Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 82
Filter
1.
Nat Commun ; 13(1): 7864, 2022 12 21.
Article in English | MEDLINE | ID: mdl-36543789

ABSTRACT

Contemporary influenza A H3N2 viruses circulating since 2016 have acquired a glycosylation site in the neuraminidase in close proximity to the enzymatic active site. Here, we investigate if this S245N glycosylation site, as a result of antigenic evolution, can impact binding and function of human monoclonal antibodies that target the conserved active site. While we find that a reduction in the inhibitory ability of neuraminidase active site binders is measurable, this class of broadly reactive monoclonal antibodies maintains protective efficacy in vivo.


Subject(s)
Antibodies, Monoclonal , Influenza A Virus, H3N2 Subtype , Neuraminidase , Humans , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Antibodies, Viral/chemistry , Antibodies, Viral/metabolism , Catalytic Domain/immunology , Catalytic Domain/physiology , Glycosylation , Hemagglutinin Glycoproteins, Influenza Virus , Influenza A virus , Influenza A Virus, H3N2 Subtype/immunology , Influenza A Virus, H3N2 Subtype/metabolism , Influenza, Human/immunology , Influenza, Human/metabolism , Neuraminidase/chemistry , Neuraminidase/immunology
2.
Front Immunol ; 12: 786617, 2021.
Article in English | MEDLINE | ID: mdl-34868073

ABSTRACT

Neuraminidase of influenza A and B viruses plays a critical role in the virus life cycle and is an important target of the host immune system. Here, we highlight the current understanding of influenza neuraminidase structure, function, antigenicity, immunogenicity, and immune protective potential. Neuraminidase inhibiting antibodies have been recognized as correlates of protection against disease caused by natural or experimental influenza A virus infection in humans. In the past years, we have witnessed an increasing interest in the use of influenza neuraminidase to improve the protective potential of currently used influenza vaccines. A number of well-characterized influenza neuraminidase-specific monoclonal antibodies have been described recently, most of which can protect in experimental challenge models by inhibiting the neuraminidase activity or by Fc receptor-dependent mechanisms. The relative instability of the neuraminidase poses a challenge for protein-based antigen design. We critically review the different solutions that have been proposed to solve this problem, ranging from the inclusion of stabilizing heterologous tetramerizing zippers to the introduction of inter-protomer stabilizing mutations. Computationally engineered neuraminidase antigens have been generated that offer broad, within subtype protection in animal challenge models. We also provide an overview of modern vaccine technology platforms that are compatible with the induction of robust neuraminidase-specific immune responses. In the near future, we will likely see the implementation of influenza vaccines that confront the influenza virus with a double punch: targeting both the hemagglutinin and the neuraminidase.


Subject(s)
Influenza Vaccines/immunology , Influenza, Human/prevention & control , Neuraminidase/immunology , Viral Proteins/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Antigenic Drift and Shift , Antigens, Viral/immunology , Antigens, Viral/ultrastructure , Catalytic Domain/genetics , Catalytic Domain/immunology , Cross Protection , Evolution, Molecular , Humans , Immunogenicity, Vaccine , Influenza Vaccines/administration & dosage , Influenza Vaccines/genetics , Influenza, Human/immunology , Influenza, Human/virology , Alphainfluenzavirus/enzymology , Alphainfluenzavirus/genetics , Alphainfluenzavirus/immunology , Betainfluenzavirus/enzymology , Betainfluenzavirus/genetics , Betainfluenzavirus/immunology , Mutation , Nanoparticles , Neuraminidase/administration & dosage , Neuraminidase/genetics , Neuraminidase/ultrastructure , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Vaccines, Synthetic/ultrastructure , Viral Proteins/administration & dosage , Viral Proteins/genetics , Viral Proteins/ultrastructure
3.
Sci Rep ; 11(1): 13865, 2021 07 05.
Article in English | MEDLINE | ID: mdl-34226629

ABSTRACT

Staphylococcus aureus is an opportunistic pathogen causing high morbidity and mortality. Since multi-drug resistant S. aureus lineages are nowadays omnipresent, alternative tools for preventive or therapeutic interventions, like immunotherapy, are urgently needed. However, there are currently no vaccines against S. aureus. Surface-exposed and secreted proteins are regarded as potential targets for immunization against S. aureus infections. Yet, many potential staphylococcal antigens of this category do not elicit protective immune responses. To obtain a better understanding of this problem, we compared the binding of serum IgGs from healthy human volunteers, highly S. aureus-colonized patients with the genetic blistering disease epidermolysis bullosa (EB), or immunized mice to the purified S. aureus peptidoglycan hydrolases Sle1, Aly and LytM and their different domains. The results show that the most abundant serum IgGs from humans and immunized mice target the cell wall-binding domain of Sle1, and the catalytic domains of Aly and LytM. Interestingly, in a murine infection model, these particular IgGs were not protective against S. aureus bacteremia. In contrast, relatively less abundant IgGs against the catalytic domain of Sle1 and the N-terminal domains of Aly and LytM were almost exclusively detected in sera from EB patients and healthy volunteers. These latter IgGs may contribute to the protection against staphylococcal infections, as previous studies suggest that serum IgGs protect EB patients against severe S. aureus infection. Together, these observations focus attention on the use of particular protein domains for vaccination to direct potentially protective immune responses towards the most promising epitopes within staphylococcal antigens.


Subject(s)
Immunoglobulin G/immunology , Methicillin-Resistant Staphylococcus aureus/immunology , N-Acetylmuramoyl-L-alanine Amidase/immunology , Staphylococcal Infections/immunology , Animals , Antibodies, Bacterial/immunology , Antigens, Bacterial/immunology , Catalytic Domain/genetics , Catalytic Domain/immunology , Cell Wall/genetics , Cell Wall/immunology , Epitopes/genetics , Epitopes/immunology , Humans , Immunoglobulin G/genetics , Methicillin-Resistant Staphylococcus aureus/genetics , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Mice , N-Acetylmuramoyl-L-alanine Amidase/chemistry , Peptidoglycan/genetics , Peptidoglycan/immunology , Staphylococcal Infections/genetics , Staphylococcal Infections/prevention & control
4.
Immunity ; 53(4): 852-863.e7, 2020 10 13.
Article in English | MEDLINE | ID: mdl-32976769

ABSTRACT

Influenza B virus (IBV) infections can cause severe disease in children and the elderly. Commonly used antivirals have lower clinical effectiveness against IBV compared to influenza A viruses (IAV). Neuraminidase (NA), the second major surface protein on the influenza virus, is emerging as a target of broadly protective antibodies that recognize the NA active site of IAVs. However, similarly broadly protective antibodies against IBV NA have not been identified. Here, we isolated and characterized human monoclonal antibodies (mAbs) that target IBV NA from an IBV-infected patient. Two mAbs displayed broad and potent capacity to inhibit IBV NA enzymatic activity, neutralize the virus in vitro, and protect against lethal IBV infection in mice in prophylactic and therapeutic settings. These mAbs inserted long CDR-H3 loops into the NA active site, engaging residues highly conserved among IBV NAs. These mAbs provide a blueprint for the development of improved vaccines and therapeutics against IBVs.


Subject(s)
Antibodies, Viral/immunology , Catalytic Domain/immunology , Influenza B virus/immunology , Neuraminidase/immunology , Viral Proteins/immunology , Animals , Antibodies, Monoclonal/immunology , Cell Line , Dogs , Female , HEK293 Cells , Humans , Influenza A virus/immunology , Influenza, Human/immunology , Leukocytes, Mononuclear/immunology , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred BALB C , Middle Aged , Orthomyxoviridae Infections/immunology
5.
Sci Rep ; 10(1): 13932, 2020 08 18.
Article in English | MEDLINE | ID: mdl-32811892

ABSTRACT

Botulinum neurotoxins (BoNTs) represent a family of bacterial toxins responsible for neuroparalytic disease 'botulism' in human and animals. Their potential use as biological weapon led to their classification in category 'A' biowarfare agent by Centers for Disease Control and Prevention (CDC), USA. In present study, gene encoding full length catalytic domain of BoNT/E-LC was cloned, expressed and protein was purified using Ni-NTA chromatography. Humoral immune response was confirmed by Ig isotyping and cell-mediated immunity by cytokine profiling and intracellular staining for enumeration of IFN-γ secreting CD4+ and CD8+ T cells. Increased antibody titer with the predominance of IgG subtype was observed. An interaction between antibodies produced against rBoNT/E-LC was established that showed the specificity against BoNT/E in SPR assay. Animal protection with rBoNT/E-LC was conferred through both humoral and cellular immune responses. These findings were supported by cytokine profiling and flow cytometric analysis. Splenocytes stimulated with rBoNT/E-LC showed a 3.27 and 2.8 times increase in the IFN-γ secreting CD4+ and CD8+ T cells, respectively; in immunized group (P < 0.05). Protection against BoNT/E challenge tended to relate with increase in the percentage of rBoNT/E-LC specific IL-2 in the splenocytes supernatant (P = 0.034) and with IFN-γ-producing CD4+ T cell responses (P = 0.045). We have immunologically evaluated catalytically active rBoNT/E-LC. Our results provide valuable investigational report for immunoprophylactic role of catalytic domain of BoNT/E.


Subject(s)
Botulinum Toxins/genetics , Botulism/prevention & control , Animals , Antibodies, Neutralizing/immunology , Botulinum Toxins/chemistry , Botulinum Toxins/immunology , Botulinum Toxins, Type A/chemistry , Botulinum Toxins, Type A/immunology , Botulism/metabolism , CD8-Positive T-Lymphocytes/immunology , Catalytic Domain/genetics , Catalytic Domain/immunology , Cloning, Molecular/methods , Clostridium botulinum/genetics , Humans , Immunization , Male , Mice , Mice, Inbred BALB C
6.
J Mol Model ; 25(11): 337, 2019 Nov 09.
Article in English | MEDLINE | ID: mdl-31705312

ABSTRACT

Nattokinase is an enzyme produced by Bacillus subtilis subsp. natto that contains strong fibrinolytic activity. It has potential to treat cardiovascular diseases. In silico analysis revealed that nattokinase is considered as an antigen, thus hindering its application for injectable therapeutic protein. Various web servers were used to predict B-cell epitopes of nattokinase both continuously and discontinuously to determine which amino acid residues had been responsible for the immunogenicity. With the exclusion of the predicted conserved amino acids, four amino acids such as S18, Q19, T242, and Q245 were allowed for mutation. Substitution mutation was done to lower the immunogenicity of native nattokinase. Through the stability of the mutated protein with the help of Gibbs free energy difference, the proposed mutein was S18D, Q19I, T242Y, and Q245W. The 3D model of the mutated nattokinase was modeled and validated with various tools. Physicochemical properties and stability analysis of the protein indicated that the mutation brought higher stability without causing any changes in the catalytic site of nattokinase. Molecular dynamics simulation implied that the mutation indicated similar stability, conformation, and behavior compared to the native nattokinase. These results are highly likely to contribute to the wet lab experiment to develop safer nattokinase.


Subject(s)
Antibody Formation/immunology , Bacillus subtilis/immunology , Bacterial Proteins/immunology , Mutagenesis/immunology , Subtilisins/immunology , Catalytic Domain/immunology , Molecular Dynamics Simulation , Mutation/immunology
7.
J Thromb Haemost ; 17(9): 1461-1469, 2019 09.
Article in English | MEDLINE | ID: mdl-31136076

ABSTRACT

BACKGROUND: Excessive, plasmin-mediated fibrinolysis augments bleeding and contributes to death in some patients. Current therapies for fibrinolytic bleeding are limited by modest efficacy, low potency, and off-target effects. OBJECTIVES: To determine whether an antibody directed against unique loop structures of the plasmin protease domain may have enhanced specificity and potency for blocking plasmin activity, fibrinolysis, and experimental hemorrhage. METHODS: The binding specificity, affinity, protease cross-reactivity and antifibrinolytic properties of a monoclonal plasmin inhibitor antibody (Pi) were examined and compared with those of epsilon aminocaproic acid (EACA), which is a clinically used fibrinolysis inhibitor. RESULTS: Pi specifically recognized loop 5 of the protease domain, and did not bind to other serine proteases or nine other non-primate plasminogens. Pi was ~7 logs more potent in neutralizing plasmin cleavage of small-molecule substrates and >3 logs more potent in quenching fibrinolysis than EACA. Pi was similarly effective in blocking catalysis of a small-molecule substrate as α2 -antiplasmin, which is the most potent covalent inhibitor of plasmin, and was a more potent fibrinolysis inhibitor. Fab or chimerized Fab fragments of Pi were equivalently effective. In vivo, in a humanized model of fibrinolytic surgical bleeding, Pi significantly reduced bleeding to a greater extent than a clinical dose of EACA. CONCLUSIONS: A mAb directed against unique loop sequences in the protease domain is a highly specific, potent, competitive plasmin inhibitor that significantly reduces experimental surgical bleeding in vivo.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antifibrinolytic Agents/therapeutic use , Fibrinolysin/antagonists & inhibitors , Hemorrhage/drug therapy , Aminocaproic Acid/pharmacology , Aminocaproic Acid/therapeutic use , Animals , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/pharmacology , Antibody Affinity , Binding, Competitive , Catalytic Domain/immunology , Cross Reactions , Drug Evaluation, Preclinical , Female , Fibrinolysin/chemistry , Fibrinolysin/immunology , Fibrinolysis/drug effects , Hemorrhage/blood , Humans , Mice , Mice, Inbred C57BL , Models, Molecular , Protein Conformation , Protein Domains , Random Allocation , Recombinant Fusion Proteins/immunology , Species Specificity , Substrate Specificity
8.
Science ; 364(6442): 775-778, 2019 05 24.
Article in English | MEDLINE | ID: mdl-31072904

ABSTRACT

G protein-coupled receptors (GPCRs) in the G protein-coupled active state have higher affinity for agonists as compared with when they are in the inactive state, but the molecular basis for this is unclear. We have determined four active-state structures of the ß1-adrenoceptor (ß1AR) bound to conformation-specific nanobodies in the presence of agonists of varying efficacy. Comparison with inactive-state structures of ß1AR bound to the identical ligands showed a 24 to 42% reduction in the volume of the orthosteric binding site. Potential hydrogen bonds were also shorter, and there was up to a 30% increase in the number of atomic contacts between the receptor and ligand. This explains the increase in agonist affinity of GPCRs in the active state for a wide range of structurally distinct agonists.


Subject(s)
Adrenergic beta-1 Receptor Agonists/chemistry , Drug Design , Receptors, G-Protein-Coupled/agonists , Adrenergic beta-1 Receptor Agonists/pharmacology , Allosteric Site/immunology , Catalytic Domain/immunology , Hydrogen Bonding , Ligands , Protein Binding , Protein Structure, Secondary , Receptors, Adrenergic, beta-1/chemistry , Receptors, Adrenergic, beta-1/immunology , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/immunology , Single-Domain Antibodies/immunology
9.
PLoS One ; 12(3): e0174187, 2017.
Article in English | MEDLINE | ID: mdl-28323873

ABSTRACT

Human botulism is primarily caused by botulinum neurotoxin (BoNT) serotypes A, B and E, with around 1% caused by serotype F (BoNT/F). BoNT/F comprises at least seven different subtypes with the amino acid sequence difference between subtypes as high as 36%. The sequence differences present a significant challenge for generating monoclonal antibodies (mAbs) that can bind, detect and neutralize all BoNT/F subtypes. We used repertoire cloning of immune mouse antibody variable (V) regions and yeast display to generate a panel of 33 lead single chain Fv (scFv) mAbs that bound one or more BoNT/F subtypes with a median equilibrium dissociation constant (KD) of 4.06 × 10-9 M. By diversifying the V-regions of the lead mAbs and selecting for cross reactivity we generated five mAbs that bound each of the seven subtypes. Three scFv binding non-overlapping epitopes were converted to IgG that had KD for the different BoNT/F subtypes ranging from 2.2×10-8 M to 1.47×10-12 pM. An equimolar combination of the mAbs was able to potently neutralize BoNT/F1, F2, F4 and F7 in the mouse neutralization assay. The mAbs have potential utility as diagnostics capable of recognizing the known BoNT/F subtypes and could be developed as antitoxins to prevent and treat type F botulism.


Subject(s)
Antibodies, Monoclonal/immunology , Antitoxins/immunology , Botulinum Toxins/immunology , Single-Chain Antibodies/immunology , Amino Acid Sequence , Animals , Antitoxins/genetics , Botulism/diagnosis , Botulism/therapy , Catalytic Domain/immunology , Clostridium botulinum/metabolism , Cross Reactions/immunology , Epitope Mapping , Epitopes/immunology , Escherichia coli/genetics , Immunization , Mice , Saccharomyces cerevisiae/genetics , Single-Chain Antibodies/genetics
10.
Biochem J ; 471(3): 391-401, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26303525

ABSTRACT

Adamalysin-like metalloproteinases with thrombospondin (TS) motifs (ADAMTS)-5 is the multi-domain metalloproteinase that most potently degrades aggrecan proteoglycan in the cartilage and its activity is implicated in the development of osteoarthritis (OA). To generate specific exosite inhibitors for it, we screened a phage display antibody library in the presence of the zinc-chelating active site-directed inhibitor GM6001 (Ilomastat) and isolated four highly selective inhibitory antibodies. Two antibodies were mapped to react with exosites in the catalytic/disintegrin domains (Cat/Dis) of the enzyme, one in the TS domain and one in the spacer domain (Sp). The antibody reacting with the Sp blocked the enzyme action only when aggrecan or the Escherichia coli-expressed aggrecan core protein were substrates, but not against a peptide substrate. The study with this antibody revealed the importance of the Sp for effective aggrecanolytic activity of ADAMTS-5 and that this domain does not interact with sulfated glycosaminoglycans (GAGs) but with the protein moiety of the proteoglycan. An antibody directed against the Cat/Dis of ADAMTS-5 was effective in a cell-based model of aggrecan degradation; however, the anti-Sp antibody was ineffective. Western blot analysis of endogenous ADAMTS-5 expressed by human chondrocytes showed the presence largely of truncated forms of ADAMTS-5, thus explaining the lack of efficacy of the anti-Sp antibody. The possibility of ADAMTS-5 truncation must then be taken into account when considering developing anti-ancillary domain antibodies for therapeutic purposes.


Subject(s)
ADAM Proteins/immunology , Antibodies/immunology , Cartilage/immunology , Osteoarthritis/genetics , ADAM Proteins/antagonists & inhibitors , ADAM Proteins/therapeutic use , ADAMTS5 Protein , Aggrecans/biosynthesis , Aggrecans/immunology , Antibodies/therapeutic use , Binding Sites/immunology , Cartilage/pathology , Catalytic Domain/immunology , Cell Surface Display Techniques , Chondrocytes/immunology , Chondrocytes/pathology , Dipeptides/administration & dosage , Humans , Osteoarthritis/immunology , Osteoarthritis/pathology , Protein Binding/immunology , Protein Structure, Tertiary/genetics , Substrate Specificity
11.
J Infect Chemother ; 21(9): 639-46, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26134278

ABSTRACT

Isolation of Bordetella pertussis and detection of the pertussis genome are not always successful because of low bacterial loads in adult patients with pertussis. Antibodies against pertussis toxin (PT) are measured but have low sensitivity in vaccinated subjects. There is no reliable diagnostic method at present. In this study, a fluorescent-EIA against several pertussis antigens and genome detection were investigated to establish clinical laboratory diagnostic methods for pertussis. The study was conducted in an outpatient clinic between September 2007 and 2013. Subjects consisted of 209 patients including adults suspected of pertussis and 35 staff members of the clinic. Loop-mediated isothermal amplification (LAMP) was performed to detect the pertussis genome in 5' UTR of the pertussis toxin (PT) gene. The catalytic region of the adenylate cyclase toxin (catACT), C-terminal of filamentous hemagglutinin (cFHA), and type 3 fimbria (Fim3) were selected, which are not pertussis vaccine component. Conventional PT and FHA antibodies were examined together with type 2 fimbria (Fim2) antibodies, and these are vaccine antigens. Pertussis DNA was detected in 23 (11%) out of 209. Detection sensitivity was high in young infants. Antibodies against Fim3 showed a higher positive rate in all age groups. Staff members at the pediatric outpatient clinic showed serological booster responses in Fim2 and Fim3 antibodies more sensitively than those in PT antibodies during outbreaks. LAMP was useful for detecting the pertussis genome in young infants, whereas a serological assay for fluorescent-EIA against Fim2 and Fim3 was preferable for adolescents and adults.


Subject(s)
Antibodies, Bacterial/blood , Antigens, Bacterial/immunology , Bordetella pertussis/immunology , Fimbriae Proteins/immunology , Virulence Factors, Bordetella/immunology , Whooping Cough/diagnosis , Whooping Cough/immunology , 5' Untranslated Regions , Adenylate Cyclase Toxin/immunology , Adhesins, Bacterial/immunology , Adolescent , Adult , Ambulatory Care Facilities , Bordetella pertussis/genetics , Catalytic Domain/immunology , Child , Child, Preschool , DNA, Bacterial/blood , Enzyme-Linked Immunosorbent Assay , Fimbriae, Bacterial/immunology , Fluorescence , Health Personnel , Humans , Infant , Middle Aged , Nucleic Acid Amplification Techniques , Pertussis Toxin/genetics , Pertussis Toxin/immunology , Whooping Cough/blood , Young Adult
12.
J Virol ; 88(16): 9072-85, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24899176

ABSTRACT

UNLABELLED: Rotaviruses and orbiviruses are nonturreted Reoviridae members. The rotavirus VP3 protein is a multifunctional capping enzyme and antagonist of the interferon-induced cellular oligoadenylate synthetase-RNase L pathway. Despite mediating important processes, VP3 is the sole protein component of the rotavirus virion whose structure remains unknown. In the current study, we used sequence alignment and homology modeling to identify features common to nonturreted Reoviridae capping enzymes and to predict the domain organization, structure, and active sites of rotavirus VP3. Our results suggest that orbivirus and rotavirus capping enzymes share a domain arrangement similar to that of the bluetongue virus capping enzyme. Sequence alignments revealed conserved motifs and suggested that rotavirus and orbivirus capping enzymes contain a variable N-terminal domain, a central guanine-N7-methyltransferase domain that contains an additional inserted domain, and a C-terminal guanylyltransferase and RNA 5'-triphosphatase domain. Sequence conservation and homology modeling suggested that the insertion in the guanine-N7-methyltransferase domain is a ribose-2'-O-methyltransferase domain for most rotavirus species. Our analyses permitted putative identification of rotavirus VP3 active-site residues, including those that form the ribose-2'-O-methyltransferase catalytic tetrad, interact with S-adenosyl-l-methionine, and contribute to autoguanylation. Previous reports have indicated that group A rotavirus VP3 contains a C-terminal 2H-phosphodiesterase domain that can cleave 2'-5' oligoadenylates, thereby preventing RNase L activation. Our results suggest that a C-terminal phosphodiesterase domain is present in the capping enzymes from two additional rotavirus species. Together, these findings provide insight into a poorly understood area of rotavirus biology and are a springboard for future biochemical and structural studies of VP3. IMPORTANCE: Rotaviruses are an important cause of severe diarrheal disease. The rotavirus VP3 protein caps viral mRNAs and helps combat cellular innate antiviral defenses, but little is known about its structure or enzymatic mechanisms. In this study, we used sequence- and structure-based alignments with related proteins to predict the structure of VP3 and identify enzymatic domains and active sites therein. This work provides insight into the mechanisms of rotavirus transcription and evasion of host innate immune defenses. An improved understanding of these processes may aid our ability to develop rotavirus vaccines and therapeutics.


Subject(s)
Capsid Proteins/genetics , Capsid Proteins/immunology , Immunity, Innate/immunology , Protein Structure, Tertiary/genetics , Rotavirus Infections/immunology , Rotavirus/genetics , Rotavirus/immunology , Amino Acid Sequence , Animals , Catalytic Domain/genetics , Catalytic Domain/immunology , Cell Line , Molecular Sequence Data , Orbivirus/genetics , Orbivirus/immunology , Phylogeny , Rotavirus Infections/virology , Sequence Alignment , Sf9 Cells , Spodoptera , Transcription, Genetic/genetics , Transcription, Genetic/immunology , Virion/genetics , Virion/immunology
13.
Immunol Lett ; 158(1-2): 7-13, 2014.
Article in English | MEDLINE | ID: mdl-24269767

ABSTRACT

Efforts to develop a vaccine for ricin toxin are focused on identifying highly immunogenic, safe, and thermostable recombinant derivatives of ricin's enzymatic A subunit (RTA). As a means to guide vaccine design, we have embarked on an effort to generate a comprehensive neutralizing and non-neutralizing B cell epitope map of RTA. In a series of previous studies, we identified three spatially distinct linear (continuous), neutralizing epitopes on RTA, as defined by monoclonal antibodies (mAbs) PB10 (and R70), SyH7, and GD12. In this report we now describe a new collection of 19 toxin-neutralizing mAbs that bind non-linear epitopes on RTA. The most potent toxin-neutralizing mAbs in this new collection, namely WECB2, TB12, PA1, PH12 and IB2 each had nanamolar (or sub-nanomolar) affinities for ricin and were each capable of passively protecting mice against a 5-10xLD50 toxin challenge. Competitive binding assays by surface plasmon resonance revealed that WECB2 binds an epitope that overlaps with PB10 and R70; TB12, PA1, PH12 recognize epitope(s) close to or overlapping with SyH7's epitope; and GD12 and IB2 recognize epitopes that are spatially distinct from all other toxin-neutralizing mAbs. We estimate that we have now accounted for ∼75% of the predicted epitopes on the surface of RTA and that toxin-neutralizing mAbs are directed against a very limited number of these epitopes. Having this information provides a framework for further refinement of RTA mutagenesis and vaccine design.


Subject(s)
Antibodies, Blocking/immunology , B-Lymphocytes/immunology , Epitopes, B-Lymphocyte/metabolism , Hemorrhage/prevention & control , Ricin/metabolism , Animals , Catalytic Domain/immunology , Epitope Mapping , Epitopes, B-Lymphocyte/chemistry , Epitopes, B-Lymphocyte/immunology , Female , Hemorrhage/chemically induced , Hemorrhage/immunology , Humans , Hybridomas , Mice , Mice, Inbred BALB C , Molecular Structure , Ricin/chemistry , Ricin/immunology , Vaccines
14.
Life Sci ; 93(18-19): 700-6, 2013 Nov 04.
Article in English | MEDLINE | ID: mdl-24044887

ABSTRACT

AIMS: The 3-phosphoinositide-dependent protein kinase-1 (PDK1) activates a number of protein kinases of the AGC subfamily, including protein kinase B and ribosomal S6 protein kinase by phosphorylating these kinases at the activation-loop. PDK1 activity is regulated by auto-phosphorylation and is further increased by stimulation of cells. PDK1 has been shown to have several phosphorylation sites including 5 serine and 3 tyrosine residues. However, Ser241 and Tyr373/376 are only involved in the regulation of PDK1 activity. MAIN METHODS: In this study, we found the putative fragments of PDK1 by using anti-Myc and anti-PDK1 antibodies. Furthermore, the existence of four different sizes of PDK1 were confirmed with other phosphosite specific antibodies. KEY FINDINGS: Taken together, the catalytic domain of PDK1 (42 kDa and 37 kDa) is separately existed in the cells and might be important for the regulation of subset of PDK1 substrate. Because the crystal structural studies suggested that PIF-pocket is located at the catalytic domain and plays a critical role on substrate recognition. SIGNIFICANCE: These suggested importance and roles of this fragment are needed to be determined. Further study on these fragments of PDK1 will provide new insight on the regulatory mechanism of PDK1 in patho-physiological condition.


Subject(s)
3-Phosphoinositide-Dependent Protein Kinases/immunology , 3-Phosphoinositide-Dependent Protein Kinases/metabolism , Antibodies, Catalytic/biosynthesis , Antibody Specificity , Peptide Fragments/metabolism , 3-Phosphoinositide-Dependent Protein Kinases/genetics , Amino Acid Sequence , Catalytic Domain/immunology , HEK293 Cells , Humans , Molecular Sequence Data , Peptide Fragments/genetics , Phosphorylation/immunology , Protein Binding/immunology
15.
Monoclon Antib Immunodiagn Immunother ; 32(3): 200-4, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23750478

ABSTRACT

Tyrosyl-tRNA synthetase (TyrRS) is one of the key enzymes of protein biosynthesis. In addition to its basic role, this enzyme reveals some important non-canonical functions. Under apoptotic conditions, the full-length enzyme splits into two fragments having distinct cytokine activities, thereby linking protein synthesis to cytokine signaling pathways. The NH2-terminal catalytic fragment, known as miniTyrRS, binds strongly to the CXC-chemokine receptor CXCR1 and, like interleukin 8, functions as a chemoattractant for polymorphonuclear leukocytes. On the other hand, an extra COOH-terminal domain of human TyrRS has cytokine activities like those of a mature human endothelial monocyte-activating polypeptide II (EMAP II). Moreover, the etiology of specific diseases (cancer, neuronal pathologies, autoimmune disorders, and disrupted metabolic conditions) is connected to specific aminoacyl-tRNA synthetases. Here we report the generation and characterization of monoclonal antibodies specific to N- and C-terminal domains of TyrRS. Recombinant TyrRS and its N- and C-terminal domains were expressed as His-tag fusion proteins in bacteria. Affinity purified proteins have been used as antigens for immunization and hybridoma cell screening. Monoclonal antibodies specific to catalytic N-terminal module and C-terminal EMAP II-like domain of TyrRS may be useful as tools in various aspects of TyrRS function and cellular localization.


Subject(s)
Antibodies, Monoclonal/immunology , Catalytic Domain/immunology , Recombinant Proteins/immunology , Tyrosine-tRNA Ligase/immunology , 3T3 Cells , Amino Acid Sequence , Animals , Cell Line , HEK293 Cells , Humans , Mice , Mice, Inbred BALB C , Protein Binding , Protein Structure, Tertiary
16.
Annu Rev Immunol ; 31: 675-704, 2013.
Article in English | MEDLINE | ID: mdl-23330955

ABSTRACT

Phosphoinositide 3-kinases (PI3Ks) control many important aspects of immune cell development, differentiation, and function. Mammals have eight PI3K catalytic subunits that are divided into three classes based on similarities in structure and function. Specific roles for the class I PI3Ks have been broadly investigated and are relatively well understood, as is the function of their corresponding phosphatases. More recently, specific roles for the class II and class III PI3Ks have emerged. Through vertebrate evolution and in parallel with the evolution of adaptive immunity, there has been a dramatic increase not only in the genes for PI3K subunits but also in genes for phosphatases that act on 3-phosphoinositides and in 3-phosphoinositide-binding proteins. Our understanding of the PI3Ks in immunity is guided by fundamental discoveries made in simpler model organisms as well as by appreciating new adaptations of this signaling module in mammals in general and in immune cells in particular.


Subject(s)
Multigene Family/immunology , Phosphatidylinositol 3-Kinases/physiology , Signal Transduction/immunology , Animals , Catalytic Domain/immunology , Endocytosis/immunology , Humans , Multiprotein Complexes/immunology , Phagocytosis/immunology , Phosphatidylinositol 3-Kinases/classification , Protein Transport/immunology
17.
Protein Pept Lett ; 20(2): 205-12, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22894159

ABSTRACT

Lethal toxin of Clostridium sordellii (MLD 150 ng/kg) is one of the most potent Clostridial toxins and is responsible for most of the diseases including sudden death syndrome in cattle, sheep and toxic shock syndrome, necrotizing faciitis, neonatal omphalitis and gangrene in humans. Lethal toxin (TcsL) is a single chain protein of about 270 kDa. In the present study, 1.6 kb DNA fragment encoding for the catalytic domain of TcsL was PCR amplified, cloned in pQE30 UA vector and expressed in E. coli SG 13009. The expression of recombinant lethal toxin protein (rTcsL) was optimized and it was purified under native conditions using a single step Ni-NTA affinity chromatography. The purified recombinant protein was used for the production of polyclonal antibodies in mice and rabbit. The raised antibodies reacted specifically with the purified rTcsL and intact native lethal toxin on Western blot. The biological activity of the recombinant protein was tested in HeLa cells where it showed the cytotoxicity. Further, the polyclonal antibodies were used for in-vitro neutralization of purified rTcsL, acid precipitated C. sordellii and C. difficile native toxins in HeLa cells. Mice and rabbit anti-rTcsL sera effectively neutralized the cytotoxicity of rTcsL and C. sordellii native toxin but it did not neutralize the cytotoxicity of C. difficile toxin in HeLa cells.


Subject(s)
Antibodies/pharmacology , Bacterial Toxins/pharmacology , Clostridium sordellii/metabolism , Antibodies/immunology , Bacterial Toxins/antagonists & inhibitors , Bacterial Toxins/immunology , Catalytic Domain/immunology , Cell Survival/drug effects , Chromatography, Affinity , HeLa Cells , Humans
18.
J Immunol ; 189(5): 2365-73, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22855709

ABSTRACT

The classical pathway of complement is crucial to the immune system, but it also contributes to inflammatory diseases when dysregulated. Binding of the C1 complex to ligands activates the pathway by inducing autoactivation of associated C1r, after which C1r activates C1s. C1s cleaves complement component C4 and then C2 to cause full activation of the system. The interaction between C1s and C4 involves active site and exosite-mediated events, but the molecular details are unknown. In this study, we identified four positively charged amino acids on the serine protease domain that appear to form a catalytic exosite that is required for efficient cleavage of C4. These residues are coincidentally involved in coordinating a sulfate ion in the crystal structure of the protease. Together with other evidence, this pointed to the involvement of sulfate ions in the interaction with the C4 substrate, and we showed that the protease interacts with a peptide from C4 containing three sulfotyrosine residues. We present a molecular model for the interaction between C1s and C4 that provides support for the above data and poses questions for future research into this aspect of complement activation.


Subject(s)
Catalytic Domain/immunology , Complement Activation/immunology , Complement C1s/metabolism , Complement C4/metabolism , Complement Pathway, Classical/immunology , Serine Proteases/metabolism , Amino Acid Sequence , Binding Sites, Antibody/immunology , Complement C4/immunology , Humans , Molecular Sequence Data , Peptide Fragments/metabolism
19.
J Immunol ; 189(6): 2975-84, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22908330

ABSTRACT

The progression of autoimmune responses is associated with an avidity maturation process driven by preferential expansion of high avidity clonotypes at the expense of their low avidity counterparts. Central and peripheral tolerance hinder the contribution of high-avidity clonotypes targeting residues 206-214 of islet-specific glucose-6-phosphatase catalytic subunit-related protein (IGRP(206-214)) during the earliest stages of autoimmune diabetes. In this study, we probe the molecular determinants and biochemical consequences of IGRP(206-214)/K(d) recognition by high-, intermediate-, and low-avidity autoreactive CD8+ T cells, and we investigate the effects of genetic IGRP(206-214) silencing on their developmental biology. We find that differences in avidity for IGRP(206-214)/K(d) map to CDR1α and are associated with quantitative differences in CD3ε proline-rich sequence exposure and Nck recruitment. Unexpectedly, we find that tolerance of high-avidity CD8+ T cells, unlike their activation and recruitment into the pancreas, is dissociated from recognition of IGRP(206-214), particularly in adult mice. This finding challenges the view that tolerance of pathogenic autoreactive T cells is invariably triggered by recognition of the peptide-MHC complex that drives their activation in the periphery, indicating the existence of mechanisms of tolerance that are capable of sensing the avidity, hence pathogenicity of autoreactive T cells without the need to rely on local autoantigen availability.


Subject(s)
Autoantigens/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Catalytic Domain/immunology , Cell Movement/immunology , Glucose-6-Phosphatase/metabolism , Immune Tolerance , Proteins/metabolism , Aging/genetics , Aging/immunology , Amino Acid Sequence , Animals , Animals, Newborn , Autoantigens/genetics , Catalytic Domain/genetics , Cell Line , Cell Movement/genetics , Gene Silencing , Glucose-6-Phosphatase/antagonists & inhibitors , Glucose-6-Phosphatase/genetics , Immune Tolerance/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , Proteins/antagonists & inhibitors , Proteins/genetics
20.
Hybridoma (Larchmt) ; 31(3): 209-13, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22741586

ABSTRACT

Protein tyrosine phosphatase 1B (PTP1B), a member of the protein tyrosine phosphatase (PTP) family, plays a crucial role in metabolic signaling, with insulin and leptin signaling being well studied. New evidence indicates that PTP1B is also involved in cancer. In the present study, we report on the establishment of a monoclonal antibody specific for catalytic domain of PTP1B (PTP1Bc) generated through the hybridoma method. The monoclonal antibody is measured to have a titer of 4.1×10(6) against PTP1Bc in indirect ELISA. Western blot and immunofluorescent analyses indicated that this antibody can specifically combine native PTP1B in MDA-MB-231 and MDA-MB-453 cells. This monoclonal antibody against PTP1Bc can help enhance the understanding of PTP1B-related physiological and pathological mechanisms and may act as a therapeutic agent for diabetes, obesity, and cancer in the future.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/immunology , Immunoglobulin G/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/immunology , Recombinant Fusion Proteins/immunology , Animals , Antibodies, Monoclonal, Murine-Derived/biosynthesis , Antibody Specificity , Blotting, Western , Catalytic Domain/immunology , Cell Line, Tumor , Female , Fluorescent Antibody Technique, Indirect , Humans , Hybridomas , Immunoglobulin G/biosynthesis , Mice , Mice, Inbred BALB C , Protein Tyrosine Phosphatase, Non-Receptor Type 1/isolation & purification , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Titrimetry
SELECTION OF CITATIONS
SEARCH DETAIL
...