Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
Proc Natl Acad Sci U S A ; 112(32): 9860-5, 2015 Aug 11.
Article in English | MEDLINE | ID: mdl-26216971

ABSTRACT

Antibody-based therapeutics have now had success in the clinic. The affinity and specificity of the antibody for the target ligand determines the specificity of therapeutic delivery and off-target side effects. The discovery and optimization of high-affinity antibodies to important therapeutic targets could be significantly improved by the availability of a robust, eukaryotic display technology comparable to phage display that would overcome the protein translation limitations of microorganisms. The use of eukaryotic cells would improve the diversity of the displayed antibodies that can be screened and optimized as well as more seamlessly transition into a large-scale mammalian expression system for clinical production. In this study, we demonstrate that the replication and polypeptide display characteristics of a eukaryotic retrovirus, avian leukosis virus (ALV), offers a robust, eukaryotic version of bacteriophage display. The binding affinity of a model single-chain Fv antibody was optimized by using ALV display, improving affinity >2,000-fold, from micromolar to picomolar levels. We believe ALV display provides an extension to antibody display on microorganisms and offers virus and cell display platforms in a eukaryotic expression system. ALV display should enable an improvement in the diversity of properly processed and functional antibody variants that can be screened and affinity-optimized to improve promising antibody candidates.


Subject(s)
Antibodies/metabolism , Avian Leukosis Virus/metabolism , Cell Surface Display Techniques/methods , Eukaryotic Cells/metabolism , Amino Acid Sequence , Animals , Cell Line , Chickens , Complementarity Determining Regions , Glycoproteins/metabolism , Humans , Kinetics , Laminin/metabolism , Molecular Sequence Data , Mutagenesis , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Protein Binding , Recombinant Fusion Proteins/metabolism , Single-Chain Antibodies/metabolism , Viral Envelope Proteins/metabolism , Virion/metabolism , Virus Replication
2.
Virol J ; 14(1): 211, 2017 11 03.
Article in English | MEDLINE | ID: mdl-29100527

ABSTRACT

BACKGROUND: Condyloma acuminatum, infected by low-risk human papillomaviruses (e.g., HPV6 and HPV11), is one of the most widespread sexually transmitted diseases. Apolipoprotein B mRNA-editing enzyme catalytic polypeptide-like 3 proteins (APOBEC3s, A3s) are cellular cytidine deaminases acting as antiviral factors through hypermutation of viral genome. However, it remains unknown whether A3s results in HPV11 gene mutations and interferon-ω (IFN-ω) exhibits antiviral activities through the A3s system. Here we investigated whether enhanced APOBEC3A (A3A) resulted in the E6 gene mutations and explore the effects of recombinant human interferon-ω (rhIFN-ω) on A3s/E6 expression in HaCaT keratinocytes containing the genome of HPV 11 (HPV11.HaCaT cells). METHODS: A3A-overexpressed HPV11.HaCaT (A3A-HPV11.HaCaT) cells were established by lentiviral infection and verified by immunofluorescence and western-blotting. Cell cycle, E6 gene mutations, APOBEC3s/E6 gene expression and subcellular localization were detected by FACS, 3D-PCR and sequencing, qRT-PCR and immunofluorescence respectively. RESULTS: The results suggested that A3A-HPV11.HaCaT cells were successfully established. Enhanced A3A induced S-phase arrest, G > A/C > T mutations and obvious reduction of E6 mRNA expression. A3A/A3B mRNA expression was up-regulated at 6 h and 12 h and obvious A3A staining existed throughout HPV11.HaCaT cells after rhIFN-ω treatment. RhIFN-ω could also inhibit mRNA expression of HPV11 E6 significantly. CONCLUSIONS: Enhanced A3A repressed HPV11 E6 expression through gene hypermutation, and rhIFN-ω might be an effective agent against HPV11 infection by up-regulation of A3A.


Subject(s)
Condylomata Acuminata/immunology , Cytidine Deaminase/metabolism , Human papillomavirus 11/genetics , Interferon Type I/metabolism , Oncogene Proteins, Viral/genetics , Proteins/metabolism , Cell Cycle , Cell Line , Condylomata Acuminata/virology , Cytidine Deaminase/genetics , Gene Expression , Humans , Keratinocytes/virology , Mutation , Proteins/genetics , RNA, Messenger , Up-Regulation
3.
Small ; 10(5): 964-72, 2014 Mar 12.
Article in English | MEDLINE | ID: mdl-24108667

ABSTRACT

Hydrogen peroxide (H2 O2 ) is a prominent member of the reactive oxygen species family and plays crucial roles in living organisms, thus detecting H2 O2 and elucidating its biological functions has become an important area of biological and biomedical research. Herein, a multifunctional fluorescent nanoprobe is demonstrated for detecting mitochondrial H2 O2 . The nanoprobe is prepared by covalently linking a mitochondria-targeting ligand (triphenylphosphonium, TPP) and a H2 O2 recognition element (PFl) onto carbon dots (CDs). For this nanoprobe, the CD serves as the carrier and the FRET donor. In the presence of H2 O2 , the PFl moieties on a CD undergo structural and spectral conversion, affording the nanoplatform a FRET-based ratiometric probe for H2 O2 . The nanoprobe displays excellent water dispersibility, high sensitivity and selectivity, satisfactory cell permeability, and very low cytotoxicity. Following the living cell uptake, this nanoprobe can specifically target and stain the mitochondria; and it can detect the exogenous H2 O2 in L929 cells, as well as the endogenously produced mitochondrial H2 O2 in Raw 264.7 cells upon stimulation by PMA. This study shows that CDs can serve as promising nano-carriers for fabricating practical multifunctional fluorescent nanosensors.


Subject(s)
Fluorescence Resonance Energy Transfer/methods , Fluorescent Dyes/metabolism , Hydrogen Peroxide/metabolism , Mitochondria/metabolism , Molecular Imaging/methods , Molecular Probes/metabolism , Nanoparticles/chemistry , Animals , Buffers , Cell Death , Cell Survival , Mice , Solutions , Spectrometry, Fluorescence , Water
4.
Emerg Microbes Infect ; : 2373307, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38953857

ABSTRACT

ABSTRACTSARS-CoV-2 has been evolving into a large number of variants, including the highly pathogenic Delta variant, and the currently prevalent Omicron subvariants with extensive evasion capability, which raises an urgent need to develop new broad-spectrum neutralizing antibodies. Herein, we engineer two IgG-(scFv)2 form bispecific antibodies with overlapping epitopes (bsAb1) or non-overlapping epitopes (bsAb2). Both bsAbs are significantly superior to the parental monoclonal antibodies in terms of their antigen-binding and virus-neutralizing activities against all tested circulating SARS-CoV-2 variants including currently dominant JN.1. The bsAb1 can efficiently neutralize all variants insensitive to parental monoclonal antibodies or the cocktail with IC50 lower than 20 ng/mL, even slightly better than bsAb2. Furthermore, the cryo-EM structures of bsAb1 in complex with the Omicron spike protein revealed that bsAb1 with overlapping epitopes effectively locked the S protein, which accounts for its conserved neutralization against Omicron variants. The bispecific antibody strategy engineered from overlapping epitopes provides a novel solution for dealing with viral immune evasion.

5.
Nat Commun ; 15(1): 2987, 2024 Apr 06.
Article in English | MEDLINE | ID: mdl-38582870

ABSTRACT

Nipah virus (NiV) is a World Health Organization priority pathogen and there are currently no approved drugs for clinical immunotherapy. Through the use of a naïve human phage-displayed Fab library, two neutralizing antibodies (NiV41 and NiV42) targeting the NiV receptor binding protein (RBP) were identified. Following affinity maturation, antibodies derived from NiV41 display cross-reactivity against both NiV and Hendra virus (HeV), whereas the antibody based on NiV42 is only specific to NiV. Results of immunogenetic analysis reveal a correlation between the maturation of antibodies and their antiviral activity. In vivo testing of NiV41 and its mature form (41-6) show protective efficacy against a lethal NiV challenge in hamsters. Furthermore, a 2.88 Å Cryo-EM structure of the tetrameric RBP and antibody complex demonstrates that 41-6 blocks the receptor binding interface. These findings can be beneficial for the development of antiviral drugs and the design of vaccines with broad spectrum against henipaviruses.


Subject(s)
Henipavirus Infections , Nipah Virus , Humans , Antibodies, Neutralizing/metabolism , Nipah Virus/metabolism , Antibodies, Viral
6.
Nat Commun ; 15(1): 4330, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38773072

ABSTRACT

The Hendra and Nipah viruses (HNVs) are highly pathogenic pathogens without approved interventions for human use. In addition, the interaction pattern between the attachment (G) and fusion (F) glycoproteins required for virus entry remains unclear. Here, we isolate a panel of Macaca-derived G-specific antibodies that cross-neutralize HNVs via multiple mechanisms. The most potent antibody, 1E5, confers adequate protection against the Nipah virus challenge in female hamsters. Crystallography demonstrates that 1E5 has a highly similar binding pattern to the receptor. In cryo-electron microscopy studies, the tendency of 1E5 to bind to the upper or lower heads results in two distinct quaternary structures of G. Furthermore, we identify the extended outer loop ß1S2-ß1S3 of G and two pockets on the apical region of fusion (F) glycoprotein as the essential sites for G-F interactions. This work highlights promising drug candidates against HNVs and contributes deeper insights into the viruses.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Cryoelectron Microscopy , Henipavirus Infections , Viral Fusion Proteins , Animals , Antibodies, Neutralizing/immunology , Female , Antibodies, Viral/immunology , Henipavirus Infections/virology , Henipavirus Infections/immunology , Viral Fusion Proteins/immunology , Viral Fusion Proteins/chemistry , Humans , Viral Envelope Proteins/immunology , Viral Envelope Proteins/chemistry , Nipah Virus/immunology , Virus Internalization/drug effects , Henipavirus/immunology , Cricetinae , Cross Reactions/immunology , Hendra Virus/immunology , Macaca , Mesocricetus , Crystallography, X-Ray
7.
Front Microbiol ; 14: 1114226, 2023.
Article in English | MEDLINE | ID: mdl-36925463

ABSTRACT

Instruction: Rift valley fever virus (RVFV) is a mosquito-transmitted bunyavirus that causes severe disease in animals and humans. Nevertheless, there are no vaccines applied to prevent RVFV infection for human at present. Therefore, it is necessary to develop a safe and effective RVFV vaccine. Methods: We generated Ad5-GnGcopt, a replication-deficient recombinant Ad5 vector (human adenovirus serotype 5) expressing codon-optimized RVFV glycoproteins Gn and Gc, and evaluated its immunogenicity and protective efficacy in mice. Results and Discussion: Intramuscular immunization of Ad5-GnGcopt in mice induces strong and durable antibody production and robust cellular immune responses. Additionally, a single vaccination with Ad5-GnGcopt vaccination can completely protect interferon-α/ß receptor-deficient A129 mice from lethal RVFV infection. Our work indicates that Ad5-GnGcopt might represent a potential vaccine candidate against RVFV. However, further research is needed, first to confirm its efficacy in a natural animal host, and ultimately escalate as a potential vaccine candidate for humans.

8.
Cell Discov ; 9(1): 37, 2023 Apr 04.
Article in English | MEDLINE | ID: mdl-37015915

ABSTRACT

The pandemic of COVID-19 caused by SARS-CoV-2 continues to spread around the world. Mutant strains of SARS-CoV-2 are constantly emerging. At present, Omicron variants have become mainstream. In this work, we carried out a systematic and comprehensive analysis of the reported spike protein antibodies, counting the epitopes and genotypes of these antibodies. We further comprehensively analyzed the impact of Omicron mutations on antibody epitopes and classified these antibodies according to their binding patterns. We found that the epitopes of the H-RBD class antibodies were significantly less affected by Omicron mutations than other classes. Binding and virus neutralization experiments showed that such antibodies could effectively inhibit the immune escape of Omicron. Cryo-EM results showed that this class of antibodies utilized a conserved mechanism to neutralize SARS-CoV-2. Our results greatly help us deeply understand the impact of Omicron mutations. Meanwhile, it also provides guidance and insights for developing Omicron antibodies and vaccines.

9.
NPJ Vaccines ; 8(1): 164, 2023 Oct 27.
Article in English | MEDLINE | ID: mdl-37891181

ABSTRACT

Rift Valley fever virus (RVFV) is listed as a priority pathogen by the World Health Organization (WHO) because it causes serious and fatal disease in humans, and there are currently no effective countermeasures. Therefore, it is urgent to develop a safe and efficacious vaccine. Here, we developed six nucleotide-modified mRNA vaccines encoding different regions of the Gn and Gc proteins of RVFV encapsulated in lipid nanoparticles, compared their ability to induce immune responses in mice and found that mRNA vaccine encoding the full-length Gn and Gc proteins had the strongest ability to induce cellular and humoral immune responses. IFNAR(-/-) mice vaccinated with mRNA-GnGc were protected from lethal RVFV challenge. In addition, mRNA-GnGc induced high levels of neutralizing antibodies and cellular responses in rhesus macaques, as well as antigen-specific memory B cells. These data demonstrated that mRNA-GnGc is a potent and promising vaccine candidate for RVFV.

10.
Int J Biol Macromol ; 253(Pt 3): 126817, 2023 Dec 31.
Article in English | MEDLINE | ID: mdl-37690653

ABSTRACT

SARS-CoV-2, a type of respiratory virus, has exerted a great impact on global health and economy over the past three years. Antibody-based therapy was initially successful but later failed due to the accumulation of mutations in the spike protein of the virus. Strategies that enable antibodies to resist virus escape are therefore of great significance. Here, we engineer a bispecific SARS-CoV-2 neutralizing nanobody in secretory Immunoglobulin A (SIgA) format, named S2-3-IgA2m2, which shows broad and potent neutralization against SARS-CoV-1, SARS-CoV-2 and its variants of concern (VOCs) including XBB and BQ.1.1. S2-3-IgA2m2 is ∼1800-fold more potent than its parental IgG counterpart in neutralizing XBB. S2-3-IgA2m2 is stable in mouse lungs at least for three days when administrated by nasal delivery. In hamsters infected with BA.5, three intranasal doses of S2-3-IgA2m2 at 1 mg/kg significantly reduce viral RNA loads and completely eliminate infectious particles in the trachea and lungs. Notably, even at single dose of 1 mg/kg, S2-3-IgA2m2 prophylactically administered through the intranasal route drastically reduces airway viral RNA loads and infectious particles. This study provides an effective weapon combating SARS-CoV-2, proposes a new strategy overcoming the virus escape, and lays strategic reserves for rapid response to potential future outbreaks of "SARS-CoV-3".


Subject(s)
Antibodies , SARS-CoV-2 , Animals , Cricetinae , Mice , Disease Outbreaks , Immunoglobulin A, Secretory , RNA, Viral , Antibodies, Viral , Antibodies, Neutralizing
11.
Microbiol Immunol ; 56(6): 406-10, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22420921

ABSTRACT

Staphylococcus aureus is the most common cause of hospital-acquired bacteremia. Due to emergence of antibiotic-resistant strains, these infections present a serious public health threat. In this study, to develop a broadly protective vaccine, we tested whether immune responses induced by several proteins associated with S. aureus toxicity could protect mice from lethal challenge with human clinical S. aureus isolate USA300. We found that the surface protein A (SasA) of S. aureus could protect mice from lethal challenge of the bacteria.


Subject(s)
Bacterial Proteins/immunology , Membrane Proteins/immunology , Staphylococcal Infections/prevention & control , Staphylococcal Vaccines/immunology , Staphylococcus aureus/immunology , Animals , Bacterial Proteins/administration & dosage , Female , Membrane Proteins/administration & dosage , Mice , Mice, Inbred BALB C , Staphylococcal Infections/immunology , Staphylococcal Infections/mortality , Staphylococcal Vaccines/administration & dosage , Staphylococcus aureus/pathogenicity , Survival Analysis
12.
Viruses ; 14(6)2022 06 18.
Article in English | MEDLINE | ID: mdl-35746803

ABSTRACT

The global spread of SARS-CoV-2 and its variants poses a serious threat to human health worldwide. Recently, the emergence of Omicron has presented a new challenge to the prevention and control of the COVID-19 pandemic. A convenient and reliable in vitro neutralization assay is an important method for validating the efficiency of antibodies, vaccines, and other potential drugs. Here, we established an effective assay based on a pseudovirus carrying a full-length spike (S) protein of SARS-CoV-2 variants in the HIV-1 backbone, with a luciferase reporter gene inserted into the non-replicate pseudovirus genome. The key parameters for packaging the pseudovirus were optimized, including the ratio of the S protein expression plasmids to the HIV backbone plasmids and the collection time for the Alpha, Beta, Gamma, Kappa, and Omicron pseudovirus particles. The pseudovirus neutralization assay was validated using several approved or developed monoclonal antibodies, underscoring that Omicron can escape some neutralizing antibodies, such as REGN10987 and REGN10933, while S309 and ADG-2 still function with reduced neutralization capability. The neutralizing capacity of convalescent plasma from COVID-19 convalescent patients in Wuhan was tested against these pseudoviruses, revealing the immune evasion of Omicron. Our work established a practical pseudovirus-based neutralization assay for SARS-CoV-2 variants, which can be conducted safely under biosafety level-2 (BSL-2) conditions, and this assay will be a promising tool for studying and characterizing vaccines and therapeutic candidates against Omicron-included SARS-CoV-2 variants.


Subject(s)
COVID-19 , SARS-CoV-2 , Antibodies, Neutralizing , Antibodies, Viral , COVID-19/therapy , Humans , Immunization, Passive , Neutralization Tests/methods , Pandemics , SARS-CoV-2/genetics , Spike Glycoprotein, Coronavirus , COVID-19 Serotherapy
13.
Front Immunol ; 13: 907675, 2022.
Article in English | MEDLINE | ID: mdl-36439179

ABSTRACT

Rift Valley fever virus (RVFV) is one of the most important virulent pathogens causing severe disease in animals and humans. However, there is currently no approved vaccine to prevent RVFV infection in humans. The use of human adenovirus serotype 4 (Ad4) as a vector for an RVFV vaccine has not been reported. Here, we report the generation of a replication-competent recombinant Ad4 vector expressing codon-optimized forms of the RVFV glycoproteins Gn and Gc (named Ad4-GnGc). Intramuscular immunization with Ad4-GnGc elicited robust neutralizing antibodies against RVFV and cellular immune responses in mice. A single low-dose vaccination with Ad4-GnGc completely protected interferon-α/ß receptor-deficient A129 mice from lethal RVFV infection. More importantly, Ad4-GnGc efficacy was not affected by pre-existing immunity to adenovirus serotype 5, which currently exists widely in populations. These results suggest that Ad4-GnGc is a promising vaccine candidate against RVFV.


Subject(s)
Adenoviridae Infections , Adenovirus Vaccines , Rift Valley Fever , Rift Valley fever virus , Viral Vaccines , Mice , Humans , Animals , Rift Valley fever virus/genetics , Rift Valley Fever/prevention & control , Adenoviridae/genetics
14.
Cell Discov ; 8(1): 132, 2022 Dec 09.
Article in English | MEDLINE | ID: mdl-36494344

ABSTRACT

Current SARS-CoV-2 Omicron subvariants impose a heavy burden on global health systems by evading immunity from most developed neutralizing antibodies and vaccines. Here, we identified a nanobody (aSA3) that strongly cross-reacts with the receptor binding domain (RBD) of both SARS-CoV-1 and wild-type (WT) SARS-CoV-2. The dimeric construct of aSA3 (aSA3-Fc) tightly binds and potently neutralizes both SARS-CoV-1 and WT SARS-CoV-2. Based on X-ray crystallography, we engineered a bispecific nanobody dimer (2-3-Fc) by fusing aSA3-Fc to aRBD-2, a previously identified broad-spectrum nanobody targeting an RBD epitope distinct from aSA3. 2-3-Fc exhibits single-digit ng/mL neutralizing potency against all major variants of concerns including BA.5. In hamsters, a single systemic dose of 2-3-Fc at 10 mg/kg conferred substantial efficacy against Omicron infection. More importantly, even at three low doses of 0.5 mg/kg, 2-3-Fc prophylactically administered through the intranasal route drastically reduced viral RNA loads and completely eliminated infectious Omicron particles in the trachea and lungs. Finally, we discovered that 2(Y29G)-3-Fc containing a Y29G substitution in aRBD-2 showed better activity than 2-3-Fc in neutralizing BA.2.75, a recent Omicron subvariant that emerged in India. This study expands the arsenal against SARS-CoV-1, provides potential therapeutic and prophylactic candidates that fully cover major SARS-CoV-2 variants, and may offer a simple preventive approach against Omicron and its subvariants.

15.
Signal Transduct Target Ther ; 7(1): 139, 2022 04 27.
Article in English | MEDLINE | ID: mdl-35478188

ABSTRACT

The SARS-CoV-2 Omicron variant shows substantial resistance to neutralization by infection- and vaccination-induced antibodies, highlighting the demands for research on the continuing discovery of broadly neutralizing antibodies (bnAbs). Here, we developed a panel of bnAbs against Omicron and other variants of concern (VOCs) elicited by vaccination of adenovirus-vectored COVID-19 vaccine (Ad5-nCoV). We also investigated the human longitudinal antibody responses following vaccination and demonstrated how the bnAbs evolved over time. A monoclonal antibody (mAb), named ZWD12, exhibited potent and broad neutralization against SARS-CoV-2 variants Alpha, Beta, Gamma, Kappa, Delta, and Omicron by blocking the spike protein binding to the angiotensin-converting enzyme 2 (ACE2) and provided complete protection in the challenged prophylactic and therapeutic K18-hACE2 transgenic mouse model. We defined the ZWD12 epitope by determining its structure in complex with the spike (S) protein via cryo-electron microscopy. This study affords the potential to develop broadly therapeutic mAb drugs and suggests that the RBD epitope bound by ZWD12 is a rational target for the design of a broad spectrum of vaccines.


Subject(s)
COVID-19 , SARS-CoV-2 , Animals , Antibodies, Monoclonal/genetics , Antibodies, Viral , Broadly Neutralizing Antibodies , COVID-19/prevention & control , COVID-19 Vaccines/genetics , Cryoelectron Microscopy , Epitopes , Humans , Mice , SARS-CoV-2/genetics , Spike Glycoprotein, Coronavirus/genetics , Vaccination , Viral Envelope Proteins
16.
Viruses ; 13(3)2021 03 15.
Article in English | MEDLINE | ID: mdl-33804206

ABSTRACT

Lassa virus (LASV) is a rodent-borne arenavirus circulating in West African regions that causes Lassa fever (LF). LF is normally asymptomatic at the initial infection stage, but can progress to severe disease with multiorgan collapse and hemorrhagic fever. To date, the therapeutic choices are limited, and there is no approved vaccine for avoiding LASV infection. Adenoviral vector-based vaccines represent an effective countermeasure against LASV because of their safety and adequate immunogenicity, as demonstrated in use against other emerging viral infections. Here, we constructed and characterized a novel Ad5 (E1-, E3-) vectored vaccine containing the glycoprotein precursor (GPC) of LASV. Ad5-GPCLASV elicited both humoral and cellular immune responses in BALB/c mice. Moreover, a bioluminescent imaging-based BALB/c mouse model infected with GPC-bearing and luciferase-expressing replication-incompetent LASV pseudovirus was utilized to evaluate the vaccine efficacy. The bioluminescence intensity of immunized mice was significantly lower than that of control mice after being inoculated with LASV pseudovirus. This study suggests that Ad5-GPCLASV represents a potential vaccine candidate against LF.


Subject(s)
Adenoviridae , Genetic Vectors/immunology , Lassa Fever , Viral Vaccines/immunology , Africa, Western , Animals , HEK293 Cells , Humans , Immunity, Cellular , Lassa Fever/immunology , Lassa Fever/prevention & control , Lassa virus/immunology , Mice , Mice, Inbred BALB C
17.
Int Immunopharmacol ; 91: 107297, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33360088

ABSTRACT

Clostridium tetani causes life-threatening disease by producing tetanus neurotoxin (TeNT), one of the most toxic protein substances. Toxicosis can be prevented and cured by administration of anti-TeNT neutralizing antibodies. Here, we identified a series of monoclonal antibodies (mAbs) derived from memory B cells of a healthy adult immunized with the C-terminal domain of TeNT (TeNT-Hc). Thirteen mAbs bound to both tetanus toxoid (TT) and TeNT-Hc, while two mAbs recognized only TT. VH3-23 was the most frequently used germline gene in these TT-binding mAbs, and the pairwise identity values of the VH gene sequences ranged from 27% to 69%. Three of these mAbs-T3, T7, and T9-6-completely protected mice from challenge with 2× LD50 of TeNT, and two (T2 and T18) significantly prolonged the survival time. The five neutralizing mAbs recognized distinct epitopes on TT, with binding affinities ranging from 0.123 to 11.9 nM. Our study provides promising therapeutic candidates for tetanus.


Subject(s)
Antibodies, Bacterial/administration & dosage , Antibodies, Monoclonal/administration & dosage , Antibodies, Neutralizing/administration & dosage , Clostridium tetani/immunology , Diphtheria-Tetanus Vaccine/administration & dosage , Diphtheria-Tetanus-acellular Pertussis Vaccines/administration & dosage , Immunogenicity, Vaccine , Tetanus Toxoid/administration & dosage , Tetanus/prevention & control , Animals , Antibodies, Bacterial/blood , Antibodies, Monoclonal/blood , Antibodies, Neutralizing/blood , Antibody Specificity , Clostridium tetani/pathogenicity , Disease Models, Animal , Epitopes , Female , Humans , Mice, Inbred BALB C , Tetanus/immunology , Tetanus/microbiology , Time Factors , Vaccination
18.
J Am Chem Soc ; 132(37): 12780-1, 2010 Sep 22.
Article in English | MEDLINE | ID: mdl-20795619

ABSTRACT

Bioluminescence, due to its high sensitivity, has been exploited in various analytical and imaging applications. In this work, we report a highly stable, cell-transductable, and wavelength-tunable bioluminescence system achieved with an elegant and simple design. Using aqueous in situ polymerization on a bioluminescent enzyme anchored with polymerizable vinyl groups, we obtained nanosized core-shell nanocapsules with the enzyme as the core and a cross-linked thin polymer net as the shell. These nanocapsules possess greatly enhanced stability, retained bioactivity, and a readily engineered surface. In particular, by incorporating polymerizable amines in the polymerization, we endowed the nanocapsules with efficient cell-transduction and sufficient conjugation sites for follow-up modification. Following in situ polymerization, decorating the polymer shell with fluorescent quantum dots allowed us to access a continuous tunable wavelength, which extends the application of such bioluminescent nanocapsules, especially in deep tissue. In addition, the unique core-shell structure and adequate conjugation sites on surface enabled us to maximize the BRET efficiency by adjusting the QD/enzyme conjugation ratio.


Subject(s)
Luminescent Proteins/chemistry , Nanocapsules/chemistry , Quantum Dots , Horseradish Peroxidase/chemistry , Horseradish Peroxidase/metabolism , Luminescent Proteins/metabolism , Models, Molecular , Molecular Conformation , Permeability
19.
MAbs ; 12(1): 1742457, 2020.
Article in English | MEDLINE | ID: mdl-32213108

ABSTRACT

Ebola virus (EBOV) can cause severe hemorrhagic fever in humans, and no approved treatment is currently available. Although several antibodies have achieved good protection in animal models, the potential emerging isolates of ebolavirus and the unknown effects of experimental antibodies in humans underscore the need to develop additional antibodies to address the threat of Ebola. Here, we isolated a series of memory B cell-derived monoclonal antibodies from healthy Chinese adults vaccinated with Ad5-EBOV. These antibodies were encoded by diverse germline genes and had high levels of somatic hypermutation. Most antibodies were cross-reactive and could bind at least two ebolavirus glycoproteins (GPs). Seven neutralizing antibodies were identified using HIV-EBOV GP-Luc pseudovirus, and they effectively neutralized authentic EBOV. In particular, monoclonal antibody 2G1 exhibited potent cross-neutralization against HIV-EBOV/SUDV/BDBV GP-Luc bearing different ebolavirus GPs. We used truncated GPs, competition assays, and software prediction to analyze seven neutralizing antibodies, which bound four different epitopes on GP. Importantly, three of these antibodies provided complete protection in mice when administered one day post-infection. Our study expands the list of candidate antibodies and the options for successfully treating ebolavirus infection.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Ebola Vaccines/immunology , Ebolavirus/immunology , Animals , Female , Hemorrhagic Fever, Ebola/immunology , Hemorrhagic Fever, Ebola/prevention & control , Humans , Male , Mice
20.
Viruses ; 12(3)2020 02 27.
Article in English | MEDLINE | ID: mdl-32120864

ABSTRACT

The Rift Valley fever virus (RVFV) is an arthropod-borne virus that can not only cause severe disease in domestic animals but also in humans. However, the licensed vaccines or available therapeutics for humans do not exist. Here, we report two Gn-specific neutralizing antibodies (NAbs), isolated from a rhesus monkey immunized with recombinant human adenoviruses type 4 expressing Rift Valley fever virus Gn and Gc protein (rHAdV4-GnGcopt). The two NAbs were both able to protect host cells from RVFV infection. The interactions between NAbs and Gn were then characterized to demonstrate that these two NAbs might preclude RVFV glycoprotein rearrangement, hindering the exposure of fusion loops in Gc to endosomal membranes after the virus invades the host cell. The target region for the two NAbs is located in the Gn domain III, implying that Gn is a desired target for developing vaccines and neutralizing antibodies against RVFV.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Rift Valley Fever/immunology , Rift Valley fever virus/immunology , Viral Envelope Proteins/immunology , Animals , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/chemistry , Antibodies, Viral/blood , Antibodies, Viral/chemistry , Cell Line , Enzyme-Linked Immunosorbent Assay , Epitopes , Humans , Immunization , Macaca mulatta , Models, Molecular , Molecular Conformation , Neutralization Tests , Protein Binding , Rift Valley Fever/virology , Structure-Activity Relationship , Viral Envelope Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL