1
|
Konishi T, Kawakami R, Vozenilek AE, Ghosh SKB, Xu W, Grogan A, Shah P, Tanaka T, Sekimoto T, Shiraki T, Kawai K, Sato Y, Mori M, Sakamoto A, Hisadome H, Ashida K, Bellissard A, Williams D, Dryanovski D, Kutys R, Cheng Q, Romero M, Chahal D, Virmani R, Finn AV. Mechanisms of Medial Wall Thinning in Chronic Total Occlusion. JACC Cardiovasc Interv 2024; 17:1719-1728. [PMID: 38970581 DOI: 10.1016/j.jcin.2024.05.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 04/25/2024] [Accepted: 05/07/2024] [Indexed: 07/08/2024]
Abstract
BACKGROUND The success rate of percutaneous coronary intervention (PCI) for chronic total occlusion (CTO) is lower and the risk for complications higher compared with other non-CTO PCI. Although interventionalists focus on intimal plaque characteristics, the coronary media is an important (especially for techniques involving antegrade dissection and re-entry) but poorly understood structure in CTO PCI. OBJECTIVES The aim of the present study was to investigate coronary medial wall thinning in CTO lesions and determine how this thinning might affect CTO PCI. METHODS A total of 2,586 sections were investigated, from arteries with evidence of CTO from 54 subjects (1,383 sections) and arteries without evidence of CTO from 54 subjects with non-coronary-related deaths (1,203 sections) after matching for age, gender, body weight, and body height. RESULTS The medial thickness in subjects with CTO was lower than that in those with non-coronary-related death (P < 0.001). In subjects with CTO, CTO lesions had thinner medial walls compared with those with lower luminal narrowing (P < 0.001). At the CTO distal segments, the 6- to 12-mm distal segment from the distal end of the CTO had significantly less luminal narrowing (P < 0.001), and similar medial thickness, compared with the distal end of the CTO. Immunohistochemical analysis revealed that short-duration CTO had more cleaved caspase-3-positive cells in media and had significantly more CD3+, CD4+, CD8+, and CD4+CD28null T cells compared with long-duration CTO. CONCLUSIONS CTO lesions demonstrated coronary medial thinning compared with non-CTO lesions. Further investigation of the cause-and-effect relationship among inflammation, apoptosis, and coronary medial wall thinning is warranted in future mechanistic studies.
Collapse
Affiliation(s)
- Takao Konishi
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA; Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Rika Kawakami
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Aimee E Vozenilek
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Saikat Kumar B Ghosh
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Weili Xu
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Alyssa Grogan
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Palak Shah
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Takamasa Tanaka
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Teruo Sekimoto
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Tatsuya Shiraki
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Kenji Kawai
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Yu Sato
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Masayuki Mori
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Atsushi Sakamoto
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | | | - Kazuhiro Ashida
- Cardiovascular Center, Seirei Yokohama Hospital, Yokohama, Japan
| | - Arielle Bellissard
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Desiree Williams
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Dilyan Dryanovski
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Robert Kutys
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Qi Cheng
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Maria Romero
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Diljon Chahal
- School of Medicine, University of Maryland, Baltimore, Maryland, USA
| | - Renu Virmani
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Aloke V Finn
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA; School of Medicine, University of Maryland, Baltimore, Maryland, USA.
| |
Collapse
|
2
|
Chen X, Yan Z, Pan Q, Zhang C, Chen Y, Liang X, Li S, Wang L. Bibliometric analysis of T-cells immunity in pulmonary hypertension from 1992 to 2022. Immun Inflamm Dis 2024; 12:e1280. [PMID: 38967362 PMCID: PMC11225084 DOI: 10.1002/iid3.1280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 04/25/2024] [Accepted: 05/03/2024] [Indexed: 07/06/2024] Open
Abstract
BACKGROUND Adaptive immunity is an important disease mediator of pulmonary vascular remodeling during pulmonary hypertension (PH) development, especially T-cells lymphocytes. However, data for bibliometric analysis of T cell immunity in PH is currently vacant. This aimed to provide a comprehensive and visualized view of T-cells research in PH pathogenesis and to lay a solid foundation for further studies. METHODS The data was acquired from the Web of Science Core Collection database. Web of Science analytic tool was used to analysis the publication years, authors, journals, countries, and organizations. CiteSpace 6.2.R3, VOSviewer 1.6.16, and Scimago Graphica 1.0.35.0 were applied to conduct a visualization bibliometric analysis about authors, countries, institutions, journals, references, and keywords. RESULTS Nine hundred and eight publications from 1992 to 2022 were included in the analysis. The results showed that Humbert Marc was the most prolific author. American Journal of Physiology Lung Cellular and Molecular Physiology had the most related articles. The institution with the most articles was Udice French Research University. The United States was far ahead in the article output. Keywords analysis showed that "Pulmonary hypertension" was the most usually appeared keyword in the relevant literature, and included "T-cells", "Regulatory T cells", and "Activated T cell." "miRNA" of reference co-citation clustering analysis demonstrated the possible T-cell immunity activation mechanisms in PH. The most cited literature was published in the European Heart Journal by Galie N in 2016. The strongest citation burst of keyword is "gene expression" and terms such as "vascular remodeling," "growth," "proliferation," and "fibrosis" are among the list, indicating that T-cells interact with stromal vascular cells to induce pulmonary vascular remodeling. The strongest burst of cited reference is "Galie N, 2016." CONCLUSIONS T-cell immunity is an important pathogenesis mechanism for PH development, which may have interaction with miRNAs and stromal vascular cells, but the possible T-cell immunity activation mechanisms in PH need to be investigated further.
Collapse
Affiliation(s)
- Xian Chen
- Department of NephrologySecond Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Zhe Yan
- Department of NephrologySecond Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Qing Pan
- Department of NephrologySecond Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Chunxia Zhang
- Department of NephrologySecond Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Yakun Chen
- Department of NephrologySecond Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Xuzhi Liang
- Department of NephrologySecond Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Shaomei Li
- Department of NephrologySecond Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Lei Wang
- Department of Pulmonary and Critical Care MedicineSecond Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| |
Collapse
|
3
|
Liu Q, Zheng Y, Goronzy JJ, Weyand CM. T cell aging as a risk factor for autoimmunity. J Autoimmun 2023; 137:102947. [PMID: 36357240 PMCID: PMC10164202 DOI: 10.1016/j.jaut.2022.102947] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 10/23/2022] [Indexed: 11/09/2022]
Abstract
Immune aging is a complex process rendering the host susceptible to cancer, infection, and insufficient tissue repair. Many autoimmune diseases preferentially occur during the second half of life, counterintuitive to the concept of excess adaptive immunity driving immune-mediated tissue damage. T cells are particularly susceptible to aging-imposed changes, as they are under extreme proliferative pressure to fulfill the demands of clonal expansion and of homeostatic T cell repopulation. T cells in older adults have a footprint of genetic and epigenetic changes, lack mitochondrial fitness, and fail to maintain proteostasis, diverging them from host protection to host injury. Here, we review recent progress in understanding how the human T-cell system ages and the evidence detailing how T cell aging contributes to autoimmune conditions. T cell aging is now recognized as a risk determinant in two prototypic autoimmune syndromes; rheumatoid arthritis and giant cell arteritis. The emerging concept adds susceptibility to autoimmune and autoinflammatory disease to the spectrum of aging-imposed adaptations and opens new opportunities for immunomodulatory therapy by restoring the functional intactness of aging T cells.
Collapse
Affiliation(s)
- Qingxiang Liu
- Department of Medicine, Mayo Clinic Alix School of Medicine, Rochester, MN 55905, USA; Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Yanyan Zheng
- Department of Medicine, Mayo Clinic Alix School of Medicine, Rochester, MN 55905, USA; Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA; Department of Cardiovascular Medicine, Mayo Alix School of Medicine, Rochester, MN, USA
| | - Jorg J Goronzy
- Department of Medicine, Mayo Clinic Alix School of Medicine, Rochester, MN 55905, USA; Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA; Department of Medicine, Stanford University School of Medicine, Stanford, CA 94306, USA
| | - Cornelia M Weyand
- Department of Medicine, Mayo Clinic Alix School of Medicine, Rochester, MN 55905, USA; Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA; Department of Cardiovascular Medicine, Mayo Alix School of Medicine, Rochester, MN, USA; Department of Medicine, Stanford University School of Medicine, Stanford, CA 94306, USA.
| |
Collapse
|
4
|
Endothelial glycocalyx and microvascular perfusion are associated with carotid intima-media thickness and impaired myocardial deformation in psoriatic disease. J Hum Hypertens 2022; 36:1113-1120. [PMID: 34819613 DOI: 10.1038/s41371-021-00640-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 11/09/2021] [Accepted: 11/11/2021] [Indexed: 02/08/2023]
Abstract
Psoriatic disease is associated with vascular and myocardial dysfunction. We aimed to evaluate endothelial glycocalyx barrier properties and microvascular perfusion in psoriatic patients, as well as their correlation with carotid intima-media thickness (cIMT) and markers of left ventricular (LV) myocardial deformation. We examined 297 psoriatic patients and 150 controls, adjusted for age, sex, and atherosclerotic risk factors. The severity of psoriatic disease was estimated using the psoriasis area and severity index (PASI). Perfused boundary region (PBR), a marker of glycocalyx barrier function, was measured non-invasively in sublingual microvessels with a diameter 5-25 μm using Sidestream Dark Field camera (Microscan, GlycoCheck). Increased PBR indicates reduced glycocalyx thickness. Indexes of microvascular perfusion, including red blood cells filling (RBCF) and functional microvascular density, were also calculated. We measured cIMT, coronary flow reserve (CFR) and markers of myocardial deformation by speckle-tracking imaging, namely global longitudinal strain (GLS) and percentage changes between peak twisting and untwisting at mitral valve opening (%dpTw-UtwMVO). Compared to controls, psoriatic patients had higher PBR5-25μm (2.13 ± 0.29μm versus 1.78 ± 0.25μm, p < 0.001) and lower RBCF and functional microvascular density (p < 0.001). Increased PASI was associated with elevated PBR and more impaired cIMT and GLS (p < 0.05). There was an inverse association of PBR with RBCF and functional microvascular density (p < 0.001). In psoriatic population, increased PBR was related to increased cIMT, reduced CFR, impaired GLS and decreased %dpTw-UtwMVO (p < 0.001). Glycocalyx thickness is reduced in psoriatic patients, which in turn impairs microvascular perfusion, and is associated with carotid IMT and impaired coronary and myocardial function.Clinical Trial Registration-URL: http://www.clinicaltrials.gov . Unique identifier: NCT02144857.
Collapse
|
5
|
Zhao TV, Sato Y, Goronzy JJ, Weyand CM. T-Cell Aging-Associated Phenotypes in Autoimmune Disease. FRONTIERS IN AGING 2022; 3:867950. [PMID: 35821833 PMCID: PMC9261367 DOI: 10.3389/fragi.2022.867950] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 03/28/2022] [Indexed: 01/10/2023]
Abstract
The aging process causes profound restructuring of the host immune system, typically associated with declining host protection against cancer and infection. In the case of T cells, aging leads to the accumulation of a diverse set of T-cell aging-associated phenotypes (TASP), some of which have been implicated in driving tissue inflammation in autoimmune diseases. T cell aging as a risk determinant for autoimmunity is exemplified in two classical autoimmune conditions: rheumatoid arthritis (RA), a disease predominantly affecting postmenopausal women, and giant cell arteritis (GCA), an inflammatory vasculopathy exclusively occurring during the 6th-9th decade of life. Pathogenic T cells in RA emerge as a consequence of premature immune aging. They have shortening and fragility of telomeric DNA ends and instability of mitochondrial DNA. As a result, they produce a distinct profile of metabolites, disproportionally expand their endoplasmic reticulum (ER) membranes and release excess amounts of pro-inflammatory effector cytokines. Characteristically, they are tissue invasive, activate the inflammasome and die a pyroptotic death. Patients with GCA expand pathogenic CD4+ T cells due to aberrant expression of the co-stimulatory receptor NOTCH1 and the failure of the PD-1/PD-L1 immune checkpoint. In addition, GCA patients lose anti-inflammatory Treg cells, promoting tissue-destructive granulomatous vasculitis. In summary, emerging data identify T cell aging as a risk factor for autoimmune disease and directly link TASPs to the breakdown of T cell tolerance and T-cell-induced tissue inflammation.
Collapse
Affiliation(s)
- Tuantuan V. Zhao
- Mayo Clinic Alix School of Medicine, College of Medicine and Science, Rochester, MN, United States
| | - Yuki Sato
- Mayo Clinic Alix School of Medicine, College of Medicine and Science, Rochester, MN, United States
| | - Jorg J. Goronzy
- Mayo Clinic Alix School of Medicine, College of Medicine and Science, Rochester, MN, United States
- School of Medicine, Stanford University, Stanford, CA, United States
| | - Cornelia M. Weyand
- Mayo Clinic Alix School of Medicine, College of Medicine and Science, Rochester, MN, United States
- School of Medicine, Stanford University, Stanford, CA, United States
| |
Collapse
|
6
|
The impact of CD4 +CD28 null T lymphocytes on atrial fibrillation: a potential pathophysiological pathway. Inflamm Res 2021; 70:1011-1014. [PMID: 34536081 PMCID: PMC8572810 DOI: 10.1007/s00011-021-01502-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/04/2021] [Accepted: 09/07/2021] [Indexed: 11/12/2022] Open
Abstract
Introduction Atrial fibrillation (AF) represents the most common cardiac arrhythmia in daily clinical practice and substantially impacts affected patients by elevation of both morbidity and mortality. Previous investigations proved that inflammatory processes are closely linked to this multifactorial pathogenesis—especially autoreactive CD4+CD28null T cells received in-depth attention. Purpose Consequently, a potential pathophysiological pathway of the impact of CD4+CD28null T lymphocytes on the development and progression AF can be outlined. Conclusion Considering the available data in the literature, it needs to be assumed that CD4+CD28null T lymphocytes are mainly involved in the development of AF and disease progression. Of utmost importance, it can be considered as the result of a T-cell-mediated auto-immune reaction among myocardial tissue. However, mechanisms which recruit CD4+CD28null cells in cardiac tissue remain unclear and need further investigation.
Collapse
|
7
|
Oxidative stress-responsive apoptosis-inducing protein in patients with heterozygous familial hypercholesterolemia. Heart Vessels 2021; 36:1923-1932. [PMID: 34308503 DOI: 10.1007/s00380-021-01898-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 07/02/2021] [Indexed: 10/20/2022]
Abstract
Oxidative stress, an inducer of apoptosis, plays a critical role in ischemia/reperfusion injury and atherosclerosis. We previously identified an apoptosis-inducing ligand, the post-translationally modified secreted form of eukaryotic translation initiation factor 5A (eIF5A), 'oxidative stress-responsive apoptosis-inducing protein' (ORAIP). In this study, we investigated the role of ORAIP in patients with heterozygous familial hypercholesterolemia (HeFH), a leading cause of premature cardiovascular disease. We analyzed plasma ORAIP and oxidized low-density lipoprotein (oxLDL) levels in 60 patients with HeFH (60% male, 57.0 ± 13.6 years of age) and 20 patients with LDL-C hypercholesterolemia (DL, 85% male, 64.1 ± 13.3 years of age). The coronary artery atherosclerosis from the patients with HeFH who had a coronary artery bypass graft was investigated by double immunostaining. The plasma ORAIP levels in the patients with HeFH were significantly elevated compared to those in the patients with DL (73.5 ± 46.0 vs. 48.3 ± 21.4 ng/mL, p = 0.0277). The plasma oxLDL levels in HeFH patients were also elevated (156.8 ± 65.2 vs. 123.7 ± 46.6 mg/dL, p = 0.0461) compared to those in DL patients and correlated with maxLDL-C levels (R = 0.4454, p = 0.00648). Double-immunostaining of ORAIP and oxLDL in the coronary artery from patients with HeFH who had a coronary artery bypass graft showed that ORAIP and oxLDL were colocalized with apoptotic vascular smooth muscle cells in the atherosclerotic plaque. ORAIP plays a role in the development of oxidative stress-induced atherosclerosis and may be an important therapeutic target for plaque rupture in patients with HeFH.
Collapse
|
8
|
Paccosi S, Pala L, Cresci B, Silvano A, Cecchi M, Caporale R, Maria Rotella C, Parenti A. Insulin resistance and obesity affect monocyte-derived dendritic cell phenotype and function. Diabetes Res Clin Pract 2020; 170:108528. [PMID: 33157116 DOI: 10.1016/j.diabres.2020.108528] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 09/10/2020] [Accepted: 10/26/2020] [Indexed: 12/12/2022]
Abstract
AIM Cardiovascular disease (CVD) is prevalent in women after menopause, which may be associated with obesity, insulin resistance and metaflammation. Despite the recognized role of immunological mechanisms in vascular remodeling, the role of dendritic cells (DCs) is still unclear. The aim was to characterize monocyte-derived DCs (Mo-DC) in post-menopausal patients with type 2 diabetes (T2DM) and obese woman, without clinical manifestations of atherosclerosis. METHODS Obese post-menopausal women with or without T2DM were enrolled and were compared to age-matched healthy women. DCs obtained from patients were phenotypically and functionally characterized by flow cytometry and mixed lymphocyte reaction. MRNA integrins expression was assessed by real time RT-PCR; circulating fetuin-A and adiponectin levels were measured by ELISA. RESULTS Phenotypic dysregulation of Mo-DC reported was related to a defective allogenic lymphocyte stimulation and to an increased mRNA of CD11c, CD18 and DC-SIGN/CD209 which regulate their adhesion to vascular wall cells. Fetuin-A and adiponectin levels were significantly altered and negatively correlated. Hyperglycaemia significantly impaired CD14+ transdifferentiation into Mo-DC. CONCLUSIONS These data show a dysfunction of Mo-DCs obtained from precursors isolated from T2DM obese post-menopausal woman without any documented clinical CV event. Association of obesity to diabetes seems to worsen DC's phenotype and function and increase vascular inflammation.
Collapse
Affiliation(s)
- Sara Paccosi
- Department of Health Sciences, Clinical Pharmacology and Oncology Section, University of Florence, Florence, Italy
| | - Laura Pala
- Diabetology, Careggi University Hospital, Florence, Italy
| | - Barbara Cresci
- Diabetology, Careggi University Hospital, Florence, Italy
| | - Angela Silvano
- Department of Health Sciences, Clinical Pharmacology and Oncology Section, University of Florence, Florence, Italy
| | - Marta Cecchi
- Department of Health Sciences, Clinical Pharmacology and Oncology Section, University of Florence, Florence, Italy
| | - Roberto Caporale
- Cytofluorimetry and Immunotherapy Diagnostic Center, Careggi University Hospital, Florence, Italy
| | - Carlo Maria Rotella
- Department of Biomedical Clinical and Experimental Sciences, Endocrine Unit, University of Florence, Florence, Italy
| | - Astrid Parenti
- Department of Health Sciences, Clinical Pharmacology and Oncology Section, University of Florence, Florence, Italy.
| |
Collapse
|
9
|
Kosmaczewska A, Ciszak L, Stosio M, Szteblich A, Madej M, Frydecka I, Wiland P, Szmyrka M. CD4 +CD28 null T cells are expanded in moderately active systemic lupus erythematosus and secrete pro-inflammatory interferon gamma, depending on the Disease Activity Index. Lupus 2020; 29:705-714. [PMID: 32279585 DOI: 10.1177/0961203320917749] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND Pathogenic CD4+CD28null cells are characterized by inflammatory cytokine synthesis and tropism to the inflamed tissues. Recent studies showed the involvement of CD28null T cells in a severe clinical outcome of lupus. However, their role in moderately active disease is still unresolved. METHODS We examined the levels of circulating CD4+CD28null cells and CD8+CD28null suppressor T cells. We also compared the CD4+CD28null and CD4+CD28+ T-cell functional properties, including the expression of interferon gamma (IFN-γ) and Ki67 among systemic lupus erythematosus (SLE) patients (n = 20) and healthy controls (n = 20). All the patients were under immunosuppressive treatment and exhibited moderate SLE activity (median SLE Disease Activity Index (SLEDAI) = 6). RESULTS In patients, we found elevated CD4+CD28null and unchanged levels of suppressor CD8+CD28null T cells. There was no difference between patients and controls in IFN-γ and Ki67-expressing CD4+, CD4+CD28+, and CD4+CD28null T cells, except for higher IFN-γ levels in CD4+CD28+ T cells in SLE. In each studied group, we observed a higher preponderance of IFN-γ- and Ki67-expressing cells among CD4+CD28null T cells and lower levels of IFN-γ in CD4+CD28null T cells compared to the CD28+ subset. Similarly, Ki67 intensity was decreased in healthy CD4+CD28null cells, whereas in patients, comparably high expression was observed in both subsets. IFN-γ intensity in CD4+CD28null T cells correlated with SLEDAI. CONCLUSION SLE with a moderately active clinical course is characterized by peripheral blood expansion of CD4+CD28null T cells and a normal abundance of suppressor CD8+CD28null T cells. The demonstration that these pathogenic CD4+ T cells, despite the lack of CD28, maintain the ability to produce pro-inflammatory IFN-γ positively correlated with disease activity as well as relatively high proliferative capacity may suggest their potentially predictive role in SLE flares.
Collapse
Affiliation(s)
- Agata Kosmaczewska
- Department of Experimental Therapy, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Lidia Ciszak
- Department of Experimental Therapy, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Malgorzata Stosio
- Faculty of Chemistry, Wroclaw University of Science and Technology, Wroclaw, Poland
| | - Aleksandra Szteblich
- Department of Experimental Therapy, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Marta Madej
- Department of Rheumatology and Internal Diseases, Wroclaw Medical University, Wroclaw, Poland
| | - Irena Frydecka
- Department of Experimental Therapy, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Piotr Wiland
- Department of Rheumatology and Internal Diseases, Wroclaw Medical University, Wroclaw, Poland
| | - Magdalena Szmyrka
- Department of Rheumatology and Internal Diseases, Wroclaw Medical University, Wroclaw, Poland
| |
Collapse
|
10
|
Lazou A, Ikonomidis I, Bartekova M, Benedek T, Makavos G, Palioura D, Cabrera Fuentes H, Andreadou I. Chronic inflammatory diseases, myocardial function and cardioprotection. Br J Pharmacol 2020; 177:5357-5374. [PMID: 31943142 DOI: 10.1111/bph.14975] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 11/22/2019] [Accepted: 11/26/2019] [Indexed: 12/15/2022] Open
Abstract
The association between chronic inflammatory diseases (CIDs) and increased cardiovascular (CV) risk is well documented and can be a most threatening complication in these patients. However, the pathogenetic mechanisms underlying increased CV risk remain elusive, especially in their cellular and biochemical pathways. Using animal models to understand mechanisms underlying cardiac involvement are limited. Additionally, treatments may influence cardiovascular events through different outcomes. Some drugs used to treat CIDs can negatively affect cardiac function by a direct toxicity, whereas others may protect the myocardium. In the present article, we focus on the cardiac manifestations and risk factors, the pathogenetic mechanisms, and the effect of treatments on myocardial function and cardioprotection for five common worldwide CIDs (rheumatoid arthritis, systemic lupus erythematosus, systemic sclerosis, psoriasis and inflammatory bowel disease). We also give recommendations in order to evaluate common targets between CID and CV disease (CVD) and to design therapies to alleviate CID-related CVD. LINKED ARTICLES: This article is part of a themed issue on Risk factors, comorbidities, and comedications in cardioprotection. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v177.23/issuetoc.
Collapse
Affiliation(s)
- Antigone Lazou
- School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Ignatios Ikonomidis
- Second Cardiology Department, Attikon Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Monika Bartekova
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Theodora Benedek
- Clinic of Cardiology, Cardiac Critical Care Unit, University of Medicine and Pharmacy, Târgu Mureş, Romania
| | - George Makavos
- Second Cardiology Department, Attikon Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitra Palioura
- School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Hector Cabrera Fuentes
- SingHealth Duke-NUS Cardiovascular Sciences Academic Clinical Programme and Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore.,National Heart Research Institute Singapore, National Heart Centre, Singapore.,Institute of Physiology, Medical School, Justus-Liebig University, Giessen, Germany.,Tecnologico de Monterrey, Centro de Biotecnologia-FEMSA, Monterrey, NL, Mexico.,Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Kazan, Russian Federation
| | - Ioanna Andreadou
- Laboratory of Pharmacology, School of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
11
|
SULT2B1b inhibits reverse cholesterol transport and promotes cholesterol accumulation and inflammation in lymphocytes from AMI patients with low LDL-C levels. Clin Sci (Lond) 2020; 134:273-287. [PMID: 31957803 DOI: 10.1042/cs20190459] [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: 05/08/2019] [Revised: 01/16/2020] [Accepted: 01/17/2020] [Indexed: 01/19/2023]
Abstract
The current main treatment for coronary artery disease (CAD) is to reduce low-density lipoprotein cholesterol (LDL-C) by statins, which could decrease the incidence of major adverse cardiovascular events (MACEs) by 30%. However, many residual risks still remain. To clarify the mechanism involved, we studied patients with acute myocardial infarction (AMI) with low LDL-C levels. Lymphocytes were isolated, and it was found that despite no difference in plasma LDL-C level, the lymphocyte cholesterol content was higher in AMI patient than those in non-CAD patients; thus, the decrease in intracellular cholesterol content was inconsistent with that in the plasma. Additionally, [3H]-cholesterol efflux rates were lower and mRNA levels of the inflammatory factors tumour necrosis factor-α (TNF-α) and interferon-γ (IFN-γ) higher in AMI lymphocytes. It was found that sulphotransferase 2B1b (SULT2B1b) expression was higher in AMI lymphocytes. Further research using Jurkat T lymphocytes confirmed that SULT2B1b knockdown increased cholesterol efflux capacity and decreased mRNA levels of TNF-α and IFN-γ by increasing liver X receptor (LXR)-β levels. Furthermore, the degree of CpG island methylation in the SULT2B1b promoter was reduced in cells from AMI patients. In conclusion, SULT2B1b up-regulation due to hypomethylation of its promoter promotes cholesterol accumulation and inflammation by inhibiting LXR-β in lymphocytes of AMI patients with low LDL-C levels. Therefore, reducing intracellular cholesterol is also important as plasma cholesterol levels. Therapeutic approaches to decrease SULT2B1b expression might be potentially beneficial for CAD prevention by decreasing intracellular cholesterol.
Collapse
|
12
|
El-Aswad BEDW, Sonbol AA, Moharm IM, El-Refai SA, Seleem HEDM, Soliman SS. Circulating endothelial cells in severe Plasmodium falciparum infection. Parasitol Int 2019; 72:101926. [PMID: 31100355 DOI: 10.1016/j.parint.2019.101926] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Revised: 05/11/2019] [Accepted: 05/13/2019] [Indexed: 12/28/2022]
Abstract
Plasmodium falciparum infection is associated with diffuse vascular dys-regulation. Levels of blood circulating endothelial cells (CECs; CD146+CD45-) are a marker of vascular injury. This study aimed to measure blood CECs by flow cytometery in patients with acute malaria infection before and after treatment and to evaluate the prognostic value of that measurement for that disease. The subjects were allocated into: Group I: uncomplicated malaria (UM, n = 32), Group II: severe malaria (SM, n = 12), Group III: the treated UM (TUM, n = 32), Group IV: the treated SM (TSM, n = 12) and Group V: healthy controls (HC, n = 25). Before treatment, SM patients showed the highest mean number of CECs (30,658.3 ± 2658.2/5 × 106 peripheral blood mononuclear cells (PBMCs)), followed by UM patients (19,481.56 ± 866.83/5x106PBMCs) and both groups were significantly higher than HC (2034 ± 300.59/5x106PBMCs, P < .001). The level of CECs decreased significantly in both infected groups after treatment; in TUM it became 5602.18 ± 509.72/5 × 106PBMCs and in TSM it reached 8457.5 ± 452.4/5 × 106 PBMCs (both values P < .001 in comparison with SM). By receiver operating characteristic curve analysis, the best cut-off count for CECs which enables prediction of the occurrence of severe malaria infection was 27,150/5 × 106 PBMCs or more, with 100% sensitivity, 100% specificity, and 100% accuracy. CECs had a significant positive correlation with parasitemia index and serum creatinine and a significant negative correlation with hemoglobin concentration in patients with acute malaria. In conclusion, the level of CECs could be used as a biomarker denoting endothelium damage during acute P. falciparum infection, and it correlated with infection severity and predicted its prognosis.
Collapse
Affiliation(s)
- Bahaa El-Deen W El-Aswad
- Department of Medical Parasitology, Faculty of Medicine, Menoufia University, Shebin al-Kom, Egypt.
| | - Ahmed A Sonbol
- Department of Clinical Pathology, Faculty of Medicine, Menoufia University, Shebin al-Kom, Egypt
| | - Ismail M Moharm
- Department of Medical Parasitology, Faculty of Medicine, Menoufia University, Shebin al-Kom, Egypt
| | - Samar A El-Refai
- Department of Medical Parasitology, Faculty of Medicine, Menoufia University, Shebin al-Kom, Egypt
| | - Hosam El-Din M Seleem
- Department of Tropical medicine, Faculty of Medicine, Menoufia University, Shebin al-Kom, Egypt
| | - Shiamaa S Soliman
- Department of Public Health and Community Medicine, Faculty of Medicine, Menoufia University, Shebin al-Kom, Egypt
| |
Collapse
|
13
|
Abstract
The diminished capacity for wound healing in patients with diabetes contributes to morbidity through ulceration and recurrent infections, loss of function and decreased workplace productivity, increased hospitalisation rates, and rising health-care costs. These are due to diabetes' effects on signalling molecules, cellular cascades, different cell populations, and the vasculature. The function of multiple immune system components including cellular response, blood factors, and vascular tone are all negatively impacted by diabetes. The purpose of this paper is to review the current understanding of immune and vascular dysfunction contributing to impaired wound healing mechanisms in the diabetic population. Normal wound healing mechanisms are reviewed followed by diabetic aberrations to immune and inflammatory function and atherogenesis and angiopathy. DECLARATION OF INTEREST The authors have no financial or personal relationships to people or organisations that could potentially and inappropriately influence their work and conclusions.
Collapse
Affiliation(s)
- A S Ahmed
- Baylor College of Medicine, 1 Baylor Plaza, Houston, Texas 77030
| | - E L Antonsen
- Baylor College of Medicine, 1 Baylor Plaza, Houston, Texas 77030.,Department of Medicine - Section of Emergency Medicine, 1 Baylor Plaza, Houston, Texas.,Department of Medicine - Section of Space Medicine, 1 Baylor Plaza, Houston, Texas
| |
Collapse
|
14
|
Kitamura K, Sato K, Sawabe M, Yoshida M, Hagiwara N. P-Selectin Glycoprotein Ligand-1 (PSGL-1) Expressing CD4 T Cells Contribute Plaque Instability in Acute Coronary Syndrome. Circ J 2018; 82:2128-2135. [PMID: 29962384 DOI: 10.1253/circj.cj-17-1270] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Adhesion molecules have essential roles in the development of atherosclerosis. We investigated whether P-selectin glycoprotein ligand-1 (PSGL-1)-expressing CD4 T cells contribute to plaque instability in acute coronary syndrome (ACS). METHODS AND RESULTS We studied the adhesion molecules on CD4 T cells from consecutive patients with ACS treated with thrombus-aspirating device and compared them with healthy controls (n=48 each). Blood, thrombi, and plaque samples from the culprit coronary arteries were collected by thrombus aspiration performed during emergency coronary artery angiography. According to flow cytometry results, peripheral CD4 T cells from ACS patients strongly expressed PSGL-1 and integrin β2 (P<0.05 for both) more than those from controls; culprit coronary arteries contained an abundance of PSGL-1+(P<0.001) but not integrin β2+CD4 T cells. In addition, immunohistochemical analysis of the thrombus-aspirating device samples revealed numerous PSGL-1+CD4 T cells in plaques from the culprit lesions. Results from the selectin-binding assay demonstrated that activated PSGL-1+CD4 T cells from ACS patients bound to P- or E-selectin after triggering the T-cell receptor, and adhered to endothelial cells under laminar flow conditions (P<0.05 and P<0.05, respectively), inducing their apoptosis (P<0.01) via activated caspase-3, which correlated with PSGL-1 expression (R=0.788, P=0.021) and was suppressed by application of a PSGL-1-specific antibody (P<0.05). CONCLUSIONS PSGL-1 contributed to cytotoxic CD4 T cell homing to the culprit coronary artery and promoted plaque instability in ACS.
Collapse
Affiliation(s)
| | - Kayoko Sato
- Department of Cardiology, Tokyo Women's Medical University
| | - Motoji Sawabe
- Section of Molecular Pathology, Graduate School of Health Care Sciences, Tokyo Medical and Dental University
| | - Masayuki Yoshida
- Life Sciences and Bioethics Research Center, Tokyo Medical and Dental University
| | | |
Collapse
|
15
|
CD4 +CD28 null T Lymphocytes are Associated with the Development of Atrial Fibrillation after Elective Cardiac Surgery. Sci Rep 2018; 8:9624. [PMID: 29941960 PMCID: PMC6018098 DOI: 10.1038/s41598-018-28046-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 05/25/2018] [Indexed: 11/17/2022] Open
Abstract
Post-operative atrial fibrillation (POAF) is postulated as a complex interaction of different pathogenic factors, suggesting inflammatory processes as a main trigger of this particular type of atrial fibrillation. Therefore, the study sought to assess the impact of cellular immunity on the development of POAF. Comparing patients developing POAF to individuals free of POAF the fraction of CD4+CD28null T Lymphocytes was significantly higher in individuals developing POAF (11.1% [POAF] vs. 1.9% [non-POAF]; p < 0.001). CD4+CD28null cells were independently associated with the development of POAF with an adjusted odds ratio per one standard deviation of 4.89 (95% CI: 2.68–8.97; p < 0.001). Compared to N-terminal Pro-Brain Natriuretic Peptide, the fraction of CD4+CD28null cells demonstrated an increased discriminatory power for the development of POAF (NRI: 87.9%, p < 0.001; IDI: 30.9%, p < 0.001). Interestingly, a pre-operative statin-therapy was associated with a lower fraction of CD4+CD28null cells (p < 0.001) and showed an inverse association with POAF (p < 0.001). CD4+CD28null cells proved to be predictive for the development of POAF after cardiac surgery. Our results potentially indicate an auto-immune impact of this preexisting, highly cytotoxic T cell subset in the pathogenesis of POAF, which might be modified via the anti-inflammatory potential of a pre-operative statin-therapy.
Collapse
|
16
|
Rakic M, Persic V, Kehler T, Bastiancic AL, Rosovic I, Laskarin G, Sotosek Tokmadzic V. Possible role of circulating endothelial cells in patients after acute myocardial infarction. Med Hypotheses 2018; 117:42-46. [PMID: 30077195 DOI: 10.1016/j.mehy.2018.06.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 05/22/2018] [Accepted: 06/05/2018] [Indexed: 01/16/2023]
Abstract
Acute myocardial infarction (AMI) occurs as a result of insufficient myocardial perfusion leading to cell necrosis. This is most commonly due to the obstruction of the coronary artery by ruptured atherosclerotic plaque and thrombosis. Damaged ischemic and necrotic myocardial cells release pro-inflammatory substances in tissue and plasma, leading to a systemic inflammatory response. Profound systemic inflammatory response during ischemia/reperfusion injury causes disruption of endothelial glycocalyx and detachment of endothelial cells that express von Willebrant factor (vWF). We hypothesize that circulating vWF+ endothelial cells could act as antigen presenting cells which interact with T and NK cells directly, by cell to cell contact and indirectly by cytokine and chemokine secretion, leading to the immune response towards inflammation. Analyzing the frequency, phenotype and pro-inflammatory substances produced in circulating vWF positive (+) cells in patients with AMI could be beneficial to determine the severity of the pro-inflammatory response, according to the level of endothelial dysfunction in the early period of AMI. To evaluate these hypotheses, we suggest to determine frequency, phenotype, and ability of cytokine/chemokine production in circulating vWF+ endothelial cells by simultaneous surface and intracellular cell staining, and flow cytometry analysis. Secretion of pro-inflammatory cytokines and chemokines, pro-atherogenic substances and the components of glycocalyx might be measured in supernatants of magnetically separated or sorted vWF+ endothelial cells, as well as in the serum of a patient with acute AMI by enzyme linked-immunoassay tests. The interaction of increasing concentrations of isolated circulating vWF+ endothelial cells and cognate T and NK cells might be investigated by lymphocyte proliferation rate, cytotoxic mediators' expression, and cytokine production. If our hypothesis is correct, characterization of circulating vWF+ endothelial cells could grant us greater insight into their role in pathophysiology of AMI and the degree of myocardial damage.
Collapse
Affiliation(s)
- Marijana Rakic
- Division of Cardiology, Hospital for Medical Rehabilitation of the Hearth and Lung Diseases and Rheumatism "Thalassotherapia" Opatija, 51410 Opatija, M. Tita 188, Croatia
| | - Viktor Persic
- Division of Cardiology, Hospital for Medical Rehabilitation of the Hearth and Lung Diseases and Rheumatism "Thalassotherapia" Opatija, 51410 Opatija, M. Tita 188, Croatia; Department of Medical Rehabilitation, Medical Faculty, University of Rijeka, 51000 Rijeka, B. Branchetta 20, Croatia
| | - Tatjana Kehler
- Department of Rheumatology, Rehabilitation, and Physical Medicine, Hospital for Medical Rehabilitation of Hearth and Lung Diseases and Rheumatism "Thalassotherapia-Opatija", 51410 Opatija, M. Tita 188, Croatia
| | - Ana Lanca Bastiancic
- Division of Cardiology, Hospital for Medical Rehabilitation of the Hearth and Lung Diseases and Rheumatism "Thalassotherapia" Opatija, 51410 Opatija, M. Tita 188, Croatia
| | - Ivan Rosovic
- Division of Cardiology, Hospital for Medical Rehabilitation of the Hearth and Lung Diseases and Rheumatism "Thalassotherapia" Opatija, 51410 Opatija, M. Tita 188, Croatia
| | - Gordana Laskarin
- Department of Rheumatology, Rehabilitation, and Physical Medicine, Hospital for Medical Rehabilitation of Hearth and Lung Diseases and Rheumatism "Thalassotherapia-Opatija", 51410 Opatija, M. Tita 188, Croatia; Department of Physiology and Immunology, Medical Faculty University of Rijeka, B.Branchetta 20, 51000 Rijeka, Croatia
| | - Vlatka Sotosek Tokmadzic
- Department of Anesthesiology, Reanimatology and Intensive Care Medicine, Faculty of Medicine, University of Rijeka, Brace Branchetta 20, 51000 Rijeka, Croatia.
| |
Collapse
|
17
|
Weyand CM, Berry GJ, Goronzy JJ. The immunoinhibitory PD-1/PD-L1 pathway in inflammatory blood vessel disease. J Leukoc Biol 2017; 103:565-575. [PMID: 28848042 DOI: 10.1189/jlb.3ma0717-283] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 08/03/2017] [Indexed: 12/16/2022] Open
Abstract
Because of their vital function, the wall structures of medium and large arteries are immunoprivileged and protected from inflammatory attack. That vascular immunoprivilege is broken in atherosclerosis and in vasculitis, when wall-invading T cells and macrophages (Mϕ) promote tissue injury and maladaptive repair. Historically, tissue-residing T cells were studied for their antigen specificity, but recent progress has refocused attention to antigen-nonspecific regulation, which determines tissue access, persistence, and functional differentiation of T cells. The coinhibitory receptor PD-1, expressed on T cells, delivers negative signals when engaged by its ligand PD-L1, expressed on dendritic cells, Mϕ, and endothelial cells to attenuate T cell activation, effector functions, and survival. Through mitigating signals, the PD-1 immune checkpoint maintains tissue tolerance. In line with this concept, dendritic cells and Mϕs from patients with the vasculitic syndrome giant cell arteritis (GCA) are PD-L1lo ; including vessel-wall-embedded DCs that guard the vascular immunoprivilege. GCA infiltrates in the arterial walls are filled with PD-1+ T cells that secrete IFN-γ, IL-17, and IL-21; drive inflammation-associated angiogenesis; and facilitate intimal hyperplasia. Conversely, chronic tissue inflammation in the atherosclerotic plaque is associated with an overreactive PD-1 checkpoint. Plaque-residing Mϕs are PD-L1hi , a defect induced by their addiction to glucose and glycolytic breakdown. PD-L1hi Mϕs render patients with coronary artery disease immunocompromised and suppress antiviral immunity, including protective anti-varicella zoster virus T cells. Thus, immunoinhibitory signals affect several domains of vascular inflammation; failing PD-L1 in vasculitis enables unopposed immunostimulation and opens the flood gates for polyfunctional inflammatory T cells, and excess PD-L1 in the atherosclerotic plaque disables tissue-protective T cell immunity.
Collapse
Affiliation(s)
- Cornelia M Weyand
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Gerald J Berry
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA
| | - Jörg J Goronzy
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
18
|
Sulzgruber P, Koller L, Winter MP, Richter B, Blum S, Korpak M, Hülsmann M, Goliasch G, Wojta J, Niessner A. The impact of CD4+CD28null T-lymphocytes on atrial fibrillation and mortality in patients with chronic heart failure. Thromb Haemost 2017; 117:349-356. [DOI: 10.1160/th16-07-0531] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 11/03/2016] [Indexed: 12/28/2022]
Abstract
SummaryAtrial fibrillation (AF) represents the most common cardiac arrhythmia. Especially in patients with chronic heart failure (CHF) the development of AF represents a severe complication resulting in haemodynamic deterioration. While pro-inflammatory cytokines proved to have a pivotal role in the development and progression of both AF and CHF, less attention has been paid to the cellular immunity. Therefore we prospectively enrolled 112 patients with CHF and performed fluorescein-activated cell sorting (FACS). Patients were stratified in two subgroups according to patients presenting with AF (n=56) and patients free of AF (n=56). Comparing AF to non-AF patients we found a significantly lower fraction of regulatory T cells (p<0.001) in patients presenting with AF. However there was a higher fraction of CD4+ cells (p=0.007) and more specifically a significantly higher number of cytotoxic T cells characterised by the loss of CD28 within CD4 T cells (CD4+CD28null; p=0.035) in individuals with AF. After a mean follow-up time of 4.5 years 32 (28.6 %) patients died due to cardiovascular causes. CD4+CD28null cells were significantly associated with cardiovascular mortality in patients presenting with AF, with an adjusted HR per one standard deviation (1-SD) of 1.59 (95 % CI 1.13–2.24; p=0.008), but not in patients free of AF with an adjusted HR per 1-SD of 1.27 (95 % CI 0.86–1.87; p=0.216). We found that the fraction of CD4+CD28null cells proved to be predictive on outcome in CHF-patients presenting with AF. Our results might indicate a potential role of CD4+CD28null cells in the pathogenesis of AF which needs to be confirmed in future studies.Supplementary Material to this article is available online at www.thrombosis-online.com.
Collapse
|
19
|
Ikonomidis I, Papadavid E, Makavos G, Andreadou I, Varoudi M, Gravanis K, Theodoropoulos K, Pavlidis G, Triantafyllidi H, Moutsatsou P, Panagiotou C, Parissis J, Iliodromitis E, Lekakis J, Rigopoulos D. Lowering Interleukin-12 Activity Improves Myocardial and Vascular Function Compared With Tumor Necrosis Factor-a Antagonism or Cyclosporine in Psoriasis. Circ Cardiovasc Imaging 2017; 10:CIRCIMAGING.117.006283. [DOI: 10.1161/circimaging.117.006283] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 07/13/2017] [Indexed: 12/16/2022]
Abstract
Background—
Interleukin (IL)-12 activity is involved in the pathogenesis of psoriasis and acute coronary syndromes. We investigated the effects of IL-12 inhibition on vascular and left ventricular (LV) function in psoriasis.
Methods and Results—
One hundred fifty psoriasis patients were randomized to receive an anti–IL-12/23 (ustekinumab, n=50), anti–tumor necrosis factor-a (TNF-α; etanercept, n=50), or cyclosporine treatment (n=50). At baseline and 4 months post-treatment, we measured (1) LV global longitudinal strain, twisting, and percent difference between peak twisting and untwisting at mitral valve opening (%untwMVO) using speckle-tracking echocardiography, (2) coronary flow reserve, (3) pulse wave velocity and augmentation index, (4) circulating NT-proBNP (N-terminal pro-B-type natriuretic peptide), TNF-α, IL-6, IL-12, IL-17, malondialdehyde, and fetuin-a. Compared with baseline, all patients had improved global longitudinal strain (median values: −17.7% versus −19.5%), LV twisting (12.4° versus 14°), %untwMVO (27.8% versus 35%), and coronary flow reserve (2.8 versus 3.1) and reduced circulating NT-proBNP, IL-17, TNF-α, and IL-6 post-treatment (
P
<0.05). Compared with anti–TNF-α and cyclosporine, anti–IL-12/23 treatment resulted in a greater improvement of global longitudinal strain (25% versus 17% versus 6%,), LV twist (27% versus 17% versus 1%), %untwMVO (31% versus 27% versus 17%), and coronary flow reserve (14% versus 11% versus 4%), as well as a greater reduction of IL-12 (−25% versus −4% versus −2%), malondialdehyde (−27% versus +5% versus +26%), and NT-proBNP(−26% versus −13.6% versus 9.1%) and increase of fetuin-a (
P
<0.01). Pulse wave velocity and augmentation index were improved only after anti–IL-12/23 treatment and correlated with changes in global longitudinal strain, LV twisting–untwisting (
P
<0.05).
Conclusions—
In psoriasis, IL-12/23 inhibition results in a greater improvement of coronary, arterial, and myocardial function than TNF-α inhibition or cyclosporine treatment.
Clinical Trial Registration—
URL:
http://www.clinicaltrials.gov
. Unique identifier: NCT02144857.
Collapse
Affiliation(s)
- Ignatios Ikonomidis
- From the Second Department of Cardiology (I.I., G.M., M.V., G.P., H.T., J.P., E.I., J.L.), Second Department of Dermatology and Venereology (E.P., K.T., D.R.), Department of Biological Chemistry (P.M., C.P.), and Department of Clinical Biochemistry (P.M., C.P.), Attikon Hospital, National and Kapodistrian University of Athens Medical School, Greece; and Department of Pharmaceutical Chemistry, National and Kapodistrian University of Athens School of Pharmacy, Greece (I.A., K.G.)
| | - Evangelia Papadavid
- From the Second Department of Cardiology (I.I., G.M., M.V., G.P., H.T., J.P., E.I., J.L.), Second Department of Dermatology and Venereology (E.P., K.T., D.R.), Department of Biological Chemistry (P.M., C.P.), and Department of Clinical Biochemistry (P.M., C.P.), Attikon Hospital, National and Kapodistrian University of Athens Medical School, Greece; and Department of Pharmaceutical Chemistry, National and Kapodistrian University of Athens School of Pharmacy, Greece (I.A., K.G.)
| | - George Makavos
- From the Second Department of Cardiology (I.I., G.M., M.V., G.P., H.T., J.P., E.I., J.L.), Second Department of Dermatology and Venereology (E.P., K.T., D.R.), Department of Biological Chemistry (P.M., C.P.), and Department of Clinical Biochemistry (P.M., C.P.), Attikon Hospital, National and Kapodistrian University of Athens Medical School, Greece; and Department of Pharmaceutical Chemistry, National and Kapodistrian University of Athens School of Pharmacy, Greece (I.A., K.G.)
| | - Ioanna Andreadou
- From the Second Department of Cardiology (I.I., G.M., M.V., G.P., H.T., J.P., E.I., J.L.), Second Department of Dermatology and Venereology (E.P., K.T., D.R.), Department of Biological Chemistry (P.M., C.P.), and Department of Clinical Biochemistry (P.M., C.P.), Attikon Hospital, National and Kapodistrian University of Athens Medical School, Greece; and Department of Pharmaceutical Chemistry, National and Kapodistrian University of Athens School of Pharmacy, Greece (I.A., K.G.)
| | - Maria Varoudi
- From the Second Department of Cardiology (I.I., G.M., M.V., G.P., H.T., J.P., E.I., J.L.), Second Department of Dermatology and Venereology (E.P., K.T., D.R.), Department of Biological Chemistry (P.M., C.P.), and Department of Clinical Biochemistry (P.M., C.P.), Attikon Hospital, National and Kapodistrian University of Athens Medical School, Greece; and Department of Pharmaceutical Chemistry, National and Kapodistrian University of Athens School of Pharmacy, Greece (I.A., K.G.)
| | - Kostas Gravanis
- From the Second Department of Cardiology (I.I., G.M., M.V., G.P., H.T., J.P., E.I., J.L.), Second Department of Dermatology and Venereology (E.P., K.T., D.R.), Department of Biological Chemistry (P.M., C.P.), and Department of Clinical Biochemistry (P.M., C.P.), Attikon Hospital, National and Kapodistrian University of Athens Medical School, Greece; and Department of Pharmaceutical Chemistry, National and Kapodistrian University of Athens School of Pharmacy, Greece (I.A., K.G.)
| | - Kostas Theodoropoulos
- From the Second Department of Cardiology (I.I., G.M., M.V., G.P., H.T., J.P., E.I., J.L.), Second Department of Dermatology and Venereology (E.P., K.T., D.R.), Department of Biological Chemistry (P.M., C.P.), and Department of Clinical Biochemistry (P.M., C.P.), Attikon Hospital, National and Kapodistrian University of Athens Medical School, Greece; and Department of Pharmaceutical Chemistry, National and Kapodistrian University of Athens School of Pharmacy, Greece (I.A., K.G.)
| | - George Pavlidis
- From the Second Department of Cardiology (I.I., G.M., M.V., G.P., H.T., J.P., E.I., J.L.), Second Department of Dermatology and Venereology (E.P., K.T., D.R.), Department of Biological Chemistry (P.M., C.P.), and Department of Clinical Biochemistry (P.M., C.P.), Attikon Hospital, National and Kapodistrian University of Athens Medical School, Greece; and Department of Pharmaceutical Chemistry, National and Kapodistrian University of Athens School of Pharmacy, Greece (I.A., K.G.)
| | - Helen Triantafyllidi
- From the Second Department of Cardiology (I.I., G.M., M.V., G.P., H.T., J.P., E.I., J.L.), Second Department of Dermatology and Venereology (E.P., K.T., D.R.), Department of Biological Chemistry (P.M., C.P.), and Department of Clinical Biochemistry (P.M., C.P.), Attikon Hospital, National and Kapodistrian University of Athens Medical School, Greece; and Department of Pharmaceutical Chemistry, National and Kapodistrian University of Athens School of Pharmacy, Greece (I.A., K.G.)
| | - Paraskevi Moutsatsou
- From the Second Department of Cardiology (I.I., G.M., M.V., G.P., H.T., J.P., E.I., J.L.), Second Department of Dermatology and Venereology (E.P., K.T., D.R.), Department of Biological Chemistry (P.M., C.P.), and Department of Clinical Biochemistry (P.M., C.P.), Attikon Hospital, National and Kapodistrian University of Athens Medical School, Greece; and Department of Pharmaceutical Chemistry, National and Kapodistrian University of Athens School of Pharmacy, Greece (I.A., K.G.)
| | - Christina Panagiotou
- From the Second Department of Cardiology (I.I., G.M., M.V., G.P., H.T., J.P., E.I., J.L.), Second Department of Dermatology and Venereology (E.P., K.T., D.R.), Department of Biological Chemistry (P.M., C.P.), and Department of Clinical Biochemistry (P.M., C.P.), Attikon Hospital, National and Kapodistrian University of Athens Medical School, Greece; and Department of Pharmaceutical Chemistry, National and Kapodistrian University of Athens School of Pharmacy, Greece (I.A., K.G.)
| | - John Parissis
- From the Second Department of Cardiology (I.I., G.M., M.V., G.P., H.T., J.P., E.I., J.L.), Second Department of Dermatology and Venereology (E.P., K.T., D.R.), Department of Biological Chemistry (P.M., C.P.), and Department of Clinical Biochemistry (P.M., C.P.), Attikon Hospital, National and Kapodistrian University of Athens Medical School, Greece; and Department of Pharmaceutical Chemistry, National and Kapodistrian University of Athens School of Pharmacy, Greece (I.A., K.G.)
| | - Efstathios Iliodromitis
- From the Second Department of Cardiology (I.I., G.M., M.V., G.P., H.T., J.P., E.I., J.L.), Second Department of Dermatology and Venereology (E.P., K.T., D.R.), Department of Biological Chemistry (P.M., C.P.), and Department of Clinical Biochemistry (P.M., C.P.), Attikon Hospital, National and Kapodistrian University of Athens Medical School, Greece; and Department of Pharmaceutical Chemistry, National and Kapodistrian University of Athens School of Pharmacy, Greece (I.A., K.G.)
| | - John Lekakis
- From the Second Department of Cardiology (I.I., G.M., M.V., G.P., H.T., J.P., E.I., J.L.), Second Department of Dermatology and Venereology (E.P., K.T., D.R.), Department of Biological Chemistry (P.M., C.P.), and Department of Clinical Biochemistry (P.M., C.P.), Attikon Hospital, National and Kapodistrian University of Athens Medical School, Greece; and Department of Pharmaceutical Chemistry, National and Kapodistrian University of Athens School of Pharmacy, Greece (I.A., K.G.)
| | - Dimitrios Rigopoulos
- From the Second Department of Cardiology (I.I., G.M., M.V., G.P., H.T., J.P., E.I., J.L.), Second Department of Dermatology and Venereology (E.P., K.T., D.R.), Department of Biological Chemistry (P.M., C.P.), and Department of Clinical Biochemistry (P.M., C.P.), Attikon Hospital, National and Kapodistrian University of Athens Medical School, Greece; and Department of Pharmaceutical Chemistry, National and Kapodistrian University of Athens School of Pharmacy, Greece (I.A., K.G.)
| |
Collapse
|
20
|
Flego D, Liuzzo G, Weyand CM, Crea F. Adaptive Immunity Dysregulation in Acute Coronary Syndromes: From Cellular and Molecular Basis to Clinical Implications. J Am Coll Cardiol 2016; 68:2107-2117. [PMID: 27810051 PMCID: PMC5651170 DOI: 10.1016/j.jacc.2016.08.036] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 08/09/2016] [Accepted: 08/16/2016] [Indexed: 11/25/2022]
Abstract
Although the early outcome of acute coronary syndrome (ACS) has considerably improved in the last decade, cardiovascular diseases still represent the main cause of morbidity and mortality worldwide. This is mainly because recurrence of ACS eventually leads to the pandemics of heart failure and sudden cardiac death, thus calling for a reappraisal of the mechanisms responsible for coronary instability. This review discusses recent advances in our understanding of how adaptive immunity contributes to the pathogenesis of ACS and the clinical implications that arise from these new pathogenic concepts.
Collapse
Affiliation(s)
- Davide Flego
- Institute of Cardiology, Catholic University, Rome, Italy
| | | | - Cornelia M Weyand
- Division of Immunology and Rheumatology, Stanford University, Stanford, California
| | - Filippo Crea
- Institute of Cardiology, Catholic University, Rome, Italy
| |
Collapse
|
21
|
Luo T, Hu J, Xi D, Xiong H, He W, Liu J, Li M, Lu H, Zhao J, Lai W, Guo Z. Lck Inhibits Heat Shock Protein 65-Mediated Reverse Cholesterol Transport in T Cells. THE JOURNAL OF IMMUNOLOGY 2016; 197:3861-3870. [PMID: 27742830 DOI: 10.4049/jimmunol.1502710] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 09/20/2016] [Indexed: 02/05/2023]
Abstract
Previously, we reported that heat shock protein (HSP)65 impairs the effects of high-density lipoprotein on macrophages. We also showed that immune response activation adversely affects reverse cholesterol transport (RCT). In this study, we investigated the effects of the Src family kinase lymphocyte-specific protein tyrosine kinase (Lck) and elucidated the mechanism underlying HSP65-regulated cholesterol efflux in T cells. We evaluated cell proliferation, Lck expression, and inflammatory cytokine production in Jurkat cells and CD4+ T cells. HSP65-mediated inhibition of RCT was assessed by evaluating ABCA1, ABCG1, SR-BI, PPAR-γ, and liver X receptor-α expression. A dose-dependent relationship was found between the levels of these proteins and the suppression of cholesterol efflux. Stimulation of Lck-silenced T cells with ionomycin resulted in a decrease in intracellular calcium levels. Treatment of Jurkat cells with PP2, an inhibitor of Src family kinase, inhibited calcium-induced, but not PMA-induced, ERK phosphorylation. NF-κB activation in response to PMA was minimally inhibited in cells stimulated with PP2. HSP65 failed to trigger downstream ERK or JNK phosphorylation or to activate NF-κB or protein kinase C-γ in Lck-silenced cells. Additionally, elevation of intracellular calcium was also impaired. However, HSP65 significantly enhanced cholesterol efflux and decreased cellular cholesterol content by inducing the expression of cholesterol transport proteins in Lck-silenced cells. The treatment of Jurkat cells with PP2 also inhibited cell proliferation and promoted RCT. In conclusion, Lck is a key molecule in the TCR signaling cascade that inhibits cholesterol efflux and upregulates intracellular cholesterol ester content in T cells. Our results demonstrate that the immune response plays a previously unrecognized role in RCT.
Collapse
Affiliation(s)
- Tiantian Luo
- Division of Cardiology, HuiQiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Jing Hu
- Division of Cardiology, HuiQiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Dan Xi
- Division of Cardiology, HuiQiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Haowei Xiong
- Division of Cardiology, HuiQiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Wenshuai He
- Division of Cardiology, HuiQiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Jichen Liu
- Division of Cardiology, HuiQiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Menghao Li
- Division of Cardiology, HuiQiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Hao Lu
- Division of Cardiology, HuiQiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Jinzhen Zhao
- Division of Cardiology, HuiQiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Wenyan Lai
- Division of Cardiology, HuiQiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Zhigang Guo
- Division of Cardiology, HuiQiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| |
Collapse
|
22
|
Sayed NM, Abdel-Rahman SM, Esmat I, Nammas W. CD4+CD28null T cells in acute coronary syndrome: lower with ST-elevation myocardial infarction. SCAND CARDIOVASC J 2015; 49:325-30. [PMID: 26375412 DOI: 10.3109/14017431.2015.1088167] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
OBJECTIVES We explored any possible difference in the percentage of CD4 + CD28null T cells between patients with ST-segment-elevation myocardial infarction (STEMI) and those with non-ST-segment-elevation acute coronary syndrome. DESIGN We enrolled 55 consecutive patients admitted with acute coronary syndrome, and 16 healthy control subjects. CD4 + CD28null T cell percentage was assayed by flow cytometry from blood samples withdrawn at the time of enrollment. RESULTS Mean age was 55.2 ± 10.8 years (69.1% males). Mean CD4 + CD28null T cell percentage was 5.9 ± 3.8% in the study cohort versus 0.8 ± 0.7% in controls (p < 0.001). Mean CD4 + CD28null T cell percentage was higher in patients presenting with non-ST-segment-elevation acute coronary syndrome versus those presenting with STEMI: 7.3 ± 4.1% versus 4.6 ± 3.1%, respectively (p = 0.008). Multivariable regression analysis identified the category of acute coronary syndrome as the only variable independently associated with CD4 + CD28null T cell percentage (p = 0.007). CONCLUSIONS In patients presenting with acute coronary syndrome, the CD4 + CD28null T cell percentage was higher in patients with non-ST-segment-elevation acute coronary syndrome versus those with STEMI. The category of acute coronary syndrome was the only variable independently associated with the CD4 + CD28null T cell percentage in multivariable regression analysis.
Collapse
Affiliation(s)
- Nehad M Sayed
- a Department of Medical Microbiology and Immunology , Faculty of Medicine, Ain Shams University , Cairo , Egypt
| | - Safaa M Abdel-Rahman
- a Department of Medical Microbiology and Immunology , Faculty of Medicine, Ain Shams University , Cairo , Egypt
| | - Iman Esmat
- b Cardiology Department, Faculty of Medicine , Ain Shams University , Cairo , Egypt
| | - Wail Nammas
- b Cardiology Department, Faculty of Medicine , Ain Shams University , Cairo , Egypt
| |
Collapse
|
23
|
Dumitriu IE. The life (and death) of CD4+ CD28(null) T cells in inflammatory diseases. Immunology 2015; 146:185-93. [PMID: 26190355 DOI: 10.1111/imm.12506] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 07/09/2015] [Accepted: 07/10/2015] [Indexed: 02/06/2023] Open
Abstract
Inflammation contributes to the development and perpetuation of several disorders and T lymphocytes orchestrate the inflammatory immune response. Although the role of T cells in inflammation is widely recognized, specific therapies that tackle inflammatory networks in disease are yet to be developed. CD4(+) CD28(null) T cells are a unique subset of helper T lymphocytes that recently shot back into the limelight as potential catalysts of inflammation in several inflammatory disorders such as autoimmunity, atherosclerosis and chronic viral infections. In contrast to conventional helper T cells, CD4(+) CD28(null) T cells have an inbuilt ability to release inflammatory cytokines and cytotoxic molecules that can damage tissues and amplify inflammatory pathways. It comes as no surprise that patients who have high numbers of these cells have more severe disease and poor prognosis. In this review, I provide an overview on the latest advances in the biology of CD4(+) CD28(null) T cells. Understanding the complex functions and dynamics of CD4(+) CD28(null) T cells may open new avenues for therapeutic intervention to prevent progression of inflammatory diseases.
Collapse
Affiliation(s)
- Ingrid E Dumitriu
- Cardiovascular and Cell Sciences Research Institute, St George's, University of London, London, UK
| |
Collapse
|
24
|
Persic V, Ruzic A, Miletic B, Samsa DT, Rakic M, Raljevic D, Pejcinovic VP, Eminovic S, Zaputovic L, Laskarin G. Granulysin Expression in Lymphocytes that Populate the Peripheral Blood and the Myocardium after an Acute Coronary Event. Scand J Immunol 2015; 75:231-42. [PMID: 21967803 DOI: 10.1111/j.1365-3083.2011.02646.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
We aimed to analyse granulysin (GNLY)-mediated cytotoxicity in the peripheral blood of patients with non-ST-segment elevation myocardial infarction (NSTEMI) treated with anti-ischaemic drug therapy. Thirty-nine NSTEMI patients with a median age of 70 years and 28 age-matched healthy subjects were enrolled in this study. On day 7 after MI, the number of GNLY(+) lymphocytes in the peripheral blood increased approximately six-fold of that in the healthy subjects, measured by flow cytometry. On day 14, the number of GNLY(+) cells significantly decreased in T, NKT, and both CD56(+dim) and CD56(+bright) NK subsets. GNLY(+) CD3(+) and GNLY(+) CD56(+) cells infiltrated central zone of myocardial infarction (MI). In persons who died in the first week after MI, GNLY(+) cells were found within accumulation of apoptotic leucocytes and reached the apoptotic cardiomyocytes in border MI zones probably due to the influence of interleukin-15 in peri-necrotic cardiomyocytes, as it is was shown by immunohistology. By day 28, the percentage of GNLY(+) lymphocytes in peripheral blood returned to the levels similar to that of the healthy subjects. Anti-GNLY mAb decreased apoptosis of K562 targets using peripheral blood NK cells from days 7 and 28 after MI, while in assays using cells from days 1 and 21, both anti-GNLY and anti-perforin mAbs were required to significantly decrease apoptosis. Using NK cells from day 14, K562 apoptosis was nearly absent. In conclusion, it seems that GNLY(+) lymphocytes, probably attracted by IL-15, not only participate partially in myocardial cell apoptosis, but also hasten resolution of cardiac leucocyte infiltration in patients with NSTEMI.
Collapse
Affiliation(s)
- V Persic
- Division of Cardiology, Hospital for Medical Rehabilitation of the Hearth and Lung Diseases and Rheumatism "Thalassotherapija" Opatija, Opatija, CroatiaDepartment of Medical Rehabilitation, Medical Faculty, University of Rijeka, Rijeka, CroatiaDepartment of Pathology, Medical Faculty, University of Rijeka, Rijeka, CroatiaDivision of Cardiology Clinical Hospital Center Rijeka, Rijeka, CroatiaDepartment of Internal Medicine, Medical Faculty, University of Rijeka, Rijeka, CroatiaDepartment of Physiology and Immunology, Medical Faculty, University of Rijeka, Rijeka, Croatia
| | - A Ruzic
- Division of Cardiology, Hospital for Medical Rehabilitation of the Hearth and Lung Diseases and Rheumatism "Thalassotherapija" Opatija, Opatija, CroatiaDepartment of Medical Rehabilitation, Medical Faculty, University of Rijeka, Rijeka, CroatiaDepartment of Pathology, Medical Faculty, University of Rijeka, Rijeka, CroatiaDivision of Cardiology Clinical Hospital Center Rijeka, Rijeka, CroatiaDepartment of Internal Medicine, Medical Faculty, University of Rijeka, Rijeka, CroatiaDepartment of Physiology and Immunology, Medical Faculty, University of Rijeka, Rijeka, Croatia
| | - B Miletic
- Division of Cardiology, Hospital for Medical Rehabilitation of the Hearth and Lung Diseases and Rheumatism "Thalassotherapija" Opatija, Opatija, CroatiaDepartment of Medical Rehabilitation, Medical Faculty, University of Rijeka, Rijeka, CroatiaDepartment of Pathology, Medical Faculty, University of Rijeka, Rijeka, CroatiaDivision of Cardiology Clinical Hospital Center Rijeka, Rijeka, CroatiaDepartment of Internal Medicine, Medical Faculty, University of Rijeka, Rijeka, CroatiaDepartment of Physiology and Immunology, Medical Faculty, University of Rijeka, Rijeka, Croatia
| | - D Travica Samsa
- Division of Cardiology, Hospital for Medical Rehabilitation of the Hearth and Lung Diseases and Rheumatism "Thalassotherapija" Opatija, Opatija, CroatiaDepartment of Medical Rehabilitation, Medical Faculty, University of Rijeka, Rijeka, CroatiaDepartment of Pathology, Medical Faculty, University of Rijeka, Rijeka, CroatiaDivision of Cardiology Clinical Hospital Center Rijeka, Rijeka, CroatiaDepartment of Internal Medicine, Medical Faculty, University of Rijeka, Rijeka, CroatiaDepartment of Physiology and Immunology, Medical Faculty, University of Rijeka, Rijeka, Croatia
| | - M Rakic
- Division of Cardiology, Hospital for Medical Rehabilitation of the Hearth and Lung Diseases and Rheumatism "Thalassotherapija" Opatija, Opatija, CroatiaDepartment of Medical Rehabilitation, Medical Faculty, University of Rijeka, Rijeka, CroatiaDepartment of Pathology, Medical Faculty, University of Rijeka, Rijeka, CroatiaDivision of Cardiology Clinical Hospital Center Rijeka, Rijeka, CroatiaDepartment of Internal Medicine, Medical Faculty, University of Rijeka, Rijeka, CroatiaDepartment of Physiology and Immunology, Medical Faculty, University of Rijeka, Rijeka, Croatia
| | - D Raljevic
- Division of Cardiology, Hospital for Medical Rehabilitation of the Hearth and Lung Diseases and Rheumatism "Thalassotherapija" Opatija, Opatija, CroatiaDepartment of Medical Rehabilitation, Medical Faculty, University of Rijeka, Rijeka, CroatiaDepartment of Pathology, Medical Faculty, University of Rijeka, Rijeka, CroatiaDivision of Cardiology Clinical Hospital Center Rijeka, Rijeka, CroatiaDepartment of Internal Medicine, Medical Faculty, University of Rijeka, Rijeka, CroatiaDepartment of Physiology and Immunology, Medical Faculty, University of Rijeka, Rijeka, Croatia
| | - V Pehar Pejcinovic
- Division of Cardiology, Hospital for Medical Rehabilitation of the Hearth and Lung Diseases and Rheumatism "Thalassotherapija" Opatija, Opatija, CroatiaDepartment of Medical Rehabilitation, Medical Faculty, University of Rijeka, Rijeka, CroatiaDepartment of Pathology, Medical Faculty, University of Rijeka, Rijeka, CroatiaDivision of Cardiology Clinical Hospital Center Rijeka, Rijeka, CroatiaDepartment of Internal Medicine, Medical Faculty, University of Rijeka, Rijeka, CroatiaDepartment of Physiology and Immunology, Medical Faculty, University of Rijeka, Rijeka, Croatia
| | - S Eminovic
- Division of Cardiology, Hospital for Medical Rehabilitation of the Hearth and Lung Diseases and Rheumatism "Thalassotherapija" Opatija, Opatija, CroatiaDepartment of Medical Rehabilitation, Medical Faculty, University of Rijeka, Rijeka, CroatiaDepartment of Pathology, Medical Faculty, University of Rijeka, Rijeka, CroatiaDivision of Cardiology Clinical Hospital Center Rijeka, Rijeka, CroatiaDepartment of Internal Medicine, Medical Faculty, University of Rijeka, Rijeka, CroatiaDepartment of Physiology and Immunology, Medical Faculty, University of Rijeka, Rijeka, Croatia
| | - L Zaputovic
- Division of Cardiology, Hospital for Medical Rehabilitation of the Hearth and Lung Diseases and Rheumatism "Thalassotherapija" Opatija, Opatija, CroatiaDepartment of Medical Rehabilitation, Medical Faculty, University of Rijeka, Rijeka, CroatiaDepartment of Pathology, Medical Faculty, University of Rijeka, Rijeka, CroatiaDivision of Cardiology Clinical Hospital Center Rijeka, Rijeka, CroatiaDepartment of Internal Medicine, Medical Faculty, University of Rijeka, Rijeka, CroatiaDepartment of Physiology and Immunology, Medical Faculty, University of Rijeka, Rijeka, Croatia
| | - G Laskarin
- Division of Cardiology, Hospital for Medical Rehabilitation of the Hearth and Lung Diseases and Rheumatism "Thalassotherapija" Opatija, Opatija, CroatiaDepartment of Medical Rehabilitation, Medical Faculty, University of Rijeka, Rijeka, CroatiaDepartment of Pathology, Medical Faculty, University of Rijeka, Rijeka, CroatiaDivision of Cardiology Clinical Hospital Center Rijeka, Rijeka, CroatiaDepartment of Internal Medicine, Medical Faculty, University of Rijeka, Rijeka, CroatiaDepartment of Physiology and Immunology, Medical Faculty, University of Rijeka, Rijeka, Croatia
| |
Collapse
|
25
|
Duque JC, Martinez L, Mesa A, Wei Y, Tabbara M, Salman LH, Vazquez-Padron RI. CD4(+) lymphocytes improve venous blood flow in experimental arteriovenous fistulae. Surgery 2015; 158:529-36. [PMID: 25999254 DOI: 10.1016/j.surg.2015.02.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 02/16/2015] [Accepted: 02/21/2015] [Indexed: 02/02/2023]
Abstract
BACKGROUND The role of immune cells in arteriovenous fistulae (AVF) maturation is poorly understood and has received, until quite recently, little attention. This study examines the function of T lymphocytes in AVF vascular remodeling. METHODS Experimental fistulae were created in athymic rnu nude rats lacking mature T lymphocytes and euthymic control animals by anastomosing the left superior epigastric vein to the nearby femoral artery. Blood flow rates, wall morphology, and histologic changes were assessed in AVF 21 days after creation. The effect of CD4(+) lymphocytes on AVF maturation in athymic animals was analyzed by adoptive transfer of cells after fistula creation. RESULTS The absence of T lymphocytes compromised blood flow in experimental fistulae. Histopathologic inspection of AVF from athymic rats revealed that T-cell immunodeficiency negatively affected venous vascular remodeling, as evidenced by a reduced lumen, a thick muscular layer, and a low number of inflammatory cells compared with control animals. Adoptive transfer of CD4(+) lymphocytes from euthymic rats into athymic animals after fistula creation improved blood flow and reduced intima-media thickness. CONCLUSION These results point at the protective role of CD4(+) lymphocytes in the remodeling of the AVF vascular wall.
Collapse
Affiliation(s)
- Juan C Duque
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL
| | - Laisel Martinez
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL
| | - Annia Mesa
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL
| | - Yuntao Wei
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL
| | - Marwan Tabbara
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL
| | - Loay H Salman
- Section of Interventional Nephrology, University of Miami Miller School of Medicine, Miami, FL
| | - Roberto I Vazquez-Padron
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL.
| |
Collapse
|
26
|
Virtue A, Mai J, Wang H, Yang X. Lymphocytes and Atherosclerosis. Atherosclerosis 2015. [DOI: 10.1002/9781118828533.ch13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
27
|
Chistiakov DA, Orekhov AN, Bobryshev YV. Contribution of neovascularization and intraplaque haemorrhage to atherosclerotic plaque progression and instability. Acta Physiol (Oxf) 2015; 213:539-53. [PMID: 25515699 DOI: 10.1111/apha.12438] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Revised: 11/13/2014] [Accepted: 12/10/2014] [Indexed: 12/14/2022]
Abstract
Atherosclerosis is a continuous pathological process that starts early in life and progresses frequently to unstable plaques. Plaque rupture leads to deleterious consequences such as acute coronary syndrome, stroke and atherothrombosis. The vulnerable lesion has several structural and functional hallmarks that distinguish it from the stable plaque. The unstable plaque has large necrotic core (over 40% plaque volume) composed of cholesterol crystals, cholesterol esters, oxidized lipids, fibrin, erythrocytes and their remnants (haeme, iron, haemoglobin), and dying macrophages. The fibrous cap is thin, depleted of smooth muscle cells and collagen, and is infiltrated with proinflammatory cells. In unstable lesion, formation of neomicrovessels is increased. These neovessels have weak integrity and leak thereby leading to recurrent haemorrhages. Haemorrhages deliver erythrocytes to the necrotic core where they degrade promoting inflammation and oxidative stress. Inflammatory cells mostly presented by monocytes/macrophages, neutrophils and mast cells extravagate from bleeding neovessels and infiltrate adventitia where they support chronic inflammation. Plaque destabilization is an evolutionary process that could start at early atherosclerotic stages and whose progression is influenced by many factors including neovascularization, intraplaque haemorrhages, formation of cholesterol crystals, inflammation, oxidative stress and intraplaque protease activity.
Collapse
Affiliation(s)
- D. A. Chistiakov
- Department of Medical Nanobiotechnology; Pirogov Russian State Medical University; Moscow Russia
- The Mount Sinai Community Clinical Oncology Program; Mount Sinai Comprehensive Cancer Center; Mount Sinai Medical Center; Miami Beach FL USA
- Research Center for Children's Health; Moscow Russia
| | - A. N. Orekhov
- Laboratory of Angiopathology; Institute of General Pathology and Pathophysiology; Russian Academy of Sciences; Moscow Russia
- Skolkovo Innovative Center; Institute for Atherosclerosis Research; Moscow Russia
| | - Y. V. Bobryshev
- Laboratory of Angiopathology; Institute of General Pathology and Pathophysiology; Russian Academy of Sciences; Moscow Russia
- Faculty of Medicine and St Vincent's Centre for Applied Medical Research; University of New South Wales; Sydney NSW Australia
- School of Medicine; University of Western Sydney; Campbelltown NSW Australia
| |
Collapse
|
28
|
Chistiakov DA, Orekhov AN, Sobenin IA, Bobryshev YV. Plasmacytoid dendritic cells: development, functions, and role in atherosclerotic inflammation. Front Physiol 2014; 5:279. [PMID: 25120492 PMCID: PMC4110479 DOI: 10.3389/fphys.2014.00279] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 07/08/2014] [Indexed: 12/21/2022] Open
Abstract
Plasmacytoid dendritic cells (pDCs) are a specialized subset of DCs that links innate and adaptive immunity. They sense viral and bacterial pathogens and release high levels of Type I interferons (IFN-I) in response to infection. pDCs were shown to contribute to inflammatory responses in the steady state and in pathology. In atherosclerosis, pDCs are involved in priming vascular inflammation and atherogenesis through production of IFN-I and chemokines that attract inflammatory cells to inflamed sites. pDCs also contribute to the proinflammatory activation of effector T cells, cytotoxic T cells, and conventional DCs. However, tolerogenic populations of pDCs are found that suppress atherosclerosis-associated inflammation through down-regulation of function and proliferation of proinflammatory T cell subsets and induction of regulatory T cells with potent immunomodulatory properties. Notably, atheroprotective tolerogenic DCs could be induced by certain self-antigens or bacterial antigens that suggests for great therapeutic potential of these DCs for development of DC-based anti-atherogenic vaccines.
Collapse
Affiliation(s)
- Dimitry A. Chistiakov
- Department of Medical Nanobiotechnology, Pirogov Russian State Medical UniversityMoscow, Russia
| | - Alexander N. Orekhov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical SciencesMoscow, Russia
- Institute for Atherosclerosis Research, Skolkovo Innovative CenterMoscow, Russia
| | - Igor A. Sobenin
- Institute for Atherosclerosis Research, Skolkovo Innovative CenterMoscow, Russia
- Laboratory of Medical Genetics, Russian Cardiology Research and Production ComplexMoscow, Russia
| | - Yuri V. Bobryshev
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical SciencesMoscow, Russia
- Faculty of Medicine, University of New South WalesSydney, NSW, Australia
- School of Medicine, University of Western SydneyCampbelltown, NSW, Australia
| |
Collapse
|
29
|
Human metapneumovirus SH and G glycoproteins inhibit macropinocytosis-mediated entry into human dendritic cells and reduce CD4+ T cell activation. J Virol 2014; 88:6453-69. [PMID: 24672038 DOI: 10.1128/jvi.03261-13] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Human metapneumovirus (HMPV) is a major etiologic agent of respiratory disease worldwide. HMPV reinfections are common in healthy adults and children, suggesting that the protective immune response to HMPV is incomplete and short-lived. We used gene-deletion viruses to evaluate the role of the attachment G and small hydrophobic SH glycoproteins on virus uptake by primary human monocyte-derived dendritic cells (MDDC) in vitro and on subsequent MDDC maturation and activation of autologous T cells. HMPV with deletion of G and SH (ΔSHG) exhibited increased infectivity but had little effect on MDDC maturation. However, MDDC stimulated with ΔSHG induced increased proliferation of autologous Th1-polarized CD4(+) T cells. This effect was independent of virus replication. Increased T cell proliferation was strictly dependent on contact between virus-stimulated MDDC and CD4(+) T cells. Confocal microscopy revealed that deletion of SH and G was associated with an increased number of immunological synapses between memory CD4(+) T cells and virus-stimulated MDDC. Uptake of HMPV by MDDC was found to be primarily by macropinocytosis. Uptake of wild-type (WT) virus was reduced compared to that of ΔSHG, indicative of inhibition by the SH and G glycoproteins. In addition, DC-SIGN-mediated endocytosis provided a minor alternative pathway that depended on SH and/or G and thus operated only for WT. Altogether, our results show that SH and G glycoproteins reduce the ability of HMPV to be internalized by MDDC, resulting in a reduced ability of the HMPV-stimulated MDDC to activate CD4(+) T cells. This study describes a previously unknown mechanism of virus immune evasion. IMPORTANCE Human metapneumovirus (HMPV) is a major etiologic agent of respiratory disease worldwide. HMPV reinfections are common in healthy adults and children, suggesting that the protective immune response to HMPV is incomplete and short-lived. We found that HMPV attachment G and small hydrophobic SH glycoproteins reduce the ability of HMPV to be internalized by macropinocytosis into human dendritic cells (DC). This results in a reduced ability of the HMPV-stimulated DC to activate Th1-polarized CD4(+) T cells. These results contribute to a better understanding of the nature of incomplete protection against this important human respiratory virus, provide new information on the entry of HMPV into human cells, and describe a new mechanism of virus immune evasion.
Collapse
|
30
|
Advances in mechanisms, imaging and management of the unstable plaque. Atherosclerosis 2014; 233:467-477. [PMID: 24530781 DOI: 10.1016/j.atherosclerosis.2014.01.036] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 01/14/2014] [Accepted: 01/15/2014] [Indexed: 01/09/2023]
Abstract
Post-mortem observations demonstrated that plaque fissure was the final event leading to coronary thrombosis and occlusion in about two-thirds of cases of sudden coronary death. Plaques prone to fissure have, therefore, been defined "vulnerable plaques" and are identified by specific anatomic features including thin inflamed fibrous cap, large lipidic core and positive remodeling. Accordingly, elegant imaging modalities have been developed in order to identify this "holy grail". However, the results of prognostic studies based on the identification of vulnerable plaques have not been encouraging because of the low positive predictive value for major cardiovascular events. This observation is not surprising as the pathogenesis of acute coronary syndromes is complex and multifactorial. In this review we propose a pathogenetic classification of acute coronary syndromes in the attempt to identify homogeneous groups of patients with a common mechanism of coronary instability which can be identified by using specific biomarkers and imaging techniques, and become a specific therapeutic target.
Collapse
|
31
|
Moro F, Morciano A, Tropea A, Sagnella F, Palla C, Scarinci E, Ciardulli A, Martinez D, Familiari A, Liuzzo G, Tritarelli A, Cosentino N, Niccoli G, Crea F, Lanzone A, Apa R. Effects of drospirenone-ethinylestradiol and/or metformin on CD4(+)CD28(null) T lymphocytes frequency in women with hyperinsulinemia having polycystic ovary syndrome: a randomized clinical trial. Reprod Sci 2013; 20:1508-17. [PMID: 23690338 DOI: 10.1177/1933719113488444] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
OBJECTIVE To evaluate the long-term effects of drospirenone (DRSP)/ethinylestradiol (EE) alone, metformin alone, and DRSP/EE-metformin on CD4(+)CD28(null) T lymphocytes frequency, a cardiovascular risk marker, in patients with hyperinsulinemic polycystic ovary syndrome (PCOS). DESIGN Randomized clinical trial. INTERVENTIONS Ninety three patients with hyperinsulinemic PCOS were age matched and body mass index matched and randomized to receive a 6 months daily treatment with DRSP (3 mg)/EE (0.03 mg), or metformin (1500 mg), or DRSP/EE combined with metformin. MAIN OUTCOME MEASURES CD4(+)CD28(null) T-cell frequencies. RESULTS The DRSP/EE and metformin groups did not show any significant change in the CD4(+)CD28(null) frequency compared to the baseline. Interestingly, a statistically significant decrease in CD4(+)CD28(null) frequency occurred after 6 months of DRSP/EE-metformin (median 3-1.5; P < .01). Of note, this statistically significant association was confirmed after adjusting for baseline values in DRSP/EE-metformin group by analysis of covariance (P < .05). CONCLUSIONS In women with hyperinsulinemic PCOS, combined therapy with DRSP/EE and metformin may reduce cardiovascular risk.
Collapse
Affiliation(s)
- Francesca Moro
- 1Institute of Obstetrics and Gynaecology, Università Cattolica del Sacro Cuore, Rome, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Polymorphonuclear neutrophils and instability of the atherosclerotic plaque: a causative role? Inflamm Res 2013; 62:537-50. [DOI: 10.1007/s00011-013-0617-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Revised: 03/18/2013] [Accepted: 03/20/2013] [Indexed: 12/20/2022] Open
|
33
|
Lymphocyte subset characterization in patients with early clinical presentation of coronary heart disease. J Thromb Thrombolysis 2013; 34:475-82. [PMID: 22903683 DOI: 10.1007/s11239-012-0761-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
This study aimed to investigate lymphocyte populations in non-diabetic patients with early clinical presentation of coronary heart disease (CHD). Twenty-five consecutive middle-aged (<55 years) out-patients with CHD (acute myocardial infarction in the previous 3 months) and stable clinical conditions (>1 month) underwent venous blood sampling in order to determinate CD3+ (T-lymphocytes), CD19+ (B-lymphocytes), CD4+ (helper/inducer lymphocytes) and CD8+ (suppressor/cytotoxic lymphocytes) populations. Patients with diabetes, heart failure symptoms, recent revascularization (<30 days) were excluded. Twenty-five patients matched for age, gender and risk factors were enrolled as controls. All patients with CHD previously underwent coronary angiography. CHD patients showed lower CD3+ levels (70.96 ± 4.72 vs. 74.12 ± 4.93 %, p < 0.05) and CD8+ (37.80 ± 7.15 vs. 46.36 ± 5.22 %, p < 0.001) but higher CD4+ (37.32 ± 7.99 vs. 31.64 ± 4.72 %, p < 0.01) and CD4+/CD8+ ratio (1.06 ± 0.43 vs. 0.69 ± 0.13, p < 0.001). Difference in CD19+ levels was not statistically significant. Subjects with an impaired (≤55 %) left ventricular ejection fraction were characterized by lower levels of CD8+ (33.23 ± 7.04 vs. 43.76 ± 7.40 %, p < 0.05) and higher levels of CD4+ (38.31 ± 8.23 vs. 32.73 ± 6.08 %, p < 0.05) and CD4+/CD8+ ratio (1.06 ± 0.38 vs. 0.79 ± 0.34, p < 0.05). CD8+ levels inversely related to severity of coronary atherosclerosis (r = -0.37, p < 0.01). In conclusion, subjects with early clinical presentation of CHD are characterized by an altered CD4+/CD8+ ratio and lower CD3+ levels.
Collapse
|
34
|
Pathogenesis of Acute Coronary Syndromes. J Am Coll Cardiol 2013; 61:1-11. [DOI: 10.1016/j.jacc.2012.07.064] [Citation(s) in RCA: 170] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2012] [Revised: 07/05/2012] [Accepted: 07/10/2012] [Indexed: 02/02/2023]
|
35
|
Pedicino D, Liuzzo G, Trotta F, Giglio AF, Giubilato S, Martini F, Zaccardi F, Scavone G, Previtero M, Massaro G, Cialdella P, Cardillo MT, Pitocco D, Ghirlanda G, Crea F. Adaptive immunity, inflammation, and cardiovascular complications in type 1 and type 2 diabetes mellitus. J Diabetes Res 2013; 2013:184258. [PMID: 23762872 PMCID: PMC3676957 DOI: 10.1155/2013/184258] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Accepted: 04/05/2013] [Indexed: 12/28/2022] Open
Abstract
Diabetes mellitus (DM) is a pandemics that affects more than 170 million people worldwide, associated with increased mortality and morbidity due to coronary artery disease (CAD). In type 1 (T1) DM, the main pathogenic mechanism seems to be the destruction of pancreatic β -cells mediated by autoreactive T-cells resulting in chronic insulitis, while in type 2 (T2) DM primary insulin resistance, rather than defective insulin production due to β -cell destruction, seems to be the triggering alteration. In our study, we investigated the role of systemic inflammation and T-cell subsets in T1- and T2DM and the possible mechanisms underlying the increased cardiovascular risk associated with these diseases.
Collapse
Affiliation(s)
- Daniela Pedicino
- Institute of Cardiology, Catholic University, Largo A. Gemelli, 8-00168 Rome, Italy
| | - Giovanna Liuzzo
- Institute of Cardiology, Catholic University, Largo A. Gemelli, 8-00168 Rome, Italy
- *Giovanna Liuzzo:
| | - Francesco Trotta
- Institute of Cardiology, Catholic University, Largo A. Gemelli, 8-00168 Rome, Italy
| | - Ada Francesca Giglio
- Institute of Cardiology, Catholic University, Largo A. Gemelli, 8-00168 Rome, Italy
| | - Simona Giubilato
- Institute of Cardiology, Catholic University, Largo A. Gemelli, 8-00168 Rome, Italy
| | - Francesca Martini
- Diabetes Care Unit, Internal Medicine, Catholic University, Largo A. Gemelli, 8-00168 Rome, Italy
| | - Francesco Zaccardi
- Diabetes Care Unit, Internal Medicine, Catholic University, Largo A. Gemelli, 8-00168 Rome, Italy
| | - Giuseppe Scavone
- Diabetes Care Unit, Internal Medicine, Catholic University, Largo A. Gemelli, 8-00168 Rome, Italy
| | - Marco Previtero
- Institute of Cardiology, Catholic University, Largo A. Gemelli, 8-00168 Rome, Italy
| | - Gianluca Massaro
- Institute of Cardiology, Catholic University, Largo A. Gemelli, 8-00168 Rome, Italy
| | - Pio Cialdella
- Institute of Cardiology, Catholic University, Largo A. Gemelli, 8-00168 Rome, Italy
| | | | - Dario Pitocco
- Diabetes Care Unit, Internal Medicine, Catholic University, Largo A. Gemelli, 8-00168 Rome, Italy
| | - Giovanni Ghirlanda
- Diabetes Care Unit, Internal Medicine, Catholic University, Largo A. Gemelli, 8-00168 Rome, Italy
| | - Filippo Crea
- Institute of Cardiology, Catholic University, Largo A. Gemelli, 8-00168 Rome, Italy
| |
Collapse
|
36
|
Shah PK. Inflammation and atherothrombosis. Clin Immunol 2013. [DOI: 10.1016/b978-0-7234-3691-1.00082-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
37
|
Laskarin G, Persic V, Miškulin R, Ruzic A, Zaputovic L. Can we assess an acute myocardial infarction in patients with acute coronary syndrome according to diagnostic accuracy of heat shock proteins? Med Hypotheses 2012; 79:592-4. [DOI: 10.1016/j.mehy.2012.07.026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2012] [Accepted: 07/24/2012] [Indexed: 10/27/2022]
|
38
|
Moro F, Morciano A, Tropea A, Sagnella F, Palla C, Scarinci E, Cosentino N, Niccoli G, Liuzzo G, Crea F, Lanzone A, Apa R. CD4(+)CD28(null) T lymphocyte frequency, a new marker of cardiovascular risk: relationship with polycystic ovary syndrome phenotypes. Fertil Steril 2012; 98:1609-15. [PMID: 22963806 DOI: 10.1016/j.fertnstert.2012.08.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Revised: 08/10/2012] [Accepted: 08/10/2012] [Indexed: 11/29/2022]
Abstract
OBJECTIVE To study the frequency of CD4(+)CD28(null) T cells, which are aggressive T lymphocytes associated with recurrent coronary instability and type 2 diabetes mellitus, in different polycystic ovary syndrome (PCOS) phenotypes and in age- and body mass index-matched healthy women. DESIGN Retrospective cohort observational study. SETTING Unit of human reproductive pathophysiology, university hospital. PATIENT(S) A total of 167 PCOS patients and 102 control subjects. INTERVENTION(S) None. MAIN OUTCOME MEASURE(S) CD4(+)CD28(null) T cell frequency, high-sensitive C-reactive protein levels, and other glucose-metabolic parameters. RESULT(S) CD4(+)CD28(null) frequency was significantly higher in all PCOS groups than in control subjects. CD4(+)CD28(null) frequency was significantly higher in nonhyperandrogenic phenotype (5.7%, range 3.2-7.1) than in phenotypes with hyperandrogenism (H) + oligoamenorrhea (O) + polycystic ovary (PCO) (3.5%, range 1-5.8), H + O (3%, range 1.8-4.7), and H + PCO (2.63%, range 1.2-4.1). The relative risk of non-H phenotype for PCOS women in the highest quartile for CD4(+)CD28(null) frequency compared with PCOS women with the lowest quartile was 3.2 (95% confidence interval 1.9-5.8). CONCLUSION(S) Cardiovascular risk evaluation should be performed in all PCOS phenotypes. In particular, we demonstrated that the non-H phenotype has potentially increased cardiovascular risk in terms of CD4(+)CD28(null) frequency.
Collapse
Affiliation(s)
- Francesca Moro
- Institute of Obstetrics and Gynecology, Università Cattolica del Sacro Cuore, Rome, Italy.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Laskarin G, Zaputovic L, Persic V, Ruzic A, Sotosek Tokmadzic V. Harmful immune reactions during acute myocardial infarction. Med Hypotheses 2012; 78:703-6. [DOI: 10.1016/j.mehy.2012.02.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2011] [Accepted: 02/12/2012] [Indexed: 10/28/2022]
|
40
|
Immune aging and autoimmunity. Cell Mol Life Sci 2012; 69:1615-23. [PMID: 22466672 DOI: 10.1007/s00018-012-0970-0] [Citation(s) in RCA: 192] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2012] [Revised: 03/13/2012] [Accepted: 03/13/2012] [Indexed: 01/09/2023]
Abstract
Age is an important risk for autoimmunity, and many autoimmune diseases preferentially occur in the second half of adulthood when immune competence has declined and thymic T cell generation has ceased. Many tolerance checkpoints have to fail for an autoimmune disease to develop, and several of those are susceptible to the immune aging process. Homeostatic T cell proliferation which is mainly responsible for T cell replenishment during adulthood can lead to the selection of T cells with increased affinity to self- or neoantigens and enhanced growth and survival properties. These cells can acquire a memory-like phenotype, in particular under lymphopenic conditions. Accumulation of end-differentiated effector T cells, either specific for self-antigen or for latent viruses, have a low activation threshold due to the expression of signaling and regulatory molecules and generate an inflammatory environment with their ability to be cytotoxic and to produce excessive amounts of cytokines and thereby inducing or amplifying autoimmune responses.
Collapse
|
41
|
Bennett M, Yu H, Clarke M. Signalling from dead cells drives inflammation and vessel remodelling. Vascul Pharmacol 2012; 56:187-92. [PMID: 22306421 DOI: 10.1016/j.vph.2012.01.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2011] [Revised: 01/17/2012] [Accepted: 01/21/2012] [Indexed: 11/16/2022]
Abstract
Death of vascular smooth muscle cells (VSMCs) has been demonstrated in vessel development and in disease, most notably in atherosclerosis, but also after injury and remodelling. VSMC death promotes multiple features of vulnerable plaques, but also induces features of normal vessel ageing and cystic medial necrosis, including loss of VSMCs, elastin fragmentation and loss, increased glycosaminoglycans and speckled calcification. VSMC apoptosis in the absence of efficient phagocytosis also produces inflammation due to secondary necrosis; in contrast, VSMC apoptosis in normal vessels can be silent. We have investigated the consequences of VSMC apoptosis in both disease and during vessel remodelling. We find that VSMCs release specific cytokines dependent upon the mode of cell death; IL-1β predominates during apoptosis, whilst IL-1α predominates during necrosis. Both IL-1α and β promote release of further cytokines from adjacent live cells, in particular IL-6 and MCP-1. The balance of cytokines results in pathology with differing compositions, including inflammation or neointima formation/vascular repair, via direct promotion of VSMC proliferation and migration. Thus, VSMC death can promote either pathology or repair, depending upon the context and cytokine signalling.
Collapse
Affiliation(s)
- Martin Bennett
- Division of Cardiovascular Medicine, University of Cambridge, Addenbrooke's Centre for Clinical Investigation, Addenbrooke's Hospital, Cambridge, United Kingdom.
| | | | | |
Collapse
|
42
|
Alberts-Grill N, Rezvan A, Son DJ, Qiu H, Kim CW, Kemp ML, Weyand CM, Jo H. Dynamic immune cell accumulation during flow-induced atherogenesis in mouse carotid artery: an expanded flow cytometry method. Arterioscler Thromb Vasc Biol 2012; 32:623-32. [PMID: 22247254 DOI: 10.1161/atvbaha.111.242180] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Inflammation plays a central role in atherosclerosis. However, the detailed changes in the composition and quantity of leukocytes in the arterial wall during atherogenesis are not fully understood in part because of the lack of suitable methods and animal models. METHODS AND RESULTS We developed a 10-fluorochrome, 13-parameter flow cytometry method to quantitate 7 major leukocyte subsets in a single digested arterial wall sample. Apolipoprotein E-deficient mice underwent left carotid artery (LCA) partial ligation and were fed a high-fat diet for 4 to 28 days. Monocyte/macrophages, dendritic cells, granulocytes, natural killer cells, and CD4 T cells significantly infiltrated the LCA as early as 4 days. Monocyte/macrophages and dendritic cells decreased between 7 and 14 days, whereas T-cell numbers remained steady. Leukocyte numbers peaked at 7 days, preceding atheroma formation at 14 days. B cells entered LCA by 14 days. Control right carotid and sham-ligated LCAs showed no significant infiltrates. Polymerase chain reaction and ELISA arrays showed that expression of proinflammatory cytokines and chemokines peaked at 7 and 14 days postligation, respectively. CONCLUSION This is the first quantitative description of leukocyte number and composition over the life span of murine atherosclerosis. These results show that disturbed flow induces rapid and dynamic leukocyte accumulation in the arterial wall during the initiation and progression of atherosclerosis.
Collapse
Affiliation(s)
- Noah Alberts-Grill
- School of Medicine, Emory University, Woodruff Memorial Bldg, Rm 2005, Atlanta, GA 30322, USA
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Laskarin G, Persic V, Ruzic A, Miletic B, Rakic M, Samsa DT, Raljevic D, Pejcinovic VP, Miskulin R, Rukavina D. Perforin-mediated cytotoxicity in non-ST elevation myocardial infarction. Scand J Immunol 2011; 74:195-204. [PMID: 21388427 DOI: 10.1111/j.1365-3083.2011.02554.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The aim of this investigation was to examine the role of perforin (P)-mediated cytotoxicity in the dynamics of tissue damage in patients with non-ST-segment elevation myocardial infarction (NSTEMI) treated with anti-ischaemic drugs. We enrolled 48 patients with NSTEMI in this study [age, 71.5 years; 61.5/76 (median, 25th/75th percentiles)]. The percentage of total peripheral blood P(+) lymphocytes was elevated owing to the increased frequency of P(+) cells within natural killer (NK) subsets, T and NKT cells in patients on day 1 after NSTEMI when compared with healthy controls. Positive correlations were found between cardiac troponin I plasma concentrations and the frequency of P(+) cells, P(+) T cells, P(+) NK cells and their CD56(+dim) and CD56(+bright) subsets during the first week after the NSTEMI. The expression of P in NK cells was accompanied by P-mediated cytotoxicity against K-562 targets at all days examined, except day 21, when an anti-perforin monoclonal antibody did not completely abolish the killing. The percentage of P(+) T cells, P(+) NKT cells and P(+) NK subsets was the highest on the day 1 after NSTEMI and decreased in the post-infarction period. CD56(+) lymphocytes were found in damaged myocardium, suggesting their tissue recruitment. In conclusion, patients with NSTEMI have a strong and prolonged P-mediated systemic inflammatory reaction, which may sustain autoaggressive reactions towards myocardial tissue during the development of myocardial infarction.
Collapse
Affiliation(s)
- G Laskarin
- Division of Cardiology, Hospital for Medical Rehabilitation of Hearth and Lung Diseases and Rheumatism Thalassotherapia-Opatija, Opatija, Croatia.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Sun W, Zheng L, Huang L. Role of unusual CD4+ CD28- T cells in acute coronary syndrome. Mol Biol Rep 2011; 39:3337-42. [PMID: 21695428 DOI: 10.1007/s11033-011-1103-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2011] [Accepted: 06/15/2011] [Indexed: 12/13/2022]
Abstract
Acute coronary syndrome (ACS) is a group of clinical symptoms that results from complete or partial occlusive thrombus, which is caused by coronary an atherosclerotic plaque rupture or erosion. According to a recent study, CD4(+) CD28(-) T cells are found in atherosclerotic plaques and the peripheral circulation blood in patients with ACS, these cells play an important role in plaque ruptures. CD4(+) CD28(-) T cells are an unusual subset of helper cells, which expand and have harmful effects in ACS. In this review, we discuss the current issues on the generation of CD4(+) CD28(-) T cells and focus on their phenotypic and functional characteristics relevant to the development of cardiovascular events. Targeting the CD4(+) CD28(-) T cells subset in ACS could provide novel therapeutic means to prevent acute life-threatening coronary events.
Collapse
Affiliation(s)
- Wenjie Sun
- Department of Laboratory Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, People's Republic of China
| | | | | |
Collapse
|
45
|
Giubilato S, Liuzzo G, Brugaletta S, Pitocco D, Graziani F, Smaldone C, Montone RA, Pazzano V, Pedicino D, Biasucci LM, Ghirlanda G, Crea F. Expansion of CD4+CD28null T-lymphocytes in diabetic patients: exploring new pathogenetic mechanisms of increased cardiovascular risk in diabetes mellitus. Eur Heart J 2011; 32:1214-26. [DOI: 10.1093/eurheartj/ehq499] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
46
|
Zuidema MY, Zhang C. Ischemia/reperfusion injury: The role of immune cells. World J Cardiol 2010; 2:325-32. [PMID: 21160610 PMCID: PMC2999044 DOI: 10.4330/wjc.v2.i10.325] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2010] [Revised: 08/19/2010] [Accepted: 08/26/2010] [Indexed: 02/06/2023] Open
Abstract
Ischemia/reperfusion (I/R) injury is an inflammatory condition that is characterized by innate immunity and an adaptive immune response. This review is focused on the acute inflammatory response in I/R injury, and also the adaptive immunological mechanisms in chronic ischemic disease that lead to increased vulnerability during acute events, in relation to the cell types that have been shown to mediate innate immunity to an adaptive immune response in I/R, specifically myocardial infarction. Novel aspects are also highlighted in respect to the mechanisms within the cardiovascular system and cardiovascular risk factors that may be involved in the inflammatory response accompanying myocardial infarction. Experimental myocardial I/R has suggested that immune cells may mediate reperfusion injury. Specifically, monocytes, macrophages, T-cells, mast cells, platelets and endothelial cells are discussed with reference to the complement cascade, toll-like receptors, cytokines, oxidative stress, renin-angiotensin system, and in reference to the microvascular system in the signaling mechanisms of I/R. Finally, the findings of the data summarized in this review are most important for possible translation into clinical cardiology practice and possible avenues for drug development.
Collapse
Affiliation(s)
- Mozow Y Zuidema
- Mozow Y Zuidema, Cuihua Zhang, Division of Cardiovascular Disease, Dalton Cardiovascular Research Center, Department of Internal Medicine, Medical Pharmacology and Physiology and Nutritional Science, Dalton Cardiovascular Research Center, University of Missouri, Columbia, School of Medicine, Columbia, MO 65212, United States
| | | |
Collapse
|
47
|
Abstract
Immunologic models of rheumatoid arthritis (RA) have to take into account that the disease occurs at an age when immunocompetence is declining and in a host whose immune system shows evidence of accelerated immune aging. By several immune aging biomarkers, the immune system in patients with RA is prematurely aged by more than 20 years. One major pathogenetic mechanism is a defect in telomere maintenance and DNA repair that causes accelerated cell death. These findings in RA are reminiscent of murine autoimmunity models, in which lymphopenia was identified as a major risk factor for autoimmunity. Progress in the understanding of how accelerated immune aging is pathogenetically involved in RA may allow development of new therapeutic approaches that go beyond the use of anti-inflammatory agents and eventually could open new avenues for preventive intervention.
Collapse
Affiliation(s)
- Jorg J Goronzy
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University, 269 West Campus Drive, Stanford, CA 94305-5166, USA
| | | | | |
Collapse
|
48
|
Sato K, Nuki T, Gomita K, Weyand CM, Hagiwara N. Statins reduce endothelial cell apoptosis via inhibition of TRAIL expression on activated CD4 T cells in acute coronary syndrome. Atherosclerosis 2010; 213:33-9. [PMID: 20430391 DOI: 10.1016/j.atherosclerosis.2010.03.034] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2010] [Revised: 03/24/2010] [Accepted: 03/28/2010] [Indexed: 01/12/2023]
Abstract
OBJECTIVE Statins reduce cardiovascular-related morbidity and mortality, but their effects on inflammation in atherosclerosis are not fully understood. We investigated whether statins can modulate cytotoxic functions of CD4 T cells in acute coronary syndrome (ACS). METHODS AND RESULTS Fresh CD4 T cells were isolated from 55 patients with ACS without statin treatment on admission and from 34 age-matched controls. CD4 T cells collected from ACS patients induced endothelial cell apoptosis significantly more than control T cells. The TNF-related apoptosis-inducing ligand (TRAIL) receptor DR5 was strongly upregulated on endothelial cells, and TRAIL-specific antibodies effectively blocked CD4 T cell-mediated apoptosis, indicating that T cell-mediated endothelial death was dependent on the TRAIL pathway. Expression of both the activating antigen CD69 and TRAIL was enhanced on ACS T cells. In in-vitro assays rosuvastatin, fluvastatin, and pitavastatin directly blocked CD4 T cell-mediated endothelial cell apoptosis and reduced T cell-expression of CD69 and TRAIL through TCR-induced Extracellar signal-Regulated Kinases (ERK) activation. CONCLUSIONS Activated CD4 T cells expressing TRAIL are enriched in the blood of ACS patients and induce endothelial injury, which may contribute to the destabilization of the plaque. Early statin therapy may suppress T cell-mediated endothelial cell damage in atherosclerotic plaques and thus prevent cardiovascular events.
Collapse
Affiliation(s)
- Kayoko Sato
- Department of Cardiology, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjyuku-ku, Tokyo 162-8666, Japan.
| | | | | | | | | |
Collapse
|
49
|
Pryshchep S, Goronzy JJ, Parashar S, Weyand CM. Insufficient deactivation of the protein tyrosine kinase lck amplifies T-cell responsiveness in acute coronary syndrome. Circ Res 2009; 106:769-78. [PMID: 20035083 DOI: 10.1161/circresaha.109.206052] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
RATIONALE In the vulnerable atherosclerotic plaque, T cells may destabilize the tissue structure through direct cell-injurious effector functions. T cells transmit environmental signals, such as recognition of antigen, into cellular responses through regulated phosphorylation of cytoplasmic proteins, with the Src family kinase Lck (lymphocyte-specific protein tyrosine kinase) in critical membrane-proximal position of the T-cell receptor (TCR) signaling cascade. The balance between protein phosphorylation and dephosphorylation defines the signal transduction threshold and determines appropriate T-cell responses. OBJECTIVE We have examined whether abnormal calibration of intracellular signaling pathways renders acute coronary syndrome (ACS) patients susceptible to disproportionate T-cell responses. METHODS AND RESULTS Intracellular signaling cascades were quantified in CD4 T cells from ACS patients and control individuals after stimulation with major histocompatibility complex class II-superantigen complexes. ACS T cells mobilized more intracellular calcium and accumulated higher levels of phosphotyrosine than control T cells. Proximal steps in TCR signaling, such as recruitment of ZAP-70 and clustering of TCR complexes in the immune synapse, were abnormally enhanced in ACS T cells. Acceleration of the signaling cascade derived from a proximal defect in ACS T cells, which failed to phosphorylate Lck at Tyr505, extending activation of the Src kinase. Abnormalities in TCR signaling did not correlate with systemic inflammation as measured by C-reactive protein. CONCLUSIONS An intrinsic abnormality in the signaling machinery of ACS T cells resulting in the accumulation of active Lck lowers the TCR threshold and renders lymphocytes hyperreactive and capable of unwanted immune responses.
Collapse
Affiliation(s)
- Sergey Pryshchep
- Lowance Center for Human Immunology and Rheumatology, Emory University, Atlanta, GA, USA
| | | | | | | |
Collapse
|
50
|
Abstract
Giant cell arteritis (GCA), is a systemic vasculitis which preferentially targets large and medium branches of the upper-body aorta. Typical clinical manifestations result from arterial stenosis/occlusion causing blindness, stroke and aortic arch syndrome. Aortic involvement leads to dissection and aneurysm. On the cellular and molecular level, GCA is a sequel of abnormal innate and adaptive immune responses that occur in the specialized tissue niche of the arterial wall. Based on recent pathogenic studies, a novel disease model for GCA is emerging. It is now understood that the series of pathogenic events begins with dendritic cells (DC) indigenous to the artery's outer wall, leading to inflammatory vasculopathy. Placed close to the vasa vasorum, vascular DC are highly sensitive in recognizing pathogen-associated motifs assigning immune monitoring functions to blood vessels. Thus the large vessels are actively involved in immune monitoring. Each vascular territory expresses a unique profile of pathogen-sensing receptors, emphasizing functional diversity amongst structurally similar arteries. Innate immune stimulators can transform vascular DC into efficient antigen-presenting cells, attracting, activating, and instructing T lymphocytes to acquire tissue-invasive features. Macrophages provide critical tissue-damaging effector functions, directly injuring wall-residing cells and promoting a remodeling process that leads to intimal hyperplasia and luminal occlusion. Novel diagnostic and therapeutic approaches to GCA need to focus on the key position of vascular DC and the signals that break the immunoprivileged state of the vessel wall.
Collapse
|