Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Viruses ; 16(1)2024 01 22.
Article in English | MEDLINE | ID: mdl-38275973

ABSTRACT

(1) Background: Avian influenza has attracted widespread attention because of its severe effect on the poultry industry and potential threat to human health. The H9N2 subtype of avian influenza viruses was the most prevalent in chickens, and there are several commercial vaccines available for the prevention of the H9N2 subtype of avian influenza viruses. However, due to the prompt antigenic drift and antigenic shift of influenza viruses, outbreaks of H9N2 viruses still continuously occur, so surveillance and vaccine updates for H9N2 subtype avian influenza viruses are particularly important. (2) Methods: In this study, we constructed a stable Chinese hamster ovary cell line (CHO) to express the H9 hemagglutinin (HA) protein of the major prevalent H9N2 strain A/chicken/Daye/DY0602/2017 with genetic engineering technology, and then a subunit H9 avian influenza vaccine was prepared using the purified HA protein with a water-in-oil adjuvant. (3) Results: The results showed that the HI antibodies significantly increased after vaccination with the H9 subunit vaccine in specific-pathogen-free (SPF) chickens with a dose-dependent potency of the immunized HA protein, and the 50 µg or more per dose HA protein could provide complete protection against the H9N2 virus challenge. (4) Conclusions: These results indicate that the CHO expression system could be a platform used to develop the subunit vaccine against H9 influenza viruses in chickens.


Subject(s)
Influenza A Virus, H9N2 Subtype , Influenza Vaccines , Influenza in Birds , Animals , Humans , Cricetinae , Influenza A Virus, H9N2 Subtype/genetics , Chickens , Hemagglutinins , Cricetulus , CHO Cells , Antibodies, Viral , Vaccines, Subunit , Hemagglutinin Glycoproteins, Influenza Virus/genetics
2.
Sheng Wu Gong Cheng Xue Bao ; 31(11): 1623-31, 2015 Nov.
Article in Chinese | MEDLINE | ID: mdl-26939445

ABSTRACT

Staphylococcus aureus is a major cause of hospital-acquired infection. Because the bacteria are very easy to become resistant to antibiotics, vaccination is a main method against S. aureus infection. Clumping factor B (ClfB) is an adhesion molecule essential for S. aureus to colonize in the host mucosa and is regarded as an important target antigen. In this study, we successfully used Escherichia coli to express a segment encoding the N1-N3 regions of ClfB protein (Truncated-ClfB) cloned from S. aureus. The protein was purified by affinity and ion exchange chromatographies and gel filtration. Rabbits were immunized three times with purified Truncated-ClfB. After that, blood was collected to prepare serum which were then used for measurement of antibody level. Phagocytosis of S. aureus opsonized by the serum was determined by a flow cytometry. Results show that the serum IgG titer reached 1:640 000. Phagocytosed S. aureus by polymorphonuclear leukocytes were significantly more when the bacteria were opsonized by the serum from Truncated-ClfB immunized rabbits than those from no immunized group (P < 0.01). Therefore, the results indicated that Truncated-ClfB could be a promising vaccine candidate against S. aureus infection.


Subject(s)
Adhesins, Bacterial/immunology , Immune Sera , Opsonin Proteins/immunology , Staphylococcal Infections/immunology , Animals , Antibodies, Bacterial/blood , Escherichia coli , Flow Cytometry , Immunoglobulin G/blood , Phagocytosis , Rabbits , Staphylococcus aureus
3.
Neoplasia ; 14(11): 994-1004, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23226093

ABSTRACT

Complement activation plays a critical role in controlling inflammatory responses. To assess the role of complement during ovarian cancer progression, we crossed two strains of mice with genetic complement deficiencies with transgenic mice that develop epithelial ovarian cancer (TgMISIIR-TAg). TgMISIIR-TAg mice fully or partially deficient for complement factor 3 (C3) (Tg(+)C3(KO) and Tg(+)C3(HET), respectively) or fully deficient for complement factor C5a receptor (C5aR) (Tg(+)C5aR(KO)) develop either no ovarian tumors or tumors that were small and poorly vascularized compared to wild-type littermates (Tg(+)C3(WT), Tg(+)C5aR(WT)). The percentage of tumor infiltrating immune cells in Tg(+)C3(HET) tumors compared to Tg(+)C3(WT) controls was either similar (macrophages, B cells, myeloid-derived suppressor cells), elevated (effector T cells), or decreased (regulatory T cells). Regardless of these ratios, cytokine production by immune cells taken from Tg(+)C3(HET) tumors was reduced on stimulation compared to Tg(+)C3(WT) controls. Interestingly, CD31(+) endothelial cell (EC) function in angiogenesis was significantly impaired in both C3(KO) and C5aR(KO) mice. Further, using the C5aR antagonist PMX53, tube formation of ECs was shown to be C5a-dependent, possibly through interactions with the VEGF(165) but not VEGF(121) isoform. Finally, the mouse VEGF(164) transcript was underexpressed in C3(KO) livers compare to C3(WT) livers. Thus, we conclude that complement inhibition blocks tumor outgrowth by altering EC function and VEGF(165) expression.


Subject(s)
Complement System Proteins/genetics , Endothelial Cells/immunology , Endothelial Cells/metabolism , Neovascularization, Pathologic/immunology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/immunology , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/immunology , Complement Activation/immunology , Complement C3/deficiency , Complement C5a/antagonists & inhibitors , Complement C5a/immunology , Complement System Proteins/drug effects , Endothelial Cells/drug effects , Female , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Male , Mice , Mice, Transgenic , Ovarian Neoplasms/blood supply , Peptides, Cyclic/pharmacology , Protein Isoforms , Vascular Endothelial Growth Factor A/metabolism
4.
J Immunol ; 186(11): 6568-75, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21531894

ABSTRACT

Neonatal respiratory distress syndrome can progress to bronchopulmonary dysplasia (BPD), a serious pulmonary fibrotic disorder. Given the involvement of the extrinsic coagulation cascade in animal models of lung fibrosis, we examined its role in BPD. We observed a higher number of neutrophils expressing tissue factor (TF) in bronchoalveolar lavage fluid (BALF) from infants with BPD than from those with uncomplicated respiratory distress syndrome together with a parallel decrease in TF and connective tissue growth factor (CTGF) in BALF supernatants during the disease course. The involvement of coagulation in the fibrotic process associated with BPD was further evaluated by treating primary human colonic myofibroblasts with BALF supernatants from infants with BPD. These human colonic myofibroblasts demonstrated an enhanced C5a- and thrombin-dependent migration. Moreover, they expressed TF in an endothelin-1-dependent manner, with subsequent activation of the extrinsic coagulation cascade and CTGF production mediated by protease-activator receptor-1 signaling. These data provide a novel mechanism for the development of BPD and indicate that endothelin-1 signaling contributes to fibrosis by upregulating a TF/thrombin amplification loop responsible for CTGF production, and offer novel and specific therapeutic targets for pulmonary fibrotic disease.


Subject(s)
Bronchopulmonary Dysplasia/metabolism , Endothelin-1/metabolism , Respiratory Distress Syndrome, Newborn/metabolism , Signal Transduction , Blotting, Western , Bronchoalveolar Lavage Fluid/chemistry , Bronchopulmonary Dysplasia/genetics , Bronchopulmonary Dysplasia/pathology , Cells, Cultured , Colon/metabolism , Colon/pathology , Complement C5a/genetics , Complement C5a/metabolism , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/metabolism , Endothelin-1/genetics , Female , Fibrosis , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Immunohistochemistry , Infant, Newborn , Lung/metabolism , Lung/pathology , Male , Microscopy, Fluorescence , Myofibroblasts/metabolism , Myofibroblasts/pathology , Receptor, Anaphylatoxin C5a , Receptor, PAR-1/genetics , Receptor, PAR-1/metabolism , Receptors, Complement/genetics , Receptors, Complement/metabolism , Respiratory Distress Syndrome, Newborn/genetics , Respiratory Distress Syndrome, Newborn/pathology , Reverse Transcriptase Polymerase Chain Reaction , Thrombin/genetics , Thrombin/metabolism , Thromboplastin/genetics , Thromboplastin/metabolism
5.
J Immunol ; 186(7): 4269-77, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21339361

ABSTRACT

Exposure of nonself surfaces such as those of biomaterials or transplanted cells and organs to host blood frequently triggers innate immune responses, thereby affecting both their functionality and tolerability. Activation of the alternative pathway of complement plays a decisive role in this unfavorable reaction. Whereas previous studies demonstrated that immobilization of physiological regulators of complement activation (RCA) can attenuate this foreign body-induced activation, simple and efficient approaches for coating artificial surfaces with intact RCA are still missing. The conjugation of small molecular entities that capture RCA with high affinity is an intriguing alternative, as this creates a surface with autoregulatory activity upon exposure to blood. We therefore screened two variable cysteine-constrained phage-displayed peptide libraries for factor H-binding peptides. We discovered three peptide classes that differed with respect to their main target binding areas. Peptides binding to the broad middle region of factor H (domains 5-18) were of particular interest, as they do not interfere with either regulatory or binding activities. One peptide in this group (5C6) was further characterized and showed high factor H-capturing activity while retaining its functional integrity. Most importantly, when 5C6 was coated to a model polystyrene surface and exposed to human lepirudin-anticoagulated plasma, the bound peptide captured factor H and substantially inhibited complement activation by the alternative pathway. Our study therefore provides a promising and novel approach to produce therapeutic materials with enhanced biocompatibility.


Subject(s)
Complement Pathway, Alternative/immunology , Peptide Fragments/metabolism , Peptide Fragments/therapeutic use , Biocompatible Materials/metabolism , Cloning, Molecular , Complement C3b/antagonists & inhibitors , Complement C3b/metabolism , Complement Factor H/metabolism , Complement Factor H/therapeutic use , Complement Factor I/antagonists & inhibitors , Complement Factor I/metabolism , Complement Inactivator Proteins/metabolism , Hemolysis , Humans , Peptide Library , Protein Binding/immunology , Surface Properties
6.
Mol Immunol ; 48(4): 481-9, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21067811

ABSTRACT

Compstatin is a 13-residue disulfide-bridged peptide that inhibits a key step in the activation of the human complement system. Compstatin and its derivatives have shown great promise for the treatment of many clinical disorders associated with unbalanced complement activity. To obtain more potent compstatin analogues, we have now performed an N-methylation scan of the peptide backbone and amino acid substitutions at position 13. One analogue (Ac-I[CVW(Me)QDW-Sar-AHRC](NMe)I-NH(2)) displayed a 1000-fold increase in both potency (IC(50) = 62 nM) and binding affinity for C3b (K(D) = 2.3 nM) over that of the original compstatin. Biophysical analysis using surface plasmon resonance and isothermal titration calorimetry suggests that the improved binding originates from more favorable free conformation and stronger hydrophobic interactions. This study provides a series of significantly improved drug leads for therapeutic applications in complement-related diseases, and offers new insights into the structure-activity relationships of compstatin analogues.


Subject(s)
Complement Inactivator Proteins/metabolism , Peptides, Cyclic/metabolism , Amino Acid Sequence , Complement Activation/drug effects , Complement Inactivator Proteins/chemistry , Complement Inactivator Proteins/pharmacology , Humans , Kinetics , Molecular Sequence Data , Peptides, Cyclic/chemistry , Protein Binding/drug effects , Thermodynamics
7.
Proc Natl Acad Sci U S A ; 107(41): 17621-6, 2010 Oct 12.
Article in English | MEDLINE | ID: mdl-20876141

ABSTRACT

The complement system is a major target of immune evasion by Staphylococcus aureus. Although many evasion proteins have been described, little is known about their molecular mechanisms of action. Here we demonstrate that the extracellular fibrinogen-binding protein (Efb) from S. aureus acts as an allosteric inhibitor by inducing conformational changes in complement fragment C3b that propagate across several domains and influence functional regions far distant from the Efb binding site. Most notably, the inhibitor impaired the interaction of C3b with complement factor B and, consequently, formation of the active C3 convertase. As this enzyme complex is critical for both activation and amplification of the complement response, its allosteric inhibition likely represents a fundamental contribution to the overall immune evasion strategy of S. aureus.


Subject(s)
Bacterial Proteins/metabolism , Complement C3b/metabolism , Complement Inactivator Proteins/metabolism , Immune Evasion/physiology , Models, Molecular , Protein Conformation , Staphylococcus aureus/physiology , Allosteric Regulation/physiology , Bacterial Proteins/chemistry , Complement C3-C5 Convertases/metabolism , Complement C3b/chemistry , Complement Factor B/metabolism , Complement Inactivator Proteins/chemistry , Mass Spectrometry , Scattering, Small Angle , Staphylococcus aureus/metabolism , Surface Plasmon Resonance
8.
J Mol Recognit ; 22(6): 495-505, 2009.
Article in English | MEDLINE | ID: mdl-19658192

ABSTRACT

Compstatin is a 13-residue peptide that inhibits activation of the complement system by binding to the central component C3 and its fragments C3b and C3c. A combination of theoretical and experimental approaches has previously allowed us to develop analogs of the original compstatin peptide with up to 264-fold higher activity; one of these analogs is now in clinical trials for the treatment of age-related macular degeneration (AMD). Here we used functional assays, surface plasmon resonance (SPR), and isothermal titration calorimetry (ITC) to assess the effect of modifications at three key residues (Trp-4, Asp-6, Ala-9) on the affinity and activity of compstatin and its analogs, and we correlated our findings to the recently reported co-crystal structure of compstatin and C3c. The K(D) values for the panel of tested analogs ranged from 10(-6) to 10(-8) M. These differences in binding affinity could be attributed mainly to differences in dissociation rather than association rates, with a >4-fold range in k(on) values (2-10 x 10(5) M(-1) s(-1)) and a k(off) variation of >35-fold (1-37 x 10(-2) s(-1)) being observed. The stability of the C3b-compstatin complex seemed to be highly dependent on hydrophobic effects at position 4, and even small changes at position 6 resulted in a loss of complex formation. Induction of a beta-turn shift by an A9P modification resulted in a more favorable entropy but a loss of binding specificity and stability. The results obtained by the three methods utilized here were highly correlated with regard to the activity/affinity of the analogs. Thus, our analyses have identified essential structural features of compstatin and provided important information to support the development of analogs with improved efficacy.


Subject(s)
Complement C3/chemistry , Peptides, Cyclic/chemistry , Calorimetry/methods , Indoles/chemistry , Kinetics , Models, Molecular , Molecular Conformation , Nitrogen/chemistry , Peptides/chemistry , Protein Structure, Secondary , Sensitivity and Specificity , Structure-Activity Relationship , Surface Plasmon Resonance , Tryptophan/chemistry , Water/chemistry
9.
J Immunol ; 183(4): 2565-74, 2009 Aug 15.
Article in English | MEDLINE | ID: mdl-19625656

ABSTRACT

Staphylococcus aureus possesses an impressive arsenal of complement evasion proteins that help the bacterium escape attack of the immune system. The staphylococcal complement inhibitor (SCIN) protein exhibits a particularly high potency and was previously shown to block complement by acting at the level of the C3 convertases. However, many details about the exact binding and inhibitory mechanism remained unclear. In this study, we demonstrate that SCIN directly binds with nanomolar affinity to a functionally important area of C3b that lies near the C terminus of its beta-chain. Direct competition of SCIN with factor B for C3b slightly decreased the formation of surface-bound convertase. However, the main inhibitory effect can be attributed to an entrapment of the assembled convertase in an inactive state. Whereas native C3 is still able to bind to the blocked convertase, no generation and deposition of C3b could be detected in the presence of SCIN. Furthermore, SCIN strongly competes with the binding of factor H to C3b and influences its regulatory activities: the SCIN-stabilized convertase was essentially insensitive to decay acceleration by factor H and the factor I- and H-mediated conversion of surface-bound C3b to iC3b was significantly reduced. By targeting a key area on C3b, SCIN is able to block several essential functions within the alternative pathway, which explains the high potency of the inhibitor. Our findings provide an important insight into complement evasion strategies by S. aureus and may act as a base for further functional studies.


Subject(s)
Complement C3b/metabolism , Complement Inactivator Proteins/physiology , Multigene Family/immunology , Staphylococcus aureus/immunology , Complement C3 Convertase, Alternative Pathway/metabolism , Complement C3 Convertase, Alternative Pathway/physiology , Complement C3b/chemistry , Complement Factor B/metabolism , Complement Factor H/metabolism , Complement Inactivator Proteins/metabolism , Humans , Protein Binding/immunology , Staphylococcus aureus/pathogenicity , Virulence
10.
Nat Immunol ; 10(7): 728-33, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19503104

ABSTRACT

Factor H (FH) is an abundant regulator of complement activation and protects host cells from self-attack by complement. Here we provide insight into the regulatory activity of FH by solving the crystal structure of the first four domains of FH in complex with its target, complement fragment C3b. FH interacted with multiple domains of C3b, covering a large, extended surface area. The structure indicated that FH destabilizes the C3 convertase by competition and electrostatic repulsion and that FH enables proteolytic degradation of C3b by providing a binding platform for protease factor I while stabilizing the overall domain arrangement of C3b. Our results offer general models for complement regulation and provide structural explanations for disease-related mutations in the genes encoding both FH and C3b.


Subject(s)
Complement C3b/chemistry , Complement Factor H/chemistry , Protein Structure, Tertiary , Binding Sites , Complement C3b/genetics , Complement C3b/metabolism , Complement Factor H/genetics , Complement Factor H/metabolism , Crystallography, X-Ray , Humans , Macromolecular Substances/chemistry , Macromolecular Substances/metabolism , Models, Molecular , Mutation , Protein Binding
11.
J Mol Biol ; 357(1): 82-99, 2006 Mar 17.
Article in English | MEDLINE | ID: mdl-16426633

ABSTRACT

Human immunodeficiency virus (HIV) entry into cells is initiated by the binding of its envelope glycoprotein (Env) gp120 to receptor CD4. Antibodies that bind to epitopes overlapping the CD4-binding site (CD4bs) on gp120 can prevent HIV entry by competing with cell-associated CD4; their ability to outcompete CD4 is a major determinant of their neutralizing potency and is proportional to their avidity. The breadth of neutralization and the likelihood of the emergence of antibody-resistant virus are critically dependent on the structure of their epitopes. Because CD4bs is highly conserved, it is reasonable to hypothesize that antibodies closely mimicking CD4 could exhibit relatively broad cross-reactivity and a high probability of preventing the emergence of resistant viruses. Previously, in a search for antibodies that mimic CD4 or the co-receptor, we identified and characterized a broadly cross-reactive HIV-neutralizing CD4bs human monoclonal antibody (hmAb), m18. Here, we describe the crystal structure of Fab m18 at 2.03 A resolution, which reveals unique conformations of heavy chain complementarity-determining regions (CDRs) 2 and 3 (H2 and H3). H2 is highly bulged and lacks cross-linking interstrand hydrogen bonds observed in all four canonical structures. H3 is 17.5 A long and rigid, forming an extended beta-sheet decorated with an alpha-turn motif bearing a phenylalanine-isoleucine fork at the apex. It shows striking similarity to the Ig CDR2-like C'C'' region of the CD4 first domain D1 that dominates the binding of CD4 to gp120. Docking simulations suggest significant similarity between the m18 epitope and the CD4bs on gp120. Fab m18 does not enhance binding of CD4-induced (CD4i) antibodies, nor does it induce CD4-independent fusion mediated by the HIV Env. Thus, vaccine immunogens based on the m18 epitope structure are unlikely to elicit antibodies that could enhance infection. The structure can also serve as a basis for the design of novel, highly efficient inhibitors of HIV entry.


Subject(s)
Antibodies, Monoclonal/chemistry , CD4 Antigens/chemistry , HIV Antibodies/chemistry , HIV-1/immunology , Molecular Mimicry , Protein Structure, Tertiary , Amino Acid Sequence , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Binding Sites , CD4 Antigens/immunology , Complementarity Determining Regions , Crystallography, X-Ray , Epitopes , HIV Antibodies/genetics , HIV Antibodies/immunology , HIV Envelope Protein gp120/chemistry , HIV Envelope Protein gp120/metabolism , Humans , Hydrogen Bonding , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/genetics , Immunoglobulin Fab Fragments/immunology , Models, Molecular , Molecular Sequence Data , Protein Structure, Secondary , Sequence Alignment
SELECTION OF CITATIONS
SEARCH DETAIL
...