1
|
Kreutzberger MAB, Yu LT, Hancu MC, Purdy MD, Osinski T, Kasson P, Egelman EH, Hartgerink JD. A Collagen Triple Helix without the Super Helical Twist. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.26.615199. [PMID: 39386471 PMCID: PMC11463431 DOI: 10.1101/2024.09.26.615199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Collagens are ubiquitous in biology functioning as the backbone of the extracellular matrix, forming the primary structural components of key immune system complexes, and fulfilling numerous other structural roles in a variety of systems. Despite this, there is limited understanding of how triple helices, the basic collagen structural units, pack into collagenous assemblies. Here we use a peptide self-assembly system to design collagenous assemblies based on the C1q collagen-like region. Using cryo-EM we solve a structure of one assembly to 3.5 Å resolution and build an atomic model. From this, we identify a triple helix conformation with no superhelical twist, starkly in contrast to the canonical right-handed triple helix. This non-twisting region allows for unique hydroxyproline stacking between adjacent triple helices and also results in the formation of an exposed cavity with rings of hydrophobic amino acids packed symmetrically. We find no precedent for such an arrangement of collagen triple helices and have designed mutant assemblies to probe key stabilizing amino acid interactions in the complex. The mutations behave as predicted by our atomic model. Our findings, combined with the extremely limited experimental structural data on triple helix packing in the literature, suggest that collagen and collagen-like assemblies may adopt a far more varied conformational landscape than previously appreciated. We hypothesize that this is particularly likely adjacent to the termini of these helices and at discontinuities to the required Xaa-Yaa-Gly repeating primary sequence; a discontinuity found in the majority of this class of proteins and in many collagen-associated diseases.
Collapse
Affiliation(s)
- Mark A. B. Kreutzberger
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA, 22903, USA
| | - Le Tracy Yu
- Department of Chemistry, Rice University, 6100 Main Street, Houston, TX, 77005, USA
| | - Maria C. Hancu
- Department of Chemistry, Rice University, 6100 Main Street, Houston, TX, 77005, USA
| | - Michael D. Purdy
- Molecular Electron Microscopy Core, University of Virginia School of Medicine, Charlottesville, VA, 22903, USA
| | - Tomasz Osinski
- Center for Advanced Research Computing, University of Southern California, Los Angeles, CA 90089
| | - Peter Kasson
- Departments of Chemistry & Biochemistry and Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332
| | - Edward H. Egelman
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA, 22903, USA
| | - Jeffrey D. Hartgerink
- Department of Chemistry, Rice University, 6100 Main Street, Houston, TX, 77005, USA
- Department of Bioengineering, Rice University, 6100 Main Street, Houston, TX, 77005, USA
| |
Collapse
|
2
|
Bhattad S, Singh N, Janardhanan J, Kumar H, Ali SMN, Arigela K, Kundaragi N, Vidyashankar P, Kotecha U, Ginigeri C. Profile of juvenile systemic lupus erythematosus patients with a special reference to monogenic lupus and lupus nephritis: a cross-sectional study. Rheumatol Int 2024:10.1007/s00296-024-05696-0. [PMID: 39180524 DOI: 10.1007/s00296-024-05696-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 08/09/2024] [Indexed: 08/26/2024]
Abstract
To study the clinical, laboratory profile and outcome of juvenile Systemic Lupus Erythematosus (jSLE) patients at a tertiary care centre in South India. A retrospective review of the medical records of all jSLE patients visiting the Pediatric Immunology and Rheumatology Unit, Aster CMI Hospital, India from February 2017 to December 2023 was performed. The clinical characteristics, treatment and outcomes were recorded and tabulated. Seventy patients diagnosed with jSLE were included in the study. The female-to-male ratio was 4.4:1. Mean age at onset and delay in diagnosis were 120.1 (+/- 56.8) and 11.7 (+/- 22.7) months respectively. The median follow-up period was 13 months (range 4, 29 months). Nine patients presented with early onset SLE (< 5 years). Most common manifestations were constitutional symptoms (n = 56), followed by haematologic (n = 55), and mucocutaneous(n = 50) involvement. Immunological workup showed SLE-specific antibody positivity in 38 patients, hypocomplementemia in 40 patients, and anti-phospholipid antibody positivity in 13 patients. Mortality was observed in five patients with LN while there was no mortality in the non-nephritis group (p 0.004). C1q deficiency was the most common cause of monogenic lupus seen in 5/9 patients; protein kinase C delta (PRKCD) defect and chronic granulomatous disease (CYBB mutation) were seen in one patient each. We describe a large cohort of jSLE from Southern India. Lupus nephritis was noted in 35.7% of our cohort and had a direct correlation with mortality. 10% of patients had monogenic lupus. Serious infections were more frequent in patients with monogenic lupus.
Collapse
Affiliation(s)
- Sagar Bhattad
- Paediatric Immunology and Rheumatology Unit, Aster CMI Hospital, Bangalore, India.
| | - Neha Singh
- Paediatric Immunology and Rheumatology Unit, Aster CMI Hospital, Bangalore, India
| | - Jyothi Janardhanan
- Paediatric Immunology and Rheumatology Unit, Aster CMI Hospital, Bangalore, India
| | - Harish Kumar
- Pediatric Intensive Care Unit, Department of Paediatrics, Aster CMI Hospital, Bengaluru, India
| | | | - Karthik Arigela
- Pediatric Intensive Care Unit, Department of Paediatrics, Aster CMI Hospital, Bengaluru, India
| | | | - P Vidyashankar
- Department of Nephrology, Aster CMI Hospital, Bengaluru, India
| | | | - Chetan Ginigeri
- Pediatric Intensive Care Unit, Department of Paediatrics, Aster CMI Hospital, Bengaluru, India
| |
Collapse
|
3
|
St. Louis BM, Quagliato SM, Su YT, Dyson G, Lee PC. The Hippo kinases control inflammatory Hippo signaling and restrict bacterial infection in phagocytes. mBio 2024; 15:e0342923. [PMID: 38624208 PMCID: PMC11078001 DOI: 10.1128/mbio.03429-23] [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: 12/20/2023] [Accepted: 03/22/2024] [Indexed: 04/17/2024] Open
Abstract
The Hippo kinases MST1 and MST2 initiate a highly conserved signaling cascade called the Hippo pathway that limits organ size and tumor formation in animals. Intriguingly, pathogens hijack this host pathway during infection, but the role of MST1/2 in innate immune cells against pathogens is unclear. In this report, we generated Mst1/2 knockout macrophages to investigate the regulatory activities of the Hippo kinases in immunity. Transcriptomic analyses identified differentially expressed genes (DEGs) regulated by MST1/2 that are enriched in biological pathways, such as systemic lupus erythematosus, tuberculosis, and apoptosis. Surprisingly, pharmacological inhibition of the downstream components LATS1/2 in the canonical Hippo pathway did not affect the expression of a set of immune DEGs, suggesting that MST1/2 control these genes via alternative inflammatory Hippo signaling. Moreover, MST1/2 may affect immune communication by influencing the release of cytokines, including TNFα, CXCL10, and IL-1ra. Comparative analyses of the single- and double-knockout macrophages revealed that MST1 and MST2 differentially regulate TNFα release and expression of the immune transcription factor MAF, indicating that the two homologous Hippo kinases individually play a unique role in innate immunity. Notably, both MST1 and MST2 can promote apoptotic cell death in macrophages upon stimulation. Lastly, we demonstrate that the Hippo kinases are critical factors in mammalian macrophages and single-cell amoebae to restrict infection by Legionella pneumophila, Escherichia coli, and Pseudomonas aeruginosa. Together, these results uncover non-canonical inflammatory Hippo signaling in macrophages and the evolutionarily conserved role of the Hippo kinases in the anti-microbial defense of eukaryotic hosts. IMPORTANCE Identifying host factors involved in susceptibility to infection is fundamental for understanding host-pathogen interactions. Clinically, individuals with mutations in the MST1 gene which encodes one of the Hippo kinases experience recurrent infection. However, the impact of the Hippo kinases on innate immunity remains largely undetermined. This study uses mammalian macrophages and free-living amoebae with single- and double-knockout in the Hippo kinase genes and reveals that the Hippo kinases are the evolutionarily conserved determinants of host defense against microbes. In macrophages, the Hippo kinases MST1 and MST2 control immune activities at multiple levels, including gene expression, immune cell communication, and programmed cell death. Importantly, these activities controlled by MST1 and MST2 in macrophages are independent of the canonical Hippo cascade that is known to limit tissue growth and tumor formation. Together, these findings unveil a unique inflammatory Hippo signaling pathway that plays an essential role in innate immunity.
Collapse
Affiliation(s)
- Brendyn M. St. Louis
- Department of Biological Sciences, College of Liberal Arts and Sciences, Wayne State University, Detroit, Michigan, USA
| | - Sydney M. Quagliato
- Department of Biological Sciences, College of Liberal Arts and Sciences, Wayne State University, Detroit, Michigan, USA
| | - Yu-Ting Su
- Department of Biological Sciences, College of Liberal Arts and Sciences, Wayne State University, Detroit, Michigan, USA
| | - Gregory Dyson
- Department of Oncology, School of Medicine, Wayne State University, Detroit, Michigan, USA
| | - Pei-Chung Lee
- Department of Biological Sciences, College of Liberal Arts and Sciences, Wayne State University, Detroit, Michigan, USA
| |
Collapse
|
4
|
Ramanathan S, Brilot F, Irani SR, Dale RC. Origins and immunopathogenesis of autoimmune central nervous system disorders. Nat Rev Neurol 2023; 19:172-190. [PMID: 36788293 DOI: 10.1038/s41582-023-00776-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/13/2023] [Indexed: 02/16/2023]
Abstract
The field of autoimmune neurology is rapidly evolving, and recent discoveries have advanced our understanding of disease aetiologies. In this article, we review the key pathogenic mechanisms underlying the development of CNS autoimmunity. First, we review non-modifiable risk factors, such as age, sex and ethnicity, as well as genetic factors such as monogenic variants, common variants in vulnerability genes and emerging HLA associations. Second, we highlight how interactions between environmental factors and epigenetics can modify disease onset and severity. Third, we review possible disease mechanisms underlying triggers that are associated with the loss of immune tolerance with consequent recognition of self-antigens; these triggers include infections, tumours and immune-checkpoint inhibitor therapies. Fourth, we outline how advances in our understanding of the anatomy of lymphatic drainage and neuroimmune interfaces are challenging long-held notions of CNS immune privilege, with direct relevance to CNS autoimmunity, and how disruption of B cell and T cell tolerance and the passage of immune cells between the peripheral and intrathecal compartments have key roles in initiating disease activity. Last, we consider novel therapeutic approaches based on our knowledge of the immunopathogenesis of autoimmune CNS disorders.
Collapse
Affiliation(s)
- Sudarshini Ramanathan
- Translational Neuroimmunology Group, Kids Neuroscience Centre, Children's Hospital at Westmead, Sydney, New South Wales, Australia
- Sydney Medical School, Faculty of Medicine and Health and Brain and Mind Centre, University of Sydney, Sydney, New South Wales, Australia
- Department of Neurology, Concord Hospital, Sydney, New South Wales, Australia
| | - Fabienne Brilot
- Translational Neuroimmunology Group, Kids Neuroscience Centre, Children's Hospital at Westmead, Sydney, New South Wales, Australia
- School of Medical Science, Faculty of Medicine and Health and Brain and Mind Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Sarosh R Irani
- Oxford Autoimmune Neurology Group, Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Russell C Dale
- Translational Neuroimmunology Group, Kids Neuroscience Centre, Children's Hospital at Westmead, Sydney, New South Wales, Australia.
- Sydney Medical School, Faculty of Medicine and Health and Brain and Mind Centre, University of Sydney, Sydney, New South Wales, Australia.
- TY Nelson Department of Paediatric Neurology, Children's Hospital Westmead, Sydney, New South Wales, Australia.
| |
Collapse
|
5
|
Govender S, Nayak NR, Nandlal L, Naicker T. Gene polymorphisms within regions of complement component C1q in HIV associated preeclampsia. Eur J Obstet Gynecol Reprod Biol 2023; 282:133-139. [PMID: 36716536 DOI: 10.1016/j.ejogrb.2023.01.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 01/24/2023] [Indexed: 01/26/2023]
Abstract
OBJECTIVE This study investigates the association of C1q gene (rs292001 and rs294183) polymorphisms in HIV infected and uninfected preeclamptic women of African ancestry. MATERIALS AND METHODS The study population consisted of 325 pregnant women of African ancestry grouped into 145 normotensive pregnant women (72 HIV uninfected normotensive, 73 HIV infected normotensive) and 180 preeclamptic pregnant women (103 HIV uninfected preeclamptics, 77 HIV infected preeclamptics). Preeclamptic pregnant women were further sub-grouped into 79 early-onset preeclampsia (EOPE) (40 HIV uninfected EOPE, 39 HIV infected EOPE) and 101 late-onset preeclampsia (LOPE) (63 HIV uninfected LOPE, 38 HIV infected LOPE). Genotyping of complement C1q gene polymorphisms (rs292001 and rs294183) was detected using a TaqMan® SNP Genotyping assay from purified DNA. RESULTS No significant differences in allelic and genotype frequencies of rs292001 and rs294183 between preeclamptic and normotensive women were observed. Likewise, there were no significant differences in allelic and genotype frequencies between HIV infected normotensive vs HIV infected preeclampsia and HIV uninfected normotensive vs HIV uninfected preeclampsia for both SNPs. However, the odds ratio of preeclamptic women having the GA genotype was 1:2. CONCLUSION We demonstrate that SNPs of the C1q gene (rs292001 and rs294183) are not associated with the pathogenesis of PE development in women of African ancestry. The role ofC1qrs292001 heterozygous GA is highlighted (with and without HIV infection) may affect susceptibility to PE development. Notably, this dysregulation may affect C1q translation and protein output thus influencing the downstream role of the complement system and functional immunology in HIV infection comorbid with PE.
Collapse
Affiliation(s)
- Sumeshree Govender
- Optics and Imaging Centre, Doris Duke Medical Research Institute, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa.
| | - Nihar R Nayak
- Department of Obstetrics and Gynaecology, School of Medicine, University of Missouri, Kansas City, United States
| | - Louansha Nandlal
- Optics and Imaging Centre, Doris Duke Medical Research Institute, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Thajasvarie Naicker
- Optics and Imaging Centre, Doris Duke Medical Research Institute, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa.
| |
Collapse
|
6
|
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: 0] [Impact Index Per Article: 0] [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
|
7
|
Weisnicht AM, Byrne R, Henkel EB, Harding SA, Kostelyna SP, Schady D, Lai J, Stubbs LA. Recurrent Rash in an 11-Year-Old Boy With Pericardial and Pleural Effusions. Pediatrics 2022; 150:189805. [PMID: 36305212 DOI: 10.1542/peds.2021-055524] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/07/2022] [Indexed: 11/06/2022] Open
Abstract
An 11-year-old, previously healthy boy presented to the emergency center (EC) for acute respiratory distress in the setting of 5 months of recurrent and worsening rash with progressive fatigue, shortness of breath, chest pain, and cough. At the onset of his rash, he and his younger brothers were diagnosed with roseola. Although his brothers' symptoms resolved, the patient's rash recurred, prompting his primary care provider to prescribe amoxicillin. The rash subsequently worsened, so amoxicillin was stopped; a prednisone course was prescribed which alleviated the rash. Upon completion of the prednisone course, the rash returned more diffusely with associated symptoms of shortness of breath, chest pain, and cough. Because of these symptoms, his mother brought him to the EC, where his vitals were notable for tachypnea and tachycardia. His initial EC imaging workup was remarkable for an echocardiogram with a mild to moderate circumferential pericardial effusion, chest x-ray (CXR) with a large right pleural effusion, and chest computerized tomography significant for prominent and diffuse mediastinal and hilar lymphadenopathy with numerous enlarged axillary lymph nodes. Laboratory results were notable for elevated liver enzymes, inflammatory markers, d-dimer, and brain natriuretic peptide. Differential diagnosis remained broad, including infectious, oncologic, and rheumatologic etiologies. Our panel of experts reviews the evaluation, hospital course, and treatment of this patient presenting with an unusual rash and serositis.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jamie Lai
- Pediatric Rheumatology, Baylor College of Medicine and Texas Children's Hospital, Houston, Texas
| | - Leigh A Stubbs
- Pediatric Rheumatology, Baylor College of Medicine and Texas Children's Hospital, Houston, Texas
| |
Collapse
|
8
|
Schulz K, Trendelenburg M. C1q as a target molecule to treat human disease: What do mouse studies teach us? Front Immunol 2022; 13:958273. [PMID: 35990646 PMCID: PMC9385197 DOI: 10.3389/fimmu.2022.958273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 06/24/2022] [Indexed: 11/17/2022] Open
Abstract
The complement system is a field of growing interest for pharmacological intervention. Complement protein C1q, the pattern recognition molecule at the start of the classical pathway of the complement cascade, is a versatile molecule with additional non-canonical actions affecting numerous cellular processes. Based on observations made in patients with hereditary C1q deficiency, C1q is protective against systemic autoimmunity and bacterial infections. Accordingly, C1q deficient mice reproduce this phenotype with susceptibility to autoimmunity and infections. At the same time, beneficial effects of C1q deficiency on disease entities such as neurodegenerative diseases have also been described in murine disease models. This systematic review provides an overview of all currently available literature on the C1q knockout mouse in disease models to identify potential target diseases for treatment strategies focusing on C1q, and discusses potential side-effects when depleting and/or inhibiting C1q.
Collapse
Affiliation(s)
- Kristina Schulz
- Laboratory of Clinical Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
- Division of Internal Medicine, University Hospital Basel, Basel, Switzerland
- *Correspondence: Kristina Schulz,
| | - Marten Trendelenburg
- Laboratory of Clinical Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
- Division of Internal Medicine, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
9
|
Trzeciak A, Mongre RK, Kim MR, Lim K, Madero RA, Parkhurst CN, Pietropaoli AP, Kim M. Neutrophil heterogeneity in complement C1q expression associated with sepsis mortality. Front Immunol 2022; 13:965305. [PMID: 35983035 PMCID: PMC9380571 DOI: 10.3389/fimmu.2022.965305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 07/08/2022] [Indexed: 11/13/2022] Open
Abstract
Sepsis is a life-threatening systemic inflammatory condition causing approximately 11 million annual deaths worldwide. Although key hyperinflammation-based organ dysfunctions that drive disease pathology have been recognized, our understanding of the factors that predispose patients to septic mortality is limited. Due to the lack of reliable prognostic measures, the development of appropriate clinical management that improves patient survival remains challenging. Here, we discovered that a subpopulation of CD49chigh neutrophils with dramatic upregulation of the complement component 1q (C1q) gene expression arises during severe sepsis. We further found that deceased septic patients failed to maintain C1q protein expression in their neutrophils, whereas septic survivors expressed higher levels of C1q. In mouse sepsis models, blocking C1q with neutralizing antibodies or conditionally knocking out C1q in neutrophils led to a significant increase in septic mortality. Apoptotic neutrophils release C1q to control their own clearance in critically injured organs during sepsis; thus, treatment of septic mice with C1q drastically increased survival. These results suggest that neutrophil C1q is a reliable prognostic biomarker of septic mortality and a potential novel therapeutic target for the treatment of sepsis.
Collapse
Affiliation(s)
- Alissa Trzeciak
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY, United States
| | - Raj Kumar Mongre
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY, United States
| | - Ma Rie Kim
- Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY, United States
| | - Kihong Lim
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY, United States
| | - Rafael A. Madero
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY, United States
| | - Christopher N. Parkhurst
- Division of Pulmonary and Critical Care Medicine, Weill-Cornell Medicine, New York, NY, United States
| | - Anthony P. Pietropaoli
- Pulmonary and Critical Care Medicine Division, University of Rochester, Rochester, NY, United States
| | - Minsoo Kim
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY, United States
| |
Collapse
|
10
|
Vadászi H, Kiss B, Micsonai A, Schlosser G, Szaniszló T, Kovács RÁ, Györffy BA, Kékesi KA, Goto Y, Uzonyi B, Liliom K, Kardos J. Competitive inhibition of the classical complement pathway using exogenous single-chain C1q recognition proteins. J Biol Chem 2022; 298:102113. [PMID: 35690144 PMCID: PMC9270254 DOI: 10.1016/j.jbc.2022.102113] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 06/03/2022] [Accepted: 06/06/2022] [Indexed: 11/26/2022] Open
Abstract
Complement component 1q (C1q) is a protein complex of the innate immune system with well-characterized binding partners that constitutes part of the classical complement pathway (CP). In addition, C1q was recently described in the central nervous system as having a role in synapse elimination both in the healthy brain and in neurodegenerative diseases. However, the molecular mechanism of C1q-associated synapse phagocytosis is still unclear. Here, we designed monomer and multimer protein constructs which comprised the globular interaction recognition parts of mouse C1q (gC1q) as single-chain molecules (sc-gC1q proteins) lacking the collagen-like effector region. These molecules, which can competitively inhibit the function of C1q, were expressed in an E. coli expression system, and their structure and capabilities to bind known CP activators were validated by mass spectrometry, analytical size exclusion chromatography, analytical ultracentrifugation, circular dichroism spectroscopy, and ELISA. We further characterized the interactions between these molecules and immunoglobulins and neuronal pentraxins using surface plasmon resonance spectroscopy. We demonstrated that sc-gC1qs potently inhibited the function of C1q. Furthermore, these sc-gC1qs competed with C1q in binding to the embryonal neuronal cell membrane. We conclude that the application of sc-gC1qs can reveal neuronal localization and functions of C1q in assays in vivo and might serve as a basis for engineering inhibitors for therapeutic purposes.
Collapse
Affiliation(s)
- Henrietta Vadászi
- ELTE NAP Neuroimmunology Research Group, Department of Biochemistry, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Bence Kiss
- Department of Biochemistry, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - András Micsonai
- ELTE NAP Neuroimmunology Research Group, Department of Biochemistry, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Gitta Schlosser
- MTA ELTE Lendu¨let Ion Mobility Mass Spectrometry Research Group, Department of Analytical Chemistry, Institute of Chemistry, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Tamás Szaniszló
- Department of Biochemistry, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Réka Á Kovács
- ELTE NAP Neuroimmunology Research Group, Department of Biochemistry, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Balázs A Györffy
- ELTE NAP Neuroimmunology Research Group, Department of Biochemistry, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Katalin A Kékesi
- ELTE NAP Neuroimmunology Research Group, Department of Biochemistry, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary; Department of Physiology and Neurobiology, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Yuji Goto
- Global Center for Medical Engineering and Informatics, Osaka University, Osaka, Japan
| | - Barbara Uzonyi
- Department of Immunology, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary; MTA-ELTE Complement Research Group, Eötvös Loránd Research Network (ELKH), Department of Immunology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Károly Liliom
- Department of Biophysics and Radiation Biology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - József Kardos
- ELTE NAP Neuroimmunology Research Group, Department of Biochemistry, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary.
| |
Collapse
|
11
|
Dijkstra DJ, Lokki AI, Gierman LM, Borggreven NV, van der Keur C, Eikmans M, Gelderman KA, Laivuori H, Iversen AC, van der Hoorn MLP, Trouw LA. Circulating Levels of Anti-C1q and Anti-Factor H Autoantibodies and Their Targets in Normal Pregnancy and Preeclampsia. Front Immunol 2022; 13:842451. [PMID: 35432365 PMCID: PMC9009242 DOI: 10.3389/fimmu.2022.842451] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 03/07/2022] [Indexed: 11/10/2022] Open
Abstract
Preeclampsia (PE) generally manifests in the second half of pregnancy with hypertension and proteinuria. The understanding of the origin and mechanism behind PE is incomplete, although there is clearly an immune component to this disorder. The placenta constitutes a complicated immune interface between fetal and maternal cells, where regulation and tolerance are key. Stress factors from placental dysfunction in PE are released to the maternal circulation evoking the maternal response. Several complement factors play a role within this intricate landscape, including C1q in vascular remodeling and Factor H (FH) as the key regulator of alternative pathway complement activation. We hypothesize that decreased levels of C1q or FH, or disturbance of their function by autoantibodies, may be associated with PE. Autoantibodies against C1q and FH and the concentrations of C1q and FH were measured by ELISA in maternal sera from women with preeclamptic and normal pregnancies. Samples originated from cohorts collected in the Netherlands (n=63 PE; n=174 control pregnancies, n=51 nonpregnant), Finland (n=181 PE; n=63 control pregnancies) and Norway (n=59 PE; n=27 control pregnancies). Serum C1q and FH concentrations were higher in control pregnancy than in nonpregnant women. No significant differences were observed for serum C1q between preeclamptic and control pregnancy in any of the three cohorts. Serum levels of FH were lower in preeclamptic pregnancies compared to control pregnancies in two of the cohorts, this effect was driven by the early onset PE cases. Neither anti-C1q autoantibodies nor anti-FH autoantibodies levels differed between women with PE and normal pregnancies. In conclusion, levels of anti-C1q and anti-FH autoantibodies are not increased in PE. C1q and FH are increased in pregnancy, but importantly, a decrease in FH concentration is associated with PE.
Collapse
Affiliation(s)
- Douwe Jan Dijkstra
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands,*Correspondence: Douwe Jan Dijkstra, ; Leendert Adrianus Trouw,
| | - A. Inkeri Lokki
- Department of Bacteriology and Immunology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland,Medical and Clinical Genetics, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Lobke Marijn Gierman
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | | | - Carin van der Keur
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Michael Eikmans
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Kyra Andrea Gelderman
- Department of Immunopathology and Haemostasis, Sanquin Diagnostic Services, Amsterdam, Netherlands
| | - Hannele Laivuori
- Medical and Clinical Genetics, University of Helsinki and Helsinki University Hospital, Helsinki, Finland,Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland,Department of Obstetrics and Gynecology, Tampere University Hospital and Tampere University, Faculty of Medicine and Health Technology, Tampere Center for Child, Adolescent, and Maternal Health Research, Tampere, Finland
| | | | - Ann-Charlotte Iversen
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | | | - Leendert Adrianus Trouw
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands,*Correspondence: Douwe Jan Dijkstra, ; Leendert Adrianus Trouw,
| |
Collapse
|
12
|
Lindahl H, Bryceson YT. Neuroinflammation Associated With Inborn Errors of Immunity. Front Immunol 2022; 12:827815. [PMID: 35126383 PMCID: PMC8807658 DOI: 10.3389/fimmu.2021.827815] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 12/27/2021] [Indexed: 01/16/2023] Open
Abstract
The advent of high-throughput sequencing has facilitated genotype-phenotype correlations in congenital diseases. This has provided molecular diagnosis and benefited patient management but has also revealed substantial phenotypic heterogeneity. Although distinct neuroinflammatory diseases are scarce among the several thousands of established congenital diseases, elements of neuroinflammation are increasingly recognized in a substantial proportion of inborn errors of immunity, where it may even dominate the clinical picture at initial presentation. Although each disease entity is rare, they collectively can constitute a significant proportion of neuropediatric patients in tertiary care and may occasionally also explain adult neurology patients. We focus this review on the signs and symptoms of neuroinflammation that have been reported in association with established pathogenic variants in immune genes and suggest the following subdivision based on proposed underlying mechanisms: autoinflammatory disorders, tolerance defects, and immunodeficiency disorders. The large group of autoinflammatory disorders is further subdivided into IL-1β-mediated disorders, NF-κB dysregulation, type I interferonopathies, and hemophagocytic syndromes. We delineate emerging pathogenic themes underlying neuroinflammation in monogenic diseases and describe the breadth of the clinical spectrum to support decisions to screen for a genetic diagnosis and encourage further research on a neglected phenomenon.
Collapse
Affiliation(s)
- Hannes Lindahl
- Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
- Center for Molecular Medicine, Department of Clinical Neuroscience, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - Yenan T. Bryceson
- Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
- Center for Hematology and Regenerative Medicine, Department of Medicine, Karolinska Institute, Karolinska University Hospital Huddinge, Stockholm, Sweden
- Brogelmann Research Laboratory, Department of Clinical Sciences, University of Bergen, Bergen, Norway
| |
Collapse
|
13
|
Tsubouchi S, Hayashi H, Tahara K, Ishii K, Yasuda T, Yamamoto Y, Mizuuchi T, Mori H, Tago M, Kato E, Sawada T. Clinical presentation of a neuropsychiatric lupus patient with symmetrical basal ganglia lesions containing cytotoxic oedema cores surrounded by vasogenic oedema. Mod Rheumatol Case Rep 2020; 4:39-46. [PMID: 33086978 DOI: 10.1080/24725625.2019.1651955] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Neuropsychiatric (NP) manifestations of systemic lupus erythematosus (SLE) are diverse, but involvement of basal ganglia is rare. We describe here a 28-year-old woman with NPSLE presenting aseptic meningitis accompanied by elevated interleukin-6 levels in the cerebrospinal fluid, who developed symmetrical basal ganglia lesions, containing a cytotoxic oedematous core, surrounded by vasogenic oedema upon magnetic resonance imaging. We were able to observe these lesions from a de novo appearance during the disease onset to its disappearance during immunosuppressive treatment. Reversibility upon immunosuppressive treatment indicated that autoimmune mediated mechanisms could contribute to the basal ganglia lesions in NPSLE.
Collapse
Affiliation(s)
- Syoko Tsubouchi
- Department of Rheumatology, Tokyo Medical University Hospital, Tokyo, Japan
| | - Haeru Hayashi
- Department of Rheumatology, Tokyo Medical University Hospital, Tokyo, Japan
| | - Koichiro Tahara
- Department of Rheumatology, Tokyo Medical University Hospital, Tokyo, Japan
| | - Kayo Ishii
- Department of Rheumatology, Tokyo Medical University Hospital, Tokyo, Japan
| | - Takuya Yasuda
- Department of Rheumatology, Tokyo Medical University Hospital, Tokyo, Japan
| | - Yusuke Yamamoto
- Department of Rheumatology, Tokyo Medical University Hospital, Tokyo, Japan
| | - Takahiro Mizuuchi
- Department of Rheumatology, Tokyo Medical University Hospital, Tokyo, Japan
| | - Hiroaki Mori
- Department of Rheumatology, Tokyo Medical University Hospital, Tokyo, Japan
| | - Mayu Tago
- Department of Rheumatology, Tokyo Medical University Hospital, Tokyo, Japan
| | - Eri Kato
- Department of Rheumatology, Tokyo Medical University Hospital, Tokyo, Japan
| | - Tetsuji Sawada
- Department of Rheumatology, Tokyo Medical University Hospital, Tokyo, Japan
| |
Collapse
|
14
|
Schrijver B, Dijkstra DJ, Borggreven NV, La Distia Nora R, Huijser E, Versnel MA, van Hagen PM, Joosten SA, Trouw LA, Dik WA. Inverse correlation between serum complement component C1q levels and whole blood type-1 interferon signature in active tuberculosis and QuantiFERON-positive uveitis: implications for diagnosis. Clin Transl Immunology 2020; 9:e1196. [PMID: 33088504 PMCID: PMC7563643 DOI: 10.1002/cti2.1196] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 08/20/2020] [Accepted: 09/25/2020] [Indexed: 01/16/2023] Open
Abstract
Objectives To examine the relation between serum C1q levels and blood type‐1 interferon signature (type‐1 IFN signature) in active pulmonary tuberculosis (APTB) and to determine whether combined measurement of serum C1q and type‐1 IFN signature may add to the diagnosis of QuantiFERON‐positive (QFT+) patients with uveitis of unknown cause. Methods C1q was determined (ELISA) in serum from two distinct Indonesian cohorts, and in total, APTB (n = 72), QFT+ uveitis of unknown aetiology (n = 58), QFT− uveitis (n = 51) patients and healthy controls (HC; n = 73) were included. The type‐1 IFN signature scores were previously determined. Results Serum C1q was higher in APTB than HC (P < 0.001). APTB patients with uveitis had higher serum C1q than APTB patients without uveitis (P = 0.0207). Serum C1q correlated inversely with type‐1 IFN signature scores in APTB (P = 0.0036, r2 = 0.3526), revealing that these biomarkers for active TB disease can be mutually exclusive. Stratification of QFT+ patients with uveitis of unknown cause, by serum C1q and type‐1 IFN signature, yielded four groups with different likelihood of suffering from active TB uveitis. Conclusion Serum C1q is elevated in APTB, especially in those cases with uveitis. We propose that combined measurement of blood type‐1 IFN signature and serum C1q may provide added value in the diagnosis of active TB disease. Combined measurement of type‐1 IFN signature and serum C1q in QFT+ patients without signs of active TB disease, but suffering from uveitis of unknown cause, may be of help to identify cases with low or high likelihood of having active TB uveitis, which may facilitate clinical management decisions.
Collapse
Affiliation(s)
- Benjamin Schrijver
- Department of Immunology Laboratory Medical Immunology Erasmus MC University Medical Center Rotterdam Rotterdam The Netherlands
| | - Douwe J Dijkstra
- Department of Immunohematology and Blood Transfusion Leiden University Medical Center Leiden The Netherlands
| | - Nicole V Borggreven
- Department of Immunohematology and Blood Transfusion Leiden University Medical Center Leiden The Netherlands
| | - Rina La Distia Nora
- Department of Ophthalmology Faculty of Medicine University of Indonesia and Cipto Mangunkusumo Hospital Jakarta Indonesia
| | - Erika Huijser
- Department of Immunology Erasmus MC University Medical Center Rotterdam Rotterdam The Netherlands
| | - Marjan A Versnel
- Department of Immunology Erasmus MC University Medical Center Rotterdam Rotterdam The Netherlands
| | - P Martin van Hagen
- Department of Immunology Laboratory Medical Immunology Erasmus MC University Medical Center Rotterdam Rotterdam The Netherlands.,Department of Internal Medicine Division Clinical Immunology Erasmus MC University Medical Center Rotterdam Rotterdam The Netherlands
| | - Simone A Joosten
- Department of Infectious Diseases Leiden University Medical Center Leiden The Netherlands
| | - Leendert A Trouw
- Department of Immunohematology and Blood Transfusion Leiden University Medical Center Leiden The Netherlands
| | - Willem A Dik
- Department of Immunology Laboratory Medical Immunology Erasmus MC University Medical Center Rotterdam Rotterdam The Netherlands.,Department of Internal Medicine Division Clinical Immunology Erasmus MC University Medical Center Rotterdam Rotterdam The Netherlands
| |
Collapse
|
15
|
Li J, Ma Q, Liu H, Song X, Pang Y, Su P, Sun F, Gou M, Lu J, Shan Y, Liu X, Li Q, Han Y. Complement component C1q plays a critical role in VLRA/VLRC-mediated immune response. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2020; 111:103750. [PMID: 32447013 DOI: 10.1016/j.dci.2020.103750] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 05/18/2020] [Accepted: 05/18/2020] [Indexed: 05/07/2023]
Abstract
In jawless vertebrates, the lamprey complement component C1q (LC1q) acts as a lectin and activates lamprey complement component C3 (LC3) in association with mannose-binding lectin (MBL)-associated serine protease (MASP) via the lectin pathway. Furthermore, LC1q may interact with variable lymphocyte receptor B (VLRB) in a complex with antigens and mediate the activation of LC3, leading to cytolysis. In the present study, we found, for the first time, that LC1q plays a critical role in VLRA/VLRC-mediated immune response. Escherichia coli, Shigella flexneri, Aeromonas hydrophila, Pseudomonas plecoglossicida, Aeromonas allosaccharophila, P. luteola, Brevundimonas diminuta, and Bacillus cereus were isolated from infected Lampetra morii in our laboratory and identified using the 16s rRNA method. A. hydrophila was confirmed as a rapidly spreading lethal pathogen in the larvae of L. morii and was used in subsequent immune stimulation experiments. The results of real-time quantitative polymerase chain reaction (Q-PCR) and immunofluorescence analyses indicated that the RNA and protein expression levels of LC1q were upregulated following exposure to 107 cfu/mL of A. hydrophila, compared to the levels of the naïve group. We obtained LC1q morphants (LC1q MO) of lamprey larvae by morpholino-mediated knockdowns. We found that LC1q played key roles in the embryonic development of lamprey. The median lethal time (LT50) of LC1q MO larvae was 2 d after being exposed to the pathogens, whereas the LT50 of control MO was 5 d. The drastic decrease in LT50 values after LC1q knockdown implies that LC1q plays a critical role in lamprey immune response. Gene expression profiles of LC1q-deficient A. hydrophila, control MO A. hydrophila, wild type A. hydrophila, and naive 1-month-old ammocoetes larvae were compared by examining the expression levels of a selected panel of orthologous genes. It is worth mentioning that LC1q MO affected the VLRA+/VLRC + population genes but did not affect the VLRB + populations. Immunohistochemical data indicated that LC1q deficiency also affected VLRA and VLRC but not VLRB. Thus, LC1q plays a critical role in VLRA/VLRC-mediated immune response in lamprey.
Collapse
Affiliation(s)
- Jun Li
- College of Life Science, Liaoning Normal University, Dalian, 116029, China; Lamprey Research Center, Liaoning Normal University, Dalian, 116029, China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034, China; Liaoning Key Laboratory of Aquatic Animal Infectious Diseases Control and Prevention, Liaoning Institute of Freshwater Fisheries Sciences, Liaoyang, 111000, China
| | - Qinghua Ma
- College of Life Science, Liaoning Normal University, Dalian, 116029, China; Lamprey Research Center, Liaoning Normal University, Dalian, 116029, China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034, China
| | - Huaixiu Liu
- College of Life Science, Liaoning Normal University, Dalian, 116029, China; Lamprey Research Center, Liaoning Normal University, Dalian, 116029, China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034, China
| | - Xiaoping Song
- College of Life Science, Liaoning Normal University, Dalian, 116029, China; Lamprey Research Center, Liaoning Normal University, Dalian, 116029, China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034, China; Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, China
| | - Yue Pang
- College of Life Science, Liaoning Normal University, Dalian, 116029, China; Lamprey Research Center, Liaoning Normal University, Dalian, 116029, China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034, China
| | - Peng Su
- College of Life Science, Liaoning Normal University, Dalian, 116029, China; Lamprey Research Center, Liaoning Normal University, Dalian, 116029, China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034, China
| | - Feng Sun
- College of Life Science, Liaoning Normal University, Dalian, 116029, China; Lamprey Research Center, Liaoning Normal University, Dalian, 116029, China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034, China
| | - Meng Gou
- College of Life Science, Liaoning Normal University, Dalian, 116029, China; Lamprey Research Center, Liaoning Normal University, Dalian, 116029, China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034, China
| | - Jingjing Lu
- College of Life Science, Liaoning Normal University, Dalian, 116029, China; Lamprey Research Center, Liaoning Normal University, Dalian, 116029, China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034, China
| | - Yue Shan
- College of Life Science, Liaoning Normal University, Dalian, 116029, China; Lamprey Research Center, Liaoning Normal University, Dalian, 116029, China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034, China
| | - Xin Liu
- College of Life Science, Liaoning Normal University, Dalian, 116029, China; Lamprey Research Center, Liaoning Normal University, Dalian, 116029, China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034, China
| | - Qingwei Li
- College of Life Science, Liaoning Normal University, Dalian, 116029, China; Lamprey Research Center, Liaoning Normal University, Dalian, 116029, China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034, China
| | - Yinglun Han
- College of Life Science, Liaoning Normal University, Dalian, 116029, China; Lamprey Research Center, Liaoning Normal University, Dalian, 116029, China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034, China.
| |
Collapse
|
16
|
Sharma M, Vignesh P, Tiewsoh K, Rawat A. Revisiting the complement system in systemic lupus erythematosus. Expert Rev Clin Immunol 2020; 16:397-408. [PMID: 32228236 DOI: 10.1080/1744666x.2020.1745063] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Introduction: Systemic lupus erythematosus (SLE) is a multi-system autoimmune disease, characterized by the production of autoantibodies. Numerous mechanisms contribute to the pathogenesis and autoimmunity in SLE. One of the most important mechanisms is the defective function of the early complement components that are involved in clearing the immune-complexes and apoptotic debris. Major evidence supporting this hypothesis is the development of severe lupus in individuals with monogenic defects in any one of the early complement components such as C1q, C1 s, C1 r, C2, or C4.Areas covered: In this review, we discuss hereditary defects in classical complement components and their clinical manifestations, acquired defects of complements in lupus, the role of complements in the pathogenesis of antiphospholipid antibody syndrome and lupus nephritis, and laboratory assessment of complement components and their functions. Articles from the last 20 years were retrieved from PubMed for this purpose.Expert opinion: Complements have a dual role in the pathogenesis of SLE. On one hand, deficiency of complement components predisposes to lupus, while, on the other, excess complement activation plays a role in the organ damage. Understanding the intricacies of the role of complements in SLE can pave way for the development of targeted therapies.
Collapse
Affiliation(s)
- Madhubala Sharma
- Allergy Immunology Unit, Department of Pediatrics, Advanced Pediatrics Centre, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Pandiarajan Vignesh
- Allergy Immunology Unit, Department of Pediatrics, Advanced Pediatrics Centre, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Karalanglin Tiewsoh
- Allergy Immunology Unit, Department of Pediatrics, Advanced Pediatrics Centre, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Amit Rawat
- Allergy Immunology Unit, Department of Pediatrics, Advanced Pediatrics Centre, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
17
|
Demirkaya E, Sahin S, Romano M, Zhou Q, Aksentijevich I. New Horizons in the Genetic Etiology of Systemic Lupus Erythematosus and Lupus-Like Disease: Monogenic Lupus and Beyond. J Clin Med 2020; 9:E712. [PMID: 32151092 PMCID: PMC7141186 DOI: 10.3390/jcm9030712] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 02/12/2020] [Accepted: 02/21/2020] [Indexed: 02/05/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is a clinically and genetically heterogeneous autoimmune disease. The etiology of lupus and the contribution of genetic, environmental, infectious and hormonal factors to this phenotype have yet to be elucidated. The most straightforward approach to unravel the molecular pathogenesis of lupus may rely on studies of patients who present with early-onset severe phenotypes. Typically, they have at least one of the following clinical features: childhood onset of severe disease (<5 years), parental consanguinity, and presence of family history for autoimmune diseases in a first-degree relative. These patients account for a small proportion of patients with lupus but they inform considerable knowledge about cellular pathways contributing to this inflammatory phenotype. In recent years with the aid of new sequencing technologies, novel or rare pathogenic variants have been reported in over 30 genes predisposing to SLE and SLE-like diseases. Future studies will likely discover many more genes with private variants associated to lupus-like phenotypes. In addition, genome-wide association studies (GWAS) have identified a number of common alleles (SNPs), which increase the risk of developing lupus in adult age. Discovery of a possible shared immune pathway in SLE patients, either with rare or common variants, can provide important clues to better understand this complex disorder, it's prognosis and can help guide new therapeutic approaches. The aim of this review is to summarize the current knowledge of the clinical presentation, genetic diagnosis and mechanisms of disease in patents with lupus and lupus-related phenotypes.
Collapse
Affiliation(s)
- Erkan Demirkaya
- Schulich School of Medicine & Dentistry, Department of Paediatrics, Division of Paediatric Rheumatology, University of Western Ontario, London, ON N6A 5W9, Canada;
| | - Sezgin Sahin
- Van Training and Research Hospital, Department of Paediatric Rheumatology, 65000 Van, Turkey;
| | - Micol Romano
- Schulich School of Medicine & Dentistry, Department of Paediatrics, Division of Paediatric Rheumatology, University of Western Ontario, London, ON N6A 5W9, Canada;
- Department of Pediatric Rheumatology, ASST-PINI-CTO, 20122 Milano, Italy
| | - Qing Zhou
- Life Sciences Institute, Zhejiang University, Hang Zhou 310058, China;
| | - Ivona Aksentijevich
- Inflammatory Disease Section, National Human Genome Research Institute, Bethesda, MD 20892, USA;
| |
Collapse
|
18
|
Complement activation and regulation in rheumatic disease. Semin Immunol 2019; 45:101339. [DOI: 10.1016/j.smim.2019.101339] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 10/24/2019] [Accepted: 10/24/2019] [Indexed: 01/02/2023]
|
19
|
Tirosh I, Spielman S, Barel O, Ram R, Stauber T, Paret G, Rubinsthein M, Pessach IM, Gerstein M, Anikster Y, Shukrun R, Dagan A, Adler K, Pode-Shakked B, Volkov A, Perelman M, Greenberger S, Somech R, Lahav E, Majmundar AJ, Padeh S, Hildebrandt F, Vivante A. Whole exome sequencing in childhood-onset lupus frequently detects single gene etiologies. Pediatr Rheumatol Online J 2019; 17:52. [PMID: 31362757 PMCID: PMC6668194 DOI: 10.1186/s12969-019-0349-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Accepted: 07/08/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) comprise a diverse range of clinical manifestations. To date, more than 30 single gene causes of lupus/lupus like syndromes in humans have been identified. In the clinical setting, identifying the underlying molecular diagnosis is challenging due to phenotypic and genetic heterogeneity. METHODS We employed whole exome sequencing (WES) in patients presenting with childhood-onset lupus with severe and/or atypical presentations to identify cases that are explained by a single-gene (monogenic) cause. RESULTS From January 2015 to June 2018 15 new cases of childhood-onset SLE were diagnosed in Edmond and Lily Safra Children's Hospital. By WES we identified causative mutations in four subjects in five different genes: C1QC, SLC7A7, MAN2B1, PTEN and STAT1. No molecular diagnoses were established on clinical grounds prior to genetic testing. CONCLUSIONS We identified a significant fraction of monogenic SLE etiologies using WES and confirm the genetic locus heterogeneity in childhood-onset lupus. These results highlight the importance of establishing a genetic diagnosis for children with severe or atypical lupus by providing accurate and early etiology-based diagnoses and improving subsequent clinical management.
Collapse
Affiliation(s)
- Irit Tirosh
- 0000 0001 2107 2845grid.413795.dDepartment of Pediatrics B, Edmond and Lily Safra Children’s Hospital, Sackler Faculty of Medicine, Sheba Medical Center, Tel-Hashomer, 5265601 Ramat Gan, Israel ,0000 0001 2107 2845grid.413795.dRheumatology Unit, Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Tel-Hashomer, Israel ,0000 0004 1937 0546grid.12136.37Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Shiri Spielman
- 0000 0001 2107 2845grid.413795.dRheumatology Unit, Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Tel-Hashomer, Israel ,0000 0004 1937 0546grid.12136.37Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Ortal Barel
- 0000 0001 2107 2845grid.413795.dThe Genomic Unit, Sheba Cancer Research Center, Sheba Medical Center, Tel Hashomer, Israel
| | - Reut Ram
- 0000 0001 2107 2845grid.413795.dDepartment of Pediatrics B, Edmond and Lily Safra Children’s Hospital, Sackler Faculty of Medicine, Sheba Medical Center, Tel-Hashomer, 5265601 Ramat Gan, Israel
| | - Tali Stauber
- 0000 0001 2107 2845grid.413795.dDepartment of Pediatrics A Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Tel-Hashomer, Israel ,0000 0004 1937 0546grid.12136.37Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Gideon Paret
- 0000 0001 2107 2845grid.413795.dIntensive care unit, Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Tel-Hashomer, Israel ,0000 0004 1937 0546grid.12136.37Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Marina Rubinsthein
- 0000 0001 2107 2845grid.413795.dIntensive care unit, Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Tel-Hashomer, Israel ,0000 0004 1937 0546grid.12136.37Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Itai M. Pessach
- 0000 0001 2107 2845grid.413795.dIntensive care unit, Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Tel-Hashomer, Israel ,0000 0004 1937 0546grid.12136.37Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Maya Gerstein
- 0000 0001 2107 2845grid.413795.dDepartment of Pediatrics B, Edmond and Lily Safra Children’s Hospital, Sackler Faculty of Medicine, Sheba Medical Center, Tel-Hashomer, 5265601 Ramat Gan, Israel ,0000 0004 1937 0546grid.12136.37Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Yair Anikster
- 0000 0001 2107 2845grid.413795.dMetabolic Disease Unit, Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Tel-Hashomer, Israel ,0000 0004 1937 0546grid.12136.37Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Rachel Shukrun
- 0000 0001 2107 2845grid.413795.dDepartment of Pediatrics B, Edmond and Lily Safra Children’s Hospital, Sackler Faculty of Medicine, Sheba Medical Center, Tel-Hashomer, 5265601 Ramat Gan, Israel ,0000 0004 1937 0546grid.12136.37Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Adi Dagan
- 0000 0001 2107 2845grid.413795.dDepartment of Pediatrics B, Edmond and Lily Safra Children’s Hospital, Sackler Faculty of Medicine, Sheba Medical Center, Tel-Hashomer, 5265601 Ramat Gan, Israel ,0000 0004 1937 0546grid.12136.37Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Katerina Adler
- 0000 0001 2107 2845grid.413795.dThe Genomic Unit, Sheba Cancer Research Center, Sheba Medical Center, Tel Hashomer, Israel
| | - Ben Pode-Shakked
- 0000 0001 2107 2845grid.413795.dDepartment of Pediatrics B, Edmond and Lily Safra Children’s Hospital, Sackler Faculty of Medicine, Sheba Medical Center, Tel-Hashomer, 5265601 Ramat Gan, Israel ,0000 0001 2107 2845grid.413795.dMetabolic Disease Unit, Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Tel-Hashomer, Israel ,0000 0004 1937 0546grid.12136.37Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Alexander Volkov
- 0000 0001 2107 2845grid.413795.dPathology Department, Sheba Medical Center, Tel-Hashomer, Israel ,0000 0004 1937 0546grid.12136.37Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Marina Perelman
- 0000 0001 2107 2845grid.413795.dPathology Department, Sheba Medical Center, Tel-Hashomer, Israel ,0000 0004 1937 0546grid.12136.37Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Shoshana Greenberger
- 0000 0001 2107 2845grid.413795.dDepartment of Dermatology, Sheba Medical Center, Tel-Hashomer, Israel ,0000 0004 1937 0546grid.12136.37Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Raz Somech
- 0000 0001 2107 2845grid.413795.dDepartment of Pediatrics A Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Tel-Hashomer, Israel ,0000 0004 1937 0546grid.12136.37Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Einat Lahav
- 0000 0001 2107 2845grid.413795.dDepartment of Pediatrics A Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Tel-Hashomer, Israel ,0000 0004 1937 0546grid.12136.37Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel ,0000 0001 2107 2845grid.413795.dNephrology Unit, Edmond and Lily Safra Children’s Hospital, Sackler Faculty of Medicine, Sheba Medical Center, Tel Hashomer, 5265601 Ramat Gan, Israel
| | - Amar J. Majmundar
- 000000041936754Xgrid.38142.3cDivision of Nephrology, Department of Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, MA USA
| | - Shai Padeh
- 0000 0001 2107 2845grid.413795.dDepartment of Pediatrics B, Edmond and Lily Safra Children’s Hospital, Sackler Faculty of Medicine, Sheba Medical Center, Tel-Hashomer, 5265601 Ramat Gan, Israel ,0000 0004 1937 0546grid.12136.37Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Friedhelm Hildebrandt
- 000000041936754Xgrid.38142.3cDivision of Nephrology, Department of Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, MA USA
| | - Asaf Vivante
- Department of Pediatrics B, Edmond and Lily Safra Children's Hospital, Sackler Faculty of Medicine, Sheba Medical Center, Tel-Hashomer, 5265601, Ramat Gan, Israel. .,Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel. .,Nephrology Unit, Edmond and Lily Safra Children's Hospital, Sackler Faculty of Medicine, Sheba Medical Center, Tel Hashomer, 5265601, Ramat Gan, Israel.
| |
Collapse
|
20
|
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: 0.6] [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
|
21
|
Fan X, Bialecka M, Moustakas I, Lam E, Torrens-Juaneda V, Borggreven NV, Trouw L, Louwe LA, Pilgram GSK, Mei H, van der Westerlaken L, Chuva de Sousa Lopes SM. Single-cell reconstruction of follicular remodeling in the human adult ovary. Nat Commun 2019; 10:3164. [PMID: 31320652 PMCID: PMC6639403 DOI: 10.1038/s41467-019-11036-9] [Citation(s) in RCA: 119] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 06/13/2019] [Indexed: 12/13/2022] Open
Abstract
The ovary is perhaps the most dynamic organ in the human body, only rivaled by the uterus. The molecular mechanisms that regulate follicular growth and regression, ensuring ovarian tissue homeostasis, remain elusive. We have performed single-cell RNA-sequencing using human adult ovaries to provide a map of the molecular signature of growing and regressing follicular populations. We have identified different types of granulosa and theca cells and detected local production of components of the complement system by (atretic) theca cells and stromal cells. We also have detected a mixture of adaptive and innate immune cells, as well as several types of endothelial and smooth muscle cells to aid the remodeling process. Our results highlight the relevance of mapping whole adult organs at the single-cell level and reflect ongoing efforts to map the human body. The association between complement system and follicular remodeling may provide key insights in reproductive biology and (in)fertility.
Collapse
Affiliation(s)
- X Fan
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZC, Leiden, Netherlands
| | - M Bialecka
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZC, Leiden, Netherlands
| | - I Moustakas
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZC, Leiden, Netherlands.,Sequencing Analysis Support Core, Leiden University Medical Center, 2333 ZC, Leiden, Netherlands
| | - E Lam
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZC, Leiden, Netherlands
| | - V Torrens-Juaneda
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZC, Leiden, Netherlands
| | - N V Borggreven
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2333 ZA, Leiden, Netherlands
| | - L Trouw
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2333 ZA, Leiden, Netherlands
| | - L A Louwe
- Department of Gynaecology, Division of Reproductive Medicine, Leiden University Medical Center, 2333 ZA, Leiden, Netherlands
| | - G S K Pilgram
- Department of Gynaecology, Division of Reproductive Medicine, Leiden University Medical Center, 2333 ZA, Leiden, Netherlands
| | - H Mei
- Sequencing Analysis Support Core, Leiden University Medical Center, 2333 ZC, Leiden, Netherlands
| | - L van der Westerlaken
- Department of Gynaecology, Division of Reproductive Medicine, Leiden University Medical Center, 2333 ZA, Leiden, Netherlands
| | - S M Chuva de Sousa Lopes
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZC, Leiden, Netherlands. .,Department for Reproductive Medicine, Ghent University Hospital, 9000, Ghent, Belgium.
| |
Collapse
|
22
|
Neo JYJ, Wee SYK, Bonne I, Tay SH, Raida M, Jovanovic V, Fairhurst AM, Lu J, Hanson BJ, MacAry PA. Characterisation of a human antibody that potentially links cytomegalovirus infection with systemic lupus erythematosus. Sci Rep 2019; 9:9998. [PMID: 31292492 PMCID: PMC6620320 DOI: 10.1038/s41598-019-46329-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 06/26/2019] [Indexed: 11/08/2022] Open
Abstract
Human cytomegalovirus (HCMV) is a ubiquitous herpesvirus that has been linked with the development of systemic lupus erythematosus (SLE). Thus far, molecular mimicry has been implicated as the principal mechanism that explains this association. In this study, we characterise a potential alternative process whereby HCMV contributes to SLE. In a cohort of SLE patients, we show a significant association between HCMV infection and SLE through a human antibody response that targets UL44. UL44 is an obligate nuclear-resident, non-structural viral protein vital for HCMV DNA replication. The intracellular nature of this viral protein complicates its targeting by the humoral response - the mechanism remains unresolved. To characterise this response, we present a thorough molecular analysis of the first human monoclonal antibody specific for UL44 derived from a HCMV seropositive donor. This human antibody immunoprecipitates UL44 from HCMV-infected cells together with known nuclear-resident SLE autoantigens - namely, nucleolin, dsDNA and ku70. We also show that UL44 is redistributed to the cell surface during virus-induced apoptosis as part of a complex with these autoantigens. This phenomenon represents a potential mechanism for the bystander presentation of SLE autoantigens to the humoral arm of our immune system under circumstances that favour a break in peripheral tolerance.
Collapse
Affiliation(s)
- Jie Ying Jacklyn Neo
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
- Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Seng Yin Kelly Wee
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
- Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Isabelle Bonne
- Electron Microscopy Laboratory, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Sen Hee Tay
- Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Medicine, National University Health System, Singapore, Singapore
- Division of Rheumatology, Department of Medicine, National University Hospital, National University Health System, Singapore, Singapore
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Manfred Raida
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Vojislav Jovanovic
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Anna-Marie Fairhurst
- Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Jinhua Lu
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
- Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | | | - Paul A MacAry
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore.
- Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
23
|
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: 57] [Impact Index Per Article: 11.4] [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
|
24
|
Nikitin PA, Rose EL, Byun TS, Parry GC, Panicker S. C1s Inhibition by BIVV009 (Sutimlimab) Prevents Complement-Enhanced Activation of Autoimmune Human B Cells In Vitro. THE JOURNAL OF IMMUNOLOGY 2019; 202:1200-1209. [PMID: 30635392 DOI: 10.4049/jimmunol.1800998] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 12/10/2018] [Indexed: 12/17/2022]
Abstract
The classical pathway of complement (CP) can mediate C3 opsonization of Ags responsible for the costimulation and activation of cognate B lymphocytes. In this manner, the complement system acts as a bridge between the innate and adaptive immune systems critical for establishing a humoral response. However, aberrant complement activation is often observed in autoimmune diseases in which C3 deposition on self-antigens may serve to activate self-reactive B cell clones. In this study, we use BIVV009 (Sutimlimab), a clinical stage, humanized mAb that specifically inhibits the CP-specific serine protease C1s to evaluate the impact of upstream CP antagonism on activation and proliferation of normal and autoimmune human B cells. We report that BIVV009 significantly inhibited complement-mediated activation and proliferation of primary human B cells. Strikingly, CP antagonism suppressed human Ig-induced activation of B cells derived from patients with rheumatoid arthritis. These results suggest that clinical use of CP inhibitors in autoimmune patients may not only block complement-mediated tissue damage, but may also prevent the long-term activation of autoimmune B cells and the production of autoantibodies that contribute to the underlying pathologic condition of these diseases.
Collapse
Affiliation(s)
| | - Eileen L Rose
- Bioverativ, a Sanofi company, South San Francisco, CA 94080
| | - Tony S Byun
- Bioverativ, a Sanofi company, South San Francisco, CA 94080
| | - Graham C Parry
- Bioverativ, a Sanofi company, South San Francisco, CA 94080
| | | |
Collapse
|
25
|
Lubbers R, Sutherland JS, Goletti D, de Paus RA, van Moorsel CHM, Veltkamp M, Vestjens SMT, Bos WJW, Petrone L, Del Nonno F, Bajema IM, Dijkman K, Verreck FAW, Walzl G, Gelderman KA, Groeneveld GH, Geluk A, Ottenhoff THM, Joosten SA, Trouw LA. Complement Component C1q as Serum Biomarker to Detect Active Tuberculosis. Front Immunol 2018; 9:2427. [PMID: 30405622 PMCID: PMC6206241 DOI: 10.3389/fimmu.2018.02427] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 10/02/2018] [Indexed: 02/03/2023] Open
Abstract
Background: Tuberculosis (TB) remains a major threat to global health. Currently, diagnosis of active TB is hampered by the lack of specific biomarkers that discriminate active TB disease from other (lung) diseases or latent TB infection (LTBI). Integrated human gene expression results have shown that genes encoding complement components, in particular different C1q chains, were expressed at higher levels in active TB compared to LTBI. Methods: C1q protein levels were determined using ELISA in sera from patients, from geographically distinct populations, with active TB, LTBI as well as disease controls. Results: Serum levels of C1q were increased in active TB compared to LTBI in four independent cohorts with an AUC of 0.77 [0.70; 0.83]. After 6 months of TB treatment, levels of C1q were similar to those of endemic controls, indicating an association with disease rather than individual genetic predisposition. Importantly, C1q levels in sera of TB patients were significantly higher as compared to patients with sarcoidosis or pneumonia, clinically important differential diagnoses. Moreover, exposure to other mycobacteria, such as Mycobacterium leprae (leprosy patients) or BCG (vaccinees) did not result in elevated levels of serum C1q. In agreement with the human data, in non-human primates challenged with Mycobacterium tuberculosis, increased serum C1q levels were detected in animals that developed progressive disease, not in those that controlled the infection. Conclusions: In summary, C1q levels are elevated in patients with active TB compared to LTBI in four independent cohorts. Furthermore, C1q levels from patients with TB were also elevated compared to patients with sarcoidosis, leprosy and pneumonia. Additionally, also in NHP we observed increased C1q levels in animals with active progressive TB, both in serum and in broncho-alveolar lavage. Therefore, we propose that the addition of C1q to current biomarker panels may provide added value in the diagnosis of active TB.
Collapse
Affiliation(s)
- Rosalie Lubbers
- Department of Rheumatology, Leiden University Medical Center, Leiden, Netherlands
| | - Jayne S Sutherland
- Medical Research Council Unit The Gambia at the London School of Hygiene and Tropical Medicine, Banjul, Gambia
| | - Delia Goletti
- Translational Research Unit, Department of Epidemiology and Preclinical Research, National Institute for Infectious Diseases, Rome, Italy
| | - Roelof A de Paus
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | | | - Marcel Veltkamp
- Department of Pulmonology, St. Antonius Hospital Nieuwegein, Nieuwegein, Netherlands
| | - Stefan M T Vestjens
- Department of Internal Medicine, St. Antonius Hospital Nieuwegein, Nieuwegein, Netherlands
| | - Willem J W Bos
- Department of Internal Medicine, St. Antonius Hospital Nieuwegein, Nieuwegein, Netherlands.,Department of Nephrology, Leiden University Medical Center, Leiden, Netherlands
| | - Linda Petrone
- Translational Research Unit, Department of Epidemiology and Preclinical Research, National Institute for Infectious Diseases, Rome, Italy
| | - Franca Del Nonno
- Pathology Service, National Institute for Infectious Diseases, Rome, Italy
| | - Ingeborg M Bajema
- Department of Pathology, Leiden University Medical Center, Leiden, Netherlands
| | - Karin Dijkman
- Section of TB Research & Immunology, Biomedical Primate Research Centre, Rijswijk, Netherlands
| | - Frank A W Verreck
- Section of TB Research & Immunology, Biomedical Primate Research Centre, Rijswijk, Netherlands
| | - Gerhard Walzl
- Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, DST/NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
| | | | - Geert H Groeneveld
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Annemieke Geluk
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Tom H M Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Simone A Joosten
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Leendert A Trouw
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
26
|
Habib A, Sawmiller D, Hou H, Kanithi M, Tian J, Zeng J, Zi D, He ZX, Sanberg PR, Tan J. Human Cord Blood Serum-Derived APP α-Secretase Cleavage Activity is Mediated by C1 Complement. Cell Transplant 2018; 27:666-676. [PMID: 29871524 PMCID: PMC7020233 DOI: 10.1177/0963689718775941] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Alzheimer's Disease (AD) is the leading cause of dementia in the elderly. In healthy individuals, amyloid precursor protein (APP) is cleaved by α-secretase, generating soluble α-amyloid precursor protein (sAPPα), which contributes neuroprotective functions in the neuronal environment. In contrast, in the neurodegenerative environment of AD patients, amyloid-β-peptide (Aβ) of either 40 or 42 residues are generated by increased activity of β- and γ-secretase. These proteins amalgamate in specific regions of the brain, which disrupts neuronal functions and leads to cognitive impairment. Human umbilical cord blood cells (HUCBC) have proven useful as potential immunomodulatory therapies in various models of neurodegenerative diseases, including AD. Our most recent work studied the impact of umbilical cord blood serum (CBS) on modulation of sAPPα production. Heat-sensitive CBS significantly promoted sAPPα production, indicating that heat-sensitive factor(s) play(s) a role in this process. Liquid chromatography with tandem mass spectrometry (LC-MS/MS) analysis was used to determine the molecular source of α-secretase in purified CBS and aged blood serum (AgBS) fraction. Of the proteins identified, the subunits of C1 complex (C1q, C1r, and C1s) and alpha-2-macroglobulin showed significantly greater levels in purified α-CBS fraction (α-CBSF) compared with the AgBS fraction (AgBSF). Specifically, C1 markedly increased sAPPα and alpha-carboxyl-terminal fragment (α-CTF) production in a dose-dependent fashion, whereas C1q alone only minimally increased and C3 did not increase sAPPα production in the absence of sera. Furthermore, C1q markedly increased sAPPα and α-CTF, while decreasing Aβ, in CHO/APPwt cells cultured in the presence of whole sera. These results confirm our initial assumption that APP α-secretase activity in human blood serum is mediated by complement C1, opening a potential therapeutic modality for the future of AD.
Collapse
Affiliation(s)
- Ahsan Habib
- 1 Department of Psychiatry and Behavioral Neurosciences, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Darrell Sawmiller
- 1 Department of Psychiatry and Behavioral Neurosciences, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Huayan Hou
- 1 Department of Psychiatry and Behavioral Neurosciences, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Manasa Kanithi
- 1 Department of Psychiatry and Behavioral Neurosciences, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Jun Tian
- 1 Department of Psychiatry and Behavioral Neurosciences, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Jin Zeng
- 1 Department of Psychiatry and Behavioral Neurosciences, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Dan Zi
- 2 Guizhou Provincial Key Laboratory for Regenerative Medicine, Stem Cell and Tissue Engineering Research Center & Sino-US Joint Laboratory for Medical Sciences, Laboratory Animal Center, Guizhou Medical University Guiyang, China
| | - Zhi-Xu He
- 2 Guizhou Provincial Key Laboratory for Regenerative Medicine, Stem Cell and Tissue Engineering Research Center & Sino-US Joint Laboratory for Medical Sciences, Laboratory Animal Center, Guizhou Medical University Guiyang, China
| | - Paul R Sanberg
- 3 Center for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Jun Tan
- 1 Department of Psychiatry and Behavioral Neurosciences, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| |
Collapse
|
27
|
Zhi H, Xie J, Skare JT. The Classical Complement Pathway Is Required to Control Borrelia burgdorferi Levels During Experimental Infection. Front Immunol 2018; 9:959. [PMID: 29867944 PMCID: PMC5949333 DOI: 10.3389/fimmu.2018.00959] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 04/18/2018] [Indexed: 01/02/2023] Open
Abstract
Activation of the classical complement pathway occurs to varying degrees within strains of the Borrelia burgdorferi sensu lato complex, which contain a group of pathogenic spirochetes that cause tick-borne Lyme borreliosis, including the agent of Lyme disease in the United States, B. burgdorferi. Despite this information, details related to the control of B. burgdorferi by the classical pathway are not clear. To address this question, we infected C1qα-/- mice, which cannot assemble the C1 complex and thus fail to activate the classical pathway, with B. burgdorferi sensu stricto strain B31. Using bioluminescent in vivo imaging, we found that C1qα-/- mice harbored more B. burgdorferi following 10 days of infection relative to their isogenic C57BL/6 parent. Quantitative PCR (qPCR) demonstrated that C1qα-/- mice harbored significantly more B. burgdorferi than parent mice did within lymph nodes, skin, heart, and joints. The increased B. burgdorferi load in C1qα-/- mice was observed at 21 and 28 days of infection, consistent with the classical pathway promoting complement-dependent, antibody-mediated killing following the development of a B. burgdorferi-specific humoral immune response. In addition, circulating borrelial-specific IgM was higher in C1qα-/- mice relative to their parent mouse strain and did not decrease at 21 and 28 days post-infection, indicating that IgG class switching was delayed in C1qα-/- mice. At day 28, both Borrelia-specific IgG1 and IgG3 levels were higher in infected C1qα-/- mice, but that these antibodies were not sufficient to control borrelial infection in the absence of the classical pathway. Furthermore, the lack of C1q also altered the balance of the Th1/Th2 response, as both circulating Th1 (MIP-1α, IL-2, IL-12, and TNFα), Th2 (IL-4, IL-10, and MCP-1), and Th17 (IL-17) cytokines were elevated in infected C1qα-/- mice. These data imply that C1q and the classical pathway play important roles in controlling borrelial infection via antibody and complement-dependent killing, as well as altering both antibody maturation processes and the T cell response following exposure to infectious B. burgdorferi.
Collapse
Affiliation(s)
- Hui Zhi
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M Health Science Center, Bryan, TX, United States
| | - Jialei Xie
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M Health Science Center, Bryan, TX, United States
| | - Jon T Skare
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M Health Science Center, Bryan, TX, United States
| |
Collapse
|
28
|
Barturen G, Alarcón-Riquelme ME. SLE redefined on the basis of molecular pathways. Best Pract Res Clin Rheumatol 2017; 31:291-305. [PMID: 29224672 DOI: 10.1016/j.berh.2017.09.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 09/01/2017] [Indexed: 12/11/2022]
Abstract
The implementation of precision medicine requires the recruiting of patients in statistically enough numbers, the possibility of obtaining enough materials, and the integration of data from various platforms, which are all real limitations. These types of studies have been performed extensively in cancer but barely on systemic lupus erythematosus (SLE) or other rheumatic diseases. To consider the practical use of the information obtained from such studies, we have to take into account the best biological fluid to use, the ease to perform the analysis in clinical practice, and its relevance to clinical practice. Here we review the most relevant studies that have performed analyses that attempt to classify or stratify SLE. We focus on two types of studies: those that stratify individuals diagnosed with SLE and those that compare SLE with other autoimmune diseases, defining differences and similarities that may be clinically relevant in the future.
Collapse
Affiliation(s)
- Guillermo Barturen
- Pfizer - University of Granada - Andalusian Government Center for Genomics and Oncological Research (GENYO), Av de la Ilustración 114, PTS, 18016, Granada, Spain.
| | - Marta E Alarcón-Riquelme
- Pfizer - University of Granada - Andalusian Government Center for Genomics and Oncological Research (GENYO), Av de la Ilustración 114, PTS, 18016, Granada, Spain; Unit of Inflammatory Chronic Diseases, Institute of Environmental Medicine, Karolinska Institutet, Solna, 17777, Sweden.
| |
Collapse
|