1
|
Lage SL, Rocco JM, Laidlaw E, Rupert A, Galindo F, Kellogg A, Kumar P, Poon R, Wortmann GW, Lisco A, Manion M, Sereti I. Activation of Complement Components on Circulating Blood Monocytes From COVID-19 Patients. Front Immunol 2022; 13:815833. [PMID: 35250994 PMCID: PMC8892247 DOI: 10.3389/fimmu.2022.815833] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 01/25/2022] [Indexed: 12/13/2022] Open
Abstract
The coronavirus disease-2019 (COVID-19) caused by the SARS-CoV-2 virus may vary from asymptomatic to severe infection with multi-organ failure and death. Increased levels of circulating complement biomarkers have been implicated in COVID-19-related hyperinflammation and coagulopathy. We characterized systemic complement activation at a cellular level in 49-patients with COVID-19. We found increases of the classical complement sentinel C1q and the downstream C3 component on circulating blood monocytes from COVID-19 patients when compared to healthy controls (HCs). Interestingly, the cell surface-bound complement inhibitor CD55 was also upregulated in COVID-19 patient monocytes in comparison with HC cells. Monocyte membrane-bound C1q, C3 and CD55 levels were associated with plasma inflammatory markers such as CRP and serum amyloid A during acute infection. Membrane-bounds C1q and C3 remained elevated even after a short recovery period. These results highlight systemic monocyte-associated complement activation over a broad range of COVID-19 disease severities, with a compensatory upregulation of CD55. Further evaluation of complement and its interaction with myeloid cells at the membrane level could improve understanding of its role in COVID-19 pathogenesis.
Collapse
Affiliation(s)
- Silvia Lucena Lage
- HIV Pathogenesis Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
- *Correspondence: Silvia Lucena Lage, ; Joseph M. Rocco,
| | - Joseph M. Rocco
- HIV Pathogenesis Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
- *Correspondence: Silvia Lucena Lage, ; Joseph M. Rocco,
| | - Elizabeth Laidlaw
- HIV Pathogenesis Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Adam Rupert
- AIDS Monitoring Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD, United States
| | - Frances Galindo
- HIV Pathogenesis Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Anela Kellogg
- Clinical Monitoring Research Program Directorate, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD, United States
| | - Princy Kumar
- Division of Infectious Diseases and Tropical Medicine, Georgetown University Medical Center, Washington, DC, United States
| | - Rita Poon
- Division of Hospital Medicine at MedStar Georgetown University Hospital, Washington, DC, United States
| | - Glenn W. Wortmann
- Section of Infectious Diseases, MedStar Washington Hospital Center, Washington, DC, United States
| | - Andrea Lisco
- HIV Pathogenesis Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Maura Manion
- HIV Pathogenesis Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Irini Sereti
- HIV Pathogenesis Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
2
|
Cortes C, Desler C, Mazzoli A, Chen JY, Ferreira VP. The role of properdin and Factor H in disease. Adv Immunol 2022; 153:1-90. [PMID: 35469595 DOI: 10.1016/bs.ai.2021.12.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The complement system consists of three pathways (alternative, classical, and lectin) that play a fundamental role in immunity and homeostasis. The multifunctional role of the complement system includes direct lysis of pathogens, tagging pathogens for phagocytosis, promotion of inflammatory responses to control infection, regulation of adaptive cellular immune responses, and removal of apoptotic/dead cells and immune complexes from circulation. A tight regulation of the complement system is essential to avoid unwanted complement-mediated damage to the host. This regulation is ensured by a set of proteins called complement regulatory proteins. Deficiencies or malfunction of these regulatory proteins may lead to pro-thrombotic hematological diseases, renal and ocular diseases, and autoimmune diseases, among others. This review focuses on the importance of two complement regulatory proteins of the alternative pathway, Factor H and properdin, and their role in human diseases with an emphasis on: (a) characterizing the main mechanism of action of Factor H and properdin in regulating the complement system and protecting the host from complement-mediated attack, (b) describing the dysregulation of the alternative pathway as a result of deficiencies, or mutations, in Factor H and properdin, (c) outlining the clinical findings, management and treatment of diseases associated with mutations and deficiencies in Factor H, and (d) defining the unwanted and inadequate functioning of properdin in disease, through a discussion of various experimental research findings utilizing in vitro, mouse and human models.
Collapse
Affiliation(s)
- Claudio Cortes
- Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI, United States.
| | - Caroline Desler
- Oakland University William Beaumont School of Medicine, Rochester, MI, United States
| | - Amanda Mazzoli
- Oakland University William Beaumont School of Medicine, Rochester, MI, United States
| | - Jin Y Chen
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Viviana P Ferreira
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States.
| |
Collapse
|
3
|
Lage SL, Wong CS, Amaral EP, Sturdevant D, Hsu DC, Rupert A, Wilson EMP, Qasba SS, Naqvi NS, Laidlaw E, Lisco A, Manion M, Sereti I. Classical complement and inflammasome activation converge in CD14highCD16- monocytes in HIV associated TB-immune reconstitution inflammatory syndrome. PLoS Pathog 2021; 17:e1009435. [PMID: 33788899 PMCID: PMC8041190 DOI: 10.1371/journal.ppat.1009435] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 04/12/2021] [Accepted: 03/01/2021] [Indexed: 02/07/2023] Open
Abstract
Inflammasome-derived cytokines, IL-1β and IL-18, and complement cascade have been independently implicated in the pathogenesis of tuberculosis (TB)-immune reconstitution inflammatory syndrome (TB-IRIS), a complication affecting HIV+ individuals starting antiretroviral therapy (ART). Although sublytic deposition of the membrane attack complex (MAC) has been shown to promote NLRP3 inflammasome activation, it is unknown whether these pathways may cooperatively contribute to TB-IRIS. To evaluate the activation of inflammasome, peripheral blood mononuclear cells (PBMCs) from HIV-TB co-infected patients prior to ART and at the IRIS or equivalent timepoint were incubated with a probe used to assess active caspase-1/4/5 followed by screening of ASC (apoptosis-associated speck-like protein containing a CARD domain) specks as a readout of inflammasome activation by imaging flow cytometry. We found higher numbers of monocytes showing spontaneous caspase-1/4/5+ASC-speck formation in TB-IRIS compared to TB non-IRIS patients. Moreover, numbers of caspase-1/4/5+ASC-speck+ monocytes positively correlated with IL-1β/IL-18 plasma levels. Besides increased systemic levels of C1q and C5a, TB-IRIS patients also showed elevated C1q and C3 deposition on monocyte cell surface, suggesting aberrant classical complement activation. A clustering tSNE analysis revealed TB-IRIS patients are enriched in a CD14highCD16- monocyte population that undergoes MAC deposition and caspase-1/4/5 activation compared to TB non-IRIS patients, suggesting complement-associated inflammasome activation during IRIS events. Accordingly, PBMCs from patients were more sensitive to ex-vivo complement-mediated IL-1β secretion than healthy control cells in a NLRP3-dependent manner. Therefore, our data suggest complement-associated inflammasome activation may fuel the dysregulated TB-IRIS systemic inflammatory cascade and targeting this pathway may represent a novel therapeutic approach for IRIS or related inflammatory syndromes. Tuberculosis (TB) associated-immune reconstitution inflammatory syndrome (TB-IRIS) is a clinical complication affecting HIV+ individuals previously co-infected with Mycobacterium tuberculosis (Mtb), upon antiretroviral therapy (ART) initiation. TB-IRIS is characterized by an exacerbated inflammatory response and can be associated with high morbidity and mortality rates in resource-limited countries with high TB prevalence. So far, there is no targeted TB-IRIS therapy, and corticosteroids are frequently used to prevent or alleviate IRIS related-symptoms. Here we found inflammasome activation (i.e. caspase1/4/5+ASC speck complex formation) on circulating classical CD14highCD16- monocytes may contribute to TB-IRIS immunopathology, since it correlates with pro-inflammatory cytokine plasma levels and its decay is associated with dampening in IRIS-related symptoms promoted by anti-inflammatory therapy. We also found TB-IRIS monocytes display higher surface complement deposition, being more sensitive to external complement-mediated NLRP3 inflammasome activation than healthy control cells. In fact, complement MAC molecule C9 and caspase-1/4/5 activation were associated on classical monocytes in TB-IRIS patients, suggesting complement-mediated inflammasome activation may lead to a positive feedback loop in the inflammatory responses observed in TB-IRIS. Therefore, our findings support that complement-NLRP3/ASC/caspase1/4/5 axis may be considered as a potential target for host-directed therapy of TB-IRIS.
Collapse
Affiliation(s)
- Silvia Lucena Lage
- HIV Pathogenesis Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States of America
- * E-mail: (SLL); (IS)
| | - Chun-Shu Wong
- HIV Pathogenesis Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States of America
| | - Eduardo Pinheiro Amaral
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States of America
| | - Daniel Sturdevant
- RML Genomics Unit, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, United States of America
| | - Denise C. Hsu
- HIV Pathogenesis Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States of America
| | - Adam Rupert
- Applied and Developmental Research Directorate, AIDS Monitoring Laboratory, Leidos Biomedical Research, Inc, Frederick, MD, United States of America
| | - Eleanor M. P. Wilson
- HIV Pathogenesis Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States of America
| | - S. Sonia Qasba
- HIV Pathogenesis Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States of America
| | - Nuha Sultana Naqvi
- HIV Pathogenesis Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States of America
| | - Elizabeth Laidlaw
- HIV Pathogenesis Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States of America
| | - Andrea Lisco
- HIV Pathogenesis Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States of America
| | - Maura Manion
- HIV Pathogenesis Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States of America
| | - Irini Sereti
- HIV Pathogenesis Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States of America
- * E-mail: (SLL); (IS)
| |
Collapse
|
4
|
Hosszu KK, Valentino A, Peerschke EI, Ghebrehiwet B. SLE: Novel Postulates for Therapeutic Options. Front Immunol 2020; 11:583853. [PMID: 33117397 PMCID: PMC7575694 DOI: 10.3389/fimmu.2020.583853] [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: 07/15/2020] [Accepted: 09/10/2020] [Indexed: 12/19/2022] Open
Abstract
Genetic deficiency in C1q is a strong susceptibility factor for systemic lupus erythematosus (SLE). There are two major hypotheses that potentially explain the role of C1q in SLE. The first postulates that C1q deficiency abrogates apoptotic cell clearance, leading to persistently high loads of potentially immunogenic self-antigens that trigger autoimmune responses. While C1q undoubtedly plays an important role in apoptotic clearance, an essential biological process such as removal of self- waste is so critical for host survival that multiple ligand-receptor combinations do fortunately exist to ensure that proper disposal of apoptotic debris is accomplished even in the absence of C1q. The second hypothesis is based on the observation that locally synthesized C1q plays a critical role in regulating the earliest stages of monocyte to dendritic cell (DC) differentiation and function. Indeed, circulating C1q has been shown to keep monocytes in a pre-dendritic state by silencing key molecular players and ensuring that unwarranted DC-driven immune responses do not occur. Monocytes are also able to display macromolecular C1 on their surface, representing a novel mechanism for the recognition of circulating "danger." Translation of this danger signal in turn, provides the requisite "license" to trigger a differentiation pathway that leads to adaptive immune response. Based on this evidence, the second hypothesis proposes that deficiency in C1q dysregulates monocyte-to-DC differentiation and causes inefficient or defective maintenance of self-tolerance. The fact that C1q receptors (cC1qR and gC1qR) are also expressed on the surface of both monocytes and DCs, suggests that C1q/C1qR may regulate DC differentiation and function through specific cell-signaling pathways. While their primary ligand is C1q, C1qRs can also independently recognize a vast array of plasma proteins as well as pathogen-associated molecular ligands, indicating that these molecules may collaborate in antigen recognition and processing, and thus regulate DC-differentiation. This review will therefore focus on the role of C1q and C1qRs in SLE and explore the gC1qR/C1q axis as a potential target for therapy.
Collapse
Affiliation(s)
- Kinga K Hosszu
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Alisa Valentino
- Department of Lab Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Ellinor I Peerschke
- Department of Lab Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Berhane Ghebrehiwet
- The Department of Medicine, Stony Brook University, Stony Brook, NY, United States
| |
Collapse
|
5
|
Skopelja-Gardner S, Colonna L, Hermanson P, Sun X, Tanaka L, Tai J, Nguyen Y, Snyder JM, Alpers CE, Hudkins KL, Salant DJ, Peng Y, Elkon KB. Complement Deficiencies Result in Surrogate Pathways of Complement Activation in Novel Polygenic Lupus-like Models of Kidney Injury. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 204:2627-2640. [PMID: 32238460 PMCID: PMC7365257 DOI: 10.4049/jimmunol.1901473] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 03/15/2020] [Indexed: 12/27/2022]
Abstract
Lupus nephritis (LN) is a major contributor to morbidity and mortality in lupus patients, but the mechanisms of kidney damage remain unclear. In this study, we introduce, to our knowledge, novel models of LN designed to resemble the polygenic nature of human lupus by embodying three key genetic alterations: the Sle1 interval leading to anti-chromatin autoantibodies; Mfge8-/- , leading to defective clearance of apoptotic cells; and either C1q-/- or C3-/- , leading to low complement levels. We report that proliferative glomerulonephritis arose only in the presence of all three abnormalities (i.e., in Sle1.Mfge8 -/- C1q -/- and Sle1.Mfge8 -/- C3 -/- triple-mutant [TM] strains [C1q -/-TM and C3-/- TM, respectively]), with structural kidney changes resembling those in LN patients. Unexpectedly, both TM strains had significant increases in autoantibody titers, Ag spread, and IgG deposition in the kidneys. Despite the early complement component deficiencies, we observed assembly of the pathogenic terminal complement membrane attack complex in both TM strains. In C1q-/- TM mice, colocalization of MASP-2 and C3 in both the glomeruli and tubules indicated that the lectin pathway likely contributed to complement activation and tissue injury in this strain. Interestingly, enhanced thrombin activation in C3-/- TM mice and reduction of kidney injury following attenuation of thrombin generation by argatroban in a serum-transfer nephrotoxic model identified thrombin as a surrogate pathway for complement activation in C3-deficient mice. These novel mouse models of human lupus inform the requirements for nephritis and provide targets for intervention.
Collapse
Affiliation(s)
| | - Lucrezia Colonna
- Division of Rheumatology, University of Washington, Seattle, WA 98109
| | - Payton Hermanson
- Division of Rheumatology, University of Washington, Seattle, WA 98109
| | - Xizhang Sun
- Division of Rheumatology, University of Washington, Seattle, WA 98109
| | - Lena Tanaka
- Division of Rheumatology, University of Washington, Seattle, WA 98109
| | - Joyce Tai
- Division of Rheumatology, University of Washington, Seattle, WA 98109
| | - Yenly Nguyen
- Division of Rheumatology, University of Washington, Seattle, WA 98109
| | - Jessica M Snyder
- Department of Comparative Medicine, University of Washington, Seattle, WA 98109
| | - Charles E Alpers
- Department of Nephrology, University of Washington, Seattle, WA 98109
| | - Kelly L Hudkins
- Department of Nephrology, University of Washington, Seattle, WA 98109
| | - David J Salant
- Division of Nephrology, Boston University, Boston, MA 02215; and
| | - YuFeng Peng
- Division of Rheumatology, University of Washington, Seattle, WA 98109;
| | - Keith B Elkon
- Division of Rheumatology, University of Washington, Seattle, WA 98109;
- Department of Immunology, University of Washington, Seattle, WA 98109
| |
Collapse
|
6
|
Fletcher EAK, van Maren W, Cordfunke R, Dinkelaar J, Codee JDC, van der Marel G, Melief CJM, Ossendorp F, Drijfhout JW, Mangsbo SM. Formation of Immune Complexes with a Tetanus-Derived B Cell Epitope Boosts Human T Cell Responses to Covalently Linked Peptides in an Ex Vivo Blood Loop System. THE JOURNAL OF IMMUNOLOGY 2018; 201:87-97. [PMID: 29752315 DOI: 10.4049/jimmunol.1700911] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 04/19/2018] [Indexed: 12/22/2022]
Abstract
Enhancing T cell responses against both viral and tumor Ags requires efficient costimulation and directed delivery of peptide Ags into APCs. Long peptide vaccines are considered favorable vaccine moieties from a clinical perspective, as they can harbor more than one immunogenic epitope enabling treatment of a broader target population. In addition, longer peptides are not extracellularly loaded on MHC class I; rather, they require intracellular processing and will thereby be presented to T cells mainly by professional APCs, thereby avoiding the risk of tolerance induction. The drawback of peptide vaccines regardless of peptide length is that naked peptides are not actively targeted to and taken up by APCs, and the standard nonconjugated adjuvant-peptide mixtures do not ensure cotargeting of the two to the same APC. We have identified a tetanus toxin-derived B cell epitope that can mediate the formation of immune complexes in the presence of circulating Abs. In this study, we show that these immune complexes improve both Ag uptake by APCs (blood monocytes and CD1c+ dendritic cells) and consequently improve CD8+ T cell recall responses in a human ex vivo blood loop system. The uptake of the peptide conjugate by blood monocytes is dependent on Abs and the complement component C1q. We envision that this strategy can be used to facilitate active uptake of Ags into APCs to improve T cell responses against pathogens or cancer.
Collapse
Affiliation(s)
- Erika A K Fletcher
- Department of Pharmaceutical Biosciences, Science for Life Laboratory, 751 24 Uppsala University, Uppsala, Sweden.,Immuneed AB, 752 37 Uppsala, Sweden
| | - Wendy van Maren
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2300 RC Leiden, the Netherlands; and
| | - Robert Cordfunke
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2300 RC Leiden, the Netherlands; and
| | - Jasper Dinkelaar
- Department of Bio-organic Synthesis, Leiden Institute of Chemistry, Leiden University, 2300 RA Leiden, the Netherlands
| | - Jeroen D C Codee
- Department of Bio-organic Synthesis, Leiden Institute of Chemistry, Leiden University, 2300 RA Leiden, the Netherlands
| | - Gijs van der Marel
- Department of Bio-organic Synthesis, Leiden Institute of Chemistry, Leiden University, 2300 RA Leiden, the Netherlands
| | - Cornelis J M Melief
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2300 RC Leiden, the Netherlands; and
| | - Ferry Ossendorp
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2300 RC Leiden, the Netherlands; and
| | - Jan Wouter Drijfhout
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2300 RC Leiden, the Netherlands; and
| | - Sara M Mangsbo
- Department of Pharmaceutical Biosciences, Science for Life Laboratory, 751 24 Uppsala University, Uppsala, Sweden; .,Immuneed AB, 752 37 Uppsala, Sweden
| |
Collapse
|
7
|
Xavier S, Sahu RK, Landes SG, Yu J, Taylor RP, Ayyadevara S, Megyesi J, Stallcup WB, Duffield JS, Reis ES, Lambris JD, Portilla D. Pericytes and immune cells contribute to complement activation in tubulointerstitial fibrosis. Am J Physiol Renal Physiol 2017; 312:F516-F532. [PMID: 28052876 PMCID: PMC5374314 DOI: 10.1152/ajprenal.00604.2016] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 12/07/2016] [Accepted: 01/03/2017] [Indexed: 12/22/2022] Open
Abstract
We have examined the pathogenic role of increased complement expression and activation during kidney fibrosis. Here, we show that PDGFRβ-positive pericytes isolated from mice subjected to obstructive or folic acid injury secrete C1q. This was associated with increased production of proinflammatory cytokines, extracellular matrix components, collagens, and increased Wnt3a-mediated activation of Wnt/β-catenin signaling, which are hallmarks of myofibroblast activation. Real-time PCR, immunoblots, immunohistochemistry, and flow cytometry analysis performed in whole kidney tissue confirmed increased expression of C1q, C1r, and C1s as well as complement activation, which is measured as increased synthesis of C3 fragments predominantly in the interstitial compartment. Flow studies localized increased C1q expression to PDGFRβ-positive pericytes as well as to CD45-positive cells. Although deletion of C1qA did not prevent kidney fibrosis, global deletion of C3 reduced macrophage infiltration, reduced synthesis of C3 fragments, and reduced fibrosis. Clodronate mediated depletion of CD11bF4/80 high macrophages in UUO mice also reduced complement gene expression and reduced fibrosis. Our studies demonstrate local synthesis of complement by both PDGFRβ-positive pericytes and CD45-positive cells in kidney fibrosis. Inhibition of complement activation represents a novel therapeutic target to ameliorate fibrosis and progression of chronic kidney disease.
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
| | - Susan G Landes
- Division of Nephrology, Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia, Charlottesville, Virginia
| | - Jing Yu
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia
| | - Ronald P Taylor
- Department of Biochemistry, University of Virginia, Charlottesville, Virginia
| | | | - Judit Megyesi
- University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - William B Stallcup
- Sanford Burnham Prebys Medical Discovery Institute, Tumor Metastasis and Cancer Immunology Program, La Jolla, California
| | | | - Edimara S Reis
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; and
| | - John D Lambris
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; and
| | - 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
|
8
|
Abstract
The complement system, which consists of three independent but interacting pathways, constitutes a powerful arm of innate immunity. Its major function is to recognize and destroy pathogenic microorganisms as well as eliminate modified self-antigens. Although it is a fine-tuned system with innate capacity to discriminate self from non-self as well as danger from non-danger signals, an unwarranted activation can nonetheless occur and cause tissue destruction. To prevent such activation, specific regulators present both in plasma and on the cell surface tightly control it. Data accumulated over the past four decades have also shown that the complement system is capable of not only cross-talk with the activation cascades of plasma––i.e. blood coagulation, contact activation, and the kinin/kallikrein system––but also serving as a bridge between innate and adaptive immunity. It is for these reasons that the various activation steps of the complement system have been recently targeted for therapy to treat diseases in which the role of complement is beyond doubt. This trend will certainly continue for years to come, especially as novel concepts guiding the field into areas never contemplated before are continuing to be discovered.
Collapse
Affiliation(s)
- Berhane Ghebrehiwet
- The Departments of Medicine and Pathology, Stony Brook University School of Medicine, Health Sciences Center, New York, USA
| |
Collapse
|
9
|
C1q as an autocrine and paracrine regulator of cellular functions. Mol Immunol 2016; 84:26-33. [PMID: 27914690 DOI: 10.1016/j.molimm.2016.11.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Revised: 11/01/2016] [Accepted: 11/03/2016] [Indexed: 12/20/2022]
Abstract
Most of the complement proteins in circulation are, by and large, synthesized in the liver. However data accumulated over the past several decades provide incontrovertible evidence that some if not most of the individual complement proteins are also synthesized extrahepatically by activated as well as non-activated cells. The question that is finally being addressed by various investigators is: are the locally synthesized proteins solely responsible for the myriad of biological functions in situ without the contribution of systemic complement? The answer is probably "yes". Among the proteins that are synthesized locally, C1q takes center stage for several reasons. First, it is synthesized predominantly by potent antigen presenting cells such as monocytes, macrophages and dendritic cells (DCs), which by itself is a clue that it plays an important role in antigen presentation and/or DC maturation. Second, it is transiently anchored on the cell surface via a transmembrane domain located in its A chain before it is cleaved off and released into the pericellular milieu. The membrane-associated C1q in turn, is able to sense danger patterns via its versatile antigen-capturing globular head domains. More importantly, locally synthesized C1q has been shown to induce a plethora of biological functions through the induction of immunomodulatory molecules by an autocrine- or paracrine- mediated signaling in a manner that mimics those of TNFα. These include recognition of pathogen- and danger- associated molecular patterns, phagocytosis, angiogenesis, apoptosis and induction of cytokines or chemokines that are important in modulating the inflammatory response. The functional convergence between C1q and TNFα in turn is attributed to their shared genetic ancestry. In this paper, we will infer to the aforementioned "local-synthesis-for-local function" paradigm using as an example, the role played by locally synthesized C1q in autoimmunity in general and in systemic lupus erythematosus in particular.
Collapse
|
10
|
C1q and HMGB1 reciprocally regulate human macrophage polarization. Blood 2016; 128:2218-2228. [PMID: 27683415 DOI: 10.1182/blood-2016-05-719757] [Citation(s) in RCA: 122] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 09/20/2016] [Indexed: 01/26/2023] Open
Abstract
A healthy immune system results from a balance of stimulatory and inhibitory pathways that allow effective responses to acute insults, without descending into chronic inflammation. Failed homeostasis is characteristic of autoimmune diseases such as systemic lupus erythematosus. Although HMGB1 induces proinflammatory M1-like macrophage differentiation, we describe a mechanism by which C1q modulates this activity and collaborates with HMGB1 to induce the differentiation of monocytes to anti-inflammatory M2-like macrophages. These anti-inflammatory macrophages are unresponsive to dendritic cell induction factors, effectively removing them from participation in an adaptive immune response. This pathway is mediated through a complex with RAGE and LAIR-1 and depends on relative levels of C1q and HMGB1. Importantly, these data provide insight into a homeostatic mechanism in which C1q and HMGB1 can cooperate to terminate inflammation, and which may be impaired in C1q-deficient patients with autoimmune disease.
Collapse
|
11
|
Macedo ACL, Isaac L. Systemic Lupus Erythematosus and Deficiencies of Early Components of the Complement Classical Pathway. Front Immunol 2016; 7:55. [PMID: 26941740 PMCID: PMC4764694 DOI: 10.3389/fimmu.2016.00055] [Citation(s) in RCA: 178] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 02/05/2016] [Indexed: 12/24/2022] Open
Abstract
The complement system plays an important role in the innate and acquired immune response against pathogens. It consists of more than 30 proteins found in soluble form or attached to cell membranes. Most complement proteins circulate in inactive forms and can be sequentially activated by the classical, alternative, or lectin pathways. Biological functions, such as opsonization, removal of apoptotic cells, adjuvant function, activation of B lymphocytes, degranulation of mast cells and basophils, and solubilization and clearance of immune complex and cell lysis, are dependent on complement activation. Although the activation of the complement system is important to avoid infections, it also can contribute to the inflammatory response triggered by immune complex deposition in tissues in autoimmune diseases. Paradoxically, the deficiency of early complement proteins from the classical pathway (CP) is strongly associated with development of systemic lupus erythematous (SLE) - mainly C1q deficiency (93%) and C4 deficiency (75%). The aim of this review is to focus on the deficiencies of early components of the CP (C1q, C1r, C1s, C4, and C2) proteins in SLE patients.
Collapse
Affiliation(s)
- Ana Catarina Lunz Macedo
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil; Faculty of Medicine, Children's Hospital, Clinics Hospital, University of São Paulo, São Paulo, Brazil
| | - Lourdes Isaac
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo , São Paulo , Brazil
| |
Collapse
|
12
|
Kouser L, Madhukaran SP, Shastri A, Saraon A, Ferluga J, Al-Mozaini M, Kishore U. Emerging and Novel Functions of Complement Protein C1q. Front Immunol 2015; 6:317. [PMID: 26175731 PMCID: PMC4484229 DOI: 10.3389/fimmu.2015.00317] [Citation(s) in RCA: 121] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 06/02/2015] [Indexed: 02/02/2023] Open
Abstract
Complement protein C1q, the recognition molecule of the classical pathway, performs a diverse range of complement and non-complement functions. It can bind various ligands derived from self, non-self, and altered self and modulate the functions of immune and non-immune cells including dendritic cells and microglia. C1q involvement in the clearance of apoptotic cells and subsequent B cell tolerance is more established now. Recent evidence appears to suggest that C1q plays an important role in pregnancy where its deficiency and dysregulation can have adverse effects, leading to preeclampsia, missed abortion, miscarriage or spontaneous loss, and various infections. C1q is also produced locally in the central nervous system, and has a protective role against pathogens and possible inflammatory functions while interacting with aggregated proteins leading to neurodegenerative diseases. C1q role in synaptic pruning, and thus CNS development, its anti-cancer effects as an immune surveillance molecule, and possibly in aging are currently areas of extensive research.
Collapse
Affiliation(s)
- Lubna Kouser
- Centre for Infection, Immunity and Disease Mechanisms, College of Health and Life Sciences, Brunel University London , Uxbridge , UK
| | - Shanmuga Priyaa Madhukaran
- Centre for Infection, Immunity and Disease Mechanisms, College of Health and Life Sciences, Brunel University London , Uxbridge , UK ; Centre for Biotechnology and Bioinformatics, Jawaharlal Nehru Institute for Advanced Studies, School of Life Sciences , Secunderabad , India
| | - Abhishek Shastri
- St. Ann's Hospital, Dorset Healthcare University NHS Foundation Trust , Poole , UK
| | - Anuvinder Saraon
- Centre for Infection, Immunity and Disease Mechanisms, College of Health and Life Sciences, Brunel University London , Uxbridge , UK
| | - Janez Ferluga
- Centre for Infection, Immunity and Disease Mechanisms, College of Health and Life Sciences, Brunel University London , Uxbridge , UK
| | - Maha Al-Mozaini
- Department of Infection and Immunity, King Faisal Specialist Hospital and Research Centre , Riyadh , Saudi Arabia
| | - Uday Kishore
- Centre for Infection, Immunity and Disease Mechanisms, College of Health and Life Sciences, Brunel University London , Uxbridge , UK
| |
Collapse
|
13
|
Fossati-Jimack L, Ling GS, Baudino L, Szajna M, Manivannan K, Zhao JC, Midgley R, Chai JG, Simpson E, Botto M, Scott D. Intranasal peptide-induced tolerance and linked suppression: consequences of complement deficiency. Immunology 2015; 144:149-57. [PMID: 25039245 PMCID: PMC4264918 DOI: 10.1111/imm.12358] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 07/11/2014] [Accepted: 07/15/2014] [Indexed: 12/27/2022] Open
Abstract
A role for complement, particularly the classical pathway, in the regulation of immune responses is well documented. Deficiencies in C1q or C4 predispose to autoimmunity, while deficiency in C3 affects the suppression of contact sensitization and generation of oral tolerance. Complement components including C3 have been shown to be required for both B-cell and T-cell priming. The mechanisms whereby complement can mediate these diverse regulatory effects are poorly understood. Our previous work, using the mouse minor histocompatibility (HY) model of skin graft rejection, showed that both C1q and C3 were required for the induction of tolerance following intranasal peptide administration. By comparing tolerance induction in wild-type C57BL/6 and C1q-, C3-, C4- and C5-deficient C57BL/6 female mice, we show here that the classical pathway components including C3 are required for tolerance induction, whereas C5 plays no role. C3-deficient mice failed to generate a functional regulatory T (Treg) -dendritic cell (DC) tolerogenic loop required for tolerance induction. This was related to the inability of C3-deficient DC to up-regulate the arginine-consuming enzyme, inducible nitric oxide synthase (Nos-2), in the presence of antigen-specific Treg cells and peptide, leading to reduced Treg cell generation. Our findings demonstrate that the classical pathway and C3 play a critical role in the peptide-mediated induction of tolerance to HY by modulating DC function.
Collapse
|
14
|
Zimmer J, Hobkirk J, Mohamed F, Browning MJ, Stover CM. On the Functional Overlap between Complement and Anti-Microbial Peptides. Front Immunol 2015; 5:689. [PMID: 25646095 PMCID: PMC4298222 DOI: 10.3389/fimmu.2014.00689] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 12/22/2014] [Indexed: 12/19/2022] Open
Abstract
Intriguingly, activated complement and anti-microbial peptides share certain functionalities; lytic, phagocytic, and chemo-attractant activities and each may, in addition, exert cell instructive roles. Each has been shown to have distinct LPS detoxifying activity and may play a role in the development of endotoxin tolerance. In search of the origin of complement, a functional homolog of complement C3 involved in opsonization has been identified in horseshoe crabs. Horseshoe crabs possess anti-microbial peptides able to bind to acyl chains or phosphate groups/saccharides of endotoxin, LPS. Complement activity as a whole is detectable in marine invertebrates. These are also a source of anti-microbial peptides with potential pharmaceutical applicability. Investigating the locality for the production of complement pathway proteins and their role in modulating cellular immune responses are emerging fields. The significance of local synthesis of complement components is becoming clearer from in vivo studies of parenchymatous disease involving specifically generated, complement-deficient mouse lines. Complement C3 is a central component of complement activation. Its provision by cells of the myeloid lineage varies. Their effector functions in turn are increased in the presence of anti-microbial peptides. This may point to a potentiating range of activities, which should serve the maintenance of health but may also cause disease. Because of the therapeutic implications, this review will consider closely studies dealing with complement activation and anti-microbial peptide activity in acute inflammation (e.g., dialysis-related peritonitis, appendicitis, and ischemia).
Collapse
Affiliation(s)
- Jana Zimmer
- Department of Infectious Diseases - Medical Microbiology and Hygiene, Ruprecht-Karls-University of Heidelberg , Heidelberg , Germany
| | - James Hobkirk
- Department of Academic Endocrinology, Diabetes and Metabolism, Hull York Medical School, University of Hull , Hull , UK
| | - Fatima Mohamed
- Department of Infection, Immunity and Inflammation, University of Leicester , Leicester , UK
| | - Michael J Browning
- Department of Infection, Immunity and Inflammation, University of Leicester , Leicester , UK ; Department of Immunology, Leicester Royal Infirmary , Leicester , UK
| | - Cordula M Stover
- Department of Infection, Immunity and Inflammation, University of Leicester , Leicester , UK
| |
Collapse
|
15
|
Ghebrehiwet B, Hosszu KK, Valentino A, Ji Y, Peerschke EIB. Monocyte Expressed Macromolecular C1 and C1q Receptors as Molecular Sensors of Danger: Implications in SLE. Front Immunol 2014; 5:278. [PMID: 25018754 PMCID: PMC4071343 DOI: 10.3389/fimmu.2014.00278] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Accepted: 05/28/2014] [Indexed: 01/07/2023] Open
Abstract
The ability of circulating blood monocytes to express C1q receptors (cC1qR and gC1qR) as well as to synthesize and secrete the classical pathway proteins C1q, C1r, and C1s and their regulator, C1-INH is very well established. What is intriguing, however, is that, in addition to secretion of the individual C1 proteins monocytes are also able to display macromolecular C1 on their surface in a manner that is stable and functional. The cell surface C1 complex is presumably formed by a Ca2+-dependent association of the C1r2⋅C1s2 tetramer to C1q, which in turn is anchored via a membrane-binding domain located in the N-terminus of its A-chain as shown previously. Monocytes, which circulate in the blood for 1–3 days before they move into tissues throughout the body, not only serve as precursors of macrophages and dendritic cells (DCs), but also fulfill three main functions in the immune system: phagocytosis, antigen presentation, and cytokine production. Since the globular heads of C1q within the membrane associated C1 are displayed outwardly, we hypothesize that their main function – especially in circulating monocytes – is to recognize and capture circulating immune complexes or pathogen-associated molecular patterns in the blood. This in turn may give crucial signal, which drives the monocytes to migrate into tissues, differentiate into macrophages or DCs, and initiate the process of antigen elimination. Unoccupied C1q on the other hand may serve to keep monocytes in a pre-dendritic phenotype by silencing key molecular players thus ensuring that unwarranted DC-driven immune response does not occur. In this paper, we will discuss the role of monocyte/DC-associated C1q receptors, macromolecular C1 as well as secreted C1q in both innate and acquired immune responses.
Collapse
Affiliation(s)
- Berhane Ghebrehiwet
- Departments of Medicine and Pathology, Stony Brook University , Stony Brook, NY , USA
| | - Kinga K Hosszu
- Departments of Medicine and Pathology, Stony Brook University , Stony Brook, NY , USA
| | - Alisa Valentino
- Departments of Medicine and Pathology, Stony Brook University , Stony Brook, NY , USA
| | - Yan Ji
- Departments of Medicine and Pathology, Stony Brook University , Stony Brook, NY , USA
| | - Ellinor I B Peerschke
- Departments of Laboratory Medicine, Memorial Sloan-Kettering Cancer Center, and Laboratory Medicine and Pathology, Weill-Cornell Medical College , New York, NY , USA
| |
Collapse
|
16
|
Courtois A, Berthou C, Guézennec J, Boisset C, Bordron A. Exopolysaccharides isolated from hydrothermal vent bacteria can modulate the complement system. PLoS One 2014; 9:e94965. [PMID: 24736648 PMCID: PMC3988086 DOI: 10.1371/journal.pone.0094965] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Accepted: 03/20/2014] [Indexed: 11/19/2022] Open
Abstract
The complement system is involved in the defence against bacterial infection, or in the elimination of tumour cells. However, disturbances in this system contributes to the pathogenesis of various inflammatory diseases. The efficiency of therapeutic anti-tumour antibodies is enhanced when the complement system is stimulated. In contrast, cancer cells are able to inhibit the complement system and thus proliferate. Some marine molecules are currently being developed as new drugs for use in humans. Among them, known exopolyssacharides (EPSs) generally originate from fungi, but few studies have been performed on bacterial EPSs and even fewer on EPSs extracted from deep-sea hydrothermal vent microbes. For use in humans, these high molecular weight EPSs must be depolymerised. Furthermore, the over-sulphation of EPSs can modify their biological activity. The aim of this study was to investigate the immunodulation of the complement system by either native or over-sulphated low molecular weight EPSs isolated from vent bacteria in order to find pro or anti-activators of complement.
Collapse
Affiliation(s)
- Anthony Courtois
- Biotechnology and Marine Molecules Laboratory, IFREMER, Brest, France
- Cellular Therapy and Immunobiology of Cancer Laboratory, University Hospital of Brest, Brest, France
| | - Christian Berthou
- Cellular Therapy and Immunobiology of Cancer Laboratory, University Hospital of Brest, Brest, France
| | - Jean Guézennec
- Biotechnology and Marine Molecules Laboratory, IFREMER, Brest, France
| | - Claire Boisset
- Biotechnology and Marine Molecules Laboratory, IFREMER, Brest, France
| | - Anne Bordron
- Cellular Therapy and Immunobiology of Cancer Laboratory, University Hospital of Brest, Brest, France
- * E-mail:
| |
Collapse
|
17
|
Bally I, Ancelet S, Moriscot C, Gonnet F, Mantovani A, Daniel R, Schoehn G, Arlaud GJ, Thielens NM. Expression of recombinant human complement C1q allows identification of the C1r/C1s-binding sites. Proc Natl Acad Sci U S A 2013; 110:8650-5. [PMID: 23650384 PMCID: PMC3666734 DOI: 10.1073/pnas.1304894110] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Complement C1q is a hexameric molecule assembled from 18 polypeptide chains of three different types encoded by three genes. This versatile recognition protein senses a wide variety of immune and nonimmune ligands, including pathogens and altered self components, and triggers the classical complement pathway through activation of its associated proteases C1r and C1s. We report a method for expression of recombinant full-length human C1q involving stable transfection of HEK 293-F mammalian cells and fusion of an affinity tag to the C-terminal end of the C chain. The resulting recombinant (r) C1q molecule is similar to serum C1q as judged from biochemical and structural analyses and exhibits the characteristic shape of a bunch of flowers. Analysis of its interaction properties by surface plasmon resonance shows that rC1q retains the ability of serum C1q to associate with the C1s-C1r-C1r-C1s tetramer, to recognize physiological C1q ligands such as IgG and pentraxin 3, and to trigger C1r and C1s activation. Functional analysis of rC1q variants carrying mutations of LysA59, LysB61, and/or LysC58, in the collagen-like stems, demonstrates that LysB61 and LysC58 each play a key role in the interaction with C1s-C1r-C1r-C1s, with LysA59 being involved to a lesser degree. We propose that LysB61 and LysC58 both form salt bridges with outer acidic Ca(2+) ligands of the C1r and C1s CUB (complement C1r/C1s, Uegf, bone morphogenetic protein) domains. The expression method reported here opens the way for deciphering the molecular basis of the unusual binding versatility of C1q by mapping the residues involved in the sensing of its targets and the binding of its receptors.
Collapse
Affiliation(s)
- Isabelle Bally
- Commissariat à l’Energie Atomique et aux Energies Alternatives, Direction des Sciences du Vivant, Institut de Biologie Structurale, F-38027 Grenoble, France
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 5075, F-38027 Grenoble, France
- Université Grenoble Alpes, F-38000 Grenoble, France
| | - Sarah Ancelet
- Commissariat à l’Energie Atomique et aux Energies Alternatives, Direction des Sciences du Vivant, Institut de Biologie Structurale, F-38027 Grenoble, France
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 5075, F-38027 Grenoble, France
- Université Grenoble Alpes, F-38000 Grenoble, France
| | - Christine Moriscot
- Commissariat à l’Energie Atomique et aux Energies Alternatives, Direction des Sciences du Vivant, Institut de Biologie Structurale, F-38027 Grenoble, France
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 5075, F-38027 Grenoble, France
- Université Grenoble Alpes, F-38000 Grenoble, France
- CNRS, Unité Mixte Internationale 3265, Unit for Virus Host Cell Interactions, F-38042-Grenoble, France
| | - Florence Gonnet
- CNRS, UMR 8587, Laboratoire Analyse et Modélisation pour la Biologie et l‘Environnement, F-91025 Evry, France
- Université d’Evry-Val-d’Essonne, Laboratoire Analyse et Modélisation pour la Biologie et l’Environnement, F-91025 Evry, France; and
| | | | - Régis Daniel
- CNRS, UMR 8587, Laboratoire Analyse et Modélisation pour la Biologie et l‘Environnement, F-91025 Evry, France
- Université d’Evry-Val-d’Essonne, Laboratoire Analyse et Modélisation pour la Biologie et l’Environnement, F-91025 Evry, France; and
| | - Guy Schoehn
- Commissariat à l’Energie Atomique et aux Energies Alternatives, Direction des Sciences du Vivant, Institut de Biologie Structurale, F-38027 Grenoble, France
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 5075, F-38027 Grenoble, France
- Université Grenoble Alpes, F-38000 Grenoble, France
- CNRS, Unité Mixte Internationale 3265, Unit for Virus Host Cell Interactions, F-38042-Grenoble, France
| | - Gérard J. Arlaud
- Commissariat à l’Energie Atomique et aux Energies Alternatives, Direction des Sciences du Vivant, Institut de Biologie Structurale, F-38027 Grenoble, France
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 5075, F-38027 Grenoble, France
- Université Grenoble Alpes, F-38000 Grenoble, France
| | - Nicole M. Thielens
- Commissariat à l’Energie Atomique et aux Energies Alternatives, Direction des Sciences du Vivant, Institut de Biologie Structurale, F-38027 Grenoble, France
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 5075, F-38027 Grenoble, France
- Université Grenoble Alpes, F-38000 Grenoble, France
| |
Collapse
|
18
|
C1q limits dendritic cell differentiation and activation by engaging LAIR-1. Proc Natl Acad Sci U S A 2012; 109:E3160-7. [PMID: 23093673 DOI: 10.1073/pnas.1212753109] [Citation(s) in RCA: 124] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
C1q, the first component of complement, and leukocyte-associated Ig-like receptor 1 (LAIR-1; CD305), an inhibitory receptor expressed on hematopoietic cells, have both been associated with arrest of monocyte-derived dendritic cell (DC) differentiation and inhibition of Toll-like receptor activity in plasmacytoid DCs. Defects in both molecules have been implicated in susceptibility to, and progression of, systemic lupus erythematosus. Inhibitory signaling partners for C1q on monocytes and DCs remain undefined. Because C1q contains collagen-like motifs and LAIR-1 is a universal collagen receptor, we hypothesized that C1q is a functional ligand for LAIR-1. Binding analyses in cell-free systems and on the cell membrane demonstrate that C1q and its collagen tail associate with LAIR-1 and LAIR-2 (CD306), a soluble inhibitor of LAIR-1. Both C1q and its collagen tail trigger phosphorylation of LAIR-1 immunoreceptor tyrosine-based inhibitory motifs (ITIMs) in monocytes. Functional analyses show that C1q-mediated inhibition of monocyte-DC differentiation and C1q-mediated inhibition of IFN-α production by plasmacytoid DCs were both reversed by LAIR-2. Moreover, C1q-mediated inhibition of DC differentiation was reversed by LAIR-1 siRNA. Thus, C1q is a functional ligand for LAIR-1 restricting immune cell differentiation and activation. The discovery of C1q interactions with LAIR-1 and LAIR-2 lends much needed insight into molecular mechanisms operating to prevent the loss of tolerance, particularly in systemic lupus erythematosus.
Collapse
|