Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 75
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Semin Immunol ; 45: 101338, 2019 10.
Article in English | MEDLINE | ID: mdl-31744753

ABSTRACT

In the past several years, a number of C1q binding surface proteins or receptors have been described. This is not of course surprising considering the complexity of the C1q molecule and its ability to bind to a wide range of cellular and plasma proteins via both its collagen-like [cC1q] region and its heterotrimeric globular heads [gC1q] each of which in turn is capable of binding a specific ligand. However, while each of these "receptor" molecules undoubtedly plays a specific function within its restricted microenvironment, and therefore merits full attention, this review nonetheless, will singularly focus on the structure and function of gC1qR-a multi-functional and multi-compartmental protein, which plays an important role in inflammation, infection, and cancer. Although first identified as a receptor for C1q, gC1qR has been shown to bind to a plethora of proteins found in plasma, on the cell surface and on pathogenic microorganisms. The plasma proteins that bind to gC1qR are mostly blood coagulation proteins and include high molecular weight kininogen [HK], Factor XII [Hageman factor], fibrinogen, thrombin [FII], and multimeric vitronectin. This suggests that gC1qR can play an important role in modulating not only of fibrin formation, particularly at local sites of immune injury and/or inflammation, but by activating the kinin/kallikrein system, it is also able to generate, bradykinin, a powerful vasoactive peptide that is largely responsible for the swelling seen in angioedema. Another important function of gC1qR is in cancer, where it has been shown to play a role in tumor cell survival, growth and metastatic invasion by interacting with critical molecules in the tumor cell microenvironment including those of the complement system and kinin system. Finally, by virtue of its ability to interact with a growing list of pathogen-associated molecules, including bacterial and viral ligands, gC1qR is becoming recognized as an important pathogen recognition receptor [PRR]. Given the numerous roles it plays in a growing list of disease settings, gC1qR has now become a potential target for the development of monoclonal antibody-based and/or small molecule-based therapies.


Subject(s)
Membrane Glycoproteins/metabolism , Receptors, Complement/metabolism , Animals , Carrier Proteins/chemistry , Carrier Proteins/genetics , Carrier Proteins/metabolism , Chromosome Mapping , Disease Susceptibility , Host-Parasite Interactions , Host-Pathogen Interactions , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Molecular Structure , Receptors, Complement/chemistry , Receptors, Complement/genetics , Structure-Activity Relationship
2.
Immunol Rev ; 274(1): 281-289, 2016 11.
Article in English | MEDLINE | ID: mdl-27782339

ABSTRACT

The blood plasma contains four biologically important proteolytic cascades, which probably evolved from the same ancestral gene. This in part may explain why each cascade has very similar "initiating trigger" followed by sequential and cascade-like downstream enzymatic activation pattern. The four cascades are: the complement system, the blood clotting cascade, the fibrinolytic system, and the kallikrein-kinin system. Although much has been written about the interplay between all these enzymatic cascades, the cross-talk between the complement and the kinin generating systems has become particularly relevant as this interaction results in the generation of nascent molecules that have significant impact in various inflammatory diseases including angioedema and cancer. In this review, we will focus on the consequences of the interplay between the two systems by highlighting the role of a novel molecular link called gC1qR. Although this protein was first identified as a receptor for C1q, it is now recognized as a multiligand binding cellular protein, which serves not only as C1q receptor, but also as high affinity (KD  ≤ 0.8 nM) binding site for both high molecular weight kininogen (HK) and factor XII (FXII). At inflammatory sites, where atherogenic factors such as immune complexes and/or pathogens can activate the endothelial cell into a procoagulant and proinflammatory surface, the two pathways are activated to generate vasoactive peptides that contribute in various ways to the inflammatory processes associated with numerous diseases. More importantly, since recent observations strongly suggest an important role for both pathways in cancer, we will focus on how a growing tumor cluster can employ the byproducts derived from the two activation systems to ensure not only its survival and growth, but also its escape into distal sites of colonization.


Subject(s)
Angioedema/immunology , Atherosclerosis/immunology , Complement C1q/metabolism , Factor XIa/metabolism , Immunity, Innate , Inflammation/immunology , Neoplasms/immunology , Animals , Humans , Proteolysis , Receptor Cross-Talk , Signal Transduction
3.
J Immunol ; 192(1): 377-84, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24319267

ABSTRACT

Bradykinin (BK) is one of the most potent vasodilator agonists known and belongs to the kinin family of proinflammatory peptides. BK induces its activity via two G protein-coupled receptors: BK receptor 1 (B1R) and BK receptor 2. Although BK receptor 2 is constitutively expressed on endothelial cells (ECs), B1R is induced by IL-1ß. The C1q receptor, receptor for the globular heads of C1q (gC1qR), which plays a role in BK generation, is expressed on activated ECs and is also secreted as soluble gC1qR (sgC1qR). Because sgC1qR can bind to ECs, we hypothesized that it may also serve as an autocrine/paracrine signal for the induction of B1R expression. In this study, we show that gC1qR binds to ECs via a highly conserved domain consisting of residues 174-180, as assessed by solid-phase binding assay and deconvolution fluorescence microscopy. Incubation of ECs (24 h, 37 °C) with sgC1qR resulted in enhancement of B1R expression, whereas incubation with gC1qR lacking aa 174-180 and 154-162 had a diminished effect. Binding of sgC1qR to ECs was through surface-bound fibrinogen and was inhibited by anti-fibrinogen. In summary, our data suggest that, at sites of inflammation, sgC1qR can enhance vascular permeability by upregulation of B1R expression through de novo synthesis, as well as rapid translocation of preformed B1R.


Subject(s)
Autocrine Communication , Carrier Proteins/metabolism , Endothelial Cells/metabolism , Mitochondrial Proteins/metabolism , Receptor, Bradykinin B1/metabolism , Signal Transduction , Autocrine Communication/drug effects , Binding Sites , Carrier Proteins/chemistry , Carrier Proteins/pharmacology , Cell Line , Cell Membrane/metabolism , Endothelial Cells/drug effects , Fibrinogen/metabolism , Gene Expression Regulation/drug effects , Humans , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/pharmacology , Protein Binding , Protein Interaction Domains and Motifs , Protein Transport , Receptor, Bradykinin B1/genetics , Signal Transduction/drug effects
4.
Neurocrit Care ; 24(1): 6-46, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26714677

ABSTRACT

BACKGROUND: The use of antithrombotic agents, including anticoagulants, antiplatelet agents, and thrombolytics has increased over the last decade and is expected to continue to rise. Although antithrombotic-associated intracranial hemorrhage can be devastating, rapid reversal of coagulopathy may help limit hematoma expansion and improve outcomes. METHODS: The Neurocritical Care Society, in conjunction with the Society of Critical Care Medicine, organized an international, multi-institutional committee with expertise in neurocritical care, neurology, neurosurgery, stroke, hematology, hemato-pathology, emergency medicine, pharmacy, nursing, and guideline development to evaluate the literature and develop an evidence-based practice guideline. Formalized literature searches were conducted, and studies meeting the criteria established by the committee were evaluated. RESULTS: Utilizing the GRADE methodology, the committee developed recommendations for reversal of vitamin K antagonists, direct factor Xa antagonists, direct thrombin inhibitors, unfractionated heparin, low-molecular weight heparin, heparinoids, pentasaccharides, thrombolytics, and antiplatelet agents in the setting of intracranial hemorrhage. CONCLUSIONS: This guideline provides timely, evidence-based reversal strategies to assist practitioners in the care of patients with antithrombotic-associated intracranial hemorrhage.


Subject(s)
Critical Care/standards , Fibrinolytic Agents/administration & dosage , Fibrinolytic Agents/adverse effects , Intracranial Hemorrhages/chemically induced , Intracranial Hemorrhages/drug therapy , Neurology/standards , Practice Guidelines as Topic/standards , Societies, Medical/standards , Humans
5.
Transfusion ; 55(8): 2001-9, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25808236

ABSTRACT

BACKGROUND: Successful peripheral blood stem cell transplantation (PBSCT) depends on the collection and infusion of adequate numbers of peripheral blood progenitor cells (PBPCs). Several predictors of PBPC yield are used currently, including white blood cell (WBC) count and CD34 analysis. This study evaluated the utility of the new automated hematopoietic progenitor cell count available on Sysmex XN hematology analyzers (XN-HPCs) in PBSCT. STUDY DESIGN AND METHODS: The performance characteristics of XN-HPC, CD34+, and WBC analysis were compared using 107 matched peripheral blood and apheresis samples. RESULTS: Good correlation was observed between XN-HPC and CD34+ cell counts in peripheral blood (r = 0.88; slope, 0.81) and apheresis collections (r = 0.91; slope, 0.89). Moreover, peripheral blood XN-HPC and CD34 analysis showed comparable ability to predict successful PBPC harvests (≥2 × 10(6) CD34+ cells/kg). At a cutoff of 20 × 10(6) progenitor cells/L, peripheral blood XN- HPC and CD34 analysis both showed negative predictive values (NPVs) of 100% and positive predictive values (PPVs) of 55.4 and 63%, respectively. Using an optimized cutoff of 38 × 10(6) progenitor cells/L, derived from receiver operating characteristic analysis, the PPV for XN-HPC and CD34 analysis increased to 71.4 and 78.9%, respectively, with relatively unchanged NPVs (XN-HPC 97.7%, CD34+ 98.0%). In contrast, the correlation between peripheral blood WBC and CD34 analysis was poor (r = 0.48; slope, 669.85), and the peripheral blood WBC count (cutoff, 10 × 10(9) /L) was a poor predictor of PBPC harvest (NPV 60%, PPV 43.1%). CONCLUSION: XN-HPC compares favorably with CD34 analysis and may be a surrogate for CD34 analysis to predict optimal timing of PBPC collections.


Subject(s)
Blood Cell Count/instrumentation , Hematopoietic Stem Cells/cytology , Peripheral Blood Stem Cell Transplantation , Adolescent , Adult , Aged , Antigens, CD34/blood , Automation , Blood Component Removal , Cell Nucleus/ultrastructure , Cell Size , Child , Child, Preschool , Cytoplasmic Granules/ultrastructure , Equipment Design , Female , Flow Cytometry/instrumentation , Hematopoietic Stem Cell Mobilization , Humans , Infant , Leukocyte Count , Male , Middle Aged , ROC Curve , Reproducibility of Results , Young Adult
6.
Blood ; 120(6): 1228-36, 2012 Aug 09.
Article in English | MEDLINE | ID: mdl-22700724

ABSTRACT

C1q modulates the differentiation and function of cells committed to the monocyte-derived dendritic cell (DC) lineage. Because the 2 C1q receptors found on the DC surface-gC1qR and cC1qR-lack a direct conduit into intracellular elements, we postulated that the receptors must form complexes with transmembrane partners. In the present study, we show that DC-SIGN, a C-type lectin expressed on DCs, binds directly to C1q, as assessed by ELISA, flow cytometry, and immunoprecipitation experiments. Surface plasmon resonance analysis revealed that the interaction was specific, and both intact C1q and the globular portion of C1q bound to DC-SIGN. Whereas IgG reduced this binding significantly, the Arg residues (162-163) of the C1q-A chain, which are thought to contribute to the C1q-IgG interaction, were not required for C1q binding to DC-SIGN. Binding was reduced significantly in the absence of Ca(2+) and by preincubation of DC-SIGN with mannan, suggesting that C1q binds to DC-SIGN at its principal Ca(2+)-binding pocket, which has increased affinity for mannose residues. Antigen-capture ELISA and immunofluorescence microscopy revealed that C1q and gC1qR associate with DC-SIGN on blood DC precursors and immature DCs. The results of the present study suggest that C1q/gC1qR may regulate DC differentiation and function through the DC-SIGN-mediated induction of cell-signaling pathways.


Subject(s)
Carrier Proteins/metabolism , Cell Adhesion Molecules/metabolism , Complement C1q/metabolism , Dendritic Cells/metabolism , Lectins, C-Type/metabolism , Mitochondrial Proteins/metabolism , Multiprotein Complexes/metabolism , Receptors, Cell Surface/metabolism , Binding, Competitive , Calcium/pharmacology , Cell Differentiation/immunology , Cells, Cultured , Dendritic Cells/physiology , Humans , Models, Biological , Monocytes/metabolism , Monocytes/physiology , Multiprotein Complexes/physiology , Protein Binding/drug effects , Protein Multimerization/drug effects , Protein Multimerization/physiology
7.
Front Immunol ; 15: 1351656, 2024.
Article in English | MEDLINE | ID: mdl-38711524

ABSTRACT

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.


Subject(s)
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
8.
Adv Exp Med Biol ; 735: 97-110, 2013.
Article in English | MEDLINE | ID: mdl-23402021

ABSTRACT

Abstract The receptor for the globular heads of C1q, gC1qR/p33, is a widely expressed cellular protein, which binds to diverse ligands including plasma proteins, cellular proteins, and microbial ligands. In addition to C1q, gC1qR also binds high molecular weight kininogen (HK), which also has two other cell surface sites, namely, cytokeratin 1 and urokinase plasminogen activator receptor (uPAR). On endothelial cells (ECs), the three molecules form two closely associated bimolecular complexes of gC1qR/cytokeratin 1 and uPAR/cytokeratin 1. However, by virtue of its high affinity for HK, gC1qR plays a central role in the assembly of the kallikrein-kinin system, leading to the generation of bradykinin (BK). BK in turn is largely responsible for the vascular leakage and associated inflammation seen in angioedema patients. Therefore, blockade of gC1qR by inhibitory peptides or antibodies may not only prevent the generation of BK but also reduce Clq-induced or microbial-ligand-induced inflammatory responses. Employing synthetic peptides and gClqR deletion mutants, we confirmed previously predicted sites for C1q (residues 75-96) and HK (residues 204-218) and identified additional sites for both C1q and HK (residues 190-202), for C1q (residues 144-162), and for HIV-1 gp41 (residues 174-180). With the exception of residues 75-96, which is located in the alphaA coiled-coil N-terminal segment, most of the identified residues form part of the highly charged loops connecting the various beta-strands in the crystal structure. Taken together, the data support the notion that gC1qR could serve as a novel molecular target for the design of antibody-based and/or peptide-based therapy to attenuate acute and/or chronic inflammation associated with vascular leakage and infection.


Subject(s)
Complement C1q/drug effects , Infections/drug therapy , Inflammation/drug therapy , Receptors, Complement/drug effects , Animals , Humans , Ligands , Neoplasms/drug therapy , Neoplasms/physiopathology , Receptors, Complement/chemistry , Receptors, Complement/genetics
9.
J Mol Diagn ; 24(8): 867-877, 2022 08.
Article in English | MEDLINE | ID: mdl-35934321

ABSTRACT

Detection of serum embryonic miRNAs miR-371a-3p and miR-372-3p has been proposed to aid in diagnosis, prognosis, and management of patients with testicular germ cell tumors (GCTs). This study describes the analytical validation and performance of a laboratory-developed test to detect these miRNA targets by stem loop real-time quantitative RT-PCR (RT-qPCR) in serum from patients with GCTs. The assay was standardized using an exogenous spike-in control of nonhuman miRNA from Caenorhabditis elegans (cel-miR-39-3p) to assess extraction efficiency, and an endogenous housekeeping miRNA, miR-30b-5p, to control for miRNA normalization. miRNA results were expressed as relative expression level, using the comparative threshold cycle method (2ΔΔCT). Analytical sensitivity of miR-371a-3p and miR-372-3p was 12.5 and 1.25 copies/µL, respectively. Clinical accuracy was evaluated using GCT patients with (n = 34) and without (n = 17) active disease. Positive/negative cutoffs and indeterminate findings were established on the basis of results from healthy volunteers (n = 25) and assay precision. miR-371a-3p and miR-372-3p exhibited a sensitivity of 81.8% and 87.5%, respectively, and a specificity of 100% and 94%, respectively, and an area under the receiver operating characteristic curve of 0.93 and 0.95, respectively. Taken together, RT-qPCR testing for serum miR-371a-3p and miR-372-3p represents a robust, sensitive, and specific clinical assay to aid in the clinical management of patients with GCT.


Subject(s)
MicroRNAs , Neoplasms, Germ Cell and Embryonal , Biomarkers, Tumor/genetics , Humans , Laboratories, Clinical , Male , Neoplasms, Germ Cell and Embryonal/diagnosis , Neoplasms, Germ Cell and Embryonal/genetics , Testicular Neoplasms
12.
Microvasc Res ; 82(1): 66-72, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21539847

ABSTRACT

Endovascular infections with Staphylococcus aureus (S. aureus) are associated with high mortality. gC1qR/p33 (gC1qR), a receptor for the complement component C1q expressed on endothelial cells, interacts with protein A of S. aureus and gC1qR blockade reduces S. aureus colonization during infective endocarditis. The aim of this study was to analyze in vivo whether this observation is due to a decreased interaction of S. aureus with the microvascular endothelium. A dorsal skinfold chamber was prepared in Syrian golden hamsters, which were treated with the monoclonal antibody (MAb) 74.5.2 directed against gC1qR or vehicle. The interaction of fluorescein isothiocyanate (FITC)-labeled staphylococci and leukocytes with the endothelium was analyzed under physiological conditions as well as after TNF-α-induced inflammation using intravital fluorescence microscopy. Administration of MAb 74.5.2 significantly reduced adherence of S. aureus to the endothelium in untreated and TNF-α-exposed tissue. In addition, we could demonstrate in vitro that S. aureus adherence to human endothelial cells was inhibited by MAb 74.5.2. Blockade of gC1qR did not affect leukocyte-endothelial cell interaction. In conclusion, our findings indicate that immunological inhibition of gC1qR may be therapeutically used to decrease the interaction of S. aureus with the microvascular endothelium.


Subject(s)
Cell Adhesion/physiology , Endothelium, Vascular/microbiology , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/metabolism , Microvessels/microbiology , Receptors, Complement/antagonists & inhibitors , Receptors, Complement/metabolism , Staphylococcus aureus/cytology , Animals , Antibodies, Monoclonal/blood , Antibodies, Monoclonal/pharmacology , Cell Adhesion/drug effects , Cell Line , Cricetinae , Dermatologic Surgical Procedures , Endothelial Cells/drug effects , Endothelial Cells/microbiology , Endothelium, Vascular/drug effects , Fluorescein-5-isothiocyanate/chemistry , Humans , Inflammation/chemically induced , Inflammation/microbiology , Inflammation/pathology , Leukocyte Rolling/drug effects , Leukocytes/pathology , Male , Membrane Glycoproteins/immunology , Mesocricetus , Microscopy, Fluorescence , Microvessels/drug effects , Receptors, Complement/immunology , Regional Blood Flow/drug effects , Regional Blood Flow/physiology , Skin/blood supply , Staining and Labeling , Tumor Necrosis Factor-alpha/pharmacology
13.
J Immunol ; 183(9): 6013-20, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19828637

ABSTRACT

Mixed cryoglobulinemia (MC) is a lymphoproliferative disorder observed in approximately 10 to 15% of hepatitis C virus (HCV)-infected patients. Circulating, nonenveloped HCV core protein, which has been detected in cryoprecipitable immune complexes, interacts with immunocytes through the receptor for the globular domain of C1q protein (gC1q-R). In this study, we have evaluated circulating gC1q-R levels in chronically HCV-infected patients, with and without MC. These levels were significantly higher in MC patients than in those without MC and in healthy controls and paralleled specific mRNA expression in PBL. Soluble gC1q-R circulates as a complexed form containing both C1q and HCV core proteins. Higher serum gC1q-R levels negatively correlated with circulating concentrations of the C4d fragment. The presence of sequestered C4d in the vascular bed of skin biopsies from MC patients was indicative of in situ complement activation. In vitro studies showed that release of soluble gC1q-R is regulated by HCV core-mediated inhibition of cell proliferation. Our results indicate that up-regulation of gC1q-R expression is a distinctive feature of MC, and that dysregulated shedding of C1q-R molecules contributes to vascular cryoglobulin-induced damage via the classic complement-mediated pathway.


Subject(s)
Complement C1q/metabolism , Cryoglobulinemia/immunology , Cryoglobulins/adverse effects , Hepacivirus/immunology , Hepatitis C, Chronic/immunology , Membrane Glycoproteins/physiology , Receptors, Complement/physiology , Vasculitis/immunology , Complement Pathway, Classical/immunology , Cryoglobulinemia/metabolism , Cryoglobulinemia/virology , Female , Hepatitis C, Chronic/metabolism , Hepatitis C, Chronic/virology , Humans , Male , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/blood , Middle Aged , Protein Structure, Tertiary , Receptors, Complement/biosynthesis , Receptors, Complement/blood , Up-Regulation/immunology , Vasculitis/metabolism , Vasculitis/virology , Viral Core Proteins/immunology , Viral Core Proteins/metabolism
14.
Front Immunol ; 12: 716361, 2021.
Article in English | MEDLINE | ID: mdl-34491250

ABSTRACT

Background: COVID-19 pathology is associated with exuberant inflammation, vascular damage, and activation of coagulation. In addition, complement activation has been described and is linked to disease pathology. However, few studies have been conducted in cancer patients. Objective: This study examined complement activation in response to COVID-19 in the setting of cancer associated thromboinflammation. Methods: Markers of complement activation (C3a, C5a, sC5b-9) and complement inhibitors (Factor H, C1-Inhibitor) were evaluated in plasma of cancer patients with (n=43) and without (n=43) COVID-19 and stratified based on elevated plasma D-dimer levels (>1.0 µg/ml FEU). Markers of vascular endothelial cell dysfunction and platelet activation (ICAM-1, thrombomodulin, P-selectin) as well as systemic inflammation (pentraxin-3, serum amyloid A, soluble urokinase plasminogen activator receptor) were analyzed to further evaluate the inflammatory response. Results: Increases in circulating markers of endothelial cell dysfunction, platelet activation, and systemic inflammation were noted in cancer patients with COVID-19. In contrast, complement activation increased in cancer patients with COVID-19 and elevated D-dimers. This was accompanied by decreased C1-Inhibitor levels in patients with D-dimers > 5 ug/ml FEU. Conclusion: Complement activation in cancer patients with COVID-19 is significantly increased in the setting of thromboinflammation. These findings support a link between coagulation and complement cascades in the setting of inflammation.


Subject(s)
COVID-19/immunology , Complement Activation/immunology , Inflammation/immunology , Neoplasms/immunology , SARS-CoV-2/immunology , Thrombosis/immunology , Adolescent , Adult , Aged , Aged, 80 and over , Biomarkers/blood , COVID-19/blood , COVID-19/virology , Complement Inactivating Agents/blood , Female , Fibrin Fibrinogen Degradation Products/metabolism , Humans , Inflammation/blood , Male , Middle Aged , Neoplasms/blood , Platelet Activation/immunology , Retrospective Studies , SARS-CoV-2/physiology , Thrombosis/blood , Young Adult
15.
Front Immunol ; 12: 767347, 2021.
Article in English | MEDLINE | ID: mdl-34804054

ABSTRACT

Infection with SARS-CoV-2 triggers the simultaneous activation of innate inflammatory pathways including the complement system and the kallikrein-kinin system (KKS) generating in the process potent vasoactive peptides that contribute to severe acute respiratory syndrome (SARS) and multi-organ failure. The genome of SARS-CoV-2 encodes four major structural proteins - the spike (S) protein, nucleocapsid (N) protein, membrane (M) protein, and the envelope (E) protein. However, the role of these proteins in either binding to or activation of the complement system and/or the KKS is still incompletely understood. In these studies, we used: solid phase ELISA, hemolytic assay and surface plasmon resonance (SPR) techniques to examine if recombinant proteins corresponding to S1, N, M and E: (a) bind to C1q, gC1qR, FXII and high molecular weight kininogen (HK), and (b) activate complement and/or the KKS. Our data show that the viral proteins: (a) bind C1q and activate the classical pathway of complement, (b) bind FXII and HK, and activate the KKS in normal human plasma to generate bradykinin and (c) bind to gC1qR, the receptor for the globular heads of C1q (gC1q) which in turn could serve as a platform for the activation of both the complement system and KKS. Collectively, our data indicate that the SARS-CoV-2 viral particle can independently activate major innate inflammatory pathways for maximal damage and efficiency. Therefore, if efficient therapeutic modalities for the treatment of COVID-19 are to be designed, a strategy that includes blockade of the four major structural proteins may provide the best option.


Subject(s)
Antigens, Viral/immunology , COVID-19/immunology , Complement System Proteins/immunology , Kallikrein-Kinin System , SARS-CoV-2/immunology , Viral Structural Proteins/immunology , Carrier Proteins/genetics , Carrier Proteins/immunology , Hemolysis , Humans , Mitochondrial Proteins/genetics , Mitochondrial Proteins/immunology , Recombinant Proteins/immunology , Viral Structural Proteins/genetics
16.
Cell Mol Gastroenterol Hepatol ; 12(1): 229-250, 2021.
Article in English | MEDLINE | ID: mdl-33515804

ABSTRACT

BACKGROUND & AIMS: Cell differentiation in the colonic crypt is driven by a metabolic switch from glycolysis to mitochondrial oxidation. Mitochondrial and goblet cell dysfunction have been attributed to the pathology of ulcerative colitis (UC). We hypothesized that p32/gC1qR/HABP1, which critically maintains oxidative phosphorylation, is involved in goblet cell differentiation and hence in the pathogenesis of UC. METHODS: Ex vivo, goblet cell differentiation in relation to p32 expression and mitochondrial function was studied in tissue biopsies from UC patients versus controls. Functional studies were performed in goblet cell-like HT29-MTX cells in vitro. Mitochondrial respiratory chain complex V-deficient, ATP8 mutant mice were utilized as a confirmatory model. Nutritional intervention studies were performed in C57BL/6 mice. RESULTS: In UC patients in remission, colonic goblet cell differentiation was significantly decreased compared to controls in a p32-dependent manner. Plasma/serum L-lactate and colonic pAMPK level were increased, pointing at high glycolytic activity and energy deficiency. Consistently, p32 silencing in mucus-secreting HT29-MTX cells abolished butyrate-induced differentiation and induced a shift towards glycolysis. In ATP8 mutant mice, colonic p32 expression correlated with loss of differentiated goblet cells, resulting in a thinner mucus layer. Conversely, feeding mice an isocaloric glucose-free, high-protein diet increased mucosal energy supply that promoted colonic p32 level, goblet cell differentiation and mucus production. CONCLUSION: We here describe a new molecular mechanism linking mucosal energy deficiency in UC to impaired, p32-dependent goblet cell differentiation that may be therapeutically prevented by nutritional intervention.


Subject(s)
Carrier Proteins/metabolism , Colitis, Ulcerative/metabolism , Colon/metabolism , Goblet Cells/metabolism , Mitochondrial Proteins/metabolism , Animals , Carrier Proteins/genetics , Cell Differentiation , Colitis, Ulcerative/pathology , Goblet Cells/pathology , Humans , Male , Mice , Mice, Inbred C57BL , Mitochondrial Proteins/genetics , Tumor Cells, Cultured
17.
Br J Haematol ; 148(4): 638-45, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19925495

ABSTRACT

The role of the complement system in immune thrombocytopenic purpura (ITP) is not well defined. We examined plasma from 79 patients with ITP, 50 healthy volunteers, and 25 patients with non-immune mediated thrombocytopenia, to investigate their complement activation/fixation capacity (CAC) on immobilized heterologous platelets. Enhanced CAC was found in 46 plasma samples (59%) from patients with ITP, but no samples from patients with non-immune mediated thrombocytopenia. Plasma from healthy volunteers was used for comparison. In patients with ITP, an enhanced plasma CAC was associated with a decreased circulating absolute immature platelet fraction (A-IPF) (<15 x 10(9)/l) (P = 0.027) and thrombocytopenia (platelet count < 100 x 10(9)/l) (P = 0.024). The positive predictive value of an enhanced CAC for a low A-IPF was 93%, with a specificity of 77%. The specificity and positive predictive values increased to 100% when plasma CAC was defined strictly by enhanced C1q and/or C4d deposition on test platelets. Although no statistically significant correlation emerged between CAC and response to different pharmacological therapies, an enhanced response to splenectomy was noted (P < 0.063). Thus, complement fixation may contribute to the thrombocytopenia of ITP by enhancing clearance of opsonized platelets from the circulation, and/or directly damaging platelets and megakaryocytes.


Subject(s)
Blood Platelets/immunology , Complement Activation/immunology , Purpura, Thrombocytopenic, Idiopathic/immunology , Adolescent , Adult , Aged , Child , Child, Preschool , Complement Fixation Tests/methods , Female , Humans , Male , Middle Aged , Platelet Count , Predictive Value of Tests , Purpura, Thrombocytopenic, Idiopathic/blood , Sensitivity and Specificity , Thrombocytopenia/immunology , Young Adult
18.
Am J Hematol ; 85(9): 726, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20645431

ABSTRACT

APTT testing is integral to hemostasis testing. A prolonged result, however, can be difficult to interpret, depending on the APTT reagent's sensitivity to the lupus anticoagulant. This often generates additional laboratory testing for both factor deficiencies and the presence of a lupus anticoagulant, and in so doing, delays patient management. We have found it useful to provide APTT testing with both a lupus anticoagulant sensitive and insensitive reagent, to facilitate the rapid exclusion of significant factor deficiencies. The following case report illustrates the utility of this approach and provides a backdrop for necessary discussions between laboratories and clinicians regarding which APTT reagent best meets their clinical need for screening hemostasis testing.


Subject(s)
Hemorrhage/blood , Lupus Coagulation Inhibitor/blood , Organometallic Compounds/chemistry , Triazoles/chemistry , Blood Coagulation Tests , Female , Humans , Middle Aged
19.
J Allergy Clin Immunol ; 124(6): 1303-10.e4, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19796797

ABSTRACT

BACKGROUND: Activation of bradykinin-mediated B2 receptor has been shown to play an important role in the onset of angioedema associated with C1 inhibitor deficiency. This finding has led to the development of novel therapeutic drugs such as the B2 receptor antagonist icatibant. However, it is unclear whether other receptors expressed on endothelial cells contribute to the release of kinins and vascular leakage in these patients. The recognition of their role may have obvious therapeutic implications. OBJECTIVE: Our aim was to investigate the involvement of B1 and gC1q receptors in in vitro and in vivo models of vascular leakage induced by plasma samples obtained from patients with C1 inhibitor deficiency. METHODS: The vascular leakage was evaluated in vitro on endothelial cells by a transwell model system and in vivo on rat mesentery microvessels by intravital microscopy. RESULTS: We observed that the attack phase plasma from C1 inhibitor-deficient patients caused a delayed fluorescein-labeled albumin leakage as opposed to the rapid effect of bradykinin, whereas remission plasma elicited a modest effect compared with control plasma. The plasma permeabilizing effect was prevented by blocking the gC1q receptor-high-molecular-weight kininogen interaction, was partially inhibited by B2 receptor or B1 receptor antagonists, and was totally prevented by the mixture of the 2 antagonists. Involvement of B1 receptor was supported by the finding that albumin leakage caused by attack phase plasma was enhanced by IL-1beta and was markedly reduced by brefeldin A. CONCLUSION: Our data suggest that both B1 receptor and gC1q receptor are involved in the vascular leakage induced by hereditary and acquired angioedema plasma.


Subject(s)
Angioedema/physiopathology , Capillary Permeability/drug effects , Complement C1 Inhibitor Protein/metabolism , Membrane Glycoproteins/metabolism , Receptor, Bradykinin B1/metabolism , Receptors, Complement/metabolism , Adrenergic beta-Antagonists/pharmacology , Adrenergic beta-Antagonists/therapeutic use , Angioedema/drug therapy , Animals , Antibodies, Monoclonal/pharmacology , Blood Vessels/drug effects , Blood Vessels/immunology , Blood Vessels/metabolism , Bradykinin/analogs & derivatives , Bradykinin/pharmacology , Bradykinin/therapeutic use , Bradykinin B1 Receptor Antagonists , Brefeldin A/pharmacology , Capillary Permeability/physiology , Cell Line, Tumor , Complement C1 Inhibitor Protein/genetics , Hereditary Angioedema Types I and II/drug therapy , Hereditary Angioedema Types I and II/physiopathology , Humans , Immunologic Factors/pharmacology , Interleukin-1beta/pharmacology , Male , Protein Synthesis Inhibitors/pharmacology , Rats , Rats, Inbred WKY
20.
J Clin Invest ; 130(11): 5674-5676, 2020 11 02.
Article in English | MEDLINE | ID: mdl-32925166

ABSTRACT

In a stunningly short period of time, the unexpected coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has turned the unprepared world topsy-turvy. Although the rapidity with which the virus struck was indeed overwhelming, scientists throughout the world have been up to the task of deciphering the mechanisms by which SARS-CoV-2 induces the multisystem and multiorgan inflammatory responses that, collectively, contribute to the high mortality rate in affected individuals. In this issue of the JCI, Skendros and Mitsios et al. is one such team who report that the complement system plays a substantial role in creating the hyperinflammation and thrombotic microangiopathy that appear to contribute to the severity of COVID-19. In support of the hypothesis that the complement system along with neutrophils and platelets contributes to COVID-19, the authors present empirical evidence showing that treatment with the complement inhibitor compstatin Cp40 inhibited the expression of tissue factor in neutrophils. These results confirm that the complement axis plays a critical role and suggest that targeted therapy using complement inhibitors is a potential therapeutic option to treat COVID-19-induced inflammation.


Subject(s)
Betacoronavirus/metabolism , Complement Activation/drug effects , Coronavirus Infections , Pandemics , Peptides, Cyclic/pharmacology , Pneumonia, Viral , Thromboplastin/biosynthesis , Thrombotic Microangiopathies , Blood Platelets/metabolism , Blood Platelets/pathology , COVID-19 , Coronavirus Infections/drug therapy , Coronavirus Infections/metabolism , Coronavirus Infections/pathology , Humans , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Inflammation/virology , Neutrophils/metabolism , Neutrophils/pathology , Pneumonia, Viral/drug therapy , Pneumonia, Viral/metabolism , Pneumonia, Viral/pathology , SARS-CoV-2 , Severity of Illness Index , Thrombotic Microangiopathies/drug therapy , Thrombotic Microangiopathies/metabolism , Thrombotic Microangiopathies/pathology , Thrombotic Microangiopathies/virology
SELECTION OF CITATIONS
SEARCH DETAIL