1
|
Xiang P, Latif R, Morshed S, Davies TF. Hypothyroidism Induced by a TSH Receptor Peptide-Implications for Thyroid Autoimmunity. Thyroid 2024. [PMID: 39435685 DOI: 10.1089/thy.2024.0089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2024]
Abstract
Background: The "neutral" thyrotropin receptor autoantibodies (N-TSHR-Ab) directed at the TSHR ectodomain's hinge region have been shown to induce thyroid cell damage in vitro. During these earlier studies, we developed a mouse monoclonal antibody (MC1) specific for a peptide (amino acid 322-340) in the region (MC1-Mab) which was able to induce thyroid cell stress and apoptosis when administered in vivo. Methods: In order to examine the effect of in vivo generated N-TSHR-Abs, rather than an acutely administered monoclonal antibody, we immunized Balb/c mice with the hinge region peptide over 18 weeks. Serum TSHR antibodies, specific TSHR hinge region antibodies, serum thyroglobulin (TG) and anti-TG as well as thyroxine and thyrotropin (TSH) levels were examined to evaluate the response to the immunization. Histological examination of the thyroid glands and flow cytometry of spleen T cells, B cells and macrophages were also performed to explore the underlying mechanisms. Results: We found that TSHR-peptide immunized mice developed N-TSHR-Abs against the peptide which resulted in thyroid damage shown by thyroid follicular destruction with follicular cell apoptosis, M1 macrophage infiltration, thyroglobulin release, and induction of thyroglobulin antibodies. This resulted in hypothyroidism with increased TSH levels. Conclusion: This study demonstrated that endogenous neutral antibodies to the TSHR could induce thyroid cell damage from apoptosis and M1 macrophage infiltration and resulted in hypothyroidism.
Collapse
Affiliation(s)
- Pingping Xiang
- Thyroid Research Unit, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Rauf Latif
- Thyroid Research Unit, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- James J. Peters VA Medical Center, New York, New York, USA
| | - Syed Morshed
- Thyroid Research Unit, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- James J. Peters VA Medical Center, New York, New York, USA
| | - Terry F Davies
- Thyroid Research Unit, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- James J. Peters VA Medical Center, New York, New York, USA
| |
Collapse
|
2
|
Richter P, Badescu MC, Rezus C, Ouatu A, Dima N, Popescu D, Burlui AM, Bratoiu I, Mihai IR, Rezus E. Antiphospholipid Antibodies as Key Players in Systemic Lupus Erythematosus: The Relationship with Cytokines and Immune Dysregulation. Int J Mol Sci 2024; 25:11281. [PMID: 39457063 PMCID: PMC11509045 DOI: 10.3390/ijms252011281] [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: 09/17/2024] [Revised: 10/10/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease characterized by an overproduction of cytokines, such as interleukins and interferons, contributing to systemic inflammation and tissue damage. Antiphospholipid syndrome is a thrombo-inflammatory autoimmune disease affecting a third of SLE patients. We performed an in-depth analysis of the available literature, and we highlighted the complex interplay between immunity, inflammation, and thrombosis, the three major pathogenic pathways that are trapped in a mutually reinforcing destructive loop.
Collapse
Affiliation(s)
- Patricia Richter
- Department of Rheumatology and Physiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (P.R.); (A.M.B.); (I.B.); (I.R.M.); (E.R.)
| | - Minerva Codruta Badescu
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (A.O.); , (D.P.)
- IIIrd Internal Medicine Clinic, “St. Spiridon” County Emergency Clinical Hospital, 1 Independence Boulevard, 700111 Iasi, Romania
| | - Ciprian Rezus
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (A.O.); , (D.P.)
- IIIrd Internal Medicine Clinic, “St. Spiridon” County Emergency Clinical Hospital, 1 Independence Boulevard, 700111 Iasi, Romania
| | - Anca Ouatu
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (A.O.); , (D.P.)
- IIIrd Internal Medicine Clinic, “St. Spiridon” County Emergency Clinical Hospital, 1 Independence Boulevard, 700111 Iasi, Romania
| | - Nicoleta Dima
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (A.O.); , (D.P.)
- IIIrd Internal Medicine Clinic, “St. Spiridon” County Emergency Clinical Hospital, 1 Independence Boulevard, 700111 Iasi, Romania
| | - Diana Popescu
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (A.O.); , (D.P.)
| | - Alexandra Maria Burlui
- Department of Rheumatology and Physiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (P.R.); (A.M.B.); (I.B.); (I.R.M.); (E.R.)
- I Rheumatology Clinic, Clinical Rehabilitation Hospital, 14 Pantelimon Halipa Street, 700661 Iasi, Romania
| | - Ioana Bratoiu
- Department of Rheumatology and Physiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (P.R.); (A.M.B.); (I.B.); (I.R.M.); (E.R.)
- I Rheumatology Clinic, Clinical Rehabilitation Hospital, 14 Pantelimon Halipa Street, 700661 Iasi, Romania
| | - Ioana Ruxandra Mihai
- Department of Rheumatology and Physiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (P.R.); (A.M.B.); (I.B.); (I.R.M.); (E.R.)
- I Rheumatology Clinic, Clinical Rehabilitation Hospital, 14 Pantelimon Halipa Street, 700661 Iasi, Romania
| | - Elena Rezus
- Department of Rheumatology and Physiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (P.R.); (A.M.B.); (I.B.); (I.R.M.); (E.R.)
- I Rheumatology Clinic, Clinical Rehabilitation Hospital, 14 Pantelimon Halipa Street, 700661 Iasi, Romania
| |
Collapse
|
3
|
Lee YS, Woo JS, Jhun J, Choi JW, Lee AR, Lee KH, Choi H, Park SH, Cho ML. SARS-CoV-2 spike aggravates lupus nephritis and lung fibrosis in systemic lupus erythematosus. Lupus Sci Med 2024; 11:e001104. [PMID: 39349051 PMCID: PMC11448135 DOI: 10.1136/lupus-2023-001104] [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: 11/13/2023] [Accepted: 08/29/2024] [Indexed: 10/02/2024]
Abstract
OBJECTIVE COVID-19 induces the development of autoimmune diseases, including SLE, which are characterised by inflammation, autoantibodies and thrombosis. However, the effects of COVID-19 on SLE remain unclear. METHODS We investigated the effects of COVID-19 on SLE development and progression in three animal models. Plasmids encoding SARS-CoV-2 spike protein and ACE2 receptor were injected into R848-induced BALB/C lupus mice, R848-induced IL-1 receptor antagonist knockout (KO) lupus mice and MRL/lpr mice. Serum levels of albumin and autoantibodies, lymphocyte phenotypes and tissue histology were evaluated. RESULTS In R848-induced BALB/C lupus mice, the SARS-CoV-2 spike protein increased autoantibody and albumin levels compared with vehicle and mock treatments. These mice also exhibited splenomegaly, which was further exacerbated by the spike protein. Flow cytometric analysis revealed elevated T helper 1 cell counts, and histological analysis indicated increased levels of the fibrosis marker protein α-smooth muscle actin. In KO mice, the spike protein induced splenomegaly, severe kidney damage and pronounced lung fibrosis. In the MRL/lpr group, spike protein increased the serum levels of autoantibodies, albumin and the thrombosis marker chemokine (C-X-C motif) ligand 4. CONCLUSION COVID-19 accelerated the development and progression of lupus by inducing autoantibody production, fibrosis and thrombosis.
Collapse
Affiliation(s)
- Yeon Su Lee
- Lab of Translational ImmunoMedicine (LaTIM), Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, South Korea
- Department of Medical Sciences, Graduate School of The Catholic University of Korea, The Catholic University of Korea, Seoul, South Korea
| | - Jin Seok Woo
- Lab of Translational ImmunoMedicine (LaTIM), Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - JooYeon Jhun
- Lab of Translational ImmunoMedicine (LaTIM), Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, South Korea
- Department of Medical Sciences, Graduate School of The Catholic University of Korea, The Catholic University of Korea, Seoul, South Korea
| | - Jeong Won Choi
- Lab of Translational ImmunoMedicine (LaTIM), Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - A Ram Lee
- Lab of Translational ImmunoMedicine (LaTIM), Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, South Korea
- Department of Medical Sciences, Graduate School of The Catholic University of Korea, The Catholic University of Korea, Seoul, South Korea
| | - Kun Hee Lee
- Lab of Translational ImmunoMedicine (LaTIM), Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, South Korea
- Department of Medical Sciences, Graduate School of The Catholic University of Korea, The Catholic University of Korea, Seoul, South Korea
| | - Haeyoun Choi
- Department of Microbiology, The Catholic University of Korea, Seoul, South Korea
- Catholic Hematopoietic Stem Cell Bank, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Sung-Hwan Park
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Mi-La Cho
- Lab of Translational ImmunoMedicine (LaTIM), Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, South Korea
- Department of Medical Sciences, Graduate School of The Catholic University of Korea, The Catholic University of Korea, Seoul, South Korea
| |
Collapse
|
4
|
Perge B, Papp G, Bói B, Nagy N, Gáspár-Kiss E, Tarr T. Clinical Features and Survival Analysis of Lupus Nephritis among Patients with Systemic Lupus Erythematosus: A Three-Decade-Long Retrospective Cohort Study. Biomedicines 2024; 12:2117. [PMID: 39335630 PMCID: PMC11429493 DOI: 10.3390/biomedicines12092117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 09/09/2024] [Accepted: 09/14/2024] [Indexed: 09/30/2024] Open
Abstract
Background/Objectives: Lupus nephritis (LN) is one of the most severe organ manifestations of systemic lupus erythematosus (SLE). The aim of our retrospective cohort study was to compare the clinical characteristics, therapy, survival, causes of death, and prognostic factors of LN and non-LN lupus patients. Moreover, we compared a wide spectrum of clinical data of LN patients diagnosed before and since 2005 to determine any changes in disease course and outcomes. Methods: We assessed the clinical and laboratory data of 384 SLE patients, out of whom, 127 patients were diagnosed with LN between 1990 and 2020. Results: Based on our observations, discoid LE, subacute cutaneous LE, antiphospholipid syndrome, Sjögren's syndrome, and rheumatoid arthritis were more common in non-LN patients, while anemia and anti-RNP positivity were more frequent in LN patients. Development of LN did not affect survival rates; male sex and presence of APS were negative prognostic parameters in the non-LN group while achieving remission was a positive prognostic factor in both groups. Death caused by sepsis was more prevalent in the LN group. Serositis and neurological manifestations occurred less frequently in LN patients diagnosed after 2005. The use of mycophenolate mofetil became more common, and the cumulative corticosteroid dose decreased. The SLICC Damage Index score also decreased. Conclusions: Our study demonstrated that the disease course has changed in recent years, and the main therapeutic goal in both SLE and lupus nephritis should be to achieve remission because this significantly improves long-term prognosis and patient survival.
Collapse
Affiliation(s)
- Bianka Perge
- Division of Clinical Immunology, Institute of Internal Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - Gábor Papp
- Division of Clinical Immunology, Institute of Internal Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - Bernadett Bói
- Department of Public Health and Epidemiology, Faculty of Medicine, University of Debrecen, H-4028 Debrecen, Hungary
| | - Nikolett Nagy
- Division of Clinical Immunology, Institute of Internal Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - Eszter Gáspár-Kiss
- Division of Clinical Immunology, Institute of Internal Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - Tünde Tarr
- Division of Clinical Immunology, Institute of Internal Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| |
Collapse
|
5
|
Kundnani NR, Levai MC, Popa MD, Borza C, Iacob M, Mederle AL, Blidisel A. Biologics in Systemic Lupus Erythematosus: Recent Evolutions and Benefits. Pharmaceutics 2024; 16:1176. [PMID: 39339212 PMCID: PMC11434659 DOI: 10.3390/pharmaceutics16091176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 08/26/2024] [Accepted: 08/30/2024] [Indexed: 09/30/2024] Open
Abstract
INTRODUCTION Systemic lupus erythematosus (SLE) is a multifaceted autoimmune disorder characterized by significant autoantibodies, particularly targeting nuclear antigens. SLE pathogenesis involves genetic, environmental, and hormonal factors. The disease course includes flares and remission and involves various organs. Recent therapeutic progresses, including biologics, have improved management and prognosis, though the long-term impact of novel therapies remains to be determined. Biologics in SLE: Rituximab, the earliest B-cell-oriented biologic, binds CD20 and depletes CD20+ B cells, leading to remission in some SLE patients. Belimumab is a B-cell-activating factor (BAFF) inhibitor with a recent additional indication for lupus nephritis. The CALIBRATE and BLISS-BELIEVE studies investigated combinations of these drugs with conventional therapies, showing varied efficacy. Ocrelizumab and obinutuzumab, newer CD20-oriented SLE therapies, together with ofatumumab and veltuzumab, are also promising. The latest trials highlight their efficacy and safety. Anifrolumab, targeting type-I interferon receptors, was evaluated in the TULIP 1/2 trials. The ongoing TULIP LTE trial supports the long-term safety and efficacy of anifrolumab. Additionally, the IRIS Phase III trial is exploring anifrolumab for lupus nephritis, showing favorable renal responses. Tocilizumab and secukinumab are being assessed for SLE, with mixed outcomes. Several biologics targeting the C5 complement protein, together with immunomodulators and immunotherapeutics, are also under investigation for potential benefits in SLE. DISCUSSION Biologics in SLE target specific immune components, aiming to improve disease control and reduce the side effects of conventional therapy. However, trial outcomes vary due to factors like inclusion criteria and trial design. CONCLUSIONS Biotechnology progress enables targeted biologic therapies for SLE, reducing disease activity and improving patients' quality of life.
Collapse
Affiliation(s)
- Nilima Rajpal Kundnani
- Department of Cardiology-Internal Medicine and Ambulatory Care, Prevention and Cardiovascular Recovery, "Victor Babeș" University of Medicine and Pharmacy, 300041 Timisoara, Romania
- Research Centre of Timișoara Institute of Cardiovascular Diseases, "Victor Babeș" University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Mihaela Codrina Levai
- Discipline of Medical Communications, Department 2-Microscopic Morphology, "Victor Babeș" University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Mihaela-Diana Popa
- Department of Microbiology, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Claudia Borza
- Discipline of Pathophysiology, Department of Functional Science, "Victor Babeș" University of Medicine and Pharmacy, 300041 Timisoara, Romania
- Center for Translational Research and Systems Medicine, "Victor Babeș" University of Medicine and Pharmacy, 300041 Timisoara, Romania
- Centre of Cognitive Research in Pathological Neuro-Psychiatry NEUROPSY-COG, "Victor Babeș" University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Mihai Iacob
- Advitam Medical Center, 300150 Timisoara, Romania
| | - Alexandra Laura Mederle
- Faculty of Medicine, "Victor Babeș" University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Alexandru Blidisel
- Clinic of Surgical Semiotics and Thoracic Surgery-1, Department IX-Surgery-1, Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania
- Center for Hepato-Biliary-Pancreatic Surgery (CHBP), "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania
| |
Collapse
|
6
|
Bende RJ, Donner N, Wormhoudt TA, Beentjes A, Scantlebery A, Grobben M, Tejjani K, Chandler F, Sikkema RS, Langerak AW, Guikema JE, van Noesel CJ. Distinct groups of autoantigens as drivers of ocular adnexal MALT lymphoma pathogenesis. Life Sci Alliance 2024; 7:e202402841. [PMID: 38977312 PMCID: PMC11231493 DOI: 10.26508/lsa.202402841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 06/28/2024] [Accepted: 06/28/2024] [Indexed: 07/10/2024] Open
Abstract
Chronic B-cell receptor signals incited by cognate antigens are believed to play a crucial role in the pathogenesis of mucosa-associated lymphoid tissue lymphomas. We have explored the immunoglobulin variable regions (IGHV) expressed by 124 ocular adnexal MALT lymphomas (OAML) and tested the in vitro reactivity of recombinant IgM derived from 23 OAMLs. Six of 124 OAMLs (5%) were found to express a high-affinity stereotyped rheumatoid factor. OAMLs have a biased IGHV4-34 usage, which confers intrinsic super auto-antigen reactivity with poly-N-acetyllactosamine (NAL) epitopes, present on cell surface glycoproteins of erythrocytes and B cells. Twenty-one OAMLs (17%) expressed IGHV4-34-encoded B-cell receptors. Five of the 23 recombinant OAML IgMs expressed IGHV4-34, four of which bound to the linear NAL i epitope expressed on B cells but not to the branched NAL I epitope on erythrocytes. One non-IGHV4-34-encoded OAML IgM was also reactive with B cells. Interestingly, three of the 23 OAML IgMs (13%) specifically reacted with proteins of U1-/U-snRNP complexes, which have been implicated as cognate-antigens in various autoimmune diseases such as systemic lupus erythematosus and mixed connective tissue disease. The findings indicate that local autoimmune reactions are instrumental in the pathogenesis of a substantial fraction of OAMLs.
Collapse
MESH Headings
- Humans
- Lymphoma, B-Cell, Marginal Zone/immunology
- Lymphoma, B-Cell, Marginal Zone/genetics
- Autoantigens/immunology
- Immunoglobulin M/immunology
- Immunoglobulin M/metabolism
- Eye Neoplasms/immunology
- Eye Neoplasms/genetics
- Female
- Middle Aged
- Receptors, Antigen, B-Cell/metabolism
- Receptors, Antigen, B-Cell/immunology
- Receptors, Antigen, B-Cell/genetics
- B-Lymphocytes/immunology
- B-Lymphocytes/metabolism
- Male
- Aged
- Immunoglobulin Variable Region/genetics
- Immunoglobulin Variable Region/immunology
- Aged, 80 and over
- Epitopes/immunology
- Adult
- Rheumatoid Factor/immunology
Collapse
Affiliation(s)
- Richard J Bende
- Department of Pathology, Amsterdam UMC, Location University of Amsterdam, Amsterdam, Netherlands
- Lymphoma and Myeloma Center (LYMMCARE), Amsterdam, Netherlands
- Cancer Center Amsterdam (CCA), Amsterdam, Netherlands
| | - Naomi Donner
- Department of Pathology, Amsterdam UMC, Location University of Amsterdam, Amsterdam, Netherlands
| | - Thera Am Wormhoudt
- Department of Pathology, Amsterdam UMC, Location University of Amsterdam, Amsterdam, Netherlands
- Lymphoma and Myeloma Center (LYMMCARE), Amsterdam, Netherlands
- Cancer Center Amsterdam (CCA), Amsterdam, Netherlands
| | - Anna Beentjes
- Department of Pathology, Amsterdam UMC, Location University of Amsterdam, Amsterdam, Netherlands
| | - Angelique Scantlebery
- Department of Pathology, Amsterdam UMC, Location University of Amsterdam, Amsterdam, Netherlands
| | - Marloes Grobben
- Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam UMC, Location University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, Netherlands
| | - Khadija Tejjani
- Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam UMC, Location University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, Netherlands
| | - Felicity Chandler
- https://ror.org/018906e22 Department of Viroscience, Erasmus MC, Rotterdam, Netherlands
| | - Reina S Sikkema
- https://ror.org/018906e22 Department of Viroscience, Erasmus MC, Rotterdam, Netherlands
| | - Anton W Langerak
- https://ror.org/018906e22 Department of Immunology, Laboratory Medical Immunology, Erasmus MC, Rotterdam, Netherlands
| | - Jeroen Ej Guikema
- Department of Pathology, Amsterdam UMC, Location University of Amsterdam, Amsterdam, Netherlands
- Lymphoma and Myeloma Center (LYMMCARE), Amsterdam, Netherlands
- Cancer Center Amsterdam (CCA), Amsterdam, Netherlands
| | - Carel Jm van Noesel
- Department of Pathology, Amsterdam UMC, Location University of Amsterdam, Amsterdam, Netherlands
- Lymphoma and Myeloma Center (LYMMCARE), Amsterdam, Netherlands
- Cancer Center Amsterdam (CCA), Amsterdam, Netherlands
| |
Collapse
|
7
|
Lempicki MD, Gray JA, Abuna G, Murata RM, Divanovic S, McNamara CA, Meher AK. BAFF neutralization impairs the autoantibody-mediated clearance of dead adipocytes and aggravates obesity-induced insulin resistance. Front Immunol 2024; 15:1436900. [PMID: 39185417 PMCID: PMC11341376 DOI: 10.3389/fimmu.2024.1436900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 07/22/2024] [Indexed: 08/27/2024] Open
Abstract
B cell-activating factor (BAFF) is a critical TNF-family cytokine that regulates homeostasis and peripheral tolerance of B2 cells. BAFF overproduction promotes autoantibody generation and autoimmune diseases. During obesity, BAFF is predominantly produced by white adipose tissue (WAT), and IgG autoantibodies against adipocytes are identified in the WAT of obese humans. However, it remains to be determined if the autoantibodies formed during obesity affect WAT remodeling and systemic insulin resistance. Here, we show that IgG autoantibodies are generated in high-fat diet (HFD)-induced obese mice that bind to apoptotic adipocytes and promote their phagocytosis by macrophages. Next, using murine models of obesity in which the gonadal WAT undergoes remodeling, we found that BAFF neutralization depleted IgG autoantibodies, increased the number of dead adipocytes, and exacerbated WAT inflammation and insulin resistance. RNA sequencing of the stromal vascular fraction from the WAT revealed decreased expression of immunoglobulin light-chain and heavy-chain variable genes suggesting a decreased repertoire of B cells after BAFF neutralization. Further, the B cell activation and the phagocytosis pathways were impaired in the WAT of BAFF-neutralized mice. In vitro, plasma IgG fractions from BAFF-neutralized mice reduced the phagocytic clearance of apoptotic adipocytes. Altogether, our study suggests that IgG autoantibodies developed during obesity, at least in part, dampens exacerbated WAT inflammation and systemic insulin resistance.
Collapse
Affiliation(s)
- Melissa D. Lempicki
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, United States
| | - Jake A. Gray
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, United States
| | - Gabriel Abuna
- School of Dental Medicine, East Carolina University, Greenville, NC, United States
| | - Ramiro M. Murata
- School of Dental Medicine, East Carolina University, Greenville, NC, United States
| | - Senad Divanovic
- Department of Pediatrics University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Center for Inflammation and Tolerance, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Coleen A. McNamara
- Cardiovascular Research Center, Cardiovascular Division, Department of Medicine, University of Virginia, Charlottesville, VA, United States
| | - Akshaya K. Meher
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, United States
| |
Collapse
|
8
|
Azzam M, Fahim S, ElMonier A, Maurice N. Functional analysis of a panel of molecular markers for diagnosis of systemic lupus erythematosus in rats. Biosci Rep 2024; 44:BSR20240318. [PMID: 38967046 PMCID: PMC11263041 DOI: 10.1042/bsr20240318] [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: 03/14/2024] [Revised: 06/04/2024] [Accepted: 06/12/2024] [Indexed: 07/06/2024] Open
Abstract
INTRODUCTION Systemic lupus erythematosus (SLE) is a diverse autoimmune disease that arises from a combination of complex genetic factors and environmental influences. While circRNAs and miRNAs have recently been identified as promising biomarkers for disease diagnosis, their specific expression patterns, and clinical implications in SLE are not yet fully understood. AIM OF THE WORK The aim of the present study was to determine the role of a panel of noncoding-RNAs specifically circRNAs (circ-TubD1, circ-CDC27, and circ-Med14), along with miRNA (rno-miR-146a-5p) and mRNA (TRAF6), as novel minimally invasive diagnostic biomarkers for experimentally induced SLE. Additionally, the study involved an insilico bioinformatics analysis to explore potential pathways involved in the pathogenesis of SLE, aiming to enhance our understanding of the disease, enable early diagnosis, and facilitate improved treatment strategies. MATERIALS AND METHODS SLE was induced in rats using single IP injection of incomplete Freund's adjuvant (IFA). The Induction was confirmed by assessing the ANA and anti-ds DNA levels using ELSA technique. qPCR analysis was conducted to assess the expression of selected RNAs in sera collected from a group of 10 rats with induced SLE and a control group of 10 rats. In addition, bioinformatics and functional analysis were used to construct a circRNA-miRNA-mRNA network and to determine the potential function of these differentially expressed circRNAs. RESULTS SLE rats demonstrated significantly higher expression levels of circ-CDC27, circ-Med14, and rno-miR-146a-5p as well as TRAF6, with lower expression level of circ-TubD1 in sera of SLE rats relative to controls. ROC curve analysis indicated that all the selected non-coding RNAs could serve as potential early diagnostic markers for SLE. In addition, the expression level of circ-TubD1 was negatively correlated with rno-miR-146a-5p, however, rno-miR-146a-5p was positively correlated with TRAF6. Bioinformatic analysis revealed the incorporation of the circRNAs targeted genes in various immune system and neurodegeneration pathways. CONCLUSIONS Therefore, circRNAs; circ-TubD1, circ-CDC27, and circ-Med14, in addition to the miRNA (rno-miR-146a-5p) and mRNA (TRAF6) may be involved in the development of SLE and may have promising roles for future diagnosis and targeted therapy.
Collapse
Affiliation(s)
- May A. Azzam
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, 11562, Cairo, Egypt
| | - Sally A. Fahim
- Department of Biochemistry, School of Pharmacy, Newgiza University (NGU), Newgiza, Km 22 Cairo-Alexandria Desert Road, 12577, Giza, Egypt
| | - Asmaa A. ElMonier
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, 11562, Cairo, Egypt
| | - Nadine W. Maurice
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, 11562, Cairo, Egypt
| |
Collapse
|
9
|
Waterman HR, Dufort MJ, Posso SE, Ni M, Li LZ, Zhu C, Raj P, Smith KD, Buckner JH, Hamerman JA. Lupus IgA1 autoantibodies synergize with IgG to enhance plasmacytoid dendritic cell responses to RNA-containing immune complexes. Sci Transl Med 2024; 16:eadl3848. [PMID: 38959329 PMCID: PMC11418372 DOI: 10.1126/scitranslmed.adl3848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Accepted: 06/12/2024] [Indexed: 07/05/2024]
Abstract
Autoantibodies to nuclear antigens are hallmarks of systemic lupus erythematosus (SLE) where they contribute to pathogenesis. However, there remains a gap in our knowledge regarding how different isotypes of autoantibodies contribute to this autoimmune disease, including the production of the critical type I interferon (IFN) cytokines by plasmacytoid dendritic cells (pDCs) in response to immune complexes (ICs). We focused on IgA, which is the second-most prevalent isotype in serum and, along with IgG, is deposited in glomeruli in individuals with lupus nephritis. We show that individuals with SLE have serum IgA autoantibodies against most nuclear antigens, correlating with IgG against the same antigen. We investigated whether IgA autoantibodies against a major SLE autoantigen, Smith ribonucleoprotein (Sm/RNP), played a role in IC activation of pDCs. We found that pDCs expressed the IgA-specific Fc receptor, FcαR, and IgA1 autoantibodies synergized with IgG in RNA-containing ICs to generate robust primary blood pDC IFN-α responses in vitro. pDC responses to these ICs required both FcαR and FcγRIIa, showing synergy between these Fc receptors. Sm/RNP IC binding to and internalization by pDCs were greater when ICs contained both IgA1 and IgG. Circulating pDCs from individuals with SLE had higher binding of IgA1-containing ICs and higher expression of FcαR than pDCs from healthy control individuals. Although pDC FcαR expression correlated with the blood IFN-stimulated gene signature in SLE, Toll-like receptor 7 agonists, but not IFN-α, up-regulated pDC FcαR expression in vitro. Together, we show a mechanism by which IgA1 autoantibodies contribute to SLE pathogenesis.
Collapse
Affiliation(s)
- Hayley R. Waterman
- Molecular and Cell Biology Program, University of Washington; Seattle, 98195, USA
- Center for Fundamental Immunology, Benaroya Research Institute; Seattle, 98101, USA
| | - Matthew J. Dufort
- Center for Systems Immunology, Benaroya Research Institute; Seattle, 98101, USA
| | - Sylvia E. Posso
- Center for Translational Immunology, Benaroya Research Institute, 98101, USA
| | - Minjian Ni
- Center for Fundamental Immunology, Benaroya Research Institute; Seattle, 98101, USA
| | - Lucy Z. Li
- Molecular and Cell Biology Program, University of Washington; Seattle, 98195, USA
- Center for Fundamental Immunology, Benaroya Research Institute; Seattle, 98101, USA
| | - Chengsong Zhu
- Department of Immunology, Microarray and Immune Phenotyping Core Facility, University of Texas Southwestern Medical Center; Dallas, 75390, USA
| | - Prithvi Raj
- Department of Immunology, Microarray and Immune Phenotyping Core Facility, University of Texas Southwestern Medical Center; Dallas, 75390, USA
| | - Kelly D. Smith
- Department of Laboratory Medicine and Pathology, University of Washington; Seattle, 98195, USA
| | - Jane H. Buckner
- Center for Translational Immunology, Benaroya Research Institute, 98101, USA
| | - Jessica A. Hamerman
- Molecular and Cell Biology Program, University of Washington; Seattle, 98195, USA
- Center for Fundamental Immunology, Benaroya Research Institute; Seattle, 98101, USA
- Department of Immunology, University of Washington; Seattle, 98195, USA
| |
Collapse
|
10
|
Iperi C, Fernández-Ochoa Á, Pers JO, Barturen G, Alarcón-Riquelme M, Quirantes-Piné R, Borrás-Linares I, Segura-Carretero A, Cornec D, Bordron A, Jamin C. Integration of multi-omics analysis reveals metabolic alterations of B lymphocytes in systemic lupus erythematosus. Clin Immunol 2024; 264:110243. [PMID: 38735509 DOI: 10.1016/j.clim.2024.110243] [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: 03/20/2024] [Revised: 04/25/2024] [Accepted: 05/03/2024] [Indexed: 05/14/2024]
Abstract
OBJECTIVE To link changes in the B-cell transcriptome from systemic lupus erythematosus (SLE) patients with those in their macroenvironment, including cellular and fluidic components. METHODS Analysis was performed on 363 patients and 508 controls, encompassing transcriptomics, metabolomics, and clinical data. B-cell and whole-blood transcriptomes were analysed using DESeq and GSEA. Plasma and urine metabolomics peak changes were quantified and annotated using Ceu Mass Mediator database. Common sources of variation were identified using MOFA integration analysis. RESULTS Cellular macroenvironment was enriched in cytokines, stress responses, lipidic synthesis/mobility pathways and nucleotide degradation. B cells shared these pathways, except nucleotide degradation diverted to nucleotide salvage pathway, and distinct glycosylation, LPA receptors and Schlafen proteins. CONCLUSIONS B cells showed metabolic changes shared with their macroenvironment and unique changes directly or indirectly induced by IFN-α signalling. This study underscores the importance of understanding the interplay between B cells and their macroenvironment in SLE pathology.
Collapse
Affiliation(s)
| | | | | | - Guillermo Barturen
- GENYO, Centre for Genomics and Oncological Research Pfizer, University of Granada, Andalusian Regional Government, PTS Granada, Granada, Spain; Department of Genetics, Faculty of Sciences, University of Granada, Granada, Spain
| | - Marta Alarcón-Riquelme
- GENYO, Centre for Genomics and Oncological Research Pfizer, University of Granada, Andalusian Regional Government, PTS Granada, Granada, Spain; Institute for Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Rosa Quirantes-Piné
- Research and Development of Functional Food Centre (CIDAF), Health Science Technological Park, Granada, Spain
| | | | | | - Divi Cornec
- LBAI, UMR1227, Univ Brest, Inserm, Brest, France
| | - Anne Bordron
- LBAI, UMR1227, Univ Brest, Inserm, Brest, France
| | | |
Collapse
|
11
|
Ramírez-Valle F, Maranville JC, Roy S, Plenge RM. Sequential immunotherapy: towards cures for autoimmunity. Nat Rev Drug Discov 2024; 23:501-524. [PMID: 38839912 DOI: 10.1038/s41573-024-00959-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/24/2024] [Indexed: 06/07/2024]
Abstract
Despite major progress in the treatment of autoimmune diseases in the past two decades, most therapies do not cure disease and can be associated with increased risk of infection through broad suppression of the immune system. However, advances in understanding the causes of autoimmune disease and clinical data from novel therapeutic modalities such as chimeric antigen receptor T cell therapies provide evidence that it may be possible to re-establish immune homeostasis and, potentially, prolong remission or even cure autoimmune diseases. Here, we propose a 'sequential immunotherapy' framework for immune system modulation to help achieve this ambitious goal. This framework encompasses three steps: controlling inflammation; resetting the immune system through elimination of pathogenic immune memory cells; and promoting and maintaining immune homeostasis via immune regulatory agents and tissue repair. We discuss existing drugs and those in development for each of the three steps. We also highlight the importance of causal human biology in identifying and prioritizing novel immunotherapeutic strategies as well as informing their application in specific patient subsets, enabling precision medicine approaches that have the potential to transform clinical care.
Collapse
|
12
|
Subramani J, Kumar GS, Gadekallu TR. Gene-Based Predictive Modelling for Enhanced Detection of Systemic Lupus Erythematosus Using CNN-Based DL Algorithm. Diagnostics (Basel) 2024; 14:1339. [PMID: 39001231 PMCID: PMC11240797 DOI: 10.3390/diagnostics14131339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 06/13/2024] [Accepted: 06/20/2024] [Indexed: 07/16/2024] Open
Abstract
Systemic Lupus Erythematosus (SLE) is a multifaceted autoimmune disease that presents with a diverse array of clinical signs and unpredictable disease progression. Conventional diagnostic methods frequently fall short in terms of sensitivity and specificity, which can result in delayed diagnosis and less-than-optimal management. In this study, we introduce a novel approach for improving the identification of SLE through the use of gene-based predictive modelling and Stacked deep learning classifiers. The study proposes a new method for diagnosing SLE using Stacked Deep Learning Classifiers (SDLC) trained on Gene Expression Omnibus (GEO) database data. By combining transcriptomic data from GEO with clinical features and laboratory results, the SDLC model achieves a remarkable accuracy value of 0.996, outperforming traditional methods. Individual models within the SDLC, such as SBi-LSTM and ACNN, achieved accuracies of 92% and 95%, respectively. The SDLC's ensemble learning approach allows for identifying complex patterns in multi-modal data, enhancing accuracy in diagnosing SLE. This study emphasises the potential of deep learning methods, in conjunction with open repositories like GEO, to advance the diagnosis and management of SLE. Overall, this research shows strong performance and potential for improving precision medicine in managing SLE.
Collapse
Affiliation(s)
- Jothimani Subramani
- Department of Information Technology, Bannari Amman Institute of Technology, Sathyamangalam 638401, Tamil Nadu, India
| | - G Sathish Kumar
- Department of Artificial Intelligence and Data Science, Sri Eshwar College of Engineering, Coimbatore 641202, Tamil Nadu, India
| | - Thippa Reddy Gadekallu
- Division of Research and Development, Lovely Professional University, Phagwara 144411, Punjab, India
- Center of Research Impact and Outcome, Chitkara University, Rajpura 140401, Punjab, India
| |
Collapse
|
13
|
Akatsu C, Tsuneshige T, Numoto N, Long W, Uchiumi T, Kaneko Y, Asano M, Ito N, Tsubata T. CD72 is an inhibitory pattern recognition receptor that recognizes ribosomes and suppresses production of anti-ribosome autoantibody. J Autoimmun 2024; 146:103245. [PMID: 38754236 DOI: 10.1016/j.jaut.2024.103245] [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: 01/08/2024] [Revised: 04/22/2024] [Accepted: 05/06/2024] [Indexed: 05/18/2024]
Abstract
B cell responses to nucleic acid-containing self-antigens that involve intracellular nucleic acid sensors play a crucial role in autoantibody production in SLE. CD72 is an inhibitory B cell co-receptor that down-regulates BCR signaling, and prevents the development of SLE. We previously showed that CD72 recognizes the RNA-containing self-antigen Sm/RNP, a target of SLE-specific autoantibodies, and induces B cell tolerance to Sm/RNP by specifically inhibiting B cell response to this self-antigen. Here, we address whether CD72 inhibits B cell response to ribosomes because the ribosome is an RNA-containing self-antigen and is a target of SLE-specific autoantibodies as well as Sm/RNP. We demonstrate that CD72 recognizes ribosomes as a ligand, and specifically inhibits BCR signaling induced by ribosomes. Although conventional protein antigens by themselves do not induce proliferation of specific B cells, ribosomes induce proliferation of B cells reactive to ribosomes in a manner dependent on RNA. This proliferative response is down-regulated by CD72. These results suggest that ribosomes activate B cells by inducing dual signaling through BCR and intracellular RNA sensors and that CD72 inhibits B cell response to ribosomes. Moreover, CD72-/- but not CD72+/+ mice spontaneously produce anti-ribosome autoantibodies. Taken together, CD72 induces B cell self-tolerance to ribosomes by recognizing ribosomes and inhibiting RNA-dependent B cell response to this self-antigen. CD72 appears to prevent development of SLE by inhibiting autoimmune B cell responses to multiple RNA-containing self-antigens. Because these self-antigens but not protein self-antigens induce RNA-dependent B cell activation, self-tolerance to RNA-containing self-antigens may require a distinct tolerance mechanism mediated by CD72.
Collapse
MESH Headings
- Animals
- Ribosomes/metabolism
- Ribosomes/immunology
- Mice
- Receptors, Antigen, B-Cell/metabolism
- Receptors, Antigen, B-Cell/immunology
- Autoantibodies/immunology
- Lupus Erythematosus, Systemic/immunology
- Lupus Erythematosus, Systemic/metabolism
- Antigens, Differentiation, B-Lymphocyte/immunology
- Antigens, Differentiation, B-Lymphocyte/metabolism
- Antigens, CD/metabolism
- Antigens, CD/immunology
- B-Lymphocytes/immunology
- B-Lymphocytes/metabolism
- Signal Transduction/immunology
- Autoantigens/immunology
- Mice, Knockout
- Lymphocyte Activation/immunology
- Cell Proliferation
- Immune Tolerance
- Humans
Collapse
Affiliation(s)
- Chizuru Akatsu
- Department of Immunology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Takahiro Tsuneshige
- Department of Immunology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan; Nihon University School of Dentistry, Tokyo, Japan
| | - Nobutaka Numoto
- Department of Structural Biology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Wang Long
- Nihon University School of Dentistry, Tokyo, Japan
| | - Toshio Uchiumi
- Department of Biology, Niigata University School of Science, Niigata, Japan
| | - Yoshikatsu Kaneko
- Division of Clinical Nephrology and Rheumatology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | | | - Nobutoshi Ito
- Department of Structural Biology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Takeshi Tsubata
- Department of Immunology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan; Nihon University School of Dentistry, Tokyo, Japan.
| |
Collapse
|
14
|
Lorca-Arce D, Pérez-Isidro A, Becerra J, Martínez MJ, De Moner N, Ríos-Garcés R, Prieto-González S, Espinosa G, Cervera R, Andalucía C, Viñas-Gomis O, Ruiz-Ortiz E. Evaluation of a novel particle-based assay for detecting SLE-related autoantibodies. Heliyon 2024; 10:e30767. [PMID: 38778929 PMCID: PMC11108844 DOI: 10.1016/j.heliyon.2024.e30767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 04/18/2024] [Accepted: 05/04/2024] [Indexed: 05/25/2024] Open
Abstract
Anti-dsDNA, anti-Sm, and anti-ribosomal-P autoantibodies are hallmarks of systemic lupus erythematosus (SLE), being anti-dsDNA and anti-Sm included in 2019-ACR/EULAR SLE-Classification Criteria. Enzyme-linked (ELISA) and chemiluminescence assays (CIA) are widely established in immunology laboratories, but new technologies, such as particle-based multi-analyte technology (PMAT), are nowadays available. The present study aimed to compare the presence of anti-dsDNA and anti-Sm autoantibodies measured by CIA and PMAT and analyze diagnostic and clinical SLE activity performance. Anti-ribosomal-P autoantibodies by PMAT were also included. Consequently, anti-dsDNA and anti-Sm detected by CIA showed substantial agreement with PMAT (Cohen's kappa = 0.662 and 0.671, respectively). Anti-dsDNA autoantibodies measured by PMAT showed a positive correlation with clinical SLEDAI-2K (p < 0.001) and a negative correlation with complement consumption (p < 0.001). Anti-Sm and anti-ribosomal-P autoantibodies showed a positive correlation with SLEDAI-2K (p < 0.001 and p = 0.001, respectively) and a negative correlation with complement consumption (p < 0.001 and p = 0.001, respectively). Finally, anti-Sm autoantibodies were associated with renal involvement (p < 0.05).
Collapse
Affiliation(s)
- Daniel Lorca-Arce
- Immunology Department, Centre Diagnostic Biomèdic CDB, Hospital Clínic de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Albert Pérez-Isidro
- Immunology Department, Centre Diagnostic Biomèdic CDB, Hospital Clínic de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Judit Becerra
- Immunology Department, Centre Diagnostic Biomèdic CDB, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Maria José Martínez
- Immunology Department, Centre Diagnostic Biomèdic CDB, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Noemí De Moner
- Immunology Department, Centre Diagnostic Biomèdic CDB, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Roberto Ríos-Garcés
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
- Department of Autoimmune Diseases, Reference Centre for Systemic Autoimmune Diseases (UEC/CSUR) of the Catalan and Spanish Health Systems-Member of ERNReCONNET, Hospital Clínic, Barcelona, Catalonia, Spain
| | - Sergio Prieto-González
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
- Department of Autoimmune Diseases, Reference Centre for Systemic Autoimmune Diseases (UEC/CSUR) of the Catalan and Spanish Health Systems-Member of ERNReCONNET, Hospital Clínic, Barcelona, Catalonia, Spain
| | - Gerard Espinosa
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
- Department of Autoimmune Diseases, Reference Centre for Systemic Autoimmune Diseases (UEC/CSUR) of the Catalan and Spanish Health Systems-Member of ERNReCONNET, Hospital Clínic, Barcelona, Catalonia, Spain
| | - Ricard Cervera
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
- Department of Autoimmune Diseases, Reference Centre for Systemic Autoimmune Diseases (UEC/CSUR) of the Catalan and Spanish Health Systems-Member of ERNReCONNET, Hospital Clínic, Barcelona, Catalonia, Spain
| | - Carmen Andalucía
- Research and Development, Headquarters & Technology Center Autoimmunity, Werfen, San Diego, CA, USA
| | - Odette Viñas-Gomis
- Immunology Department, Centre Diagnostic Biomèdic CDB, Hospital Clínic de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Estibaliz Ruiz-Ortiz
- Immunology Department, Centre Diagnostic Biomèdic CDB, Hospital Clínic de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
15
|
Goh RCW, Maharajan MK, Gopinath D, Fang CM. The Therapeutic Effects of Probiotic on Systemic Lupus Erythematosus in Lupus Mice Models: A Systematic Review. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10297-1. [PMID: 38806966 DOI: 10.1007/s12602-024-10297-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/18/2024] [Indexed: 05/30/2024]
Abstract
Increasing evidence suggests the beneficial immunomodulatory effects of probiotics can reduce inflammation in systemic lupus erythematosus (SLE). However, there is no summary of the existing evidence available. This study aims to investigate the therapeutic effects of probiotics on SLE in a lupus mouse model by examining various markers, including inflammatory cytokines, Treg cells, disease activity, and gut microbiota. A systematic search was conducted using three databases (Web of Science, PubMed, and Scopus) to identify animal studies that reported the therapeutic benefits of probiotics against SLE. Data extracted from the selected articles were qualitatively synthesized. The SYRCLE risk of bias tool was used to evaluate the risk of bias. Out of a total of 3205 articles, 12 met the inclusion criteria. Probiotic strains, quantities, and routes of administration varied among the studies. The treatment ranged from 8 to 47 weeks. Probiotic strains such as L. fermentum CECT5716, L. casei B255, L. reuteri DSM 17509, L. plantarum LP299v, and L. acidophilus can significantly reduce pro-inflammatory cytokines (TNF-α, IL-12, IL-6, IL-1β, IL-17, and IFN-γ) levels while increasing anti-inflammatory IL-10 and Treg cells. Probiotics also delay the production of autoantibodies, thus prolonging the remission period, decreasing flare frequency, and delaying disease progression. Furthermore, probiotic administration prevents gut dysbiosis, increases intestinal stability, and prevents pathogen colonization. In conclusion, probiotics can be considered a new alternative therapeutic approach for the management of SLE. Further clinical studies are required to investigate and validate the safety and effectiveness of probiotics in humans.
Collapse
Affiliation(s)
- Rachael Chaeh-Wen Goh
- Division of Biomedical Sciences, School of Pharmacy, University of Nottingham Malaysia, Jalan Broga, 43500, Semenyih, Malaysia
| | - Mari Kannan Maharajan
- School of Pharmacy, University of Nottingham Malaysia, Jalan Broga, 43500, Semenyih, Malaysia
| | - Divya Gopinath
- Basic Medical and Dental Sciences Department, College of Dentistry, Ajman University, P.O. Box 346, Ajman, United Arab Emirates
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Chee-Mun Fang
- Division of Biomedical Sciences, School of Pharmacy, University of Nottingham Malaysia, Jalan Broga, 43500, Semenyih, Malaysia.
| |
Collapse
|
16
|
Lovell CD, Jiwrajka N, Amerman HK, Cancro MP, Anguera MC. Xist Deletion in B Cells Results in Systemic Lupus Erythematosus Phenotypes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.15.594175. [PMID: 38798403 PMCID: PMC11118349 DOI: 10.1101/2024.05.15.594175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease preferentially observed in females. X-linked gene expression in XX females is normalized to that of XY males by X-Chromosome Inactivation (XCI). However, B cells from female SLE patients and mouse models of SLE exhibit mislocalization of Xist RNA, a critical regulator of XCI, and aberrant expression of X-linked genes, suggesting that impairment of XCI may contribute to disease. Here, we find that a subset of female mice harboring a conditional deletion of Xis t in B cells ("Xist cKO") spontaneously develop SLE phenotypes, including expanded activated B cell subsets, disease-specific autoantibodies, and glomerulonephritis. Moreover, pristane-induced SLE-like disease is more severe in Xist cKO mice. Activated B cells from Xist cKO mice with SLE phenotypes have increased expression of proinflammatory X-linked genes implicated in SLE. Together, this work indicates that impaired XCI maintenance in B cells directly contributes to the female-bias of SLE.
Collapse
|
17
|
von Hofsten S, Fenton KA, Pedersen HL. Human and Murine Toll-like Receptor-Driven Disease in Systemic Lupus Erythematosus. Int J Mol Sci 2024; 25:5351. [PMID: 38791389 PMCID: PMC11120885 DOI: 10.3390/ijms25105351] [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: 04/26/2024] [Revised: 05/10/2024] [Accepted: 05/12/2024] [Indexed: 05/26/2024] Open
Abstract
The pathogenesis of systemic lupus erythematosus (SLE) is linked to the differential roles of toll-like receptors (TLRs), particularly TLR7, TLR8, and TLR9. TLR7 overexpression or gene duplication, as seen with the Y-linked autoimmune accelerator (Yaa) locus or TLR7 agonist imiquimod, correlates with increased SLE severity, and specific TLR7 polymorphisms and gain-of-function variants are associated with enhanced SLE susceptibility and severity. In addition, the X-chromosome location of TLR7 and its escape from X-chromosome inactivation provide a genetic basis for female predominance in SLE. The absence of TLR8 and TLR9 have been shown to exacerbate the detrimental effects of TLR7, leading to upregulated TLR7 activity and increased disease severity in mouse models of SLE. The regulatory functions of TLR8 and TLR9 have been proposed to involve competition for the endosomal trafficking chaperone UNC93B1. However, recent evidence implies more direct, regulatory functions of TLR9 on TLR7 activity. The association between age-associated B cells (ABCs) and autoantibody production positions these cells as potential targets for treatment in SLE, but the lack of specific markers necessitates further research for precise therapeutic intervention. Therapeutically, targeting TLRs is a promising strategy for SLE treatment, with drugs like hydroxychloroquine already in clinical use.
Collapse
Affiliation(s)
- Susannah von Hofsten
- Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, 9019 Tromsø, Norway;
| | - Kristin Andreassen Fenton
- Centre of Clinical Research and Education, University Hospital of North Norway, Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, 9019 Tromsø, Norway;
| | - Hege Lynum Pedersen
- Centre of Clinical Research and Education, University Hospital of North Norway, Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, 9019 Tromsø, Norway;
| |
Collapse
|
18
|
Fernández-Cladera Y, García-González M, Hernández-Díaz M, Gómez-Bernal F, Quevedo-Abeledo JC, González-Rivero AF, de Vera-González A, Gómez-Moreno C, González-Gay MÁ, Ferraz-Amaro I. Relationship of Hematological Profiles with the Serum Complement System in Patients with Systemic Lupus Erythematosus. Biomedicines 2024; 12:967. [PMID: 38790929 PMCID: PMC11117834 DOI: 10.3390/biomedicines12050967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 04/12/2024] [Accepted: 04/25/2024] [Indexed: 05/26/2024] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disorder identified by hematological abnormalities including anemia, leukopenia, and thrombocytopenia. Complement system disturbance is implicated in the pathogenesis of SLE. In this work, we aim to study how a full assessment of the complement system, which includes the evaluation of its three pathways, relates to blood cell counts in a population of patients with SLE. New-generation functional assays of the classical, alternative, and lectin pathways of the complement system were conducted in 284 patients with SLE. Additionally, serum levels of inactive molecules (C1q, C2, C3, C4, factor D) and activated molecules (C3a), as well as regulators (C1-inhibitor and factor H), were evaluated. Complete blood cell counts were analyzed. Multivariable linear regression analysis was performed to study the relationship of hematological profiles with this full characterization of the complement system. After multivariable adjustments that included age, sex, SLICC-DI (damage), and SLEDAI (activity) scores, as well as the use of aspirin, prednisone, methotrexate, azathioprine, and mycophenolate mofetil, several relationships were observed between the C pathways and the individual products and blood cells profile. Lower values of C1q and C2 were associated with lower hemoglobin levels. Lower leukocyte counts showed significantly lower values of C4, C1 inhibitor, C3, factor D, and alternative pathway functional levels. Neutrophil counts showed significant negative relationships only with the alternative pathway and C1-inh. In the case of lymphocytes, associations were found, especially with functional tests of the classical and alternative pathways, as well as with C2, C4, C3, and C3a. On the contrary, for platelets, significance was only observed, after multivariable adjustment, with lower C2 concentrations. In conclusion, the serum complement system and hematological profile in SLE are independently linked, after adjustment for disease activity and damage. These relationships are basically negative and are predominantly found in lymphocytes.
Collapse
Affiliation(s)
- Yolanda Fernández-Cladera
- Division of Central Laboratory, Hospital Universitario de Canarias, 38320 Tenerife, Spain; (Y.F.-C.); (F.G.-B.); (A.F.G.-R.); (A.d.V.-G.)
| | - María García-González
- Division of Rheumatology, Hospital Universitario de Canarias, 38320 Tenerife, Spain; (M.G.-G.); (M.H.-D.)
| | - Marta Hernández-Díaz
- Division of Rheumatology, Hospital Universitario de Canarias, 38320 Tenerife, Spain; (M.G.-G.); (M.H.-D.)
| | - Fuensanta Gómez-Bernal
- Division of Central Laboratory, Hospital Universitario de Canarias, 38320 Tenerife, Spain; (Y.F.-C.); (F.G.-B.); (A.F.G.-R.); (A.d.V.-G.)
| | | | - Agustín F. González-Rivero
- Division of Central Laboratory, Hospital Universitario de Canarias, 38320 Tenerife, Spain; (Y.F.-C.); (F.G.-B.); (A.F.G.-R.); (A.d.V.-G.)
| | - Antonia de Vera-González
- Division of Central Laboratory, Hospital Universitario de Canarias, 38320 Tenerife, Spain; (Y.F.-C.); (F.G.-B.); (A.F.G.-R.); (A.d.V.-G.)
| | - Cristina Gómez-Moreno
- School of Nursing, Fundación Jiménez Díaz, Autonomous University of Madrid, 28040 Madrid, Spain;
| | - Miguel Á. González-Gay
- Division of Rheumatology, Instituto de Investigación Sanitaria Fundación Jiménez Díaz, 28040 Madrid, Spain
- Department of Internal Medicine, University of Cantabria, 39005 Santander, Spain
| | - Iván Ferraz-Amaro
- Division of Rheumatology, Hospital Universitario de Canarias, 38320 Tenerife, Spain; (M.G.-G.); (M.H.-D.)
- Department of Internal Medicine, University of La Laguna (ULL), 38200 Tenerife, Spain
| |
Collapse
|
19
|
Li Q, Jia C, Pan W, Liu H, Tang C, Weber D, Chen K, Long H, Byrne-Steele ML, Han J, He N, Xiao R, Zhao M, Che N, Guo Q, Gui G, Li S, Si H, Guo S, Liu H, Wang G, Zhu G, Yang B, Wang Y, Ding Y, Yang X, Akihiko Y, Lu L, Chang C, Chan V, Lau CS, Qi H, Liu W, Li S, Wu H, Lu Q. Multi-omics study reveals different pathogenesis of the generation of skin lesions in SLE and IDLE patients. J Autoimmun 2024; 146:103203. [PMID: 38643729 DOI: 10.1016/j.jaut.2024.103203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 02/28/2024] [Accepted: 03/04/2024] [Indexed: 04/23/2024]
Abstract
Lupus erythematosus (LE) is a heterogeneous, antibody-mediated autoimmune disease. Isolate discoid LE (IDLE) and systematic LE (SLE) are traditionally regarded as the two ends of the spectrum, ranging from skin-limited damage to life-threatening multi-organ involvement. Both belong to LE, but IDLE and SLE differ in appearance of skin lesions, autoantibody panels, pathological changes, treatments, and immunopathogenesis. Is discoid lupus truly a form of LE or is it a completely separate entity? This question has not been fully elucidated. We compared the clinical data of IDLE and SLE from our center, applied multi-omics technology, such as immune repertoire sequencing, high-resolution HLA alleles sequencing and multi-spectrum pathological system to explore cellular and molecular phenotypes in skin and peripheral blood from LE patients. Based on the data from 136 LE patients from 8 hospitals in China, we observed higher damage scores and fewer LE specific autoantibodies in IDLE than SLE patients, more uCDR3 sharing between PBMCs and skin lesion from SLE than IDLE patients, elevated diversity of V-J recombination in IDLE skin lesion and SLE PBMCs, increased SHM frequency and class switch ratio in IDLE skin lesion, decreased SHM frequency but increased class switch ratio in SLE PBMCs, HLA-DRB1*03:01:01:01, HLA-B*58:01:01:01, HLA-C*03:02:02:01, and HLA-DQB1*02:01:01:01 positively associated with SLE patients, and expanded Tfh-like cells with ectopic germinal center structures in IDLE skin lesions. These findings suggest a significant difference in the immunopathogenesis of skin lesions between SLE and IDLE patients. SLE is a B cell-predominate systemic immune disorder, while IDLE appears limited to the skin. Our findings provide novel insights into the pathogenesis of IDLE and other types of LE, which may direct more accurate diagnosis and novel therapeutic strategies.
Collapse
Affiliation(s)
- Qianwen Li
- Department of Dermatology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, Hunan, 410011, China
| | - Chen Jia
- Department of Dermatology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, Hunan, 410011, China
| | - Wenjing Pan
- Nanjing ARP Biotechnology Co., Ltd, Nanjing, Jiangsu, China; iRepertoire Inc., Huntsville, AL, USA
| | - Hongmei Liu
- Hunan University of Technology, Hunan Key Laboratory of Biomedical Nanomaterials and Devices, Zhuzhou, Hunan, China
| | - Congli Tang
- Nanjing ARP Biotechnology Co., Ltd, Nanjing, Jiangsu, China
| | | | - Kaili Chen
- Department of Dermatology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, Hunan, 410011, China
| | - Hai Long
- Department of Dermatology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, Hunan, 410011, China
| | | | - Jian Han
- iRepertoire Inc., Huntsville, AL, USA
| | - Nongyue He
- Nanjing ARP Biotechnology Co., Ltd, Nanjing, Jiangsu, China
| | - Rong Xiao
- Department of Dermatology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, Hunan, 410011, China
| | - Ming Zhao
- Department of Dermatology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, Hunan, 410011, China; Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, 210042, China; Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China; Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, China
| | - Nan Che
- Department of Rheumatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qing Guo
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510000, China
| | - Guangji Gui
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510000, China
| | - Shanshan Li
- Department of Dermatology, The First Bethune Hospital of Jilin University, Changchun, Jilin, 130000, China
| | - Henan Si
- Department of Dermatology, The First Bethune Hospital of Jilin University, Changchun, Jilin, 130000, China
| | - Shuping Guo
- Department of Dermatology, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030000, China
| | - Hongye Liu
- Department of Dermatology, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030000, China
| | - Gang Wang
- Department of Dermatology, Xijing Hospital, Xi'an, Shaanxi, 710000, China
| | - Guannan Zhu
- Department of Dermatology, Xijing Hospital, Xi'an, Shaanxi, 710000, China
| | - Bin Yang
- Dermatology Hospital of Southern Medical University, Guangzhou, Guangdong, 510000, China
| | - Yu Wang
- Dermatology Hospital of Southern Medical University, Guangzhou, Guangdong, 510000, China
| | - Yan Ding
- Hainan Provincial Hospital of Skin Disease, Haikou, Hainan, 570100, China
| | - Xianxu Yang
- Hainan Provincial Hospital of Skin Disease, Haikou, Hainan, 570100, China
| | - Yoshimura Akihiko
- Department of Microbiology and Immunology, Keio University School of Medicine 35 Shinanoomachi, Shinjyuku-ku, Tokyo, 160-8582, Japan
| | - Liwei Lu
- Department of Pathology and Shenzhen Institute of Research and Innovation, The University of Hong Kong, Hong Kong, 999077, China
| | - Christopher Chang
- Division of Immunology, Allergy and Rheumatology, Memorial Healthcare System, Joe DiMaggio Children's Hospital, Hollywood, FL, USA
| | - Vera Chan
- Division of Rheumatology & Clinical Immunology, Department of Medicine, University of Hong Kong, Hong Kong, China
| | - Chak-Sing Lau
- Division of Rheumatology & Clinical Immunology, Department of Medicine, University of Hong Kong, Hong Kong, China
| | - Hai Qi
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Wanli Liu
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Song Li
- Hunan University of Technology, Hunan Key Laboratory of Biomedical Nanomaterials and Devices, Zhuzhou, Hunan, China.
| | - Haijing Wu
- Department of Dermatology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, Hunan, 410011, China.
| | - Qianjin Lu
- Department of Dermatology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, Hunan, 410011, China; Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, 210042, China; Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China; Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, China.
| |
Collapse
|
20
|
Corona-Meraz FI, Vázquez-Del Mercado M, Sandoval-García F, Robles-De Anda JA, Tovar-Cuevas AJ, Rosales-Gómez RC, Guzmán-Ornelas MO, González-Inostroz D, Peña-Nava M, Martín-Márquez BT. Biomarkers in Systemic Lupus Erythematosus along with Metabolic Syndrome. J Clin Med 2024; 13:1988. [PMID: 38610754 PMCID: PMC11012563 DOI: 10.3390/jcm13071988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 03/25/2024] [Accepted: 03/26/2024] [Indexed: 04/14/2024] Open
Abstract
Metabolic syndrome (MetS) is a group of physiological abnormalities characterized by obesity, insulin resistance (IR), and hypertriglyceridemia, which carry the risk of developing cardiovascular disease (CVD) and type 2 diabetes (T2D). Immune and metabolic alterations have been observed in MetS and are associated with autoimmune development. Systemic lupus erythematosus (SLE) is an autoimmune disease caused by a complex interaction of environmental, hormonal, and genetic factors and hyperactivation of immune cells. Patients with SLE have a high prevalence of MetS, in which elevated CVD is observed. Among the efforts of multidisciplinary healthcare teams to make an early diagnosis, a wide variety of factors have been considered and associated with the generation of biomarkers. This review aimed to elucidate some primary biomarkers and propose a set of assessments to improve the projection of the diagnosis and evolution of patients. These biomarkers include metabolic profiles, cytokines, cardiovascular tests, and microRNAs (miRs), which have been observed to be dysregulated in these patients and associated with outcomes.
Collapse
Affiliation(s)
- Fernanda Isadora Corona-Meraz
- Multidisciplinary Health Research Center, Department of Biomedical Sciences, University Center of Tonala, University of Guadalajara, Guadalajara 45425, Jalisco, Mexico; (A.-J.T.-C.); (R.-C.R.-G.); (M.-O.G.-O.)
- Department of Molecular Biology and Genomics, Institute of Rheumatology and Musculoskeletal System Research, University Center of Health Sciences, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico; (M.V.-D.M.); (F.S.-G.); (J.-A.R.-D.A.); (D.G.-I.); (M.P.-N.)
| | - Mónica Vázquez-Del Mercado
- Department of Molecular Biology and Genomics, Institute of Rheumatology and Musculoskeletal System Research, University Center of Health Sciences, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico; (M.V.-D.M.); (F.S.-G.); (J.-A.R.-D.A.); (D.G.-I.); (M.P.-N.)
- Rheumatology Service, Internal Medicine Division, Civil Hospital of Guadalajara “Dr. Juan I. Menchaca”, Guadalajara 44340, Jalisco, Mexico
- Academic Group UDG-CA-703, “Immunology and Rheumatology”, University Center of Health Sciences, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Flavio Sandoval-García
- Department of Molecular Biology and Genomics, Institute of Rheumatology and Musculoskeletal System Research, University Center of Health Sciences, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico; (M.V.-D.M.); (F.S.-G.); (J.-A.R.-D.A.); (D.G.-I.); (M.P.-N.)
- Academic Group UDG-CA-703, “Immunology and Rheumatology”, University Center of Health Sciences, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Jesus-Aureliano Robles-De Anda
- Department of Molecular Biology and Genomics, Institute of Rheumatology and Musculoskeletal System Research, University Center of Health Sciences, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico; (M.V.-D.M.); (F.S.-G.); (J.-A.R.-D.A.); (D.G.-I.); (M.P.-N.)
| | - Alvaro-Jovanny Tovar-Cuevas
- Multidisciplinary Health Research Center, Department of Biomedical Sciences, University Center of Tonala, University of Guadalajara, Guadalajara 45425, Jalisco, Mexico; (A.-J.T.-C.); (R.-C.R.-G.); (M.-O.G.-O.)
| | - Roberto-Carlos Rosales-Gómez
- Multidisciplinary Health Research Center, Department of Biomedical Sciences, University Center of Tonala, University of Guadalajara, Guadalajara 45425, Jalisco, Mexico; (A.-J.T.-C.); (R.-C.R.-G.); (M.-O.G.-O.)
| | - Milton-Omar Guzmán-Ornelas
- Multidisciplinary Health Research Center, Department of Biomedical Sciences, University Center of Tonala, University of Guadalajara, Guadalajara 45425, Jalisco, Mexico; (A.-J.T.-C.); (R.-C.R.-G.); (M.-O.G.-O.)
| | - Daniel González-Inostroz
- Department of Molecular Biology and Genomics, Institute of Rheumatology and Musculoskeletal System Research, University Center of Health Sciences, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico; (M.V.-D.M.); (F.S.-G.); (J.-A.R.-D.A.); (D.G.-I.); (M.P.-N.)
| | - Miguel Peña-Nava
- Department of Molecular Biology and Genomics, Institute of Rheumatology and Musculoskeletal System Research, University Center of Health Sciences, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico; (M.V.-D.M.); (F.S.-G.); (J.-A.R.-D.A.); (D.G.-I.); (M.P.-N.)
| | - Beatriz-Teresita Martín-Márquez
- Department of Molecular Biology and Genomics, Institute of Rheumatology and Musculoskeletal System Research, University Center of Health Sciences, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico; (M.V.-D.M.); (F.S.-G.); (J.-A.R.-D.A.); (D.G.-I.); (M.P.-N.)
- Academic Group UDG-CA-703, “Immunology and Rheumatology”, University Center of Health Sciences, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
| |
Collapse
|
21
|
McDonnell SRP, Nguyen VA, Walton NM, Merkwirth C, Hong F, Berg D, Muensterman ET, Furie RA. Mezagitamab in systemic lupus erythematosus: clinical and mechanistic findings of CD38 inhibition in an autoimmune disease. Lupus Sci Med 2024; 11:e001112. [PMID: 38453421 PMCID: PMC10921479 DOI: 10.1136/lupus-2023-001112] [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: 11/22/2023] [Accepted: 02/15/2024] [Indexed: 03/09/2024]
Abstract
OBJECTIVE To evaluate safety and mechanism of action of mezagitamab (TAK-079), an anti-CD38 monoclonal antibody, in patients with moderate to severe systemic lupus erythematosus (SLE). METHODS A phase 1b double-blind, placebo-controlled, multicentre study was conducted in patients with SLE receiving standard background therapy. Eligible patients were adults who met the 2012 SLICC or ACR criteria for diagnosis, had a baseline SLE Disease Activity Index 2000 (SLEDAI-2K) score of ≥6 and were positive for anti-double-stranded DNA antibodies and/or anti-extractable nuclear antigens antibodies. Patients received 45 mg, 90 mg or 135 mg of mezagitamab or placebo every 3 weeks over 12 weeks. Primary endpoints were safety and tolerability. Secondary endpoints included pharmacokinetics and pharmacodynamics. Exploratory assessments included disease activity scales, deep immune profiling and interferon pathway analysis. RESULTS 22 patients received at least one dose of either mezagitamab or placebo. In patients exposed to mezagitamab (n=17), drug was well tolerated. Adverse event (AEs) were balanced across treatment groups, with no treatment emergent AEs exceeding grade 2. Responder analyses for Cutaneous Lupus Erythematosus Disease Area and Severity Index (CLASI) and SLEDAI-2K did not reveal any observable differences across treatment groups. However, there was a trend for more profound skin responses among patients with higher CLASI scores (>10) at baseline. Pharmacodynamic analysis showed median CD38 receptor occupancy up to 88.4% on CD38+ natural killer cells with concurrent depletion of these cells up to 90% in the 135 mg group. Mean reductions in IgG and autoantibodies were less than 20% in all dose groups. Cytometry by time of flight and type 1 interferon gene analysis revealed unique fingerprints that are indicative of a broad immune landscape shift following CD38 targeting. CONCLUSIONS Mezagitamab had a favourable safety profile in patients with moderate to severe SLE and elicited a pharmacodynamic effect consistent with CD38+ cell depletion. These findings reveal novel insights into the drug's mechanism of action and support the continued investigation of mezagitamab in autoimmune diseases.
Collapse
Affiliation(s)
| | - Van Anh Nguyen
- Takeda Development Center Americas Inc, Lexington, Massachusetts, USA
| | - Noah M Walton
- Takeda Development Center Americas Inc, Lexington, Massachusetts, USA
| | - Carsten Merkwirth
- Takeda Development Center Americas Inc, Lexington, Massachusetts, USA
| | - Feng Hong
- Takeda Development Center Americas Inc, Lexington, Massachusetts, USA
| | - Deborah Berg
- Clinical Sciences, Takeda Pharmaceuticals America Inc, Lexington, Massachusetts, USA
| | | | - Richard A Furie
- Department of Rheumatology, Northwell Health, Great Neck, New York, USA
| |
Collapse
|
22
|
Vitzthum von Eckstaedt H, Singh A, Reid P, Trotter K. Immune Checkpoint Inhibitors and Lupus Erythematosus. Pharmaceuticals (Basel) 2024; 17:252. [PMID: 38399467 PMCID: PMC10892070 DOI: 10.3390/ph17020252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 01/16/2024] [Accepted: 02/01/2024] [Indexed: 02/25/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) are the standard of care for a growing number of malignancies. Unfortunately, they are associated with a broad range of unique toxicities that mimic the presentations of primary autoimmune conditions. These adverse events are termed immune-related adverse events (irAEs), of which ICI-lupus erythematosus (ICI-LE) constitutes a small percentage. Our review aims to describe the available literature on ICI-LE and ICI treatment for patients with pre-existing lupus. Most diagnoses of ICI-LE had findings of only cutaneous lupus; four diagnoses of ICI-LE had systemic lupus manifestations. Over 90% (27 of 29) of cases received anti-PD-1/PDL-1 monotherapy, 1 received combination therapy, and 1 received only anti-CTLA-4 treatment. About three-fourths (22 of 29 or 76%) of patients with ICI-lupus were managed with topical steroids, 13 (45%) received hydroxychloroquine, and 10 (34%) required oral corticosteroids. In our case series, none of the patients with pre-existing lupus receiving ICI therapy for cancer had a flare of their lupus, but few had de novo irAE manifestations, all of which were characterized as low-grade. The review of the literature yielded seven ICI-LE flares from a total of 27 patients with pre-existing lupus who received ICI. Most flares were manageable without need for ICI cessation.
Collapse
Affiliation(s)
| | - Arohi Singh
- College of the University of Chicago, University of Chicago, Chicago, IL 60637, USA;
| | - Pankti Reid
- Committee on Clinical Pharmacology and Pharmacogenomics, University of Chicago Medicine, Chicago, IL 60637, USA
- Department of Medicine, Section of Rheumatology, University of Chicago Medicine, Chicago, IL 60637, USA;
| | - Kimberly Trotter
- Department of Medicine, Section of Rheumatology, University of Chicago Medicine, Chicago, IL 60637, USA;
| |
Collapse
|
23
|
Sotoodeh A, Nguyen Hoang M, Hellgren K, Forss A. Prevalence of coeliac disease in patients with systemic lupus erythematosus: a systematic review and meta-analysis. Lupus Sci Med 2024; 11:e001106. [PMID: 38351098 PMCID: PMC10868192 DOI: 10.1136/lupus-2023-001106] [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: 11/17/2023] [Accepted: 02/03/2024] [Indexed: 02/16/2024]
Abstract
BACKGROUND There is some evidence of a higher prevalence of coeliac disease (CD) among patients with SLE than in the general population. However, the prevalence estimates vary substantially. OBJECTIVE To investigate the prevalence of CD among patients with SLE through systematic review and meta-analysis. METHODS We performed searches in the databases of Medline, Embase, Cochrane and Web of Science Core Collection between 1 January 1990 and 9 July 2023. A total of 2053 publications were rendered in the searches, of which 68 were reviewed in full text and 14 included in the analyses. Primary analysis estimated the pooled prevalence of biopsy-verified CD in patients with SLE. In the secondary analysis, the prevalence of serological markers indicative of CD was investigated. The quality of studies was appraised using the Joanna Briggs Institute Critical Appraisal Tool. We conducted meta-regression analyses to investigate associations between the prevalence of CD in individuals with SLE and publication year, study population size, CD prevalence in the general population, proportion of females and quality assessment score. RESULTS A total of 14 studies met the inclusion criteria, of which 11 were included in the primary analysis of biopsy-verified CD. Among 1238 patients with SLE, 14 had CD. The weighted pooled prevalence of CD was 0.7% (95% CI 0.0 to 1.8). The weighted pooled prevalence of CD serological markers in 1063 patients with SLE was 3.7% (95% CI 1.4 to 6.7). In meta-regression analyses, no associations between CD prevalence and study characteristics, demographics and quality assessment scores were found. CONCLUSIONS In this meta-analysis, we found a weighted pooled prevalence of biopsy-verified CD in patients with SLE comparable with the prevalence in the general population. Our findings do not support routine screening for CD in patients with SLE. However, individual screening could be considered in cases of clinical suspicion and additional risk factors for CD. PROSPERO REGISTRATION NUMBER CRD42022339594.
Collapse
Affiliation(s)
- Adonis Sotoodeh
- Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | | | - Karin Hellgren
- Gastroenterology Unit, Department of Gastroenterology, Dermatovenereology and Rheumatology, Karolinska University Hospital, Stockholm, Sweden
| | - Anders Forss
- Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Medicine, Solna, Division of Clinical Epidemiology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
24
|
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
|
25
|
Mehmandar-Oskuie A, Jahankhani K, Rostamlou A, Mardafkan N, Karamali N, Razavi ZS, Mardi A. Molecular mechanism of lncRNAs in pathogenesis and diagnosis of auto-immune diseases, with a special focus on lncRNA-based therapeutic approaches. Life Sci 2024; 336:122322. [PMID: 38042283 DOI: 10.1016/j.lfs.2023.122322] [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: 10/05/2023] [Revised: 11/24/2023] [Accepted: 11/29/2023] [Indexed: 12/04/2023]
Abstract
Autoimmune diseases are a diverse set of conditions defined by organ damage due to abnormal innate and acquired immune system responses. The pathophysiology of autoimmune disorders is exceedingly intricate and has yet to be fully understood. The study of long non-coding RNAs (lncRNAs), non-protein-coding RNAs with at least 200 nucleotides in length, has gained significant attention due to the completion of the human genome project and the advancement of high-throughput genomic approaches. Recent research has demonstrated how lncRNA alters disease development to different degrees. Although lncRNA research has made significant progress in cancer and generative disorders, autoimmune illnesses are a relatively new research area. Moreover, lncRNAs play crucial functions in differentiating various immune cells, and their potential relationships with autoimmune diseases have received growing attention. Because of the importance of Th17/Treg axis in auto-immune disease development, in this review, we discuss various molecular mechanisms by which lncRNAs regulate the differentiation of Th17/Treg cells. Also, we reviewed recent findings regarding the several approaches in the application of lncRNAs in the diagnosis and treatment of human autoimmune diseases, as well as current challenges in lncRNA-based therapeutic approaches to auto-immune diseases.
Collapse
Affiliation(s)
- Amirreza Mehmandar-Oskuie
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kasra Jahankhani
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arman Rostamlou
- Department of Medical Biology, Faculty of Medicine, University of EGE, Izmir, Turkey
| | - Nasibeh Mardafkan
- Department of Laboratory Science, Faculty of Paramedicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Negin Karamali
- Student Research Committee, Tabriz University of Medical Science, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Science, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Science, Tabriz, Iran
| | - Zahra Sadat Razavi
- Department of Immunology, Faculty of Medicine, Tarbiat Modares University, Tehran, Iran
| | - Amirhossein Mardi
- Student Research Committee, Tabriz University of Medical Science, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Science, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Science, Tabriz, Iran.
| |
Collapse
|
26
|
Charles N, Kortekaas-Krohn I, Kocaturk E, Scheffel J, Altrichter S, Steinert C, Xiang YK, Gutermuth J, Reber LL, Maurer M. Autoreactive IgE: Pathogenic role and therapeutic target in autoimmune diseases. Allergy 2023; 78:3118-3135. [PMID: 37555488 DOI: 10.1111/all.15843] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 07/08/2023] [Accepted: 07/27/2023] [Indexed: 08/10/2023]
Abstract
Autoimmunity is the break of tolerance to self-antigens that leads to organ-specific or systemic diseases often characterized by the presence of pathogenic autoreactive antibodies (AAb) produced by plasmablast and/or plasma cells. AAb are prevalent in the general population and not systematically associated with clinical symptoms. In contrast, in some individuals, these AAb are pathogenic and drive the development of signs and symptoms of antibody-mediated autoimmune diseases (AbAID). AAb production, isotype profiles, and glycosylations are promoted by pro-inflammatory triggers linked to genetic, environmental, and hormonal parameters. Recent evidence supports a role for pathogenic AAb of the IgE isotype in a number of AbAID. Autoreactive IgE can drive the activation of mast cells, basophils, and other types of FcεRI-bearing cells and may play a role in promoting autoantibody production and other pro-inflammatory pathways. In this review, we discuss the current knowledge on the pathogenicity of autoreactive IgE in AbAID and their status as therapeutic targets. We also highlight unresolved issues including the need for assays that reproducibly quantify IgE AAbs, to validate their diagnostic and prognostic value, and to further study their pathophysiological contributions to AbAID.
Collapse
Affiliation(s)
- Nicolas Charles
- Faculté de Médecine site Bichat, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS EMR8252, Université Paris Cité, Paris, France
- Laboratoire d'Excellence Inflamex, Université Paris Cité, Paris, France
| | - Inge Kortekaas-Krohn
- Vrije Universiteit Brussel (VUB), Skin Immunology & Immune Tolerance (SKIN) Research Group, Brussels, Belgium
- Department of Dermatology, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Emek Kocaturk
- Department of Dermatology, Koç University School of Medicine, Istanbul, Turkey
- Institute of Allergology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Allergology and Immunology, Berlin, Germany
| | - Jörg Scheffel
- Institute of Allergology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Allergology and Immunology, Berlin, Germany
| | - Sabine Altrichter
- Institute of Allergology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Allergology and Immunology, Berlin, Germany
- Departement of Dermatology and Venerology, Kepler University Hospital, Linz, Austria
| | - Carolin Steinert
- Institute of Allergology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Allergology and Immunology, Berlin, Germany
- Freie Universität Berlin, Department of Biology, Chemistry and Pharmacy, Berlin, Germany
| | - Yi-Kui Xiang
- Institute of Allergology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Allergology and Immunology, Berlin, Germany
| | - Jan Gutermuth
- Vrije Universiteit Brussel (VUB), Skin Immunology & Immune Tolerance (SKIN) Research Group, Brussels, Belgium
- Department of Dermatology, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Laurent L Reber
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), UMR 1291, University of Toulouse, INSERM, CNRS, Toulouse, France
| | - Marcus Maurer
- Institute of Allergology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Allergology and Immunology, Berlin, Germany
| |
Collapse
|
27
|
Al-Karaja L, Alshayeb FO, Amro D, Khdour YF, Alamlih L. Shrinking Lung Syndrome in a Systemic Lupus Erythematous Patient Improved by Rituximab: A Case Report With Literature Review. Cureus 2023; 15:e50229. [PMID: 38192926 PMCID: PMC10773592 DOI: 10.7759/cureus.50229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/09/2023] [Indexed: 01/10/2024] Open
Abstract
Shrinking lung syndrome (SLS) is a rare complication of autoimmune and connective tissue diseases like systemic lupus erythematosus (SLE). A 35-year-old female patient, diagnosed with SLE, came to the hospital complaining of severe dyspnea and pleuritic pain for several months that was worsening on exertion. Imaging (X-ray and CT scan) of the chest at the time of presentation showed bilateral basal atelectasis with elevated diaphragm. Pulmonary function test (PFT) showed restrictive findings including forced expiratory volume in the first second (FEV1) of 37%, total lung capacity of 40%, and vital capacity of 32% predicted with a restrictive pattern on flow volume loop confirming the diagnosis of SLS. The treatment focused on methotrexate and rituximab. Patients with a known history of SLE who start respiratory symptoms like cough and dyspnea should be ruled out of SLS at the earliest as it can be deadly in the later stages.
Collapse
Affiliation(s)
| | - Fatima O Alshayeb
- General Practice, Jordan University of Science and Technology, Amman, JOR
| | - Dana Amro
- Allergy and Immunology, Jordan University of Science and Technology, Amman, JOR
| | - Yazan F Khdour
- Rheumatology, Princess Alia Governmental Hospital, Hebron, PSE
| | | |
Collapse
|
28
|
Kielbassa K, Van der Weele L, Voskuyl AE, de Vries N, Eldering E, Kuijpers TW. Differential expression pattern of Bcl-2 family members in B and T cells in systemic lupus erythematosus and rheumatoid arthritis. Arthritis Res Ther 2023; 25:225. [PMID: 37993903 PMCID: PMC10664305 DOI: 10.1186/s13075-023-03203-7] [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: 03/13/2023] [Accepted: 10/31/2023] [Indexed: 11/24/2023] Open
Abstract
OBJECTIVE This study aimed to evaluate the expression level of anti-apoptotic Bcl-2 family proteins in B and T cells in patients with systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA) in relation to disease activity and the effect of various Bcl-2 family inhibitors (BH3 mimetics) as potential treatment. METHODS We included 14 SLE patients, 12 RA patients, and 13 healthy controls to study anti-apoptotic Bcl-2, Bcl-XL, and Mcl-1 expression and cell survival in different B and T cell subsets using stimulation assays and intracellular flow cytometry. Effect of various BH3 mimetics was assessed by cell viability analyses. RESULTS In SLE, significant differences in Bcl-2 family members were confined to the B cell compartment with decreased induction of Bcl-XL (p ≤ 0.05) and Mcl-1 (p ≤ 0.001) upon CpG stimulation. In RA, we did not observe any differences in expression levels of Bcl-2 family proteins. Expression patterns did not correlate with disease activity apart from decreased induction of Mcl-1 in B cells in active SLE. After in vitro stimulation with CpG, plasmablasts were more viable after treatment with three different BH3 mimetics compared to naïve or memory B cells in control and patient cells. After activation, Mcl-1 inhibition was most effective in reducing plasmablast and T cell viability, however, less in patients than controls. CONCLUSION Our study provides evidence for the increased differential expression pattern of Bcl-2 family members in B and T cell subsets of patients with SLE compared to controls. Tested BH3 mimetics showed higher efficacy in controls compared to both autoimmune diseases, though nonsignificant due to low patient numbers.
Collapse
Affiliation(s)
- K Kielbassa
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute (AIII), Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Department of Lymphoma and Myeloma Center Amsterdam, Amsterdam, The Netherlands
| | - L Van der Weele
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute (AIII), Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.
- Department of Rheumatology and Clinical Immunology, Amsterdam Rheumatology and Immunology Centre (ARC), Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.
| | - A E Voskuyl
- Department of Rheumatology and Clinical Immunology, Amsterdam Rheumatology and Immunology Centre (ARC), Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
| | - N de Vries
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute (AIII), Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Department of Rheumatology and Clinical Immunology, Amsterdam Rheumatology and Immunology Centre (ARC), Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - E Eldering
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute (AIII), Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Department of Lymphoma and Myeloma Center Amsterdam, Amsterdam, The Netherlands
| | - T W Kuijpers
- Department of Pediatric Immunology, Rheumatology and Infectious Diseases, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
29
|
Perdomo J, Leung HHL. Immune Thrombosis: Exploring the Significance of Immune Complexes and NETosis. BIOLOGY 2023; 12:1332. [PMID: 37887042 PMCID: PMC10604267 DOI: 10.3390/biology12101332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 09/26/2023] [Accepted: 10/11/2023] [Indexed: 10/28/2023]
Abstract
Neutrophil extracellular traps (NETs) are major contributors to inflammation and autoimmunity, playing a key role in the development of thrombotic disorders. NETs, composed of DNA, histones, and numerous other proteins serve as scaffolds for thrombus formation and promote platelet activation, coagulation, and endothelial dysfunction. Accumulating evidence indicates that NETs mediate thrombosis in autoimmune diseases, viral and bacterial infections, cancer, and cardiovascular disease. This article reviews the role and mechanisms of immune complexes in NETs formation and their contribution to the generation of a prothrombotic state. Immune complexes are formed by interactions between antigens and antibodies and can induce NETosis by the direct activation of neutrophils via Fc receptors, via platelet activation, and through endothelial inflammation. We discuss the mechanisms by which NETs induced by immune complexes contribute to immune thrombotic processes and consider the potential development of therapeutic strategies. Targeting immune complexes and NETosis hold promise for mitigating thrombotic events and reducing the burden of immune thrombosis.
Collapse
Affiliation(s)
- José Perdomo
- Haematology Research Group, Faculty Medicine and Health, Central Clinical School, University of Sydney, Sydney, NSW 2006, Australia
| | - Halina H. L. Leung
- Haematology Research Unit, St George & Sutherland Clinical Campuses, Faculty of Medicine & Health, School of Clinical Medicine, University of New South Wales, Kogarah, NSW 2217, Australia;
| |
Collapse
|
30
|
Wang T, Wei L, Meng S, Song W, Chen Y, Li H, Zhao Q, Jiang Z, Liu D, Ren H, Hong X. Coordinated Priming of NKG2D Pathway by IL-15 Enhanced Functional Properties of Cytotoxic CD4 +CD28 - T Cells Expanded in Systemic Lupus Erythematosus. Inflammation 2023; 46:1587-1601. [PMID: 37415045 PMCID: PMC10567942 DOI: 10.1007/s10753-023-01860-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 05/27/2023] [Accepted: 06/16/2023] [Indexed: 07/08/2023]
Abstract
Systemic lupus erythematosus (SLE) is a systemic autoimmune disorder, and numerous aberrations of T cell responses have been reported and were implicated in its pathophysiology. Recently, CD4-positive T cells with cytotoxic potential were shown to be involved in autoimmune disease progression and tissue damage. However, the effector functions of this cell type and their potential molecular mechanisms in SLE patients remain to be elucidated. In this study, we find that cytotoxic CD4+CD28- T cells are expanded in SLE patients with flow cytometry analysis, and the percentage of CD4+CD28- T cells positively correlates with the Systemic Lupus International Collaborating Clinics/ACR Damage Index (SDI). Furthermore, our study suggests that interleukin-15 (IL-15) promotes the expansion, proliferation, and cytotoxic function of CD4+CD28- T cells in SLE patients through activation of the Janus kinase3-STAT5 pathway. Further study indicates that IL-15 not only mediates the upregulation of NKG2D, but also cooperates with the NKG2D pathway to regulate the activation of the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) pathway. Together, our study demonstrated that proinflammatory and cytolytic CD4+CD28- T cells expand in SLE patients. The pathogenic potential of these CD4+CD28- T cells is driven by the coupling of the IL-15/IL-15R signaling pathway and the NKG2D/DAP10 signaling pathway, which may open new avenues for therapeutic intervention to prevent SLE progression.
Collapse
Affiliation(s)
- Tingting Wang
- Department of Rheumatology and Immunology, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, 518020, China
- Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, 510632, China
| | - Laiyou Wei
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, China
- Shenzhen People's Hospital, The Frist Affiliated Hospital of Southern University of Science and Technology, Shenzhen, 518020, China
| | - Shuhui Meng
- Department of Rheumatology and Immunology, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, 518020, China
| | - Wencong Song
- Department of Rheumatology and Immunology, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, 518020, China
| | - Yulan Chen
- Department of Rheumatology and Immunology, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, 518020, China
| | - Heng Li
- Department of Rheumatology and Immunology, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, 518020, China
- Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, 510632, China
| | - Qianqian Zhao
- Department of Rheumatology and Immunology, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, 518020, China
- Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, 510632, China
| | - Zhenyou Jiang
- Department of Microbiology and Immunology, College of Basic Medicine and Public Hygiene, Jinan University, Guangzhou, 510632, China
| | - Dongzhou Liu
- Department of Rheumatology and Immunology, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, 518020, China
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, China
- Shenzhen People's Hospital, The Frist Affiliated Hospital of Southern University of Science and Technology, Shenzhen, 518020, China
| | - Huan Ren
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, China.
| | - Xiaoping Hong
- Department of Rheumatology and Immunology, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, 518020, China.
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, China.
- Shenzhen People's Hospital, The Frist Affiliated Hospital of Southern University of Science and Technology, Shenzhen, 518020, China.
| |
Collapse
|
31
|
García-González M, Gómez-Bernal F, Quevedo-Abeledo JC, Fernández-Cladera Y, González-Rivero AF, López-Mejías R, Díaz-González F, González-Gay MÁ, Ferraz-Amaro I. HDL Cholesterol Efflux and the Complement System Are Linked in Systemic Lupus Erythematosus. J Clin Med 2023; 12:5405. [PMID: 37629447 PMCID: PMC10455830 DOI: 10.3390/jcm12165405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 08/07/2023] [Accepted: 08/17/2023] [Indexed: 08/27/2023] Open
Abstract
Cholesterol efflux capacity (CEC), the ability of high-density lipoprotein (HDL) cholesterol to accept cholesterol from macrophages, has been linked to cardiovascular events. Systemic lupus erythematosus (SLE) is characterized by the consumption of complement (C) proteins and has been associated with an increased risk of cardiovascular disease. CEC is reduced in SLE patients compared to controls. In the present work, our objective was to analyze whether the disruption of C influences CEC in patients with SLE. New-generation functional assays of the three pathways of the C system were performed in 207 patients with SLE. Additionally, serum levels of inactive (C1q, C2, C3, C4, and factor D) and activated (C3a) molecules, and regulators (C1-inhibitor and factor H) of C system were measured. CEC, using an in vitro assay, and lipoprotein serum concentrations were assessed. Multivariable linear regression analysis was performed to assess the relationship between C system and CEC. After full multivariable analysis, the alternative C cascade functional test showed a significant and negative relationship with CEC. This was also the case for C2 and C3, in which the associations were found to be positive and statistically significant, after adjustment for covariates. In conclusion, C system and CEC are interconnected in patients with SLE.
Collapse
Affiliation(s)
- María García-González
- Division of Rheumatology, Hospital Universitario de Canarias, 38320 Tenerife, Spain; (M.G.-G.); (F.D.-G.)
| | - Fuensanta Gómez-Bernal
- Division of Central Laboratory, Hospital Universitario de Canarias, 38320 Tenerife, Spain; (F.G.-B.); (Y.F.-C.); (A.F.G.-R.)
| | | | - Yolanda Fernández-Cladera
- Division of Central Laboratory, Hospital Universitario de Canarias, 38320 Tenerife, Spain; (F.G.-B.); (Y.F.-C.); (A.F.G.-R.)
| | - Agustín F. González-Rivero
- Division of Central Laboratory, Hospital Universitario de Canarias, 38320 Tenerife, Spain; (F.G.-B.); (Y.F.-C.); (A.F.G.-R.)
| | - Raquel López-Mejías
- Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Instituto de Investigación sanitaria Marqués de Valdecilla (IDIVAL), 39011 Santander, Spain;
| | - Federico Díaz-González
- Division of Rheumatology, Hospital Universitario de Canarias, 38320 Tenerife, Spain; (M.G.-G.); (F.D.-G.)
- Department of Internal Medicine, University of La Laguna (ULL), 38200 Tenerife, Spain
| | - Miguel Á. González-Gay
- Division of Rheumatology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain
- Department of Medicine and Psychiatry, University of Cantabria, 39005 Santander, Spain
| | - Iván Ferraz-Amaro
- Division of Rheumatology, Hospital Universitario de Canarias, 38320 Tenerife, Spain; (M.G.-G.); (F.D.-G.)
- Department of Internal Medicine, University of La Laguna (ULL), 38200 Tenerife, Spain
| |
Collapse
|
32
|
Lodde V, Floris M, Zoroddu E, Zarbo IR, Idda ML. RNA-binding proteins in autoimmunity: From genetics to molecular biology. WILEY INTERDISCIPLINARY REVIEWS. RNA 2023; 14:e1772. [PMID: 36658783 DOI: 10.1002/wrna.1772] [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: 06/10/2022] [Revised: 11/18/2022] [Accepted: 12/05/2022] [Indexed: 01/21/2023]
Abstract
Autoimmune diseases (ADs) are chronic pathologies generated by the loss of immune tolerance to the body's own cells and tissues. There is growing recognition that RNA-binding proteins (RBPs) critically govern immunity in healthy and pathological conditions by modulating gene expression post-transcriptionally at all levels: nuclear mRNA splicing and modification, export to the cytoplasm, as well as cytoplasmic mRNA transport, storage, editing, stability, and translation. Despite enormous efforts to identify new therapies for ADs, definitive solutions are not yet available in many instances. Recognizing that many ADs have a strong genetic component, we have explored connections between the molecular biology and the genetics of RBPs in ADs. Here, we review the genetics and molecular biology of RBPs in four major ADs, multiple sclerosis (MS), type 1 diabetes mellitus (T1D), systemic lupus erythematosus (SLE), and rheumatoid arthritis (RA). We anticipate that gaining insights into the genetics and biology of ADs can facilitate the discovery of new therapies. This article is categorized under: RNA in Disease and Development > RNA in Disease.
Collapse
Affiliation(s)
- Valeria Lodde
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Matteo Floris
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Enrico Zoroddu
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Ignazio Roberto Zarbo
- Department of Medical, Surgical and Experimental Sciences, University of Sassari - Neurology Unit Azienza Ospedaliera Universitaria (AOU), Sassari, Italy
| | - Maria Laura Idda
- Institute for Genetic and Biomedical Research - National Research Council (IRGB-CNR), Sassari, Italy
| |
Collapse
|
33
|
Gong M, Dai L, Xie Z, Hong D, Li N, Fan X, Xie C. Serological and clinical associations of autoantibodies in Chinese patients with new-onset systemic lupus erythematosus. Sci Rep 2023; 13:10101. [PMID: 37344560 DOI: 10.1038/s41598-023-37100-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 06/15/2023] [Indexed: 06/23/2023] Open
Abstract
To study the clinical significance of autoantibodies in Chinese patients with new-onset systemic lupus erythematosus (SLE), we enrolled 526 new-onset patients who met the 1997 Updated American College of Rheumatology SLE Classification Criteria for a retrospective cohort study. Chi-square test and Wilcoxon rank-sum test were used to detect the relationship of autoantibodies with clinical manifestations and serological results respectively. Our results demonstrated that the positive rate of anti-ribosomal P protein (anti-P) antibody in female patients was higher than that in male patients (41.2% vs. 22%, P = 0.008). Patients with anti-SSB (43.95 ± 73.12 vs. 40.92 ± 75.75, P = 0.004; 63.93 ± 103.56 vs. 55.06 ± 120.84, P = 0.008 respectively) antibodies had higher levels of alanine aminotransferase (ALT) and aspartate transaminase (AST), whereas those with anti-P antibody (28.90 ± 25.70 vs. 50.08 ± 93.00, P = 0.014; 38.51 ± 48.19 vs. 69.95 ± 142.67, P = 0.047, respectively) had lower levels of them. Anti-dsDNA antibody (P = 0.021) was associated with pulmonary arterial hypertension (PAH). The patients with anti-Ro60 (P = 0.044), anti-P (P = 0.012) and anti-dsDNA (P = 0.013) antibodies were less likely to develop Interstitial lung disease. Anti-SmRNP antibody was correlated to lower prevalence of neuropsychiatric symptoms (P = 0.037), and patients with anti-centromere antibody (ACA) were more likely to develop serositis (P = 0.016).We identified five clusters of SLE-related autoantibodies, confirmed previously reported associations of autoantibodies, and discovered new associations.
Collapse
Affiliation(s)
- Muxue Gong
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, China
| | - Li Dai
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, China
| | - Zhuobei Xie
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, China
| | - Dengxiao Hong
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, China
| | - Ning Li
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, China
| | - Xiaoyun Fan
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, China
| | - Changhao Xie
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, China.
- Anhui Provincial Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Bengbu, 233003, China.
| |
Collapse
|
34
|
Choi D, Kim J, Yang JW, Kim JH, Park S, Shin JI. Dysregulated MicroRNAs in the Pathogenesis of Systemic Lupus Erythematosus: A Comprehensive Review. Int J Biol Sci 2023; 19:2495-2514. [PMID: 37215992 PMCID: PMC10197884 DOI: 10.7150/ijbs.74315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 12/11/2022] [Indexed: 05/24/2023] Open
Abstract
Systemic lupus erythematosus is a chronic autoimmune disease of which clinical presentation is vastly heterogeneous, ranging from mild skin rashes to severe renal diseases. Treatment goal of this illness is to minimize disease activity and prevent further organ damage. In recent years, much research has been done on the epigenetic aspects of SLE pathogenesis, for among the various factors known to contribute to the pathogenic process, epigenetic factors, especially microRNAs, bear the most therapeutic potential that can be altered unlike congenital genetic factors. This article reviews and updates what has been discovered so far about the pathogenesis of lupus, while focusing on the dysregulation of microRNAs in lupus patients in comparison to healthy controls along with the potentially pathogenic roles of the microRNAs commonly reported to be either upregulated or downregulated. Furthermore, this review includes microRNAs of which results are controversial, suggesting possible explanations for such discrepancies and directions for future research. Moreover, we aimed to emphasize the point that had been overlooked so far in studies regarding microRNA expression levels; that is, which specimen was used to assess the dysregulation of microRNAs. To our surprise, a vast number of studies have not considered this factor and have analyzed the potential role of microRNAs in general. Despite extensive investigations done on microRNA levels, their significance and potential role remain a mystery, which calls for further studies on this particular subject in regard of which specimen is used for assessment.
Collapse
Affiliation(s)
- Daeun Choi
- Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jimin Kim
- Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jae Won Yang
- Department of Nephrology, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Ji Hong Kim
- Department of Pediatrics, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seoyeon Park
- Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jae Il Shin
- Department of Pediatrics, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
35
|
Abozaid HSM, Hefny HM, Abualfadl EM, Ismail MA, Noreldin AK, Eldin ANN, Goda AM, Ali AH. Negative ANA-IIF in SLE patients: what is beyond? Clin Rheumatol 2023:10.1007/s10067-023-06577-w. [PMID: 37016193 DOI: 10.1007/s10067-023-06577-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 03/07/2023] [Accepted: 03/09/2023] [Indexed: 04/06/2023]
Abstract
The antinuclear antibody (ANA) test has high sensitivity in diagnosing and classifying systemic lupus erythematosus (SLE). OBJECTIVES To describe the immunological pattern of SLE patients through investigating specific antinuclear autoantibodies by enzyme dot immunoassay and studying their frequency in both positive and negative ANA indirect immunofluorescence assay (IIF) cases. METHODS In a cross-sectional study, blood samples from 393 newly diagnosed SLE patients were analyzed using (IIF) on HEp-2 cells and ANA dot immunoassay by automated enzyme immunoassay (EIA) to detect 19 antibodies. RESULTS Ninety-one percent of the patients are females; their mean age was 37 ± 12.28. Antinuclear antibody (ANA) was detected by IIF in 82.4% of cases, with 181 (46.1%) speckled and 167 (42.4%) homogeneous ANA patterns. The majority of patients (96%) demonstrated autoantibodies via EIA. Among the ANA-IIF-negative patients, 97.2% demonstrated autoantibodies. There was a significant difference in the frequency of certain autoantibodies between SLE patients with negative and positive ANA-IIF (1.44 0.73, 3.12 2.09, p = 0.00) respectively. CONCLUSION The results of analyzing 19 autoantibodies with the ANA staining pattern increased the significance of analyzing the immune profile even if IIF is negative when clinical symptoms strongly suggest SLE diagnosis. Certain autoantibodies may evade staining by the IFA approach while they are present in the patient's serum, and they may not be detected by the ANA EIA profile if it does not contain that antigenic substrate. Key Points • Indirect immunofluorescence on Hep-2 is the conventional method for ANA detection and is regarded as the "gold standard" for testing in clinical practice for SLE. • In our study, ANA profile dot enzyme immunoassay (EIA)-based test was performed to evaluate 19 autoantibodies in SLE patients either positive or negative for ANA-IIF. • The presence of anti-dsDNA with ANA-IIF-negative serum in 32.4% of SLE patients provides evidence that not all anti-dsDNA antibodies are identified on standard HEp-2 substrates. • certain autoantibodies can evade staining by the ANA-IIF method despite being present in the SLE patient's blood; this supports the ANA profile enzyme dot immunoassay as a more sensitive test.
Collapse
Affiliation(s)
- Hanan Sayed M Abozaid
- Rheumatology & Rehabilitation Department, Sohag Faculty of Medicine, Sohag University Hospital, Sohag, 82524, Egypt.
| | - Hesham M Hefny
- Clinical Pathology Department, Sohag University Hospital, Sohag, Egypt
| | - Esam M Abualfadl
- Rheumatology & Rehabilitation Department, Sohag Faculty of Medicine, Sohag University Hospital, Sohag, 82524, Egypt
| | - Mohamad A Ismail
- Rheumatology & Rehabilitation Department, Sohag Faculty of Medicine, Sohag University Hospital, Sohag, 82524, Egypt
| | - Amal K Noreldin
- Internal Medicine Department, Sohag University Hospital, Sohag, Egypt
| | | | - Asmaa M Goda
- Medical Microbiology and Immunology, Faculty of Medicine, Sohag University, Sohag, Egypt
| | - Amal H Ali
- Medical Microbiology and Immunology, Faculty of Medicine, Aswan University, Aswan, Egypt
| |
Collapse
|
36
|
Joy GM, Arbiv OA, Wong CK, Lok SD, Adderley NA, Dobosz KM, Johannson KA, Ryerson CJ. Prevalence, imaging patterns and risk factors of interstitial lung disease in connective tissue disease: a systematic review and meta-analysis. Eur Respir Rev 2023; 32:32/167/220210. [PMID: 36889782 PMCID: PMC10032591 DOI: 10.1183/16000617.0210-2022] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 01/09/2023] [Indexed: 03/10/2023] Open
Abstract
INTRODUCTION Interstitial lung disease (ILD) is a frequent manifestation of connective tissue disease (CTD) with substantial variability in prevalence and outcomes reported across CTD subtypes. This systematic review summarises the prevalence, risk factors and ILD patterns on chest computed tomography of CTD-ILD. METHODS A comprehensive search was performed in Medline and Embase to identify eligible studies. Meta-analyses were completed using a random effects model to determine the pooled prevalence of CTD-ILD and ILD patterns. RESULTS 11 582 unique citations were identified with 237 articles included. Pooled prevalence of ILD was 11% in rheumatoid arthritis (95% CI 7-15%), 47% in systemic sclerosis (44-50%), 41% in idiopathic inflammatory myositis (33-50%), 17% in primary Sjögren's syndrome (12-21%), 56% in mixed connective tissue disease (39-72%) and 6% in systemic lupus erythematosus (3-10%). Usual interstitial pneumonia was the most prevalent ILD pattern in rheumatoid arthritis (pooled prevalence of 46%), while nonspecific interstitial pneumonia was the most common ILD pattern in all other CTD subtypes (pooled prevalence range 27-76%). Across all CTDs with available data, positive serology and higher inflammatory markers were risk factors for development of ILD. DISCUSSION We identified substantial variability in ILD across CTD subtypes suggesting that CTD-ILD is too heterogenous to be considered a single entity.
Collapse
Affiliation(s)
- Greta M Joy
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Omri A Arbiv
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Carmen K Wong
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Stacey D Lok
- Department of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | | | - Krzysztof M Dobosz
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | | | - Christopher J Ryerson
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
37
|
Chang SH, Choi Y. Gut dysbiosis in autoimmune diseases: Association with mortality. Front Cell Infect Microbiol 2023; 13:1157918. [PMID: 37065187 PMCID: PMC10102475 DOI: 10.3389/fcimb.2023.1157918] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 03/15/2023] [Indexed: 04/03/2023] Open
Abstract
To better understand the impact of gut dysbiosis on four autoimmune diseases [Sjögren’s syndrome (SS), systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), and multiple sclerosis (MS)], this review investigated the altered gut bacteria in each disease and the shared ones among the four diseases. The enriched gut bacteria shared by three of the four autoimmune diseases were Streptococcus, Prevotella, and Eggerthella, which are associated with autoantibody production or activation of Th17 cells in immune-related diseases. On the other hand, Faecalibacterium comprises depleted gut bacteria shared by patients with SLE, MS, and SS, which is associated with various anti-inflammatory activities. The indexes of gut dysbiosis, defined as the number of altered gut bacterial taxa divided by the number of studies in SLE, MS, RA, and SS, were 1.7, 1.8, 0.7, and 1.3, respectively. Interestingly, these values presented a positive correlation trend with the standardized mortality rates —2.66, 2.89, 1.54, and 1.41, respectively. In addition, shared altered gut bacteria among the autoimmune diseases may correlate with the prevalence of polyautoimmunity in patients with SLE, SS, RA, and MS, that is, 41 percent, 32.6 percent, 14 percent, and 1–16.6 percent, respectively. Overall, this review suggests that gut dysbiosis in autoimmune diseases may be closely related to the failure of the gut immune system to maintain homeostasis.
Collapse
|
38
|
Demers-Mathieu V. Optimal Selection of IFN-α-Inducible Genes to Determine Type I Interferon Signature Improves the Diagnosis of Systemic Lupus Erythematosus. Biomedicines 2023; 11:biomedicines11030864. [PMID: 36979843 PMCID: PMC10045398 DOI: 10.3390/biomedicines11030864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 02/24/2023] [Accepted: 03/09/2023] [Indexed: 03/14/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease characterized by the production of autoantibodies specific to self-molecules in the nucleus, cytoplasm, and cell surface. The diversity of serologic and clinical manifestations observed in SLE patients challenges the development of diagnostics and tools for monitoring disease activity. Elevated type I interferon signature (IFN- I) in SLE leads to dysregulation of innate and adaptive immune function, resulting in autoantibodies production. The most common method to determine IFN-I signature is measuring the gene expression of several IFN-α-inducible genes (IFIGs) in blood samples and calculating a score. Optimal selection of IFIGs improves the sensitivity, specificity, and accuracy of the diagnosis of SLE. We describe the mechanisms of the immunopathogenesis of IFN-I signature (IFNα production) and its clinical consequences in SLE. In addition, we explore the association between IFN-I signature, the presence of autoantibodies, disease activity, medical therapy, and ethnicity. We discuss the presence of IFN-I signature in some patients with other autoimmune diseases, including rheumatoid arthritis, systemic and multiple sclerosis, Sjogren’s syndrome, and dermatomyositis. Prospective studies are required to assess the role of IFIG and the best combination of IFIGs to monitor SLE disease activity and drug treatments.
Collapse
|
39
|
Saleh BH, Lugaajju A, Storry JR, Persson KEM. Autoantibodies against red blood cell antigens are common in a malaria endemic area. Microbes Infect 2023; 25:105060. [PMID: 36270601 DOI: 10.1016/j.micinf.2022.105060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 10/12/2022] [Accepted: 10/13/2022] [Indexed: 11/06/2022]
Abstract
Plasmodium falciparum malaria can cause severe anemia. Even after treatment, hematocrit can decrease. The role of autoantibodies against erythrocytes is not clearly elucidated and how common they are, or what they are directed against, is still largely unknown. We have investigated antibodies against erythrocytes in healthy adult men living in a highly malaria endemic area in Uganda. We found antibodies in more than half of the individuals, which is significantly more than in a non-endemic area (Sweden). Some of the Ugandan samples had a broad reactivity where it was not possible to determine the exact target of the autoantibodies, but we also found specific antibodies directed against erythrocyte surface antigens known to be of importance for merozoite invasion such as glycophorin A (anti-Ena, anti-M) and glycophorin B (anti-U, anti-S). In addition, several autoantibodies had partial specificities against glycophorin C and the blood group systems Rh, Diego (located on Band 3), Duffy (located on ACKR1), and Cromer (located on CD55), all of which have been described to be important for malaria and therefore of interest for understanding how autoantibodies could potentially stop parasites from entering the erythrocyte. In conclusion, specific autoantibodies against erythrocytes are common in a malaria endemic area.
Collapse
Affiliation(s)
- Bandar Hasan Saleh
- Department of Laboratory Medicine, Lund University, Skåne University Hospital Lund, Klinikgatan 19, 22185 Lund, Sweden; Faculty of Medicine, Department of Medical Microbiology and Parasitology, King Abdulaziz University, Building 7, 21589 Jeddah, Saudi Arabia
| | - Allan Lugaajju
- Department of Laboratory Medicine, Lund University, Skåne University Hospital Lund, Klinikgatan 19, 22185 Lund, Sweden; School of Biomedical Sciences, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Jill R Storry
- Division of Hematology and Transfusion Medicine, Department of Laboratory Medicine, Lund University, Klinikgatan 26, Lund, Sweden; Clinical Immunology and Transfusion Medicine, Laboratory Medicine, Office for Medical Services, Region Skåne, Akutgatan 8, Lund, Sweden
| | - Kristina E M Persson
- Department of Laboratory Medicine, Lund University, Skåne University Hospital Lund, Klinikgatan 19, 22185 Lund, Sweden.
| |
Collapse
|
40
|
Hurst C, Soto M, Vina ER, Rodgers KE. Renin-Angiotensin System-Modifying Antihypertensive Drugs Can Reduce the Risk of Cardiovascular Complications in Lupus: A Retrospective Cohort Study. Am J Med 2023; 136:284-293.e4. [PMID: 36495935 PMCID: PMC9957968 DOI: 10.1016/j.amjmed.2022.11.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 11/11/2022] [Accepted: 11/14/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND Patients with systemic lupus erythematosus have a higher incidence of cardiovascular disease than the general population. Antihypertensive drugs that modify the renin-angiotensin system (RAS) are used to protect renal function in lupus nephritis and may also have extrarenal effects that lower cardiovascular disease risk due to their anti-inflammatory properties. In this study, we compared the effects of RAS vs non-RAS antihypertensive drugs on cardiovascular disease incidence in patients with lupus. METHODS Using a medical insurance claims dataset, 220,168 patients with lupus were identified, of which 31,647 patients (4018 patients prescribed RAS drugs, 27,629 patients prescribed non-RAS drugs) were eligible for the study. Patients had a mean age of 46.1 years, were 93.0% female, and healthy (96.9% Charlson Comorbidity Index score 0-4). Patients in the 2 drug groups were propensity score matched using demographic data, risk factors, and comorbidities. RESULTS Use of RAS vs non-RAS drugs lowered the relative risk (RR) of diagnosis of cardiovascular disease (RR 0.80; 95% confidence interval [CI], 0.74-0.87), which was more pronounced after propensity score matching (RR 0.62; 95% CI, 0.57-0.68). The decreased risk in cardiovascular disease occurred regardless of lupus nephritis status (with lupus nephritis: RR 0.51; 95% CI, 0.39-0.65; without lupus nephritis: RR 0.65; 95% CI, 0.59-0.72). RAS-modifying therapies significantly increased cardiovascular disease-free survival probability over a 5-year period (86.0% vs 78.3% probability). CONCLUSIONS RAS-modifying drugs reduced the risk of cardiovascular disease in patients with systemic lupus erythematosus in this dataset. These findings have the potential to impact clinical decision-making with regards to hypertension management in patients with lupus.
Collapse
Affiliation(s)
- Chelsie Hurst
- Department of Pharmacology, Center for Innovation in Brain Science, College of Medicine, University of Arizona, Tucson
| | - Maira Soto
- Department of Pharmacology, Center for Innovation in Brain Science, College of Medicine, University of Arizona, Tucson
| | - Ernest R Vina
- Section of Rheumatology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pa
| | - Kathleen E Rodgers
- Department of Pharmacology, Center for Innovation in Brain Science, College of Medicine, University of Arizona, Tucson.
| |
Collapse
|
41
|
Olewicz-Gawlik A, Kowala-Piaskowska A. Self-reactive IgE and anti-IgE therapy in autoimmune diseases. Front Pharmacol 2023; 14:1112917. [PMID: 36755957 PMCID: PMC9899859 DOI: 10.3389/fphar.2023.1112917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 01/06/2023] [Indexed: 01/24/2023] Open
Abstract
Growing evidence indicates the pathogenic role of autoreactive IgE in autoimmune diseases. Incidence of autoimmune and allergic diseases in the industrialized countries is consistently icreasing, thus leading to concerted efforts to comprehend the regulation of IgE-mediated mechanisms. The first reports of a presence of IgE autoantibodies in patients with autoimmune diseases have been published a long time ago, and it is now recognized that self-reactive IgE can mediate inflammatory response in bullous pemhigoid, systemic lupus erythematosus, chronic urticaria, and atopic dermatitis. The advances in understanding the pathomechanisms of these disorders brought to a successful use of anti-IgE strategies in their management. The present review discusses the current state of knowledge on the IgE-mediated autoimmunity and anti-IgE treatment, and pave the way for further exploration of the subject.
Collapse
Affiliation(s)
- Anna Olewicz-Gawlik
- Department of Immunology, Poznan University of Medical Sciences, Poznan, Poland,Department of Infectious Diseases, Hepatology and Acquired Immunodeficiencies, Poznan University of Medical Sciences, Poznan, Poland,*Correspondence: Anna Olewicz-Gawlik,
| | - Arleta Kowala-Piaskowska
- Department of Infectious Diseases, Hepatology and Acquired Immunodeficiencies, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
42
|
Korn MA, Steffensen M, Brandl C, Royzman D, Daniel C, Winkler TH, Nitschke L. Epistatic effects of Siglec-G and DNase1 or DNase1l3 deficiencies in the development of systemic lupus erythematosus. Front Immunol 2023; 14:1095830. [PMID: 36969253 PMCID: PMC10030676 DOI: 10.3389/fimmu.2023.1095830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 02/20/2023] [Indexed: 03/29/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is a severe autoimmune disease that displays considerable heterogeneity not only in its symptoms, but also in its environmental and genetic causes. Studies in SLE patients have revealed that many genetic variants contribute to disease development. However, often its etiology remains unknown. Existing efforts to determine this etiology have focused on SLE in mouse models revealing not only that mutations in specific genes lead to SLE development, but also that epistatic effects of several gene mutations significantly amplify disease manifestation. Genome-wide association studies for SLE have identified loci involved in the two biological processes of immune complex clearance and lymphocyte signaling. Deficiency in an inhibitory receptor expressed on B lymphocytes, Siglec-G, has been shown to trigger SLE development in aging mice, as have mutations in DNA degrading DNase1 and DNase1l3, that are involved in clearance of DNA-containing immune complexes. Here, we analyze the development of SLE-like symptoms in mice deficient in either Siglecg and DNase1 or Siglecg and DNase1l3 to evaluate potential epistatic effects of these genes. We found that germinal center B cells and follicular helper T cells were increased in aging Siglecg -/- x Dnase1 -/- mice. In contrast, anti-dsDNA antibodies and anti-nuclear antibodies were strongly increased in aging Siglecg-/- x Dnase1l3-/- mice, when compared to single-deficient mice. Histological analysis of the kidneys revealed glomerulonephritis in both Siglecg -/- x Dnase1 -/- and Siglecg-/- x Dnase1l3-/- mice, but with a stronger glomerular damage in the latter. Collectively, these findings underscore the impact of the epistatic effects of Siglecg with DNase1 and Dnase1l3 on disease manifestation and highlight the potential combinatory effects of other gene mutations in SLE.
Collapse
Affiliation(s)
- Marina A. Korn
- Division of Genetics, Department of Biology, University of Erlangen, Erlangen, Germany
| | - Marie Steffensen
- Division of Genetics, Department of Biology, University of Erlangen, Erlangen, Germany
| | - Carolin Brandl
- Division of Genetics, Department of Biology, University of Erlangen, Erlangen, Germany
| | - Dmytro Royzman
- Division of Genetics, Department of Biology, University of Erlangen, Erlangen, Germany
- Department of Immune Modulation, University Hospital of Erlangen, Erlangen, Germany
| | - Christoph Daniel
- Department of Nephropathology, University Hospital of Erlangen, Erlangen, Germany
| | - Thomas H. Winkler
- Division of Genetics, Department of Biology, University of Erlangen, Erlangen, Germany
| | - Lars Nitschke
- Division of Genetics, Department of Biology, University of Erlangen, Erlangen, Germany
- *Correspondence: Lars Nitschke,
| |
Collapse
|
43
|
Anis S, Fatima A, Abdul Jabbar S, Arain T. ANA-specific antibodies, ANA patterns, anti-ds-DNA results, and clinical diagnosis: a laboratory and clinical audit. Immunol Res 2022; 71:267-275. [PMID: 36456720 DOI: 10.1007/s12026-022-09347-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 11/23/2022] [Indexed: 12/03/2022]
Abstract
The diagnosis of systemic autoimmune diseases (SAID) is challenging, due to overlapping features with other non-immune disorders. Anti-nuclear antibodies (ANA)/anti-cellular antibodies are the sensitive screening tests but anti-double-stranded-deoxyribonucleic acid-antibody (anti-ds-DNA) and ANA-specific antibodies are specific for SAID. We aimed to look at ANA-specific antibodies in our patients and correlated them with ANA patterns, anti-ds-DNA, and clinical diagnosis for proper interpretation and better patient management cost-effectively. A retrospective data analysis of 641 patients was done (1st of February 2019 to 31st of July 2021) who were tested for ANA-specific antibodies at the Immunology Department of Indus Hospital and Health Network. ANA and anti-ds-DNA results and clinical diagnosis were also analyzed for ANA-specific antibody-positive patients. Descriptive data were presented in mean ± standard deviation and frequency percentages whereas inferential data were analyzed with a chi-square test for association between ANA-specific antibodies status, ANA, anti-ds-DNA, and clinical features. ANA-specific antibodies test revealed positivity for at least one autoantibody in 245 (38.2%) patients. Of these, ANA was tested in 206 patients reactive for ANA-specific antibodies and found positive in 195 (95%) as compared to negative (< 0.001). Speckled and homogenous were predominant ANA patterns in ANA-specific antibody-positives (56% and 42% respectively). Multiple ANA patterns were found in 18 patients most commonly with systemic lupus erythematosus (SLE) and mixed connective tissue disorder (MCTD). Anti-SSA were the most common ANA-specific antibodies (50%) and were mostly found in sera with speckled (61/97) and homogenous (38/97) patterns and associated mostly with SLE (48%) and Sjogren's syndrome (86%). Among ANA-negative patients, anti-SSA were the most common antibodies (n = 5). Anti-ds-DNA was found in 66% of SLE patients along with another ANA-specific antibody. This study showed that testing for ANA-specific antibodies cannot be gated on ANA patterns. Also, there is a redundancy of these antibodies with various clinical diagnoses. Moreover, they are useful in making a diagnosis in ANA-negative patients as well with clinical suspicion.
Collapse
Affiliation(s)
- Sabiha Anis
- Department of Pathology and Department of Medicine and Allied, The Indus Hospital and Health Network (IHHN), Karachi, Pakistan.
| | - Areej Fatima
- Department of Pathology, The Indus Hospital and Health Network (IHHN), Karachi, Pakistan
| | - Sidra Abdul Jabbar
- Department of Pathology, The Indus Hospital and Health Network (IHHN), Karachi, Pakistan
| | - Tayyab Arain
- Department of Pathology, The Indus Hospital and Health Network (IHHN), Karachi, Pakistan
| |
Collapse
|
44
|
Dodd KC, Menon M. Sex bias in lymphocytes: Implications for autoimmune diseases. Front Immunol 2022; 13:945762. [PMID: 36505451 PMCID: PMC9730535 DOI: 10.3389/fimmu.2022.945762] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 11/03/2022] [Indexed: 11/25/2022] Open
Abstract
Autoimmune diseases are characterized by a significant sex dimorphism, with women showing increased susceptibility to disease. This is, at least in part, due to sex-dependent differences in the immune system that are influenced by the complex interplay between sex hormones and sex chromosomes, with contribution from sociological factors, diet and gut microbiota. Sex differences are evident in the number and function of lymphocyte populations. Women mount a stronger pro-inflammatory response than males, with increased lymphocyte proliferation, activation and pro-inflammatory cytokine production, whereas men display expanded regulatory cell subsets. Ageing alters the immune landscape of men and women in differing ways, resulting in changes in autoimmune disease susceptibility. Here we review the current literature on sex differences in lymphocyte function, the factors that influence this, and the implications for autoimmune disease. We propose that improved understanding of sex bias in lymphocyte function can provide sex-specific tailoring of treatment strategies for better management of autoimmune diseases.
Collapse
Affiliation(s)
- Katherine C. Dodd
- Lydia Becker Institute of Immunology and Inflammation, Division of Immunology, Immunity to Infection and Respiratory Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom,Manchester Centre for Clinical Neurosciences, Salford Royal Hospital, Salford, United Kingdom
| | - Madhvi Menon
- Lydia Becker Institute of Immunology and Inflammation, Division of Immunology, Immunity to Infection and Respiratory Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom,*Correspondence: Madhvi Menon,
| |
Collapse
|
45
|
Xu L, Li Y, Ji J, Lai Y, Chen J, Ding T, Li H, Ding B, Ge W. The anti-inflammatory effects of Hedyotis diffusa Willd on SLE with STAT3 as a key target. JOURNAL OF ETHNOPHARMACOLOGY 2022; 298:115597. [PMID: 35940466 DOI: 10.1016/j.jep.2022.115597] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 07/26/2022] [Accepted: 07/29/2022] [Indexed: 06/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Hedyotis diffusa Willd, also named Scleromitrion diffusum (Willd.) R.J. Wang, is one medical herb, which has been traditionally used by the She nationality in China. And H. diffusa represents a beneficial effect on Systemic lupus erythematosus (SLE) treatment in clinic. AIM OF THE STUDY The underlying mechanisms of the protective effects of H. diffusa on SLE remain unclear. In this study, we treated MRL/lpr mice with H. diffusa water extract (HDW) to assess its therapeutic effects and verified its regulating signalling pathway through cytological experiments. MATERIALS AND METHODS In the present study, the constituents of HDW were analysed through ultra-performance liquid chromatography/quadrupole time-of-flight mass spectrometry (UPLC-Q-TOF-MS) and SCIEX OS software. The protective activity and underlying mechanisms were studied in a MRL/lpr lupus mouse model. The blood cells, autoantibodies, metabolites and the cytokines in serum were identified with a hematology analyzer, specific ELISA kit, GC/MS system and cytometric assays. The histological and immunohistochemical analysis were engaged in the morphologic, and the expression and translocation of the crucial protein observation. The dual luciferase reporter assay was applied to identifying the regulative activity of HDW. The transcription and translation expression of the protein was studied by real-time PCR and Western blot assays. The network pharmacology analysis was employed to predict the IL-6/STAT3 pathway regulators and the screen the STAT3 inhibitors in HDW. RESULTS The results revealed the capability of HDW to attenuate the production of autoantibodies, secretion of inflammatory cytokines (IL-6 and IFN-γ), and suppressed the IgG and C3 deposition, the development of glomerular lesions in MRL/lpr mice. Serum metabolomics study showed the improvement in serum metabolites, especially aminoacyl-tRNA biosynthesis, by HDW. IL-6 was clarified to be highly associated with the significantly changed metabolites in network analysis. We further demonstrated the effects of HDW on the IL-6/STAT3 pathway in vivo and in vitro. CONCLUSIONS This study suggested that HDW exerts a therapeutic effect in SLE model mice by suppressing the IL-6/STAT3 pathway.
Collapse
Affiliation(s)
- Li Xu
- College of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, PR China.
| | - Ying Li
- College of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, PR China.
| | - Jinjun Ji
- College of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, PR China.
| | - Yahui Lai
- College of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, PR China.
| | - Jing Chen
- College of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, PR China.
| | - Tao Ding
- College of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, PR China.
| | - Haichang Li
- College of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, PR China.
| | - Bin Ding
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, PR China.
| | - Weihong Ge
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, PR China.
| |
Collapse
|
46
|
Chen H, Huang L, Jiang X, Wang Y, Bian Y, Ma S, Liu X. Establishment and analysis of a disease risk prediction model for the systemic lupus erythematosus with random forest. Front Immunol 2022; 13:1025688. [PMID: 36405750 PMCID: PMC9667742 DOI: 10.3389/fimmu.2022.1025688] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 10/17/2022] [Indexed: 09/25/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is a latent, insidious autoimmune disease, and with the development of gene sequencing in recent years, our study aims to develop a gene-based predictive model to explore the identification of SLE at the genetic level. First, gene expression datasets of SLE whole blood samples were collected from the Gene Expression Omnibus (GEO) database. After the datasets were merged, they were divided into training and validation datasets in the ratio of 7:3, where the SLE samples and healthy samples of the training dataset were 334 and 71, respectively, and the SLE samples and healthy samples of the validation dataset were 143 and 30, respectively. The training dataset was used to build the disease risk prediction model, and the validation dataset was used to verify the model identification ability. We first analyzed differentially expressed genes (DEGs) and then used Lasso and random forest (RF) to screen out six key genes (OAS3, USP18, RTP4, SPATS2L, IFI27 and OAS1), which are essential to distinguish SLE from healthy samples. With six key genes incorporated and five iterations of 10-fold cross-validation performed into the RF model, we finally determined the RF model with optimal mtry. The mean values of area under the curve (AUC) and accuracy of the models were over 0.95. The validation dataset was then used to evaluate the AUC performance and our model had an AUC of 0.948. An external validation dataset (GSE99967) with an AUC of 0.810, an accuracy of 0.836, and a sensitivity of 0.921 was used to assess the model's performance. The external validation dataset (GSE185047) of all SLE patients yielded an SLE sensitivity of up to 0.954. The final high-throughput RF model had a mean value of AUC over 0.9, again showing good results. In conclusion, we identified key genetic biomarkers and successfully developed a novel disease risk prediction model for SLE that can be used as a new SLE disease risk prediction aid and contribute to the identification of SLE.
Collapse
Affiliation(s)
- Huajian Chen
- School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
| | - Li Huang
- School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
| | - Xinyue Jiang
- School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
| | - Yue Wang
- School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
| | - Yan Bian
- School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
| | - Shumei Ma
- School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
| | - Xiaodong Liu
- School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
- South Zhejiang Institute of Radiation Medicine and Nuclear Technology, Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Watershed Science and Health of Zhejiang Province, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
47
|
Gómez-Bañuelos E, Shi J, Wang H, Danila MI, Bridges SL, Giles JT, Sims GP, Andrade F, Darrah E. Heavy Chain Constant Region Usage in Antibodies to Peptidylarginine Deiminase 4 as a Marker of Disease Subsets in Rheumatoid Arthritis. Arthritis Rheumatol 2022; 74:1746-1754. [PMID: 35675168 PMCID: PMC9617771 DOI: 10.1002/art.42262] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/03/2022] [Accepted: 06/02/2022] [Indexed: 11/11/2022]
Abstract
OBJECTIVE The study of autoantibody isotypes in autoimmune diseases is useful for identifying clinically relevant endotypes. This study was undertaken to study the prevalence and clinical significance of different isotypes and IgG subclasses of anti-peptidylarginine deiminase 4 (anti-PAD4) autoantibodies in individuals with rheumatoid arthritis (RA). METHODS In 196 RA subjects and 64 healthy controls, anti-PAD4 antibody types were determined using enzyme-linked immunosorbent assay. We investigated associations between anti-PAD4 antibodies and clinical outcomes, and relevant features were confirmed in an independent RA cohort. RESULTS Anti-PAD4 IgG1, anti-PAD4 IgG2, anti-PAD4 IgG3, anti-PAD4 IgG4, anti-PAD4 IgA, and anti-PAD4 IgE antibodies were more frequent in RA patients than healthy controls (P < 0.001). Anti-PAD4 IgG1, anti-PAD4 IgG3, and anti-PAD4 IgE were associated with distinct clinical features. Anti-PAD4 IgG1 was predictive of progressive radiographic joint damage (odds ratio [OR] 4.88, P = 0.005), especially in RA patients without baseline joint damage (40% versus 0%, P = 0.003) or in those negative for anti-cyclic citrullinated peptide and/or rheumatoid factor (OR 32; P = 0.009). IgG1 was also associated with higher levels of C-reactive protein (P = 0.006) and interleukin-6 (P = 0.021). RA patients with anti-PAD4 IgG3 had higher baseline joint damage scores (median Sharp/van der Heijde score 13 versus 7, P = 0.046), while those with anti-PAD4 IgE had higher Disease Activity Score in 28 joints (median 4.0 versus 3.5, P = 0.025), more frequent rheumatoid nodules (31% versus 16%, P = 0.025), and more frequent interstitial lung disease (ground-glass opacification) (24% versus 9%, P = 0.014). Anti-PAD4 IgG1 antibody associations with joint damage were corroborated in an independent RA cohort. CONCLUSION Anti-PAD4 IgG1, anti-PAD4 IgG3, and anti-PAD4 IgE antibodies identify discrete disease subsets in RA, suggesting that heavy chain usage drives distinct effector mechanisms of anti-PAD4 antibodies in RA.
Collapse
Affiliation(s)
- E Gómez-Bañuelos
- Division of Rheumatology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - J Shi
- Division of Rheumatology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - H Wang
- Division of Rheumatology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - MI Danila
- Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - SL Bridges
- Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - JT Giles
- Division of Rheumatology, Columbia University, College of Physicians and Surgeons, New York, NY, USA
| | - GP Sims
- Early Respiratory & Inflammation, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - F Andrade
- Division of Rheumatology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - E Darrah
- Division of Rheumatology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
48
|
Palomares O, Elewaut D, Irving PM, Jaumont X, Tassinari P. Regulatory T cells and immunoglobulin E: A new therapeutic link for autoimmunity? Allergy 2022; 77:3293-3308. [PMID: 35852798 DOI: 10.1111/all.15449] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 07/07/2022] [Accepted: 07/14/2022] [Indexed: 01/28/2023]
Abstract
Autoimmune diseases have a prevalence of approximately 7 to 9% and are classified as either organ-specific diseases, including type I diabetes, multiple sclerosis, inflammatory bowel disease and myasthenia gravis, or systemic diseases, including systemic lupus erythematosus, rheumatoid arthritis and Sjögren's syndrome. While many advancements have been made in understanding of the mechanisms of autoimmune disease, including the nature of self-tolerance and its breakdown, there remain unmet needs in terms of effective and highly targeted treatments. T regulatory cells (Tregs) are key mediators of peripheral tolerance and are implicated in many autoimmune diseases, either as a result of reduced numbers or altered function. Tregs may be broadly divided into those generated in the thymus (tTregs) and those generated in the periphery (pTregs). Tregs target many different immune cell subsets and tissues to suppress excessive inflammation and to support tissue repair and homeostasis: there is a fine balance between Treg cell stability and the plasticity that is required to adjust Tregs' regulatory purposes to particular immune responses. The central role of immunoglobulin E (IgE) in allergic disease is well recognized, and it is becoming increasingly apparent that this immunoglobulin also has a wider role encompassing other diseases including autoimmune disease. Anti-IgE treatment restores the capacity of plasmacytoid dendritic cells (pDCs) impaired by IgE- high-affinity IgE receptor (FcεR1) cross-linking to induce Tregs in vitro in atopic patients. The finding that anti-IgE therapy restores Treg cell homeostasis, and that this mechanism is associated with clinical improvement in asthma and chronic spontaneous urticaria suggests that anti-IgE therapy may also have a potential role in the treatment of autoimmune diseases in which Tregs are involved.
Collapse
Affiliation(s)
| | - Dirk Elewaut
- Department of Rheumatology, VIB Center for Inflammation Research, Ghent University, Ghent University Hospital, Ghent, Belgium
| | - Peter M Irving
- Guy's and St Thomas' Hospital Foundation Trust, London, UK
- King's College London, London, UK
| | | | | |
Collapse
|
49
|
Vílchez-Oya F, Balastegui Martin H, García-Martínez E, Corominas H. Not all autoantibodies are clinically relevant. Classic and novel autoantibodies in Sjögren’s syndrome: A critical review. Front Immunol 2022; 13:1003054. [PMID: 36325321 PMCID: PMC9619091 DOI: 10.3389/fimmu.2022.1003054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 09/20/2022] [Indexed: 11/26/2022] Open
Abstract
Sjögren’s syndrome (SjS) is a heterogeneous systemic disease. The abnormal responses to La/SSB and Ro/SSA of both B-cells and T-cells are implicated as well as others, in the destruction of the epithelium of the exocrine glands, whose tissue characteristically shows a peri-epithelial lymphocytic infiltration that can vary from sicca syndrome to systemic disease and lymphoma. Despite the appearance of new autoantibodies, anti-Ro/SSA is still the only autoantibody included in the American College of Rheumatology/European League Against Rheumatism (ACR/EULAR) classification criteria and is used extensively as a traditional biomarker in clinical practice. The study and findings of new autoantibodies in SjS has risen in the previous decade, with a central role given to diagnosis and elucidating new aspects of SjS physiopathology, while raising the opportunity to establish clinical phenotypes with the goal of predicting long-term complications. In this paper, we critically review the classic and the novel autoantibodies in SjS, analyzing the methods employed for detection, the pathogenic role and the wide spectrum of clinical phenotypes.
Collapse
Affiliation(s)
- Francisco Vílchez-Oya
- Department of Anaesthesiology, Pain Medicine Section, Hospital Clínic de Barcelona, Barcelona, Spain
| | | | - E. García-Martínez
- Department of Immunology, Hospital Universitario Gregorio Marañón, Madrid, Spain
| | - Hèctor Corominas
- Department of Rheumatology and Autoimmune Diseases, Hospital de la Santa Creu i Sant, Barcelona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
- Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
- *Correspondence: Hèctor Corominas,
| |
Collapse
|
50
|
Peck AB, Ambrus JL. A Temporal Comparative RNA Transcriptome Profile of the Annexin Gene Family in the Salivary versus Lacrimal Glands of the Sjögren's Syndrome-Susceptible C57BL/6.NOD- Aec1Aec2 Mouse. Int J Mol Sci 2022; 23:11709. [PMID: 36233010 PMCID: PMC9570365 DOI: 10.3390/ijms231911709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/29/2022] [Accepted: 09/29/2022] [Indexed: 11/29/2022] Open
Abstract
A generally accepted hypothesis for the initial activation of an immune or autoimmune response argues that alarmins are released from injured, dying and/or activated immune cells, and these products complex with receptors that activate signal transduction pathways and recruit immune cells to the site of injury where the recruited cells are stimulated to initiate immune and/or cellular repair responses. While there are multiple diverse families of alarmins such as interleukins (IL), heat-shock proteins (HSP), Toll-like receptors (TLR), plus individual molecular entities such as Galectin-3, Calreticulin, Thymosin, alpha-Defensin-1, RAGE, and Interferon-1, one phylogenetically conserved family are the Annexin proteins known to promote an extensive range of biomolecular and cellular products that can directly and indirectly regulate inflammation and immune activities. For the present report, we examined the temporal expression profiles of the 12 mammalian annexin genes (Anxa1-11 and Anxa13), applying our temporal genome-wide transcriptome analyses of ex vivo salivary and lacrimal glands from our C57BL/6.NOD-Aec1Aec2 mouse model of Sjögren's Syndrome (SS), a human autoimmune disease characterized primarily by severe dry mouth and dry eye symptoms. Results indicate that annexin genes Anax1-7 and -11 exhibited upregulated expressions and the initial timing for these upregulations occurred as early as 8 weeks of age and prior to any covert signs of a SS-like disease. While the profiles of the two glands were similar, they were not identical, suggesting the possibility that the SS-like disease may not be uniform in the two glands. Nevertheless, this early pre-clinical and concomitant upregulated expression of this specific set of alarmins within the immune-targeted organs represents a potential target for identifying the pre-clinical stage in human SS as well, a fact that would clearly impact future interventions and therapeutic strategies.
Collapse
Affiliation(s)
- Ammon B Peck
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, P.O. Box 100125, Gainesville, FL 32610, USA
| | - Julian L Ambrus
- Division of Allergy, Immunology and Rheumatology, SUNY Buffalo School of Medicine, 875 Ellicott Street, Buffalo, NY 14203, USA
| |
Collapse
|