Sera of Patients with Atherosclerosis-Related Coronary Artery Disease, Cerebral Infarction and Diabetes Mellitus. J Circ Biomark 2016; 5:8. [PMID: 28936256 PMCID: PMC5548317 DOI: 10.5772/63218] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 03/15/2016] [Indexed: 12/15/2022] Open
Abstract
Adiponectin secreted from the adipocytes plays pleiotropic, anti-atherosclerotic roles, such as enhancement of insulin secretion and an increase in energy expenditure. The measurement of levels of circulating adiponectin is useful to evaluate the progression of atherosclerosis-related diseases, such as coronary artery disease (CAD), cerebral infarction (CI) and diabetes mellitus (DM). We examined the serum antibody levels against recombinant adiponectin protein via the amplified luminescent proximity homogeneous assay-linked immunosorbent assay (AlphaLISA) method. The results revealed that the antibody levels were significantly higher in patients with CAD, CI and type 2 DM, than in healthy donors. Receiver operating curve analysis showed that the sensitivity was in a range of 41–48% for CAD, CI and DM. Thus, the serum anti-adiponectin antibody levels could be a common marker for atherosclerosis-related diseases.
Collapse
Affiliation(s)
- Takaki Hiwasa
- Department of Biochemistry and Genetics, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Xiao-Meng Zhang
- Department of Biochemistry and Genetics, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Risa Kimura
- Department of Biochemistry and Genetics, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Mikiko Ohno
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Po-Min Chen
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Eiichiro Nishi
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Koh Ono
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takeshi Kimura
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | | - Takeshi Wada
- Department of Internal Medicine, Chiba Aoba Municipal Hospital, Chiba, Japan
| | - Akiyo Aotsuka
- Department of Internal Medicine, Chiba Aoba Municipal Hospital, Chiba, Japan
| | - Seiichiro Mine
- Department of Neurological Surgery, Chiba Prefectural Sawara Hospital, Chiba, Japan.,Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Hirotaka Takizawa
- Port Square Kashiwado Clinic, Kashiwado Memorial Foundation, Chiba, Japan
| | | | - Minoru Takemoto
- Department of Clinical Cell Biology and Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Kazuki Kobayashi
- Department of Clinical Cell Biology and Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Harukiyo Kawamura
- Department of Clinical Cell Biology and Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Ryoichi Ishibashi
- Department of Clinical Cell Biology and Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Koutaro Yokote
- Department of Clinical Cell Biology and Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Rika Nakamura
- Department of Biochemistry and Genetics, Graduate School of Medicine, Chiba University, Chiba, Japan.,Medical Project Division, Research Development Center, Fujikura Kasei Co., Saitama, Japan
| | - Go Tomiyoshi
- Department of Biochemistry and Genetics, Graduate School of Medicine, Chiba University, Chiba, Japan.,Medical Project Division, Research Development Center, Fujikura Kasei Co., Saitama, Japan
| | - Natsuko Shinmen
- Department of Biochemistry and Genetics, Graduate School of Medicine, Chiba University, Chiba, Japan.,Medical Project Division, Research Development Center, Fujikura Kasei Co., Saitama, Japan
| | - Hideyuki Kuroda
- Medical Project Division, Research Development Center, Fujikura Kasei Co., Saitama, Japan
| |
Collapse
|
30
|
Rodríguez-Carrio J, Alperi-López M, López P, Ballina-García FJ, Abal F, Suárez A. Antibodies to high-density lipoproteins are associated with inflammation and cardiovascular disease in rheumatoid arthritis patients. Transl Res 2015; 166:529-39. [PMID: 26279255 DOI: 10.1016/j.trsl.2015.07.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 07/01/2015] [Accepted: 07/23/2015] [Indexed: 11/17/2022]
Abstract
Several lines of evidence suggest that chronic inflammation and immune dysregulation are related to altered lipid profiles in rheumatoid arthritis (RA), but the actual mechanisms are still unclear. We wondered whether the development of antibodies against high-density lipoprotein (HDL) can be found in RA patients linked to clinical and cardiovascular (CV) risk factors. To this end, immunoglobulin G (IgG) anti-HDL antibodies and total IgG serum levels were quantified in 212 RA patients, 131 sex- and age-matched healthy controls (HC), and 52 subjects with traditional CV risk factors (tCVRs). A subgroup of 13 RA patients was prospectively followed on TNFα-blockade. TNFα, interferon (IFN)α, MIP1α, IFNγ, IL-8, VEGF, GM-CSF, IL-17, MCP-1, SDF-1α, resistin, and leptin serum levels were quantified by immunoassays. IgG anti-HDL levels were higher in RA patients compared with HC (P < 0.0001) and tCVR subjects (P = 0.015). Differences with HC remained after correction for total IgG levels (P < 0.003). Anti-HDL/IgG were negatively associated with HDL levels in RA (-1.182 [-1.823 to -0.541], P = 0.0003) after adjusting for demographical, clinical, inflammatory parameters, and treatments. RA patients with high levels of anti-HDL/IgG (n = 40, 18.8%) were more likely to have experienced a CV event (P < 0.0001) and exhibited increased levels of several proinflammatory mediators (C-reactive protein, IFNα, MIP1α, IFNγ, IL-8, GM-CSF, IL-17 and MCP-1). Finally, change in anti-HDL antibodies on TNFα-blockade was independently associated with increasing HDL levels. Overall, IgG anti-HDL antibodies are increased in RA independently of tCVRs and associated with a proinflammatory milieu and impaired lipid blood profile, which may contribute to the increased rate of CV events in these patients.
Collapse
Affiliation(s)
- Javier Rodríguez-Carrio
- Area of Immunology, Department of Functional Biology, University of Oviedo, Oviedo, Asturias, Spain
| | - Mercedes Alperi-López
- Department of Rheumatology, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Patricia López
- Area of Immunology, Department of Functional Biology, University of Oviedo, Oviedo, Asturias, Spain
| | | | - Francisco Abal
- Centro de Salud Sariego, Servicio de Salud del Principado de Asturias, Pola de Siero, Spain
| | - Ana Suárez
- Area of Immunology, Department of Functional Biology, University of Oviedo, Oviedo, Asturias, Spain.
| |
Collapse
|
31
|
Carbone F, Teixeira PC, Braunersreuther V, Mach F, Vuilleumier N, Montecucco F. Pathophysiology and Treatments of Oxidative Injury in Ischemic Stroke: Focus on the Phagocytic NADPH Oxidase 2. Antioxid Redox Signal 2015; 23:460-89. [PMID: 24635113 PMCID: PMC4545676 DOI: 10.1089/ars.2013.5778] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
SIGNIFICANCE Phagocytes play a key role in promoting the oxidative stress after ischemic stroke occurrence. The phagocytic NADPH oxidase (NOX) 2 is a membrane-bound enzyme complex involved in the antimicrobial respiratory burst and free radical production in these cells. RECENT ADVANCES Different oxidants have been shown to induce opposite effects on neuronal homeostasis after a stroke. However, several experimental models support the detrimental effects of NOX activity (especially the phagocytic isoform) on brain recovery after stroke. Therapeutic strategies selectively targeting the neurotoxic ROS and increasing neuroprotective oxidants have recently produced promising results. CRITICAL ISSUES NOX2 might promote carotid plaque rupture and stroke occurrence. In addition, NOX2-derived reactive oxygen species (ROS) released by resident and recruited phagocytes enhance cerebral ischemic injury, activating the inflammatory apoptotic pathways. The aim of this review is to update evidence on phagocyte-related oxidative stress, focusing on the role of NOX2 as a potential therapeutic target to reduce ROS-related cerebral injury after stroke. FUTURE DIRECTIONS Radical scavenger compounds (such as Ebselen and Edaravone) are under clinical investigation as a therapeutic approach against stroke. On the other hand, NOX inhibition might represent a promising strategy to prevent the stroke-related injury. Although selective NOX inhibitors are not yet available, nonselective compounds (such as apocynin and fasudil) provided encouraging results in preclinical studies. Whereas additional studies are needed to better evaluate this therapeutic potential in human beings, the development of specific NOX inhibitors (such as monoclonal antibodies, small-molecule inhibitors, or aptamers) might further improve brain recovery after stroke.
Collapse
Affiliation(s)
- Federico Carbone
- 1 Division of Cardiology, Foundation for Medical Researches, Department of Medical Specialties, University of Geneva , Geneva, Switzerland .,2 Department of Internal Medicine, University of Genoa School of Medicine , IRCCS Azienda Ospedaliera Universitaria San Martino-IST Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| | - Priscila Camillo Teixeira
- 3 Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals , Geneva, Switzerland
| | - Vincent Braunersreuther
- 1 Division of Cardiology, Foundation for Medical Researches, Department of Medical Specialties, University of Geneva , Geneva, Switzerland
| | - François Mach
- 1 Division of Cardiology, Foundation for Medical Researches, Department of Medical Specialties, University of Geneva , Geneva, Switzerland
| | - Nicolas Vuilleumier
- 3 Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals , Geneva, Switzerland
| | - Fabrizio Montecucco
- 1 Division of Cardiology, Foundation for Medical Researches, Department of Medical Specialties, University of Geneva , Geneva, Switzerland .,2 Department of Internal Medicine, University of Genoa School of Medicine , IRCCS Azienda Ospedaliera Universitaria San Martino-IST Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy .,3 Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals , Geneva, Switzerland
| |
Collapse
|
32
|
Machida T, Kubota M, Kobayashi E, Iwadate Y, Saeki N, Yamaura A, Nomura F, Takiguchi M, Hiwasa T. Identification of stroke-associated-antigens via screening of recombinant proteins from the human expression cDNA library (SEREX). J Transl Med 2015; 13:71. [PMID: 25890248 PMCID: PMC4344740 DOI: 10.1186/s12967-015-0393-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 01/13/2015] [Indexed: 12/24/2022] Open
Abstract
Background Because circulating antibodies against a variety of antigens have been detected in patients with coronary heart disease, carotid atherosclerosis and those who have suffered a stroke, it is suspected that immune response may be one of the mechanisms of atherogenesis The objective of this study is to identify novel antibodies in ischemic stroke patients by screening the expressed recombinant proteins using a human cDNA library (SEREX). Methods To identify the candidate antigens, cDNA library was screened by SEREX using plasma from ten patients with ischemic stroke. Subsequently, via ELISA using recombinant proteins and synthetic peptides, the serum antibody levels were measured in two independent patient/healthy donor (HD) cohorts (142 and 78 in the 2nd screening and a validation cohort, respectively). Results The initial screening resulted in the identification of six candidate antigens. Of these antigens, replication protein A2 (RPA2) was determined to be the antigen associated with stroke (P < 0.05) by ELISA with 2nd screening and validation cohort. Multifactorial logistic regression analysis showed that the increased levels of the RPA2 antibodies (RPA2-Abs) associated with stroke independent of other risk factors for stroke (P < 0.05). Receiver operating curve analysis demonstrated that the area under the curve from ELISA using GST fusion RPA2 and synthetic peptides (bRPA2-132) were 0.867 (95% CI: 0.798-0.936) and 0.971 (95% CI: 0.940-1.00), respectively. If the cut-off value of the bRPA2-132-Ab level was determined to be 0.334, the sensitivity and specificity of the antibody level as the diagnostic marker for stroke were 0.323 (95% CI: 0.209-0.453) and 1.00 (95% CI: 0.713-1.00), respectively. Conclusions SEREX identified RPA2 as the antigen associated with ischemic stroke and serum auto-antibodies against RPA2 elevates in stroke patients. RPA2-Abs could become a biomarker for the evaluation of ischemic stroke at risk.
Collapse
Affiliation(s)
- Toshio Machida
- Departments of Neurosurgery, Chiba Cardiovascular Center, Ichihara, Chiba, Japan.
| | - Motoo Kubota
- Department of Neurosurgery, Kameda Medical Center, Chiba, Japan.
| | - Eiichi Kobayashi
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan.
| | - Yasuo Iwadate
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan.
| | - Naokatsu Saeki
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan.
| | - Akira Yamaura
- Chiba Prefectural University of Health Sciences, Chiba, Japan.
| | - Fumio Nomura
- Department of Molecular Diagnosis, Graduate School of Medicine, Chiba University, Chuo-ku, Chiba, 260-8670, Japan.
| | - Masaki Takiguchi
- Department of Biochemistry and Genetics, Graduate School of Medicine, Chiba University, Chiba, Japan.
| | - Takaki Hiwasa
- Department of Biochemistry and Genetics, Graduate School of Medicine, Chiba University, Chiba, Japan.
| |
Collapse
|
33
|
Teixeira PC, Ferber P, Vuilleumier N, Cutler P. Biomarkers for cardiovascular risk assessment in autoimmune diseases. Proteomics Clin Appl 2015; 9:48-57. [DOI: 10.1002/prca.201400125] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 11/30/2014] [Accepted: 12/15/2014] [Indexed: 12/20/2022]
Affiliation(s)
- Priscila Camillo Teixeira
- Pharma Research and Early Development; Roche Innovation Center Basel; Basel; Switzerland
- Division of Laboratory Medicine; Department of Genetics and Laboratory Medicine; Geneva University Hospitals; Geneva; Switzerland
| | - Philippe Ferber
- Pharma Research and Early Development; Roche Innovation Center Basel; Basel; Switzerland
| | - Nicolas Vuilleumier
- Division of Laboratory Medicine; Department of Genetics and Laboratory Medicine; Geneva University Hospitals; Geneva; Switzerland
| | - Paul Cutler
- Pharma Research and Early Development; Roche Innovation Center Basel; Basel; Switzerland
| |
Collapse
|
34
|
Carbone F, Montecucco F. Inflammation in arterial diseases. IUBMB Life 2015; 67:18-28. [PMID: 25631520 DOI: 10.1002/iub.1344] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 12/28/2014] [Indexed: 12/26/2022]
Affiliation(s)
- Federico Carbone
- First Clinic of Internal Medicine; Department of Internal Medicine; University of Genoa School of Medicine, IRCCS Azienda Ospedaliera Universitaria San Martino-IST Istituto Nazionale per la Ricerca sul Cancro; Genoa Italy
- Division of Cardiology; Foundation for Medical Researches; Department of Medical Specialties; University of Geneva; Geneva Switzerland
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine; Department of Internal Medicine; University of Genoa School of Medicine, IRCCS Azienda Ospedaliera Universitaria San Martino-IST Istituto Nazionale per la Ricerca sul Cancro; Genoa Italy
- Division of Cardiology; Foundation for Medical Researches; Department of Medical Specialties; University of Geneva; Geneva Switzerland
- Division of Laboratory Medicine; Department of Genetics and Laboratory Medicine; Geneva University Hospitals; Geneva Switzerland
| |
Collapse
|
35
|
Montecucco F, Favari E, Norata GD, Ronda N, Nofer JR, Vuilleumier N. Impact of systemic inflammation and autoimmune diseases on apoA-I and HDL plasma levels and functions. Handb Exp Pharmacol 2015; 224:455-82. [PMID: 25522998 DOI: 10.1007/978-3-319-09665-0_14] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The cholesterol of high-density lipoproteins (HDLs) and its major proteic component, apoA-I, have been widely investigated as potential predictors of acute cardiovascular (CV) events. In particular, HDL cholesterol levels were shown to be inversely and independently associated with the risk of acute CV diseases in different patient populations, including autoimmune and chronic inflammatory disorders. Some relevant and direct anti-inflammatory activities of HDL have been also recently identified targeting both immune and vascular cell subsets. These studies recently highlighted the improvement of HDL function (instead of circulating levels) as a promising treatment strategy to reduce inflammation and associated CV risk in several diseases, such as systemic lupus erythematosus and rheumatoid arthritis. In these diseases, anti-inflammatory treatments targeting HDL function might improve both disease activity and CV risk. In this narrative review, we will focus on the pathophysiological relevance of HDL and apoA-I levels/functions in different acute and chronic inflammatory pathophysiological conditions.
Collapse
Affiliation(s)
- Fabrizio Montecucco
- Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals, 4 rue Gabrielle Perret-Gentil, 1211, Geneva, Switzerland
| | | | | | | | | | | |
Collapse
|
36
|
Abstract
Atherosclerosis is a degenerative inflammatory disease of the vascular wall, which is characterized by the formation of atherosclerotic plaques that contain lipids, activated smooth muscle cells, immune cells, foam cells, a necrotic core and calcified sites. In atherosclerosis pathology, monocytes and macrophages play the most important role by accumulating redundant LDL particles in their oxidized form and producing proinflammatory cytokines. Atherosclerotic plaque macrophages reveal distinct phenotypes that are distinguished into M1 (pro-inflammatory) and M2 (anti-inflammatory) macrophages. Numerous environmental signals (cytokines, microbial cell molecules) that are received by macrophages drive their polarization, but it must be determined whether this classification reflects different macrophage subtypes or plasticity and phenotypic tissue changes, but the balance between subsets is crucial. M1 macrophages are dominant in symptomatic atherosclerotic plaques, while M2 macrophages are more frequent in asymptomatic plaques. Nevertheless, a positive correlation of both M1 and M2 macrophages with atherosclerotic lesion severity was also observed.
Collapse
Affiliation(s)
- A. KRÁLOVÁ
- Laboratory for Atherosclerosis Research, Centre of Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | | | | |
Collapse
|
37
|
Teixeira PC, Ducret A, Ferber P, Gaertner H, Hartley O, Pagano S, Butterfield M, Langen H, Vuilleumier N, Cutler P. Definition of human apolipoprotein A-I epitopes recognized by autoantibodies present in patients with cardiovascular diseases. J Biol Chem 2014; 289:28249-59. [PMID: 25170076 PMCID: PMC4192480 DOI: 10.1074/jbc.m114.589002] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Autoantibodies to apolipoprotein A-I (anti-apoA-I IgG) have been shown to be both markers and mediators of cardiovascular disease, promoting atherogenesis and unstable atherosclerotic plaque. Previous studies have shown that high levels of anti-apoA-I IgGs are independently associated with major adverse cardiovascular events in patients with myocardial infarction. Autoantibody responses to apoA-I can be polyclonal and it is likely that more than one epitope may exist. To identify the specific immunoreactive peptides in apoA-I, we have developed a set of methodologies and procedures to isolate, purify, and identify novel apoA-I endogenous epitopes. First, we generated high purity apoA-I from human plasma, using thiophilic interaction chromatography followed by enzymatic digestion specifically at lysine or arginine residues. Immunoreactivity to the different peptides generated was tested by ELISA using serum obtained from patients with acute myocardial infarction and high titers of autoantibodies to native apoA-I. The immunoreactive peptides were further sequenced by mass spectrometry. Our approach successfully identified two novel immunoreactive peptides, recognized by autoantibodies from patients suffering from myocardial infarction, who contain a high titer of anti-apoA-I IgG. The discovery of these epitopes may open innovative prognostic and therapeutic opportunities potentially suitable to improve current cardiovascular risk stratification.
Collapse
Affiliation(s)
- Priscila Camillo Teixeira
- From the Pharma Research and Early Development, Roche Innovation Center, 4070 Basel, the Department of Genetics and Laboratory Medicine, Division of Laboratory Medicine, 1205 Geneva University Hospitals, 1205 Geneva, and
| | - Axel Ducret
- From the Pharma Research and Early Development, Roche Innovation Center, 4070 Basel
| | - Philippe Ferber
- From the Pharma Research and Early Development, Roche Innovation Center, 4070 Basel
| | - Hubert Gaertner
- the Department of Immunopathology, Faculty of Medicine, University of Geneva, 1205 Geneva, Switzerland
| | - Oliver Hartley
- the Department of Immunopathology, Faculty of Medicine, University of Geneva, 1205 Geneva, Switzerland
| | - Sabrina Pagano
- the Department of Genetics and Laboratory Medicine, Division of Laboratory Medicine, 1205 Geneva University Hospitals, 1205 Geneva, and
| | - Michelle Butterfield
- From the Pharma Research and Early Development, Roche Innovation Center, 4070 Basel
| | - Hanno Langen
- From the Pharma Research and Early Development, Roche Innovation Center, 4070 Basel
| | - Nicolas Vuilleumier
- the Department of Genetics and Laboratory Medicine, Division of Laboratory Medicine, 1205 Geneva University Hospitals, 1205 Geneva, and
| | - Paul Cutler
- From the Pharma Research and Early Development, Roche Innovation Center, 4070 Basel
| |
Collapse
|
38
|
Fuller JP, Stavenhagen JB, Teeling JL. New roles for Fc receptors in neurodegeneration-the impact on Immunotherapy for Alzheimer's Disease. Front Neurosci 2014; 8:235. [PMID: 25191216 PMCID: PMC4139653 DOI: 10.3389/fnins.2014.00235] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 07/17/2014] [Indexed: 12/11/2022] Open
Abstract
There are an estimated 18 million Alzheimer's disease (AD) sufferers worldwide and with no disease modifying treatment currently available, development of new therapies represents an enormous unmet clinical need. AD is characterized by episodic memory loss followed by severe cognitive decline and is associated with many neuropathological changes. AD is characterized by deposits of amyloid beta (Aβ), neurofibrillary tangles, and neuroinflammation. Active immunization or passive immunization against Aβ leads to the clearance of deposits in transgenic mice expressing human Aβ. This clearance is associated with reversal of associated cognitive deficits, but these results have not translated to humans, with both active and passive immunotherapy failing to improve memory loss. One explanation for these observations is that certain anti-Aβ antibodies mediate damage to the cerebral vasculature limiting the top dose and potentially reducing efficacy. Fc gamma receptors (FcγR) are a family of immunoglobulin-like receptors which bind to the Fc portion of IgG, and mediate the response of effector cells to immune complexes. Data from both mouse and human studies suggest that cross-linking FcγR by therapeutic antibodies and the subsequent pro-inflammatory response mediates the vascular side effects seen following immunotherapy. Increasing evidence is emerging that FcγR expression on CNS resident cells, including microglia and neurons, is increased during aging and functionally involved in the pathogenesis of age-related neurodegenerative diseases. Therefore, we propose that increased expression and ligation of FcγR in the CNS, either by endogenous IgG or therapeutic antibodies, has the potential to induce vascular damage and exacerbate neurodegeneration. To produce safe and effective immunotherapies for AD and other neurodegenerative diseases it will be vital to understand the role of FcγR in the healthy and diseased brain. Here we review the literature on FcγR expression, function and proposed roles in multiple age-related neurological diseases. Lessons can be learnt from therapeutic antibodies used for the treatment of cancer where antibodies have been engineered for optimal efficacy.
Collapse
Affiliation(s)
- James P. Fuller
- CNS Inflammation Group, Centre for Biological Sciences, University of SouthamptonSouthampton, UK
| | | | - Jessica L. Teeling
- CNS Inflammation Group, Centre for Biological Sciences, University of SouthamptonSouthampton, UK
| |
Collapse
|
39
|
Abstract
The development of atherosclerosis is the major etiological factor causing cardiovascular disease and constitutes a lipid-induced, chronic inflammatory and autoimmune disease of the large arteries. A long-standing view of the protective role of B cells in atherosclerosis has been challenged by recent studies using B cell depletion in animal models. Whereas complete B cell deficiency increases atherosclerosis, depletion of B2 but not B1 cells reduces atherosclerosis. This has led to a re-evaluation of the multiple potential pathways by which B cells can regulate atherosclerosis, and the apparent opposing roles of B1 and B2 cells. B cells, in addition to having the unique ability to produce antibodies, are now recognized to play a number of important roles in the immune system, including cytokine production and direct regulation of T cell responses. This review summarizes current knowledge on B cell subsets and functions, and how these could distinctly influence atherosclerosis development.
Collapse
Affiliation(s)
- Andrew P Sage
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge , Cambridge , UK
| | | |
Collapse
|
40
|
Vuilleumier N, Montecucco F, Hartley O. Autoantibodies to apolipoprotein A-1 as a biomarker of cardiovascular autoimmunity. World J Cardiol 2014; 6:314-326. [PMID: 24944761 PMCID: PMC4062126 DOI: 10.4330/wjc.v6.i5.314] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Accepted: 03/18/2014] [Indexed: 02/06/2023] Open
Abstract
Immune-driven inflammation plays an important part in atherogenesis and is therefore believed to be key to the development of cardiovascular disease (CVD), which is currently the leading cause of death in the Western world. By fulfilling some of the Koch postulates, atherogenesis has even been proposed to be considered as an autoimmune disease, raising the hope that CVD could be prevented by immunomodulation. Nevertheless, the role of the immune system and autoimmune reactions in atherosclerosis appear to be a double edged-sword, with both pro-atherogenic and anti-atherogenic attributes. Hence, if immunomodulation is to become a therapeutic option for atherosclerosis and CVD, it will be crucial to correctly identify patients who might benefit from targeted suppression of deleterious autoimmune responses. This could be achieved, for example, by the detection of disease-associated autoantibodies. In this work, we will review the currently available clinical, in vitro, and animal studies dedicated to autoantibodies against apolipoprotein A-1 (anti-apoA-1 IgG), the major proteic fraction of high density lipoprotein. Current clinical studies indicate that high levels of anti-apoA-1 IgG are associated with a worse cardiovascular prognosis. In addition, in vitro and animal studies indicate a pro-inflammatory and pro-atherogenic role, supporting the hypothesis that these autoantibodies may play a direct causal role in CVD, and furthermore that they could potentially represent a therapeutic target for CVD in the future.
Collapse
|
41
|
Chronic exposure to oral pathogens and autoimmune reactivity in acute coronary atherothrombosis. Autoimmune Dis 2014; 2014:613157. [PMID: 24839554 PMCID: PMC4003799 DOI: 10.1155/2014/613157] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 01/18/2014] [Accepted: 01/20/2014] [Indexed: 01/22/2023] Open
Abstract
Background. It has been hypothesized that various infective agents may activate immune reactions as part of the atherosclerotic process. We aimed to investigate the interrelationship between chronic exposure to oral pathogens and immune-inflammatory response in patients with acute coronary atherothrombosis.
Patients and Methods. The study included 200 participants from Serbia: 100 patients with acute myocardial infarction (MI), and 100 age- and sex-matched controls. Antibodies to oral anaerobes and aerobes were determined as well as autoantibodies to endothelial cells, beta-2 glycoprotein I, platelet glycoprotein IIb/IIIa and anticardiolipin. Interleukin-6 (IL-6) and C-reactive protein (CRP) were measured. Results. The mean serum antibodies to oral anaerobes tended to be higher among subjects with MI (0.876 ± 0.303 versus 0.685 ± 0.172 OD, P < 0.001). Similarly, antibody levels against oral aerobes in patients were significantly different from controls. Antibodies against endothelial cell, beta-2 glycoprotein I, platelet glycoprotein IIb/IIIa, anticardiolipin along with CRP and IL-6 were highly elevated in patients. The levels of antibodies to oral bacteria showed linear correlation with tissue antibodies, CRP and IL-6.
Conclusion. Antibody response to chronic oral bacterial infections and host immune response against them may be responsible for the elevation of tissue antibodies and biomarkers of inflammation which are involved in acute coronary thrombosis development.
Collapse
|
42
|
'The way to a man's heart is through his gut microbiota'--dietary pro- and prebiotics for the management of cardiovascular risk. Proc Nutr Soc 2014; 73:172-85. [PMID: 24495527 DOI: 10.1017/s0029665113003911] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The human gut microbiota has been identified as a possible novel CVD risk factor. This review aims to summarise recent insights connecting human gut microbiome activities with CVD and how such activities may be modulated by diet. Aberrant gut microbiota profiles have been associated with obesity, type 1 and type 2 diabetes and non-alcoholic fatty liver disease. Transfer of microbiota from obese animals induces metabolic disease and obesity in germ-free animals. Conversely, transfer of pathogen-free microbiota from lean healthy human donors to patients with metabolic disease can increase insulin sensitivity. Not only are aberrant microbiota profiles associated with metabolic disease, but the flux of metabolites derived from gut microbial metabolism of choline, phosphatidylcholine and l-carnitine has been shown to contribute directly to CVD pathology, providing one explanation for increased disease risk of eating too much red meat. Diet, especially high intake of fermentable fibres and plant polyphenols, appears to regulate microbial activities within the gut, supporting regulatory guidelines encouraging increased consumption of whole-plant foods (fruit, vegetables and whole-grain cereals), and providing the scientific rationale for the design of efficacious prebiotics. Similarly, recent human studies with carefully selected probiotic strains show that ingestion of viable microorganisms with the ability to hydrolyse bile salts can lower blood cholesterol, a recognised risk factor in CVD. Taken together such observations raise the intriguing possibility that gut microbiome modulation by whole-plant foods, probiotics and prebiotics may be at the base of healthy eating pyramids advised by regulatory agencies across the globe. In conclusion, dietary strategies which modulate the gut microbiota or their metabolic activities are emerging as efficacious tools for reducing CVD risk and indicate that indeed, the way to a healthy heart may be through a healthy gut microbiota.
Collapse
|
43
|
Reifenberg K, Cheng F, Twardowski L, Küpper I, Wiese E, Bollmann F, Kleinert H, Blessing M, Lackner KJ, Torzewski M. T cell-specific overexpression of TGFß1 fails to influence atherosclerosis in ApoE-deficient mice. PLoS One 2013; 8:e81444. [PMID: 24339930 PMCID: PMC3855303 DOI: 10.1371/journal.pone.0081444] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Accepted: 10/21/2013] [Indexed: 01/24/2023] Open
Abstract
Clinical data have indicated a negative correlation between plasma TGFß1 concentrations and the extent of atherosclerosis and have thus led to the hypothesis that the pleiotropic cytokine may have anti-atherogenic properties. T-cells are currently discussed to significantly participate in atherogenesis, but the precise role of adaptive immunity in atherogenesis remains to be elucidated. TGFß1 is known to strongly modulate the function of T-cells, however, inhibition of TGFß1 signalling in T-cells of atherosclerosis-prone knock-out mice failed to unequivocally clarify the role of the cytokine for the development of atherosclerosis. In the present study, we thus tried to specify the role of TGFß1 in atherogenesis by using the murine CD2-TGFß1 transgenic strain which represents a well characterized model of T-cell specific TGFß1 overexpression. The CD2-TGFß1 transgenic mice were crossed to ApoE knock-out mice and quantity and quality of atherosclerosis regarding number of macrophages, smooth muscle cells, CD3 positive T-cells and collagen was analyzed in CD2-TGFß1 ApoE double mutants as well as non-transgenic ApoE controls on both normal and atherogenic diet of a duration of 8, 16 or 24 weeks, respectively. In all experimental groups investigated, we failed to detect any influence of TGFß1 overexpression on disease. Total number of CD3-positive T-lymphocytes was not significantly different in atherosclerotic lesions of CD2-TGFß1 ApoE−/− females and isogenic ApoE−/− controls, even after 24 weeks on the atherogenic diet. The synopsis of these data and our previous study on TGFß1 overexpressing macrophages suggests that potential effects of TGFß1 on atherosclerosis are most probably mediated by macrophages rather than T-cells.
Collapse
Affiliation(s)
- Kurt Reifenberg
- Animal Laboratory Services, German Cancer Research Center, Heidelberg, Germany
| | - Fei Cheng
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Laura Twardowski
- Department of Laboratory Medicine, Robert-Bosch-Hospital, Stuttgart, Germany
| | - Ines Küpper
- Central Laboratory Animal Facility, Johannes Gutenberg-University, Mainz, Germany
| | - Elena Wiese
- Central Laboratory Animal Facility, Johannes Gutenberg-University, Mainz, Germany
| | - Franziska Bollmann
- Department of Pharmacology, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Hartmut Kleinert
- Department of Pharmacology, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Manfred Blessing
- Center for Biotechnology and Biomedicine, Veterinary Faculty, University of Leipzig, Leipzig, Germany
| | - Karl J. Lackner
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Michael Torzewski
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
- Department of Laboratory Medicine, Robert-Bosch-Hospital, Stuttgart, Germany
- * E-mail:
| |
Collapse
|