Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 847
1.
Front Immunol ; 15: 1351656, 2024.
Article En | MEDLINE | ID: mdl-38711524

Understanding at the molecular level of the cell biology of tumors has led to significant treatment advances in the past. Despite such advances however, development of therapy resistance and tumor recurrence are still unresolved major challenges. This therefore underscores the need to identify novel tumor targets and develop corresponding therapies to supplement existing biologic and cytotoxic approaches so that a deeper and more sustained treatment responses could be achieved. The complement system is emerging as a potential novel target for cancer therapy. Data accumulated to date show that complement proteins, and in particular C1q and its receptors cC1qR/CR and gC1qR/p33/HABP1, are overexpressed in most cancer cells and together are involved not only in shaping the inflammatory tumor microenvironment, but also in the regulation of angiogenesis, metastasis, and cell proliferation. In addition to the soluble form of C1q that is found in plasma, the C1q molecule is also found anchored on the cell membrane of monocytes, macrophages, dendritic cells, and cancer cells, via a 22aa long leader peptide found only in the A-chain. This orientation leaves its 6 globular heads exposed outwardly and thus available for high affinity binding to a wide range of molecular ligands that enhance tumor cell survival, migration, and proliferation. Similarly, the gC1qR molecule is not only overexpressed in most cancer types but is also released into the microenvironment where it has been shown to be associated with cancer cell proliferation and metastasis by activation of the complement and kinin systems. Co-culture of either T cells or cancer cells with purified C1q or anti-gC1qR has been shown to induce an anti-proliferative response. It is therefore postulated that in the tumor microenvironment, the interaction between C1q expressing cancer cells and gC1qR bearing cytotoxic T cells results in T cell suppression in a manner akin to the PD-L1 and PD-1 interaction.


Carrier Proteins , Complement C1q , Immune Checkpoint Inhibitors , Membrane Glycoproteins , Mitochondrial Proteins , Neoplasms , Receptors, Complement , Humans , Complement C1q/metabolism , Complement C1q/immunology , Neoplasms/immunology , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Receptors, Complement/metabolism , Animals , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/pharmacology , Tumor Microenvironment/immunology
2.
Front Immunol ; 15: 1404752, 2024.
Article En | MEDLINE | ID: mdl-38690267

Helminths produce calreticulin (CRT) to immunomodulate the host immune system as a survival strategy. However, the structure of helminth-derived CRT and the structural basis of the immune evasion process remains unclarified. Previous study found that the tissue-dwelling helminth Trichinella spiralis produces calreticulin (TsCRT), which binds C1q to inhibit activation of the complement classical pathway. Here, we used x-ray crystallography to resolve the structure of truncated TsCRT (TsCRTΔ), the first structure of helminth-derived CRT. TsCRTΔ was observed to share the same binding region on C1q with IgG based on the structure and molecular docking, which explains the inhibitory effect of TsCRT on C1q-IgG-initiated classical complement activation. Based on the key residues in TsCRTΔ involved in the binding activity to C1q, a 24 amino acid peptide called PTsCRT was constructed that displayed strong C1q-binding activity and inhibited C1q-IgG-initiated classical complement activation. This study is the first to elucidate the structural basis of the role of TsCRT in immune evasion, providing an approach to develop helminth-derived bifunctional peptides as vaccine target to prevent parasite infections or as a therapeutic agent to treat complement-related autoimmune diseases.


Calreticulin , Complement C1q , Immune Evasion , Trichinella spiralis , Trichinella spiralis/immunology , Complement C1q/immunology , Complement C1q/metabolism , Complement C1q/chemistry , Animals , Calreticulin/immunology , Calreticulin/chemistry , Calreticulin/metabolism , Crystallography, X-Ray , Protein Binding , Molecular Docking Simulation , Helminth Proteins/immunology , Helminth Proteins/chemistry , Complement Activation/immunology , Immunoglobulin G/immunology , Humans , Antigens, Helminth/immunology , Antigens, Helminth/chemistry , Trichinellosis/immunology , Trichinellosis/parasitology , Complement Pathway, Classical/immunology , Protein Conformation
3.
Scand J Immunol ; 99(6): e13368, 2024 Jun.
Article En | MEDLINE | ID: mdl-38527944

Cellular debris resulting from large trauma might overwhelm the scavenger mechanisms and lead to autoimmune reactions. We analysed whether a major well-defined trauma in humans induces laboratory signs of transient autoimmunity in the months after the insult. We included 50 patients with pertrochanteric femur fracture undergoing intramedullary nail osteosynthesis in a prospective cohort study and followed them at 3-4 days, 6 weeks, 12 weeks and 12 months postoperatively. By standard techniques, we assessed levels of total immunoglobulins, anti-nuclear antibodies (ANA), anti-cardiolipin antibodies, anti-dsDNA antibodies and anti-C1q antibodies, as well as antibodies against cytomegalovirus (CMV) as a control. Blood leukocyte differential and lymphocyte subpopulations were determined at baseline and in the first two postoperative samples. The mean age of the patients reached 80.1 years, and 23 (46%) completed all visits. Serum concentrations of total IgG, IgM and IgA increased at all follow-up time points. The ANA fluorescence light intensity units increased at 12 weeks and 12 months postoperatively (p < 0.0001), but the proportion of ANA-positive patients did not change (35%). The values of anti-C1q mildly increased at all follow-up visits, but not the ratio to total IgG. Anti-dsDNA remained negative in all patients, and anti-cardiolipin IgG/IgM antibodies did not change. Anti-CMV IgG antibodies increased significantly at all follow-up visits, without change in the ratio to total IgG. Flow cytometry showed an increased proportion of B-cells 3-4 days postoperatively. In conclusion, major musculoskeletal trauma in elderly patients induces a generalized non-specific increase in immunoglobulin production without laboratory signs for enhanced systemic autoimmunity.


Autoantibodies , Humans , Male , Female , Prospective Studies , Autoantibodies/blood , Autoantibodies/immunology , Aged , Aged, 80 and over , Antibodies, Antinuclear/blood , Antibodies, Antinuclear/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Complement C1q/immunology , Immunoglobulin M/blood , Cohort Studies , Autoimmunity , Immunoglobulins/blood
4.
Proc Natl Acad Sci U S A ; 119(26): e2204289119, 2022 06 28.
Article En | MEDLINE | ID: mdl-35727985

Behçet's disease (BD) is a chronic vasculitis characterized by systemic immune aberrations. However, a comprehensive understanding of immune disturbances in BD and how they contribute to BD pathogenesis is lacking. Here, we performed single-cell and bulk RNA sequencing to profile peripheral blood mononuclear cells (PBMCs) and isolated monocytes from BD patients and healthy donors. We observed prominent expansion and transcriptional changes in monocytes in PBMCs from BD patients. Deciphering the monocyte heterogeneity revealed the accumulation of C1q-high (C1qhi) monocytes in BD. Pseudotime inference indicated that BD monocytes markedly shifted their differentiation toward inflammation-accompanied and C1qhi monocyte-ended trajectory. Further experiments showed that C1qhi monocytes enhanced phagocytosis and proinflammatory cytokine secretion, and multiplatform analyses revealed the significant clinical relevance of this subtype. Mechanistically, C1qhi monocytes were induced by activated interferon-γ (IFN-γ) signaling in BD patients and were decreased by tofacitinib treatment. Our study illustrates the BD immune landscape and the unrecognized contribution of C1qhi monocytes to BD hyperinflammation, showing their potential as therapeutic targets and clinical assessment indexes.


Behcet Syndrome , Complement C1q , Monocytes , Behcet Syndrome/genetics , Behcet Syndrome/immunology , Complement C1q/genetics , Complement C1q/immunology , Humans , Monocytes/immunology , RNA-Seq , Single-Cell Analysis
5.
Proc Natl Acad Sci U S A ; 119(13): e2117770119, 2022 03 29.
Article En | MEDLINE | ID: mdl-35312359

Spirochetal pathogens, such as the causative agent of Lyme disease, Borrelia burgdorferi sensu lato, encode an abundance of lipoproteins; however, due in part to their evolutionary distance from more well-studied bacteria, such as Proteobacteria and Firmicutes, few spirochetal lipoproteins have assigned functions. Indeed, B. burgdorferi devotes almost 8% of its genome to lipoprotein genes and interacts with its environment primarily through the production of at least 80 surface-exposed lipoproteins throughout its tick vector­vertebrate host lifecycle. Several B. burgdorferi lipoproteins have been shown to serve roles in cellular adherence or immune evasion, but the functions for most B. burgdorferi surface lipoproteins remain unknown. In this study, we developed a B. burgdorferi lipoproteome screening platform utilizing intact spirochetes that enables the identification of previously unrecognized host interactions. As spirochetal survival in the bloodstream is essential for dissemination, we targeted our screen to C1, the first component of the classical (antibody-initiated) complement pathway. We identified two high-affinity C1 interactions by the paralogous lipoproteins, ElpB and ElpQ (also termed ErpB and ErpQ, respectively). Using biochemical, microbiological, and biophysical approaches, we demonstrate that ElpB and ElpQ bind the activated forms of the C1 proteases, C1r and C1s, and represent a distinct mechanistic class of C1 inhibitors that protect the spirochete from antibody-mediated complement killing. In addition to identifying a mode of complement inhibition, our study establishes a lipoproteome screening methodology as a discovery platform for identifying direct host­pathogen interactions that are central to the pathogenesis of spirochetes, such as the Lyme disease agent.


Bacterial Proteins , Borrelia burgdorferi , Complement C1q , Immune Evasion , Lipoproteins , Lyme Disease , Bacterial Proteins/immunology , Borrelia burgdorferi/immunology , Complement C1q/immunology , Humans , Immunoglobulins/immunology , Lipoproteins/immunology , Lyme Disease/immunology , Lyme Disease/microbiology , Proteome/immunology
6.
PLoS One ; 16(12): e0260954, 2021.
Article En | MEDLINE | ID: mdl-34932587

Elimination of the binding of immunoglobulin Fc to Fc gamma receptors (FcγR) is highly desirable for the avoidance of unwanted inflammatory responses to therapeutic antibodies and fusion proteins. Many different approaches have been described in the literature but none of them completely eliminates binding to all of the Fcγ receptors. Here we describe a set of novel variants having specific amino acid substitutions in the Fc region at L234 and L235 combined with the substitution G236R. They show no detectable binding to Fcγ receptors or to C1q, are inactive in functional cell-based assays and do not elicit inflammatory cytokine responses. Meanwhile, binding to FcRn, manufacturability, stability and potential for immunogenicity are unaffected. These variants have the potential to improve the safety and efficacy of therapeutic antibodies and Fc fusion proteins.


Antibody-Dependent Cell Cytotoxicity , Complement C1q/metabolism , Histocompatibility Antigens Class I/metabolism , Immunoglobulin Fc Fragments/metabolism , Immunoglobulin G/metabolism , Receptors, Fc/metabolism , Receptors, IgG/metabolism , Amino Acid Substitution , Antibody Affinity , Complement C1q/genetics , Complement C1q/immunology , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Humans , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/immunology , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Protein Binding , Protein Engineering , Receptors, Fc/genetics , Receptors, Fc/immunology , Receptors, IgG/genetics , Receptors, IgG/immunology
7.
Int J Mol Sci ; 22(21)2021 Nov 05.
Article En | MEDLINE | ID: mdl-34769432

Antibodies capable of activating the complement system (CS) when bound with antigen are referred to as "complement-fixing antibodies" and are involved in protection against Flaviviruses. A complement-fixing antibody test has been used in the past to measure the ability of dengue virus (DENV)-specific serum antibodies to activate the CS. As originally developed, the test is time-consuming, cumbersome, and has limited sensitivity for DENV diagnosis. Here, we developed and characterized a novel multiplex anti-DENV complement-fixing assay based on the Luminex platform to quantitate serum antibodies against all four serotypes (DENV1-4) that activate the CS based on their ability to fix the complement component 1q (C1q). The assay demonstrated good reproducibility and showed equivalent performance to a DENV microneutralization assay that has been used to determine DENV serostatus. In non-human primates, antibodies produced in response to primary DENV1-4 infection induced C1q fixation on homologous and heterologous serotypes. Inter-serotype cross-reactivity was associated with homology of the envelope protein. Interestingly, the antibodies produced following vaccination against Zika virus fixed C1q on DENV. The anti-DENV complement fixing antibody assay represents an alternative approach to determine the quality of functional antibodies produced following DENV natural infection or vaccination and a biomarker for dengue serostatus, while providing insights about immunological cross-reactivity among different Flaviviruses.


Antibodies, Viral/immunology , Complement C1q/immunology , Complement Fixation Tests/methods , Dengue Virus/immunology , Dengue/immunology , Animals , Antibodies, Viral/blood , Biological Assay , Cross Reactions/immunology , Dengue/metabolism , Dengue/virology , Humans , Macaca , Male , Reproducibility of Results , Serogroup
8.
Chest ; 160(5): e513-e518, 2021 11.
Article En | MEDLINE | ID: mdl-34743855

CASE PRESENTATION: A 56-year-old man presented to the pulmonary clinic with dyspnea and hypoxemia on exertion. He was an avid biker and skier who had noticed a significant decrease in high-level physical activity over the past 3 years. He reported dyspnea, desaturations at altitudes higher than 9,000 feet, dry cough, tachycardia, and palpitations with exercise. Review of systems was also notable for gluten-intolerance, Raynaud's phenomenon, recurrent skin lesions and joint swelling, pain, and stiffness in the areas overlying the jaw, wrists, knees, and ankles (after capsaicin exposure). He denied fever, chills, anorexia, weight loss, hair loss, ocular symptoms, jaw claudication, chest pain, or lower extremity swelling. He had a five pack-year smoking history, no history of prematurity, childhood asthma, recurrent infections, or environmental and occupational exposure. Based on pulmonary function tests from an outside provider, he had received a diagnosis of exercise-induced asthma and had been prescribed an albuterol inhaler to use on an as-needed basis, which failed to improve his symptoms. He was later prescribed a mometasone-formoterol inhaler, still with no symptomatic improvement.


Arthralgia , Complement C1q , Complement C4/analysis , Emphysema , Exanthema , Prednisolone/administration & dosage , Pulmonary Arterial Hypertension , Vasculitis, Leukocytoclastic, Cutaneous , Arthralgia/diagnosis , Arthralgia/etiology , Autoantibodies/blood , Bronchodilator Agents/administration & dosage , Complement C1q/analysis , Complement C1q/immunology , Diagnosis, Differential , Emphysema/diagnosis , Emphysema/etiology , Exanthema/diagnosis , Exanthema/etiology , Humans , Immunologic Factors/administration & dosage , Male , Middle Aged , Patient Care Management/methods , Pulmonary Arterial Hypertension/diagnosis , Pulmonary Arterial Hypertension/etiology , Rituximab/administration & dosage , Vasculitis, Leukocytoclastic, Cutaneous/blood , Vasculitis, Leukocytoclastic, Cutaneous/diagnosis , Vasculitis, Leukocytoclastic, Cutaneous/physiopathology
9.
Mol Microbiol ; 116(6): 1476-1488, 2021 12.
Article En | MEDLINE | ID: mdl-34725868

Pathogenic Rickettsia are obligate intracellular bacteria and the etiologic agents of many life-threatening infectious diseases. Due to the serious nature of these infections, it is imperative to both identify the responsive immune sensory pathways and understand the associated immune mechanisms that restrict Rickettsia proliferation. Previous studies have demonstrated that the mammalian complement system is both activated during Rickettsia infection and contributes to the immune response to infection. To further define this component of the mammalian anti-Rickettsia immune response, we sought to identify the mechanism(s) of complement activation during Rickettsia infection. We have employed a series of in vitro and in vivo models of infection to investigate the role of the classical complement activation pathway during Rickettsia infection. Depletion or elimination of complement activity demonstrates that both C1q and pre-existing IgM contribute to complement activation; thus implicating the classical complement system in Rickettsia-mediated complement activation. Elimination of the classical complement pathway from mice increases susceptibility to R. australis infection with both increased bacterial loads in multiple tissues and decreased immune activation markers. This study highlights the role of the classical complement pathway in immunity against Rickettsia and implicates resident Rickettsia-responsive IgM in the response to infection.


Antibodies, Bacterial/immunology , Complement C1q/immunology , Immunoglobulin M/immunology , Rickettsia Infections/immunology , Rickettsia/immunology , Animals , Complement Pathway, Classical , Humans , Mice , Mice, Inbred C57BL , Rickettsia/genetics , Rickettsia Infections/microbiology
10.
Int J Mol Sci ; 22(15)2021 Aug 01.
Article En | MEDLINE | ID: mdl-34361054

We addressed the issue of C1q autoantigenicity by studying the structural features of the autoepitopes recognized by the polyclonal anti-C1q antibodies present in Lupus Nephritis (LN) sera. We used six fractions of anti-C1q as antigens and selected anti-idiotypic scFv antibodies from the phage library "Griffin.1". The monoclonal scFv A1 was the most potent inhibitor of the recognition of C1q and its fragments ghA, ghB and ghC, comprising the globular domain gC1q, by the lupus autoantibodies. It was sequenced and in silico folded by molecular dynamics into a 3D structure. The generated 3D model of A1 elucidated CDR similarity to the apical region of gC1q, thus mapping indirectly for the first time a globular autoepitope of C1q. The VH CDR2 of A1 mimicked the ghA sequence GSEAD suggested as a cross-epitope between anti-DNA and anti-C1q antibodies. Other potential inhibitors of the recognition of C1q by the LN autoantibodies among the selected recombinant antibodies were the monoclonal scFv F6, F9 and A12.


Antibodies, Anti-Idiotypic/immunology , Autoantibodies/blood , Autoantigens/immunology , Complement C1q/immunology , Epitopes/immunology , Lupus Nephritis/immunology , Single-Chain Antibodies/immunology , Humans , Lupus Nephritis/blood , Protein Structure, Tertiary , Protein Subunits
11.
J Neurosci ; 41(41): 8508-8531, 2021 10 13.
Article En | MEDLINE | ID: mdl-34417332

Axon regenerative failure in the mature CNS contributes to functional deficits following many traumatic injuries, ischemic injuries, and neurodegenerative diseases. The complement cascade of the innate immune system responds to pathogen threat through inflammatory cell activation, pathogen opsonization, and pathogen lysis, and complement is also involved in CNS development, neuroplasticity, injury, and disease. Here, we investigated the involvement of the classical complement cascade and microglia/monocytes in CNS repair using the mouse optic nerve injury (ONI) model, in which axons arising from retinal ganglion cells (RGCs) are disrupted. We report that central complement C3 protein and mRNA, classical complement C1q protein and mRNA, and microglia/monocyte phagocytic complement receptor CR3 all increase in response to ONI, especially within the optic nerve itself. Importantly, genetic deletion of C1q, C3, or CR3 attenuates RGC axon regeneration induced by several distinct methods, with minimal effects on RGC survival. Local injections of C1q function-blocking antibody revealed that complement acts primarily within the optic nerve, not retina, to support regeneration. Moreover, C1q opsonizes and CR3+ microglia/monocytes phagocytose growth-inhibitory myelin debris after ONI, a likely mechanism through which complement and myeloid cells support axon regeneration. Collectively, these results indicate that local optic nerve complement-myeloid phagocytic signaling is required for CNS axon regrowth, emphasizing the axonal compartment and highlighting a beneficial neuroimmune role for complement and microglia/monocytes in CNS repair.SIGNIFICANCE STATEMENT Despite the importance of achieving axon regeneration after CNS injury and the inevitability of inflammation after such injury, the contributions of complement and microglia to CNS axon regeneration are largely unknown. Whereas inflammation is commonly thought to exacerbate the effects of CNS injury, we find that complement proteins C1q and C3 and microglia/monocyte phagocytic complement receptor CR3 are each required for retinal ganglion cell axon regeneration through the injured mouse optic nerve. Also, whereas studies of optic nerve regeneration generally focus on the retina, we show that the regeneration-relevant role of complement and microglia/monocytes likely involves myelin phagocytosis within the optic nerve. Thus, our results point to the importance of the innate immune response for CNS repair.


Axons/metabolism , Complement C1q/metabolism , Complement C3/metabolism , Myeloid Cells/metabolism , Optic Nerve Injuries/metabolism , Retinal Ganglion Cells/metabolism , Animals , Axons/immunology , Complement C1q/immunology , Complement C3/immunology , Female , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/immunology , Nerve Regeneration/physiology , Optic Nerve Injuries/immunology , Optic Nerve Injuries/pathology , Retinal Ganglion Cells/immunology
12.
Neurobiol Dis ; 159: 105493, 2021 11.
Article En | MEDLINE | ID: mdl-34464705

The amyloid beta (Aß) peptide is believed to play a central role in Alzheimer's disease (AD), the most common age-related neurodegenerative disorder. However, the natural, evolutionarily selected functions of Aß are incompletely understood. Here, we report that nanomolar concentrations of Aß act synergistically with known cytokines to promote pro-inflammatory activation in primary human astrocytes (a cell type increasingly implicated in brain aging and AD). Using transcriptomics (RNA-seq), we show that Aß can directly substitute for the complement component C1q in a cytokine cocktail previously shown to induce astrocyte immune activation. Furthermore, we show that astrocytes synergistically activated by Aß have a transcriptional signature similar to neurotoxic "A1" astrocytes known to accumulate with age and in AD. Interestingly, we find that this biological action of Aß at low concentrations is distinct from the transcriptome changes induced by the high/supraphysiological doses of Aß often used in in vitro studies. Collectively, our results suggest an important, cytokine-like function for Aß and a novel mechanism by which it may directly contribute to the neuroinflammation associated with brain aging and AD.


Aging/immunology , Alzheimer Disease/immunology , Amyloid beta-Peptides/immunology , Astrocytes/immunology , Brain/immunology , Cytokines/immunology , Neuroinflammatory Diseases/immunology , Amyloid beta-Peptides/pharmacology , Astrocytes/drug effects , Complement C1q/immunology , Complement C1q/pharmacology , Cytokines/pharmacology , Gene Expression Profiling , Humans , Interleukin-1alpha/immunology , Interleukin-1alpha/pharmacology , Peptide Fragments/pharmacology , Primary Cell Culture , RNA-Seq , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/pharmacology
13.
Viruses ; 13(7)2021 06 30.
Article En | MEDLINE | ID: mdl-34209320

Herpes simplex virus (HSV) prevention is a global health priority but, despite decades of research, there is no effective vaccine. Prior efforts focused on generating glycoprotein D (gD) neutralizing antibodies, but clinical trial outcomes were disappointing. The deletion of gD yields a single-cycle candidate vaccine (∆gD-2) that elicits high titer polyantigenic non-gD antibodies that exhibit little complement-independent neutralization but mediate antibody-dependent cellular cytotoxicity (ADCC) and phagocytosis (ADCP). Active or passive immunization with DgD-2 completely protects mice from lethal disease and latency following challenge with clinical isolates of either serotype. The current studies evaluated the role of complement in vaccine-elicited protection. The immune serum from the DgD-2 vaccinated mice exhibited significantly greater C1q binding compared to the serum from the gD protein vaccinated mice with infected cell lysates from either serotype as capture antigens. The C1q-binding antibodies recognized glycoprotein B. This resulted in significantly greater antibody-mediated complement-dependent cytolysis and neutralization. Notably, complete protection was preserved when the DgD-2 immune serum was passively transferred into C1q knockout mice, suggesting that ADCC and ADCP are sufficient in mice. We speculate that the polyfunctional responses elicited by DgD-2 may prove more effective in preventing HSV, compared to the more restrictive responses elicited by adjuvanted gD protein vaccines.


Antibodies, Viral/metabolism , Complement Activation , Complement C1q/metabolism , Immunization, Passive , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Viral Vaccines/genetics , Viral Vaccines/immunology , Adjuvants, Immunologic , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Antibody-Dependent Cell Cytotoxicity , Complement C1q/genetics , Complement C1q/immunology , Female , Gene Deletion , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Phagocytosis , Vaccination , Viral Envelope Proteins/metabolism , Viral Vaccines/administration & dosage
14.
Front Immunol ; 12: 654652, 2021.
Article En | MEDLINE | ID: mdl-34276649

Objectives: This study aimed to determine the prevalence and localization of complement factor C4d in renal biopsies from patients with lupus nephritis (LN), as well as its associations with the disease's clinico-pathological features. The correlation between arteriolar C4d deposition and renal microvascular lesions (RVLs) was further analyzed. Methods: A total of 325 biopsy-proven LN patients were enrolled, and their clinico-pathological data were collected. C4d staining of renal biopsies was performed by immunohistochemistry. The associations between C4d deposition and the clinico-pathological features were further analyzed. Results: C4d deposition was present in most (98.8%) renal specimens in our cohort. These deposits were localized in the glomeruli (98.2%), tubular basement membrane (TBM) (43.7%), arterioles (31.4%), and peritubular capillary (33.8%). Patients with TBM C4d staining had higher disease activity (measured with the Systemic Lupus Erythematous Disease Activity Index) and higher National Institutes of Health pathological activity and chronicity indices (all P < 0.01). Patients with arteriolar C4d deposition were more likely to develop RVLs (91.2%) compared to those with no arteriolar C4d deposition (78.0%; P = 0.004), especially with two or more types of RVLs (P < 0.001). During the mean follow-up of 55.8 months, arteriolar C4d was related to worse renal outcomes [hazard ration (HR): 2.074, 95% confidence interval (CI) 1.056-4.075, P = 0.034]. Multivariate Cox hazard analysis showed that co-deposition of arteriolar C4d and C3c was an independent risk factor (HR: 3.681, 95% CI 1.519-8.921, P = 0.004) for predicting renal outcomes. Conclusions: C4d deposition was common in renal tissues from LN patients. TBM C4d deposition was related to the disease activity, and arteriolar C4d deposition was associated with RVLs and worse renal outcomes.


Complement C4b/immunology , Complement C4b/metabolism , Disease Susceptibility , Lupus Nephritis/etiology , Lupus Nephritis/metabolism , Peptide Fragments/immunology , Peptide Fragments/metabolism , Adult , Biomarkers , Biopsy , Complement C1q/immunology , Complement C1q/metabolism , Complement C3c/immunology , Complement C3c/metabolism , Female , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Kidney Glomerulus/immunology , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Lupus Nephritis/diagnosis , Lupus Nephritis/mortality , Male , Middle Aged , Prognosis , Young Adult
15.
Front Immunol ; 12: 704072, 2021.
Article En | MEDLINE | ID: mdl-34249009

Complement impacts innate and adaptive immunity. Using a model in which the human KEL glycoprotein is expressed on murine red blood cells (RBCs), we have shown that polyclonal immunoprophylaxis (KELIg) prevents alloimmunization to transfused RBCs when a recipient is in their baseline state of heath but with immunoprophylaxis failure occurring in the presence of a viral-like stimulus. As complement can be detected on antibody coated KEL RBCs following transfusion, we hypothesized that recipient complement synergizes with viral-like inflammation to reduce immunoprophylaxis efficacy. Indeed, we found recipient C3 and C1q were critical to immunoprophylaxis failure in the setting of a viral-like stimulus, with no anti-KEL IgG alloantibodies generated in C3-/- or C1q-/- mice following KELIg treatment and KEL RBC transfusion. Differences in RBC uptake were noted in mice lacking C3, with lower consumption by splenic and peripheral blood inflammatory monocytes. Finally, no alloantibodies were detected in the setting of a viral-like stimulus following KELIg treatment and KEL RBC transfusion in mice lacking complement receptors (CR1/2-/-), narrowing key cells for immunoprophylaxis failure to those expressing these complement receptors. In-vitro studies showed complement fixed opsonized RBCs were significantly less likely to bind to B-cells from CR1/2-/- than wild type mice, potentially implicating lowered B-cell activation threshold in the presence of complement as being responsible for these findings. We thus propose a two-hit model for inflammation-induced immunoprophylaxis failure, where the first "hit" is recipient inflammation and the second "hit" is complement production/sensing. These results may have translational relevance to antigen-antibody interactions in humans.


Complement C1q/immunology , Complement C3/immunology , Erythrocyte Transfusion/adverse effects , Membrane Glycoproteins/immunology , Metalloendopeptidases/immunology , Transfusion Reaction/prevention & control , Animals , B-Lymphocytes/immunology , Complement C1q/genetics , Complement C3/genetics , Erythrocytes , Immunoglobulin G/immunology , Isoantibodies/immunology , Membrane Glycoproteins/genetics , Metalloendopeptidases/genetics , Mice , Mice, Knockout , Transfusion Reaction/genetics , Transfusion Reaction/immunology
16.
Mol Neurobiol ; 58(9): 4323-4336, 2021 Sep.
Article En | MEDLINE | ID: mdl-34002346

Recent studies have identified roles for complement in synaptic pruning, both physiological during development and pathological in Alzheimer's disease (AD). These reports suggest that C1q initiates complement activation on synapses and C3 fragments then tag them for removal by microglia. There is an urgent need to characterise these processes in rodent AD models; this requires the development of reagents and methods for detection and quantification of rodent C1q in fluids and pathological tissues. These will enable better evaluation of the role of C1q in disease and its value as disease biomarker. We describe the generation in C1q-deficient mice of novel monoclonal antibodies against mouse and rat C1q that enabled development of a sensitive, specific, and quantitative ELISA for mouse and rat C1q capable of measuring C1q in biological fluids and tissue extracts. Serum C1q levels were measured in wild-type (WT), C1q knockout (KO), C3 KO, C7 KO, Crry KO, and 3xTg and APPNL-G-F AD model mice through ageing. C1q levels significantly decreased in WT, APPNL-G-F, and C7 KO mice with ageing. C1q levels were reduced in APPNL-G-F compared to WT at all ages and in 3xTg at 12 months; C3 KO and C7 KO, but not Crry KO mice, also demonstrated significantly lower C1q levels compared to matched WT. In brain homogenates, C1q levels increased with age in both WT and APPNL-G-F mice. This robust and adaptable assay for quantification of mouse and rat C1q provides a vital tool for investigating the expression of C1q in rodent models of AD and other complement-driven pathologies.


Alzheimer Disease/immunology , Antibodies, Monoclonal , Brain/immunology , Complement C1q/immunology , Alzheimer Disease/pathology , Animals , Brain/metabolism , Disease Models, Animal , Mice , Rats
17.
Dev Comp Immunol ; 122: 104077, 2021 09.
Article En | MEDLINE | ID: mdl-33905781

As an invertebrate, the compound ascidian Botryllus schlosseri faces nonself only with innate immunity. In this species, we already identified the key components of the lectin and alternative complement activation pathways. In the present work, by mining the transcriptome, we identified a single transcript codifying for a protein, member of the C1q-domain-containing protein family, with a signal peptide followed by two globular C1q (gC1q) domains. It shares a similar domain organisation with C1q/TNF-related proteins 4, the only vertebrate protein family with two gC1q domains. Our gC1q domain-containing protein, called BsC1qDC, is actively transcribed by immunocytes. The transcription is modulated during the Botryllus blastogenetic cycle and is upregulated following the injection of Bacillus clausii cells in the circulation. Furthermore, the injection of bsc1qdc iRNA in the vasculature results in decreased transcription of the gene and a significant impairment of phagocytosis and degranulation, suggesting the involvement of this molecule in immune responses.


Complement C1q/immunology , Immunity, Innate/immunology , Opsonization/immunology , Urochordata/immunology , Amino Acid Sequence , Animals , Bacillus clausii/immunology , Cell Degranulation/immunology , Complement C1q/genetics , Hemocytes/metabolism , Phagocytosis/immunology , Protein Domains , RNA Interference , RNA, Small Interfering/genetics , Transcriptome/genetics
18.
Scand J Immunol ; 94(1): e13048, 2021 Jul.
Article En | MEDLINE | ID: mdl-33914934

Not all anti-HLA donor-specific antibodies (HLA-DSAs) are detrimental to renal allograft. In this context, the C1q complement activating ability of antibodies appears to be an important parameter to distinguish clinically inert versus detrimental DSAs. We evaluated sera of 206 consecutive primary live donor renal transplant recipients before transplant and at post-operative day 7, 30, 90, 180 and at the time of graft dysfunction for quantifying HLA-DSAs using single antigen bead assay on a Luminex platform. Patients positive for these antibodies with an MFI >500 were further screened for C1q fixing nature of DSA. Fourteen of the 18 antibody-positive patients had C1q fixing DSA with MFI value >5000. Only 4 antibody-positive patients did not have C1q fixing DSA. The MFI values of DSA detected by C1q assay were generally higher at least by 25% than those detected by the conventional IgG-SAB assay. Twelve of the 14 patients (85.71%) with C1q+ DSA developed antibody-mediated rejection during the mean follow-up period of 21.43 ± 8.03 months as compared to none of the four C1q-negative DSA (85.71% vs 0%; P = .001). These results suggest deleterious effect of C1q+ DSA vis-à-vis C1q-negative DSA on renal allograft.


Antibodies/immunology , Complement C1q/immunology , Graft Rejection/immunology , Adult , Complement Activation/immunology , Female , Graft Survival/immunology , HLA Antigens/immunology , Humans , Immunoglobulin G/immunology , Kidney Transplantation/adverse effects , Male , Middle Aged , Retrospective Studies , Tissue Donors , Transplantation, Homologous/adverse effects
19.
Methods Mol Biol ; 2227: 107-114, 2021.
Article En | MEDLINE | ID: mdl-33847935

Autoantibodies against complement C1q (anti-C1q) are an excellent marker for active nephritis in SLE patients. Here, we describe a typical protocol for the quantification of anti-C1q using immobilized C1q (important for the presentation of relevant cryptic epitopes) and a high salt buffer for the incubation steps (to prevent immune-complex binding to intact C1q). More recently, a linear epitope on the C1q A chain, that is targeted by anti-C1q, has been described (A08). The assay using this peptide seems to be more specific and more sensitive for the detection of active nephritis in SLE patients than the conventional anti-C1q assay, but further studies are required to establish the role of anti-A08 of C1q in the clinical routine.


Autoantibodies/analysis , Complement C1q/immunology , Diagnostic Tests, Routine , Animals , Autoantibodies/isolation & purification , Biomarkers/analysis , Diagnostic Tests, Routine/methods , Diagnostic Tests, Routine/standards , Diagnostic Tests, Routine/trends , Enzyme-Linked Immunosorbent Assay/methods , Enzyme-Linked Immunosorbent Assay/standards , Enzyme-Linked Immunosorbent Assay/trends , Humans , Inventions , Lupus Erythematosus, Systemic/diagnosis , Lupus Erythematosus, Systemic/immunology , Lupus Nephritis/diagnosis , Lupus Nephritis/immunology , Rabbits , Reference Standards
20.
Front Immunol ; 12: 586521, 2021.
Article En | MEDLINE | ID: mdl-33717067

Antibodies recognizing the amino-terminal domain of receptor subunit proteins modify the receptor efficiency to controlling transmitter release in isolated nerve endings (e.g., synaptosomes) indirectly confirming their presence in these particles but also allowing to speculate on their subunit composition. Western blot analysis and confocal microscopy unveiled the presence of the GluA1, GluA2, GluA3, and GluA4 receptor subunits in cortical synaptosomes. Functional studies confirmed the presence of presynaptic release-regulating AMPA autoreceptors in these terminals, whose activation releases [3H]D-aspartate ([3H]D-Asp, here used as a marker of glutamate) in a NBQX-dependent manner. The AMPA autoreceptors traffic in a constitutive manner, since entrapping synaptosomes with the pep2-SVKI peptide (which interferes with the GluA2-GRIP1/PICK1 interaction) amplified the AMPA-evoked releasing activity, while the inactive pep2-SVKE peptide was devoid of activity. Incubation of synaptosomes with antibodies recognizing the NH2 terminus of the GluA2 and the GluA3 subunits increased, although to a different extent, the GluA2 and 3 densities in synaptosomal membranes, also amplifying the AMPA-evoked glutamate release in a NBQX-dependent fashion. We then analyzed the releasing activity of complement (1:300) from both treated and untreated synaptosomes and found that the complement-induced overflow occurred in a DL-t-BOA-sensitive, NBQX-insensitive fashion. We hypothesized that anti-GluA/GluA complexes in neuronal membranes could trigger the classic pathway of activation of the complement, modifying its releasing activity. Accordingly, the complement-evoked release of [3H]D-Asp from antiGluA2 and anti-GluA3 antibody treated synaptosomes was significantly increased when compared to untreated terminals and facilitation was prevented by omitting the C1q component of the immunocomplex. Antibodies recognizing the NH2 terminus of the GluA1 or the GluA4 subunits failed to affect both the AMPA and the complement-evoked tritium overflow. Our results suggest the presence of GluA2/GluA3-containing release-regulating AMPA autoreceptors in cortical synaptosomes. Incubation of synaptosomes with commercial anti-GluA2 or anti-GluA3 antibodies amplifies the AMPA-evoked exocytosis of glutamate through a complement-independent pathway, involving an excessive insertion of AMPA autoreceptors in plasma membranes but also affects the complement-dependent releasing activity, by promoting the classic pathway of activation of the immunocomplex. Both events could be relevant to the development of autoimmune diseases typified by an overproduction of anti-GluA subunits.


Antibodies/pharmacology , Protein Interaction Domains and Motifs/drug effects , Protein Subunits/antagonists & inhibitors , Receptors, AMPA/antagonists & inhibitors , Animals , Cell Membrane/drug effects , Cell Membrane/metabolism , Cerebral Cortex/metabolism , Complement C1q/immunology , Fluorescent Antibody Technique , Male , Mice , Receptors, AMPA/chemistry , Synaptosomes/drug effects , Synaptosomes/metabolism
...