1
|
Xiao F, Guo J, Tomlinson S, Yuan G, He S. The role of the complosome in health and disease. Front Immunol 2023; 14:1146167. [PMID: 36969185 PMCID: PMC10036758 DOI: 10.3389/fimmu.2023.1146167] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 02/28/2023] [Indexed: 03/12/2023] Open
Abstract
The complement system is one of the immune system's oldest defense mechanisms and is historically regarded as a liver-derived and serum-active innate immune system that 'complements' cell-mediated and antibody-mediated immune responses against pathogens. However, the complement system is now recognized as a central component of both innate and adaptive immunity at both the systemic and local tissue levels. More findings have uncovered novel activities of an intracellularly active complement system-the complosome-that have shifted established functional paradigms in the field. The complosome has been shown to play a critical function in regulating T cell responses, cell physiology (such as metabolism), inflammatory disease processes, and cancer, which has amply proved its immense research potential and informed us that there is still much to learn about this system. Here, we summarize current understanding and discuss the emerging roles of the complosome in health and disease.
Collapse
Affiliation(s)
- Fang Xiao
- Division of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Jixu Guo
- Division of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Stephen Tomlinson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
| | - Guandou Yuan
- Division of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, Guangxi, China
| | - Songqing He
- Division of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, Guangxi, China
| |
Collapse
|
2
|
Schanzenbacher J, Köhl J, Karsten CM. Anaphylatoxins spark the flame in early autoimmunity. Front Immunol 2022; 13:958392. [PMID: 35958588 PMCID: PMC9358992 DOI: 10.3389/fimmu.2022.958392] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 06/29/2022] [Indexed: 11/13/2022] Open
Abstract
The complement system (CS) is an ancient and highly conserved part of the innate immune system with important functions in immune defense. The multiple fragments bind to specific receptors on innate and adaptive immune cells, the activation of which translates the initial humoral innate immune response (IR) into cellular innate and adaptive immunity. Dysregulation of the CS has been associated with the development of several autoimmune disorders such as systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), ANCA-associated vasculitis, and autoimmune bullous dermatoses (AIBDs), where complement drives the inflammatory response in the effector phase. The role of the CS in autoimmunity is complex. On the one hand, complement deficiencies were identified as risk factors to develop autoimmune disorders. On the other hand, activation of complement can drive autoimmune responses. The anaphylatoxins C3a and C5a are potent mediators and regulators of inflammation during the effector phase of autoimmunity through engagement of specific anaphylatoxin receptors, i.e., C3aR, C5aR1, and C5aR2 either on or in immune cells. In addition to their role in innate IRs, anaphylatoxins regulate humoral and cellular adaptive IRs including B-cell and T-cell activation, differentiation, and survival. They regulate B- and T-lymphocyte responses either directly or indirectly through the activation of anaphylatoxin receptors via dendritic cells that modulate lymphocyte function. Here, we will briefly review our current understanding of the complex roles of anaphylatoxins in the regulation of immunologic tolerance and the early events driving autoimmunity and the implications of such regulation for therapeutic approaches that target the CS.
Collapse
Affiliation(s)
- Jovan Schanzenbacher
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Jörg Köhl
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
- Division of Immunobiology, Cincinnati Childrens Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Christian M. Karsten
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
- *Correspondence: Christian M. Karsten,
| |
Collapse
|
3
|
Wang Y, Xiang M, Zhang H, Lu Y. Decreased complement 4d increases poor prognosis in patients with non‑small cell lung cancer combined with gastrointestinal lymph node metastasis. Exp Ther Med 2022; 24:560. [PMID: 35978919 PMCID: PMC9366274 DOI: 10.3892/etm.2022.11497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 06/23/2022] [Indexed: 11/18/2022] Open
Abstract
Lung cancer is a common malignancy that is difficult to treat and has a high risk of mortality. Although gastrointestinal lymph node metastasis has long been known to exert major impact on the prognosis of lung cancer, the mechanism of its occurrence and potential biological markers remain elusive. Therefore, the present study retrospectively analyzed data from 132 patients with non-small cell lung cancer (NSCLC) combined with lymph node metastasis between February 2010 and April 2019 from the First Affiliated Hospital of Soochow University (Suzhou, China) and Sichuan Cancer Hospital (Chengdu, China). Overall survival was assessed using Kaplan-Meier analysis and Cox logistic regression model. In addition, a prediction model was constructed based on immune indicators such as complement C3b and C4d (measured by ELISA), before the accuracy of this model was validated using calibration curves for 5-year OS. Among the 132 included patients, a total of 92 (70.0%) succumbed to the disease within 5 years. Multifactorial analysis revealed that complement C3b deficiency increased the risk of mortality by nearly two-fold [hazard ratio (HR)=2.23; 95% CI=1.20-4.14; P=0.017], whilst complement C4d deficiency similarly increased the risk of mortality by two-fold (HR=2.14; 95% CI=1.14-4.00; P=0.012). The variables were subsequently screened using Cox model to construct a prediction model based on complement C3b and C4d levels before a Nomogram plotted. By internal validation for the 132 patients, the Nomogram accurately estimated the risk of mortality, with a corrected C-index of 0.810. External validation of the model in another 50 patients from Sichuan Cancer Hospital revealed an accuracy of 77.0%. Overall, this mortality risk prediction model constructed based on complement levels showed accuracy in assessing the prognosis of patients with metastatic NSCLC. Therefore, complement C3b and C4d have potential for use as biomarkers to predict the risk of mortality in such patients.
Collapse
Affiliation(s)
- Yan Wang
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, P.R. China
| | - Mengqi Xiang
- Department of Medical Oncology, Sichuan Cancer Hospital, Medical School of University of Electronic Science and Technology of China, Chengdu, Sichuan 610000, P.R. China
| | - Huachuan Zhang
- Department of Thoracic Surgery, Sichuan Cancer Hospital, Medical School of University of Electronic Science and Technology of China, Chengdu, Sichuan 610000, P.R. China
| | - Yongda Lu
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, P.R. China
| |
Collapse
|
4
|
Sahu RK, Xavier S, Chauss D, Wang L, Chew C, Taylor R, Stallcup WB, Ma JZ, Kazemian M, Afzali B, Köhl J, Portilla D. Folic acid-mediated fibrosis is driven by C5a receptor 1-mediated activation of kidney myeloid cells. Am J Physiol Renal Physiol 2022; 322:F597-F610. [PMID: 35379003 PMCID: PMC9054266 DOI: 10.1152/ajprenal.00404.2021] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 03/21/2022] [Accepted: 03/30/2022] [Indexed: 11/22/2022] Open
Abstract
We have previously reported that increased expression and activation of kidney cell complement components play an important role in the pathogenesis of renal scarring. Here, we used floxed green fluorescent protein (GFP)-C5a receptor 1 (C5aR1) knockin mice (GFP-C5ar1fl/fl) and the model of folic acid (FA)-induced kidney injury to define the cell types and potential mechanisms by which increased C5aR1 activation leads to fibrosis. Using flow cytometry and confocal microscopy, we identified macrophages as the major interstitial cell type showing increased expression of C5aR1 in FA-treated mice. C5ar1fl/fl.Lyz2Cre+/- mice, in which C5aR1 has been specifically deleted in lysozyme M-expressing myeloid cells, experienced reduced fibrosis compared with control C5ar1fl/fl mice. Examination of C5aR1-expressing macrophage transcriptomes by gene set enrichment analysis demonstrated that these cells were enriched in pathways corresponding to the complement cascade, collagen formation, and the NABA matrisome, strongly pointing to their critical roles in tissue repair/scarring. Since C5aR1 was also detected in a small population of platelet-derived growth factor receptor-β+ GFP+ cells, we developed C5ar1fl/fl.Foxd1Cre+/- mice, in which C5aR1 is deleted specifically in pericytes, and found reduced FA-induced fibrosis. Primary cell cultures of platelet-derived growth factor receptor-β+ pericytes isolated from FA-treated C5ar1fl/fl.Foxd1Cre+/- mice showed reduced secretion of several cytokines, including IL-6 and macrophage inflammatory protein-2, compared with pericytes isolated from FA-treated control GFP-C5ar1fl/fl mice. Collectively, these data imply that C5a/C5aR1 axis activation primarily in interstitial cells contributes to the development of renal fibrosis.NEW & NOTEWORTHY This study used novel green fluorescent protein C5a receptor 1 floxed mice and the model of folic acid-mediated kidney fibrosis to demonstrate the pathogenic role of increased expression of this complement receptor on macrophages.
Collapse
Affiliation(s)
- Ranjit K Sahu
- Department of Medicine and Center for Immunity and Regenerative Medicine, University of Virginia, Charlottesville, Virginia
| | - Sandhya Xavier
- Department of Medicine and Center for Immunity and Regenerative Medicine, University of Virginia, Charlottesville, Virginia
| | - Daniel Chauss
- Immunoregulation Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Luopin Wang
- Departments of Biochemistry and Computer Science, Purdue University, West Lafayette, Indiana
| | - Claude Chew
- Flow Cytometry Core, University of Virginia, Charlottesville, Virginia
| | - Ronald Taylor
- Department of Biochemistry, University of Virginia, Charlottesville, Virginia
| | - William B Stallcup
- Sanford Burnham Prebys Medical Discovery Institute, Tumor Microenvironment and Cancer Immunology Program, La Jolla, California
| | - Jennie Z Ma
- Division of Biostatistics, Department of Public Health, University of Virginia, Charlottesville, Virginia
| | - Majid Kazemian
- Departments of Biochemistry and Computer Science, Purdue University, West Lafayette, Indiana
| | - Behdad Afzali
- Immunoregulation Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Jörg Köhl
- Institute for Systemic Inflammation Research, University of Lübeck, Germany
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Didier Portilla
- Department of Medicine and Center for Immunity and Regenerative Medicine, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
5
|
Westacott LJ, Wilkinson LS. Complement Dependent Synaptic Reorganisation During Critical Periods of Brain Development and Risk for Psychiatric Disorder. Front Neurosci 2022; 16:840266. [PMID: 35600620 PMCID: PMC9120629 DOI: 10.3389/fnins.2022.840266] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 03/09/2022] [Indexed: 12/12/2022] Open
Abstract
We now know that the immune system plays a major role in the complex processes underlying brain development throughout the lifespan, carrying out a number of important homeostatic functions under physiological conditions in the absence of pathological inflammation or infection. In particular, complement-mediated synaptic pruning during critical periods of early life may play a key role in shaping brain development and subsequent risk for psychopathology, including neurodevelopmental disorders such as schizophrenia and autism spectrum disorders. However, these disorders vary greatly in their onset, disease course, and prevalence amongst sexes suggesting complex interactions between the immune system, sex and the unique developmental trajectories of circuitries underlying different brain functions which are yet to be fully understood. Perturbations of homeostatic neuroimmune interactions during different critical periods in which regional circuits mature may have a plethora of long-term consequences for psychiatric phenotypes, but at present there is a gap in our understanding of how these mechanisms may impact on the structural and functional changes occurring in the brain at different developmental stages. In this article we will consider the latest developments in the field of complement mediated synaptic pruning where our understanding is beginning to move beyond the visual system where this process was first described, to brain areas and developmental periods of potential relevance to psychiatric disorders.
Collapse
Affiliation(s)
- Laura J. Westacott
- Neuroscience and Mental Health Innovation Institute, MRC Centre for Neuropsychiatric Genetic and Genomics, School of Medicine, Cardiff University, Cardiff, United Kingdom
- Hodge Centre for Neuropsychiatric Immunology, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Lawrence S. Wilkinson
- Neuroscience and Mental Health Innovation Institute, MRC Centre for Neuropsychiatric Genetic and Genomics, School of Medicine, Cardiff University, Cardiff, United Kingdom
- Hodge Centre for Neuropsychiatric Immunology, School of Medicine, Cardiff University, Cardiff, United Kingdom
- Behavioural Genetics Group, Schools of Psychology and Medicine, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
6
|
Gaboriaud C, Lorvellec M, Rossi V, Dumestre-Pérard C, Thielens NM. Complement System and Alarmin HMGB1 Crosstalk: For Better or Worse. Front Immunol 2022; 13:869720. [PMID: 35572583 PMCID: PMC9095977 DOI: 10.3389/fimmu.2022.869720] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 04/04/2022] [Indexed: 12/21/2022] Open
Abstract
Our immune system responds to infectious (PAMPs) and tissue damage (DAMPs) signals. The complement system and alarmin High-Mobility Group Box 1 (HMGB1) are two powerful soluble actors of human host defense and immune surveillance. These systems involve molecular cascades and amplification loops for their signaling or activation. Initially activated as alarm raising systems, their function can be finally switched towards inflammation resolution, where they sustain immune maturation and orchestrate repair mechanisms, opening the way back to homeostasis. However, when getting out of control, these defense systems can become deleterious and trigger serious cellular and tissue damage. Therefore, they can be considered as double-edged swords. The close interaction between the complement and HMGB1 pathways is described here, as well as their traditional and non-canonical roles, their functioning at different locations and their independent and collective impact in different systems both in health and disease. Starting from these systems and interplay at the molecular level (when elucidated), we then provide disease examples to better illustrate the signs and consequences of their roles and interaction, highlighting their importance and possible vicious circles in alarm raising and inflammation, both individually or in combination. Although this integrated view may open new therapeutic strategies, future challenges have to be faced because of the remaining unknowns regarding the molecular mechanisms underlying the fragile molecular balance which can drift towards disease or return to homeostasis, as briefly discussed at the end.
Collapse
Affiliation(s)
| | | | | | - Chantal Dumestre-Pérard
- Univ. Grenoble Alpes, CEA, CNRS, IBS, Grenoble, France
- Laboratoire d’Immunologie, Pôle de Biologie, CHU Grenoble Alpes, Grenoble, France
| | | |
Collapse
|
7
|
Westacott LJ, Humby T, Haan N, Brain SA, Bush EL, Toneva M, Baloc AI, Moon AL, Reddaway J, Owen MJ, Hall J, Hughes TR, Morgan BP, Gray WP, Wilkinson LS. Complement C3 and C3aR mediate different aspects of emotional behaviours; relevance to risk for psychiatric disorder. Brain Behav Immun 2022; 99:70-82. [PMID: 34543680 DOI: 10.1016/j.bbi.2021.09.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 09/03/2021] [Accepted: 09/13/2021] [Indexed: 12/18/2022] Open
Abstract
Complement is a key component of the immune system with roles in inflammation and host-defence. Here we reveal novel functions of complement pathways impacting on emotional reactivity of potential relevance to the emerging links between complement and risk for psychiatric disorder. We used mouse models to assess the effects of manipulating components of the complement system on emotionality. Mice lacking the complement C3a Receptor (C3aR-/-) demonstrated a selective increase in unconditioned (innate) anxiety whilst mice deficient in the central complement component C3 (C3-/-) showed a selective increase in conditioned (learned) fear. The dissociable behavioural phenotypes were linked to different signalling mechanisms. Effects on innate anxiety were independent of C3a, the canonical ligand for C3aR, consistent with the existence of an alternative ligand mediating innate anxiety, whereas effects on learned fear were due to loss of iC3b/CR3 signalling. Our findings show that specific elements of the complement system and associated signalling pathways contribute differentially to heightened states of anxiety and fear commonly seen in psychopathology.
Collapse
Affiliation(s)
- Laura J Westacott
- Neuroscience and Mental Health Research Institute, MRC Centre for Neuropsychiatric Genetic and Genomics, School of Medicine, Hadyn Ellis Building, Cardiff University, Cardiff CF24 4HQ, UK; Hodge Centre for Neuropsychiatric Immunology, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK
| | - Trevor Humby
- Neuroscience and Mental Health Research Institute, MRC Centre for Neuropsychiatric Genetic and Genomics, School of Medicine, Hadyn Ellis Building, Cardiff University, Cardiff CF24 4HQ, UK; Behavioural Genetics Group, Schools of Psychology and Medicine, Cardiff University, Cardiff CF10 3AT, UK
| | - Niels Haan
- Neuroscience and Mental Health Research Institute, MRC Centre for Neuropsychiatric Genetic and Genomics, School of Medicine, Hadyn Ellis Building, Cardiff University, Cardiff CF24 4HQ, UK
| | - Sophie A Brain
- Behavioural Genetics Group, Schools of Psychology and Medicine, Cardiff University, Cardiff CF10 3AT, UK
| | - Emma-Louise Bush
- Behavioural Genetics Group, Schools of Psychology and Medicine, Cardiff University, Cardiff CF10 3AT, UK
| | - Margarita Toneva
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, UK
| | - Andreea-Ingrid Baloc
- Behavioural Genetics Group, Schools of Psychology and Medicine, Cardiff University, Cardiff CF10 3AT, UK
| | - Anna L Moon
- Neuroscience and Mental Health Research Institute, MRC Centre for Neuropsychiatric Genetic and Genomics, School of Medicine, Hadyn Ellis Building, Cardiff University, Cardiff CF24 4HQ, UK
| | - Jack Reddaway
- Neuroscience and Mental Health Research Institute, MRC Centre for Neuropsychiatric Genetic and Genomics, School of Medicine, Hadyn Ellis Building, Cardiff University, Cardiff CF24 4HQ, UK; Hodge Centre for Neuropsychiatric Immunology, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK
| | - Michael J Owen
- Neuroscience and Mental Health Research Institute, MRC Centre for Neuropsychiatric Genetic and Genomics, School of Medicine, Hadyn Ellis Building, Cardiff University, Cardiff CF24 4HQ, UK
| | - Jeremy Hall
- Neuroscience and Mental Health Research Institute, MRC Centre for Neuropsychiatric Genetic and Genomics, School of Medicine, Hadyn Ellis Building, Cardiff University, Cardiff CF24 4HQ, UK; Hodge Centre for Neuropsychiatric Immunology, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK
| | - Timothy R Hughes
- Complement Biology Group, Systems Immunity Research Institute, School of Medicine, Cardiff University, Cardiff CF14 4XW, UK; Hodge Centre for Neuropsychiatric Immunology, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK
| | - B Paul Morgan
- Complement Biology Group, Systems Immunity Research Institute, School of Medicine, Cardiff University, Cardiff CF14 4XW, UK; Hodge Centre for Neuropsychiatric Immunology, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK; UK Dementia Research Institute, Cardiff University, Cardiff CF24 4HQ, UK
| | - William P Gray
- Neuroscience and Mental Health Research Institute, MRC Centre for Neuropsychiatric Genetic and Genomics, School of Medicine, Hadyn Ellis Building, Cardiff University, Cardiff CF24 4HQ, UK; Hodge Centre for Neuropsychiatric Immunology, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK; Brain Repair and Intracranial Therapeutics (BRAIN) Unit, School of Medicine, Cardiff University, CF24 4HQ, UK
| | - Lawrence S Wilkinson
- Neuroscience and Mental Health Research Institute, MRC Centre for Neuropsychiatric Genetic and Genomics, School of Medicine, Hadyn Ellis Building, Cardiff University, Cardiff CF24 4HQ, UK; Behavioural Genetics Group, Schools of Psychology and Medicine, Cardiff University, Cardiff CF10 3AT, UK; Hodge Centre for Neuropsychiatric Immunology, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK.
| |
Collapse
|
8
|
Xiang M, Zhang H, Kou L, Chen J, Xu Z, He J. Low level of complement factor H increases the risk of cancer-related death in patients with small-cell lung cancer. Postgrad Med J 2021; 98:919-924. [PMID: 34725230 DOI: 10.1136/postgradmedj-2021-141186] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 10/08/2021] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Pulmonary cancer is a kind of deeply invasive tumour which is difficult to treat, and its mortality rate is high. Previous research has shown that activation of complement could contribute to the progression of non-small-cell lung cancer (SCLC). However, little research has been done on SCLC. METHODS Complement factor H (CFH), complements C3 as well as C4 were measured in patients, and the prognostic impact of different parameters was assessed by log-rank function analysis and Cox multifactor models. Besides, we constructed a predictive model based on complement fractions and validated the accuracy of the model. RESULTS Among these 242 patients, 200 (82.6%) died. The median survival time was 18.3 months. We found by multifactorial analysis that high levels of CFH decreased the risk of death (HR 0.23, 95% CI 0.10 to 0.57, p<0.001), while elevated complement C4 displayed poor prognosis (HR 2.28, 95% CI 1.66 to 3.13, p<0.001). We screened variables by Cox models and constructed CFH-based prediction models to plot a nomogram by internal validation. The nomogram showed excellent accuracy in assessing the probability of death, yielding an adjusted C-statistics of 0.905. CONCLUSIONS CFH can be recognised as a biomarker to predict the risk of death in SCLC. The prediction model established based on CFH, C3 and C4 levels has good accuracy in patients' prognostic assessment.
Collapse
Affiliation(s)
- Mengqi Xiang
- Department of Medical Oncology, Sichuan Cancer Hospital and Institute, Chengdu, Sichuan, China
| | - Huachuan Zhang
- Department of Thoracic Surgery, Sichuan Cancer Hospital and Institute, Chengdu, Sichuan, China
| | - Lingna Kou
- Department of Medical Oncology, Sichuan Cancer Hospital and Institute, Chengdu, Sichuan, China
| | - Jing Chen
- Department of Medical Oncology, Sichuan Cancer Hospital and Institute, Chengdu, Sichuan, China
| | - Zhihua Xu
- General Surgery, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jintao He
- Department of Thoracic Surgery, Sichuan Cancer Hospital and Institute, Chengdu, Sichuan, China
| |
Collapse
|
9
|
Bomback AS, Appel GB, Gipson DS, Hladunewich MA, Lafayette R, Nester CM, Parikh SV, Smith RJH, Trachtman H, Heeger PS, Ram S, Rovin BH, Ali S, Arceneaux N, Ashoor I, Bailey-Wickins L, Barratt J, Beck L, Cattran DC, Cravedi P, Erkan E, Fervenza F, Frazer-Abel AA, Fremeaux-Bacchi V, Fuller L, Gbadegesin R, Hogan JJ, Kiryluk K, le Quintrec-Donnette M, Licht C, Mahan JD, Pickering MC, Quigg R, Rheault M, Ronco P, Sarwal MM, Sethna C, Spino C, Stegall M, Vivarelli M, Feldman DL, Thurman JM. Improving Clinical Trials for Anticomplement Therapies in Complement-Mediated Glomerulopathies: Report of a Scientific Workshop Sponsored by the National Kidney Foundation. Am J Kidney Dis 2021; 79:570-581. [PMID: 34571062 DOI: 10.1053/j.ajkd.2021.07.025] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 07/13/2021] [Indexed: 12/25/2022]
Abstract
Blocking the complement system as a therapeutic strategy has been proposed for numerous glomerular diseases but presents myriad questions and challenges, not the least of which is demonstrating efficacy and safety. In light of these potential issues and because there are an increasing number of anticomplement therapy trials either planned or under way, the National Kidney Foundation facilitated an all-virtual scientific workshop entitled "Improving Clinical Trials for Anti-Complement Therapies in Complement-Mediated Glomerulopathies." Attended by patient representatives and experts in glomerular diseases, complement physiology, and clinical trial design, the aim of this workshop was to develop standards applicable for designing and conducting clinical trials for anticomplement therapies across a wide spectrum of complement-mediated glomerulopathies. Discussions focused on study design, participant risk assessment and mitigation, laboratory measurements and biomarkers to support these studies, and identification of optimal outcome measures to detect benefit, specifically for trials in complement-mediated diseases. This report summarizes the discussions from this workshop and outlines consensus recommendations.
Collapse
Affiliation(s)
- Andrew S Bomback
- Division of Nephrology, Columbia University Irving Medical Center, New York.
| | - Gerald B Appel
- Division of Nephrology, New York University Langone Health, New York
| | - Debbie S Gipson
- Department of Pediatrics, Division of Nephrology, University of Michigan, Ann Arbor, Michigan
| | | | | | - Carla M Nester
- Division of Nephrology, University of Iowa, Iowa City, Iowa
| | - Samir V Parikh
- Division of Nephrology, The Ohio State University College of Medicine, Columbus, Ohio
| | - Richard J H Smith
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa
| | - Howard Trachtman
- Division of Nephrology, New York University Langone Health, New York
| | - Peter S Heeger
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York
| | - Sanjay Ram
- Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Brad H Rovin
- Division of Nephrology, The Ohio State University College of Medicine, Columbus, Ohio
| | | | | | - Isa Ashoor
- Division of Nephrology, Louisiana State University Health, New Orleans, Louisiana
| | | | | | - Laurence Beck
- Division of Nephrology, Boston University School of Medicine, Boston, Massachusetts
| | - Daniel C Cattran
- Division of Nephrology, University of Toronto, Toronto, ON, Canada
| | - Paolo Cravedi
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York
| | - Elif Erkan
- Division of Nephrology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | | | - Ashley A Frazer-Abel
- Division of Nephrology, University of Colorado School of Medicine, Aurora, Colorado
| | | | | | | | - Jonathan J Hogan
- Division of Nephrology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Krzysztof Kiryluk
- Division of Nephrology, Columbia University Irving Medical Center, New York
| | | | - Christoph Licht
- Division of Nephrology, University of Toronto, Toronto, ON, Canada
| | - John D Mahan
- Division of Nephrology, The Ohio State University College of Medicine, Columbus, Ohio
| | | | - Richard Quigg
- Division of Nephrology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York
| | - Michelle Rheault
- Division of Nephrology, University of Minnesota, Minneapolis, Minnesota
| | - Pierre Ronco
- Division of Nephrology, Sorbonne Université, Université Pierre et Marie Curie, Paris
| | - Minnie M Sarwal
- Division of Nephrology, University of California, San Francisco, California
| | - Christine Sethna
- Division of Nephrology, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York
| | - Cathie Spino
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan
| | | | - Marina Vivarelli
- Division of Nephrology, Bambino Gesu Children's Hospital, Rome, Italy
| | | | - Joshua M Thurman
- Division of Nephrology, University of Colorado School of Medicine, Aurora, Colorado
| |
Collapse
|
10
|
Shimazaki R, Takano S, Satoh M, Takada M, Miyahara Y, Sasaki K, Yoshitomi H, Kagawa S, Furukawa K, Takayashiki T, Kuboki S, Sogawa K, Motohashi S, Nomura F, Miyazaki M, Ohtsuka M. Complement factor B regulates cellular senescence and is associated with poor prognosis in pancreatic cancer. Cell Oncol (Dordr) 2021; 44:937-950. [PMID: 34075561 PMCID: PMC8338870 DOI: 10.1007/s13402-021-00614-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 05/19/2021] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND The interplay between cancer cells and stromal components, including soluble mediators released from cancer cells, contributes to the progression of pancreatic ductal adenocarcinoma (PDAC). Here, we set out to identify key secreted proteins involved in PDAC progression. METHODS We performed secretome analyses of culture media of mouse pancreatic intraepithelial neoplasia (PanIN) and PDAC cells using Stable Isotope Labeling by Amino acid in Cell culture (SILAC) with click chemistry and liquid chromatography-mass spectrometry (LC-MS/MS). The results obtained were verified in primary PDAC tissue samples and cell line models. RESULTS Complement factor B (CFB) was identified as one of the robustly upregulated proteins, and found to exhibit elevated expression in PDAC cells compared to PanIN cells. Endogenous CFB knockdown by a specific siRNA dramatically decreased the proliferation of PDAC cells, PANC-1 and MIA PaCa-II. CFB knockdown induced increases in the number of senescence-associated-β-galactosidase (SA-β-gal) positive cells exhibiting p21 expression upregulation, which promotes cellular senescence with cyclinD1 accumulation. Furthermore, CFB knockdown facilitated downregulation of proliferating cell nuclear antigen and led to cell cycle arrest in the G1 phase in PDAC cells. Using immunohistochemistry, we found that high stromal CFB expression was associated with unfavorable clinical outcomes with hematogenous dissemination after surgery in human PDAC patients. Despite the presence of enriched CD8+ tumor infiltrating lymphocytes in the PDAC tumor microenvironments, patients with a high stromal CFB expression exhibited a significantly poorer prognosis compared to those with a low stromal CFB expression. Immunofluorescence staining revealed a correlation between stromal CFB expression in the tumor microenvironment and an enrichment of immunosuppressive regulatory T-cells (Tregs), myeloid-derived suppressor cells (MDSCs) and tumor-associated macrophages (TAMs). We also found that high stromal CFB expression showed a positive correlation with high CD8+/Foxp3+ Tregs populations in PDAC tissues. CONCLUSIONS Our data indicate that CFB, a key secreted protein, promotes proliferation by preventing cellular senescence and is associated with immunological tumor promotion in PDAC. These findings suggest that CFB may be a potential target for the treatment of PDAC.
Collapse
Affiliation(s)
- Reiri Shimazaki
- Department of General Surgery, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba-shi, Chiba, 260- 8677, Japan
| | - Shigetsugu Takano
- Department of General Surgery, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba-shi, Chiba, 260- 8677, Japan.
| | - Mamoru Satoh
- Division of Clinical Mass Spectrometry, Chiba University Hospital, Chiba, 260-8677, Japan
| | - Mamoru Takada
- Department of General Surgery, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba-shi, Chiba, 260- 8677, Japan
| | - Yoji Miyahara
- Department of General Surgery, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba-shi, Chiba, 260- 8677, Japan
| | - Kosuke Sasaki
- Department of General Surgery, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba-shi, Chiba, 260- 8677, Japan
| | - Hideyuki Yoshitomi
- Department of General Surgery, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba-shi, Chiba, 260- 8677, Japan
| | - Shingo Kagawa
- Department of General Surgery, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba-shi, Chiba, 260- 8677, Japan
| | - Katsunori Furukawa
- Department of General Surgery, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba-shi, Chiba, 260- 8677, Japan
| | - Tsukasa Takayashiki
- Department of General Surgery, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba-shi, Chiba, 260- 8677, Japan
| | - Satoshi Kuboki
- Department of General Surgery, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba-shi, Chiba, 260- 8677, Japan
| | - Kazuyuki Sogawa
- Department of Biochemistry, School of Life and Environmental Science, Azabu University, 252-5201, Kanagawa, Japan
| | - Shinichiro Motohashi
- Department of Medical Immunology, Graduate School of Medicine, Chiba University, 260-8677, Chiba, Japan
| | - Fumio Nomura
- Division of Clinical Mass Spectrometry, Chiba University Hospital, Chiba, 260-8677, Japan
| | - Masaru Miyazaki
- Department of General Surgery, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba-shi, Chiba, 260- 8677, Japan
| | - Masayuki Ohtsuka
- Department of General Surgery, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba-shi, Chiba, 260- 8677, Japan
| |
Collapse
|
11
|
Portilla D, Xavier S. Role of intracellular complement activation in kidney fibrosis. Br J Pharmacol 2021; 178:2880-2891. [PMID: 33555070 DOI: 10.1111/bph.15408] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/22/2021] [Accepted: 02/02/2021] [Indexed: 02/06/2023] Open
Abstract
Increased expression of complement C1r, C1s and C3 in kidney cells plays an important role in the pathogenesis of kidney fibrosis. Our studies suggest that activation of complement in kidney cells with increased generation of C3 and its fragments occurs by activation of classical and alternative pathways. Single nuclei RNA sequencing studies in kidney tissue from unilateral ureteral obstruction mice show that increased synthesis of complement C3 and C5 occurs primarily in renal tubular epithelial cells (proximal and distal), while increased expression of complement receptors C3ar1 and C5ar1 occurs in interstitial cells including immune cells like monocytes/macrophages suggesting compartmentalization of complement components during kidney injury. Although global deletion of C3 and macrophage ablation prevent inflammation and reduced kidney tissue scarring, the development of mice with cell-specific deletion of complement components and their regulators could bring further insights into the mechanisms by which intracellular complement activation leads to fibrosis and progressive kidney disease. LINKED ARTICLES: This article is part of a themed issue on Canonical and non-canonical functions of the complement system in health and disease. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.14/issuetoc.
Collapse
Affiliation(s)
- Didier Portilla
- Department of Medicine and Center for Immunity and Regenerative Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Sandhya Xavier
- Department of Medicine and Center for Immunity and Regenerative Medicine, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
12
|
Garred P, Tenner AJ, Mollnes TE. Therapeutic Targeting of the Complement System: From Rare Diseases to Pandemics. Pharmacol Rev 2021; 73:792-827. [PMID: 33687995 PMCID: PMC7956994 DOI: 10.1124/pharmrev.120.000072] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The complement system was discovered at the end of the 19th century as a heat-labile plasma component that "complemented" the antibodies in killing microbes, hence the name "complement." Complement is also part of the innate immune system, protecting the host by recognition of pathogen-associated molecular patterns. However, complement is multifunctional far beyond infectious defense. It contributes to organ development, such as sculpting neuron synapses, promoting tissue regeneration and repair, and rapidly engaging and synergizing with a number of processes, including hemostasis leading to thromboinflammation. Complement is a double-edged sword. Although it usually protects the host, it may cause tissue damage when dysregulated or overactivated, such as in the systemic inflammatory reaction seen in trauma and sepsis and severe coronavirus disease 2019 (COVID-19). Damage-associated molecular patterns generated during ischemia-reperfusion injuries (myocardial infarction, stroke, and transplant dysfunction) and in chronic neurologic and rheumatic disease activate complement, thereby increasing damaging inflammation. Despite the long list of diseases with potential for ameliorating complement modulation, only a few rare diseases are approved for clinical treatment targeting complement. Those currently being efficiently treated include paroxysmal nocturnal hemoglobinuria, atypical hemolytic-uremic syndrome, myasthenia gravis, and neuromyelitis optica spectrum disorders. Rare diseases, unfortunately, preclude robust clinical trials. The increasing evidence for complement as a pathogenetic driver in many more common diseases suggests an opportunity for future complement therapy, which, however, requires robust clinical trials; one ongoing example is COVID-19 disease. The current review aims to discuss complement in disease pathogenesis and discuss future pharmacological strategies to treat these diseases with complement-targeted therapies. SIGNIFICANCE STATEMENT: The complement system is the host's defense friend by protecting it from invading pathogens, promoting tissue repair, and maintaining homeostasis. Complement is a double-edged sword, since when dysregulated or overactivated it becomes the host's enemy, leading to tissue damage, organ failure, and, in worst case, death. A number of acute and chronic diseases are candidates for pharmacological treatment to avoid complement-dependent damage, ranging from the well established treatment for rare diseases to possible future treatment of large patient groups like the pandemic coronavirus disease 2019.
Collapse
Affiliation(s)
- Peter Garred
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Rigshospitalet, Copenhagen, Denmark, and Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (P.G.); Departments of Molecular Biology and Biochemistry, Neurobiology and Behavior, and Pathology and Laboratory Medicine, University of California, Irvine, California (A.J.T.); and Research Laboratory, Nordland Hospital, Bodø, Norway, Faculty of Health Sciences, K.G. Jebsen TREC, University of Tromsø, Tromsø, Norway (T.E.M.); Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim, Norway (T.E.M.); and Department of Immunology, Oslo University Hospital and University of Oslo, Oslo, Norway (T.E.M.)
| | - Andrea J Tenner
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Rigshospitalet, Copenhagen, Denmark, and Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (P.G.); Departments of Molecular Biology and Biochemistry, Neurobiology and Behavior, and Pathology and Laboratory Medicine, University of California, Irvine, California (A.J.T.); and Research Laboratory, Nordland Hospital, Bodø, Norway, Faculty of Health Sciences, K.G. Jebsen TREC, University of Tromsø, Tromsø, Norway (T.E.M.); Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim, Norway (T.E.M.); and Department of Immunology, Oslo University Hospital and University of Oslo, Oslo, Norway (T.E.M.)
| | - Tom E Mollnes
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Rigshospitalet, Copenhagen, Denmark, and Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (P.G.); Departments of Molecular Biology and Biochemistry, Neurobiology and Behavior, and Pathology and Laboratory Medicine, University of California, Irvine, California (A.J.T.); and Research Laboratory, Nordland Hospital, Bodø, Norway, Faculty of Health Sciences, K.G. Jebsen TREC, University of Tromsø, Tromsø, Norway (T.E.M.); Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim, Norway (T.E.M.); and Department of Immunology, Oslo University Hospital and University of Oslo, Oslo, Norway (T.E.M.)
| |
Collapse
|
13
|
Luntzer K, Lackner I, Weber B, Mödinger Y, Ignatius A, Gebhard F, Mihaljevic SY, Haffner-Luntzer M, Kalbitz M. Increased Presence of Complement Factors and Mast Cells in Alveolar Bone and Tooth Resorption. Int J Mol Sci 2021; 22:ijms22052759. [PMID: 33803323 PMCID: PMC7967164 DOI: 10.3390/ijms22052759] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 02/24/2021] [Accepted: 03/03/2021] [Indexed: 02/07/2023] Open
Abstract
Periodontitis is the inflammatory destruction of the tooth-surrounding and -supporting tissue, resulting at worst in tooth loss. Another locally aggressive disease of the oral cavity is tooth resorption (TR). This is associated with the destruction of the dental mineralized tissue. However, the underlying pathomechanisms remain unknown. The complement system, as well as mast cells (MCs), are known to be involved in osteoclastogenesis and bone loss. The complement factors C3 and C5 were previously identified as key players in periodontal disease. Therefore, we hypothesize that complement factors and MCs might play a role in alveolar bone and tooth resorption. To investigate this, we used the cat as a model because of the naturally occurring high prevalence of both these disorders in this species. Teeth, gingiva samples and serum were collected from domestic cats, which had an appointment for dental treatment under anesthesia, as well as from healthy cats. Histological analyses, immunohistochemical staining and the CH-50 and AH-50 assays revealed increased numbers of osteoclasts and MCs, as well as complement activity in cats with TR. Calcifications score in the gingiva was highest in animals that suffer from TR. This indicates that MCs and the complement system are involved in the destruction of the mineralized tissue in this condition.
Collapse
Affiliation(s)
- Kathrin Luntzer
- Center for Trauma Research Ulm (ZTF), University of Ulm, 89081 Ulm, Germany; (K.L.); (I.L.); (B.W.); (Y.M.); (A.I.); (F.G.); (M.H.-L.)
- Department of Traumatology, Hand-, Plastic-, and Reconstructive Surgery, University Medical Center, 89081 Ulm, Germany
- Small Animal Clinic Ravensburg Evidensia GmbH, Eywiesenstraße 4, 88212 Ravensburg, Germany
| | - Ina Lackner
- Center for Trauma Research Ulm (ZTF), University of Ulm, 89081 Ulm, Germany; (K.L.); (I.L.); (B.W.); (Y.M.); (A.I.); (F.G.); (M.H.-L.)
- Department of Traumatology, Hand-, Plastic-, and Reconstructive Surgery, University Medical Center, 89081 Ulm, Germany
| | - Birte Weber
- Center for Trauma Research Ulm (ZTF), University of Ulm, 89081 Ulm, Germany; (K.L.); (I.L.); (B.W.); (Y.M.); (A.I.); (F.G.); (M.H.-L.)
- Department of Traumatology, Hand-, Plastic-, and Reconstructive Surgery, University Medical Center, 89081 Ulm, Germany
| | - Yvonne Mödinger
- Center for Trauma Research Ulm (ZTF), University of Ulm, 89081 Ulm, Germany; (K.L.); (I.L.); (B.W.); (Y.M.); (A.I.); (F.G.); (M.H.-L.)
- Institute of Orthopedic Research and Biomechanics, University of Ulm, 89081 Ulm, Germany
| | - Anita Ignatius
- Center for Trauma Research Ulm (ZTF), University of Ulm, 89081 Ulm, Germany; (K.L.); (I.L.); (B.W.); (Y.M.); (A.I.); (F.G.); (M.H.-L.)
- Institute of Orthopedic Research and Biomechanics, University of Ulm, 89081 Ulm, Germany
| | - Florian Gebhard
- Center for Trauma Research Ulm (ZTF), University of Ulm, 89081 Ulm, Germany; (K.L.); (I.L.); (B.W.); (Y.M.); (A.I.); (F.G.); (M.H.-L.)
- Department of Traumatology, Hand-, Plastic-, and Reconstructive Surgery, University Medical Center, 89081 Ulm, Germany
| | | | - Melanie Haffner-Luntzer
- Center for Trauma Research Ulm (ZTF), University of Ulm, 89081 Ulm, Germany; (K.L.); (I.L.); (B.W.); (Y.M.); (A.I.); (F.G.); (M.H.-L.)
- Institute of Orthopedic Research and Biomechanics, University of Ulm, 89081 Ulm, Germany
| | - Miriam Kalbitz
- Center for Trauma Research Ulm (ZTF), University of Ulm, 89081 Ulm, Germany; (K.L.); (I.L.); (B.W.); (Y.M.); (A.I.); (F.G.); (M.H.-L.)
- Department of Traumatology, Hand-, Plastic-, and Reconstructive Surgery, University Medical Center, 89081 Ulm, Germany
- Correspondence:
| |
Collapse
|
14
|
Schartz ND, Tenner AJ. The good, the bad, and the opportunities of the complement system in neurodegenerative disease. J Neuroinflammation 2020; 17:354. [PMID: 33239010 PMCID: PMC7690210 DOI: 10.1186/s12974-020-02024-8] [Citation(s) in RCA: 158] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 11/04/2020] [Indexed: 02/06/2023] Open
Abstract
The complement cascade is a critical effector mechanism of the innate immune system that contributes to the rapid clearance of pathogens and dead or dying cells, as well as contributing to the extent and limit of the inflammatory immune response. In addition, some of the early components of this cascade have been clearly shown to play a beneficial role in synapse elimination during the development of the nervous system, although excessive complement-mediated synaptic pruning in the adult or injured brain may be detrimental in multiple neurogenerative disorders. While many of these later studies have been in mouse models, observations consistent with this notion have been reported in human postmortem examination of brain tissue. Increasing awareness of distinct roles of C1q, the initial recognition component of the classical complement pathway, that are independent of the rest of the complement cascade, as well as the relationship with other signaling pathways of inflammation (in the periphery as well as the central nervous system), highlights the need for a thorough understanding of these molecular entities and pathways to facilitate successful therapeutic design, including target identification, disease stage for treatment, and delivery in specific neurologic disorders. Here, we review the evidence for both beneficial and detrimental effects of complement components and activation products in multiple neurodegenerative disorders. Evidence for requisite co-factors for the diverse consequences are reviewed, as well as the recent studies that support the possibility of successful pharmacological approaches to suppress excessive and detrimental complement-mediated chronic inflammation, while preserving beneficial effects of complement components, to slow the progression of neurodegenerative disease.
Collapse
Affiliation(s)
- Nicole D. Schartz
- Department of Molecular Biology and Biochemistry, University of California Irvine, 3205 McGaugh Hall, Irvine, CA 92697 USA
| | - Andrea J. Tenner
- Department of Molecular Biology and Biochemistry, University of California Irvine, 3205 McGaugh Hall, Irvine, CA 92697 USA
- Department of Neurobiology and Behavior, University of California Irvine, 3205 McGaugh Hall, Irvine, CA 92697 USA
- Department of Pathology and Laboratory Medicine, University of California Irvine, 3205 McGaugh Hall, Irvine, CA 92697 USA
| |
Collapse
|
15
|
Li J, Cao Z, Mi L, Xu Z, Wu X. Complement sC5b-9 and CH50 increase the risk of cancer-related mortality in patients with non-small cell lung cancer. J Cancer 2020; 11:7157-7165. [PMID: 33193878 PMCID: PMC7646172 DOI: 10.7150/jca.46721] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 10/08/2020] [Indexed: 12/29/2022] Open
Abstract
Objectives: Immunologic dysfunction occurred in most of patients with non-small cell lung cancer (NSCLC), which worsened the overall survival (OS) of patients. Complement activation plays a significant role in abnormal activation of immune system. However, the prognostic value of complement components such as CH50 and sC5b-9 in NSCLC patients remains unclear. This study evaluated the risk factors of NSCLC and created a prediction model. Methods: A real-world study was conducted including data from 928 patients with NSCLC between April 1, 2005 and June 1, 2015. CH50 and sC5b-9 were recorded during the admission. Cox proportional hazard model was applied for survival analyses and for assessing risk factors of cancer-related mortality and to create a nomogram for prediction. The accuracy of the model was evaluated by C-index and calibration curve. Results: In this study, the mortality in group with high CH50 level (≥ 480.56 umol/L) was 92.0%. Based on univariate analysis, we put factors (P <0.05) into a multivariate regression model, patients with high CH50 level (P <0.001, HR=1.59) and sC5b-9 >1422.18 μmol/L (P <0.001, HR=2.28) remained statistically factors for worsened OS and regarded as independent risk factors. These independently associated risk factors were applied to establish an OS estimation nomogram. Nomogram revealed good accuracy in estimating the risk, with a bootstrap-corrected C index of 0.741. Conclusion: sC5b-9 and CH50 increased the risk of cancer-related mortality in patients with NSCLC. Nomogram based on multivariate analysis demonstrated good accuracy in estimating the risk of overall mortality.
Collapse
Affiliation(s)
- Jing Li
- Department of Medicine, Respiratory, Emergency and Intensive Care Medicine, The Affiliated Dushu Lake Hospital of Soochow University, Suzhou, China
| | - Zhijun Cao
- Department of Urology, The Ninth People's Hospital of Suzhou, Suzhou, China
| | - Lijie Mi
- Department of Cardiovascular, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhihua Xu
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiangmei Wu
- Department of Endocrinology, Suzhou Xiangcheng People's Hospital, Suzhou, China
| |
Collapse
|
16
|
Barnum SR, Bubeck D, Schein TN. Soluble Membrane Attack Complex: Biochemistry and Immunobiology. Front Immunol 2020; 11:585108. [PMID: 33240274 PMCID: PMC7683570 DOI: 10.3389/fimmu.2020.585108] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 10/14/2020] [Indexed: 12/15/2022] Open
Abstract
The soluble membrane attack complex (sMAC, a.k.a., sC5b-9 or TCC) is generated on activation of complement and contains the complement proteins C5b, C6, C7, C8, C9 together with the regulatory proteins clusterin and/or vitronectin. sMAC is a member of the MACPF/cholesterol-dependent-cytolysin superfamily of pore-forming molecules that insert into lipid bilayers and disrupt cellular integrity and function. sMAC is a unique complement activation macromolecule as it is comprised of several different subunits. To date no complement-mediated function has been identified for sMAC. sMAC is present in blood and other body fluids under homeostatic conditions and there is abundant evidence documenting changes in sMAC levels during infection, autoimmune disease and trauma. Despite decades of scientific interest in sMAC, the mechanisms regulating its formation in healthy individuals and its biological functions in both health and disease remain poorly understood. Here, we review the structural differences between sMAC and its membrane counterpart, MAC, and examine sMAC immunobiology with respect to its presence in body fluids in health and disease. Finally, we discuss the diagnostic potential of sMAC for diagnostic and prognostic applications and potential utility as a companion diagnostic.
Collapse
Affiliation(s)
| | - Doryen Bubeck
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | | |
Collapse
|
17
|
Smole U, Kratzer B, Pickl WF. Soluble pattern recognition molecules: Guardians and regulators of homeostasis at airway mucosal surfaces. Eur J Immunol 2020; 50:624-642. [PMID: 32246830 PMCID: PMC7216992 DOI: 10.1002/eji.201847811] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 02/25/2020] [Accepted: 03/31/2020] [Indexed: 01/08/2023]
Abstract
Maintenance of homeostasis at body barriers that are constantly challenged by microbes, toxins and potentially bioactive (macro)molecules requires complex, highly orchestrated mechanisms of protection. Recent discoveries in respiratory research have shed light on the unprecedented role of airway epithelial cells (AEC), which, besides immune cells homing to the lung, also significantly contribute to host defence by expressing membrane‐bound and soluble pattern recognition receptors (sPRR). Recent evidence suggests that distinct, evolutionary ancient, sPRR secreted by AEC might become activated by usually innocuous proteins, commonly referred to as allergens. We here provide a systematic overview on sPRR detectable in the mucus lining of AEC. Some of them become actively produced and secreted by AECs (like the pentraxins C‐reactive protein and pentraxin 3; the collectins mannose binding protein and surfactant proteins A and D; H‐ficolin; serum amyloid A; and the complement components C3 and C5). Others are elaborated by innate and adaptive immune cells such as monocytes/macrophages and T cells (like the pentraxins C‐reactive protein and pentraxin 3; L‐ficolin; serum amyloid A; and the complement components C3 and C5). Herein we discuss how sPRRs may contribute to homeostasis but sometimes also to overt disease (e.g. airway hyperreactivity and asthma) at the alveolar–air interface.
Collapse
Affiliation(s)
- Ursula Smole
- Institute of ImmunologyCenter for PathophysiologyInfectiology and ImmunologyMedical University of ViennaViennaAustria
| | - Bernhard Kratzer
- Institute of ImmunologyCenter for PathophysiologyInfectiology and ImmunologyMedical University of ViennaViennaAustria
| | - Winfried F. Pickl
- Institute of ImmunologyCenter for PathophysiologyInfectiology and ImmunologyMedical University of ViennaViennaAustria
| |
Collapse
|
18
|
Schirmer B, Giehl K, Kubatzky KF. Report of the 23rd Meeting on Signal Transduction 2019-Trends in Cancer and Infection. Int J Mol Sci 2020; 21:ijms21082728. [PMID: 32326408 PMCID: PMC7215334 DOI: 10.3390/ijms21082728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 04/06/2020] [Indexed: 11/16/2022] Open
Abstract
The annual meeting "Signal Transduction-Receptors, Mediators and Genes" of the Signal Transduction Society (STS) is an interdisciplinary conference open to all scientists sharing the common interest in elucidating the signalling pathways underlying the physiological or pathological processes in health and disease of humans, animals, plants, fungi, prokaryotes and protists. The 23rd meeting on signal transduction was held from 4-6 November 2019 in Weimar, Germany, and focused on "Trends in Cancer and Infection". As usual, keynote presentations by invited scientists introduced the respective workshops and were followed by speakers chosen from the submitted abstracts. Ample time had been reserved for discussion of the presented data during the workshops. In this report, we provide a concise summary of the various workshops and further aspects of the scientific program.
Collapse
Affiliation(s)
- Bastian Schirmer
- Institute of Pharmacology, Hannover Medical School, 30625 Hannover, Germany
- Correspondence: ; Tel.: +49-511-532-3875
| | - Klaudia Giehl
- Signal Transduction of Cellular Motility, Internal Medicine V, Justus Liebig University Giessen, 35392 Giessen, Germany;
| | - Katharina F. Kubatzky
- Department of Medical Microbiology and Hygiene, Heidelberg University Hospital, 69120 Heidelberg, Germany;
| |
Collapse
|
19
|
Roumenina LT, Daugan MV, Petitprez F, Sautès-Fridman C, Fridman WH. Context-dependent roles of complement in cancer. Nat Rev Cancer 2019; 19:698-715. [PMID: 31666715 DOI: 10.1038/s41568-019-0210-0] [Citation(s) in RCA: 230] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/14/2019] [Indexed: 12/16/2022]
Abstract
The tumour microenvironment (TME) highly influences the growth and spread of tumours, thus impacting the patient's clinical outcome. In this context, the complement system plays a major and complex role. It may either act to kill antibody-coated tumour cells, support local chronic inflammation or hamper antitumour T cell responses favouring tumour progression. Recent studies demonstrate that these opposing effects are dependent upon the sites of complement activation, the composition of the TME and the tumour cell sensitivity to complement attack. In this Review, we present the evidence that has so far accrued showing a role for complement activation and its effects on cancer control and clinical outcome under different TME contexts. We also include a new analysis of the publicly available transcriptomic data to provide an overview of the prognostic value of complement gene expression in 30 cancer types. We argue that the interplay of complement components within each cancer type is unique, governed by the properties of the tumour cells and the TME. This concept is of critical importance for the design of efficient therapeutic strategies aimed at targeting complement components and their signalling.
Collapse
Affiliation(s)
- Lubka T Roumenina
- INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, Sorbonne Universités, Université de Paris, Paris, France.
| | - Marie V Daugan
- INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, Sorbonne Universités, Université de Paris, Paris, France
| | - Florent Petitprez
- INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, Sorbonne Universités, Université de Paris, Paris, France
- Programme Cartes d'Identité des Tumeurs, Ligue Nationale Contre le Cancer, Paris, France
| | - Catherine Sautès-Fridman
- INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, Sorbonne Universités, Université de Paris, Paris, France
| | - Wolf Herman Fridman
- INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, Sorbonne Universités, Université de Paris, Paris, France.
| |
Collapse
|
20
|
Xavier S, Sahu RK, Bontha SV, Mass V, Taylor RP, Megyesi J, Thielens NM, Portilla D. Complement C1r serine protease contributes to kidney fibrosis. Am J Physiol Renal Physiol 2019; 317:F1293-F1304. [PMID: 31509012 PMCID: PMC6879941 DOI: 10.1152/ajprenal.00357.2019] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 08/28/2019] [Accepted: 09/07/2019] [Indexed: 02/07/2023] Open
Abstract
We have previously reported that complement activation precedes the development of kidney fibrosis; however, little is known about the cellular mechanisms involved in this transition. We hypothesized that increased expression of C1 complex protease C1r, the initiator of complement activation, contributes to tubulointerstitial fibrosis and tested this idea in mice with global deletion of C1r. Although expression of C1r in untreated wild-type (WT) mice was higher in the liver compared with kidney tissue, administration of folic acid (FA) led to upregulation of C1r mRNA and protein levels only in kidney tissue. Immunohistochemistry and in situ hybridization experiments localized increased expression of C1r and C1s proteases to renal tubular epithelial cells. C1r-null mice had reduced acute tubular injury and inflammation measured 2 days after FA administration compared with WT mice. C1r deletion reduced expression of C1s, C3 fragment formation, and organ fibrosis measured 14 days after FA administration. Differential gene expression performed in kidney tissue demonstrated that C1r-null mice had reduced expression of genes associated with the acute phase response, complement, proliferation of connective tissue cells (e.g., platelet-derived growth factor receptor-β), and reduced expression of genes associated with inflammation compared with FA-treated WT mice. In vitro experiments in renal epithelial cells demonstrated that C1s expression is dependent on increased C1r expression and that interferon-γ induces the expression of these two proteases. We conclude that increased expression of C1 complex proteases is associated with increased tissue inflammation and complement C3 formation and represents an important pathogenic mechanism leading to FA-mediated tubulointerstitial fibrosis.
Collapse
Affiliation(s)
- Sandhya Xavier
- Division of Nephrology, Center for Immunity, Inflammation and Regenerative Medicine, University of Virginia, Charlottesville, Virginia
| | - Ranjit K Sahu
- Division of Nephrology, Center for Immunity, Inflammation and Regenerative Medicine, University of Virginia, Charlottesville, Virginia
| | - Sai Vineela Bontha
- Methodist University of Tennessee Transplant Institute, Memphis, Tennessee
| | - Valeria Mass
- Methodist University of Tennessee Transplant Institute, Memphis, Tennessee
| | - Ronald P Taylor
- Department of Biochemistry, University of Virginia, Charlottesville, Virginia
| | - Judit Megyesi
- University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Nicole M Thielens
- University of Grenoble Alpes, Centre National de la Recherche Scientifique, Commissariat à l'énergie Atomique et aux Énergies Alternatives, L'Institut de Biologie Structurale, Grenoble, France
| | - Didier Portilla
- Division of Nephrology, Center for Immunity, Inflammation and Regenerative Medicine, University of Virginia, Charlottesville, Virginia
- Salem Veterans Affairs Medical Center, Salem, Virginia
| |
Collapse
|
21
|
Bohlson SS, Garred P, Kemper C, Tenner AJ. Complement Nomenclature-Deconvoluted. Front Immunol 2019; 10:1308. [PMID: 31231398 PMCID: PMC6568193 DOI: 10.3389/fimmu.2019.01308] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 05/22/2019] [Indexed: 01/08/2023] Open
Abstract
In 2014, specific recommendations for complement nomenclature were presented by the complement field. There remained some unresolved designations and new areas of ambiguity, and here we propose solutions to resolve these remaining issues. To enable rapid understanding of the intricate complement system and facilitate therapeutic development and application, a uniform nomenclature for cleavage fragments, pattern recognition molecules (PRMs) and enzymes of the lectin pathway and regulatory proteins of the complement system are proposed, and a standardization of language to designate different activation states of complement components is recommended.
Collapse
Affiliation(s)
- Suzanne S Bohlson
- Department of Microbiology and Immunology, Des Moines University, Des Moines, IA, United States
| | - Peter Garred
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Rigshospitalet University Hospital of Copenhagen, Copenhagen, Denmark
| | - Claudia Kemper
- Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Andrea J Tenner
- Department of Molecular Biology and Biochemistry, Department of Neurobiology and Behavior, Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|