Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Med ; 28(9): 1802-1812, 2022 09.
Article in English | MEDLINE | ID: mdl-35927581

ABSTRACT

Clearing amyloid-ß (Aß) through immunotherapy is one of the most promising therapeutic approaches to Alzheimer's disease (AD). Although several monoclonal antibodies against Aß have been shown to substantially reduce Aß burden in patients with AD, their effects on improving cognitive function remain marginal. In addition, a significant portion of patients treated with Aß-targeting antibodies experience brain edema and microhemorrhage associated with antibody-mediated Fc receptor activation in the brain. Here, we develop a phagocytosis inducer for Aß consisting of a single-chain variable fragment of an Aß-targeting monoclonal antibody fused with a truncated receptor binding domain of growth arrest-specific 6 (Gas6), a bridging molecule for the clearance of dead cells via TAM (TYRO3, AXL, and MERTK) receptors. This chimeric fusion protein (αAß-Gas6) selectively eliminates Aß plaques through TAM receptor-dependent phagocytosis without inducing NF-kB-mediated inflammatory responses or reactive gliosis. Furthermore, αAß-Gas6 can induce synergistic clearance of Aß by activating both microglial and astrocytic phagocytosis, resulting in better behavioral outcomes with substantially reduced synapse elimination and microhemorrhage in AD and cerebral amyloid angiopathy model mice compared with Aß antibody treatment. Our results suggest that αAß-Gas6 could be a novel immunotherapeutic agent for AD that overcomes the side effects of conventional antibody therapy.


Subject(s)
Alzheimer Disease , Single-Chain Antibodies , Alzheimer Disease/drug therapy , Alzheimer Disease/therapy , Amyloid beta-Peptides/metabolism , Animals , Anti-Inflammatory Agents/therapeutic use , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Disease Models, Animal , Mice , Mice, Transgenic , NF-kappa B , Plaque, Amyloid/drug therapy , Receptors, Fc/therapeutic use , Single-Chain Antibodies/therapeutic use , c-Mer Tyrosine Kinase
2.
Mol Ther ; 30(2): 579-592, 2022 02 02.
Article in English | MEDLINE | ID: mdl-34628052

ABSTRACT

CD19-targeting chimeric antigen receptor (CAR) T cells have become an important therapeutic option for patients with relapsed and refractory B cell malignancies. However, a significant portion of patients still do not benefit from the therapy owing to various resistance mechanisms, including high expression of multiple inhibitory immune checkpoint receptors. Here, we report a lentiviral two-in-one CAR T approach in which two checkpoint receptors are downregulated simultaneously by a dual short hairpin RNA cassette integrated into a CAR vector. Using this system, we evaluated CD19-targeting CAR T cells in the context of four different checkpoint combinations-PD-1/TIM-3, PD-1/LAG-3, PD-1/CTLA-4, and PD-1/TIGIT-and found that CAR T cells with PD-1/TIGIT downregulation uniquely exerted synergistic antitumor effects. Importantly, functional and phenotypic analyses suggested that downregulation of PD-1 enhances short-term effector function, whereas downregulation of TIGIT is primarily responsible for maintaining a less differentiated/exhausted state, providing a potential mechanism for the observed synergy. The PD-1/TIGIT-downregulated CAR T cells generated from diffuse large B cell lymphoma patient-derived T cells also showed robust antitumor activity and significantly improved persistence in vivo. The efficacy and safety of PD-1/TIGIT-downregulated CD19-targeting CAR T cells are currently being evaluated in adult patients with relapsed or refractory large B cell lymphoma (ClinicalTrials.gov: NCT04836507).


Subject(s)
Lymphoma, Large B-Cell, Diffuse , Programmed Cell Death 1 Receptor , Antigens, CD19 , Down-Regulation , Humans , Immunotherapy, Adoptive , Phenotype , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , T-Lymphocytes
3.
ACS Synth Biol ; 6(9): 1635-1641, 2017 09 15.
Article in English | MEDLINE | ID: mdl-28548828

ABSTRACT

Bacteriophages are thought to be ideal vehicles for linking antibodies to nanoparticles. Here, we define the sequence of peptides exposed as a fusion protein on M13 bacteriophages to yield optimal binding of gold nanocubes and efficient bacteriophage amplification. We generated five helper bacteriophage libraries using AE(X)2DP, AE(X)3DP, AE(X)4DP, AE(X)5DP, and AE(X)6DP as the exposed portion of pVIII, in which X was a randomized amino acid residue encoded by the nucleotide sequence NNK. Efficient phage amplification was achievable only in the AE(X)2DP, AE(X)3DP, and AE(X)4DP libraries. Through biopanning with gold nanocubes, we enriched the phage clones and selected the clone with the highest fold change after enrichment. This clone displayed Pro-Asp on the surface of the bacteriophage and had amplification yields similar to those of the wild-type helper bacteriophage (VCSM13). The clone displayed even binding of gold nanocubes along its length and minimal aggregation after binding. We conclude that, for efficient amplification, the exposed pVIII amino acid length should be limited to six residues and Ala-Glu-Pro-Asp-Asp-Pro (AEPDDP) is the ideal fusion protein sequence for guaranteeing the optimal formation of a complex with gold nanocubes.


Subject(s)
Aspartic Acid/chemistry , Bacteriophage M13/genetics , Gold/chemistry , Metal Nanoparticles/chemistry , Peptide Library , Proline/chemistry , Protein Interaction Mapping/methods , Aspartic Acid/genetics , Binding Sites , Proline/genetics , Protein Binding , Sequence Analysis, Protein/methods , Staining and Labeling/methods
4.
Exp Mol Med ; 48(11): e271, 2016 11 18.
Article in English | MEDLINE | ID: mdl-27857068

ABSTRACT

The C-terminal domain of RNA polymerase II is an unusual series of repeated residues appended to the C-terminus of the largest subunit and serves as a flexible binding scaffold for numerous nuclear factors. The binding of these factors is determined by the phosphorylation patterns on the repeats in the domain. In this study, we generated a synthetic antibody library by replacing the third heavy chain complementarity-determining region of an anti-HER2 (human epidermal growth factor receptor 2) antibody (trastuzumab) with artificial sequences of 7-18 amino-acid residues. From this library, antibodies were selected that were specific to serine phosphopeptides that represent typical phosphorylation patterns on the functional unit (YSPTSPS)2 of the RNA polymerase II C-terminal domain (CTD). Antibody clones pCTD-1stS2 and pCTD-2ndS2 showed specificity for peptides with phosphoserine at the second residues of the first or second heptamer repeat, respectively. Additional clones specifically reacted to peptides with phosphoserine at the fifth serine of the first repeat (pCTD-1stS5), the seventh residue of the first repeat and fifth residue of the second repeat (pCTD-S7S5) or the seventh residue of either the first or second repeat (pCTD-S7). All of these antibody clones successfully reacted to RNA polymerase II in immunoblot analysis. Interestingly, pCTD-2ndS2 precipitated predominately RNA polymerase II from the exonic regions of genes in genome-wide chromatin immunoprecipitation sequencing analysis, which suggests that the phosphoserine at the second residue of the second repeat of the functional unit (YSPTSPS)2 is a mediator of exon definition.


Subject(s)
Antibodies/metabolism , Chromatin Immunoprecipitation/methods , Exons , RNA Polymerase II/metabolism , Antibodies/immunology , HEK293 Cells , HeLa Cells , Humans , Phosphorylation , Protein Binding , RNA Polymerase II/immunology
5.
PLoS One ; 10(10): e0141312, 2015.
Article in English | MEDLINE | ID: mdl-26512723

ABSTRACT

To date, four main mechanisms mediating inhibition of influenza infection by anti-hemagglutinin antibodies have been reported. Anti-globular-head-domain antibodies block either influenza virus receptor binding to the host cell or progeny virion release from the host cell. Anti-stem region antibodies hinder the membrane fusion process or induce antibody-dependent cytotoxicity to infected cells. In this study we identified a human monoclonal IgG1 antibody (CT302), which does not inhibit both the receptor binding and the membrane fusion process but efficiently reduced the nucleus entry of viral nucleoprotein suggesting a novel inhibition mechanism of viral infection by antibody. This antibody binds to the subtype-H3 hemagglutinin globular head domain of group-2 influenza viruses circulating throughout the population between 1997 and 2007.


Subject(s)
Antibodies, Viral/immunology , Influenza, Human/immunology , Influenza, Human/virology , Nucleoproteins/metabolism , Orthomyxoviridae/physiology , Virus Replication , Amino Acid Sequence , Animals , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antibody Affinity/immunology , Cell Line , Disease Models, Animal , Epitope Mapping/methods , Epitopes/chemistry , Epitopes/immunology , Ferrets , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Humans , Immunoglobulin G/immunology , Influenza A Virus, H3N2 Subtype/immunology , Male , Mice , Models, Molecular , Molecular Sequence Data , Neutralization Tests , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Protein Binding , Protein Conformation , Sequence Alignment
6.
Exp Mol Med ; 46: e114, 2014 Sep 19.
Article in English | MEDLINE | ID: mdl-25236766

ABSTRACT

The N-terminal fragment of prohormone brain natriuretic peptide (NT-proBNP) is a commonly used biomarker for the diagnosis of congestive heart failure, although its biological function is not well known. NT-proBNP exhibits heavy O-linked glycosylation, and it is quite difficult to develop an antibody that exhibits glycosylation-independent binding. We developed an antibody that binds to the recombinant NT-proBNP protein and its deglycosylated form with similar affinities in an enzyme immunoassay. The epitope was defined as Gly63-Lys68 based on mimetic peptide screening, site-directed mutagenesis and a competition assay with a peptide mimotope. The nearest O-glycosylation residues are Thr58 and Thr71; therefore, four amino acid residues intervene between the epitope and those residues in both directions. In conclusion, we report that an antibody reactive to Gly63-Lys68 of NT-proBNP exhibits O-glycosylation-independent binding.


Subject(s)
Antibodies/immunology , Epitopes/immunology , Natriuretic Peptide, Brain/immunology , Peptide Fragments/immunology , Amino Acid Sequence , Animals , Antigen-Antibody Reactions , Epitope Mapping , Epitopes/chemistry , Epitopes/genetics , Glycosylation , HEK293 Cells , Heart Failure/immunology , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Natriuretic Peptide, Brain/chemistry , Natriuretic Peptide, Brain/genetics , Peptide Fragments/chemistry , Peptide Fragments/genetics , Rabbits , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL