Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 231
Filter
1.
Mar Drugs ; 19(12)2021 Nov 26.
Article in English | MEDLINE | ID: mdl-34940667

ABSTRACT

C1q domain-containing (C1qDC) proteins are a group of biopolymers involved in immune response as pattern recognition receptors (PRRs) in a lectin-like manner. A new protein MkC1qDC from the hemolymph plasma of Modiolus kurilensis bivalve mollusk widespread in the Northwest Pacific was purified. The isolation procedure included ammonium sulfate precipitation followed by affinity chromatography on pectin-Sepharose. The full-length MkC1qDC sequence was assembled using de novo mass-spectrometry peptide sequencing complemented with N-terminal Edman's degradation, and included 176 amino acid residues with molecular mass of 19 kDa displaying high homology to bivalve C1qDC proteins. MkC1qDC demonstrated antibacterial properties against Gram-negative and Gram-positive strains. MkC1qDC binds to a number of saccharides in Ca2+-dependent manner which characterized by structural meta-similarity in acidic group enrichment of galactose and mannose derivatives incorporated in diversified molecular species of glycans. Alginate, κ-carrageenan, fucoidan, and pectin were found to be highly effective inhibitors of MkC1qDC activity. Yeast mannan, lipopolysaccharide (LPS), peptidoglycan (PGN) and mucin showed an inhibitory effect at concentrations three orders of magnitude greater than for the most effective saccharides. MkC1qDC localized to the mussel hemal system and interstitial compartment. Intriguingly, MkC1qDC was found to suppress proliferation of human adenocarcinoma HeLa cells in a dose-dependent manner, indicating to the biomedical potential of MkC1qDC protein.


Subject(s)
Membrane Glycoproteins/genetics , Mollusca , Proteins/genetics , Receptors, Complement/genetics , Receptors, Pattern Recognition/genetics , Animals , Aquatic Organisms , Humans , Membrane Glycoproteins/chemistry , Pacific Ocean , Proteins/chemistry , Receptors, Complement/chemistry , Receptors, Pattern Recognition/chemistry
2.
Immunol Lett ; 237: 42-57, 2021 09.
Article in English | MEDLINE | ID: mdl-34186155

ABSTRACT

The involvement of complement in the regulation of antibody responses has been known for long. By now several additional B cell functions - including cytokine production and antigen presentation - have also been shown to be regulated by complement proteins. Most of these important activities are mediated by receptors interacting with activation fragments of the central component of the complement system C3, such as C3b, iC3b and C3d, which are covalently attached to antigens and immune complexes. This review summarizes the role of complement receptors interacting with these ligands, namely CR1 (CD35), CR2 (CD21), CR3 (CD11b/CD18) and CR4 (CD11c/CD18) expressed by B cells in health and disease. Although we focus on human B lymphocytes, we also aim to call the attention to important differences between human and mouse systems.


Subject(s)
B-Lymphocytes/immunology , Complement C3/immunology , Receptors, Complement/immunology , Animals , Antibody Formation , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Cell Division , Gene Expression , Humans , Immunologic Memory , Ligands , Mice , Organ Specificity , Receptors, Antigen, B-Cell/immunology , Receptors, Complement/chemistry , Receptors, Complement/genetics , Species Specificity , Structure-Activity Relationship
3.
BMC Microbiol ; 20(1): 274, 2020 09 02.
Article in English | MEDLINE | ID: mdl-32878596

ABSTRACT

BACKGROUND: Clostridioides (formerly Clostridium) difficile infection is the leading cause of antibiotic-associated colitis. Studies have demonstrated that C. difficile toxin A (TcdA) can cause apoptosis of many human cell types. The purpose of this study was to investigate the relationships among exposure to TcdA, the role of the receptor for the globular heads of C1q (gC1qR) gene and the underlying intracellular apoptotic mechanism in human colonic epithelial cells (NCM 460). In this study, gC1qR expression was examined using real-time polymerase chain reaction (PCR), western blotting and immunohistochemical staining. Cell viability was assessed by the water-soluble tetrazolium salt (WST-1) assay, and cell apoptosis was assessed by flow cytometry and the terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling (TUNEL) assay. Mitochondrial function was assessed based on reactive oxygen species (ROS) generation, changes in the mitochondrial membrane potential (ΔΨm) and the content of ATP. RESULTS: Our study demonstrated that increasing the concentration of TcdA from 10 ng/ml to 20 ng/ml inhibited cell viability and induced cell apoptosis (p < 0.01). Moreover, the TcdA-induced gC1qR expression and enhanced expression of gC1qR caused mitochondrial dysfunction (including production of ROS and decreases in the ΔΨm and the content of ATP) and cell apoptosis. However, silencing of the gC1qR gene reversed TcdA-induced cell apoptosis and mitochondrial dysfunction. CONCLUSION: These data support a mechanism by which gC1qR plays a crucial role in TcdA-induced apoptosis of human colonic epithelial cells in a mitochondria-dependent manner.


Subject(s)
Apoptosis/drug effects , Bacterial Toxins/toxicity , Colon/cytology , Enterotoxins/toxicity , Membrane Glycoproteins/metabolism , Mitochondria/drug effects , Receptors, Complement/metabolism , Adenosine Triphosphate/metabolism , Apoptosis/physiology , Cell Line , Colon/pathology , Epithelial Cells , Gene Expression Regulation/drug effects , Gene Silencing , Humans , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Receptors, Complement/chemistry , Receptors, Complement/genetics
4.
Blood ; 136(14): 1685-1697, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32559765

ABSTRACT

The contact system is composed of factor XII (FXII), prekallikrein (PK), and cofactor high-molecular-weight kininogen (HK). The globular C1q receptor (gC1qR) has been shown to interact with FXII and HK. We reveal the FXII fibronectin type II domain (FnII) binds gC1qR in a Zn2+-dependent fashion and determined the complex crystal structure. FXIIFnII binds the gC1qR trimer in an asymmetric fashion, with residues Arg36 and Arg65 forming contacts with 2 distinct negatively charged pockets. gC1qR residues Asp185 and His187 coordinate a Zn2+ adjacent to the FXII-binding site, and a comparison with the ligand-free gC1qR crystal structure reveals the anionic G1-loop becomes ordered upon FXIIFnII binding. Additional conformational changes in the region of the Zn2+-binding site reveal an allosteric basis for Zn2+ modulation of FXII binding. Mutagenesis coupled with surface plasmon resonance demonstrate the gC1qR Zn2+ site contributes to FXII binding, and plasma-based assays reveal gC1qR stimulates coagulation in a FXII-dependent manner. Analysis of the binding of HK domain 5 (HKD5) to gC1qR shows only 1 high-affinity binding site per trimer. Mutagenesis studies identify a critical G3-loop located at the center of the gC1qR trimer, suggesting steric occlusion as the mechanism for HKD5 asymmetric binding. Gel filtration experiments reveal that gC1qR clusters FXII and HK into a higher-order 500-kDa ternary complex. These results support the conclusion that extracellular gC1qR can act as a chaperone to cluster contact factors, which may be a prelude for initiating the cascades that drive bradykinin generation and the intrinsic pathway of coagulation.


Subject(s)
Allosteric Site , Binding Sites , Carrier Proteins/chemistry , Factor XII/chemistry , Kininogens/chemistry , Membrane Glycoproteins/chemistry , Mitochondrial Proteins/chemistry , Models, Molecular , Receptors, Complement/chemistry , Aged , Carrier Proteins/metabolism , Factor XII/metabolism , Female , Humans , Kinetics , Kininogens/metabolism , Ligands , Membrane Glycoproteins/metabolism , Mitochondrial Proteins/metabolism , Models, Biological , Molecular Dynamics Simulation , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Protein Binding , Protein Conformation , Receptors, Complement/metabolism , Recombinant Proteins , Structure-Activity Relationship , Zinc/chemistry , Zinc/metabolism
5.
Trends Biochem Sci ; 45(8): 693-705, 2020 08.
Article in English | MEDLINE | ID: mdl-32402749

ABSTRACT

Complement factor C5a is an integral constituent of the complement cascade critically involved in the innate immune response, and it exerts its functions via two distinct receptors, C5aR1 and C5aR2. While C5aR1 is a prototypical G-protein-coupled receptor (GPCR), C5aR2 lacks functional coupling to heterotrimeric G proteins, although both receptors efficiently recruit ß arrestins (ßarrs). Here, we discuss the recent studies providing direct structural details of ligand-receptor interactions, and a framework of functional bias in this system, including the differences in terms of structural motifs and transducer coupling. We also discuss the functional analogy of C5aR2 with the atypical chemokine receptors (ACKRs), and highlight the future directions to elucidate the mechanistic basis of the functional divergence of these receptors activated by a common natural agonist.


Subject(s)
Complement C5a/metabolism , Receptors, Complement/chemistry , Receptors, Complement/metabolism , Animals , Humans , Structure-Activity Relationship
6.
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
7.
Fish Shellfish Immunol ; 82: 378-385, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30144564

ABSTRACT

The gC1qR is a ubiquitously expressed cell protein that interacts with the globular heads of C1q (gC1q) and many other ligands. In this study, one gC1qR homolog gene was obtained from Exopalaemon carinicauda and named EcgC1qR. The complete nucleotide sequence of EcgC1qR contained a 774 bp open reading frame (ORF) encoding EcgC1qR precursor of 257 amino acids. The deduced amino acid sequence of EcgC1qR revealed a 55-amino-acid-long mitochondrial targeting sequence at the N-terminal and a mitochondrial acidic matrix protein of 33 kDa (MAM33) domain. The genomic organization of EcgC1qR gene showed that EcgC1qR gene contained five exons and four introns. EcgC1qR could express in all of the detected tissues and its expression was much higher in hepatopancreas and hemocytes. The expression of EcgC1qR in the hepatopancreas of prawns challenged with Vibrio parahaemolyticus and Aeromonas hydrophila changed in a time-dependent manner. The expression of EcgC1qR in prawns challenged with V. parahaemolyticus was up-regulated at 6 h (p < 0.05), and significantly up-regulated at 12 h and 24 h (p < 0.01), and then returned to the control levels at 48 h post-challenge (p > 0.05). At the same time, the expression in Aeromonas-challenged group was significantly up-regulated at 6, 12 and 24 h. The recombinant EcgC1qR could inhibit the growth of two tested bacteria. In addition, we successfully deleted EcgC1qR gene through CRISPR/Cas9 technology and it was the first time to obtain the mutant of gC1qR homolog gene in crustacean. It's a great progress to study the biological function of gC1qR in crustacean in future.


Subject(s)
Gene Expression Regulation/immunology , Immunity, Innate/genetics , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Palaemonidae/genetics , Palaemonidae/immunology , Receptors, Complement/genetics , Receptors, Complement/immunology , Aeromonas hydrophila/physiology , Amino Acid Sequence , Animals , Arthropod Proteins/chemistry , Arthropod Proteins/genetics , Arthropod Proteins/immunology , Base Sequence , Gene Expression Profiling , Membrane Glycoproteins/chemistry , Phylogeny , Receptors, Complement/chemistry , Sequence Alignment , Vibrio parahaemolyticus/physiology
8.
Matrix Biol ; 64: 112-127, 2017 12.
Article in English | MEDLINE | ID: mdl-28912033

ABSTRACT

The glycoprotein CD93 has recently been recognized to play an important role in the regulation of the angiogenic process. Moreover, CD93 is highly expressed in the endothelial cells of tumor blood vessel and faintly expressed in the non-proliferating endothelium. Much evidence suggests that CD93 mediates adhesion in the endothelium. Here we identify Multimerin 2 (MMRN2), a pan-endothelial extracellular matrix protein, as a specific ligand for CD93. We found that CD93 and MMRN2 are co-expressed in the blood vessels of various human tumors. Moreover, disruption of the CD93-MMRN2 interaction reduced endothelial cell adhesion and migration, making the interaction of CD93 with MMRN2 an ideal target to block pathological angiogenesis. Model structures and docking studies served to envisage the region of CD93 and MMRN2 involved in the interaction. Site-directed mutagenesis identified different residue hotspots either directly or indirectly involved in the binding. We propose a molecular model in which the coiled-coil domain of MMRN2 is engaged by F238 of CD93. Altogether, these studies identify the key interaction surfaces of the CD93-MMRN2 complex and provide a framework for exploring how to inhibit angiogenesis by hindering the CD93-MMRN2 interaction.


Subject(s)
Antigens, Surface/metabolism , Endothelium, Vascular/cytology , Membrane Glycoproteins/metabolism , Neoplasms/blood supply , Receptors, Complement/metabolism , Antigens, Surface/chemistry , Antigens, Surface/genetics , Binding Sites , Cell Adhesion , Cell Line, Tumor , Cell Movement , Endothelium, Vascular/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Models, Molecular , Molecular Docking Simulation , Mutagenesis, Site-Directed , Neoplasms/metabolism , Protein Binding , Receptors, Complement/chemistry , Receptors, Complement/genetics
9.
Nat Commun ; 8(1): 351, 2017 08 24.
Article in English | MEDLINE | ID: mdl-28839129

ABSTRACT

Complement C3a is an important protein in innate and adaptive immunity, but its specific roles in vivo remain uncertain because C3a degrades rapidly to form the C3a-desArg protein, which does not bind to the C3a receptor and is indistinguishable from C3a using antibodies. Here we develop the most potent, stable and highly selective small molecule modulators of C3a receptor, using a heterocyclic hinge to switch between agonist and antagonist ligand conformations. This enables characterization of C3 areceptor-selective pro- vs. anti-inflammatory actions in human mast cells and macrophages, and in rats. A C3a receptor-selective agonist induces acute rat paw inflammation by first degranulating mast cells before activating macrophages and neutrophils. An orally administered C3a receptor-selective antagonist inhibits mast cell degranulation, thereby blocking recruitment and activation of macrophages and neutrophils, expression of inflammatory mediators and inflammation in a rat paw edema model. These novel tools reveal the mechanism of C3a-induced inflammation and provide new insights to complement-based medicines.Complement C3a is an important protein in innate and adaptive immunity, but its roles in vivo are unclear. Here the authors develop novel chemical agonists and antagonists for the C3a receptor, and show that they modulate mast cell degranulation and inflammation in a rat paw edema model.


Subject(s)
Complement C3a/physiology , Immunity, Innate/genetics , Receptors, Complement/chemistry , Animals , Anti-Asthmatic Agents/pharmacology , Cell Degranulation/drug effects , Cells, Cultured , Complement C3a/genetics , Complement C3a/metabolism , Cromolyn Sodium/pharmacology , Humans , Ligands , Macrophages/immunology , Male , Mast Cells/immunology , Neutrophils/immunology , Protein Conformation , Rats , Rats, Wistar , Receptors, Complement/agonists , Receptors, Complement/antagonists & inhibitors
10.
J Immunol Methods ; 439: 67-73, 2016 12.
Article in English | MEDLINE | ID: mdl-27742562

ABSTRACT

CD93 belongs to the group XIV C-type lectin like domain (CTLD) and is closely related to thrombomodulin (CD141). Although CD93 is known to be involved in the regulation of cell adhesion and phagocytosis, its role in innate immunity remains to be fully investigated. Critically, published data about CD141 suggest that CD93 CTLD could be involved in the control of inflammation. In order to address further functional and structural analyses, we expressed human CD93 CTLD with several disulfide bonds in an E. coli expression system. As the E. coli cytoplasm is a reducing compartment, production of disulfide-bond proteins remains a challenge. Hence, we decided to over express CD93 CTLD in commercially available strains of E. coli and co-expressed a sulfhydryl oxidase (Erv1p) and a disulfide isomerase (DsbC). This strategy led to high yield expression of a native form of CD93 CTLD. NMR studies revealed that Ca2+ was not able to bind to CD93 CTLD. We also showed that the recombinant protein could alter LPS pro-inflammatory activity on THP1. This work provides new tool for further functional and structural studies to decipher the functions associated to the CTLD of CD93. This approach may also be used for others members of the group XIV C-type lectin like domain (CD141, CD248 and CLec14A).


Subject(s)
Cloning, Molecular/methods , Cytoplasm/metabolism , Disulfides/metabolism , Escherichia coli/metabolism , Membrane Glycoproteins/biosynthesis , Receptors, Complement/biosynthesis , Binding Sites , Calcium/metabolism , Cell Line , Disulfides/chemistry , Escherichia coli/genetics , Escherichia coli Proteins/biosynthesis , Escherichia coli Proteins/genetics , Gene Expression Regulation, Bacterial , Humans , Inflammation/immunology , Inflammation/metabolism , Lipopolysaccharides/pharmacology , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Membrane Glycoproteins/pharmacology , Monocytes/drug effects , Monocytes/immunology , Monocytes/metabolism , Nuclear Magnetic Resonance, Biomolecular , Oxidoreductases/biosynthesis , Oxidoreductases/genetics , Protein Binding , Protein Disulfide-Isomerases/biosynthesis , Protein Disulfide-Isomerases/genetics , Protein Domains , Receptors, Complement/chemistry , Receptors, Complement/genetics , Recombinant Proteins/biosynthesis , Structure-Activity Relationship , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
11.
EBioMedicine ; 9: 207-216, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27333049

ABSTRACT

Plasmodium falciparum malaria is a deadly pathogen. The invasion of red blood cells (RBCs) by merozoites is a target for vaccine development. Although anti-merozoite antibodies can block invasion in vitro, there is no efficacy in vivo. To explain this discrepancy we hypothesized that complement activation could enhance RBC invasion by binding to the complement receptor 1 (CR1). Here we show that a monoclonal antibody directed against the merozoite and human polyclonal IgG from merozoite vaccine recipients enhanced RBC invasion in a complement-dependent manner and that soluble CR1 inhibited this enhancement. Sialic acid-independent strains, that presumably are able to bind to CR1 via a native ligand, showed less complement-dependent enhancement of RBC invasion than sialic acid-dependent strains that do not utilize native CR1 ligands. Confocal fluorescent microscopy revealed that complement-dependent invasion resulted in aggregation of CR1 at the RBC surface in contact with the merozoite. Finally, total anti-P. berghei IgG enhanced parasite growth and C3 deficiency decreased parasite growth in mice. These results demonstrate, contrary to current views, that complement activation in conjunction with antibodies can paradoxically aid parasites invade RBCs and should be considered in future design and testing of merozoite vaccines.


Subject(s)
Antibodies, Monoclonal/immunology , Immunoglobulin G/immunology , Plasmodium falciparum/pathogenicity , Receptors, Complement/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Cell Adhesion/drug effects , Cells, Cultured , Erythrocytes/cytology , Erythrocytes/metabolism , Erythrocytes/parasitology , Flow Cytometry , Humans , Malaria/parasitology , Merozoites/drug effects , Merozoites/immunology , Merozoites/physiology , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Parasitemia/parasitology , Parasitemia/pathology , Parasitemia/veterinary , Plasmodium falciparum/growth & development , Plasmodium falciparum/metabolism , Protozoan Proteins/immunology , Protozoan Proteins/metabolism , Protozoan Vaccines/immunology , Receptor Aggregation , Receptors, Complement/chemistry
12.
Interdiscip Sci ; 8(1): 35-40, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26264056

ABSTRACT

There are so many obstacles in developing a vaccine or vaccine technology for diseases like cancer and human immunodeficiency virus infection. While developing vaccines that target specific infection, molecular adjuvants are indispensable. These molecular adjuvants act as a vaccine delivery vehicle to the immune system to increase the effectiveness of the specific antigens. In the present work, a computational study has been done on molecular adjuvants like IgGFc, GMCSF and C3d to find out how efficiently they are binding to CR1. Sequence, structure and mutational analysis are performed on the molecular adjuvants to understand the features important for their binding with the receptor. Results obtained from our study indicate that the adjuvant IgGFc complexed with the receptor CR1 has the best binding efficiency, which can be used further to develop better vaccine technologies.


Subject(s)
Adjuvants, Immunologic/metabolism , Receptors, Complement/metabolism , Adjuvants, Immunologic/chemistry , Amino Acid Motifs , Humans , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Immunoglobulin G/chemistry , Models, Molecular , Molecular Docking Simulation , Protein Structure, Tertiary , Receptors, Complement/chemistry
13.
J Transl Med ; 12: 286, 2014 Oct 05.
Article in English | MEDLINE | ID: mdl-25288439

ABSTRACT

BACKGROUND: Human papillomavirus type-16 (HPV-16) E2 protein acts as a transcriptional modulator and plays a key role in regulating many biological responses. The purpose of this study was to investigate the relationship between HPV-16 E2, the receptor for the globular heads of human C1q (gC1qR) gene expression, mitochondrial dysfunction and apoptosis regulation in human cervical squamous carcinoma cells (C33a and SiHa). METHODS: HPV-16 E2 and gC1qR expression was examined using real-time PCR and western blot analysis. Apoptosis in C33a and SiHa cells was assessed by flow cytometry. Mitochondrial function was detected via ROS generation, the amount of cytosolic Ca2+, and changes in the mitochondrial membrane potential (Δψm). RESULTS: The expression of the HPV-16 E2 and gC1qR gene significantly decreased in human cervical squamous carcinoma samples relative to the non-cancerous cervix samples. C33a and SiHa cells that were transfected with a vector encoding HPV-16 E2 displayed significantly increased gC1qR gene expression and mitochondrial dysfunction as well as an up-regulation of cellular apoptosis, which was abrogated by the addition of gC1qR small-interfering RNA (siRNA). CONCLUSIONS: These data support a mechanism whereby gC1qR plays an important role in HPV-16 E2-induced human cervical squamous carcinoma cell apoptosis via a mitochondria-dependent pathway.


Subject(s)
Apoptosis , Carcinoma, Squamous Cell/pathology , DNA-Binding Proteins/metabolism , Human papillomavirus 16/physiology , Membrane Glycoproteins/metabolism , Mitochondria/pathology , Oncogene Proteins, Viral/metabolism , Receptors, Complement/metabolism , Uterine Cervical Neoplasms/pathology , Adult , Carcinoma, Squamous Cell/virology , Cell Line, Tumor , Female , Humans , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Middle Aged , Mitochondria/metabolism , Protein Structure, Tertiary , Receptors, Complement/chemistry , Receptors, Complement/genetics , Signal Transduction , Structure-Activity Relationship , Uterine Cervical Neoplasms/virology , Young Adult
14.
J Nucl Med ; 55(5): 824-9, 2014 May.
Article in English | MEDLINE | ID: mdl-24686780

ABSTRACT

UNLABELLED: An accurate and noninvasive tracer able to detect molecular events underlying the development of rheumatoid arthritis (RA) would be useful for RA diagnosis and drug efficacy assessment. A complement receptor of the Ig superfamily (CRIg) is expressed on synovial macrophages of RA patients, making it an interesting target for molecular imaging of RA. We aim to develop a radiotracer for the visualization of CRIg in a mouse model for RA using radiolabeled single-domain variable antibody VHH fragments (Nanobodies). METHODS: Quantitative polymerase chain reaction was used to locate CRIg expression in mice with collagen-induced arthritis (CIA). A Nanobody, NbV4m119, was generated to specifically target CRIg. Flow cytometry, phosphorimaging, and confocal microscopy were used to confirm NbVm119 binding to CRIg-positive cells. SPECT (SPECT/CT) was used to image arthritic lesions in the inflamed paws of 29 mice using (99m)Tc-NbV4m119 Nanobody. RESULTS: CRIg is constitutively expressed in the liver and was found to be upregulated in synovial tissues of CIA mice. SPECT/CT imaging revealed that (99m)Tc-NbV4m119 specifically targeted CRIg-positive liver macrophages in naïve wild-type but not in CRIg(-/-) (CRIg knockout) mice. In CIA mice, (99m)Tc-NbV4m119 accumulation in arthritic lesions increased according to the severity of the inflammation. In the knees of mice with CIA, (99m)Tc-NbV4m119 was found to accumulate even before the onset of macroscopic clinical symptoms. CONCLUSION: SPECT/CT imaging with (99m)Tc-NbV4m119 visualizes joint inflammation in CIA. Furthermore, imaging could predict which mice will develop clinical symptoms during CIA. Consequently, imaging of joint inflammation with CRIg-specific Nanobodies offers perspectives for clinical applications in RA patients.


Subject(s)
Arthritis, Rheumatoid/diagnosis , Arthritis, Rheumatoid/immunology , Macrophages/metabolism , Molecular Imaging/methods , Receptors, Complement/chemistry , Single-Domain Antibodies/chemistry , Animals , Arthritis, Experimental/immunology , Arthritis, Rheumatoid/metabolism , Disease Models, Animal , Immunohistochemistry , Inflammation , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , RNA, Messenger/metabolism , Radiopharmaceuticals , Real-Time Polymerase Chain Reaction , Synovial Membrane/metabolism , Technetium/chemistry , Tomography, Emission-Computed, Single-Photon , X-Ray Microtomography
15.
Methods Mol Biol ; 1100: 319-27, 2014.
Article in English | MEDLINE | ID: mdl-24218271

ABSTRACT

The recognition subunit of C1, C1q, has emerged as an important player in various pathophysiologic conditions largely in part due to its ability to interact with pathogen-associated or cell surface expressed ligands and receptors. Identification and purification of these molecules is therefore of paramount importance if we are to procure valuable information with regards to the structure, function, and cell surface distribution. Since the interaction of C1q is better served when the receptors are purified from homologous species, we discuss here a simple guideline for the purification and characterization of the two C1q receptors, cC1qR (calreticulin) and gC1qR, from human cell lines.


Subject(s)
Membrane Glycoproteins/isolation & purification , Membrane Glycoproteins/metabolism , Receptors, Complement/isolation & purification , Receptors, Complement/metabolism , Biotinylation , Cell Culture Techniques , Cell Membrane/chemistry , Cell Membrane/metabolism , Chromatography, Liquid/methods , Humans , Membrane Glycoproteins/chemistry , Receptors, Complement/chemistry , U937 Cells
16.
Neurol Res ; 35(9): 976-83, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23815859

ABSTRACT

OBJECTIVE: Complement receptor type 1 (CR1), one of the most potent inhibitors in complement activation, shows a protective effect on cerebral ischemia/reperfusion (CI/R) injury due to its ability to bind C3b and C4b and to inactivate C3/C5 convertases. So far, no study assessed the effect of the first three short consensus repeats (SCR1-3) with low molecular weight, one of the most active functional domains of CR1, binding C4b with a powerful decay-acceleration effect on classical and alternative C3/C5 convertases pathways. Therefore, we aim to assess this effect on CI/R injury in the present study. METHODS: Seventy-five adult male Sprague-Dawley rats were randomly divided into three groups: sham operation group (n = 15), CI/R group (n = 30), and CI/R group treated with CR1-SCR1-3 protein (n = 30). After middle cerebral artery occlusion (MCAO) for 1 hour and reperfusion for 24 hours, neurological motor deficits, cerebral infarct size, and biochemical parameters including myeloperoxidase (MPO), malondialdehyde (MDA), and superoxide dismutase (SOD) were assessed. Meanwhile, tissues in cerebral cortex were collected and processed for western blotting, immunohistochemistry, and HE staining. RESULTS: CR1-SCR1-3 could improve neurological functions in brain with a 26.8% decrease in neurological motor deficit score and could lead to a 63.8% reduction in cerebral infarct size. Besides, pretreatment using CR1-SCR1-3 could prevent neutrophil infiltration and alleviate inflammation severity and subsequent tissue damage. Decreased C4b expression and action, as well as improved morphological changes, were also observed in cerebral tissues of CI/R+CR1-SCR1-3 rats. CONCLUSION: CR1-SCR1-3 protein could possess a neuroprotective effect on acute CI/R injury.


Subject(s)
Infarction, Middle Cerebral Artery/drug therapy , Neuroprotective Agents/therapeutic use , Receptors, Complement/chemistry , Receptors, Complement/therapeutic use , Reperfusion Injury/drug therapy , Animals , Blotting, Western , Brain/drug effects , Brain/metabolism , Brain/pathology , Complement C4b/metabolism , Consensus Sequence , Immunohistochemistry , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/pathology , Male , Movement Disorders/drug therapy , Movement Disorders/metabolism , Movement Disorders/pathology , Neuroprotective Agents/chemistry , Neuroprotective Agents/metabolism , Neutrophils/drug effects , Neutrophils/physiology , Protein Interaction Domains and Motifs , Random Allocation , Rats , Rats, Sprague-Dawley , Receptors, Complement/isolation & purification , Receptors, Complement/metabolism , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Severity of Illness Index
17.
J Immunol ; 190(7): 3721-31, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23460739

ABSTRACT

Complement receptor type 1 (CR1) is a membrane receptor expressed on a wide range of cells. It is involved in immune complex clearance, phagocytosis, and complement regulation. Its ectodomain is composed of 30 complement control protein (CCP) modules, organized into four long homologous repeats (A-D). In addition to its main ligands C3b and C4b, CR1 was reported to interact with C1q and mannan-binding lectin (MBL) likely through its C-terminal region (CCP22-30). To decipher the interaction of human CR1 with the recognition proteins of the lectin complement pathway, a recombinant fragment encompassing CCP22-30 was expressed in eukaryotic cells, and its interaction with human MBL and ficolins was investigated using surface plasmon resonance spectroscopy. MBL and L-ficolin were shown to interact with immobilized soluble CR1 and CR1 CCP22-30 with apparent dissociation constants in the nanomolar range, indicative of high affinity. The binding site for CR1 was located at or near the MBL-associated serine protease (MASP) binding site in the collagen stalks of MBL and L-ficolin, as shown by competition experiments with MASP-3. Accordingly, the mutation of an MBL conserved lysine residue essential for MASP binding (K55) abolished binding to soluble CR1 and CCP22-30. The CR1 binding site for MBL/ficolins was mapped to CCP24-25 of long homologous repeat D using deletion mutants. In conclusion, we show that ficolins are new CR1 ligands and propose that MBL/L-ficolin binding involves major ionic interactions between conserved lysine residues of their collagen stalks and surface exposed acidic residues located in CR1 CCP24 and/or CCP25.


Subject(s)
Complement Pathway, Mannose-Binding Lectin , Mannose-Binding Lectin/metabolism , Receptors, Complement/metabolism , Binding Sites , Carrier Proteins/metabolism , Complement Pathway, Mannose-Binding Lectin/genetics , Humans , Kinetics , Lectins/metabolism , Mannose-Binding Protein-Associated Serine Proteases/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Protein Interaction Mapping , Receptors, Complement/chemistry , Receptors, Complement/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Ficolins
18.
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
19.
J Biomol Struct Dyn ; 31(2): 215-23, 2013.
Article in English | MEDLINE | ID: mdl-22831459

ABSTRACT

C5aR antagonists have been thought as potential immune mediators in various inflammatory and autoimmune diseases, and discovery of C5aR antagonists has attracted much attention in recent years. The discovery of C5aR antagonists was usually achieved through high-throughput screening, which usually suffered a high cost and a low success rate. Currently, the fast developing computer-aided virtual screening (VS) methods provide economic and rapid approaches to the lead discovery. In this account, we proposed a hybrid ligand-based VS protocol that is based on support vector machine (SVM) classification and pharmacophore models for retrieving novel C5aR antagonists. Performance evaluation of this hybrid VS protocol in virtual screening against a large independent test set, T-CHEM, showed that the hybrid VS approach significantly increased the hit rate and enrichment factor compared with the individual SVM classification model-based VS and pharmacophore model-based VS, as well as molecular docking-based VS in that the receptor structure was created by homology modeling. The hybrid VS approach was then used to screen several large chemical libraries including PubChem, Specs, and Enamine. Finally, a total of 20 compounds were selected from the top ranking hits, and shifted to the subsequent in vitro and in vivo studies, which results will be reported in the near future.


Subject(s)
Complement Inactivating Agents/chemistry , Molecular Docking Simulation , Receptors, Complement/antagonists & inhibitors , Drug Evaluation, Preclinical/methods , Humans , Inhibitory Concentration 50 , Ligands , Models, Chemical , Receptor, Anaphylatoxin C5a , Receptors, Complement/chemistry , Small Molecule Libraries , Structural Homology, Protein , Support Vector Machine
20.
J Transl Med ; 10: 255, 2012 Dec 26.
Article in English | MEDLINE | ID: mdl-23268996

ABSTRACT

BACKGROUND: The globular heads of the human C1q receptor (gC1qR) localize predominantly to the mitochondrial matrix. gC1qR mediates many biological responses, including growth perturbation, morphological abnormalities and the initiation of apoptosis. The purpose of this study was to investigate the relationship between mitochondrial dysfunction, p53 status and gC1qR expression and the regulation of apoptosis in human cervical squamous carcinoma cells (C33a and SiHa). METHODS: Here, gC1qR expression was examined in human cervical tissues using real-time PCR and Western blot analysis. Apoptotic death of C33a and SiHa cells was assessed by flow cytometric analysis that detected the subG1 population. Mitochondrial function was assessed via ROS generation, the content of cytosolic Ca2+, and the change in mitochondrial membrane potential (Δψm). The viability and migration of C33a and SiHa cells were detected via the water-soluble tetrazolium salt (WST-1) assay and the transwell assay, respectively. RESULTS: gC1qR expression was decreased in cervical squamous cell carcinoma tissues compared with normal tissues. C33a and SiHa cells transfected with a vector encoding gC1qR displayed mitochondrial dysfunction and apoptosis, which was abrogated by the addition of a mutant form of p53 or p53 small interference RNA (siRNA). Furthermore, upon overexpression of gC1qR, cell viability and migration were significantly enhanced, and the apoptosis of C33a and SiHa cells were decreased when cells were treated with mutant p53 or p53 siRNA. CONCLUSIONS: These data support a mechanism whereby gC1qR induces apoptosis through the mitochondrial and p53-dependent pathways in cervical squamous cell carcinoma.


Subject(s)
Apoptosis , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/metabolism , Receptors, Complement/chemistry , Receptors, Complement/metabolism , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Uterine Cervical Neoplasms/pathology , Apoptosis/genetics , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Movement , Cell Survival , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Membrane Glycoproteins/genetics , Mitochondria/metabolism , Protein Structure, Tertiary , Receptors, Complement/genetics , Signal Transduction/genetics , Structure-Activity Relationship , Uterine Cervical Neoplasms/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...