1
|
Erdő-Bonyár S, Rapp J, Subicz R, Böröcz K, Szinger D, Filipánits K, Minier T, Kumánovics G, Czirják L, Berki T, Simon D. Disturbed Complement Receptor Expression Pattern of B Cells Is Enhanced by Toll-like Receptor CD180 Ligation in Diffuse Cutaneous Systemic Sclerosis. Int J Mol Sci 2024; 25:9230. [PMID: 39273179 PMCID: PMC11394765 DOI: 10.3390/ijms25179230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/23/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024] Open
Abstract
Autoantibody production is a hallmark of systemic sclerosis (SSc) and the most extensively studied role of B cells in the pathogenesis of the disease. However, the potential involvement of innate immune molecules in B-cell dysfunction in SSc is less understood. B-cell activation is an early event in the pathogenesis of SSc and is influenced by complement receptors (CRs) and Toll-like receptors (TLRs), shaping antibody responses. CR2 and CR1 modulate B-cell activation, and the roles of CR3 and CR4 are associated with autoimmune conditions. We investigated the expression of CRs in B cells from patients with the more severe form of the disease, diffuse cutaneous SSc (dcSSc), and the effect of TLR CD180 ligation on their expression. We found no significant difference in the basal expression of CD21 and CD11c in B cells between dcSSc and healthy controls (HCs). However, reduced basal CD11b expression in B cells in dcSSc compared to HCs, accompanied by a decrease in CD35 and an increase in CD11c expression following CD180 ligation may promote plasma cell formation and autoantibody production. Additionally, we searched for correlations between dcSSc-associated anti-DNA topoisomerase I (Scl-70) autoantibody, anti-citrate synthase (CS) natural autoantibody and complement component 3 (C3) levels and found a negative correlation between C3 and anti-CS autoantibody in dcSSc but not in HCs, supporting the hypothesis that natural autoantibodies could activate the complement system contributing to tissue injury in SSc.
Collapse
Affiliation(s)
- Szabina Erdő-Bonyár
- Department of Immunology and Biotechnology, Clinical Center, University of Pécs Medical School, H-7624 Pécs, Hungary
| | - Judit Rapp
- Department of Immunology and Biotechnology, Clinical Center, University of Pécs Medical School, H-7624 Pécs, Hungary
| | - Rovéna Subicz
- Department of Immunology and Biotechnology, Clinical Center, University of Pécs Medical School, H-7624 Pécs, Hungary
| | - Katalin Böröcz
- Department of Immunology and Biotechnology, Clinical Center, University of Pécs Medical School, H-7624 Pécs, Hungary
| | - Dávid Szinger
- Department of Immunology and Biotechnology, Clinical Center, University of Pécs Medical School, H-7624 Pécs, Hungary
| | - Kristóf Filipánits
- Department of Rheumatology and Immunology, Clinical Center, University of Pécs Medical School, H-7632 Pécs, Hungary
| | - Tünde Minier
- Department of Rheumatology and Immunology, Clinical Center, University of Pécs Medical School, H-7632 Pécs, Hungary
| | - Gábor Kumánovics
- Department of Rheumatology and Immunology, Clinical Center, University of Pécs Medical School, H-7632 Pécs, Hungary
| | - László Czirják
- Department of Rheumatology and Immunology, Clinical Center, University of Pécs Medical School, H-7632 Pécs, Hungary
| | - Tímea Berki
- Department of Immunology and Biotechnology, Clinical Center, University of Pécs Medical School, H-7624 Pécs, Hungary
| | - Diána Simon
- Department of Immunology and Biotechnology, Clinical Center, University of Pécs Medical School, H-7624 Pécs, Hungary
| |
Collapse
|
2
|
Wu S, Yin Y, Du L. The bidirectional relationship of depression and disturbances in B cell homeostasis: Double trouble. Prog Neuropsychopharmacol Biol Psychiatry 2024; 132:110993. [PMID: 38490433 DOI: 10.1016/j.pnpbp.2024.110993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 03/08/2024] [Accepted: 03/12/2024] [Indexed: 03/17/2024]
Abstract
Major depressive disorder (MDD) is a recurrent, persistent, and debilitating neuropsychiatric syndrome with an increasing morbidity and mortality, representing the leading cause of disability worldwide. The dysregulation of immune systems (including innate and adaptive immune systems) has been identified as one of the key contributing factors in the progression of MDD. As the main force of the humoral immunity, B cells have an essential role in the defense against infections, antitumor immunity and autoimmune diseases. Several recent studies have suggested an intriguing connection between disturbances in B cell homeostasis and the pathogenesis of MDD, however, the B-cell-dependent mechanism of MDD remains largely unexplored compared to other immune cells. In this review, we provide an overview of how B cell abnormality regulates the progression of MMD and the potential consequence of the disruption of B cell homeostasis in patients with MDD. Abnormalities of B-cell homeostasis not only promote susceptibility to MDD, but also lead to an increased risk of developing infection, malignancy and autoimmune diseases in patients with MDD. A better understanding of the contribution of B cells underlying MDD would provide opportunities for identification of more targeted treatment approaches and might provide an overall therapeutic benefit to improve the long-term outcomes of patients with MDD.
Collapse
Affiliation(s)
- Shusheng Wu
- Department of Neurology, Affiliated Hospital of Yangzhou University, Jiangsu, China
| | - Yuye Yin
- College of Bioscience and Biotechnology, Yangzhou University, Yangzhou, Jiangsu, China
| | - Longfei Du
- Department of Laboratory Medicine, Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu, China.
| |
Collapse
|
3
|
Żuber-Łaskawiec K, Wilańska J, Karska-Basta I, Pociej-Marciak W, Romanowska-Dixon B, Sanak M, Kubicka-Trząska A. Circulating Anti-Endothelial Cell Antibodies in Patients with Geographic Atrophy Related to Dry Age-Related Macular Degeneration. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:810. [PMID: 38792993 PMCID: PMC11122982 DOI: 10.3390/medicina60050810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 05/13/2024] [Accepted: 05/14/2024] [Indexed: 05/26/2024]
Abstract
Background and Objectives: Age-related macular degeneration (AMD) is one of the leading causes of central vision loss among elderly patients, and its dry form accounts for the majority of cases. Although several causes and mechanisms for the development and progression of AMD have previously been identified, the pathogenesis of this complex disease is still not entirely understood. As inflammation and immune system involvement are strongly suggested to play a central role in promoting the degenerative process and stimulating the onset of complications, we aimed to analyze the frequency of serum anti-retinal (ARAs) and anti-endothelial cell antibodies (AECAs) in patients with dry AMD and to determine their relationship with the clinical features of the disease, notably the area of geographic atrophy (GA). Materials and Methods: This study included 41 patients with advanced-stage dry AMD and 50 healthy controls without AMD, matched for gender and age. ARAs were detected by indirect immunofluorescence using monkey retina as an antigen substrate, and the presence of AECAs was determined using cultivated human umbilical vein endothelial cells and primate skeletal muscle. Results: ARAs were detected in 36 (87.8%) AMD patients (titers ranged from 1:20 to 1:320) and in 16 (39.0%) (titers ranged from 1:10 to 1:40) controls (p = 0.0000). Twenty of the forty-one patients (48.8%) were positive for AECAs, while in the control group, AECAs were present only in five sera (10.0%). The titers of AECAs in AMD patients ranged from 1:100 to 1:1000, and in the control group, the AECA titers were 1:100 (p = 0.0001). There were no significant correlations between the presence of AECAs and disease activity. Conclusions: This study demonstrates a higher prevalence of circulating AECAs in patients with dry AMD; however, no correlation was found between the serum levels of these autoantibodies and the area of GA.
Collapse
Affiliation(s)
- Katarzyna Żuber-Łaskawiec
- Chair of Ophthalmology, Faculty of Medicine, Medical College, Jagiellonian University, Kopernika Str. 38, 31-501 Krakow, Poland; (K.Ż.-Ł.)
- Clinic of Ophthalmology and Ocular Oncology, University Hospital, Kopernika Str. 38, 31-501 Krakow, Poland
| | - Joanna Wilańska
- Department of Molecular Biology and Clinical Genetics, II Chair of Internal Medicine, Medical College, Faculty of Medicine, Jagiellonian University, Skawińska Str. 8, 31-066 Krakow, Poland
| | - Izabella Karska-Basta
- Chair of Ophthalmology, Faculty of Medicine, Medical College, Jagiellonian University, Kopernika Str. 38, 31-501 Krakow, Poland; (K.Ż.-Ł.)
- Clinic of Ophthalmology and Ocular Oncology, University Hospital, Kopernika Str. 38, 31-501 Krakow, Poland
| | - Weronika Pociej-Marciak
- Chair of Ophthalmology, Faculty of Medicine, Medical College, Jagiellonian University, Kopernika Str. 38, 31-501 Krakow, Poland; (K.Ż.-Ł.)
- Clinic of Ophthalmology and Ocular Oncology, University Hospital, Kopernika Str. 38, 31-501 Krakow, Poland
| | - Bożena Romanowska-Dixon
- Chair of Ophthalmology, Faculty of Medicine, Medical College, Jagiellonian University, Kopernika Str. 38, 31-501 Krakow, Poland; (K.Ż.-Ł.)
- Clinic of Ophthalmology and Ocular Oncology, University Hospital, Kopernika Str. 38, 31-501 Krakow, Poland
| | - Marek Sanak
- Department of Molecular Biology and Clinical Genetics, II Chair of Internal Medicine, Medical College, Faculty of Medicine, Jagiellonian University, Skawińska Str. 8, 31-066 Krakow, Poland
| | - Agnieszka Kubicka-Trząska
- Chair of Ophthalmology, Faculty of Medicine, Medical College, Jagiellonian University, Kopernika Str. 38, 31-501 Krakow, Poland; (K.Ż.-Ł.)
- Clinic of Ophthalmology and Ocular Oncology, University Hospital, Kopernika Str. 38, 31-501 Krakow, Poland
| |
Collapse
|
4
|
Fiorelli D, Francavilla B, Velletrani G, Maurantonio S, Passali FM, Bernardini S, Di Girolamo S, Nuccetelli M. Autoantibody profiles assessment in individuals with persistent olfactory impairment following SARS-CoV-2 infection. Int Immunopharmacol 2024; 129:111599. [PMID: 38330796 DOI: 10.1016/j.intimp.2024.111599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 01/14/2024] [Accepted: 01/24/2024] [Indexed: 02/10/2024]
Abstract
BACKGROUND Olfactory impairment, particularly hyposmia and anosmia, has emerged as a distinctive early symptom of SARS-CoV-2. Drawing on the historical association of autoimmune diseases with olfactory function, this study delves into the connections between COVID-19, autoimmunity, and persistent olfactory dysfunctions, focusing on individuals experiencing long-lasting smell disorders (3-18 months post-SARS-CoV-2 infection). METHODS The study comprised 36 Long Covid patients with persistent olfactory dysfunctions, alongside two control groups. Olfactory functionality was assessed using the Sniffin' Sticks extended test. Non-invasive olfactory mucosa brushing and nasal secretions were processed for nasal samples, while serum samples were obtained through peripheral venous sampling. A panel of autoantibodies, including Immunocirculating Complexes, ANA, ENA, and AECA, was investigated in serum and brush supernatant samples. RESULTS Contrary to expectations, the absence of traditional autoantibodies challenges the proposed autoimmune etiology of Long Covid-associated olfactory dysfunction. However, the presence and potential pathogenic role of AECA suggest viral cytopathic and inflammatory involvement in specific anatomical districts. One hypothesis explores the impact of inflammation and cytokine release induced by the viral infection, altering neuronal signaling and contributing to persistent hyposmia. CONCLUSION This research contributes to our understanding of the complex relationships between autoimmunity, olfactory impairment, and COVID-19. The absence of classical autoantibodies challenges prevailing theories, while the prominence of AECA hints at unique viral-induced pathogenic mechanisms. By unraveling these complexities, this study enhances our comprehension of post-acute sequelae, offering valuable perspectives on immune-mediated responses in the aftermath of the pandemic.
Collapse
Affiliation(s)
- Denise Fiorelli
- Department of Experimental Medicine, University of Tor Vergata, Rome, Italy
| | - Beatrice Francavilla
- Department of Otorhinolaryngology, University of Rome "Tor Vergata", Rome, Italy
| | - Gianluca Velletrani
- Department of Otorhinolaryngology, University of Rome "Tor Vergata", Rome, Italy.
| | - Sara Maurantonio
- Department of Otorhinolaryngology, University of Rome "Tor Vergata", Rome, Italy
| | | | - Sergio Bernardini
- Department of Experimental Medicine, University of Tor Vergata, Rome, Italy
| | - Stefano Di Girolamo
- Department of Otorhinolaryngology, University of Rome "Tor Vergata", Rome, Italy
| | - Marzia Nuccetelli
- Department of Experimental Medicine, University of Tor Vergata, Rome, Italy
| |
Collapse
|
5
|
Justiz-Vaillant AA, Gopaul D, Soodeen S, Arozarena-Fundora R, Barbosa OA, Unakal C, Thompson R, Pandit B, Umakanthan S, Akpaka PE. Neuropsychiatric Systemic Lupus Erythematosus: Molecules Involved in Its Imunopathogenesis, Clinical Features, and Treatment. Molecules 2024; 29:747. [PMID: 38398500 PMCID: PMC10892692 DOI: 10.3390/molecules29040747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 01/14/2024] [Accepted: 01/16/2024] [Indexed: 02/25/2024] Open
Abstract
Systemic lupus erythematosus (SLE) is an idiopathic chronic autoimmune disease that can affect any organ in the body, including the neurological system. Multiple factors, such as environmental (infections), genetic (many HLA alleles including DR2 and DR3, and genes including C4), and immunological influences on self-antigens, such as nuclear antigens, lead to the formation of multiple autoantibodies that cause deleterious damage to bodily tissues and organs. The production of autoantibodies, such as anti-dsDNA, anti-SS(A), anti-SS(B), anti-Smith, and anti-neuronal DNA are characteristic features of this disease. This autoimmune disease results from a failure of the mechanisms responsible for maintaining self-tolerance in T cells, B cells, or both. Immune complexes, circulating antibodies, cytokines, and autoreactive T lymphocytes are responsible for tissue injury in this autoimmune disease. The diagnosis of SLE is a rheumatological challenge despite the availability of clinical criteria. NPSLE was previously referred to as lupus cerebritis or lupus sclerosis. However, these terms are no longer recommended because there is no definitive pathological cause for the neuropsychiatric manifestations of SLE. Currently, the treatment options are primarily based on symptomatic presentations. These include the use of antipsychotics, antidepressants, and anxiolytic medications for the treatment of psychiatric and mood disorders. Antiepileptic drugs to treat seizures, and immunosuppressants (e.g., corticosteroids, azathioprine, and mycophenolate mofetil), are directed against inflammatory responses along with non-pharmacological interventions.
Collapse
Affiliation(s)
- Angel A. Justiz-Vaillant
- Department of Para-Clinical Sciences, University of the West Indies, St. Augustine Campus, St. Augustine 00000, Trinidad and Tobago; (S.S.); (C.U.); (R.T.); (B.P.); (P.E.A.)
| | - Darren Gopaul
- Port of Spain General Hospital, University of the West Indies, St. Augustine Campus, St. Augustine 00000, Trinidad and Tobago;
| | - Sachin Soodeen
- Department of Para-Clinical Sciences, University of the West Indies, St. Augustine Campus, St. Augustine 00000, Trinidad and Tobago; (S.S.); (C.U.); (R.T.); (B.P.); (P.E.A.)
| | - Rodolfo Arozarena-Fundora
- Eric Williams Medical Sciences Complex, North Central Regional Health Authority, Champs Fleurs, San Juan 00000, Trinidad and Tobago; (R.A.-F.); (O.A.B.)
- Department of Clinical and Surgical Sciences, Faculty of Medical Sciences, The University of the West Indies, St. Augustine 00000, Trinidad and Tobago
| | - Odette Arozarena Barbosa
- Eric Williams Medical Sciences Complex, North Central Regional Health Authority, Champs Fleurs, San Juan 00000, Trinidad and Tobago; (R.A.-F.); (O.A.B.)
| | - Chandrashehkar Unakal
- Department of Para-Clinical Sciences, University of the West Indies, St. Augustine Campus, St. Augustine 00000, Trinidad and Tobago; (S.S.); (C.U.); (R.T.); (B.P.); (P.E.A.)
| | - Reinand Thompson
- Department of Para-Clinical Sciences, University of the West Indies, St. Augustine Campus, St. Augustine 00000, Trinidad and Tobago; (S.S.); (C.U.); (R.T.); (B.P.); (P.E.A.)
| | - Bijay Pandit
- Department of Para-Clinical Sciences, University of the West Indies, St. Augustine Campus, St. Augustine 00000, Trinidad and Tobago; (S.S.); (C.U.); (R.T.); (B.P.); (P.E.A.)
| | - Srikanth Umakanthan
- Department of Para-Clinical Sciences, University of the West Indies, St. Augustine Campus, St. Augustine 00000, Trinidad and Tobago; (S.S.); (C.U.); (R.T.); (B.P.); (P.E.A.)
| | - Patrick E. Akpaka
- Department of Para-Clinical Sciences, University of the West Indies, St. Augustine Campus, St. Augustine 00000, Trinidad and Tobago; (S.S.); (C.U.); (R.T.); (B.P.); (P.E.A.)
| |
Collapse
|
6
|
罗 靓, 李 云, 王 红, 相 晓, 赵 静, 孙 峰, 张 晓, 贾 汝, 李 春. [Anti-endothelial cell antibodies in predicting early miscarriage]. BEIJING DA XUE XUE BAO. YI XUE BAN = JOURNAL OF PEKING UNIVERSITY. HEALTH SCIENCES 2023; 55:1039-1044. [PMID: 38101786 PMCID: PMC10723987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Indexed: 12/17/2023]
Abstract
OBJECTIVE To explore the clinical significance of anti-endothelial cell antibodies (AECA) in predicting early miscarriage. METHODS A total of 122 pregnant women with no history of autoimmune diseases who underwent prenatal examination at Peking University People's Hospital from January 2020 to December 2022 were selected, and they were tested for AECA. Based on the history of early miscarriage (gestational age at miscarriage < 12 weeks), the participants were divided into an early miscarriage group and a control group. t-tests, non-parametric Wilcoxon tests, Chi-square tests, and Fisher's exact probability method were used to compare general information and laboratory indicators between the two groups. A multivariate Logistic regression model was used to analyze the factors associated with early miscarriage. The natural miscarriage rates were assessed through follow-up with pregnant women, and Kaplan-Meier survival analysis was employed to compare the natural miscarriage rates between AECA-positive and AECA-negative pregnant women. RESULTS (1) A total of 122 pregnant women were enrolled, comprising 35 cases (28.7%) in the early miscarriage group, with an average age of (32.1±6.1) years, and 87 cases (71.3%) in the control group, with an average age of (30.7±5.1) years. The early miscarriage group had higher gravidity [3 (2, 4) vs. 1 (1, 2), Z=-6.402, P < 0.001] and a higher prevalence of hypertension (11.4% vs.1.1%, P=0.024). The positive rate of AECA in the early miscarriage group (34.3% vs. 8.0%, χ2=13.070, P < 0.001) and the proportion of elevated immunoglobulin G (17.1% vs. 4.6%, P=0.032) were significantly higher than that in the control group. (2) Multivariate logistic regression analysis showed that higher gravidity (OR=4.149, 95%CI: 2.287-7.529, P < 0.001), AECA positivity (OR= 4.288, 95% CI: 1.157-15.893, P=0.029), and elevated immunoglobulin G levels (OR =6.177, 95%CI: 1.156-33.015, P=0.033) were risk factors for early miscarriage. (3) The 122 pregnant women were categorized into two groups: the AECA-positive group (19 cases) and the AECA-negative group (103 cases). Survival analysis demonstrated that at the end of 12 weeks of gestation, the fetal survival rate in the AECA-positive group was significantly lower than that in the AECA-negative group (84.2% vs. 96.1%, P= 0.035). CONCLUSION Higher gravidity, AECA positivity, and elevated immunoglobulin G levels are significant risk factors for early miscarriage. The results demonstrate that AECA is a novel predicting test in early miscarriage.
Collapse
Affiliation(s)
- 靓 罗
- 北京大学人民医院风湿免疫科, 北京 100044Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing 100044, China
- 重庆市渝北区人民医院中医科, 重庆 401120Department of Chinese Medicine, the People's Hospital of Yubei District of Chongqing City, Chongqing 401120, China
| | - 云 李
- 北京大学人民医院风湿免疫科, 北京 100044Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing 100044, China
| | - 红彦 王
- 北京大学人民医院风湿免疫科, 北京 100044Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing 100044, China
| | - 晓红 相
- 北京大学人民医院风湿免疫科, 北京 100044Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing 100044, China
| | - 静 赵
- 北京大学人民医院风湿免疫科, 北京 100044Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing 100044, China
| | - 峰 孙
- 北京大学人民医院风湿免疫科, 北京 100044Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing 100044, China
| | - 晓盈 张
- 北京大学人民医院风湿免疫科, 北京 100044Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing 100044, China
| | - 汝琳 贾
- 北京大学人民医院风湿免疫科, 北京 100044Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing 100044, China
| | - 春 李
- 北京大学人民医院风湿免疫科, 北京 100044Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing 100044, China
| |
Collapse
|
7
|
Serum of Post-COVID-19 Syndrome Patients with or without ME/CFS Differentially Affects Endothelial Cell Function In Vitro. Cells 2022; 11:cells11152376. [PMID: 35954219 PMCID: PMC9367589 DOI: 10.3390/cells11152376] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 07/28/2022] [Accepted: 07/29/2022] [Indexed: 12/24/2022] Open
Abstract
A proportion of COVID-19 reconvalescent patients develop post-COVID-19 syndrome (PCS) including a subgroup fulfilling diagnostic criteria of Myalgic encephalomyelitis/Chronic Fatigue Syndrome (PCS/CFS). Recently, endothelial dysfunction (ED) has been demonstrated in these patients, but the mechanisms remain elusive. Therefore, we investigated the effects of patients’ sera on endothelia cells (ECs) in vitro. PCS (n = 17), PCS/CFS (n = 13), and healthy controls (HC, n = 14) were screened for serum anti-endothelial cell autoantibodies (AECAs) and dysregulated cytokines. Serum-treated ECs were analysed for the induction of activation markers and the release of small molecules by flow cytometry. Moreover, the angiogenic potential of sera was measured in a tube formation assay. While only marginal differences between patient groups were observed for serum cytokines, AECA binding to ECs was significantly increased in PCS/CFS patients. Surprisingly, PCS and PCS/CFS sera reduced surface levels of several EC activation markers. PCS sera enhanced the release of molecules associated with vascular remodelling and significantly promoted angiogenesis in vitro compared to the PCS/CFS and HC groups. Additionally, sera from both patient cohorts induced the release of molecules involved in inhibition of nitric oxide-mediated endothelial relaxation. Overall, PCS and PCS/CFS patients′ sera differed in their AECA content and their functional effects on ECs, i.e., secretion profiles and angiogenic potential. We hypothesise a pro-angiogenic effect of PCS sera as a compensatory mechanism to ED which is absent in PCS/CFS patients.
Collapse
|
8
|
The impact of anti-endothelial cell antibodies (AECAs) on the development of blood vessel damage in patients with systemic lupus erythematosus: the preliminary study. Rheumatol Int 2022; 42:791-801. [DOI: 10.1007/s00296-022-05104-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 02/10/2022] [Indexed: 12/13/2022]
|
9
|
Michael Henry B, Benoit SW, Vikse J, Favaloro E, Benoit JL, Lippi G. Anti-Endothelial Cell Antibodies are not frequently elevated in hospitalized patients with COVID-19. ACTA BIO-MEDICA : ATENEI PARMENSIS 2022; 93:e2022043. [PMID: 35546028 PMCID: PMC9171859 DOI: 10.23750/abm.v93i2.11196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 01/12/2021] [Indexed: 11/14/2022]
Affiliation(s)
- Brandon Michael Henry
- Cardiac Intensive Care Unit, The Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA, Division of Nephrology and Hypertension, Cincinnati Children’s Hospital Medical Center, OH, USA
| | - Stefanie W. Benoit
- Division of Nephrology and Hypertension, Cincinnati Children’s Hospital Medical Center, OH, USA, Department of Pediatrics, University of Cincinnati, College of Medicine, OH, USA
| | - Jens Vikse
- Clinical Immunology Unit, Stavanger University Hospital, Stavanger, Norway
| | - Emmanuel Favaloro
- Haematology, Sydney Centres for Thrombosis and Haemostasis, Institute of Clinical Pathology and Medical Research (ICPMR), NSW Health Pathology, Westmead Hospital, Westmead NSW, Australia
| | - Justin L. Benoit
- Department of Emergency Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Giuseppe Lippi
- Section of Clinical Biochemistry, Department of Neuroscience, Biomedicine and Movement, University of Verona, Verona, Italy
| |
Collapse
|
10
|
Henry BM, Benoit SW, Vikse J, Favaloro E, Benoit JL, Lippi G. Anti-Endothelial Cell Antibodies are not frequently elevated in hospitalized patients with COVID-19. ACTA BIO-MEDICA : ATENEI PARMENSIS 2022; 93:e2022026. [PMID: 35315426 PMCID: PMC8972892 DOI: 10.23750/abm.v93i1.10799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 05/31/2021] [Indexed: 11/15/2022]
Abstract
COVID-19 is now established to be associated with a thrombotic phenomenon, now called COVID-19 associated coagulopathy (CAC). Anti-Endothelial Cell Antibodies (AECA) are a heterogenous group of autoantibodies targeting various endothelial cell antigens or antigens adhering to endothelial cells, They are commonly observed in a variety of auto-immune and rheumatologic conditions, and were observed in patients with the severe acute respiratory syndrome (SARS) in 2005. We aimed to assess AECA status in patients with COVID-19 and their potential contributing role to endothelial injury and CAC. AECA identification was a relatively infrequent finding in COVID-19 patients on admission, and their presence, albeit in only 2/33 patients, was not associated with disease severity. However, as the autoantibodies were only measured at admission, we cannot exclude the possibility of pathogenic AECA developing later in the course of diseaseFurther studies using additional methods are needed to evaluate the presence and potential pathogenic role of AECA in later stages of COVID-19.
Collapse
Affiliation(s)
- Brandon Michael Henry
- Disease Intervention & Prevention and Population Health Programs, Texas Biomedical Research Institute, San Antonio, TX, USA, Division of Nephrology and Hypertension, Cincinnati Children’s Hospital Medical Center, OH, USA
| | - Stefanie W. Benoit
- Division of Nephrology and Hypertension, Cincinnati Children’s Hospital Medical Center, OH, USA, Department of Pediatrics, University of Cincinnati, College of Medicine, OH, USA
| | - Jens Vikse
- Clinical Immunology Unit, Stavanger University Hospital, Stavanger, Norway
| | - Emmanuel Favaloro
- Haematology, Sydney Centres for Thrombosis and Haemostasis, Institute of Clinical Pathology and Medical Research (ICPMR), NSW Health Pathology, Westmead Hospital, Westmead NSW, Australia
| | - Justin L. Benoit
- Department of Emergency Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Giuseppe Lippi
- Section of Clinical Biochemistry, Department of Neuroscience, Biomedicine and Movement, University of Verona, Verona, Italy
| |
Collapse
|
11
|
Experimental evidence for alpha enolase as one potential autoantigen in the pathogenesis of both autoimmune thyroiditis and its related encephalopathy. Int Immunopharmacol 2020; 85:106563. [PMID: 32442899 DOI: 10.1016/j.intimp.2020.106563] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 04/26/2020] [Accepted: 05/03/2020] [Indexed: 11/24/2022]
Abstract
Alpha-enolase (ENO1) is a ubiquitous protein. Patients with autoimmune thyroiditis-associated encephalopathy have high serum ENO1Ab titers. We aimed to explore whether ENO1Ab was the pathogenic antibody in the thyroid and brain. The serum ENO1Ab titers were significantly increased in the mice immunized with Thyroglobulin (Tg). And in the mice immunized with ENO1, serum levels of both TgAb and thyroid-stimulating hormone (TSH) were significantly increased. Obvious CD16+ cell infiltration, IgG deposit and cleaved caspase-3 were observed in the thyroid of ENO1-immunized mice. Spatial learning and memory abilities and synaptic functions were impaired in ENO1-immunized mice. Furthermore, the expression levels of Iba-1, GFAP, interlukin-6, CDK5, and phosphorylated tau were increased, and endothelial tight junction proteins were decreased in the brain of ENO1-immunized mice. These results suggest that ENO1Ab can cause thyrocyte damage via ADCC effect and impair cerebral function by disrupting the blood-brain barrier.
Collapse
|
12
|
Sánchez-Zapardiel E, Mancebo E, Díaz-Ordoñez M, de Jorge-Huerta L, Ruiz-Martínez L, Serrano A, Castro-Panete MJ, Utrero-Rico A, de Andrés A, Morales JM, Domínguez-Rodríguez S, Paz-Artal E. Isolated De Novo Antiendothelial Cell Antibodies and Kidney Transplant Rejection. Am J Kidney Dis 2016; 68:933-943. [PMID: 27599627 DOI: 10.1053/j.ajkd.2016.07.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 07/06/2016] [Indexed: 11/11/2022]
Abstract
BACKGROUND Studies analyzing the role of antiendothelial cell antibodies (AECAs) in large series of kidney transplant recipients are scarce, and HLA, MHC (major histocompatibility complex) class I-related chain A (MICA), and angiotensin II type 1 receptor have not been formally excluded as targets. STUDY DESIGN Retrospective study of a cohort of kidney transplant recipients. SETTING & PARTICIPANTS 324 kidney transplant recipients who were negative for anti-HLA, anti-MICA, and anti-angiotensin II type 1 receptor antibodies were tested for AECAs in pre- and posttransplantation serum samples. PREDICTORS AECA-positive (preformed [pre+/post+] vs de novo [pre-/post+]) versus AECA-negative (pre-/post-) before or after transplantation. OUTCOMES Patient mortality, transplant loss, and acute rejection events. RESULTS 66 (20%) patients were AECA positive (39 [12%] preformed, 27 [8%] de novo) and 258 (80%) were AECA negative. During a follow-up of 10 years, 7 (18%) AECA pre+/post+ patients had rejections compared with 14 (52%) AECA pre-/post+ and 57 (22%) AECA pre-/post- recipients (OR, 3.80; P=0.001). AECA pre-/post+ status emerged as an independent risk factor for transplant rejection compared to the AECA pre-/post- group (OR, 5.17; P<0.001). However, AECA pre+/post+ and AECA pre-/post+ patients did not show higher risk for either patient death (ORs of 1.49 [P=0.7] and 1.06 [P=0.9], respectively) or transplant loss (ORs of 1.22 and 0.86, respectively; P for both = 0.8) compared to the AECA pre-/post- population. LIMITATIONS Retrospective study. Posttransplantation sera were collected before or after rejection, entailing a nearly cross-sectional relationship between the exposure and outcome. Lack of identification of precise antigens for AECAs. CONCLUSIONS De novo AECAs may be associated with rejection. These antibodies might serve as biomarkers of endothelium damage in kidney transplant recipients.
Collapse
Affiliation(s)
| | - Esther Mancebo
- Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain; Research Institute I+12, Hospital Universitario 12 de Octubre, Madrid, Spain
| | | | | | | | - Antonio Serrano
- Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain; Research Institute I+12, Hospital Universitario 12 de Octubre, Madrid, Spain; Section of Immunology, Universidad San Pablo CEU, Madrid, Spain
| | - María J Castro-Panete
- Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain; Research Institute I+12, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Alberto Utrero-Rico
- Research Institute I+12, Hospital Universitario 12 de Octubre, Madrid, Spain; School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Amado de Andrés
- Department of Nephrology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - José M Morales
- Research Institute I+12, Hospital Universitario 12 de Octubre, Madrid, Spain; Department of Nephrology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | | | - Estela Paz-Artal
- Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain; Research Institute I+12, Hospital Universitario 12 de Octubre, Madrid, Spain; School of Medicine, Universidad Complutense de Madrid, Madrid, Spain; Section of Immunology, Universidad San Pablo CEU, Madrid, Spain
| |
Collapse
|
13
|
Kwok SK, Seo SH, Ju JH, Park KS, Yoon CH, Kim WU, Min JK, Park SH, Cho CS, Kim HY. Lupus enteritis: clinical characteristics, risk factor for relapse and association with anti-endothelial cell antibody. Lupus 2016; 16:803-9. [PMID: 17895303 DOI: 10.1177/0961203307082383] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The study was undertaken to evaluate clinical and laboratory characteristics of patients with lupus enteritis and to investigate its association with anti-endothelial cell antibodies (AECAs). Systemic lupus erythematosus (SLE) patients who were admitted to Kangnam St. Mary's Hospital with complaints of acute abdominal pain from January 1990 to July 2006 were reviewed retrospectively. The clinical features, laboratory data and prognosis of these patients were analyzed. Among the 706 SLE patients admitted during the study period, 87 were found to admit for acute abdominal pain. Among them, 41 patients were identified with lupus enteritis. The SLE disease activity index score at admission and the mean prednisolone dose administered during the last three months prior to admission were significantly higher in patients with lupus enteritis than those with other causes ( P < 0.001, P = 0.036). Serum anti-endothelial cell antibody (AECA-IgG) titer was also significantly higher in patients with lupus enteritis than those with other manifestations or healthy controls ( P = 0.040, P < 0.001). Four out of 13 recurrent patients had pre-existing anti-phospholipid syndrome (APS), whereas only one out of 28 non-recurrent patients had pre-existing APS ( P = 0.028). Most of the patients with lupus enteritis showed good response to high-dose intravenous steroids and there was no death directly associated with lupus enteritis. Lupus (2007) 16, 803—809.
Collapse
Affiliation(s)
- S-K Kwok
- Division of Rheumatology, Department of Internal Medicine, School of Medicine, The Catholic University of Korea, Seoul, South Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
George J, Matucci-Cerinic M, Bar I, Shimoni S. Circulating autoantibodies to endothelial progenitor cells: binding characteristics and association with risk factors for atherosclerosis. PLoS One 2014; 9:e97836. [PMID: 24945945 PMCID: PMC4063726 DOI: 10.1371/journal.pone.0097836] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 04/24/2014] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE Endothelial progenitor cells (EPC) are committed to transform into EC promoting vasculogenic ischemic repair. Anti-endothelial cells (AECA) have been described in various disorders with an associated vascular damage. Herein, we explored a novel circulating population of IgG reactive with EPC, in patients with differential risk profile for atherosclerotic vascular disease. APPROACH AND RESULTS A novel cyto-ELISA system was established where the coated cells were late outgrowth EPC. Levels of anti-EPC antibodies were determined in 100 subjects and differential risk score for atherosclerosis, as well as to circulating EPC levels and the inflammatory markers IL-6 and C-reactive protein. To study endothelial cell (EC) activating properties, sera were tested for their ability to induce VCAM-1 expression in a cell ELISA system. Detectable levels of anti-EPC antibodies, that correlated with age, Framingham risk score and CRP concentrations but did not associate with levels of LDL, HDL, hypertension or diabetes, were detected. Anti-EPC antibodies were distinct from EC binding antibodies as shown by competitive inhibition studies, and have been positively correlated with the extent of EC activation manifested by in vitro VCAM-1 expression. CONCLUSION This is the first study showing a newly defined subgroup of self-antibodies binding EPC and associating positively with the Framingham risk score. Further studies are required to characterize and test this interesting subset of EPC binding autoantibodies and their potential significance.
Collapse
Affiliation(s)
- Jacob George
- Heart Center, Kaplan Medical Center, Rehovot, Affiliated to the Hebrew University, Jerusalem, Israel
| | - Marco Matucci-Cerinic
- Department of Experimental and Clinical Medicine, Division of Rheumatology AOUC, University of Florence, Florence, Italy
| | - Iris Bar
- Heart Center, Kaplan Medical Center, Rehovot, Affiliated to the Hebrew University, Jerusalem, Israel
| | - Sara Shimoni
- Heart Center, Kaplan Medical Center, Rehovot, Affiliated to the Hebrew University, Jerusalem, Israel
| |
Collapse
|
15
|
Autoantibodies and depression. Neurosci Biobehav Rev 2014; 40:62-79. [DOI: 10.1016/j.neubiorev.2014.01.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 12/10/2013] [Accepted: 01/05/2014] [Indexed: 01/05/2023]
|
16
|
Delunardo F, Scalzi V, Capozzi A, Camerini S, Misasi R, Pierdominici M, Pendolino M, Crescenzi M, Sorice M, Valesini G, Ortona E, Alessandri C. Streptococcal-vimentin cross-reactive antibodies induce microvascular cardiac endothelial proinflammatory phenotype in rheumatic heart disease. Clin Exp Immunol 2013; 173:419-29. [PMID: 23663103 DOI: 10.1111/cei.12135] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/07/2013] [Indexed: 01/08/2023] Open
Abstract
Rheumatic heart disease (RHD) is characterized by the presence of anti-streptococcal group A antibodies and anti-endothelial cell antibodies (AECA). Molecular mimicry between streptococcal antigens and self proteins is a hallmark of the pathogenesis of rheumatic fever. We aimed to identify, in RHD patients, autoantibodies specific to endothelial autoantigens cross-reactive with streptococcal proteins and to evaluate their role in inducing endothelial damage. We used an immunoproteomic approach with endothelial cell-surface membrane proteins in order to identify autoantigens recognized by AECA of 140 RHD patients. Cross-reactivity of purified antibodies with streptococcal proteins was analysed. Homologous peptides recognized by serum cross-reactive antibodies were found through comparing the amino acid sequence of streptococcal antigens with human antigens. To investigate interleukin (IL)-1R-associated kinase (IRAK1) and nuclear factor-κB (NF-κB) activation, we performed a Western blot analysis of whole extracts proteins from unstimulated or stimulated human microvascular cardiac endothelial cells (HMVEC-C). Adhesion molecule expression and release of proinflammatory cytokines and growth factors were studied by multiplex bead based immunoassay kits. We observed anti-vimentin antibodies in sera from 49% RHD AECA-positive patients. Cross-reactivity of purified anti-vimentin antibodies with heat shock protein (HSP)70 and streptopain streptococcal proteins was shown. Comparing the amino acid sequence of streptococcal HSP70 and streptopain with human vimentin, we found two homologous peptides recognized by serum cross-reactive antibodies. These antibodies were able to stimulate HMVEC-C inducing IRAK and NF-κB activation, adhesion molecule expression and release of proinflammatory cytokines and growth factors. In conclusion, streptococcal-vimentin cross-reactive antibodies were able to activate microvascular cardiac endothelium by amplifying the inflammatory response in RHD.
Collapse
Affiliation(s)
- F Delunardo
- Dipartimento di Biologia Cellulare e Neuroscienze, Istituto Superiore di Sanità, Roma, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Wolf SI, Howat S, Abraham DJ, Pearson JD, Lawson C. Agonistic anti-ICAM-1 antibodies in scleroderma: activation of endothelial pro-inflammatory cascades. Vascul Pharmacol 2013; 59:19-26. [PMID: 23685129 PMCID: PMC3731553 DOI: 10.1016/j.vph.2013.05.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Revised: 04/23/2013] [Accepted: 05/08/2013] [Indexed: 11/30/2022]
Abstract
Background Scleroderma (SSc) is a complex autoimmune disorder that can be characterised by the presence 2of circulating autoantibodies to nuclear, cytoplasmic and cell surface antigens. In particular antibodies directed against endothelial cell antigens (anti-endothelial cell antibodies; AECA) have been detected. ICAM-1 is an adhesion molecule expressed on the surface of human endothelial cells. We have previously shown that cross-linking ICAM-1 with monoclonal antibodies leads to pro-inflammatory activation of human endothelial and vascular smooth muscle cells and that cardiac transplant recipients with transplant associated vasculopathy make antibodies directed against ICAM-1. Objectives To determine whether SSc patients make antibodies directed against ICAM-1 and whether these antibodies induce pro-inflammatory activation of human endothelial cells in vitro. Methods Using recombinant ICAM-1 as capture antigen, an ELISA was developed to measure ICAM-1 antibodies in sera from SSc patients. Antibodies were purified using ICAM-1 micro-affinity columns. HUVEC were incubated with purified anti-ICAM-1 antibodies and generation of reactive oxygen species, and expression of VCAM-1 was measured. Results Significantly elevated levels of anti-ICAM-1 antibodies were detected in patients with diffuse (dSSc; 10/31 32%) or limited (lSSc; 14/36 39%) scleroderma. Cross-linking of HUVEC with purified anti-ICAM-1 antibodies caused a significant increase in ROS production (2.471 ± 0.408 fold increase above untreated after 150 min p < 0.001), and significant increase in VCAM-1 expression (10.6 ± 1.77% vs 4.12 ± 1.33%, p < 0.01). Conclusion AECA from SSc patients target specific endothelial antigens including ICAM-1, and cause pro-inflammatory activation of human endothelial cells, suggesting that they are not only a marker of disease but that they contribute to its progression.
Collapse
Affiliation(s)
- Sabine I Wolf
- Department of Comparative Biomedical Sciences, The Royal Veterinary College, London, UK
| | | | | | | | | |
Collapse
|
18
|
Hernandez NM, Casselbrant A, Joshi M, Johansson BR, Sumitran-Holgersson S. Antibodies to kidney endothelial cells contribute to a "leaky" glomerular barrier in patients with chronic kidney diseases. Am J Physiol Renal Physiol 2011; 302:F884-94. [PMID: 22189942 DOI: 10.1152/ajprenal.00250.2011] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Anti-endothelial cell antibodies (AECA) have been reported to cause endothelial dysfunction, but their clinical importance for tissue-specific endothelial cells is not clear. We hypothesized that AECA reactive with human kidney endothelial cells (HKEC) may cause renal endothelial dysfunction in patients with chronic kidney diseases. We report that a higher fraction (56%) of end-stage renal disease (ESRD) patients than healthy controls (5%) have AECA reactive against kidney endothelial cells (P <0.001). The presence of antibodies was associated with female gender (P < 0.001), systolic hypertension (P < 0.01), and elevated TNF-α (P < 0.05). These antibodies markedly decrease expression of both adherens and tight junction proteins VE-cadherin, claudin-1, and zonula occludens-1 and provoked a rapid increase in cytosolic free Ca(2+) and rearrangement of actin filaments in HKEC compared with controls. This was followed by an enhancement in protein flux and phosphorylation of VE-cadherin, events associated with augmented endothelial cell permeability. Additionally, kidney biopsies from ESRD patients with AECA but not controls demonstrated a marked decrease in adherens and tight junctions in glomerular endothelium, confirming our in vitro data. In summary, our data demonstrate a causal link between AECA and their capacity to induce alterations in glomerular vascular permeability.
Collapse
Affiliation(s)
- Nidia Maritza Hernandez
- Dept. of Transplantation Surgery, Laboratory for Transplantation and Regenerative Medicine, Sahlgrenska Academy, University of Gothenburg, Sahlgrenska Science Park, Medicinaregatan 8A, Gothenburg, Sweden
| | | | | | | | | |
Collapse
|
19
|
Alard JE, Hillion S, Guillevin L, Saraux A, Pers JO, Youinou P, Jamin C. Autoantibodies to endothelial cell surface ATP synthase, the endogenous receptor for hsp60, might play a pathogenic role in vasculatides. PLoS One 2011; 6:e14654. [PMID: 21326874 PMCID: PMC3034716 DOI: 10.1371/journal.pone.0014654] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2010] [Accepted: 01/13/2011] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Heat shock protein (hsp) 60 that provides "danger signal" binds to the surface of resting endothelial cells (EC) but its receptor has not yet been characterized. In mitochondria, hsp60 specifically associates with adenosine triphosphate (ATP) synthase. We therefore examined the possible interaction between hsp60 and ATP synthase on EC surface. METHODOLOGY/PRINCIPAL FINDINGS Using Far Western blot approach, co-immunoprecipitation studies and surface plasmon resonance analyses, we demonstrated that hsp60 binds to the β-subunit of ATP synthase. As a cell surface-expressed molecule, ATP synthase is potentially targeted by anti-EC-antibodies (AECAs) found in the sera of patients suffering vasculitides. Based on enzyme-linked immunosorbent assay and Western blotting techniques with F1-ATP synthase as substrate, we established the presence of anti-ATP synthase antibodies at higher frequency in patients with primary vasculitides (group I) compared with secondary vasculitides (group II). Anti-ATP synthase reactivity from group I patients was restricted to the β-subunit of ATP synthase, whereas those from group II was directed to the α-, β- and γ-subunits. Cell surface ATP synthase regulates intracellular pH (pHi). In low extracellular pH medium, we detected abnormal decreased of EC pHi in the presence of anti-ATP synthase antibodies, irrespective of their fine reactivities. Interestingly, soluble hsp60 abrogated the anti-ATP synthase-induced pHi down-regulation. CONCLUSIONS/SIGNIFICANCE Our results indicate that ATP synthase is targeted by AECAs on the surface of EC that induce intracellular acidification. Such pathogenic effect in vasculitides can be modulated by hsp60 binding on ATP synthase which preserves ATP synthase activity.
Collapse
Affiliation(s)
- Jean-Eric Alard
- EA2216 “Immunology and Pathology” and IFR 148 ScInBioS, Université de Brest and Université Européenne de Bretagne, Brest, France
| | - Sophie Hillion
- EA2216 “Immunology and Pathology” and IFR 148 ScInBioS, Université de Brest and Université Européenne de Bretagne, Brest, France
| | - Loïc Guillevin
- Department of Internal Medicine, Hôpital Cochin, Paris, France
| | - Alain Saraux
- EA2216 “Immunology and Pathology” and IFR 148 ScInBioS, Université de Brest and Université Européenne de Bretagne, Brest, France
- Centre Hospitalier Universitaire, Brest, France
| | - Jacques-Olivier Pers
- EA2216 “Immunology and Pathology” and IFR 148 ScInBioS, Université de Brest and Université Européenne de Bretagne, Brest, France
- Centre Hospitalier Universitaire, Brest, France
| | - Pierre Youinou
- EA2216 “Immunology and Pathology” and IFR 148 ScInBioS, Université de Brest and Université Européenne de Bretagne, Brest, France
- Centre Hospitalier Universitaire, Brest, France
| | - Christophe Jamin
- EA2216 “Immunology and Pathology” and IFR 148 ScInBioS, Université de Brest and Université Européenne de Bretagne, Brest, France
- Centre Hospitalier Universitaire, Brest, France
| |
Collapse
|
20
|
Scalzi V, Hadi HA, Alessandri C, Croia C, Conti V, Agati L, Angelici A, Riccieri V, Meschini C, Al-Motarreb A, Al-Ansi A, Valesini G. Anti-endothelial cell antibodies in rheumatic heart disease. Clin Exp Immunol 2011; 161:570-5. [PMID: 20646009 DOI: 10.1111/j.1365-2249.2010.04207.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
To evaluate the anti-endothelial cell antibodies (AECA), anti-cardiolipin antibodies (aCL) and serum mannose-binding lectin (MBL) profiles of a large cohort of Yemeni patients with rheumatic heart disease (RHD) and to correlate these findings with clinical features of the disease. Patients (n = 140) were recruited from Al-Thawra Hospital in Sana'a, Yemen. All had RHD diagnosed according to modified Jones' criteria. We also studied 140 sex- and age-matched healthy blood donors from the same area. Echocardiography was performed according to the recommendations of the American Society of Echocardiography. Solid phase enzyme-linked immunosorbent assays (ELISAs) were used to measure AECA and aCL titres and serum MBL levels. Forty per cent of the patients were AECA-positive, but only 7·8% were positive for aCL antibodies. Serum MBL levels were significantly lower in the RHD group (median 4221 ng/ml versus 5166 ng/ml in healthy controls). AECA titres were correlated positively with patient age, duration of RHD and the severity of aortic stenosis, as determined by echocardiographic findings. In several autoimmune rheumatic diseases, such as systemic lupus erythematosus, vasculitis and scleroderma, AECA have been shown to play pathogenic roles by producing proinflammatory and procoagulant effects (increased expression of adhesion molecules and tissue factors, increased cytokine release) in endothelial cells. In RHD, these autoantibodies might represent a pathological link between activation of the valvular endothelium and valvular damage.
Collapse
Affiliation(s)
- V Scalzi
- Dipartimento di Clinica e Terapia Medica, Sapienza Università di Roma, Roma, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Ju JH, Min JK, Jung CK, Oh SN, Kwok SK, Kang KY, Park KS, Ko HJ, Yoon CH, Park SH, Cho CS, Kim HY. Lupus mesenteric vasculitis can cause acute abdominal pain in patients with SLE. Nat Rev Rheumatol 2009; 5:273-81. [PMID: 19412194 DOI: 10.1038/nrrheum.2009.53] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Lupus mesenteric vasculitis (LMV) is a unique clinical entity found in patients who present with gastrointestinal manifestations of systemic lupus erythematosus, and is the main cause of acute abdominal pain in these patients. LMV usually presents as acute abdominal pain with sudden onset, severe intensity and diffuse localization. Other causes of abdominal pain, such as acute gastroenteritis, peptic ulcers, acute pancreatitis, peritonitis, and other reasons for abdominal surgery should be ruled out. Prompt and accurate diagnosis of LMV is critical to ensure implementation of appropriate immunosuppressive therapy and avoidance of unnecessary surgical intervention. The pathology of LMV comprises immune-complex deposition and complement activation, with subsequent submucosal edema, leukocytoclastic vasculitis and thrombus formation; most of these changes are confined to small mesenteric vessels. Abdominal CT is the most useful tool for diagnosing LMV, which is characterized by the presence of target signs, comb signs, and other associated findings. The presence of autoantibodies against phospholipids and endothelial cells might provide information about the likelihood of recurrence of LMV. Immediate, high-dose, intravenous steroid therapy can lead to a favorable outcome and prevent serious complications such as bowel ischemia, necrosis and perforation.
Collapse
Affiliation(s)
- Ji Hyeon Ju
- Rheumatology Division, College of Medicine, Catholic University of Korea, Seocho-Gu, Seoul, South Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Wells R, Guth A, Lappin M, Dow S. Anti-Endothelial Cell Antibodies in Dogs with Immune-Mediated Hemolytic Anemia and Other Diseases Associated with High Risk of Thromboembolism. J Vet Intern Med 2009; 23:295-300. [DOI: 10.1111/j.1939-1676.2008.0251.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
|
23
|
Abstract
The aetiology of primary sclerosing cholangitis (PSC) is not known and controversy exists as to whether PSC should be denominated an autoimmune disease. A large number of autoantibodies have been detected in PSC patients, but the specificity of these antibodies is generally low, and the frequencies vary largely between different studies. The presence of autoantibodies in PSC may be the result of a nonspecific dysregulation of the immune system, but the literature in PSC points to the possible presence of specific antibody targets in the biliary epithelium and in neutrophil granulocytes. The present review aims to give an overview of the studies of autoantibodies in PSC, with a particular emphasis on the prevalence, clinical relevance and possible pathogenetic importance of each individual marker.
Collapse
|
24
|
Antiendothelial cell antibody levels in patients with masked hypertension. Int J Cardiol 2008; 130:405-8. [PMID: 18234377 DOI: 10.1016/j.ijcard.2007.08.122] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2007] [Accepted: 08/11/2007] [Indexed: 11/22/2022]
Abstract
UNLABELLED Recent evidence demonstrate that masked hypertension (MH) is a significant predictor of cardiovascular disease, while, elevated levels of circulating antibodies against endothelial cell surface antigens (antiendothelial cell antibodies - AECA) seem to play an important role at the early stages of atherosclerosis process and of borderline hypertension as well. Aim of this study was to investigate the presence of AECA in patients (pts) with MH and to compare the AECA title among pts with MH and healthy normotensives (HN), matched for age, sex and body mass index. METHODS One hundred-thirty (60 M, 70 F) healthy subjects mean age 45+/-12 yrs who had clinic blood pressure <140/90 mm Hg were studied. The whole study population underwent 24 hour ambulatory blood pressure monitoring (ABPM). According to the ABPM recordings, 24 individuals (8 M, 16 F) had MH (daytime systolic blood pressure >/=135 mm Hg or daytime diastolic blood pressure >/=85 mm Hg - group A) and the remainder 106 subjects (52 M, 54 F) had normal ABPM recordings, group B. IgG and IgM AECA levels were determined by ELISA method. AECA levels were expressed as mean value+/-SD. None of the study population had a history of connective tissue disease or any metabolic disorder. RESULTS Significantly increased titles of AECA class IgG were found in 8/24 pts of group A (30%) vs. 5/106 (4.6%) of group B (p<0,001). Significantly increased titles of AECA class IgM were also found in 6/24 pts of group A (25%) vs. 3/80 (3.8%) of group B (p<0,001). CONCLUSIONS Our results suggest that patients with MH have significantly higher AECA levels of both classes (IgG, IgM) compared to healthy normotensives. These findings may indicate a possible explanation of the increased cardiovascular risk in MH. The possibility that high AECA levels may be a driving mechanism for the development of MH needs further investigation.
Collapse
|