1
|
Petrow E, Feng C, Frazer-Abel A, Marangoni RG, Lutz K, Nichols WC, Holers VM, Ritchlin C, White RJ, Korman BD. Utility of factor D and other alternative complement factors as biomarkers in systemic sclerosis-associated pulmonary arterial hypertension (SSc-PAH). Semin Arthritis Rheum 2024; 69:152554. [PMID: 39298973 PMCID: PMC11606736 DOI: 10.1016/j.semarthrit.2024.152554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 08/22/2024] [Accepted: 08/23/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND Activation of the complement cascade is thought to play a role in scleroderma vasculopathy. We previously showed that complement factor D was elevated in patients with limited cutaneous SSc and pulmonary arterial hypertension (PAH). In this study, we sought to assess multiple relevant components of the complement cascade to determine if they are altered in SSc-PAH, as well as their potential utility as biomarkers of disease severity and progression. METHODS Complement components (n = 14) were measured using multiplex assays in 156 patients with SSc-PAH from a multi-site repository and were compared to 33 patients with SSc without PAH, and 40 healthy controls. Data were evaluated for correlations between complement levels, right heart catheterization measures, and clinical endpoints including 6-minute walk distance. To assess complement longitudinally, serum complement levels were assayed at 0, 4, 12, 24, 36 and 48 weeks in 52 SSc-PAH patients who participated in a prior clinical trial. RESULTS We found that factor D was significantly elevated in SSc-PAH compared to SSc without PAH (p < 0.0001) and was highly sensitive and specific for SSc-PAH (AUC=0.82, p < 0.001). In SSc-PAH patients, alterations in factor H, C4, and factor D were associated with measures of PAH disease severity including right heart catheterization measurements (cardiac output, right atrial pressure, and VO2 max), survival, and 6-minute walk distance. No significant changes in complement levels or clinical associations were seen over time or associated with treatment in the longitudinal clinical trial study. CONCLUSION Our work confirms prior studies demonstrating a role for complement activation in SSc vascular disease and elevations of factor D in a large SSc-PAH population. Further, factor H and other complement factors are associated with severity of PAH including mortality. Taken together, these findings suggest that the alternative complement pathway plays a role in SSc-PAH pathogenesis and may serve as a biomarker to inform diagnosis and prognosis.
Collapse
Affiliation(s)
- Eva Petrow
- Department of Medicine, Division of Allergy, Immunology and Rheumatology, University of Rochester Medical Center, 601 Elmwood Ave, Box 695, Rochester, NY 14642, United States.
| | - Changyong Feng
- Department of Biostatistics and Computational Biology, University of Rochester, Saunders Research Building, 265 Crittenden Boulevard, Box 630, Rochester, NY 14642, United States.
| | - Ashley Frazer-Abel
- Exsera BioLabs, University of Colorado School of Medicine, 1775 Aurora Court, Mail Stop B115, Aurora, CO 80045, United States.
| | - Roberta Goncalves Marangoni
- Department of Medicine, Division of Allergy, Immunology and Rheumatology, University of Rochester Medical Center, 601 Elmwood Ave, Box 695, Rochester, NY 14642, United States.
| | - Katie Lutz
- Division of Human Genetics, Cincinnati Children's Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, 3333 Burnet Ave, ML7016, Cincinnati, OH 45229, United States.
| | - William C Nichols
- Division of Human Genetics, Cincinnati Children's Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, 3333 Burnet Ave, ML7016, Cincinnati, OH 45229, United States.
| | - V Michael Holers
- Departments of Medicine and Immunology, Division of Rheumatology, University of Colorado School of Medicine, 1775 North Aurora Court, 3102, Aurora, CO 80045, United States.
| | - Christopher Ritchlin
- Department of Medicine, Division of Allergy, Immunology and Rheumatology, University of Rochester Medical Center, 601 Elmwood Ave, Box 695, Rochester, NY 14642, United States.
| | - R James White
- Department of Medicine, Division of Pulmonary & Critical Care Medicine, University of Rochester Medical Center, 601 Elmwood Ave, Box 692, Rochester NY 14642, United States.
| | - Benjamin D Korman
- Department of Medicine, Division of Allergy, Immunology and Rheumatology, University of Rochester Medical Center, 601 Elmwood Ave, Box 695, Rochester, NY 14642, United States.
| |
Collapse
|
2
|
Katz G, Perugino C, Wallace ZS, Jiang B, Guy T, McMahon GA, Jha I, Zhang Y, Liu H, Fernandes AD, Pillai SS, Atkinson JP, Kim AH, Stone JH. Multiorgan involvement and circulating IgG1 predict hypocomplementaemia in IgG4-related disease. Ann Rheum Dis 2024; 83:1773-1780. [PMID: 39079893 PMCID: PMC11563845 DOI: 10.1136/ard-2024-225846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 07/14/2024] [Indexed: 11/16/2024]
Abstract
OBJECTIVES Hypocomplementaemia is common in patients with IgG4-related disease (IgG4-RD). We aimed to determine the IgG4-RD features associated with hypocomplementaemia and investigate mechanisms of complement activation in this disease. METHODS We performed a single-centre cross-sectional study of 279 patients who fulfilled the IgG4-RD classification criteria, using unadjusted and multivariable-adjusted logistic regression to identify factors associated with hypocomplementaemia. RESULTS Hypocomplementaemia was observed in 90 (32%) patients. In the unadjusted model, the number of organs involved (OR 1.42, 95% CI 1.23 to 1.63) and involvement of the lymph nodes (OR 3.87, 95% CI 2.19 to 6.86), lungs (OR 3.81, 95% CI 2.10 to 6.89), pancreas (OR 1.66, 95% CI 1.001 to 2.76), liver (OR 2.73, 95% CI 1.17 to 6.36) and kidneys (OR 2.48, 95% CI 1.47 to 4.18) were each associated with hypocomplementaemia. After adjusting for age, sex and number of organs involved, only lymph node (OR 2.59, 95% CI 1.36 to 4.91) and lung (OR 2.56, 95% CI 1.35 to 4.89) involvement remained associated with hypocomplementaemia while the association with renal involvement was attenuated (OR 1.6, 95% CI 0.92 to 2.98). Fibrotic disease manifestations (OR 0.43, 95% CI 0.21 to 0.87) and lacrimal gland involvement (OR 0.53, 95% CI 0.28 to 0.999) were inversely associated with hypocomplementaemia in the adjusted analysis. Hypocomplementaemia was associated with higher concentrations of all IgG subclasses and IgE (all p<0.05). After adjusting for serum IgG1 and IgG3, only IgG1 but not IgG4 remained strongly associated with hypocomplementaemia. CONCLUSIONS Hypocomplementaemia in IgG4-RD is not unique to patients with renal involvement and may reflect the extent of disease. IgG1 independently correlates with hypocomplementaemia in IgG4-RD, but IgG4 does not. Complement activation is likely involved in IgG4-RD pathophysiology.
Collapse
Affiliation(s)
- Guy Katz
- Massachusetts General Hospital Division of Rheumatology Allergy and Immunology, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Cory Perugino
- Massachusetts General Hospital Division of Rheumatology Allergy and Immunology, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Ragon Institute, Charlestown, Massachusetts, USA
| | - Zachary S Wallace
- Massachusetts General Hospital Division of Rheumatology Allergy and Immunology, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Bohang Jiang
- Massachusetts General Hospital Division of Rheumatology Allergy and Immunology, Boston, Massachusetts, USA
| | - Thomas Guy
- Ragon Institute, Charlestown, Massachusetts, USA
| | - Grace A McMahon
- Massachusetts General Hospital Division of Rheumatology Allergy and Immunology, Boston, Massachusetts, USA
| | - Isha Jha
- Massachusetts General Hospital Division of Rheumatology Allergy and Immunology, Boston, Massachusetts, USA
| | - Yuqing Zhang
- Massachusetts General Hospital Division of Rheumatology Allergy and Immunology, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Hang Liu
- Ragon Institute, Charlestown, Massachusetts, USA
| | - Ana D Fernandes
- Massachusetts General Hospital Division of Rheumatology Allergy and Immunology, Boston, Massachusetts, USA
| | - Shiv S Pillai
- Ragon Institute, Charlestown, Massachusetts, USA
- Massachusetts General Hospital, Boston, Massachusetts, USA
| | - John Patterson Atkinson
- Medicine, Rheumatology, Washington University in Saint Louis School of Medicine, Saint Louis, Missouri, USA
| | - Alfred Hyoungju Kim
- Medicine, Rheumatology, Washington University in Saint Louis School of Medicine, Saint Louis, Missouri, USA
| | - John H Stone
- Massachusetts General Hospital Division of Rheumatology Allergy and Immunology, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
3
|
Ren Y, Tian J, Shi W, Feng J, Liu Y, Kang H, He Y. Evaluation of ocular surface inflammation and systemic conditions in patients with systemic lupus erythematosus: a cross-sectional study. BMC Ophthalmol 2024; 24:492. [PMID: 39533209 PMCID: PMC11556210 DOI: 10.1186/s12886-024-03760-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
OBJECTIVE The cross-sectional study was designed to evaluate the association of ocular surface inflammation with systemic conditions in patients with systemic lupus erythematosus (SLE). METHODS The study enrolled 30 SLE patients and 30 controls. Ocular symptoms were evaluated using the Ocular Surface Disease Index (OSDI) questionnaire. Tear samples from all participants were collected for tear multi-cytokine and chemokine concentration analysis. All participants were assessed for dry eye disease (DED), including Schirmer I test, tear break-up time (TBUT), corneal fluorescein staining (CFS), meibomian gland secretion (MGS), lid-parallel conjunctival folds (LIPCOF), corneal clarity, and symblepharon. Besides, all participants were also examined for conjunctival impression cytology to measure the density of conjunctival goblet cells (CGCs). The peripheral blood indicators from SLE patients were also collected to measure the SLE-associated autoantibody specificities and systemic inflammatory indicators. Pearson and Spearman's analysis were uesd to examine the correlation between tear cytokines, CGCs, DED-related indicators, and systemic conditions. RESULTS The two groups were matched for age and gender in this study. 36.67% of eyes (11 in 30) of SLE patients and 13.33% of eyes (4 in 30) of controls were diagnosed with DED. OSDI scores, abnormal TBUT percentages, CFS percentages, and DED grading were all higher in SLE patients than in control group, while density of CGCs was lower. There were no significant differences in Schirmer I test, MGS, LIPCOF, corneal clarity, and symblepharon between SLE patients and controls. The levels of tear chemokine (C-X-C motif) ligand 11 (CXCL11) and cytokine interleukin-7 (IL-7) in patients with SLE were significantly higher than those in control group. Moreover, among SLE patients, the severity of DED and the level of tear chemokine CXCL11 were significantly positively correlated with SLE-associated autoantibody specificities. CONCLUSION Dry eye and tear cytokines and chemokines-mediated ocular surface inflammation persist in SLE patients and are associated with systemic conditions. Therefore, it is necessary for patients with SLE to combine systemic and ocular assessments.
Collapse
Affiliation(s)
- Yuerong Ren
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, China
| | - Jing Tian
- Department of Rheumatology and Immunology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wen Shi
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, China
| | - Jianing Feng
- Xi'an People's Hospital (Xi'an Fourth Hospital), Shaanxi Eye Hospital, Northwest University Affiliated People's Hospital, Xi'an, Shaanxi Province, China
| | - Yingyi Liu
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, China
| | - Huanmin Kang
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, China
| | - Yan He
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmic and Visual Science Key Laboratory, Beijing, China.
| |
Collapse
|
4
|
Mahin A, Chikmagalur Ravindra S, Ramesh P, Naik P, Raju R, Keshava Prasad TS, Abhinand CS. Unveiling Actin Cytoskeleton Role in Mediating Chikungunya-Associated Arthritis: An Integrative Proteome-Metabolome Study. Vector Borne Zoonotic Dis 2024; 24:753-762. [PMID: 38717066 DOI: 10.1089/vbz.2024.0018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2024] Open
Abstract
Background: Chikungunya is a zoonotic disease caused by the Chikungunya virus (CHIKV), primarily transmitted to humans through infected Aedes mosquitoes. The infection is characterized by symptoms such as high fever, musculoskeletal pain, polyarthritis, and a rash, which can lead to severe complications such as encephalitis, meningitis, and even fatalities. While many disease manifestations resemble those of other viral infections, chronic arthritis caused by CHIKV is unique, and its molecular mechanisms remain ill-defined. Materials and Methods: Proteomics data from both cellular and patient levels of CHIKV infection were curated from PubMed and screened using inclusion and exclusion criteria. Patient serum proteomics data obtained from PRIDE underwent reanalysis using Proteome Discoverer 2.2. Enrichment and protein-protein interaction network analysis were conducted on differentially expressed proteins from both serum and cellular datasets. Metabolite data from CHIKV-infected patients were further retrieved, and their protein binding partners were identified using BindingDB. The protein-metabolite interaction pathway was further developed using MetaboAnalyst. Results: The proteomics data analysis revealed differential expression of proteins involved in critical host mechanisms, such as cholesterol metabolism and mRNA splicing, during CHIKV infection. Consistent upregulation of two actin cytoskeleton proteins, TAGLN2 and PFN1, was noted in both serum and cellular datasets, and their upregulations are associated with arthritis. Furthermore, alterations in purine metabolism were observed in the integrative proteome-metabolome analysis, correlating with cytoskeletal remodelling. Conclusion: Collectively, this integrative view sheds light on the involvement of actin cytoskeleton remodeling proteins and purine metabolic pathways in the development of arthritis during CHIKV infection.
Collapse
Affiliation(s)
- Althaf Mahin
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Center, Yenepoya (Deemed to Be University), Mangalore, India
| | - Sourav Chikmagalur Ravindra
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Center, Yenepoya (Deemed to Be University), Mangalore, India
- Department of Biosciences, Mangalore University, Mangalore, India
| | - Poornima Ramesh
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Center, Yenepoya (Deemed to Be University), Mangalore, India
| | - Prashantha Naik
- Department of Biosciences, Mangalore University, Mangalore, India
| | - Rajesh Raju
- Centre for Integrative Omics Data Science (CIODS), Yenepoya (Deemed to Be University), Mangalore, India
| | | | - Chandran S Abhinand
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Center, Yenepoya (Deemed to Be University), Mangalore, India
| |
Collapse
|
5
|
Tortosa-Carreres J, Cubas-Núñez L, Piqueras M, Castillo-Villalba J, Quintanilla-Bordàs C, Quiroga-Varela A, Villarrubia N, Monreal E, Álvarez G, Gasque-Rubio R, Forés-Toribio L, Carratalà-Boscà S, Lucas C, Sanz MT, Ramió-Torrentà L, Villar LM, Casanova B, Laiz B, Pérez-Miralles FC. Evaluating the complement C1q levels in serum and cerebrospinal fluid in multiple sclerosis patients: Could it serve as a valuable marker in clinical practice? J Neuroimmunol 2024; 394:578428. [PMID: 39121816 DOI: 10.1016/j.jneuroim.2024.578428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 07/16/2024] [Accepted: 08/01/2024] [Indexed: 08/12/2024]
Abstract
Immunohistochemical studies have identified complement component C1q in MS lesions. We aimed to compare serum (sC1q) and CSF (csfC1q) levels in a large cohort of MS patients (pwMS) (n = 222) with those of healthy controls (HC, n = 52), individuals with other immune (IND, n = 14), and non-immune neurological disorders (nIND, n = 15), and to analyze their correlation with other biomarkers. pwMS were divided into three series based on their origin. CSF samples were unavailable for HC. All three pwMS cohorts had lower sC1q levels compared to HC and IND. csfC1q was higher in one pwMS cohort, with a trend in another, and correlated with IgG, Free Kappa Light Chains, GFAP, and Chitinase-3 Like Protein-1 in CSF. Our findings suggest a significant role for C1q in MS pathophysiology, potentially serving as a biomarker for disease identification.
Collapse
Affiliation(s)
- Jordi Tortosa-Carreres
- Laboratory Department, La Fe University and Polytechnic Hospital, 46026, Valencia, Spain; Neuroimmunology Unit, Health Research Institute La Fe, 46026, Valencia. Spain.
| | - Laura Cubas-Núñez
- Neuroimmunology Unit, Health Research Institute La Fe, 46026, Valencia. Spain.
| | - Mónica Piqueras
- Laboratory Department, La Fe University and Polytechnic Hospital, 46026, Valencia, Spain
| | | | - Carlos Quintanilla-Bordàs
- Neuroimmunology Unit, Health Research Institute La Fe, 46026, Valencia. Spain; Neurology Department, La Fe University and Polytechnic Hospital, 46026, Valencia, Spain
| | - Ana Quiroga-Varela
- Girona Neuroimmunology and Multiple Sclerosis Unit, Neurology Department, Dr. Josep Trueta University Hospital and Santa Caterina Hospital, Girona, Spain; Neurodegeneration and Neuroinflammation Research Group, Girona Biomedical Research Institute (IDIBGI), Salt, Spain.
| | - Noelia Villarrubia
- Departments of Immunology and Neurology, Ramon y Cajal University Hospital, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Red Española de Esclerosis Múltiple (REEM), Red de Enfermedades Inflamatorias (REI), ISCIII, Madrid, Spain.
| | - Enric Monreal
- Departments of Immunology and Neurology, Ramon y Cajal University Hospital, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Red Española de Esclerosis Múltiple (REEM), Red de Enfermedades Inflamatorias (REI), ISCIII, Madrid, Spain.
| | - Gary Álvarez
- Girona Neuroimmunology and Multiple Sclerosis Unit, Neurology Department, Dr. Josep Trueta University Hospital and Santa Caterina Hospital, Girona, Spain; Neurodegeneration and Neuroinflammation Research Group, Girona Biomedical Research Institute (IDIBGI), Salt, Spain.
| | - Raquel Gasque-Rubio
- Neuroimmunology Unit, Health Research Institute La Fe, 46026, Valencia. Spain
| | | | | | - Celia Lucas
- Computer Systems, La Fe University and Polytechnic Hospital, 46026, Valencia, Spain..
| | - María T Sanz
- Department of Didactic of Mathematics. University of Valencia, Spain.
| | - Lluís Ramió-Torrentà
- Girona Neuroimmunology and Multiple Sclerosis Unit, Neurology Department, Dr. Josep Trueta University Hospital and Santa Caterina Hospital, Girona, Spain.
| | - Luisa María Villar
- Multiple Sclerosis Unit, Ramon y Cajal University Hospital, Madrid, Spain.
| | - Bonaventura Casanova
- Neuroimmunology Unit, Health Research Institute La Fe, 46026, Valencia. Spain; Neurology Department, La Fe University and Polytechnic Hospital, 46026, Valencia, Spain
| | - Begoña Laiz
- Laboratory Department, La Fe University and Polytechnic Hospital, 46026, Valencia, Spain.
| | - Francisco Carlos Pérez-Miralles
- Neuroimmunology Unit, Health Research Institute La Fe, 46026, Valencia. Spain; Neurology Department, La Fe University and Polytechnic Hospital, 46026, Valencia, Spain
| |
Collapse
|
6
|
Dijkstra DJ, van de Bovenkamp FS, Abendstein L, Zuijderduijn R, Pool J, Kramer CSM, Slot LM, Drijfhout JW, de Vor L, Gelderman KA, Rooijakkers SHM, Zaldumbide A, Vidarsson G, Sharp TH, Parren PWHI, Trouw LA. Human anti-C1q autoantibodies bind specifically to solid-phase C1q and enhance phagocytosis but not complement activation. Proc Natl Acad Sci U S A 2023; 120:e2310666120. [PMID: 38048459 PMCID: PMC10723154 DOI: 10.1073/pnas.2310666120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 10/27/2023] [Indexed: 12/06/2023] Open
Abstract
Autoantibodies directed against complement component C1q are commonly associated with autoimmune diseases, especially systemic lupus erythematosus. Importantly, these anti-C1q autoantibodies are specific for ligand-bound, solid-phase C1q and do not bind to fluid-phase C1q. In patients with anti-C1q, C1q levels are in the normal range, and the autoantibodies are thus not depleting. To study these human anti-C1q autoantibodies at the molecular level, we isolated C1q-reactive B cells and recombinantly produced nine monoclonal antibodies (mAbs) from four different healthy individuals. The isolated mAbs were of the IgG isotype, contained extensively mutated variable domains, and showed high affinity to the collagen-like region of C1q. The anti-C1q mAbs exclusively bound solid-phase C1q in complex with its natural ligands, including immobilized or antigen-bound IgG, IgM or CRP, and necrotic cells. Competition experiments reveal that at least 2 epitopes, also targeted by anti-C1q antibodies in sera from SLE patients, are recognized. Electron microscopy with hexameric IgG-C1q immune complexes demonstrated that multiple mAbs can interact with a single C1q molecule and identified the region of C1q targeted by these mAbs. The opsonization of immune complexes with anti-C1q greatly enhanced Fc-receptor-mediated phagocytosis but did not increase complement activation. We conclude that human anti-C1q autoantibodies specifically bind neo-epitopes on solid-phase C1q, which results in an increase in Fc-receptor-mediated effector functions that may potentially contribute to autoimmune disease immunopathology.
Collapse
Affiliation(s)
- Douwe J. Dijkstra
- Department of Immunology, Leiden University Medical Center, Leiden2300 RC, The Netherlands
| | - Fleur S. van de Bovenkamp
- Department of Immunology, Leiden University Medical Center, Leiden2300 RC, The Netherlands
- Lava Therapeutics, Utrecht3584 CM, The Netherlands
| | - Leoni Abendstein
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden2300 RC, The Netherlands
| | - Rob Zuijderduijn
- Department of Immunology, Leiden University Medical Center, Leiden2300 RC, The Netherlands
| | - Jos Pool
- Department of Immunology, Leiden University Medical Center, Leiden2300 RC, The Netherlands
| | - Cynthia S. M. Kramer
- Department of Immunology, Leiden University Medical Center, Leiden2300 RC, The Netherlands
| | - Linda M. Slot
- Department of Rheumatology, Leiden University Medical Center, Leiden2300 RC, The Netherlands
| | - Jan W. Drijfhout
- Department of Immunology, Leiden University Medical Center, Leiden2300 RC, The Netherlands
| | - Lisanne de Vor
- Department of Medical Microbiology, University Medical Center, Utrecht3584 CX, The Netherlands
| | | | - Suzan H. M. Rooijakkers
- Department of Medical Microbiology, University Medical Center, Utrecht3584 CX, The Netherlands
| | - Arnaud Zaldumbide
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden2300 RC, The Netherlands
| | - Gestur Vidarsson
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam1066 CX, The Netherlands
| | - Thomas H. Sharp
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden2300 RC, The Netherlands
| | - Paul W. H. I. Parren
- Department of Immunology, Leiden University Medical Center, Leiden2300 RC, The Netherlands
- Gyes BV, Naarden1411 DC, The Netherlands
| | - Leendert A. Trouw
- Department of Immunology, Leiden University Medical Center, Leiden2300 RC, The Netherlands
| |
Collapse
|
7
|
Murad D, Zafar Paracha R, Saeed MT, Ahmad J, Mushtaq A, Humayun M. Modelling and analysis of the complement system signalling pathways: roles of C3, C5a and pro-inflammatory cytokines in SARS-CoV-2 infection. PeerJ 2023; 11:e15794. [PMID: 37744234 PMCID: PMC10517668 DOI: 10.7717/peerj.15794] [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: 02/08/2023] [Accepted: 07/04/2023] [Indexed: 09/26/2023] Open
Abstract
The complement system is an essential part of innate immunity. It is activated by invading pathogens causing inflammation, opsonization, and lysis via complement anaphylatoxins, complement opsonin's and membrane attack complex (MAC), respectively. However, in SARS-CoV-2 infection overactivation of complement system is causing cytokine storm leading to multiple organs damage. In this study, the René Thomas kinetic logic approach was used for the development of biological regulatory network (BRN) to model SARS-CoV-2 mediated complement system signalling pathways. Betweenness centrality analysis in cytoscape was adopted for the selection of the most biologically plausible states in state graph. Among the model results, in strongly connected components (SCCs) pro-inflammatory cytokines (PICyts) oscillatory behaviour between recurrent generation and downregulation was found as the main feature of SARS-CoV-2 infection. Diversion of trajectories from the SCCs leading toward hyper-inflammatory response was found in agreement with in vivo studies that overactive innate immunity response caused PICyts storm during SARS-CoV-2 infection. The complex of negative regulators FI, CR1 and DAF in the inhibition of complement peptide (C5a) and PICyts was found desirable to increase immune responses. In modelling role of MAC and PICyts in lowering of SARS-CoV-2 titre was found coherent with experimental studies. Intervention in upregulation of C5a and PICyts by C3 was found helpful in back-and-forth variation of signalling pattern linked with the levels of PICyts. Moreover, intervention in upregulation of PICyts by C5a was found productive in downregulation of all activating factors in the normal SCCs. However, the computational model predictions require experimental studies to be validated by exploring the activation role of C3 and C5a which could change levels of PICyts at various phases of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Didar Murad
- School of Interdisciplinary Engineering and Sciences/Department of Sciences, National University of Science and Technology, Islamabad, Pakistan
| | - Rehan Zafar Paracha
- School of Interdisciplinary Engineering and Sciences/Department of Sciences, National University of Science and Technology, Islamabad, Pakistan
| | - Muhammad Tariq Saeed
- School of Interdisciplinary Engineering and Sciences/Department of Sciences, National University of Science and Technology, Islamabad, Pakistan
| | - Jamil Ahmad
- Department of Computer Science and Information Technology, University of Malakand, Chakdara, Malakand, Pakistan
| | - Ammar Mushtaq
- School of Interdisciplinary Engineering and Sciences/Department of Sciences, National University of Science and Technology, Islamabad, Pakistan
| | - Maleeha Humayun
- School of Interdisciplinary Engineering and Sciences/Department of Sciences, National University of Science and Technology, Islamabad, Pakistan
| |
Collapse
|
8
|
Triggianese P, Conigliaro P, De Martino E, Monosi B, Chimenti MS. Overview on the Link Between the Complement System and Auto-Immune Articular and Pulmonary Disease. Open Access Rheumatol 2023; 15:65-79. [PMID: 37214353 PMCID: PMC10198272 DOI: 10.2147/oarrr.s318826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 05/05/2023] [Indexed: 05/24/2023] Open
Abstract
Complement system (CS) dysregulation is a key factor in the pathogenesis of different autoimmune diseases playing a central role in many immune innate and adaptive processes. Rheumatoid arthritis (RA) is a chronic inflammatory disease characterized by ta breach of self-tolerance leading to a synovitis and extra-articular manifestations. The CS is activated in RA and seems not only to mediate direct tissue damage but also play a role in the initiation of RA pathogenetic mechanisms through interactions with citrullinated proteins. Interstitial lung disease (ILD) represents the most common extra-articular manifestation that can lead to progressive fibrosis. In this review, we focused on the evidence of CS dysregulation in RA and in ILD, and highlighted the role of the CS in both the innate and adaptive immune responses in the development of diseases, by using idiopathic pulmonary fibrosis as a model of lung disease. As a proof of concept, we dissected the evidence that several treatments used to treat RA and ILD such as glucocorticoids, pirfenidone, disease modifying antirheumatic drugs, targeted biologics such as tumor necrosis factor (TNF)-inhibitors, rituximab, tocilizumab, and nintedanib may act indirectly on the CS, suggesting that the CS might represent a potential therapeutic target in these complex diseases.
Collapse
Affiliation(s)
- Paola Triggianese
- Department of Systems Medicine, Rheumatology, Allergology and Clinical Immunology, University of Rome Tor Vergata, Rome, Italy
| | - Paola Conigliaro
- Department of Systems Medicine, Rheumatology, Allergology and Clinical Immunology, University of Rome Tor Vergata, Rome, Italy
| | - Erica De Martino
- Department of Systems Medicine, Rheumatology, Allergology and Clinical Immunology, University of Rome Tor Vergata, Rome, Italy
| | - Benedetta Monosi
- Department of Systems Medicine, Rheumatology, Allergology and Clinical Immunology, University of Rome Tor Vergata, Rome, Italy
| | - Maria Sole Chimenti
- Department of Systems Medicine, Rheumatology, Allergology and Clinical Immunology, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
9
|
West EE, Kemper C. Complosome - the intracellular complement system. Nat Rev Nephrol 2023:10.1038/s41581-023-00704-1. [PMID: 37055581 PMCID: PMC10100629 DOI: 10.1038/s41581-023-00704-1] [Citation(s) in RCA: 65] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/10/2023] [Indexed: 04/15/2023]
Abstract
The complement system is a recognized pillar of host defence against infection and noxious self-derived antigens. Complement is traditionally known as a serum-effective system, whereby the liver expresses and secretes most complement components, which participate in the detection of bloodborne pathogens and drive an inflammatory reaction to safely remove the microbial or antigenic threat. However, perturbations in normal complement function can cause severe disease and, for reasons that are currently not fully understood, the kidney is particularly vulnerable to dysregulated complement activity. Novel insights into complement biology have identified cell-autonomous and intracellularly active complement - the complosome - as an unexpected central orchestrator of normal cell physiology. For example, the complosome controls mitochondrial activity, glycolysis, oxidative phosphorylation, cell survival and gene regulation in innate and adaptive immune cells, and in non-immune cells, such as fibroblasts and endothelial and epithelial cells. These unanticipated complosome contributions to basic cell physiological pathways make it a novel and central player in the control of cell homeostasis and effector responses. This discovery, together with the realization that an increasing number of human diseases involve complement perturbations, has renewed interest in the complement system and its therapeutic targeting. Here, we summarize the current knowledge about the complosome across healthy cells and tissues, highlight contributions from dysregulated complosome activities to human disease and discuss potential therapeutic implications.
Collapse
Affiliation(s)
- Erin E West
- National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Complement and Inflammation Research Section (CIRS), Bethesda, MD, USA
| | - Claudia Kemper
- National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Complement and Inflammation Research Section (CIRS), Bethesda, MD, USA.
| |
Collapse
|
10
|
Effects of Cannabidiol on Innate Immunity: Experimental Evidence and Clinical Relevance. Int J Mol Sci 2023; 24:ijms24043125. [PMID: 36834537 PMCID: PMC9964491 DOI: 10.3390/ijms24043125] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/18/2023] [Accepted: 02/02/2023] [Indexed: 02/09/2023] Open
Abstract
Cannabidiol (CBD) is the main non-psychotropic cannabinoid derived from cannabis (Cannabis sativa L., fam. Cannabaceae). CBD has received approval by the Food and Drug Administration (FDA) and European Medicines Agency (EMA) for the treatment of seizures associated with Lennox-Gastaut syndrome or Dravet syndrome. However, CBD also has prominent anti-inflammatory and immunomodulatory effects; evidence exists that it could be beneficial in chronic inflammation, and even in acute inflammatory conditions, such as those due to SARS-CoV-2 infection. In this work, we review available evidence concerning CBD's effects on the modulation of innate immunity. Despite the lack so far of clinical studies, extensive preclinical evidence in different models, including mice, rats, guinea pigs, and even ex vivo experiments on cells from human healthy subjects, shows that CBD exerts a wide range of inhibitory effects by decreasing cytokine production and tissue infiltration, and acting on a variety of other inflammation-related functions in several innate immune cells. Clinical studies are now warranted to establish the therapeutic role of CBD in diseases with a strong inflammatory component, such as multiple sclerosis and other autoimmune diseases, cancer, asthma, and cardiovascular diseases.
Collapse
|
11
|
Muñoz CM, Goulden B, Ahmed K, Alijotas-Reig J, Giles I. Risk of adverse pregnancy outcomes prior to the onset of an autoimmune rheumatic disease: a systematic review. Rheumatology (Oxford) 2023; 62:497-511. [PMID: 35929796 PMCID: PMC9891407 DOI: 10.1093/rheumatology/keac417] [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: 04/19/2022] [Revised: 06/17/2022] [Accepted: 07/07/2022] [Indexed: 02/04/2023] Open
Abstract
OBJECTIVES An increased risk of adverse maternal and foetal pregnancy complications (including pre-eclampsia, intrauterine growth restriction, and small for gestational age) is well described in women with autoimmune rheumatic disease (ARD) compared with the general population (GenPop). It is less clear, however, whether this risk of adverse pregnancy outcome (APO) also exists in women with 'preclinical ARD' (pre-ARD) before they are diagnosed with an ARD many years post-partum. Therefore, we have undertaken a systematic review of the available evidence on APO in patients who subsequently were diagnosed with a rheumatic disease to identify whether there is an increased risk in pre-ARD. METHODS The present study was reported in accordance with the guidance of the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) standard. A systematic literature review was performed using the online PubMed database. Pre-SLE and pre-RA patients were defined as those who, over the subsequent years, developed SLE or RA according to international classification criteria. RESULTS A total of 176 articles were screened, and 27 original articles were selected for final analysis. Pre-RA was the most studied group, with 15 studies and a total of >1600 pregnancies, and pre-SLE was the second-most studied pre-ARD in pregnancy, with 14 studies and a total of >1000 pregnancies. We found that patients who subsequently developed SLE had an increased burden of poor pregnancy outcomes compared with pregnant women from the GenPop, but fewer APOs compared with pregnancies of women with SLE. In contrast, a similar rate of APOs was found when pre-RA pregnancies were compared with GenPop pregnancies. CONCLUSION Our findings of an increased risk of APO in certain pre-ARDs highlights the relevance of taking an obstetric history during the first rheumatology appointment and the need for novel screening strategies for the prediction of APOs. Further research is required to elucidate the immune basis of APOs in preclinical and clinical ARD.
Collapse
Affiliation(s)
- Candido Muñoz Muñoz
- Centre for Rheumatology, Department of Inflammation, Division of Medicine, University College London, London, UK.,Systemic Autoimmune Disease Unit, Department of Medicine, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Bethan Goulden
- Centre for Rheumatology, Department of Inflammation, Division of Medicine, University College London, London, UK
| | - Kawser Ahmed
- Centre of Inflammation, Division of Medicine, University College London, London, UK
| | - Jaume Alijotas-Reig
- Systemic Autoimmune Disease Unit, Department of Medicine, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Ian Giles
- Centre for Rheumatology, Department of Inflammation, Division of Medicine, University College London, London, UK
| |
Collapse
|
12
|
de Boer ECW, Thielen AJF, Langereis JD, Kamp A, Brouwer MC, Oskam N, Jongsma ML, Baral AJ, Spaapen RM, Zeerleder S, Vidarsson G, Rispens T, Wouters D, Pouw RB, Jongerius I. The contribution of the alternative pathway in complement activation on cell surfaces depends on the strength of classical pathway initiation. Clin Transl Immunology 2023; 12:e1436. [PMID: 36721662 PMCID: PMC9881211 DOI: 10.1002/cti2.1436] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 10/31/2022] [Accepted: 12/23/2022] [Indexed: 01/28/2023] Open
Abstract
Objectives The complement system is an important component of innate immunity. The alternative pathway (AP) amplification loop is considered an essential feed forward mechanism for complement activation. However, the role of the AP in classical pathway (CP) activation has only been studied in ELISA settings. Here, we investigated its contribution on physiologically relevant surfaces of human cells and bacterial pathogens and in antibody-mediated complement activation, including in autoimmune haemolytic anaemia (AIHA) setting with autoantibodies against red blood cells (RBCs). Methods We evaluated the contribution of the AP to complement responses initiated through the CP on human RBCs by serum of AIHA patients and recombinant antibodies. Moreover, we studied complement activation on Neisseria meningitidis and Escherichia coli. The effect of the AP was examined using either AP-depleted sera or antibodies against factor B and factor D. Results We show that the amplification loop is redundant when efficient CP activation takes place. This is independent of the presence of membrane-bound complement regulators. The role of the AP may become significant when insufficient CP complement activation occurs, but this depends on antibody levels and (sub)class. Our data indicate that therapeutic intervention in the amplification loop will most likely not be effective to treat antibody-mediated diseases. Conclusion The AP can be bypassed through efficient CP activation. The AP amplification loop has a role in complement activation during conditions of modest activation via the CP, when it can allow for efficient complement-mediated killing.
Collapse
Affiliation(s)
- Esther CW de Boer
- Department of Immunopathology, Sanquin Research and Landsteiner LaboratoryAmsterdam Infection and Immunity Institute, Amsterdam University Medical CentreAmsterdamThe Netherlands,Department of Pediatric Immunology, Rheumatology, and Infectious Diseases, Emma Children's HospitalAmsterdam University Medical CentreAmsterdamThe Netherlands
| | - Astrid JF Thielen
- Department of Immunopathology, Sanquin Research and Landsteiner LaboratoryAmsterdam Infection and Immunity Institute, Amsterdam University Medical CentreAmsterdamThe Netherlands
| | - Jeroen D Langereis
- Laboratory of Medical Immunology, Radboud Institute for Molecular Life SciencesRadboudumcNijmegenThe Netherlands,Radboud Center for Infectious Diseases, RadboudumcNijmegenThe Netherlands
| | - Angela Kamp
- Department of Immunopathology, Sanquin Research and Landsteiner LaboratoryAmsterdam Infection and Immunity Institute, Amsterdam University Medical CentreAmsterdamThe Netherlands
| | - Mieke C Brouwer
- Department of Immunopathology, Sanquin Research and Landsteiner LaboratoryAmsterdam Infection and Immunity Institute, Amsterdam University Medical CentreAmsterdamThe Netherlands
| | - Nienke Oskam
- Department of Immunopathology, Sanquin Research and Landsteiner LaboratoryAmsterdam Infection and Immunity Institute, Amsterdam University Medical CentreAmsterdamThe Netherlands
| | - Marlieke L Jongsma
- Department of Immunopathology, Sanquin Research and Landsteiner LaboratoryAmsterdam Infection and Immunity Institute, Amsterdam University Medical CentreAmsterdamThe Netherlands
| | - April J Baral
- Translational and Clinical Research InstituteNewcastle upon TyneUK
| | - Robbert M Spaapen
- Department of Immunopathology, Sanquin Research and Landsteiner LaboratoryAmsterdam Infection and Immunity Institute, Amsterdam University Medical CentreAmsterdamThe Netherlands
| | - Sacha Zeerleder
- Department of Immunopathology, Sanquin Research and Landsteiner LaboratoryAmsterdam Infection and Immunity Institute, Amsterdam University Medical CentreAmsterdamThe Netherlands,Department of Hematology, Luzerner KantonsspitalLuzern and University of BernBernSwitzerland,Department for BioMedical ResearchUniversity of BernBernSwitzerland
| | - Gestur Vidarsson
- Department of Experimental Immunohematology, Sanquin Research, and Landsteiner LaboratoryAmsterdam University Medical CenterAmsterdamThe Netherlands
| | - Theo Rispens
- Department of Immunopathology, Sanquin Research and Landsteiner LaboratoryAmsterdam Infection and Immunity Institute, Amsterdam University Medical CentreAmsterdamThe Netherlands
| | - Diana Wouters
- Department of Immunopathology, Sanquin Research and Landsteiner LaboratoryAmsterdam Infection and Immunity Institute, Amsterdam University Medical CentreAmsterdamThe Netherlands,Centre for Infectious Disease ControlNational Institute for Public Health and the Environment (RIVM)BilthovenThe Netherlands
| | - Richard B Pouw
- Department of Immunopathology, Sanquin Research and Landsteiner LaboratoryAmsterdam Infection and Immunity Institute, Amsterdam University Medical CentreAmsterdamThe Netherlands,Sanquin Health SolutionsAmsterdamThe Netherlands
| | - Ilse Jongerius
- Department of Immunopathology, Sanquin Research and Landsteiner LaboratoryAmsterdam Infection and Immunity Institute, Amsterdam University Medical CentreAmsterdamThe Netherlands,Department of Pediatric Immunology, Rheumatology, and Infectious Diseases, Emma Children's HospitalAmsterdam University Medical CentreAmsterdamThe Netherlands
| |
Collapse
|
13
|
Ni X, Jiao J, Yang Z, Wang Z, Nan N, Gao D, Sun L, Zhu X, Zhou Q, Zhang N, Wu Z, Zhang S, Yuan H. The general law of plasma proteome alterations occurring in the lifetime of Chinese individuals reveals the importance of immunity. Aging (Albany NY) 2022; 14:7065-7092. [PMID: 36084955 PMCID: PMC9512505 DOI: 10.18632/aging.204278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/25/2022] [Indexed: 11/25/2022]
Abstract
Background: Aging is characterized by a continuous loss of protein homeostasis. A closer examination of peripheral blood, which houses proteins from nearly all tissues and cells, helped identify several biomarkers and other aspects of aging biology. To further explore the general law of aging and identify key time nodes and associated aging biology, we collected 97 plasma samples from 253 healthy individuals aged 0-100 years without adverse outcomes to conduct nano-Ultra High Performance Liquid Chromatography-Mass Spectrometry/Mass Spectrometry (nano-UHPLC-MS/MS) and weighted gene co-expression network analysis (WGCNA). Results: Through biological processes and key biological pathways identified in discrete age group modules, our analyses highlighted a strong correlation between alterations in the immune system and aging process. We also identified hub genes associated with distinct age groups that revealed alterations not only in protein expression but also in signaling cascade. Among them, hub genes from age groups of 0-20 years old and 71-100 years old are mostly involved in infectious diseases and the immune system. In addition, CDC5L and HMGB2 were the key transcription factors (TFs) regulating genes expression in people aged between 51-60 and 71-100 years of age. They were shown to not only be independent but also mutually regulate certain hub gene expressions. Conclusions: This study reveals that the plasma proteome undergoes a complex alteration over the lifetime of a human. In this process, the immune system is crucial throughout the lifespan of a human being. However, the underlying mechanism(s) regulating differential protein expressions at distinct ages remains to be elucidated.
Collapse
Affiliation(s)
- Xiaolin Ni
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, National Center of Gerontology of National Health Commission, Beijing 100730, China.,University of Chinese Academy of Sciences, Beijing 100049, China.,State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
| | - Juan Jiao
- Department of Clinical Laboratory, The Seventh Medical Center, Chinese PLA General Hospital, Beijing 100700, China
| | - Ze Yang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, National Center of Gerontology of National Health Commission, Beijing 100730, China
| | - Zhaoping Wang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, National Center of Gerontology of National Health Commission, Beijing 100730, China
| | - Nan Nan
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Chaoyang, Beijing 100029, China
| | - Danni Gao
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, National Center of Gerontology of National Health Commission, Beijing 100730, China
| | - Liang Sun
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, National Center of Gerontology of National Health Commission, Beijing 100730, China
| | - Xiaoquan Zhu
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, National Center of Gerontology of National Health Commission, Beijing 100730, China
| | - Qi Zhou
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, National Center of Gerontology of National Health Commission, Beijing 100730, China
| | - Nan Zhang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, National Center of Gerontology of National Health Commission, Beijing 100730, China
| | - Zhu Wu
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, National Center of Gerontology of National Health Commission, Beijing 100730, China
| | - Shenqi Zhang
- Department of Joint and Sports Medicine, Zaozhuang Municipal Hospital Affiliated to Jining Medical University, Shandong 277100, China
| | - Huiping Yuan
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, National Center of Gerontology of National Health Commission, Beijing 100730, China
| |
Collapse
|
14
|
Jia C, Tan Y, Zhao M. The complement system and autoimmune diseases. Chronic Dis Transl Med 2022; 8:184-190. [PMID: 36161202 PMCID: PMC9481883 DOI: 10.1002/cdt3.24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 03/10/2022] [Indexed: 11/13/2022] Open
Abstract
The complement system plays a key role in the pathogenesis of autoimmune diseases, which usually injures the kidney. More and more studies have shown the pathogenic role and indicated that abnormal activation of the complement system was highly involved in the outbreak of autoimmune diseases. This review mainly introduced recent studies of complement system activation contributing to the pathogenesis of autoimmune diseases, including systemic lupus erythematosus, antiphospholipid syndrome, antineutrophil cytoplasmic antibody-associated vasculitides, and so on. Understanding the pathogenic roles of complement activation in various autoimmune diseases will identify potential novel therapeutic targets on complement systems.
Collapse
Affiliation(s)
- Changhao Jia
- Laboratory of Renal Disease, Department of Medicine, Renal Division, Ministry of Health of China, Peking University First HospitalPeking University Institute of NephrologyBeijingChina
- Peking‐Tsinghua Center for Life SciencesBeijingChina
| | - Ying Tan
- Laboratory of Renal Disease, Department of Medicine, Renal Division, Ministry of Health of China, Peking University First HospitalPeking University Institute of NephrologyBeijingChina
| | - Minghui Zhao
- Laboratory of Renal Disease, Department of Medicine, Renal Division, Ministry of Health of China, Peking University First HospitalPeking University Institute of NephrologyBeijingChina
- Peking‐Tsinghua Center for Life SciencesBeijingChina
- Center for Interdisciplinary Research in BiomedicinePeking UniversityBeijingChina
| |
Collapse
|
15
|
Liisborg C, Skov V, Kjær L, Hasselbalch HC, Sørensen TL. Patients with MPNs and retinal drusen show signs of complement system dysregulation and a high degree of chronic low-grade inflammation. EClinicalMedicine 2022; 43:101248. [PMID: 35128362 PMCID: PMC8808164 DOI: 10.1016/j.eclinm.2021.101248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/16/2021] [Accepted: 12/08/2021] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND The hematopoietic stem cell disorders, myeloproliferative neoplasms (MPNs), are characterised by chronic low-grade inflammation (CLI). Recently, we showed that patients with MPNs have an increased prevalence of drusen and age-related macular degeneration (AMD), and drusen prevalence seemed associated with higher CLI. Studying MPNs may reveal more about drusen pathophysiology. This study investigated CLI further by measuring cytokine levels and complement system markers, comparing these between patients with MPNs and AMD. METHODS This cross-sectional study, between July 2018 and November 2020 conducted at Zealand University Hospital (ZUH) - Roskilde, Denmark, included 29 patients with neovascular AMD (nAMD), 28 with intermediate-stage AMD (iAMD), 62 with MPNs (35 with drusen - MPNd and 27 with healthy retinas - MPNn). With flow cytometry, we measured complement-regulatory-proteins (Cregs). With immunoassays, we investigated cytokine levels combined into a summary-inflammation-score (SIS). FINDINGS The MPNd and nAMD groups had similar SIS, significantly higher than the MPNn and iAMD groups. Additionally, we found SIS to increase over the MPN biological continuum from early cancer stage, essential thrombocytaemia (ET), over polycythaemia vera (PV) to the late-stage primary myelofibrosis (PMF). MPNs showed signs of complement dysregulation, with Cregs expression lower in PV than ET and PMF and even lower in PV patients with drusen. INTERPRETATION This study suggests that MPNd have a higher CLI than MPNn and may indicate systemic CLI to play a greater part in, and even initiate drusen formation. We suggest using MPNs as a "Human Inflammation Model" of drusen development. The CLI in MPNs elicits drusen formation, triggering more CLI creating a vicious cycle, increasing the risk of developing AMD. FUNDING Fight for Sight, Denmark, and Region Zealand's research promotion fund.
Collapse
Affiliation(s)
- Charlotte Liisborg
- Department of Ophthalmology, Zealand University Hospital, Vestermarksvej 23, Roskilde DK-4000, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, Copenhagen 2200, Denmark
- Corresponding author.
| | - Vibe Skov
- Department of Haematology, Zealand University Hospital, Vestermarksvej 15-17, Roskilde 4000, Denmark
| | - Lasse Kjær
- Department of Haematology, Zealand University Hospital, Vestermarksvej 15-17, Roskilde 4000, Denmark
| | - Hans Carl Hasselbalch
- Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, Copenhagen 2200, Denmark
- Department of Haematology, Zealand University Hospital, Vestermarksvej 15-17, Roskilde 4000, Denmark
| | - Torben Lykke Sørensen
- Department of Ophthalmology, Zealand University Hospital, Vestermarksvej 23, Roskilde DK-4000, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, Copenhagen 2200, Denmark
| |
Collapse
|
16
|
Zhang P, Zhang Y, Pan M, Liu Z, Li J, Peng L, Zhou J, Hu C, Liu S, Zeng X, Ge W, Zhang W. Proteomic analyses of plasma-derived exosomes in immunoglobulin (Ig) G4-related disease and their potential roles in B cell differentiation and tissue damage. J Autoimmun 2021; 122:102650. [PMID: 34107438 DOI: 10.1016/j.jaut.2021.102650] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/28/2021] [Accepted: 04/29/2021] [Indexed: 01/06/2023]
Abstract
OBJECTIVE To investigate the proteomic profiles of plasma exosomes isolated from patients with immunoglobulin (Ig) G4-related disease (IgG4-RD) and to determine their potential roles in B cell differentiation and tissue damage. METHODS One hundred untreated IgG4-RD patients and 135 sex- and age-matched healthy controls (HCs) were enrolled in this study. A combination of liquid chromatography-tandem mass spectrometry (LC-MS/MS) and tandem mass tag (TMT)-label quantitation was used for proteomic profiling. Differentially expressed proteins were validated by Western blot, enzyme-linked immunosorbent assay (ELISA) and real-time quantitative PCR (RT-qPCR) analyses. B cell activation, apoptosis, differentiation and reactive oxygen species (ROS) production were analyzed by flow cytometry. We also analyzed the correlations between differentially expressed complement proteins and laboratory parameters. RESULTS A total of 178 differentially expressed proteins were identified in plasma exosomes in IgG4-RD patients compared with HCs, and these proteins were enriched predominantly in the complement cascade pathway. Furthermore, reduced expression levels of complement components C3 and C5 in IgG4-RD were correlated with clinical parameters. Following stimulation with IgG4-RD plasma exosomes, the percentages of naïve B cells decreased, while those of memory B cells and plasmablasts increased; the levels of cytochrome c, somatic (CYCS) and downstream complement system activation also increased. Moreover, ROS production was greater in B cells of IgG4-RD patients than in those of HCs. In affected submandibular glands, the BCR signalling pathway was activated, and exosomes were enriched. CONCLUSION Proteomic profiling revealed that plasma exosome proteins may participate in the pathogenesis of IgG4-RD through complement activation and may be involved in B cell differentiation and activation of the B cell auto-oxidative damage pathway.
Collapse
Affiliation(s)
- Panpan Zhang
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, State Key Laboratory of Complex Severe and Rare Diseases, Beijing, China; Department of Rheumatology and Immunology, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, Henan Province, China
| | - Yusheng Zhang
- State Key Laboratory of Medical Molecular Biology & Department of Immunology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Meng Pan
- State Key Laboratory of Medical Molecular Biology & Department of Immunology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Zheng Liu
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, State Key Laboratory of Complex Severe and Rare Diseases, Beijing, China
| | - Jieqiong Li
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, State Key Laboratory of Complex Severe and Rare Diseases, Beijing, China
| | - Linyi Peng
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, State Key Laboratory of Complex Severe and Rare Diseases, Beijing, China
| | - Jiaxin Zhou
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, State Key Laboratory of Complex Severe and Rare Diseases, Beijing, China
| | - Chaojun Hu
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, State Key Laboratory of Complex Severe and Rare Diseases, Beijing, China
| | - Shengyun Liu
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, Henan Province, China
| | - Xiaofeng Zeng
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, State Key Laboratory of Complex Severe and Rare Diseases, Beijing, China
| | - Wei Ge
- State Key Laboratory of Medical Molecular Biology & Department of Immunology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China.
| | - Wen Zhang
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, State Key Laboratory of Complex Severe and Rare Diseases, Beijing, China.
| |
Collapse
|
17
|
Meuleman MS, Fremeaux-Bacchi V, Roumenina LT, Chauvet S. Ex Vivo Complement Activation on Endothelial Cells: Research and Translational Value. Trends Mol Med 2021; 27:418-421. [PMID: 33648869 DOI: 10.1016/j.molmed.2021.01.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/20/2021] [Accepted: 01/28/2021] [Indexed: 01/07/2023]
Abstract
The spectrum of human diseases with complement contribution is ever increasing. Tools to study the complement contribution and the potential interest of novel complement inhibitors in clinical practice are lacking. Here we discuss a functional ex vivo assay to monitor complement activation on endothelial cells, which can answer to this need.
Collapse
Affiliation(s)
- Marie-Sophie Meuleman
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, F-75006 Paris, France
| | - Veronique Fremeaux-Bacchi
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, F-75006 Paris, France
| | - Lubka T Roumenina
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, F-75006 Paris, France.
| | - Sophie Chauvet
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, F-75006 Paris, France.
| |
Collapse
|
18
|
Chen Y, Shao S, Huang J, Gu Y, Cheng Y, Zhu X. Therapeutic Efficacy of a Trichinella Spiralis Paramyosin-Derived Peptide Modified With a Membrane-Targeting Signal in Mice With Antigen-Induced Arthritis. Front Microbiol 2020; 11:608380. [PMID: 33424810 PMCID: PMC7785802 DOI: 10.3389/fmicb.2020.608380] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 11/30/2020] [Indexed: 12/21/2022] Open
Abstract
Helminth-derived molecules have the ability to modulate the host immune system. Our previous study identified a tetradecapeptide derived from Trichinella spiralis paramyosin (Ts-pmy) that could bind to human complement component C9 to inhibit its polymerization, making the peptide a candidate therapeutic agent for complement-related immune disorders. Here, the peptide underwent an N-terminal modification with a membrane-targeting signal (a unique myristoylated peptide) to improve its therapeutic efficacy. We found that the modified peptide had a binding affinity to human C9 that was similar to that of the original peptide, as confirmed by microscale thermophoresis assays. The binding of the modified peptide to human C9 resulted in the inhibition of C9-related complement activation, as reflected by the decreased Zn2+-induced C9 polymerization and the decreased C9-dependent lysis of rabbit erythrocytes. In addition, the original and modified peptides could both bind to recombinant mouse C9 and inhibit the C9-dependent lysis of rabbit erythrocytes in normal mouse serum (NMS), which meant that the peptides could cross the species barrier to inhibit complement activity in mice. Further in vitro and in vivo analyses confirmed that the peptide modification increased the retention time of the peptide. Furthermore, intraarticular injection of the modified peptide markedly ameliorated knee swelling and joint damage in mice with antigen-induced arthritis (AIA), as assessed histologically. These results suggested that the Ts-pmy-derived peptide modified with a membrane-targeting signal was a reasonable candidate therapeutic agent for membrane attack complex (MAC)-related diseases [such as rheumatoid arthritis (RA)] and the study presented a new modification method to improve the potential therapeutic effects of the peptide.
Collapse
Affiliation(s)
- Yi Chen
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Shuai Shao
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Jingjing Huang
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yuan Gu
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yuli Cheng
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xinping Zhu
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| |
Collapse
|
19
|
Innate immune response in systemic autoimmune diseases: a potential target of therapy. Inflammopharmacology 2020; 28:1421-1438. [DOI: 10.1007/s10787-020-00762-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 09/18/2020] [Indexed: 02/06/2023]
|
20
|
Nasonov EL, Reshetnyak TM, Alekberova ZS. [Thrombotic microangiopathy in rheumatology: a link between thrombosis and autoimmunity]. TERAPEVT ARKH 2020; 92:4-14. [PMID: 32598770 DOI: 10.26442/00403660.2020.05.000697] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Indexed: 12/16/2022]
Abstract
Uncontrolled hypercoagulation and inflammation (thromboinflammation), which are both independent and closely related and amplifying each other pathological processes, form the basis for pathogenesis of a wide range of diseases and complications, including immuno-inflammatory (autoimmune) rheumatic diseases, with the development of potentially fatal injuries of internal organs. Thrombotic microangiopathy is one of the most prominent prototypes of thromboinflammatory pathological conditions. The close link between environmental factors, hemostasis genetic defects and the complement system, inflammation and autoimmunity as pathogenetic mechanisms of microthrombosis draws particular attention to studying thrombotic microangiopathy in immuno-inflammatory rheumatic diseases, primarily systemic lupus erythematosus, antiphospholipid syndrome and scleroderma renal crisis. In future, these studies may be important for expanding the idea of the role of autoimmune mechanisms in pathogenesis of critical hemostasis disorders in human diseases, and for developing new approaches to therapy. Recently, special attention has been paid to the treatment of systemic lupus erythematosus and antiphospholipid syndrome with eculizumab, which is humanized monoclonal IgG2/4k antibody that blocks the complement component C5a and the membrane attack complex (C5b-9) formation, and which is registered for the treatment of atypical hemolytic uremic syndrome, paroxysmal nocturnal hemoglobinuria, as well as severe forms of myasthenia gravis and neuromyelitis optica. Further studies in this direction will create prerequisites for improving the prognosis not only in patients with orphan disorders, but also for widespread human diseases.
Collapse
|
21
|
Halkjær L, Troldborg A, Pedersen H, Jensen L, Hansen AG, Hansen TK, Bjerre M, Østergaard JA, Thiel S. Complement Receptor 2 Based Immunoassay Measuring Activation of the Complement System at C3-Level in Plasma Samples From Mice and Humans. Front Immunol 2020; 11:774. [PMID: 32431705 PMCID: PMC7214740 DOI: 10.3389/fimmu.2020.00774] [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] [Received: 02/07/2020] [Accepted: 04/06/2020] [Indexed: 12/15/2022] Open
Abstract
We aimed at establishing a sensitive and robust assay for estimation of systemic complement activation at complement component C3 level in mouse and human plasma samples. In order to capture the activation products iC3b and C3dg in a specific and physiological relevant manner we utilized a construct consisting of the iC3b/C3dg-binding site of human complement receptor 2 (CR2) attached to an Fc-part of mouse IgG. This construct binds C3dg and iC3b from both mice and humans. We purified the CR2-IgG construct from mouse B myeloma cell line supernatants, J558L-CR2-IgG, by protein G affinity chromatography. The CR2-IgG construct was used for capturing C3 fragments in microtiter wells and an anti-mouse or an anti-human-C3 antibody was used for detection of bound C3 fragments. Initially we tested the specificity of the assays with the use of purified C3 fragments. Further, with the use of the CR2-based assay, we measured an up to three-fold higher signal in activated mouse serum as compared to non-activated mouse serum, whereas activated serum from a C3 knock-out mouse gave no signal. We tested in vivo generated samples from a mouse experiment; complement activation was induced by injecting cobra venom factor or heat aggregated IgG into C57bl6 mice, followed by withdrawal of EDTA blood samples at different time points and measurement of iC3b/C3dg. We observed a clear time-dependent distinction in signals between samples with expected high and low complement activation. Furthermore, with the use of the assay for human C3 fragments, we observed that patients with systemic lupus erythematosus (SLE) (n = 144) had significantly higher iC3b/C3dg levels as compared to healthy individuals (n = 144) (p < 0.0001). We present two functional immunoassays, that are able to measure systemic levels of the C3-activation products iC3b and C3dg in mice and humans. To our knowledge, these are the first assays for complement activation that use a physiological relevant capture construct such as CR2. These assays will be a relevant tool when investigating mouse models and human diseases involving the complement system.
Collapse
Affiliation(s)
- Lene Halkjær
- Department of Biomedicine, Faculty of Health, Aarhus University, Aarhus, Denmark.,Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark.,Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Anne Troldborg
- Department of Biomedicine, Faculty of Health, Aarhus University, Aarhus, Denmark.,Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark.,Department of Rheumatology, Aarhus University Hospital, Aarhus, Denmark
| | - Henrik Pedersen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Lisbeth Jensen
- Department of Biomedicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| | | | | | - Mette Bjerre
- Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Jakob Appel Østergaard
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark.,Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Steffen Thiel
- Department of Biomedicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| |
Collapse
|
22
|
Xiao L, Harrison DG. Inflammation in Hypertension. Can J Cardiol 2020; 36:635-647. [PMID: 32389337 DOI: 10.1016/j.cjca.2020.01.013] [Citation(s) in RCA: 143] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 12/26/2019] [Accepted: 01/02/2020] [Indexed: 12/22/2022] Open
Abstract
For more than 50 years, evidence has accumulated that inflammation contributes to the pathogenesis of hypertension. Immune cells have been observed in vessels and kidneys of hypertensive humans. Biomarkers of inflammation, including high sensitivity C-reactive protein, various cytokines, and products of the complement pathway are elevated in humans with hypertension. Emerging evidence suggests that hypertension is accompanied and indeed initiated by activation of complement, the inflammasome, and by a change in the phenotype of circulating immune cells, particularly myeloid cells. High-dimensional transcriptomic analyses are providing insight into new subclasses of immune cells that are likely injurious in hypertension. These inflammatory events are interdependent and there is ultimately engagement of the adaptive immune system through mechanisms involving oxidative stress, modification of endogenous proteins, and alterations in antigen processing and presentation. These observations suggest new therapeutic opportunities to reduce end organ damage in hypertension might be used and guided by levels of inflammatory biomarkers.
Collapse
Affiliation(s)
- Liang Xiao
- Departments of Medicine, Pharmacology, and Physiology, and Division of Clinical Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - David G Harrison
- Departments of Medicine, Pharmacology, and Physiology, and Division of Clinical Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| |
Collapse
|