Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Ther ; 28(7): 1585-1599, 2020 07 08.
Article in English | MEDLINE | ID: mdl-32454027

ABSTRACT

HIV infection preferentially depletes HIV-specific CD4+ T cells, thereby impairing antiviral immunity. In this study, we explored the therapeutic utility of adoptively transferred CD4+ T cells expressing an HIV-specific chimeric antigen receptor (CAR4) to restore CD4+ T cell function to the global HIV-specific immune response. We demonstrated that CAR4 T cells directly suppressed in vitro HIV replication and eliminated virus-infected cells. Notably, CAR4 T cells containing intracellular domains (ICDs) derived from the CD28 receptor family (ICOS and CD28) exhibited superior effector functions compared to the tumor necrosis factor receptor (TNFR) family ICDs (CD27, OX40, and 4-1BB). However, despite demonstrating limited in vitro efficacy, only HIV-resistant CAR4 T cells expressing the 4-1BBζ ICD exhibited profound expansion, concomitant with reduced rebound viremia after antiretroviral therapy (ART) cessation and protection of CD4+ T cells (CAR-) from HIV-induced depletion in humanized mice. Moreover, CAR4 T cells enhanced the in vivo persistence and efficacy of HIV-specific CAR-modified CD8+ T cells expressing the CD28ζ ICD, which alone exhibited poor survival. Collectively, these studies demonstrate that HIV-resistant CAR4 T cells can directly control HIV replication and augment the virus-specific CD8+ T cell response, highlighting the therapeutic potential of engineered CD4+ T cells to engender a functional HIV cure.


Subject(s)
CD28 Antigens/chemistry , CD4-Positive T-Lymphocytes/transplantation , HIV Infections/therapy , HIV/physiology , Inducible T-Cell Co-Stimulator Protein/chemistry , Receptors, Chimeric Antigen/metabolism , Animals , CD28 Antigens/genetics , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Disease Models, Animal , Disease Progression , Drug Resistance, Viral , HIV/immunology , HIV Infections/immunology , HIV-1/immunology , Humans , Immunotherapy, Adoptive , Inducible T-Cell Co-Stimulator Protein/genetics , Mice , Protein Domains , Receptors, Chimeric Antigen/genetics , Treatment Outcome , Virus Replication
2.
PLoS Pathog ; 13(10): e1006613, 2017 Oct.
Article in English | MEDLINE | ID: mdl-29023549

ABSTRACT

HIV is adept at avoiding naturally generated T cell responses; therefore, there is a need to develop HIV-specific T cells with greater potency for use in HIV cure strategies. Starting with a CD4-based chimeric antigen receptor (CAR) that was previously used without toxicity in clinical trials, we optimized the vector backbone, promoter, HIV targeting moiety, and transmembrane and signaling domains to determine which components augmented the ability of T cells to control HIV replication. This re-engineered CAR was at least 50-fold more potent in vitro at controlling HIV replication than the original CD4 CAR, or a TCR-based approach, and substantially better than broadly neutralizing antibody-based CARs. A humanized mouse model of HIV infection demonstrated that T cells expressing optimized CARs were superior at expanding in response to antigen, protecting CD4 T cells from infection, and reducing viral loads compared to T cells expressing the original, clinical trial CAR. Moreover, in a humanized mouse model of HIV treatment, CD4 CAR T cells containing the 4-1BB costimulatory domain controlled HIV spread after ART removal better than analogous CAR T cells containing the CD28 costimulatory domain. Together, these data indicate that potent HIV-specific T cells can be generated using improved CAR design and that CAR T cells could be important components of an HIV cure strategy.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , HIV Infections/therapy , HIV Infections/virology , HIV-1/physiology , Recoverin/immunology , Virus Replication , Antibodies, Neutralizing/immunology , HIV Infections/immunology , Humans , Signal Transduction/physiology
3.
Nat Commun ; 7: 12340, 2016 07 29.
Article in English | MEDLINE | ID: mdl-27471053

ABSTRACT

Antimicrobial peptides are an important component of the molecular arsenal employed by hosts against bacteria. Many bacteria in turn possess pathways that provide protection against these compounds. In Escherichia coli and related bacteria, the PhoQ/PhoP signalling system is a key regulator of this antimicrobial peptide defence. Here we show that treating E. coli with sublethal concentrations of antimicrobial peptides causes cells to filament, and that this division block is controlled by the PhoQ/PhoP system. The filamentation results from increased expression of QueE, an enzyme that is part of a tRNA modification pathway but that, as we show here, also affects cell division. We also find that a functional YFP-QueE fusion localizes to the division septum in filamentous cells, suggesting QueE blocks septation through interaction with the divisome. Regulation of septation by PhoQ/PhoP may protect cells from antimicrobial peptide-induced stress or other conditions associated with high-level stimulation of this signalling system.


Subject(s)
Antimicrobial Cationic Peptides/pharmacology , Escherichia coli/metabolism , Signal Transduction/drug effects , Biosynthetic Pathways/drug effects , Cell Division/drug effects , Cell Division/genetics , Chromosome Segregation/drug effects , Cytoplasm/drug effects , Cytoplasm/metabolism , Escherichia coli/drug effects , Escherichia coli Proteins/metabolism , Gene Deletion , Gene Expression Regulation, Bacterial/drug effects , Models, Biological , Nucleoside Q/biosynthesis , Transcription, Genetic/drug effects
4.
Mol Ther ; 23(7): 1149-1159, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25896251

ABSTRACT

Despite the ability of antiretroviral therapy to minimize human immunodeficiency virus type 1 (HIV-1) replication and increase the duration and quality of patients' lives, the health consequences and financial burden associated with the lifelong treatment regimen render a permanent cure highly attractive. Although T cells play an important role in controlling virus replication, they are themselves targets of HIV-mediated destruction. Direct genetic manipulation of T cells for adoptive cellular therapies could facilitate a functional cure by generating HIV-1-resistant cells, redirecting HIV-1-specific immune responses, or a combination of the two strategies. In contrast to a vaccine approach, which relies on the production and priming of HIV-1-specific lymphocytes within a patient's own body, adoptive T-cell therapy provides an opportunity to customize the therapeutic T cells prior to administration. However, at present, it is unclear how to best engineer T cells so that sustained control over HIV-1 replication can be achieved in the absence of antiretrovirals. This review focuses on T-cell gene-engineering and gene-editing strategies that have been performed in efforts to inhibit HIV-1 replication and highlights the requirements for a successful gene therapy-mediated functional cure.


Subject(s)
Cell Engineering , Cell- and Tissue-Based Therapy , HIV Infections/therapy , T-Lymphocytes/immunology , HIV Infections/virology , HIV-1/genetics , HIV-1/pathogenicity , Humans , T-Lymphocytes/metabolism , Virus Replication/genetics
5.
mBio ; 3(4): e00193-12, 2012.
Article in English | MEDLINE | ID: mdl-22829679

ABSTRACT

UNLABELLED: Enterococcus faecalis is a common Gram-positive commensal bacterium of the metazoan gastrointestinal tract capable of biofilm formation and an opportunistic pathogen of increasing clinical concern. Dogma has held that biofilms are slow-growing structures, often taking days to form mature microcolonies. Here we report that extracellular DNA (eDNA) is an integral structural component of early E. faecalis biofilms (≤4 h postinoculation). Combining cationic dye-based biofilm matrix stabilization techniques with correlative immuno-scanning electron microscopy (SEM) and fluorescent techniques, we demonstrate that--in early E. faecalis biofilms--eDNA localizes to previously undescribed intercellular filamentous structures, as well as to thick mats of extruded extracellular matrix material. Both of these results are consistent with previous reports that early biofilms are exquisitely sensitive to exogenous DNase treatment. High-resolution SEM demonstrates a punctate labeling pattern in both structures, suggesting the presence of an additional, non-DNA constituent. Notably, the previously described fratricidal or lytic mechanism reported as the source of eDNA in older (≥24 h) E. faecalis biofilms does not appear to be at work under these conditions; extensive visual examination by SEM revealed a striking lack of lysed cells, and bulk biochemical assays also support an absence of significant lysis at these early time points. In addition, some cells demonstrated eDNA labeling localized at the septum, suggesting the possibility of DNA secretion from metabolically active cells. Overall, these data are consistent with a model in which a subpopulation of viable E. faecalis cells secrete or extrude DNA into the extracellular matrix. IMPORTANCE: This paper reports the production of extracellular DNA during early biofilm formation in Enterococcus faecalis. The work is significant because the mechanism of eDNA (extracellular DNA) production is independent of cell lysis and the DNA is confined to well-defined structures, suggesting a novel form of DNA secretion by viable cells. Previous models of biofilm formation in enterococci and related species propose cell lysis as the mechanism of DNA release.


Subject(s)
Bacteriolysis , Biofilms , DNA, Bacterial/metabolism , Enterococcus faecalis/physiology , Extracellular Space/metabolism , Bacterial Secretion Systems , DNA, Bacterial/genetics , DNA, Bacterial/ultrastructure , Enterococcus faecalis/genetics , Enterococcus faecalis/ultrastructure , Extracellular Space/genetics , Microscopy, Electron, Scanning
SELECTION OF CITATIONS
SEARCH DETAIL
...