1
|
Arslanoglu Aydin E, Ceylaner S, Baglan E, Bagrul I, Kocamaz NG, Ozdel S. Two siblings with monogenic lupus due to C1qC deficiency and case based review. Clin Rheumatol 2025; 44:1355-1365. [PMID: 39843834 DOI: 10.1007/s10067-025-07333-y] [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: 11/01/2024] [Revised: 11/26/2024] [Accepted: 01/11/2025] [Indexed: 01/24/2025]
Abstract
Monogenic lupus is an extremely rare clinical condition in children. Defects in the complement pathway are the most common causes of monogenic lupus. C1qC deficiency is one of the defects in this pathway and is even rarer. Herein, we present two cases of monogenic lupus diagnosed with C1qC deficiency in siblings. In addition, a literature search was conducted for articles on monogenic lupus due to C1qC deficiency. We found 14 articles. Our literature search identified 17 paediatric patients with monogenic lupus associated with C1qC deficiency. 10 (58%) of the reported patients were female. The median age at diagnosis of patients in the literature was 3 years. Mucocutaneous involvement was remarkable in all cases of C1qC deficiency. Joint involvement was reported in about half of the cases. Approximately half of the reported cases has suffered from recurrent infections. 38% of the cases have had CNS involvement and 25% of these had nephritis. While both of our patients had mucocutaneous involvement, one of our patients had recurrent EBV infection. ANA was positive, anti-dsDNA was negative, C3-C4 levels were normal in almost all cases in the reported cases. The anti-Sm and anti-SSA positivities of these cases were also remarkable. These laboratory findings were similar in our patients. The G34R mutation of the C1qC gene is the most common genetic defect identified to date. We found a GRCh38/Hg38 1p36.12 homozygous deletion in the C1qC gene in both of our patients. It is necessary to investigate the causes of monogenic lupus in patients with early-onset lupus, history of consanguineous marriages, and antibody positivity.
Collapse
Affiliation(s)
| | - Serdar Ceylaner
- Department of Genetics, Intergen Genetics Centre, Ankara, Turkey
| | - Esra Baglan
- Department of Pediatric Rheumatology, Ankara Etlik City Hospital, Ankara, Turkey
| | - Ilknur Bagrul
- Department of Pediatric Rheumatology, Ankara Etlik City Hospital, Ankara, Turkey
| | - Nesibe Gokce Kocamaz
- Department of Pediatric Rheumatology, Ankara Etlik City Hospital, Ankara, Turkey
| | - Semanur Ozdel
- Department of Pediatric Rheumatology, Ankara Etlik City Hospital, Ankara, Turkey
| |
Collapse
|
2
|
Pinzon-Hoyos N, Li Y, McGee M, Poolos NP, Marchi N, Brewster AL. Drug-resistant epilepsy associated with peripheral complement decreases and sex-specific cytokine imbalances: a pilot study. Sci Rep 2025; 15:5096. [PMID: 39934240 PMCID: PMC11814292 DOI: 10.1038/s41598-025-88654-5] [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: 11/18/2024] [Accepted: 01/29/2025] [Indexed: 02/13/2025] Open
Abstract
Drug-resistant epilepsy (DRE) presents significant challenges in treatment and management. While seizure-related alterations in peripheral immune players are increasingly recognized, the involvement of the immune complement system remains insufficiently explored in DRE. We studied complement components and their relationship to cytokine profiles in serum samples from 46 DRE patients and 45 matched healthy controls. We examined relationships between these molecules and clinical outcomes, including epilepsy duration, intelligence scores, and age. We identified DRE-associated complement decreases, including reduced levels of C1q, Factor H, C4, C4b, C3, and C3b/iC3b, as well as elevated bFGF. DRE females showed dysregulation of the classical complement pathway and lower TNFα and interleukin-8 compared to healthy females. DRE males exhibited dysregulation of the classical, lectin, and terminal complement pathways, with trends of increased CCL2 and CCL5 compared to healthy males. Specific complement and inflammatory markers (C2, IL-8, and IL-9) correlated with full-scale IQ scores in DRE patients. Our study reveals significantly lower levels of circulating complement components in DRE and sex-specific complement dysregulation and cytokine imbalances. These findings suggest an underlying peripheral immune system vulnerability that may be sex-dependent and warrants further investigation in DRE.
Collapse
Affiliation(s)
- Nicole Pinzon-Hoyos
- Department of Biological Sciences, Dedman College of Humanities & Science, Southern Methodist University, 6501 Airline Rd, Dallas, TX, 75205, USA
| | - Yibo Li
- Department of Biological Sciences, Dedman College of Humanities & Science, Southern Methodist University, 6501 Airline Rd, Dallas, TX, 75205, USA
| | - Monnie McGee
- Department of Statistics and Data Science, Southern Methodist University, Dallas, TX, USA
| | - Nicholas P Poolos
- Department of Neurology and Regional Epilepsy Center, University of Washington, Seattle, WA, USA
| | - Nicola Marchi
- University of Montpellier, CNRS, INSERM, Institut de Génomique Fonctionnelle, Montpellier, France
| | - Amy L Brewster
- Department of Biological Sciences, Dedman College of Humanities & Science, Southern Methodist University, 6501 Airline Rd, Dallas, TX, 75205, USA.
| |
Collapse
|
3
|
Jain H, Kartik S, Kumar A, Dwivedi A, Jayaprakash S, Vasdev V, Chandwani A. Monogenic lupus with neuroregression in an infant due to rare compound heterozygous variants in C1QA gene: Case-based review. Mod Rheumatol Case Rep 2025; 9:57-62. [PMID: 39096524 DOI: 10.1093/mrcr/rxae039] [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: 05/01/2024] [Revised: 05/15/2024] [Accepted: 07/24/2024] [Indexed: 08/05/2024]
Abstract
Monogenic lupus is a rare variant of systemic lupus erythematosus (SLE) that develops in patients with a single gene disorder. Early complement component deficiencies were the first forms of monogenic lupus to be described, and C1Q gene mutations are one of the most common forms. C1QA complement deficiency has been reported to occur usually due to biallelic variants in C1QA gene, and compound heterozygous variants in C1QA gene have rarely been reported. A majority of monogenic lupus patients with C1Q deficiency present with mucocutaneous, renal, and musculoskeletal manifestations. Our patient is an unusual case of monogenic lupus with severe neurological manifestations along with cutaneous, haematological, and hepatic manifestations secondary to rare compound heterozygous variants in C1QA gene and antiribosomal P autoantibody positivity. She was treated with glucocorticoids, rituximab, and fresh frozen plasma with partial neurological recovery. Thus, we present a unique case of monogenic lupus due to a rare compound heterozygous variant in C1QA gene with a brief review of literature.
Collapse
Affiliation(s)
- Harsh Jain
- Department of Clinical Immunology and Rheumatology, Army Hospital Research and Referral, New Delhi, India
| | - S Kartik
- Department of Clinical Immunology and Rheumatology, Army Hospital Research and Referral, New Delhi, India
| | - Abhishek Kumar
- Department of Clinical Immunology and Rheumatology, Army Hospital Research and Referral, New Delhi, India
| | - Aradhana Dwivedi
- Department of Paediatrics, Geneticist, Army Hospital Research and Referral, New Delhi, India
| | - Sankar Jayaprakash
- Department of Clinical Immunology and Rheumatology, Army Hospital Research and Referral, New Delhi, India
| | - V Vasdev
- Department of Clinical Immunology and Rheumatology, Army Hospital Research and Referral, New Delhi, India
| | - Ashish Chandwani
- Department of Clinical Immunology and Rheumatology, Army Hospital Research and Referral, New Delhi, India
| |
Collapse
|
4
|
Oakes A, Liu Y, Dubielecka PM. Complement or insult: the emerging link between complement cascade deficiencies and pathology of myeloid malignancies. J Leukoc Biol 2024; 116:966-984. [PMID: 38836653 PMCID: PMC11531810 DOI: 10.1093/jleuko/qiae130] [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: 02/29/2024] [Revised: 05/01/2024] [Accepted: 05/03/2024] [Indexed: 06/06/2024] Open
Abstract
The complement cascade is an ancient and highly conserved arm of the immune system. The accumulating evidence highlights elevated activity of the complement cascade in cancer microenvironment and emphasizes its effects on the immune, cancer, and cancer stroma cells, pointing to a role in inflammation-mediated etiology of neoplasms. The role the cascade plays in development, progression, and relapse of solid tumors is increasingly recognized, however its role in hematological malignancies, especially those of myeloid origin, has not been thoroughly assessed and remains obscure. As the role of inflammation and autoimmunity in development of myeloid malignancies is becoming recognized, in this review we focus on summarizing the links that have been identified so far for complement cascade involvement in the pathobiology of myeloid malignancies. Complement deficiencies are primary immunodeficiencies that cause an array of clinical outcomes including an increased risk of a range of infectious as well as local or systemic inflammatory and thrombotic conditions. Here, we discuss the impact that deficiencies in complement cascade initiators, mid- and terminal-components and inhibitors have on the biology of myeloid neoplasms. The emergent conclusions indicate that the links between complement cascade, inflammatory signaling, and the homeostasis of hematopoietic system exist, and efforts should continue to detail the mechanistic involvement of complement cascade in the development and progression of myeloid cancers.
Collapse
Affiliation(s)
- Alissa Oakes
- Department of Medicine, Alpert Medical School, Brown University, 69 Brown St, Providence, RI 02906, USA
- Division of Hematology/Oncology, Rhode Island Hospital, 69 Brown St, Providence, RI 02906, USA
- Therapeutic Sciences Graduate program, Brown University, 69 Brown St, Providence, RI 02906, USA
| | - Yuchen Liu
- Greenebaum Comprehensive Cancer Center, University of Maryland Medical Center, 22. S. Greene St., Baltimore, MD 21201-1595, USA
| | - Patrycja M Dubielecka
- Department of Medicine, Alpert Medical School, Brown University, 69 Brown St, Providence, RI 02906, USA
- Division of Hematology/Oncology, Rhode Island Hospital, 69 Brown St, Providence, RI 02906, USA
- Therapeutic Sciences Graduate program, Brown University, 69 Brown St, Providence, RI 02906, USA
- Legorreta Cancer Center, Brown University, One Hoppin St., Coro West, Suite 5.01, Providence, RI 02903, USA
| |
Collapse
|
5
|
Buso H, Adam E, Arkwright PD, Bhattad S, Hamidieh AA, Behfar M, Belot A, Benezech S, Chan AY, Crow YJ, Dvorak CC, Flinn AM, Kapoor U, Lankester A, Kobayashi M, Matsumura R, Mottaghipisheh H, Okada S, Ouachee M, Parvaneh N, Ramprakash S, Satwani P, Sharafian S, Triaille C, Wynn RF, Movahedi N, Ziaee V, Williams E, Slatter M, Gennery AR. Hematopoietic Stem Cell Transplantation for C1q Deficiency: A Study on Behalf of the EBMT Inborn Errors Working Party. J Clin Immunol 2024; 45:35. [PMID: 39470951 PMCID: PMC11522153 DOI: 10.1007/s10875-024-01819-1] [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: 06/19/2024] [Accepted: 10/01/2024] [Indexed: 11/01/2024]
Abstract
C1q deficiency is a rare inborn error of immunity characterized by increased susceptibility to infections and autoimmune manifestations mimicking SLE, with an associated morbidity and mortality. Because C1q is synthesized by monocytes, to date, four patients treated with allogeneic HSCT have been reported, with a positive outcome in three. We conducted an international retrospective study to assess the outcome of HSCT in C1q deficiency. Eighteen patients, fourteen previously unreported, from eleven referral centres, were included. Two patients had two HSCTs, thus 20 HSCTs were performed in total, at a median age of 10 years (range 0.9-19). Indications for HSCT were autoimmune manifestations not controlled by ongoing treatment in seventeen, and early development of MALT lymphoma in one patient. Overall survival (OS) was 71% and event-free survival was 59% at two years (considering an event as acute GvHD ≥ grade III, disease recurrence and death). In eleven patients HSCT led to resolution of autoimmune features and discontinuation of immunosuppressive treatments (follow-up time range 3-84 months). Five patients died due to transplant-related complications. Patients with a severe autoimmune phenotype, defined as neurological and/or renal involvement, had the worst OS (40% vs 84%; p = 0.034). Reviewing data of 69 genetically confirmed C1q deficient patients, we found that anti-Ro antibodies are associated with neurologic involvement, and anti-RNP and anti-DNA antibodies with renal involvement. In conclusion, HSCT may be a valid curative option for C1q deficiency, but careful selection of patients, with an accurate assessment of risk and benefit, is mandatory.
Collapse
Affiliation(s)
- Helena Buso
- Department of Medicine (DIMED), University of Padova, Padua, Italy
- Paediatric Haematopoietic Stem Cell Transplant Unit, Great North Children's Hospital, Newcastle Upon Tyne, NE1 4LP, UK
| | - Etai Adam
- Sheba Medical Center, The Edmond and Lily Safra Children's Hospital, Ramat Gan, Israel
| | - Peter D Arkwright
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | - Sagar Bhattad
- Division of Paediatric Immunology and Rheumatology, Department of Paediatrics, Aster CMI Hospital, Bengaluru, India
| | - Amir Ali Hamidieh
- Pediatric Cell and Gene Therapy Research Center, Gene, Cell & Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Behfar
- Pediatric Cell and Gene Therapy Research Center, Gene, Cell & Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Alexandre Belot
- Department of Paediatric Rheumatology, Femme-Mère-Enfant Hospital, HCL, Lyon, France
| | - Sarah Benezech
- Institute of Hematology and Pediatric Oncology, 69008, Lyon, France
| | - Alice Y Chan
- Division of Pediatric Allergy, Immunology, and Blood and Marrow Transplant, UCSF Benioff Children's Hospital, University of California San Francisco, San Francisco, CA, USA
| | - Yanick J Crow
- Laboratory of Neurogenetics and Neuroinflammation, Imagine Institute, INSERM UMR1163, Paris, France
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Christopher C Dvorak
- Division of Pediatric Allergy, Immunology, and Blood and Marrow Transplant, UCSF Benioff Children's Hospital, University of California San Francisco, San Francisco, CA, USA
| | - Aisling M Flinn
- Department of Pediatric Immunology, Children's Health Ireland at Crumlin, Dublin, Ireland
| | - Urvi Kapoor
- Division of of Pediatrics Haematology, Oncology and Stem Cell Transplant, Children's Hospital New York-Presbyterian, Columbia University, 161 Fort Washington, Irving 7, New York, NY, 10032, USA
| | - Arjan Lankester
- Department of Pediatrics, Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, The Netherlands
| | - Masao Kobayashi
- Department of Pediatrics, Hiroshima University Hospital, 1‑2‑3 Kasumi, Minami‑ku, Hiroshima, 734‑8551, Japan
| | - Risa Matsumura
- Department of Pediatrics, Hiroshima University Hospital, 1‑2‑3 Kasumi, Minami‑ku, Hiroshima, 734‑8551, Japan
| | - Hadi Mottaghipisheh
- Hematology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Satoshi Okada
- Department of Pediatrics, Hiroshima University Hospital, 1‑2‑3 Kasumi, Minami‑ku, Hiroshima, 734‑8551, Japan
| | - Marie Ouachee
- Institute of Hematology and Pediatric Oncology, 69008, Lyon, France
| | - Nima Parvaneh
- Division of Allergy and Clinical Immunology, Department of Pediatrics, Tehran University of Medical Sciences, Tehran, Iran
| | - Stalin Ramprakash
- Aster International Institute of Oncology, Aster CMI Hospital, Bangalore, India
| | - Prakash Satwani
- Division of of Pediatrics Haematology, Oncology and Stem Cell Transplant, Children's Hospital New York-Presbyterian, Columbia University, 161 Fort Washington, Irving 7, New York, NY, 10032, USA
| | - Samin Sharafian
- Department of Allergy and Clinical Immunology, Mofid Children's Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Clément Triaille
- Pôle de Pathologies Rhumatismales Systémiques Et Inflammatoires, Institut de Recherche Expérimentale Et Clinique, Université Catholique de Louvain, Brussels, Belgium
- Pediatric Immunology and Rheumatology Division, Department of Pediatrics, CHU Sainte-Justine, University of Montreal, Montreal, QC, Canada
| | - Robert F Wynn
- Department of Paediatric Haematology & Oncology, Royal Manchester Children's Hospital, Manchester, UK
| | - Nasim Movahedi
- Golestan Rheumatology Research Center (GRRC), Golestan University of Medical Sciences, Gorgan, Iran
- Pediatric Rheumatology Research Group, Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Vahid Ziaee
- Pediatric Rheumatology Research Group, Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pediatrics, Tehran University of Medical Sciences, Tehran, Iran
| | - Eleri Williams
- Paediatric Haematopoietic Stem Cell Transplant Unit, Great North Children's Hospital, Newcastle Upon Tyne, NE1 4LP, UK
| | - Mary Slatter
- Paediatric Haematopoietic Stem Cell Transplant Unit, Great North Children's Hospital, Newcastle Upon Tyne, NE1 4LP, UK
- Translational and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, UK
| | - Andrew R Gennery
- Paediatric Haematopoietic Stem Cell Transplant Unit, Great North Children's Hospital, Newcastle Upon Tyne, NE1 4LP, UK.
- Translational and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, UK.
| |
Collapse
|
6
|
Pinzon-Hoyos N, Li Y, McGee M, Poolos NP, Marchi N, Brewster AL. Sex-Specific Complement and Cytokine Imbalances in Drug-Resistant Epilepsy: Biomarkers of Immune Vulnerability. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.16.612934. [PMID: 39345496 PMCID: PMC11429787 DOI: 10.1101/2024.09.16.612934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Objective Drug-resistant epilepsy (DRE) poses significant challenges in treatment and management. While seizure-related alterations in peripheral immune players are increasingly recognized, the involvement of the complement system, central to immune function, remains insufficiently explored in DRE. This study aimed to investigate the levels of complement system components and their association with cytokine profiles in patients with DRE. Methods We analyzed serum samples from DRE patients (n = 46) and age- and sex-matched healthy controls (n = 45). Complement components and cytokines were quantified using Multi- and Single-plex ELISA. Statistical analyses examined relationships between complement molecules, cytokines, and clinical outcomes including epilepsy duration, Full-Scale Intelligence Quotient (FSIQ) scores, and age. Results We found common alterations in all DRE cases, including significant complement deficiencies (C1q, Factor H, C4, C4b, C3, and C3b/iC3b) and detectable bFGF levels. DRE females showed significantly lower levels of TNFα and IL-8 compared to healthy females. We observed a trend towards elevated CCL2 and CCL5 levels in DRE males compared to healthy males. These findings suggest potential sex dimorphism in immune profiles. Our analysis also indicated associations between specific complement and inflammatory markers (C2, IL-8, and IL-9) and Full-Scale Intelligence Quotient (FSIQ) scores in DRE patients. Interpretation Our study reveals sex-specific peripheral complement deficiencies and cytokine dysregulation in DRE patients, indicating an underlying immune system vulnerability. These findings provide new insights into DRE mechanisms, potentially guiding future research on complement and cytokine signaling toward personalized treatments for DRE patients.
Collapse
Affiliation(s)
- Nicole Pinzon-Hoyos
- Department of Biological Sciences, Southern Methodist University, Dallas, TX, USA
| | - Yibo Li
- Department of Biological Sciences, Southern Methodist University, Dallas, TX, USA
| | - Monnie McGee
- Department of Statistics and Data Science, Southern Methodist University, Dallas, TX, USA
| | - Nicholas P. Poolos
- Department of Neurology and Regional Epilepsy Center, University of Washington, Seattle, WA, USA
| | - Nicola Marchi
- Institut de Génomique Fonctionnelle, University of Montpellier, CNRS, INSERM; Cerebrovascular and Glia Research, Montpellier, France
| | - Amy L. Brewster
- Department of Biological Sciences, Southern Methodist University, Dallas, TX, USA
| |
Collapse
|
7
|
Triaille C, Rao NM, Rice GI, Seabra L, Sutherland FJH, Bondet V, Duffy D, Gennery AR, Fournier B, Bader-Meunier B, Troedson C, Cleary G, Buso H, Dalby-Payne J, Ranade P, Jansen K, De Somer L, Frémond ML, Chavan PP, Wong M, Dale RC, Wouters C, Quartier P, Khubchandani R, Crow YJ. Hereditary C1q Deficiency is Associated with Type 1 Interferon-Pathway Activation and a High Risk of Central Nervous System Inflammation. J Clin Immunol 2024; 44:185. [PMID: 39196411 PMCID: PMC11358312 DOI: 10.1007/s10875-024-01788-5] [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: 03/05/2024] [Accepted: 08/07/2024] [Indexed: 08/29/2024]
Abstract
Hereditary C1q deficiency (C1QDef) is a rare monogenic disorder leading to defective complement pathway activation and systemic lupus erythematosus (SLE)-like manifestations. The link between impairment of the complement cascade and autoimmunity remains incompletely understood. Here, we assessed type 1 interferon pathway activation in patients with C1QDef. Twelve patients with genetically confirmed C1QDef were recruited through an international collaboration. Clinical, biological and radiological data were collected retrospectively. The expression of a standardized panel of interferon stimulated genes (ISGs) in peripheral blood was measured, and the level of interferon alpha (IFNα) protein in cerebrospinal fluid (CSF) determined using SIMOA technology. Central nervous system (encompassing basal ganglia calcification, encephalitis, vasculitis, chronic pachymeningitis), mucocutaneous and renal involvement were present, respectively, in 10, 11 and 2 of 12 patients, and severe infections recorded in 2/12 patients. Elevated ISG expression was observed in all patients tested (n = 10/10), and serum and CSF IFNα elevated in 2/2 patients. Three patients were treated with Janus-kinase inhibitors (JAKi), with variable outcome; one displaying an apparently favourable response in respect of cutaneous and neurological features, and two others experiencing persistent disease despite JAKi therapy. To our knowledge, we report the largest original series of genetically confirmed C1QDef yet described. Additionally, we present a review of all previously described genetically confirmed cases of C1QDef. Overall, individuals with C1QDef demonstrate many characteristics of recognized monogenic interferonopathies: particularly, cutaneous involvement (malar rash, acral vasculitic/papular rash, chilblains), SLE-like disease, basal ganglia calcification, increased expression of ISGs in peripheral blood, and elevated levels of CSF IFNα.
Collapse
Affiliation(s)
- Clément Triaille
- Division of Pediatric Rheumatology, Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium.
- Pôle de Pathologies Rhumatismales Systémiques Et Inflammatoires, Institut de Recherche Expérimentale Et Clinique, Université Catholique de Louvain, Brussels, Belgium.
| | - Neha Mohan Rao
- Department of Pediatric Rheumatology, NH SRCC Hospital, Mumbai, Maharashtra, India
| | - Gillian I Rice
- Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Luis Seabra
- Laboratory of Neurogenetics and Neuroinflammation, Imagine Institute, INSERM UMR1163, Paris, France
| | - Fraser J H Sutherland
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Vincent Bondet
- Translational Immunology Unit, Institut Pasteur, Université Paris-Cité, Paris, France
| | - Darragh Duffy
- Translational Immunology Unit, Institut Pasteur, Université Paris-Cité, Paris, France
| | - Andrew R Gennery
- Translational and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, UK
- Paediatric Stem Cell Transplant Unit, Great North Children's Hospital, Newcastle Upon Tyne, UK
| | - Benjamin Fournier
- Paediatric Immunology-Hematology and Rheumatology Unit, Necker Hospital, APHP Centre, Université Paris-Cité, Paris, France
| | - Brigitte Bader-Meunier
- Paediatric Immunology-Hematology and Rheumatology Unit, Necker Hospital, APHP Centre, Université Paris-Cité, Paris, France
| | - Christopher Troedson
- T. Y. Nelson Department of Neurology and Neurosurgery, Children's Hospital at Westmead, University of Sydney, Westmead, NSW, Australia
| | - Gavin Cleary
- Paediatric Rheumatology, Alder Hey Children's Hospital, Liverpool, UK
| | - Helena Buso
- Paediatric Stem Cell Transplant Unit, Great North Children's Hospital, Newcastle Upon Tyne, UK
- Department of Medicine - DIMED, University of Padova, Padua, Italy
| | - Jacqueline Dalby-Payne
- Specialty of Child and Adolescent Health, Faculty of Medicine, The University of Sydney, Camperdown, Australia
- Department of General Medicine, The Children's Hospital at Westmead, Westmead, Australia
| | - Prajakta Ranade
- Department of Pediatric Rheumatology, NH SRCC Hospital, Mumbai, Maharashtra, India
| | - Katrien Jansen
- Division of Pediatric Neurology, Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium
| | - Lien De Somer
- Division of Pediatric Rheumatology, Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium
| | - Marie-Louise Frémond
- Laboratory of Neurogenetics and Neuroinflammation, Imagine Institute, INSERM UMR1163, Paris, France
- Paediatric Immunology-Hematology and Rheumatology Unit, Necker Hospital, APHP Centre, Université Paris-Cité, Paris, France
| | | | - Melanie Wong
- Department of Allergy and Immunology, Children's Hospital at Westmead, Westmead, Australia
| | - Russell C Dale
- T. Y. Nelson Department of Neurology and Neurosurgery, Children's Hospital at Westmead, University of Sydney, Westmead, NSW, Australia
| | - Carine Wouters
- Division of Pediatric Rheumatology, Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium
- Paediatric Immunology-Hematology and Rheumatology Unit, Necker Hospital, APHP Centre, Université Paris-Cité, Paris, France
| | - Pierre Quartier
- Paediatric Immunology-Hematology and Rheumatology Unit, Necker Hospital, APHP Centre, Université Paris-Cité, Paris, France
| | - Raju Khubchandani
- Department of Pediatric Rheumatology, NH SRCC Hospital, Mumbai, Maharashtra, India
| | - Yanick J Crow
- Laboratory of Neurogenetics and Neuroinflammation, Imagine Institute, INSERM UMR1163, Paris, France.
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
8
|
Yuan X, Ruan W, Bobrow B, Carmeliet P, Eltzschig HK. Targeting hypoxia-inducible factors: therapeutic opportunities and challenges. Nat Rev Drug Discov 2024; 23:175-200. [PMID: 38123660 DOI: 10.1038/s41573-023-00848-6] [Citation(s) in RCA: 52] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/06/2023] [Indexed: 12/23/2023]
Abstract
Hypoxia-inducible factors (HIFs) are highly conserved transcription factors that are crucial for adaptation of metazoans to limited oxygen availability. Recently, HIF activation and inhibition have emerged as therapeutic targets in various human diseases. Pharmacologically desirable effects of HIF activation include erythropoiesis stimulation, cellular metabolism optimization during hypoxia and adaptive responses during ischaemia and inflammation. By contrast, HIF inhibition has been explored as a therapy for various cancers, retinal neovascularization and pulmonary hypertension. This Review discusses the biochemical mechanisms that control HIF stabilization and the molecular strategies that can be exploited pharmacologically to activate or inhibit HIFs. In addition, we examine medical conditions that benefit from targeting HIFs, the potential side effects of HIF activation or inhibition and future challenges in this field.
Collapse
Affiliation(s)
- Xiaoyi Yuan
- Department of Anaesthesiology, Critical Care and Pain Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA.
| | - Wei Ruan
- Department of Anaesthesiology, Critical Care and Pain Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Department of Anaesthesiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Bentley Bobrow
- Department of Emergency Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Peter Carmeliet
- Laboratory of Angiogenesis & Vascular Metabolism, Center for Cancer Biology, VIB, Department of Oncology, KU Leuven, Leuven, Belgium
- Laboratory of Angiogenesis & Vascular Heterogeneity, Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Holger K Eltzschig
- Department of Anaesthesiology, Critical Care and Pain Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA.
- Outcomes Research Consortium, Cleveland, OH, USA.
| |
Collapse
|
9
|
Wang SSY, Tang H, Loe MWC, Yeo SC, Javaid MM. Complements and Their Role in Systemic Disorders. Cureus 2024; 16:e52991. [PMID: 38406130 PMCID: PMC10894639 DOI: 10.7759/cureus.52991] [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: 01/26/2024] [Indexed: 02/27/2024] Open
Abstract
The complement system is critical to the body's innate defense against exogenous pathogens and clearance of endogenous waste, comprising the classical, alternative, and lectin pathways. Although tightly regulated, various congenital and acquired diseases can perturb the complement system, resulting in specific complement deficiencies. Systemic rheumatic, neurological, ophthalmological, renal, and hematological disorders are some prototypical complement-mediated diseases. An adequate understanding of the mechanisms of the normal complement system and the pathophysiology of complement dysregulation is critical for providing diagnostic clues and appropriately managing these conditions. This review guides clinicians in understanding the role of complement factors in systemic diseases and what diagnostic and therapeutic options are available for complement-mediated disorders.
Collapse
Affiliation(s)
| | - Haoming Tang
- Medicine, Duke-National University of Singapore Medical School, Singapore, SGP
| | | | | | - Muhammad M Javaid
- Medicine, Monash University, Melbourne, AUS
- Medicine, Deakin University, Warrnambool, AUS
- Renal Medicine, Woodlands Health, Singapore, SGP
- Nephrology, Tan Tock Seng Hospital, Singapore, SGP
| |
Collapse
|
10
|
Coss SL, Zhou D, Chua GT, Aziz RA, Hoffman RP, Wu YL, Ardoin SP, Atkinson JP, Yu CY. The complement system and human autoimmune diseases. J Autoimmun 2023; 137:102979. [PMID: 36535812 PMCID: PMC10276174 DOI: 10.1016/j.jaut.2022.102979] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 12/11/2022] [Indexed: 12/23/2022]
Abstract
Genetic deficiencies of early components of the classical complement activation pathway (especially C1q, r, s, and C4) are the strongest monogenic causal factors for the prototypic autoimmune disease systemic lupus erythematosus (SLE), but their prevalence is extremely rare. In contrast, isotype genetic deficiency of C4A and acquired deficiency of C1q by autoantibodies are frequent among patients with SLE. Here we review the genetic basis of complement deficiencies in autoimmune disease, discuss the complex genetic diversity seen in complement C4 and its association with autoimmune disease, provide guidance as to when clinicians should suspect and test for complement deficiencies, and outline the current understanding of the mechanisms relating complement deficiencies to autoimmunity. We focus primarily on SLE, as the role of complement in SLE is well-established, but will also discuss other informative diseases such as inflammatory arthritis and myositis.
Collapse
Affiliation(s)
- Samantha L Coss
- Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, USA; Department of Pediatrics, The Ohio State University, Columbus, OH, USA.
| | - Danlei Zhou
- Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, USA; Department of Pediatrics, The Ohio State University, Columbus, OH, USA
| | - Gilbert T Chua
- Department of Pediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Rabheh Abdul Aziz
- Department of Pediatrics, The Ohio State University, Columbus, OH, USA; Department of Allergy, Immunology and Rheumatology, University of Buffalo, NY, USA
| | - Robert P Hoffman
- Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, USA; Department of Pediatrics, The Ohio State University, Columbus, OH, USA
| | - Yee Ling Wu
- Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, USA; Department of Pediatrics, The Ohio State University, Columbus, OH, USA; Department of Microbiology and Immunology, Loyola University Chicago, Maywood, IL, USA
| | - Stacy P Ardoin
- Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, USA; Department of Pediatrics, The Ohio State University, Columbus, OH, USA
| | - John P Atkinson
- Department of Medicine, Division of Rheumatology, Washington University School of Medicine, St Louis, MO, USA
| | - Chack-Yung Yu
- Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, USA; Department of Pediatrics, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
11
|
Abstract
Autoreactive B cells and interferons are central players in systemic lupus erythematosus (SLE) pathogenesis. The partial success of drugs targeting these pathways, however, supports heterogeneity in upstream mechanisms contributing to disease pathogenesis. In this review, we focus on recent insights from genetic and immune monitoring studies of patients that are refining our understanding of these basic mechanisms. Among them, novel mutations in genes affecting intrinsic B cell activation or clearance of interferogenic nucleic acids have been described. Mitochondria have emerged as relevant inducers and/or amplifiers of SLE pathogenesis through a variety of mechanisms that include disruption of organelle integrity or compartmentalization, defective metabolism, and failure of quality control measures. These result in extra- or intracellular release of interferogenic nucleic acids as well as in innate and/or adaptive immune cell activation. A variety of classic and novel SLE autoantibody specificities have been found to recapitulate genetic alterations associated with monogenic lupus or to trigger interferogenic amplification loops. Finally, atypical B cells and novel extrafollicular T helper cell subsets have been proposed to contribute to the generation of SLE autoantibodies. Overall, these novel insights provide opportunities to deepen the immunophenotypic surveillance of patients and open the door to patient stratification and personalized, rational approaches to therapy.
Collapse
Affiliation(s)
- Simone Caielli
- Drukier Institute for Children's Health and Department of Pediatrics, Weill Cornell Medical Center, New York, NY, USA; , ,
| | - Zurong Wan
- Drukier Institute for Children's Health and Department of Pediatrics, Weill Cornell Medical Center, New York, NY, USA; , ,
| | - Virginia Pascual
- Drukier Institute for Children's Health and Department of Pediatrics, Weill Cornell Medical Center, New York, NY, USA; , ,
| |
Collapse
|
12
|
Patra PK, Banday AZ, Nisar R, Priyanka P, Reddy P, Bhattarai D. Persistent skin ulceration - First manifestation of lupus in a child with novel homozygous deletion in C1QC gene. Australas J Dermatol 2023; 64:e107-e108. [PMID: 36416474 DOI: 10.1111/ajd.13955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/27/2022] [Accepted: 11/10/2022] [Indexed: 11/24/2022]
Affiliation(s)
- Pratap Kumar Patra
- Department of Pediatrics, All India Institute of Medical Sciences, Patna, India
| | - Aaqib Zaffar Banday
- Rheumatology Division, Kashmir Clinics Group, Srinagar, India.,Clinical Immunology & Rheumatology Division, Department of Pediatrics, Khyber Medical Institute, Srinagar, India
| | - Rahila Nisar
- Department of Microbiology, Sher-i-Kashmir Institute of Medical Sciences, Srinagar, India
| | - Priyanka Priyanka
- Department of Pediatrics, All India Institute of Medical Sciences, Patna, India
| | - Pakkiresh Reddy
- Department of Pediatrics, All India Institute of Medical Sciences, Patna, India
| | - Dharmagat Bhattarai
- Advanced Center for Immunology & Rheumatology, Om Hospital and Research Center, Kathmandu, Nepal
| |
Collapse
|
13
|
Wang L, Yang Z, Yu H, Lin W, Wu R, Yang H, Yang K. Predicting diagnostic gene expression profiles associated with immune infiltration in patients with lupus nephritis. Front Immunol 2022; 13:839197. [PMID: 36532018 PMCID: PMC9755505 DOI: 10.3389/fimmu.2022.839197] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Accepted: 11/09/2022] [Indexed: 12/03/2022] Open
Abstract
Objective To identify potential diagnostic markers of lupus nephritis (LN) based on bioinformatics and machine learning and to explore the significance of immune cell infiltration in this pathology. Methods Seven LN gene expression datasets were downloaded from the GEO database, and the larger sample size was used as the training group to obtain differential genes (DEGs) between LN and healthy controls, and to perform gene function, disease ontology (DO), and gene set enrichment analyses (GSEA). Two machine learning algorithms, least absolute shrinkage and selection operator (LASSO) and support vector machine-recursive feature elimination (SVM-RFE), were applied to identify candidate biomarkers. The diagnostic value of LN diagnostic gene biomarkers was further evaluated in the area under the ROC curve observed in the validation dataset. CIBERSORT was used to analyze 22 immune cell fractions from LN patients and to analyze their correlation with diagnostic markers. Results Thirty and twenty-one DEGs were screened in kidney tissue and peripheral blood, respectively. Both of which covered macrophages and interferons. The disease enrichment analysis of DEGs in kidney tissues showed that they were mainly involved in immune and renal diseases, and in peripheral blood it was mainly enriched in cardiovascular system, bone marrow, and oral cavity. The machine learning algorithm combined with external dataset validation revealed that C1QA(AUC = 0.741), C1QB(AUC = 0.758), MX1(AUC = 0.865), RORC(AUC = 0.911), CD177(AUC = 0.855), DEFA4(AUC= 0.843)and HERC5(AUC = 0.880) had high diagnostic value and could be used as diagnostic biomarkers of LN. Compared to controls, pathways such as cell adhesion molecule cam, and systemic lupus erythematosus were activated in kidney tissues; cell cycle, cytoplasmic DNA sensing pathways, NOD-like receptor signaling pathways, proteasome, and RIG-1-like receptors were activated in peripheral blood. Immune cell infiltration analysis showed that diagnostic markers in kidney tissue were associated with T cells CD8 and Dendritic cells resting, and in blood were associated with T cells CD4 memory resting, suggesting that CD4 T cells, CD8 T cells and dendritic cells are closely related to the development and progression of LN. Conclusion C1QA, C1QB, MX1, RORC, CD177, DEFA4 and HERC5 could be used as new candidate molecular markers for LN. It may provide new insights into the diagnosis and molecular treatment of LN in the future.
Collapse
Affiliation(s)
- Lin Wang
- Nephrology Department, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhihua Yang
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Hangxing Yu
- Nephrology Department, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Wei Lin
- Nephrology Department, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Ruoxi Wu
- Nephrology Department, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Hongtao Yang
- Nephrology Department, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Kang Yang
- Nephrology Department, The First Affiliated Hospital of Henan University of Chinese Medicine, Henan, China
| |
Collapse
|
14
|
Schanzenbacher J, Köhl J, Karsten CM. Anaphylatoxins spark the flame in early autoimmunity. Front Immunol 2022; 13:958392. [PMID: 35958588 PMCID: PMC9358992 DOI: 10.3389/fimmu.2022.958392] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 06/29/2022] [Indexed: 11/13/2022] Open
Abstract
The complement system (CS) is an ancient and highly conserved part of the innate immune system with important functions in immune defense. The multiple fragments bind to specific receptors on innate and adaptive immune cells, the activation of which translates the initial humoral innate immune response (IR) into cellular innate and adaptive immunity. Dysregulation of the CS has been associated with the development of several autoimmune disorders such as systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), ANCA-associated vasculitis, and autoimmune bullous dermatoses (AIBDs), where complement drives the inflammatory response in the effector phase. The role of the CS in autoimmunity is complex. On the one hand, complement deficiencies were identified as risk factors to develop autoimmune disorders. On the other hand, activation of complement can drive autoimmune responses. The anaphylatoxins C3a and C5a are potent mediators and regulators of inflammation during the effector phase of autoimmunity through engagement of specific anaphylatoxin receptors, i.e., C3aR, C5aR1, and C5aR2 either on or in immune cells. In addition to their role in innate IRs, anaphylatoxins regulate humoral and cellular adaptive IRs including B-cell and T-cell activation, differentiation, and survival. They regulate B- and T-lymphocyte responses either directly or indirectly through the activation of anaphylatoxin receptors via dendritic cells that modulate lymphocyte function. Here, we will briefly review our current understanding of the complex roles of anaphylatoxins in the regulation of immunologic tolerance and the early events driving autoimmunity and the implications of such regulation for therapeutic approaches that target the CS.
Collapse
Affiliation(s)
- Jovan Schanzenbacher
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Jörg Köhl
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
- Division of Immunobiology, Cincinnati Childrens Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Christian M. Karsten
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
- *Correspondence: Christian M. Karsten,
| |
Collapse
|
15
|
Dasdemir S, Yildiz M, Celebi D, Sahin S, Aliyeva N, Haslak F, Gunalp A, Adrovic A, Barut K, Artim Esen B, Kasapcopur O. Genetic screening of early-onset patients with systemic lupus erythematosus by a targeted next-generation sequencing gene panel. Lupus 2022; 31:330-337. [PMID: 35086391 DOI: 10.1177/09612033221076733] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
OBJECTIVE In this study, we aimed to screen 31 genes (C1QA, C1QB, C1QC, C1R, C1S, C2, C3, TREX1, RNASEH2A, RNASEH2B, RNASEH2C, SAMHD1, ADAR, DNASE1, DNASE1L3, PRKCD, ACP5, SLC7A7, IFIH1, TMEM173, ISG15, CYBB, FAS, FASLG, KRAS, NRAS, MAN2B1, PEPD, PTPN11, RAG2, and SHOC2), that we have categorized under the umbrella term "monogenic lupus" using a targeted next-generation sequencing (NGS) panel in 24 individuals with early-onset (≤10 years of age) systemic lupus erythematosus (SLE) and in 24 patients with late-onset (>10 years of age) disease. METHODS A total of 48 SLE patients (24 with disease onset ≤10 years of age and 24 with disease onset >10 years of age) were included. Patients with late-onset disease have been used as patient controls. Sequencing was carried out using 400 bp kit on the Ion S5 system. RESULTS Among the 48 patients, three had one pathogenic variant and 45 patients had at least one rare variant classified as benign, likely benign or variant of unknown significance (VUS). In all three patients with a pathogenic variant, the onset of disease was before 10 years of age. Two patients (they were siblings) carried C1QA homozygote pathogenic allele (p.Gln208Ter, rs121909581), and one patient carried PEPD heterozygote pathogenic allele (p.Arg184Gln, rs121917722). CONCLUSION We demonstrated a pathogenic variant in our target gene panel with a frequency of 9.52% in patients with a disease onset ≤10 years of age. All patients with early-onset SLE phenotype, irrespective of a positive family history for SLE or parental consanguinity, should be scanned for a single-gene defect by a targeted gene panel sequencing. With the discovery of many single-gene defects and ongoing efforts to identify novel genes in SLE, similar gene panels including even more genes will possibly become more necessary and practical in the future.
Collapse
Affiliation(s)
- Selcuk Dasdemir
- Department of Medical Biology, Istanbul Faculty of Medicine, 64298Istanbul University, Istanbul, Turkey
| | - Mehmet Yildiz
- Department of Pediatric Rheumatology, Cerrahpasa Medical Faculty, 532719Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Damla Celebi
- Department of Medical Biology, Istanbul Faculty of Medicine, 64298Istanbul University, Istanbul, Turkey
| | - Sezgin Sahin
- Department of Pediatric Rheumatology, Cerrahpasa Medical Faculty, 532719Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Numune Aliyeva
- Department of Internal Medicine, Istanbul Faculty of Medicine, Division of Rheumatology, 64298Istanbul University, Istanbul, Turkey
| | - Fatih Haslak
- Department of Pediatric Rheumatology, Cerrahpasa Medical Faculty, 532719Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Aybuke Gunalp
- Department of Pediatric Rheumatology, Cerrahpasa Medical Faculty, 532719Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Amra Adrovic
- Department of Pediatric Rheumatology, Cerrahpasa Medical Faculty, 532719Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Kenan Barut
- Department of Pediatric Rheumatology, Cerrahpasa Medical Faculty, 532719Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Bahar Artim Esen
- Department of Internal Medicine, Istanbul Faculty of Medicine, Division of Rheumatology, 64298Istanbul University, Istanbul, Turkey
| | - Ozgur Kasapcopur
- Department of Pediatric Rheumatology, Cerrahpasa Medical Faculty, 532719Istanbul University-Cerrahpasa, Istanbul, Turkey
| |
Collapse
|
16
|
Disease criteria of systemic lupus erythematosus (SLE); the potential role of non-criteria autoantibodies. J Transl Autoimmun 2022; 5:100143. [PMID: 35072035 PMCID: PMC8761754 DOI: 10.1016/j.jtauto.2022.100143] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 01/04/2022] [Accepted: 01/05/2022] [Indexed: 12/22/2022] Open
Abstract
Patients with SLE show a broad spectrum of more than 200 autoantibodies. They can be pathogenic, predictive, prognostic or even an epiphenomenon. Here, we discuss different autoantibodies that have not been included in EULAR/ACR 2019 classification criteria. Most of them have been addressed to monitor and detect disease activity and not specifically as classification criteria. Indeed, markers to assess disease activity fluctuate as compared with classification criteria and their validation is different. The development of new methods will probably bring new clinical associations and be evaluated as potential classification criteria. Most of the autoantibodies described in SLE are of utility in monitoring disease activity. The validation of activity biomarkers is different from classification criteria biomarkers. The new methods coming into the clinical routine will bring new associations and potentially classification criteria.
Collapse
|
17
|
Hołubowicz R, Ożyhar A, Dobryszycki P. Natural Mutations Affect Structure and Function of gC1q Domain of Otolin-1. Int J Mol Sci 2021; 22:ijms22169085. [PMID: 34445792 PMCID: PMC8396674 DOI: 10.3390/ijms22169085] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/13/2021] [Accepted: 08/19/2021] [Indexed: 12/29/2022] Open
Abstract
Otolin-1 is a scaffold protein of otoliths and otoconia, calcium carbonate biominerals from the inner ear. It contains a gC1q domain responsible for trimerization and binding of Ca2+. Knowledge of a structure-function relationship of gC1q domain of otolin-1 is crucial for understanding the biology of balance sensing. Here, we show how natural variants alter the structure of gC1q otolin-1 and how Ca2+ are able to revert some effects of the mutations. We discovered that natural substitutions: R339S, R342W and R402P negatively affect the stability of apo-gC1q otolin-1, and that Q426R has a stabilizing effect. In the presence of Ca2+, R342W and Q426R were stabilized at higher Ca2+ concentrations than the wild-type form, and R402P was completely insensitive to Ca2+. The mutations affected the self-association of gC1q otolin-1 by inducing detrimental aggregation (R342W) or disabling the trimerization (R402P) of the protein. Our results indicate that the natural variants of gC1q otolin-1 may have a potential to cause pathological changes in otoconia and otoconial membrane, which could affect sensing of balance and increase the probability of occurrence of benign paroxysmal positional vertigo (BPPV).
Collapse
Affiliation(s)
- Rafał Hołubowicz
- Correspondence: (R.H.); (P.D.); Tel.: +48-71-320-63-34 (R.H.); +48-71-320-63-32 (P.D.)
| | | | - Piotr Dobryszycki
- Correspondence: (R.H.); (P.D.); Tel.: +48-71-320-63-34 (R.H.); +48-71-320-63-32 (P.D.)
| |
Collapse
|
18
|
Clinical and Immunological Biomarkers for Systemic Lupus Erythematosus. Biomolecules 2021; 11:biom11070928. [PMID: 34206696 PMCID: PMC8301935 DOI: 10.3390/biom11070928] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/15/2021] [Accepted: 06/17/2021] [Indexed: 12/16/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is characterized by immune system dysfunction and is clinically heterogeneous, exhibiting renal, dermatological, neuropsychiatric, and cardiovascular symptoms. Clinical and physiological assessment is usually inadequate for diagnosing and assessing pathophysiological processes in SLE. Clinical and immunological biomarkers could play a critical role in improving diagnosis, assessment, and ultimately, control of SLE. This article reviews clinical and immunological biomarkers that could diagnose and monitor disease activity in SLE, with and without organ-specific injury. In addition, novel SLE biomarkers that have been discovered through “omics” research are also reviewed.
Collapse
|
19
|
Sogkas G, Atschekzei F, Adriawan IR, Dubrowinskaja N, Witte T, Schmidt RE. Cellular and molecular mechanisms breaking immune tolerance in inborn errors of immunity. Cell Mol Immunol 2021; 18:1122-1140. [PMID: 33795850 PMCID: PMC8015752 DOI: 10.1038/s41423-020-00626-z] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 12/11/2020] [Indexed: 02/01/2023] Open
Abstract
In addition to susceptibility to infections, conventional primary immunodeficiency disorders (PIDs) and inborn errors of immunity (IEI) can cause immune dysregulation, manifesting as lymphoproliferative and/or autoimmune disease. Autoimmunity can be the prominent phenotype of PIDs and commonly includes cytopenias and rheumatological diseases, such as arthritis, systemic lupus erythematosus (SLE), and Sjogren's syndrome (SjS). Recent advances in understanding the genetic basis of systemic autoimmune diseases and PIDs suggest an at least partially shared genetic background and therefore common pathogenic mechanisms. Here, we explore the interconnected pathogenic pathways of autoimmunity and primary immunodeficiency, highlighting the mechanisms breaking the different layers of immune tolerance to self-antigens in selected IEI.
Collapse
Affiliation(s)
- Georgios Sogkas
- Department of Rheumatology and Immunology, Hannover Medical School, Hanover, Germany.
- Hannover Medical School, Cluster of Excellence RESIST (EXC 2155), Hanover, Germany.
| | - Faranaz Atschekzei
- Department of Rheumatology and Immunology, Hannover Medical School, Hanover, Germany
- Hannover Medical School, Cluster of Excellence RESIST (EXC 2155), Hanover, Germany
| | - Ignatius Ryan Adriawan
- Department of Rheumatology and Immunology, Hannover Medical School, Hanover, Germany
- Hannover Medical School, Cluster of Excellence RESIST (EXC 2155), Hanover, Germany
| | - Natalia Dubrowinskaja
- Department of Rheumatology and Immunology, Hannover Medical School, Hanover, Germany
- Hannover Medical School, Cluster of Excellence RESIST (EXC 2155), Hanover, Germany
| | - Torsten Witte
- Department of Rheumatology and Immunology, Hannover Medical School, Hanover, Germany
- Hannover Medical School, Cluster of Excellence RESIST (EXC 2155), Hanover, Germany
| | - Reinhold Ernst Schmidt
- Department of Rheumatology and Immunology, Hannover Medical School, Hanover, Germany
- Hannover Medical School, Cluster of Excellence RESIST (EXC 2155), Hanover, Germany
| |
Collapse
|
20
|
Zecevic M, Minic A, Pasic S, Perovic V, Prohászka Z. Case Report: Early Onset Systemic Lupus Erythematosus Due to Hereditary C1q Deficiency Treated With Fresh Frozen Plasma. Front Pediatr 2021; 9:756387. [PMID: 34993161 PMCID: PMC8724570 DOI: 10.3389/fped.2021.756387] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 10/29/2021] [Indexed: 11/18/2022] Open
Abstract
Background: Hereditary C1q deficiency is associated with early-onset autoimmunity causing SLE or SLE-like disease as well as increased risk for infections with encapsulated bacteria. It is a rare genetic condition inherited in an autosomal recessive manner, caused by mutations in C1q genes. Treatment and management of this rare disease are very complex and include prophylactic vaccination, antibiotics, and immunosuppressive drugs. There are two possible modalities for the replacement of the missing protein: regular fresh frozen plasma (FFP) administration and allogeneic hematopoietic stem cell transplant because the protein is derived from monocytes. Replacing C1q with FFP is being attempted in some patients with success in controlling the disease and in avoiding flare. Case Report: We report a case of sixteen-month-old girl with ulcerations in her mouth, skin erythema, and elevated liver enzymes. ANAs were positive, antibodies against dsDNA were negative, but she had positive anti-Smith antibodies. Complement complements C3 and C4 levels were normal. Total complement activity, classical pathway (hemolytic test) was deficient and C1q antigen was below the detection limit supporting the presence of C1q deficiency. The girl has pathogenic homozygous nonsense mutation in C1qC gene, Arg69Ter (c205>T). The initial response to corticosteroid therapy was good. Regular fresh frozen plasma infusions keep her disease under control, and we were able to reduce the dose of corticosteroids. Conclusion: Young patients with cutaneous lesions resembling SLE, early onset of autoimmunity, with normal C3, C4, elevated ANAs, and negative anti-dsDNA, C1q deficiency should be suspected and complement screening tests should be done. It is important to exclude secondary C1q deficiency. FFP in our patient seems to be well tolerated, without any side effects, able to control the disease.
Collapse
Affiliation(s)
- Milica Zecevic
- Clinical Immunology and Allergy Department, Institute for Health Protection of Mother and Child of Serbia "Dr Vukan Cupic", Belgrade, Serbia
| | - Aleksandra Minic
- Clinical Immunology and Allergy Department, Institute for Health Protection of Mother and Child of Serbia "Dr Vukan Cupic", Belgrade, Serbia
| | - Srdjan Pasic
- Clinical Immunology and Allergy Department, Institute for Health Protection of Mother and Child of Serbia "Dr Vukan Cupic", Belgrade, Serbia.,Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Vladimir Perovic
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Zoltán Prohászka
- Department of Internal Medicine and Haematology, Semmelweis University, Budapest, Hungary.,Research Group for Immunology and Haematology, Semmelweis University-Eötvös Loránd Research Network (Office for Supported Research Groups), Budapest, Hungary
| |
Collapse
|
21
|
González LA, Ugarte-Gil MF, Alarcón GS. Systemic lupus erythematosus: The search for the ideal biomarker. Lupus 2020; 30:181-203. [PMID: 33307987 DOI: 10.1177/0961203320979051] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
During the last decades, there has been an increased interest in the discovery and validation of biomarkers that reliably reflect specific aspects of lupus. Although many biomarkers have been developed, few of them have been validated and used in clinical practice, but with unsatisfactory performances. Thus, there is still a need to rigorously validate many of these novel promising biomarkers in large-scale longitudinal studies and also identify better biomarkers not only for lupus diagnosis but also for monitoring and predicting upcoming flares and response to treatment. Besides serological biomarkers, urinary and cerebrospinal fluid biomarkers have emerged for assessing both renal and central nervous system involvement in systemic lupus erythematosus, respectively. Also, novel omics techniques help us to understand the molecular basis of the disease and also allow the identification of novel biomarkers which may be potentially useful for guiding new therapeutic targets.
Collapse
Affiliation(s)
- Luis Alonso González
- Division of Rheumatology, Department of Internal Medicine, School of Medicine, Universidad de Antioquia, Hospital Universitario de San Vicente Fundación, Medellín, Colombia
| | - Manuel Francisco Ugarte-Gil
- Rheumatology Department, Hospital Guillermo Almenara Irigoyen, EsSalud, Lima, Perú.,School of Medicine, Universidad Científica del Sur, Lima, Perú
| | - Graciela S Alarcón
- Division of Clinical Immunology and Rheumatology, Department of Medicine, School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA.,Department of Medicine, School of Medicine, Universidad Peruana Cayetano Heredia, Lima, Perú
| |
Collapse
|
22
|
Bordron A, Bagacean C, Tempescul A, Berthou C, Bettacchioli E, Hillion S, Renaudineau Y. Complement System: a Neglected Pathway in Immunotherapy. Clin Rev Allergy Immunol 2020; 58:155-171. [PMID: 31144209 DOI: 10.1007/s12016-019-08741-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Approved for the treatment of autoimmune diseases, hematological malignancies, and solid cancers, several monoclonal antibodies (mAb) make use of complement in their mechanism of action. Such an assessment is based on comprehensive investigations that used mouse models, in vitro studies, and analyses from patients at initiation (basal level to highlight deficiencies) and after treatment initiation (mAb impact on complement), which have further provided key insights into the importance of the complement activation and/or complement deficiencies in mAb activity. Accordingly, new approaches can now be developed with the final objective of increasing the clinical efficacy of mAb. These improvements include (i) the concurrent administration of fresh frozen plasma during mAb therapy; (ii) mAb modifications such as immunoglobulin G subclass switching, Fc mutation, or IgG hexamerization to improve the fixation and activation of C1q; (iii) optimization of the target recognition to induce a higher complement-dependent cytotoxicity (CDC) and/or complement-dependant cellular cytotoxicity (CDCC); and (iv) the control of soluble and cellular complement inhibitors.
Collapse
Affiliation(s)
- Anne Bordron
- Inserm UMR1227, B lymphocytes and autoimmunity, University of Brest, Brest, France
| | - Cristina Bagacean
- Inserm UMR1227, B lymphocytes and autoimmunity, University of Brest, Brest, France.,Service d'Hématologie, CHU de Brest, Brest, France
| | - Adrian Tempescul
- Inserm UMR1227, B lymphocytes and autoimmunity, University of Brest, Brest, France.,Service d'Hématologie, CHU de Brest, Brest, France
| | - Christian Berthou
- Inserm UMR1227, B lymphocytes and autoimmunity, University of Brest, Brest, France.,Service d'Hématologie, CHU de Brest, Brest, France
| | | | - Sophie Hillion
- Inserm UMR1227, B lymphocytes and autoimmunity, University of Brest, Brest, France.,Laboratory of Immunology and Immunotherapy, CHU de Brest, Brest, France
| | - Yves Renaudineau
- Inserm UMR1227, B lymphocytes and autoimmunity, University of Brest, Brest, France. .,Laboratory of Immunology and Immunotherapy, CHU de Brest, Brest, France.
| |
Collapse
|
23
|
Bone marrow transplantation from a human leukocyte antigen-mismatched unrelated donor in a case with C1q deficiency associated with refractory systemic lupus erythematosus. Int J Hematol 2020; 113:302-307. [PMID: 33000368 DOI: 10.1007/s12185-020-03004-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 08/10/2020] [Accepted: 09/10/2020] [Indexed: 10/23/2022]
Abstract
Human C1q deficiency is frequently associated with systemic lupus erythematosus (SLE), which requires long-term systemic corticosteroid administration. We report the case of a 12-year-old female patient with C1q deficiency presenting with intractable SLE who successfully underwent bone marrow transplantation from a human leukocyte antigen (HLA)-mismatched unrelated donor with an immunosuppressive conditioning regimen based on fludarabine, melphalan, and anti-thymocyte globulin. She developed Grade I graft-versus-host disease, but did not have any transplantation-related morbidity. Complete donor chimerism has been maintained for 2 years after transplantation, leading to the restoration of C1q levels and the resolution of SLE symptoms. Normal C1q mRNA expression was observed in CD14 + cells. Hematopoietic stem cell transplantation from an HLA-mismatched donor is a feasible treatment for patients with C1q deficiency with refractory SLE that is dependent on systemic corticosteroid treatment who do not have an HLA-matched donor.
Collapse
|
24
|
Jiang SH, Stanley M, Vinuesa CG. Rare genetic variants in systemic autoimmunity. Immunol Cell Biol 2020; 98:490-499. [PMID: 32315078 DOI: 10.1111/imcb.12339] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 04/15/2020] [Accepted: 04/15/2020] [Indexed: 01/03/2023]
Abstract
Autoimmune disease is a substantial cause of morbidity and is strongly influenced by genetic risk. Extensive efforts have characterized the overall genetic basis of many autoimmune diseases, typically by investigation of common variants. While these common variants have modest effects and may cumulatively predispose to disease, it is also increasingly apparent that rare variants have significantly greater effect on phenotype and are likely to contribute to autoimmune disease. Recent advances have illustrated the next fundamental step in elucidating the genetic basis of autoimmunity, moving beyond association to demonstrate the functional consequences of these variants.
Collapse
Affiliation(s)
- Simon H Jiang
- Centre for Personalised Immunology, NHMRC Centre for Research Excellence, Acton, ACT, 2601, Australia.,Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Australian National University, Acton, ACT, 2601, Australia.,Department of Renal Medicine, The Canberra Hospital, Garran, ACT, 2601, Australia
| | - Maurice Stanley
- Centre for Personalised Immunology, NHMRC Centre for Research Excellence, Acton, ACT, 2601, Australia.,Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Australian National University, Acton, ACT, 2601, Australia
| | - Carola G Vinuesa
- Centre for Personalised Immunology, NHMRC Centre for Research Excellence, Acton, ACT, 2601, Australia.,Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Australian National University, Acton, ACT, 2601, Australia.,China Australia Centre for Personalised Immunology, Shanghai Renji Hospital, Jiao Tong University Shanghai, Huangpu Qu, 200333, China
| |
Collapse
|
25
|
Moghimi SM, Simberg D, Papini E, Farhangrazi ZS. Complement activation by drug carriers and particulate pharmaceuticals: Principles, challenges and opportunities. Adv Drug Deliv Rev 2020; 157:83-95. [PMID: 32389761 DOI: 10.1016/j.addr.2020.04.012] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 04/30/2020] [Accepted: 04/30/2020] [Indexed: 12/16/2022]
Abstract
Considering the multifaceted protective and homeostatic roles of the complement system, many consequences arise when drug carriers, and particulate pharmaceutical formulations clash with complement proteins, and trigger complement cascade. Complement activation may induce formulation destabilization, promote opsonization, and affect biological and therapeutic performance of pharmaceutical nano- and micro-particles. In some cases, complement activation is beneficial, where complement may play a role in prophylactic protection, whereas uncontrolled complement activation is deleterious, and contributes to disease progression. Accordingly, design initiatives with particulate medicines should consider complement activation properties of the end formulation within the context of administration route, dosing, systems biology, and therapeutic perspective. Here we examine current progress in mechanistic processes underlying complement activation by pre-clinical and clinical particles, identify opportunities and challenges ahead, and suggest future directions in nanomedicine-complement interface research.
Collapse
Affiliation(s)
- S Moein Moghimi
- School of Pharmacy, Newcastle University, Newcastle upon Tyne NE1 7RU, UK; Translational and Clinical Research Institute, Faculty of Health and Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; Colorado Center for Nanomedicine and Nanosafety, Skagg's School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| | - Dmitri Simberg
- Colorado Center for Nanomedicine and Nanosafety, Skagg's School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Translational Bio-Nanosciences Laboratory, School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Emanuele Papini
- Department of Biomedical Sciences, University of Padua, Padua 35121, Italy; CRIBI Biotechnology Center, University of Padua, Padua 35121, Italy
| | - Z Shadi Farhangrazi
- S. M. Discovery Group Inc., Denver, CO, USA; S. M. Discovery Group Ltd., Durham, UK
| |
Collapse
|
26
|
Hata J, Machida T, Matsuoka K, Hoshi S, Akaihata H, Hiraki H, Suzuki T, Ogawa S, Kataoka M, Haga N, Ishibashi K, Homma Y, Sekine H, Kojima Y. Complement activation by autoantigen recognition in the growth process of benign prostatic hyperplasia. Sci Rep 2019; 9:20357. [PMID: 31889151 PMCID: PMC6937285 DOI: 10.1038/s41598-019-57001-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 12/19/2019] [Indexed: 02/02/2023] Open
Abstract
The pathophysiology of benign prostatic hyperplasia (BPH) remained unclear. Here, we concentrated on the complement activation in the growth of BPH using a rat model. BPH tissues were harvested from rats after rat urogenital sinus implantation. The local expression and deposition levels of C1q, C3, mannose-binding lectin (MBL), factor B (FB), and C5b-9 in the rat and human BPH tissues were analyzed by real-time RT-PCR, western blotting and immunohistochemistry (IHC). Serum IgG levels in the rat BPH model were analyzed by ELISA, and IHC was used to assess tissue localization. Proteins binding serum IgG autoantibody in the BPH rats were isolated by immunoprecipitation. C1q, C3, MBL, FB and C5b-9 were highly localized in rat BPH tissues compared to normal tissues. In contrast, C3, FB and C5b-9, but not C1q and MBL, were abundantly detected in human BPH tissues compared to normal tissues. Diffuse localization of IgG in rat BPH tissues was found. Heat shock protein 90, annexin, α-smooth muscle actin, and β-actin were identified as targets for IgG autoantibodies in the BPH model. Our results strongly suggested the role for complement activation in the growth process of BPH, likely triggered by classical pathway activation with autoantibodies.
Collapse
Affiliation(s)
- Junya Hata
- Department of Urology, Fukushima Medical University School of Medicine, Fukushima, 960-1295, Japan.
| | - Takeshi Machida
- Department of Immunology, Fukushima Medical University School of qwMedicine, Fukushima, 960-1295, Japan
| | - Kanako Matsuoka
- Department of Urology, Fukushima Medical University School of Medicine, Fukushima, 960-1295, Japan
| | - Seiji Hoshi
- Department of Urology, Fukushima Medical University School of Medicine, Fukushima, 960-1295, Japan
| | - Hidenori Akaihata
- Department of Urology, Fukushima Medical University School of Medicine, Fukushima, 960-1295, Japan
| | - Hiroyuki Hiraki
- Department of Urology, Fukushima Medical University School of Medicine, Fukushima, 960-1295, Japan
| | - Toshiyuki Suzuki
- Department of Biomolecular Science Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima, 960-1295, Japan
| | - Soichiro Ogawa
- Department of Urology, Fukushima Medical University School of Medicine, Fukushima, 960-1295, Japan
| | - Masao Kataoka
- Department of Urology, Fukushima Medical University School of Medicine, Fukushima, 960-1295, Japan
| | - Nobuhiro Haga
- Department of Urology, Fukushima Medical University School of Medicine, Fukushima, 960-1295, Japan
| | - Kei Ishibashi
- Department of Urology, Fukushima Medical University School of Medicine, Fukushima, 960-1295, Japan
| | - Yoshimi Homma
- Department of Biomolecular Science Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima, 960-1295, Japan
| | - Hideharu Sekine
- Department of Immunology, Fukushima Medical University School of qwMedicine, Fukushima, 960-1295, Japan
| | - Yoshiyuki Kojima
- Department of Urology, Fukushima Medical University School of Medicine, Fukushima, 960-1295, Japan
| |
Collapse
|
27
|
Abstract
The transcription factor MafB regulates macrophage differentiation. However, studies on
the phenotype of Mafb-deficient macrophages are still limited. Recently,
it was shown that the specific expression of MafB permits macrophages to be distinguished
from dendritic cells. In addition, MafB has been reported to be involved in various
diseases related to macrophages. Studies using macrophage-specific
Mafb-deficient mice show that MafB is linked to atherosclerosis,
autoimmunity, obesity, and ischemic stroke, all of which exhibit macrophage abnormality.
Therefore, MafB is hypothesized to be indispensable for the regulation of macrophages to
maintain systemic homeostasis and may serve as an innovative target for treating
macrophage-related diseases.
Collapse
Affiliation(s)
- Michito Hamada
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.,Laboratory Animal Resource Center (LARC), Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Yuki Tsunakawa
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.,Ph.D. Program in Human Biology, School of Integrative and Global Majors, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Hyojung Jeon
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.,Laboratory Animal Resource Center (LARC), Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Manoj Kumar Yadav
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.,Ph.D. Program in Human Biology, School of Integrative and Global Majors, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Satoru Takahashi
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.,Laboratory Animal Resource Center (LARC), Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| |
Collapse
|
28
|
Lubbers R, Beaart-van de Voorde LJJ, van Leeuwen K, de Boer M, Gelderman KA, van den Berg MJ, Ketel AG, Simon A, de Ree J, Huizinga TWJ, Steup-Beekman GM, Trouw LA. Complex medical history of a patient with a compound heterozygous mutation in C1QC. Lupus 2019; 28:1255-1260. [PMID: 31357913 PMCID: PMC6710612 DOI: 10.1177/0961203319865029] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Introduction C1q is an essential part of the classical pathway of complement activation. Genetic deficiencies, caused by homozygous mutations in one of the C1q genes, are rare and are strongly associated with development of systemic lupus erythematosus (SLE). Here we describe a C1q-deficient patient with a compound heterozygous mutation. Material and methods Serum was analysed with enzyme-linked immunosorbent assay (ELISA) and Western blot for the presence of C1q, and DNA and RNA sequencing was performed to identify the mutations and confirm that these were located on different chromosomes. Results The medical history of the patient includes SLE diagnosis at age 11 years with cerebral involvement at age 13, various infections, osteonecrosis and hemophagocytic syndrome. Using ELISA and Western blot, we confirmed the absence of C1q in the serum of the patient. Using DNA sequencing, two mutations in the C1QC gene were identified: c.100G > A p.(Gly34Arg) and c.205C > T p.(Arg69X). With RNA sequencing we confirmed that the mutations are located on different chromosomes. Discussion The patient described in this case report has a compound heterozygous mutation in C1QC resulting in C1q deficiency.
Collapse
Affiliation(s)
- R Lubbers
- 1 Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands
| | | | - K van Leeuwen
- 2 Sanquin Diagnostic Services, Amsterdam, the Netherlands
| | - M de Boer
- 2 Sanquin Diagnostic Services, Amsterdam, the Netherlands
| | - K A Gelderman
- 2 Sanquin Diagnostic Services, Amsterdam, the Netherlands
| | - M J van den Berg
- 3 Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Pediatric Rheumatology and Immunology, Amsterdam, the Netherlands
| | - A G Ketel
- 4 Spaarnegasthuis, Hoofddorp, the Netherlands
| | - A Simon
- 5 Radboud University Medical Center, Center for Immunodeficiency and Autoinflammation, Department of Internal Medicine, Nijmegen, the Netherlands
| | - J de Ree
- 4 Spaarnegasthuis, Hoofddorp, the Netherlands
| | - T W J Huizinga
- 1 Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands
| | - G M Steup-Beekman
- 1 Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands
| | - L A Trouw
- 1 Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands.,6 Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
29
|
Almlöf JC, Nystedt S, Leonard D, Eloranta ML, Grosso G, Sjöwall C, Bengtsson AA, Jönsen A, Gunnarsson I, Svenungsson E, Rönnblom L, Sandling JK, Syvänen AC. Whole-genome sequencing identifies complex contributions to genetic risk by variants in genes causing monogenic systemic lupus erythematosus. Hum Genet 2019; 138:141-150. [PMID: 30707351 PMCID: PMC6373277 DOI: 10.1007/s00439-018-01966-7] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 12/13/2018] [Indexed: 01/01/2023]
Abstract
Systemic lupus erythematosus (SLE, OMIM 152700) is a systemic autoimmune disease with a complex etiology. The mode of inheritance of the genetic risk beyond familial SLE cases is currently unknown. Additionally, the contribution of heterozygous variants in genes known to cause monogenic SLE is not fully understood. Whole-genome sequencing of DNA samples from 71 Swedish patients with SLE and their healthy biological parents was performed to investigate the general genetic risk of SLE using known SLE GWAS risk loci identified using the ImmunoChip, variants in genes associated to monogenic SLE, and the mode of inheritance of SLE risk alleles in these families. A random forest model for predicting genetic risk for SLE showed that the SLE risk variants were mainly inherited from one of the parents. In the 71 patients, we detected a significant enrichment of ultra-rare ( ≤ 0.1%) missense and nonsense mutations in 22 genes known to cause monogenic forms of SLE. We identified one previously reported homozygous nonsense mutation in the C1QC (Complement C1q C Chain) gene, which explains the immunodeficiency and severe SLE phenotype of that patient. We also identified seven ultra-rare, coding heterozygous variants in five genes (C1S, DNASE1L3, DNASE1, IFIH1, and RNASEH2A) involved in monogenic SLE. Our findings indicate a complex contribution to the overall genetic risk of SLE by rare variants in genes associated with monogenic forms of SLE. The rare variants were inherited from the other parent than the one who passed on the more common risk variants leading to an increased genetic burden for SLE in the child. Higher frequency SLE risk variants are mostly passed from one of the parents to the offspring affected with SLE. In contrast, the other parent, in seven cases, contributed heterozygous rare variants in genes associated with monogenic forms of SLE, suggesting a larger impact of rare variants in SLE than hitherto reported.
Collapse
Affiliation(s)
- Jonas Carlsson Almlöf
- Department of Medical Sciences, Molecular Medicine and Science for Life Laboratory, Uppsala University, 751 23, Uppsala, Sweden.
| | - Sara Nystedt
- Department of Medical Sciences, Molecular Medicine and Science for Life Laboratory, Uppsala University, 751 23, Uppsala, Sweden
| | - Dag Leonard
- Department of Medical Sciences, Rheumatology and Science for Life Laboratory, Uppsala University, 751 85, Uppsala, Sweden
| | - Maija-Leena Eloranta
- Department of Medical Sciences, Rheumatology and Science for Life Laboratory, Uppsala University, 751 85, Uppsala, Sweden
| | - Giorgia Grosso
- Rheumatology Unit, Department of Medicine, Karolinska Institutet, Rheumatology, Karolinska University Hospital, 171 77, Stockholm, Sweden
| | - Christopher Sjöwall
- Division of Neuro and Inflammation Sciences, Department of Clinical and Experimental Medicine, Rheumatology, Linköping University, 581 83, Linköping, Sweden
| | - Anders A Bengtsson
- Department of Clinical Sciences, Rheumatology, Lund University, Skåne University Hospital, 222 42, Lund, Sweden
| | - Andreas Jönsen
- Department of Clinical Sciences, Rheumatology, Lund University, Skåne University Hospital, 222 42, Lund, Sweden
| | - Iva Gunnarsson
- Rheumatology Unit, Department of Medicine, Karolinska Institutet, Rheumatology, Karolinska University Hospital, 171 77, Stockholm, Sweden
| | - Elisabet Svenungsson
- Rheumatology Unit, Department of Medicine, Karolinska Institutet, Rheumatology, Karolinska University Hospital, 171 77, Stockholm, Sweden
| | - Lars Rönnblom
- Department of Medical Sciences, Rheumatology and Science for Life Laboratory, Uppsala University, 751 85, Uppsala, Sweden
| | - Johanna K Sandling
- Department of Medical Sciences, Rheumatology and Science for Life Laboratory, Uppsala University, 751 85, Uppsala, Sweden
| | - Ann-Christine Syvänen
- Department of Medical Sciences, Molecular Medicine and Science for Life Laboratory, Uppsala University, 751 23, Uppsala, Sweden
| |
Collapse
|
30
|
Bolin K, Eloranta ML, Kozyrev SV, Dahlqvist J, Nilsson B, Knight A, Rönnblom L. A case of systemic lupus erythematosus with C1q deficiency, increased serum interferon-α levels and high serum interferogenic activity. Rheumatology (Oxford) 2019; 58:918-919. [DOI: 10.1093/rheumatology/key419] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/18/2018] [Indexed: 02/03/2023] Open
Affiliation(s)
- Karin Bolin
- Department of Medical Sciences, Science for Life Laboratory, Rheumatology
| | | | - Sergey V Kozyrev
- Department of Medical Biochemistry and Microbiology, Uppsala University , Uppsala, Sweden
| | - Johanna Dahlqvist
- Department of Medical Biochemistry and Microbiology, Uppsala University , Uppsala, Sweden
- The Broad Institute of Harvard and MIT, Cambridge, MA, US
| | - Bo Nilsson
- Department of Immunology, Genetics and Pathology
| | - Ann Knight
- Department of Medical Sciences, Rheumatology, Uppsala University, Uppsala, Sweden
| | - Lars Rönnblom
- Department of Medical Sciences, Science for Life Laboratory, Rheumatology
| |
Collapse
|
31
|
Ding S, Liu C, Li Y, Liu H, Liu Z, Chen T, Zhang T, Shao Z, Fu R. Expression of C1q in the serum of patients with non‑severe aplastic anemia, and its association with disease severity. Mol Med Rep 2018; 19:1194-1202. [PMID: 30569170 PMCID: PMC6323203 DOI: 10.3892/mmr.2018.9754] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 11/13/2018] [Indexed: 01/04/2023] Open
Abstract
A type of aplastic anemia (AA), non-severe aplastic anemia (NSAA) is defined as AA that does not meet the diagnostic criteria of severe aplastic anemia (SAA). Complement component 1q (C1q) has an important role in the pathogenesis of various autoimmune diseases; however, the role of C1q in the immune pathogenesis of NSAA is not clear. The current study aimed to determine whether C1q has an important role in the pathogenesis of NSAA. Isobaric tags for relative and absolute quantitation (iTRAQ) was used to compare the protein expression in bone marrow mononuclear cells from patients with NSAA and healthy volunteers. Pathway enrichment analysis was performed to determine the biological functions involved in NSAA. The differential expression of C1q was marked compared with other proteins. Subsequently, the concentration of C1q in serum samples was determined using ELISA and the correlation of C1q levels and NSAA severity was evaluated. The serum concentrations of C1q were significantly lower in untreated patients with newly diagnosed NSAA compared with NSAA cases in remission and normal controls. Furthermore, there was no significant difference in C1q concentration between newly diagnosed patients with NSAA and patients with autoimmune hemolytic anemia or immune thrombocytopenia. The serum concentration of C1q in newly diagnosed NSAA was significantly lower in patients with SAA (P<0.0001); whereas, there was no significant difference between the patients with SAA, patients with NSAA remission and normal controls (P>0.05). Additionally, the serum C1q concentration was significantly correlated with granulocyte counts, the level of hemoglobin, platelet counts, reticulocyte percentage and remission in patients with NSAA. The serum C1q concentration was also positively correlated with the myeloid/plasmacytoid dendritic cell ratio, and negatively correlated with the CD4(+)/CD8(+) ratio. These findings suggested that C1q may be a reliable serological marker for monitoring and evaluating disease severity in patients with NSAA. C1q may have an important role in the immune pathogenesis of NSAA.
Collapse
Affiliation(s)
- Shaoxue Ding
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Chunyan Liu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Yang Li
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Hui Liu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Zhaoyun Liu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Tong Chen
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Tian Zhang
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Zonghong Shao
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Rong Fu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| |
Collapse
|
32
|
Batu ED, Koşukcu C, Taşkıran E, Sahin S, Akman S, Sözeri B, Ünsal E, Bilginer Y, Kasapcopur O, Alikaşifoğlu M, Ozen S. Whole Exome Sequencing in Early-onset Systemic Lupus Erythematosus. J Rheumatol 2018; 45:1671-1679. [PMID: 30008451 DOI: 10.3899/jrheum.171358] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/16/2018] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Systemic lupus erythematosus (SLE) is a multisystem autoimmune disorder. Early-onset, familial, and/or syndromic SLE may reveal monogenic pathologies. The aim of this study was to examine genetic associations in patients with early-onset or familial SLE. METHODS We enrolled 7 SLE cases (from different families) with disease onset ≤ 5 years of age and family history consistent with an autosomal recessive inheritance. Whole exome sequencing (WES) was performed in 6 index cases. Suspected variants were confirmed by Sanger sequencing. We did not perform WES in 1 patient who had features similar to the first 3 cases; only the exons of C1QA, C1QB, and C1QC were screened with Sanger sequencing. RESULTS We demonstrated 2 novel and 3 previously reported variants in genes associated with SLE: a homozygous non-sense alteration (c.622C>T/p.Gln208Ter) in C1QA in 2 patients; homozygous non-sense alteration (c.79C>T/p.Gln27Ter) in C1QC in 1 (novel variant); homozygous missense alteration (c.100G>A/p.Gly34Arg) in C1QC in 1; homozygous missense alteration (c.1945G>C/p.Ala649Pro) in C1S in 1 (novel variant); and homozygous frameshift alteration (c.289_290delAC/p.Thr97Ilefs*2) in DNASE1L3 in 1 patient. Further, in 1 patient, we determined a strong candidate variant in HDAC7 (histone decetylase 7). CONCLUSION Five patients had homozygous alterations in genes coding early complement proteins. This may lead to decreased clearance of apoptotic bodies. One patient had DNASE1L3 variant, which functions in the clearance of self-antigens. In 1 patient, we determined a novel gene that may be important in SLE pathogenesis. We suggest that monogenic causes/associations should be sought in early-onset and/or familial SLE.
Collapse
Affiliation(s)
- Ezgi Deniz Batu
- From the Department of Pediatrics, Division of Rheumatology, and the Department of Medical Genetics, Hacettepe University Faculty of Medicine, Ankara; Department of Bioinformatics, Institute of Health Sciences, Hacettepe University, Ankara; Department of Pediatrics, Division of Rheumatology, Istanbul University Cerrahpasa Faculty of Medicine, Istanbul; Department of Pediatrics, Division of Nephrology and Rheumatology, Akdeniz University Faculty of Medicine, Antalya; Department of Pediatrics, Division of Rheumatology, Umraniye Training and Research Center, University of Health Sciences, Istanbul; Department of Pediatrics, Division of Rheumatology, Dokuz Eylül University Faculty of Medicine, İzmir, Turkey
- E.D. Batu, MD, MSc, Department of Pediatrics, Division of Rheumatology, Hacettepe University Faculty of Medicine; C. Koşukcu, MSc, Department of Medical Genetics, Hacettepe University Faculty of Medicine, and Department of Bioinformatics, Institute of Health Sciences, Hacettepe University; E. Taşkıran, PhD, Department of Medical Genetics, Hacettepe University Faculty of Medicine; S. Sahin, MD, Department of Pediatrics, Division of Rheumatology, Istanbul University Cerrahpasa Faculty of Medicine; S. Akman, MD, Department of Pediatrics, Division of Nephrology and Rheumatology, Akdeniz University Faculty of Medicine; B. Sözeri, MD, Department of Pediatrics, Division of Rheumatology, Umraniye Training and Research Center, University of Health Sciences; E. Ünsal, MD, Department of Pediatrics, Division of Rheumatology, Dokuz Eylül University Faculty of Medicine; Y. Bilginer, MD, MSc, Department of Pediatrics, Division of Rheumatology, Hacettepe University Faculty of Medicine; O. Kasapcopur, MD, Department of Pediatrics, Division of Rheumatology, Istanbul University Cerrahpasa Faculty of Medicine; M. Alikaşifoğlu, MD, PhD, Department of Medical Genetics, Hacettepe University Faculty of Medicine; S. Ozen, MD, MSc, Department of Pediatrics, Division of Rheumatology, Hacettepe University Faculty of Medicine
| | - Can Koşukcu
- From the Department of Pediatrics, Division of Rheumatology, and the Department of Medical Genetics, Hacettepe University Faculty of Medicine, Ankara; Department of Bioinformatics, Institute of Health Sciences, Hacettepe University, Ankara; Department of Pediatrics, Division of Rheumatology, Istanbul University Cerrahpasa Faculty of Medicine, Istanbul; Department of Pediatrics, Division of Nephrology and Rheumatology, Akdeniz University Faculty of Medicine, Antalya; Department of Pediatrics, Division of Rheumatology, Umraniye Training and Research Center, University of Health Sciences, Istanbul; Department of Pediatrics, Division of Rheumatology, Dokuz Eylül University Faculty of Medicine, İzmir, Turkey
- E.D. Batu, MD, MSc, Department of Pediatrics, Division of Rheumatology, Hacettepe University Faculty of Medicine; C. Koşukcu, MSc, Department of Medical Genetics, Hacettepe University Faculty of Medicine, and Department of Bioinformatics, Institute of Health Sciences, Hacettepe University; E. Taşkıran, PhD, Department of Medical Genetics, Hacettepe University Faculty of Medicine; S. Sahin, MD, Department of Pediatrics, Division of Rheumatology, Istanbul University Cerrahpasa Faculty of Medicine; S. Akman, MD, Department of Pediatrics, Division of Nephrology and Rheumatology, Akdeniz University Faculty of Medicine; B. Sözeri, MD, Department of Pediatrics, Division of Rheumatology, Umraniye Training and Research Center, University of Health Sciences; E. Ünsal, MD, Department of Pediatrics, Division of Rheumatology, Dokuz Eylül University Faculty of Medicine; Y. Bilginer, MD, MSc, Department of Pediatrics, Division of Rheumatology, Hacettepe University Faculty of Medicine; O. Kasapcopur, MD, Department of Pediatrics, Division of Rheumatology, Istanbul University Cerrahpasa Faculty of Medicine; M. Alikaşifoğlu, MD, PhD, Department of Medical Genetics, Hacettepe University Faculty of Medicine; S. Ozen, MD, MSc, Department of Pediatrics, Division of Rheumatology, Hacettepe University Faculty of Medicine
| | - Ekim Taşkıran
- From the Department of Pediatrics, Division of Rheumatology, and the Department of Medical Genetics, Hacettepe University Faculty of Medicine, Ankara; Department of Bioinformatics, Institute of Health Sciences, Hacettepe University, Ankara; Department of Pediatrics, Division of Rheumatology, Istanbul University Cerrahpasa Faculty of Medicine, Istanbul; Department of Pediatrics, Division of Nephrology and Rheumatology, Akdeniz University Faculty of Medicine, Antalya; Department of Pediatrics, Division of Rheumatology, Umraniye Training and Research Center, University of Health Sciences, Istanbul; Department of Pediatrics, Division of Rheumatology, Dokuz Eylül University Faculty of Medicine, İzmir, Turkey
- E.D. Batu, MD, MSc, Department of Pediatrics, Division of Rheumatology, Hacettepe University Faculty of Medicine; C. Koşukcu, MSc, Department of Medical Genetics, Hacettepe University Faculty of Medicine, and Department of Bioinformatics, Institute of Health Sciences, Hacettepe University; E. Taşkıran, PhD, Department of Medical Genetics, Hacettepe University Faculty of Medicine; S. Sahin, MD, Department of Pediatrics, Division of Rheumatology, Istanbul University Cerrahpasa Faculty of Medicine; S. Akman, MD, Department of Pediatrics, Division of Nephrology and Rheumatology, Akdeniz University Faculty of Medicine; B. Sözeri, MD, Department of Pediatrics, Division of Rheumatology, Umraniye Training and Research Center, University of Health Sciences; E. Ünsal, MD, Department of Pediatrics, Division of Rheumatology, Dokuz Eylül University Faculty of Medicine; Y. Bilginer, MD, MSc, Department of Pediatrics, Division of Rheumatology, Hacettepe University Faculty of Medicine; O. Kasapcopur, MD, Department of Pediatrics, Division of Rheumatology, Istanbul University Cerrahpasa Faculty of Medicine; M. Alikaşifoğlu, MD, PhD, Department of Medical Genetics, Hacettepe University Faculty of Medicine; S. Ozen, MD, MSc, Department of Pediatrics, Division of Rheumatology, Hacettepe University Faculty of Medicine
| | - Sezgin Sahin
- From the Department of Pediatrics, Division of Rheumatology, and the Department of Medical Genetics, Hacettepe University Faculty of Medicine, Ankara; Department of Bioinformatics, Institute of Health Sciences, Hacettepe University, Ankara; Department of Pediatrics, Division of Rheumatology, Istanbul University Cerrahpasa Faculty of Medicine, Istanbul; Department of Pediatrics, Division of Nephrology and Rheumatology, Akdeniz University Faculty of Medicine, Antalya; Department of Pediatrics, Division of Rheumatology, Umraniye Training and Research Center, University of Health Sciences, Istanbul; Department of Pediatrics, Division of Rheumatology, Dokuz Eylül University Faculty of Medicine, İzmir, Turkey
- E.D. Batu, MD, MSc, Department of Pediatrics, Division of Rheumatology, Hacettepe University Faculty of Medicine; C. Koşukcu, MSc, Department of Medical Genetics, Hacettepe University Faculty of Medicine, and Department of Bioinformatics, Institute of Health Sciences, Hacettepe University; E. Taşkıran, PhD, Department of Medical Genetics, Hacettepe University Faculty of Medicine; S. Sahin, MD, Department of Pediatrics, Division of Rheumatology, Istanbul University Cerrahpasa Faculty of Medicine; S. Akman, MD, Department of Pediatrics, Division of Nephrology and Rheumatology, Akdeniz University Faculty of Medicine; B. Sözeri, MD, Department of Pediatrics, Division of Rheumatology, Umraniye Training and Research Center, University of Health Sciences; E. Ünsal, MD, Department of Pediatrics, Division of Rheumatology, Dokuz Eylül University Faculty of Medicine; Y. Bilginer, MD, MSc, Department of Pediatrics, Division of Rheumatology, Hacettepe University Faculty of Medicine; O. Kasapcopur, MD, Department of Pediatrics, Division of Rheumatology, Istanbul University Cerrahpasa Faculty of Medicine; M. Alikaşifoğlu, MD, PhD, Department of Medical Genetics, Hacettepe University Faculty of Medicine; S. Ozen, MD, MSc, Department of Pediatrics, Division of Rheumatology, Hacettepe University Faculty of Medicine
| | - Sema Akman
- From the Department of Pediatrics, Division of Rheumatology, and the Department of Medical Genetics, Hacettepe University Faculty of Medicine, Ankara; Department of Bioinformatics, Institute of Health Sciences, Hacettepe University, Ankara; Department of Pediatrics, Division of Rheumatology, Istanbul University Cerrahpasa Faculty of Medicine, Istanbul; Department of Pediatrics, Division of Nephrology and Rheumatology, Akdeniz University Faculty of Medicine, Antalya; Department of Pediatrics, Division of Rheumatology, Umraniye Training and Research Center, University of Health Sciences, Istanbul; Department of Pediatrics, Division of Rheumatology, Dokuz Eylül University Faculty of Medicine, İzmir, Turkey
- E.D. Batu, MD, MSc, Department of Pediatrics, Division of Rheumatology, Hacettepe University Faculty of Medicine; C. Koşukcu, MSc, Department of Medical Genetics, Hacettepe University Faculty of Medicine, and Department of Bioinformatics, Institute of Health Sciences, Hacettepe University; E. Taşkıran, PhD, Department of Medical Genetics, Hacettepe University Faculty of Medicine; S. Sahin, MD, Department of Pediatrics, Division of Rheumatology, Istanbul University Cerrahpasa Faculty of Medicine; S. Akman, MD, Department of Pediatrics, Division of Nephrology and Rheumatology, Akdeniz University Faculty of Medicine; B. Sözeri, MD, Department of Pediatrics, Division of Rheumatology, Umraniye Training and Research Center, University of Health Sciences; E. Ünsal, MD, Department of Pediatrics, Division of Rheumatology, Dokuz Eylül University Faculty of Medicine; Y. Bilginer, MD, MSc, Department of Pediatrics, Division of Rheumatology, Hacettepe University Faculty of Medicine; O. Kasapcopur, MD, Department of Pediatrics, Division of Rheumatology, Istanbul University Cerrahpasa Faculty of Medicine; M. Alikaşifoğlu, MD, PhD, Department of Medical Genetics, Hacettepe University Faculty of Medicine; S. Ozen, MD, MSc, Department of Pediatrics, Division of Rheumatology, Hacettepe University Faculty of Medicine
| | - Betül Sözeri
- From the Department of Pediatrics, Division of Rheumatology, and the Department of Medical Genetics, Hacettepe University Faculty of Medicine, Ankara; Department of Bioinformatics, Institute of Health Sciences, Hacettepe University, Ankara; Department of Pediatrics, Division of Rheumatology, Istanbul University Cerrahpasa Faculty of Medicine, Istanbul; Department of Pediatrics, Division of Nephrology and Rheumatology, Akdeniz University Faculty of Medicine, Antalya; Department of Pediatrics, Division of Rheumatology, Umraniye Training and Research Center, University of Health Sciences, Istanbul; Department of Pediatrics, Division of Rheumatology, Dokuz Eylül University Faculty of Medicine, İzmir, Turkey
- E.D. Batu, MD, MSc, Department of Pediatrics, Division of Rheumatology, Hacettepe University Faculty of Medicine; C. Koşukcu, MSc, Department of Medical Genetics, Hacettepe University Faculty of Medicine, and Department of Bioinformatics, Institute of Health Sciences, Hacettepe University; E. Taşkıran, PhD, Department of Medical Genetics, Hacettepe University Faculty of Medicine; S. Sahin, MD, Department of Pediatrics, Division of Rheumatology, Istanbul University Cerrahpasa Faculty of Medicine; S. Akman, MD, Department of Pediatrics, Division of Nephrology and Rheumatology, Akdeniz University Faculty of Medicine; B. Sözeri, MD, Department of Pediatrics, Division of Rheumatology, Umraniye Training and Research Center, University of Health Sciences; E. Ünsal, MD, Department of Pediatrics, Division of Rheumatology, Dokuz Eylül University Faculty of Medicine; Y. Bilginer, MD, MSc, Department of Pediatrics, Division of Rheumatology, Hacettepe University Faculty of Medicine; O. Kasapcopur, MD, Department of Pediatrics, Division of Rheumatology, Istanbul University Cerrahpasa Faculty of Medicine; M. Alikaşifoğlu, MD, PhD, Department of Medical Genetics, Hacettepe University Faculty of Medicine; S. Ozen, MD, MSc, Department of Pediatrics, Division of Rheumatology, Hacettepe University Faculty of Medicine
| | - Erbil Ünsal
- From the Department of Pediatrics, Division of Rheumatology, and the Department of Medical Genetics, Hacettepe University Faculty of Medicine, Ankara; Department of Bioinformatics, Institute of Health Sciences, Hacettepe University, Ankara; Department of Pediatrics, Division of Rheumatology, Istanbul University Cerrahpasa Faculty of Medicine, Istanbul; Department of Pediatrics, Division of Nephrology and Rheumatology, Akdeniz University Faculty of Medicine, Antalya; Department of Pediatrics, Division of Rheumatology, Umraniye Training and Research Center, University of Health Sciences, Istanbul; Department of Pediatrics, Division of Rheumatology, Dokuz Eylül University Faculty of Medicine, İzmir, Turkey
- E.D. Batu, MD, MSc, Department of Pediatrics, Division of Rheumatology, Hacettepe University Faculty of Medicine; C. Koşukcu, MSc, Department of Medical Genetics, Hacettepe University Faculty of Medicine, and Department of Bioinformatics, Institute of Health Sciences, Hacettepe University; E. Taşkıran, PhD, Department of Medical Genetics, Hacettepe University Faculty of Medicine; S. Sahin, MD, Department of Pediatrics, Division of Rheumatology, Istanbul University Cerrahpasa Faculty of Medicine; S. Akman, MD, Department of Pediatrics, Division of Nephrology and Rheumatology, Akdeniz University Faculty of Medicine; B. Sözeri, MD, Department of Pediatrics, Division of Rheumatology, Umraniye Training and Research Center, University of Health Sciences; E. Ünsal, MD, Department of Pediatrics, Division of Rheumatology, Dokuz Eylül University Faculty of Medicine; Y. Bilginer, MD, MSc, Department of Pediatrics, Division of Rheumatology, Hacettepe University Faculty of Medicine; O. Kasapcopur, MD, Department of Pediatrics, Division of Rheumatology, Istanbul University Cerrahpasa Faculty of Medicine; M. Alikaşifoğlu, MD, PhD, Department of Medical Genetics, Hacettepe University Faculty of Medicine; S. Ozen, MD, MSc, Department of Pediatrics, Division of Rheumatology, Hacettepe University Faculty of Medicine
| | - Yelda Bilginer
- From the Department of Pediatrics, Division of Rheumatology, and the Department of Medical Genetics, Hacettepe University Faculty of Medicine, Ankara; Department of Bioinformatics, Institute of Health Sciences, Hacettepe University, Ankara; Department of Pediatrics, Division of Rheumatology, Istanbul University Cerrahpasa Faculty of Medicine, Istanbul; Department of Pediatrics, Division of Nephrology and Rheumatology, Akdeniz University Faculty of Medicine, Antalya; Department of Pediatrics, Division of Rheumatology, Umraniye Training and Research Center, University of Health Sciences, Istanbul; Department of Pediatrics, Division of Rheumatology, Dokuz Eylül University Faculty of Medicine, İzmir, Turkey
- E.D. Batu, MD, MSc, Department of Pediatrics, Division of Rheumatology, Hacettepe University Faculty of Medicine; C. Koşukcu, MSc, Department of Medical Genetics, Hacettepe University Faculty of Medicine, and Department of Bioinformatics, Institute of Health Sciences, Hacettepe University; E. Taşkıran, PhD, Department of Medical Genetics, Hacettepe University Faculty of Medicine; S. Sahin, MD, Department of Pediatrics, Division of Rheumatology, Istanbul University Cerrahpasa Faculty of Medicine; S. Akman, MD, Department of Pediatrics, Division of Nephrology and Rheumatology, Akdeniz University Faculty of Medicine; B. Sözeri, MD, Department of Pediatrics, Division of Rheumatology, Umraniye Training and Research Center, University of Health Sciences; E. Ünsal, MD, Department of Pediatrics, Division of Rheumatology, Dokuz Eylül University Faculty of Medicine; Y. Bilginer, MD, MSc, Department of Pediatrics, Division of Rheumatology, Hacettepe University Faculty of Medicine; O. Kasapcopur, MD, Department of Pediatrics, Division of Rheumatology, Istanbul University Cerrahpasa Faculty of Medicine; M. Alikaşifoğlu, MD, PhD, Department of Medical Genetics, Hacettepe University Faculty of Medicine; S. Ozen, MD, MSc, Department of Pediatrics, Division of Rheumatology, Hacettepe University Faculty of Medicine
| | - Ozgur Kasapcopur
- From the Department of Pediatrics, Division of Rheumatology, and the Department of Medical Genetics, Hacettepe University Faculty of Medicine, Ankara; Department of Bioinformatics, Institute of Health Sciences, Hacettepe University, Ankara; Department of Pediatrics, Division of Rheumatology, Istanbul University Cerrahpasa Faculty of Medicine, Istanbul; Department of Pediatrics, Division of Nephrology and Rheumatology, Akdeniz University Faculty of Medicine, Antalya; Department of Pediatrics, Division of Rheumatology, Umraniye Training and Research Center, University of Health Sciences, Istanbul; Department of Pediatrics, Division of Rheumatology, Dokuz Eylül University Faculty of Medicine, İzmir, Turkey
- E.D. Batu, MD, MSc, Department of Pediatrics, Division of Rheumatology, Hacettepe University Faculty of Medicine; C. Koşukcu, MSc, Department of Medical Genetics, Hacettepe University Faculty of Medicine, and Department of Bioinformatics, Institute of Health Sciences, Hacettepe University; E. Taşkıran, PhD, Department of Medical Genetics, Hacettepe University Faculty of Medicine; S. Sahin, MD, Department of Pediatrics, Division of Rheumatology, Istanbul University Cerrahpasa Faculty of Medicine; S. Akman, MD, Department of Pediatrics, Division of Nephrology and Rheumatology, Akdeniz University Faculty of Medicine; B. Sözeri, MD, Department of Pediatrics, Division of Rheumatology, Umraniye Training and Research Center, University of Health Sciences; E. Ünsal, MD, Department of Pediatrics, Division of Rheumatology, Dokuz Eylül University Faculty of Medicine; Y. Bilginer, MD, MSc, Department of Pediatrics, Division of Rheumatology, Hacettepe University Faculty of Medicine; O. Kasapcopur, MD, Department of Pediatrics, Division of Rheumatology, Istanbul University Cerrahpasa Faculty of Medicine; M. Alikaşifoğlu, MD, PhD, Department of Medical Genetics, Hacettepe University Faculty of Medicine; S. Ozen, MD, MSc, Department of Pediatrics, Division of Rheumatology, Hacettepe University Faculty of Medicine
| | - Mehmet Alikaşifoğlu
- From the Department of Pediatrics, Division of Rheumatology, and the Department of Medical Genetics, Hacettepe University Faculty of Medicine, Ankara; Department of Bioinformatics, Institute of Health Sciences, Hacettepe University, Ankara; Department of Pediatrics, Division of Rheumatology, Istanbul University Cerrahpasa Faculty of Medicine, Istanbul; Department of Pediatrics, Division of Nephrology and Rheumatology, Akdeniz University Faculty of Medicine, Antalya; Department of Pediatrics, Division of Rheumatology, Umraniye Training and Research Center, University of Health Sciences, Istanbul; Department of Pediatrics, Division of Rheumatology, Dokuz Eylül University Faculty of Medicine, İzmir, Turkey
- E.D. Batu, MD, MSc, Department of Pediatrics, Division of Rheumatology, Hacettepe University Faculty of Medicine; C. Koşukcu, MSc, Department of Medical Genetics, Hacettepe University Faculty of Medicine, and Department of Bioinformatics, Institute of Health Sciences, Hacettepe University; E. Taşkıran, PhD, Department of Medical Genetics, Hacettepe University Faculty of Medicine; S. Sahin, MD, Department of Pediatrics, Division of Rheumatology, Istanbul University Cerrahpasa Faculty of Medicine; S. Akman, MD, Department of Pediatrics, Division of Nephrology and Rheumatology, Akdeniz University Faculty of Medicine; B. Sözeri, MD, Department of Pediatrics, Division of Rheumatology, Umraniye Training and Research Center, University of Health Sciences; E. Ünsal, MD, Department of Pediatrics, Division of Rheumatology, Dokuz Eylül University Faculty of Medicine; Y. Bilginer, MD, MSc, Department of Pediatrics, Division of Rheumatology, Hacettepe University Faculty of Medicine; O. Kasapcopur, MD, Department of Pediatrics, Division of Rheumatology, Istanbul University Cerrahpasa Faculty of Medicine; M. Alikaşifoğlu, MD, PhD, Department of Medical Genetics, Hacettepe University Faculty of Medicine; S. Ozen, MD, MSc, Department of Pediatrics, Division of Rheumatology, Hacettepe University Faculty of Medicine
| | - Seza Ozen
- From the Department of Pediatrics, Division of Rheumatology, and the Department of Medical Genetics, Hacettepe University Faculty of Medicine, Ankara; Department of Bioinformatics, Institute of Health Sciences, Hacettepe University, Ankara; Department of Pediatrics, Division of Rheumatology, Istanbul University Cerrahpasa Faculty of Medicine, Istanbul; Department of Pediatrics, Division of Nephrology and Rheumatology, Akdeniz University Faculty of Medicine, Antalya; Department of Pediatrics, Division of Rheumatology, Umraniye Training and Research Center, University of Health Sciences, Istanbul; Department of Pediatrics, Division of Rheumatology, Dokuz Eylül University Faculty of Medicine, İzmir, Turkey.
- E.D. Batu, MD, MSc, Department of Pediatrics, Division of Rheumatology, Hacettepe University Faculty of Medicine; C. Koşukcu, MSc, Department of Medical Genetics, Hacettepe University Faculty of Medicine, and Department of Bioinformatics, Institute of Health Sciences, Hacettepe University; E. Taşkıran, PhD, Department of Medical Genetics, Hacettepe University Faculty of Medicine; S. Sahin, MD, Department of Pediatrics, Division of Rheumatology, Istanbul University Cerrahpasa Faculty of Medicine; S. Akman, MD, Department of Pediatrics, Division of Nephrology and Rheumatology, Akdeniz University Faculty of Medicine; B. Sözeri, MD, Department of Pediatrics, Division of Rheumatology, Umraniye Training and Research Center, University of Health Sciences; E. Ünsal, MD, Department of Pediatrics, Division of Rheumatology, Dokuz Eylül University Faculty of Medicine; Y. Bilginer, MD, MSc, Department of Pediatrics, Division of Rheumatology, Hacettepe University Faculty of Medicine; O. Kasapcopur, MD, Department of Pediatrics, Division of Rheumatology, Istanbul University Cerrahpasa Faculty of Medicine; M. Alikaşifoğlu, MD, PhD, Department of Medical Genetics, Hacettepe University Faculty of Medicine; S. Ozen, MD, MSc, Department of Pediatrics, Division of Rheumatology, Hacettepe University Faculty of Medicine.
| |
Collapse
|
33
|
Anti-Double-Stranded DNA Isotypes and Anti-C1q Antibody Improve the Diagnostic Specificity of Systemic Lupus Erythematosus. DISEASE MARKERS 2018; 2018:4528547. [PMID: 30363714 PMCID: PMC6180986 DOI: 10.1155/2018/4528547] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 07/17/2018] [Accepted: 08/09/2018] [Indexed: 12/18/2022]
Abstract
Objectives We aimed to evaluate the value of immunoglobulin (Ig) G, IgM, and IgA isotypes of anti-double-stranded DNA (anti-dsDNA) and anti-C1q antibody in diagnosing systemic lupus erythematosus (SLE) patients and elucidate their association with disease activity and lupus nephritis. Methods Blood samples were obtained from 96 SLE patients, 62 other autoimmune disease patients, and 60 healthy blood donors. Anti-dsDNA IgG, IgM, and IgA isotypes and anti-C1q antibody were measured by enzyme-linked immunosorbent assay. Disease activity of SLE patients was assessed according to the SLE Disease Activity Index score. Results When specificity was greater than 90%, the sensitivity of anti-dsDNA IgG, IgM, and IgA isotypes and anti-C1q antibody in diagnosing SLE was 75%, 45%, 33%, and 49%, respectively. The prevalence of anti-dsDNA IgG (p = 0.002), anti-dsDNA IgA (p = 0.028), and anti-C1q antibody (p = 0.000) in active cases was significantly higher than those in inactive ones. In addition, the presence of anti-C1q antibody was associated with renal involvement (p = 0.032). Anti-dsDNA IgM showed no significant association with disease activity, but it was inversely linked with lupus nephritis (p = 0.005). When anti-dsDNA IgG and IgA and anti-C1q were combined to evaluate SLE disease activity, the specificity reached the highest level (90%). When anti-C1q positive was accompanied by anti-dsDNA IgM negative, the specificity of diagnosing lupus nephritis was up to 96%. Conclusions This study demonstrated the role of anti-dsDNA IgG, IgM, and IgA isotypes and anti-C1q antibody alone or combination in diagnosing SLE. Anti-dsDNA IgG and IgA and anti-C1q were shown to be associated with disease activity, while anti-dsDNA IgM and anti-C1q were associated with lupus nephritis. When the related antibodies were combined, the diagnostic specificity was significantly higher.
Collapse
|
34
|
Monogenic systemic lupus erythematosus: insights in pathophysiology. Rheumatol Int 2018; 38:1763-1775. [DOI: 10.1007/s00296-018-4048-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 05/10/2018] [Indexed: 01/02/2023]
|
35
|
Ghebrehiwet B, Kandov E, Kishore U, Peerschke EIB. Is the A-Chain the Engine That Drives the Diversity of C1q Functions? Revisiting Its Unique Structure. Front Immunol 2018; 9:162. [PMID: 29459870 PMCID: PMC5807628 DOI: 10.3389/fimmu.2018.00162] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 01/18/2018] [Indexed: 12/27/2022] Open
Abstract
The immunopathological functions associated with human C1q are still growing in terms of novelty, diversity, and pathologic relevance. It is, therefore, not surprising that C1q is being recognized as an important molecular bridge between innate and adaptive immunity. The secret of this functional diversity, in turn, resides in the elegant but complex structure of the C1q molecule, which is assembled from three distinct gene products: A, B, and C, each of which has evolved from a separate and unique ancestral gene template. The C1q molecule is made up of 6A, 6B, and 6C polypeptide chains, which are held together through strong covalent and non-covalent bonds to form the 18-chain, bouquet-of-flower-like protein that we know today. The assembled C1q protein displays at least two distinct structural and functional regions: the collagen-like region (cC1q) and the globular head region (gC1q), each being capable of driving a diverse range of ligand- or receptor-mediated biological functions. What is most intriguing, however, is the observation that most of the functions appear to be predominantly driven by the A-chain of the molecule, which begs the question: what are the evolutionary modifications or rearrangements that singularly shaped the primordial A-chain gene to become a pluripotent and versatile component of the intact C1q molecule? Here, we revisit and discuss some of the known unique structural and functional features of the A-chain, which may have contributed to its versatility.
Collapse
Affiliation(s)
- Berhane Ghebrehiwet
- Departments of Medicine, Stony Brook University, Stony Brook, NY, United States
| | - Evelyn Kandov
- Departments of Medicine, Stony Brook University, Stony Brook, NY, United States
| | - Uday Kishore
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| | - Ellinor I B Peerschke
- Department of Laboratory Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY, United States
| |
Collapse
|
36
|
Abstract
The transcription factor MafB is expressed by monocytes and macrophages. Efferocytosis (apoptotic cell uptake) by macrophages is important for inhibiting the development of autoimmune diseases, and is greatly reduced in Mafb-deficient macrophages. Here, we show the expression of the first protein in the classical complement pathway C1q is important for mediating efferocytosis and is reduced in Mafb-deficient macrophages. The efferocytosis defect in Mafb-deficient macrophages can be rescued by adding serum from wild-type mice, but not by adding serum from C1q-deficient mice. By hemolysis assay we also show that activation of the classical complement pathway is decreased in Mafb-deficient mice. In addition, MafB overexpression induces C1q-dependent gene expression and signals that induce C1q genes are less effective in the absence of MafB. We also show that Mafb-deficiency can increase glomerular autoimmunity, including anti-nuclear antibody deposition. These results show that MafB is an important regulator of C1q.
Collapse
|
37
|
Ekinci Z, Ozturk K. Systemic lupus erythematosus with C1q deficiency: treatment with fresh frozen plasma. Lupus 2017; 27:134-138. [PMID: 29113537 DOI: 10.1177/0961203317741565] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Treatment and outcome of systemic lupus erythematosus (SLE) in C1q deficient patients are rarely reported. The aim of this report is to share our experience about the course of management of three cases diagnosed as SLE with C1q deficiency, in light of present literature. Initial and dominant complaints of three cases from two different families were cutaneous manifestations. One patient was also diagnosed with arthritis and thrombocytopenia. Antinuclear antibody was positive in all cases, whereas anti-dsDNA was negative with normal levels of complement C3, C4 and decreased CH50 activity. C1QA gene of two patients had homozygous nonsense mutation (c.622 > T/p.Gln208Ter). Previously, all of them had been treated with steroids, hydroxychloroquine and methotrexate or azathioprine. It was learned that they had responded only to high dosage prednisolone and their symptoms flared up during dosage reduction even under methotrexate or azathioprine. All symptoms of all three cases improved by daily fresh frozen plasma (FFP) infusions, and once cutaneous lesions subsided, the infusions were reduced to a frequency that would prevent the flare up of the symptoms. Literature search revealed seven reports on fresh frozen plasma treatment in SLE with C1q deficient patients. In this report, it is concluded that severe cutaneous lesions, as seen in these C1q deficient SLE patients, cannot be controlled with conventional immunosuppressive treatment. Instead, regular fresh frozen plasma infusions are proposed as a more reasonable method of treatment.
Collapse
Affiliation(s)
- Z Ekinci
- 1 Kadıköy Florence Nightingale Medical Center, Istanbul, Turkey
| | - K Ozturk
- 2 Cengiz Gökçek Kadın Doğum ve Çocuk Hastalıkları Hastanesi, Gaziantep, Turkey
| |
Collapse
|
38
|
Autoinflammatory Diseases in Pediatric Dermatology–Part 2: Histiocytic, Macrophage Activation, and Vasculitis Syndromes. ACTAS DERMO-SIFILIOGRAFICAS 2017. [DOI: 10.1016/j.adengl.2017.06.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
39
|
Abstract
Recognition and removal of apoptotic and necrotic cells must be efficient and highly controlled to avoid excessive inflammation and autoimmune responses to self. The complement system, a crucial part of innate immunity, plays an important role in this process. Thus, apoptotic and necrotic cells are recognized by complement initiators such as C1q, mannose binding lectin, ficolins, and properdin. This triggers complement activation and opsonization of cells with fragments of C3b, which enhances phagocytosis and thus ensures silent removal. Importantly, the process is tightly controlled by the binding of complement inhibitors C4b-binding protein and factor H, which attenuates late steps of complement activation and inflammation. Furthermore, factor H becomes actively internalized by apoptotic cells, where it catalyzes the cleavage of intracellular C3 to C3b. The intracellularly derived C3b additionally opsonizes the cell surface further supporting safe and fast clearance and thereby aids to prevent autoimmunity. Internalized factor H also binds nucleosomes and directs monocytes into production of anti-inflammatory cytokines upon phagocytosis of such complexes. Disturbances in the complement-mediated clearance of dying cells result in persistence of autoantigens and development of autoimmune diseases like systemic lupus erythematosus, and may also be involved in development of age-related macula degeneration.
Collapse
Affiliation(s)
- Myriam Martin
- Division of Medical Protein Chemistry, Department of Translational Medicine, Lund University, Malmö, Sweden
| | - Anna M Blom
- Division of Medical Protein Chemistry, Department of Translational Medicine, Lund University, Malmö, Sweden.
| |
Collapse
|
40
|
Allogeneic Hematopoietic Stem Cell Transplantation in the Treatment of Human C1q Deficiency: The Karolinska Experience. Transplantation 2017; 100:1356-62. [PMID: 26516671 DOI: 10.1097/tp.0000000000000975] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND Human C1q deficiency is associated with systemic lupus erythematosus (SLE) and increased susceptibility to severe bacterial infections. These patients require extensive medical therapy and some develop treatment-resistant disease. Because C1q is produced by monocytes, it has been speculated that allogeneic hematopoietic stem cell transplantation (allo-HSCT) may cure this disorder. METHODS We have so far treated 5 patients with C1q deficiency. In 3 cases, SLE symptoms remained relatively mild after the start of medical therapy, but 2 patients developed treatment-resistant SLE, and we decided to pursue treatment with allo-HSCT. For this purpose, we chose a conditioning regimen composed of treosulfan (14 g/m) and fludarabine (30 mg/m) started on day -6 and given for 3 and 5 consecutive days, respectively. Thymoglobulin was given at a cumulative dose of 8 mg/kg, and graft-versus-host disease prophylaxis was composed of cyclosporine and methotrexate. RESULTS A 9-year-old boy and a 12-year-old girl with refractory SLE restored C1q production after allo-HSCT. This resulted in normal functional properties of the classical complement pathway followed by reduced severity of SLE symptoms. The boy developed posttransplant lymphoproliferative disease, which resolved after treatment with rituximab and donor lymphocyte infusion. Unfortunately, donor lymphocyte infusion induced severe cortisone-resistant gastrointestinal graft-versus-host disease, and the patient died from multiple organ failure 4 months after transplantation. The girl is doing well 33 months after transplantation, and clinically, all signs of SLE have resolved. CONCLUSIONS Allo-HSCT can cure SLE in human C1q deficiency and should be considered early in subjects resistant to medical therapy.
Collapse
|
41
|
Abstract
PURPOSE OF REVIEW We review select studies of newly discovered rare variants in autoimmune diseases with a focus on newly described monogenic disorders, rheumatoid arthritis, and systemic lupus erythematosus. RECENT FINDINGS Two new monogenic syndromes of inflammatory arthritis were discovered using whole exome sequencing: the coatomer subunit alpha syndrome because of rare mutations in coatomer subunit alpha and haploinsufficiency of A20 resulting from rare mutations in TNFAIP3. Targeted exon sequencing identified rare variants in IL2RA and IL2RB associated with rheumatoid arthritis. Rare variants in TREX1 and other genes associated with monogenic interferonopathies are also associated with systemic lupus erythematosus. SUMMARY Rare genetic variants contribute to the heritability of autoimmunity and provide key insight into both novel and previously implicated immunological pathways that are disrupted in autoimmune diseases.
Collapse
|
42
|
Hernández-Ostiz S, Xirotagaros G, Prieto-Torres L, Noguera-Morel L, Torrelo A. Autoinflammatory Diseases in Pediatric Dermatology-Part 2: Histiocytic, Macrophage Activation, and Vasculitis Syndromes. ACTAS DERMO-SIFILIOGRAFICAS 2017; 108:620-629. [PMID: 28438265 DOI: 10.1016/j.ad.2016.12.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 11/10/2016] [Accepted: 12/23/2016] [Indexed: 12/11/2022] Open
Abstract
The discovery of new autoinflammatory syndromes and novel mutations has advanced at breakneck speed in recent years. Part 2 of this review focuses on vasculitis syndromes and the group of histiocytic and macrophage activation syndromes. We also include a table showing the mutations associated with these autoinflammatory syndromes and treatment alternatives.
Collapse
Affiliation(s)
| | - G Xirotagaros
- Servicio de Dermatología, Hospital Niño Jesús, Madrid, España
| | - L Prieto-Torres
- Servicio de Dermatología, Hospital Niño Jesús, Madrid, España
| | - L Noguera-Morel
- Servicio de Dermatología, Hospital Niño Jesús, Madrid, España
| | - A Torrelo
- Servicio de Dermatología, Hospital Niño Jesús, Madrid, España.
| |
Collapse
|
43
|
Vignesh P, Rawat A, Sharma M, Singh S. Complement in autoimmune diseases. Clin Chim Acta 2017; 465:123-130. [PMID: 28040558 DOI: 10.1016/j.cca.2016.12.017] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 12/15/2016] [Accepted: 12/17/2016] [Indexed: 12/18/2022]
Abstract
The complement system is an ancient and evolutionary conserved element of the innate immune mechanism. It comprises of more than 20 serum proteins most of which are synthesized in the liver. These proteins are synthesized as inactive precursor proteins which are activated by appropriate stimuli. The activated forms of these proteins act as proteases and cleave other components successively in amplification pathways leading to exponential generation of final effectors. Three major pathways of complement pathways have been described, namely the classical, alternative and lectin pathways which are activated by different stimuli. However, all the 3 pathways converge on Complement C3. Cleavage of C3 and C5 successively leads to the production of the membrane attack complex which is final common effector. Excessive and uncontrolled activation of the complement has been implicated in the host of autoimmune diseases. But the complement has also been bemusedly described as the proverbial "double edged sword". On one hand, complement is the final effector of tissue injury in autoimmune diseases and on the other, deficiencies of some components of the complement can result in autoimmune diseases. Currently available tools such as enzyme based immunoassays for functional assessment of complement pathways, flow cytometry, next generation sequencing and proteomics-based approaches provide an exciting opportunity to study this ancient yet mysterious element of innate immunity.
Collapse
Affiliation(s)
- Pandiarajan Vignesh
- Pediatric Allergy and Immunology Unit, Dept. of Pediatrics, Advanced Pediatrics Centre, PGIMER, Chandigarh, India
| | - Amit Rawat
- Pediatric Allergy and Immunology Unit, Dept. of Pediatrics, Advanced Pediatrics Centre, PGIMER, Chandigarh, India.
| | - Madhubala Sharma
- Pediatric Allergy and Immunology Unit, Dept. of Pediatrics, Advanced Pediatrics Centre, PGIMER, Chandigarh, India
| | - Surjit Singh
- Pediatric Allergy and Immunology Unit, Dept. of Pediatrics, Advanced Pediatrics Centre, PGIMER, Chandigarh, India
| |
Collapse
|
44
|
Nanthapisal S, Omoyinmi E, Murphy C, Standing A, Eisenhut M, Eleftheriou D, Brogan PA. Early-Onset Juvenile SLE Associated With a Novel Mutation in Protein Kinase C δ. Pediatrics 2017; 139:peds.2016-0781. [PMID: 28003329 DOI: 10.1542/peds.2016-0781] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/06/2016] [Indexed: 11/24/2022] Open
Abstract
Juvenile systemic lupus erythematosus (jSLE) is rare before 5 years of age. Monogenic causes are suspected in cases of very early onset jSLE particularly in the context of a family history and/or consanguinity. We performed whole-exome sequencing and homozygosity mapping in the siblings presented with early-onset jSLE. A novel homozygous missense mutation in protein kinase C delta (c.1294G>T; p.Gly432Trp) was identified in both patients. One patient showed a marked clinical response and resolution inflammation with rituximab therapy. This report demonstrates the clinical importance of identifying monogenic causes of rare disease to provide a definitive diagnosis, help rationalize treatment, and facilitate genetic counseling.
Collapse
Affiliation(s)
- Sira Nanthapisal
- Infection, Inflammation, and Rheumatology Section, Infection, Immunity, Inflammation and Physiological Medicine Programme, UCL Institute of Child Health, London, United Kingdom;
| | - Ebun Omoyinmi
- Infection, Inflammation, and Rheumatology Section, Infection, Immunity, Inflammation and Physiological Medicine Programme, UCL Institute of Child Health, London, United Kingdom
| | - Claire Murphy
- Infection, Inflammation, and Rheumatology Section, Infection, Immunity, Inflammation and Physiological Medicine Programme, UCL Institute of Child Health, London, United Kingdom
| | - Ariane Standing
- Infection, Inflammation, and Rheumatology Section, Infection, Immunity, Inflammation and Physiological Medicine Programme, UCL Institute of Child Health, London, United Kingdom
| | - Michael Eisenhut
- Luton & Dunstable University Hospital NHS Foundation Trust, Luton, United Kingdom
| | - Despina Eleftheriou
- Infection, Inflammation, and Rheumatology Section, Infection, Immunity, Inflammation and Physiological Medicine Programme, UCL Institute of Child Health, London, United Kingdom
| | - Paul A Brogan
- Infection, Inflammation, and Rheumatology Section, Infection, Immunity, Inflammation and Physiological Medicine Programme, UCL Institute of Child Health, London, United Kingdom
| |
Collapse
|
45
|
van Schaarenburg RA, Magro-Checa C, Bakker JA, Teng YKO, Bajema IM, Huizinga TW, Steup-Beekman GM, Trouw LA. C1q Deficiency and Neuropsychiatric Systemic Lupus Erythematosus. Front Immunol 2016; 7:647. [PMID: 28082982 PMCID: PMC5186770 DOI: 10.3389/fimmu.2016.00647] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 12/13/2016] [Indexed: 12/25/2022] Open
Abstract
C1q deficiency is a rare immunodeficiency, which is strongly associated with the development of systemic lupus erythematosus (SLE). A mutation in one of the C1q genes can either lead to complete deficiency or to low C1q levels with C1q polypeptide in the form of low-molecular weight (LMW) C1q. Patients with C1q deficiency mainly present with cutaneous and renal involvement. Although less frequent, neuropsychiatric (NP) involvement has also been reported in 20% of the C1q-deficient patients. This involvement appears to be absent in other deficiencies of early components of the complement classical pathway (CP) (C1r/C1s, C2, or C4 deficiencies). We describe a new case with C1q deficiency with a homozygous G34R mutation in C1qC-producing LMW-C1q presenting with a severe SLE flare with NP involvement. The serum of this patient contained very low levels of a LMW variant of C1q polypeptides. Cell lysates contained the three chains of C1q, but no intact C1q was detected, consistent with the hypothesis of the existence of a LMW-C1q. Furthermore, we provide a literature overview of NP-SLE in C1q deficiency and hypothesize about the potential role of C1q in the pathogenesis of NP involvement in these patients. The onset of NP-SLE in C1q-deficient individuals is more severe when compared with complement competent NP-SLE patients. An important number of cases present with seizures and the most frequent findings in neuroimaging are changes in basal ganglia and cerebral vasculitis. A defective CP, because of non-functional C1q, does not protect against NP involvement in SLE. The absence of C1q and, subsequently, some of its biological functions may be associated with more severe NP-SLE.
Collapse
Affiliation(s)
| | - César Magro-Checa
- Department of Rheumatology, Leiden University Medical Center , Leiden , Netherlands
| | - Jaap A Bakker
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center , Leiden , Netherlands
| | - Y K Onno Teng
- Department of Nephrology, Leiden University Medical Center , Leiden , Netherlands
| | - Ingeborg M Bajema
- Department of Pathology, Leiden University Medical Center , Leiden , Netherlands
| | - Tom W Huizinga
- Department of Rheumatology, Leiden University Medical Center , Leiden , Netherlands
| | | | - Leendert A Trouw
- Department of Rheumatology, Leiden University Medical Center , Leiden , Netherlands
| |
Collapse
|
46
|
Hui-Yuen JS, Zhu L, Wong LP, Jiang K, Chen Y, Liu T, Jarvis JN. Chromatin landscapes and genetic risk in systemic lupus. Arthritis Res Ther 2016; 18:281. [PMID: 27906046 PMCID: PMC5134118 DOI: 10.1186/s13075-016-1169-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 11/02/2016] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) is a multi-system, complex disease in which the environment interacts with inherited genes to produce broad phenotypes with inter-individual variability. Of 46 single nucleotide polymorphisms (SNPs) shown to confer genetic risk for SLE in recent genome-wide association studies, 30 lie within noncoding regions of the human genome. We therefore sought to identify and describe the functional elements (aside from genes) located within these regions of interest. METHODS We used chromatin immunoprecipitation followed by sequencing to identify epigenetic marks associated with enhancer function in adult neutrophils to determine whether enhancer-associated histone marks were enriched within the linkage disequilibrium (LD) blocks encompassing the 46 SNPs of interest. We also interrogated available data in Roadmap Epigenomics for CD4+ T cells and CD19+ B cells to identify these same elements in lymphoid cells. RESULTS All three cell types demonstrated enrichment of enhancer-associated histone marks compared with genomic background within LD blocks encoded by SLE-associated SNPs. In addition, within the promoter regions of these LD blocks, all three cell types demonstrated enrichment for transcription factor binding sites above genomic background. In CD19+ B cells, all but one of the LD blocks of interest were also enriched for enhancer-associated histone marks. CONCLUSIONS Much of the genetic risk for SLE lies within or near genomic regions of disease-relevant cells that are enriched for epigenetic marks associated with enhancer function. Elucidating the specific roles of these noncoding elements within these cell-type-specific genomes will be crucial to our understanding of SLE pathogenesis.
Collapse
Affiliation(s)
- Joyce S. Hui-Yuen
- Division of Pediatric Rheumatology, Steven and Alexandra Cohen Children’s Medical Center, 1991 Marcus Avenue, Suite M100, Lake Success, NY 11042 USA
- Department of Pediatrics, Hofstra-Northwell School of Medicine, Hempstead, NY 11549 USA
| | - Lisha Zhu
- Department of Biochemistry, University at Buffalo, Buffalo, NY 14203 USA
| | - Lai Ping Wong
- Department of Pediatrics, University at Buffalo, Buffalo, NY 14203 USA
| | - Kaiyu Jiang
- Department of Pediatrics, University at Buffalo, Buffalo, NY 14203 USA
| | - Yanmin Chen
- Department of Pediatrics, University at Buffalo, Buffalo, NY 14203 USA
| | - Tao Liu
- Department of Biochemistry, and Genetics, Genomics, and Bioinformatics Program, University at Buffalo, Buffalo, NY 14203 USA
| | - James N. Jarvis
- Genetics, Genomics, and Bioinformatics Program, University at Buffalo, Buffalo, NY 14203 USA
| |
Collapse
|
47
|
C1q as an autocrine and paracrine regulator of cellular functions. Mol Immunol 2016; 84:26-33. [PMID: 27914690 DOI: 10.1016/j.molimm.2016.11.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Revised: 11/01/2016] [Accepted: 11/03/2016] [Indexed: 12/20/2022]
Abstract
Most of the complement proteins in circulation are, by and large, synthesized in the liver. However data accumulated over the past several decades provide incontrovertible evidence that some if not most of the individual complement proteins are also synthesized extrahepatically by activated as well as non-activated cells. The question that is finally being addressed by various investigators is: are the locally synthesized proteins solely responsible for the myriad of biological functions in situ without the contribution of systemic complement? The answer is probably "yes". Among the proteins that are synthesized locally, C1q takes center stage for several reasons. First, it is synthesized predominantly by potent antigen presenting cells such as monocytes, macrophages and dendritic cells (DCs), which by itself is a clue that it plays an important role in antigen presentation and/or DC maturation. Second, it is transiently anchored on the cell surface via a transmembrane domain located in its A chain before it is cleaved off and released into the pericellular milieu. The membrane-associated C1q in turn, is able to sense danger patterns via its versatile antigen-capturing globular head domains. More importantly, locally synthesized C1q has been shown to induce a plethora of biological functions through the induction of immunomodulatory molecules by an autocrine- or paracrine- mediated signaling in a manner that mimics those of TNFα. These include recognition of pathogen- and danger- associated molecular patterns, phagocytosis, angiogenesis, apoptosis and induction of cytokines or chemokines that are important in modulating the inflammatory response. The functional convergence between C1q and TNFα in turn is attributed to their shared genetic ancestry. In this paper, we will infer to the aforementioned "local-synthesis-for-local function" paradigm using as an example, the role played by locally synthesized C1q in autoimmunity in general and in systemic lupus erythematosus in particular.
Collapse
|
48
|
Xirotagaros G, Hernández-Ostiz S, Aróstegui JI, Torrelo A. Newly Described Autoinflammatory Diseases in Pediatric Dermatology. Pediatr Dermatol 2016; 33:602-614. [PMID: 27699831 DOI: 10.1111/pde.12984] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Specific gene mutations leading to dysregulation of innate immune response produce the expanding spectrum of monogenic autoinflammatory diseases (AIDs). They are characterized by seemingly unprovoked, recurrent episodes of systemic inflammation in which a myriad of manifestations usually affect skin. Novel genetic technologies have led to the discovery of new AIDs and phenotypes that were not previously clinically described. Consequently the number of AIDs is continuously growing and their recognition and the disclosure of their pathophysiology will prompt early diagnosis and targeted treatment of affected patients. The objective of the present work is to review those newly described AIDs with prominent dermatologic manifestations that may constitute a major criterion for their diagnosis.
Collapse
Affiliation(s)
| | | | | | - Antonio Torrelo
- Department of Dermatology, Hospital Infantil del Niño Jesús, Madrid, Spain
| |
Collapse
|
49
|
Goilav B, Putterman C, Rubinstein TB. Biomarkers for kidney involvement in pediatric lupus. Biomark Med 2016; 9:529-43. [PMID: 26079958 DOI: 10.2217/bmm.15.25] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Lupus nephritis (LN), the renal involvement in systemic lupus erythematosus, is currently diagnosed by histopathology obtained by percutaneous renal biopsy and is associated with increased morbidity and mortality in both adults and children. LN is more prevalent and severe in children, requiring aggressive and prolonged immunosuppression. The consequences of the diagnosis and its treatment have devastating long-term effects on the growth, well-being and quality of life of affected children. The paucity of reliable clinical indicators of the presence and severity of renal involvement have contributed to a halt in the reduction of progression to end-stage renal disease in recent years. Here, we discuss the recent development of biomarkers in the management of LN and their role as therapeutic targets.
Collapse
Affiliation(s)
- Beatrice Goilav
- Children's Hospital at Montefiore, Department of Pediatrics, Division of Nephrology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, New York, NY 10461, USA
| | - Chaim Putterman
- Division of Rheumatology & Department of Microbiology & Immunology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, New York, NY 10461, USA
| | - Tamar B Rubinstein
- Children's Hospital at Montefiore, Department of Pediatrics, Division of Rheumatology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, New York, NY 10461, USA
| |
Collapse
|
50
|
Lintner KE, Wu YL, Yang Y, Spencer CH, Hauptmann G, Hebert LA, Atkinson JP, Yu CY. Early Components of the Complement Classical Activation Pathway in Human Systemic Autoimmune Diseases. Front Immunol 2016; 7:36. [PMID: 26913032 PMCID: PMC4753731 DOI: 10.3389/fimmu.2016.00036] [Citation(s) in RCA: 115] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 01/24/2016] [Indexed: 01/06/2023] Open
Abstract
The complement system consists of effector proteins, regulators, and receptors that participate in host defense against pathogens. Activation of the complement system, via the classical pathway (CP), has long been recognized in immune complex-mediated tissue injury, most notably systemic lupus erythematosus (SLE). Paradoxically, a complete deficiency of an early component of the CP, as evidenced by homozygous genetic deficiencies reported in human, are strongly associated with the risk of developing SLE or a lupus-like disease. Similarly, isotype deficiency attributable to a gene copy-number (GCN) variation and/or the presence of autoantibodies directed against a CP component or a regulatory protein that result in an acquired deficiency are relatively common in SLE patients. Applying accurate assay methodologies with rigorous data validations, low GCNs of total C4, and heterozygous and homozygous deficiencies of C4A have been shown as medium to large effect size risk factors, while high copy numbers of total C4 or C4A as prevalent protective factors, of European and East-Asian SLE. Here, we summarize the current knowledge related to genetic deficiency and insufficiency, and acquired protein deficiencies for C1q, C1r, C1s, C4A/C4B, and C2 in disease pathogenesis and prognosis of SLE, and, briefly, for other systemic autoimmune diseases. As the complement system is increasingly found to be associated with autoimmune diseases and immune-mediated diseases, it has become an attractive therapeutic target. We highlight the recent developments and offer a balanced perspective concerning future investigations and therapeutic applications with a focus on early components of the CP in human systemic autoimmune diseases.
Collapse
Affiliation(s)
- Katherine E Lintner
- Center for Molecular and Human Genetics, Division of Pediatric Rheumatology, Department of Pediatrics, Nationwide Children's Hospital, The Ohio State University , Columbus, OH , USA
| | - Yee Ling Wu
- Center for Molecular and Human Genetics, Division of Pediatric Rheumatology, Department of Pediatrics, Nationwide Children's Hospital, The Ohio State University , Columbus, OH , USA
| | - Yan Yang
- Center for Molecular and Human Genetics, Division of Pediatric Rheumatology, Department of Pediatrics, Nationwide Children's Hospital, The Ohio State University , Columbus, OH , USA
| | - Charles H Spencer
- Center for Molecular and Human Genetics, Division of Pediatric Rheumatology, Department of Pediatrics, Nationwide Children's Hospital, The Ohio State University , Columbus, OH , USA
| | - Georges Hauptmann
- Laboratoire d'Immuno-Rhumatologie Moleculaire, INSERM UMR_S 1109, LabEx Transplantex, Faculté de Médecine, Université de Strasbourg , Strasbourg , France
| | - Lee A Hebert
- Division of Nephrology, College of Medicine, The Ohio State University , Columbus, OH , USA
| | - John P Atkinson
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine , St. Louis, MO , USA
| | - C Yung Yu
- Center for Molecular and Human Genetics, Division of Pediatric Rheumatology, Department of Pediatrics, Nationwide Children's Hospital, The Ohio State University , Columbus, OH , USA
| |
Collapse
|