1
|
Cafaro A, Schietroma I, Sernicola L, Belli R, Campagna M, Mancini F, Farcomeni S, Pavone-Cossut MR, Borsetti A, Monini P, Ensoli B. Role of HIV-1 Tat Protein Interactions with Host Receptors in HIV Infection and Pathogenesis. Int J Mol Sci 2024; 25:1704. [PMID: 38338977 PMCID: PMC10855115 DOI: 10.3390/ijms25031704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 01/19/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
Each time the virus starts a new round of expression/replication, even under effective antiretroviral therapy (ART), the transactivator of viral transcription Tat is one of the first HIV-1 protein to be produced, as it is strictly required for HIV replication and spreading. At this stage, most of the Tat protein exits infected cells, accumulates in the extracellular matrix and exerts profound effects on both the virus and neighbor cells, mostly of the innate and adaptive immune systems. Through these effects, extracellular Tat contributes to the acquisition of infection, spreading and progression to AIDS in untreated patients, or to non-AIDS co-morbidities in ART-treated individuals, who experience inflammation and immune activation despite virus suppression. Here, we review the role of extracellular Tat in both the virus life cycle and on cells of the innate and adaptive immune system, and we provide epidemiological and experimental evidence of the importance of targeting Tat to block residual HIV expression and replication. Finally, we briefly review vaccine studies showing that a therapeutic Tat vaccine intensifies ART, while its inclusion in a preventative vaccine may blunt escape from neutralizing antibodies and block early events in HIV acquisition.
Collapse
Affiliation(s)
- Aurelio Cafaro
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, 00161 Rome, Italy; (I.S.); (L.S.); (R.B.); (M.C.); (F.M.); (S.F.); (M.R.P.-C.); (A.B.); (P.M.)
| | | | | | | | | | | | | | | | | | | | - Barbara Ensoli
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, 00161 Rome, Italy; (I.S.); (L.S.); (R.B.); (M.C.); (F.M.); (S.F.); (M.R.P.-C.); (A.B.); (P.M.)
| |
Collapse
|
2
|
Chvatal-Medina M, Lopez-Guzman C, Diaz FJ, Gallego S, Rugeles MT, Taborda NA. Molecular mechanisms by which the HIV-1 latent reservoir is established and therapeutic strategies for its elimination. Arch Virol 2023; 168:218. [PMID: 37530901 DOI: 10.1007/s00705-023-05800-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 04/12/2023] [Indexed: 08/03/2023]
Abstract
The human immunodeficiency virus type 1 (HIV-1) reservoir, composed of cells harboring the latent, integrated virus, is not eliminated by antiretroviral therapy. It therefore represents a significant barrier to curing the infection. The biology of HIV-1 reservoirs, the mechanisms of their persistence, and effective strategies for their eradication are not entirely understood. Here, we review the molecular mechanisms by which HIV-1 reservoirs develop, the cells and compartments where the latent virus resides, and advancements in curative therapeutic strategies. We first introduce statistics and relevant data on HIV-1 infection, aspects of pathogenesis, the role of antiretroviral therapy, and the general features of the latent HIV reservoir. Then, the article is built on three main pillars: The molecular mechanisms related to latency, the different strategies for targeting the reservoir to obtain a cure, and the current progress in immunotherapy to counteract said reservoirs.
Collapse
Affiliation(s)
- Mateo Chvatal-Medina
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Carolina Lopez-Guzman
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Francisco J Diaz
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Salomon Gallego
- Grupo de Investigaciones Biomédicas Uniremington, Programa de Medicina, Facultad de Ciencias de la Salud, Corporación Universitaria Remington, Medellín, Colombia
| | - Maria T Rugeles
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Natalia A Taborda
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia.
- Grupo de Investigaciones Biomédicas Uniremington, Programa de Medicina, Facultad de Ciencias de la Salud, Corporación Universitaria Remington, Medellín, Colombia.
- Universidad Cooperativa de Colombia, Campus Medellin, Envigado, Colombia.
| |
Collapse
|
3
|
Tough Way In, Tough Way Out: The Complex Interplay of Host and Viral Factors in Nucleocytoplasmic Trafficking during HIV-1 Infection. Viruses 2022; 14:v14112503. [PMID: 36423112 PMCID: PMC9696704 DOI: 10.3390/v14112503] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/08/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
Abstract
Human immunodeficiency virus-1 (HIV-1) is a retrovirus that integrates its reverse-transcribed genome as proviral DNA into the host genome to establish a successful infection. The viral genome integration requires safeguarding the subviral complexes, reverse transcription complex (RTC) and preintegration complex (PIC), in the cytosol from degradation, presumably effectively secured by the capsid surrounding these complexes. An intact capsid, however, is a large structure, which raises concerns about its translocation from cytoplasm to nucleus crossing the nuclear membrane, guarded by complex nuclear pore structures, which do not allow non-specific transport of large molecules. In addition, the generation of new virions requires the export of incompletely processed viral RNA from the nucleus to the cytoplasm, an event conventionally not permitted through mammalian nuclear membranes. HIV-1 has evolved multiple mechanisms involving redundant host pathways by liaison with the cell's nucleocytoplasmic trafficking system, failure of which would lead to the collapse of the infection cycle. This review aims to assemble the current developments in temporal and spatial events governing nucleocytoplasmic transport of HIV-1 factors. Discoveries are anticipated to serve as the foundation for devising host-directed therapies involving selective abolishment of the critical interactomes between viral proteins and their host equivalents.
Collapse
|
4
|
Ensoli B, Moretti S, Borsetti A, Maggiorella MT, Buttò S, Picconi O, Tripiciano A, Sgadari C, Monini P, Cafaro A. New insights into pathogenesis point to HIV-1 Tat as a key vaccine target. Arch Virol 2021; 166:2955-2974. [PMID: 34390393 PMCID: PMC8363864 DOI: 10.1007/s00705-021-05158-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 05/09/2021] [Indexed: 02/07/2023]
Abstract
Despite over 30 years of enormous effort and progress in the field, no preventative and/or therapeutic vaccines against human immunodeficiency virus (HIV) are available. Here, we briefly summarize the vaccine strategies and vaccine candidates that in recent years advanced to efficacy trials with mostly unsatisfactory results. Next, we discuss a novel and somewhat contrarian approach based on biological and epidemiological evidence, which led us to choose the HIV protein Tat for the development of preventive and therapeutic HIV vaccines. Toward this goal, we review here the role of Tat in the virus life cycle as well as experimental and epidemiological evidence supporting its key role in the natural history of HIV infection and comorbidities. We then discuss the preclinical and clinical development of a Tat therapeutic vaccine, which, by improving the functionality and homeostasis of the immune system and by reducing the viral reservoir in virologically suppressed vaccinees, helps to establish key determinants for intensification of combination antiretroviral therapy (cART) and a functional cure. Future developments and potential applications of the Tat therapeutic vaccine are also discussed, as well as the rationale for its use in preventative strategies. We hope this contribution will lead to a reconsideration of the current paradigms for the development of HIV/AIDS vaccines, with a focus on targeting of viral proteins with key roles in HIV pathogenesis.
Collapse
Affiliation(s)
- Barbara Ensoli
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy.
| | - Sonia Moretti
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Alessandra Borsetti
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Maria Teresa Maggiorella
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Stefano Buttò
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Orietta Picconi
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Antonella Tripiciano
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Cecilia Sgadari
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Paolo Monini
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Aurelio Cafaro
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| |
Collapse
|
5
|
Tripiciano A, Picconi O, Moretti S, Sgadari C, Cafaro A, Francavilla V, Arancio A, Paniccia G, Campagna M, Pavone-Cossut MR, Sighinolfi L, Latini A, Mercurio VS, Pietro MD, Castelli F, Saracino A, Mussini C, Perri GD, Galli M, Nozza S, Ensoli F, Monini P, Ensoli B. Anti-Tat immunity defines CD4 + T-cell dynamics in people living with HIV on long-term cART. EBioMedicine 2021; 66:103306. [PMID: 33839064 PMCID: PMC8105504 DOI: 10.1016/j.ebiom.2021.103306] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/10/2021] [Accepted: 03/11/2021] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Low-level HIV viremia originating from virus reactivation in HIV reservoirs is often present in cART treated individuals and represents a persisting source of immune stimulation associated with sub-optimal recovery of CD4+ T cells. The HIV-1 Tat protein is released in the extracellular milieu and activates immune cells and latent HIV, leading to virus production and release. However, the relation of anti-Tat immunity with residual viremia, persistent immune activation and CD4+ T-cell dynamics has not yet been defined. METHODS Volunteers enrolled in a 3-year longitudinal observational study were stratified by residual viremia, Tat serostatus and frequency of anti-Tat cellular immune responses. The impact of anti-Tat immunity on low-level viremia, persistent immune activation and CD4+ T-cell recovery was investigated by test for partitions, longitudinal regression analysis for repeated measures and generalized estimating equations. FINDINGS Anti-Tat immunity is significantly associated with higher nadir CD4+ T-cell numbers, control of low-level viremia and long-lasting CD4+ T-cell recovery, but not with decreased immune activation. In adjusted analysis, the extent of CD4+ T-cell restoration reflects the interplay among Tat immunity, residual viremia and immunological determinants including CD8+ T cells and B cells. Anti-Env immunity was not related to CD4+ T-cell recovery. INTERPRETATION Therapeutic approaches aiming at reinforcing anti-Tat immunity should be investigated to improve immune reconstitution in people living with HIV on long-term cART. TRIAL REGISTRATION ISS OBS T-002 ClinicalTrials.gov identifier: NCT01024556 FUNDING: Italian Ministry of Health, special project on the Development of a vaccine against HIV based on the Tat protein and Ricerca Corrente 2019/2020.
Collapse
Affiliation(s)
- Antonella Tripiciano
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome 00161, Italy
| | - Orietta Picconi
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome 00161, Italy
| | - Sonia Moretti
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome 00161, Italy
| | - Cecilia Sgadari
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome 00161, Italy
| | - Aurelio Cafaro
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome 00161, Italy
| | - Vittorio Francavilla
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome 00161, Italy
| | - Angela Arancio
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome 00161, Italy
| | - Giovanni Paniccia
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome 00161, Italy
| | - Massimo Campagna
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome 00161, Italy
| | | | - Laura Sighinolfi
- Unit of Infectious Diseases, University Hospital of Ferrara, Ferrara, Italy
| | - Alessandra Latini
- Unit of Dermatology and Sexually Transmitted Diseases, San Gallicano Institute - Istituti Fisioterapici Ospitalieri (IFO) IRCCS, Rome, Italy
| | - Vito S Mercurio
- Department of Infectious Diseases, S. Maria Goretti Hospital, Latina, Italy
| | - Massimo Di Pietro
- Unit of Infectious Diseases, S.M. Annunziata Hospital, Florence, Italy
| | - Francesco Castelli
- University Division of Infectious and Tropical Diseases, University of Brescia and ASST Spedali Civili, Brescia, Italy
| | - Annalisa Saracino
- Division of Infectious Diseases, Policlinic Hospital, University of Bari, Bari, Italy
| | - Cristina Mussini
- Division of Infectious Diseases, University Policlinic of Modena, Modena, Italy
| | - Giovanni Di Perri
- Clinic of Infectious Diseases, Amedeo di Savoia University Hospital, Turin, Italy
| | - Massimo Galli
- Institute of Tropical and Infectious Diseases, L. Sacco University Hospital, Milan, Italy
| | - Silvia Nozza
- Division of Infectious Diseases, S. Raffaele University Hospital IRCCS, Milan, Italy
| | - Fabrizio Ensoli
- Pathology and Microbiology, San Gallicano Institute - (IFO) IRCCS, Rome, Italy
| | - Paolo Monini
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome 00161, Italy
| | - Barbara Ensoli
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome 00161, Italy.
| |
Collapse
|
6
|
Khan T, Mayuresh Patkar M, Momin M, Omri A. Macrophage targeted nanocarrier delivery systems in HIV therapeutics. Expert Opin Drug Deliv 2020; 17:903-918. [PMID: 32347124 DOI: 10.1080/17425247.2020.1762565] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Human immunodeficiency virus (HIV) targets and modulates the immune system increasing the risk of other associated infections. Highly active antiretroviral therapy (HAART) has significantly improved AIDS-associated morbidity, but has limitations of adverse effects, frequent dosing regimen leading to medical non-adherence. Drug delivery systems that target HIV reservoirs could potentially reduce dose-dependent toxicity and the duration of treatment. The major cellular HIV reservoirs are macrophages and CD4+ T cells with macrophages being responsible for carrying and spreading the virus. The crucial involvement of macrophages in the pathogenesis of HIV infection has led to development of macrophage targeted nanocarrier delivery systems. AREAS COVERED Eradication of viral reservoirs like HIV-infected macrophages has emerged to be a fundamental barrier and challenge for complete eradication of HIV from the immune system. Literature reports several macrophage targeted nanocarrier delivery systems developed as either functionalized or non-functionalized formulations such as liposomes, ethosomes, polymeric nanoparticles, dendrimers, and solid lipid nanoparticles showcasing superior efficacy over the conventional antiretroviral delivery systems. EXPERT OPINION The development of fixed dose combination of antiretroviral drugs into macrophage targeted delivery systems should factor in the inherent plasticity and heterogeneity of macrophages that is dependent on their microenvironment. A rational selection of nanocarriers will facilitate selectivity and enhanced efficacy of antiretroviral drugs accompanied by reduced dosing and toxicity. Such macrophage targeted delivery systems would positively impact the therapeutic outcomes in the management of HIV infection.
Collapse
Affiliation(s)
- Tabassum Khan
- Department of Pharmaceutical Chemistry and Quality Assurance, SVKM's Dr. Bhanuben Nanavati College of Pharmacy , Mumbai, Maharashtra, India
| | - Mayuresh Mayuresh Patkar
- Department of Quality Assurance, SVKM's Dr. Bhanuben Nanavati College of Pharmacy , Mumbai, Maharashtra, India
| | - Munira Momin
- Department of Pharmaceutics, SVKM's Dr. Bhanuben Nanavati College of Pharmacy , Mumbai, Maharashtra, India
| | - Abdelwahab Omri
- The Novel Drug & Vaccine Delivery Systems Facility, Department of Chemistry and Biochemistry, Laurentian University , Sudbury, ON, Canada
| |
Collapse
|
7
|
Ajasin D, Eugenin EA. HIV-1 Tat: Role in Bystander Toxicity. Front Cell Infect Microbiol 2020; 10:61. [PMID: 32158701 PMCID: PMC7052126 DOI: 10.3389/fcimb.2020.00061] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 02/06/2020] [Indexed: 12/21/2022] Open
Abstract
HIV Tat protein is a critical protein that plays multiple roles in HIV pathogenesis. While its role as the transactivator of HIV transcription is well-established, other non-viral replication-associated functions have been described in several HIV-comorbidities even in the current antiretroviral therapy (ART) era. HIV Tat protein is produced and released into the extracellular space from cells with active HIV replication or from latently HIV-infected cells into neighboring uninfected cells even in the absence of active HIV replication and viral production due to effective ART. Neighboring uninfected and HIV-infected cells can take up the released Tat resulting in the upregulation of inflammatory genes and activation of pathways that leads to cytotoxicity observed in several comorbidities such as HIV associated neurocognitive disorder (HAND), HIV associated cardiovascular impairment, and accelerated aging. Thus, understanding how Tat modulates host and viral response is important in designing novel therapeutic approaches to target the chronic inflammatory effects of soluble viral proteins in HIV infection.
Collapse
Affiliation(s)
- David Ajasin
- Department of Neuroscience, Cell Biology, and Anatomy, University of Texas Medical Branch, Galveston, TX, United States
| | - Eliseo A Eugenin
- Department of Neuroscience, Cell Biology, and Anatomy, University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
8
|
Moretti S, Cafaro A, Tripiciano A, Picconi O, Buttò S, Ensoli F, Sgadari C, Monini P, Ensoli B. HIV therapeutic vaccines aimed at intensifying combination antiretroviral therapy. Expert Rev Vaccines 2020; 19:71-84. [PMID: 31957513 DOI: 10.1080/14760584.2020.1712199] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Introduction: Although successful at suppressing HIV replication, combination antiretroviral therapy (cART) only partially restores immune functions and fails to reduce the latent HIV reservoir, thus requiring novel interventions for its intensification.Areas covered: Here are reviewed therapeutic vaccine candidates that are being developed to this goal. Among them, the Tat vaccine has been shown to promote immune restoration, including CD4+ T-cell recovery in low immunological responders, and to reduce the virus reservoirs well beyond what achieved with long-term suppressive cART.Expert opinion: The authors propose the Tat vaccine as a promising vaccine candidate for cART intensification toward HIV reservoirs depletion, functional cure, and eradication strategies, suggesting that targeting a key protein in the virus life cycle is pivotal to success.
Collapse
Affiliation(s)
- Sonia Moretti
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Rome, Italy
| | - Aurelio Cafaro
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Rome, Italy
| | | | - Orietta Picconi
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Rome, Italy
| | - Stefano Buttò
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Rome, Italy
| | - Fabrizio Ensoli
- Pathology and Microbiology, San Gallicano Dermatological Institute IRCCS, Rome, Italy
| | - Cecilia Sgadari
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Rome, Italy
| | - Paolo Monini
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Rome, Italy
| | - Barbara Ensoli
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
9
|
Ahmed I, Witbooi P, Christoffels A. Prediction of human-Bacillus anthracis protein-protein interactions using multi-layer neural network. Bioinformatics 2019; 34:4159-4164. [PMID: 29945178 PMCID: PMC6289132 DOI: 10.1093/bioinformatics/bty504] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Accepted: 06/24/2018] [Indexed: 12/22/2022] Open
Abstract
Motivation Triplet amino acids have successfully been included in feature selection to predict human-HPV protein-protein interactions (PPI). The utility of supervised learning methods is curtailed due to experimental data not being available in sufficient quantities. Improvements in machine learning techniques and features selection will enhance the study of PPI between host and pathogen. Results We present a comparison of a neural network model versus SVM for prediction of host-pathogen PPI based on a combination of features including: amino acid quadruplets, pairwise sequence similarity, and human interactome properties. The neural network and SVM were implemented using Python Sklearn library. The neural network model using quadruplet features and other network features outperformance the SVM model. The models are tested against published predictors and then applied to the human-B.anthracis case. Gene ontology term enrichment analysis identifies immunology response and regulation as functions of interacting proteins. For prediction of Human-viral PPI, our model (neural network) is a significant improvement in overall performance compared to a predictor using the triplets feature and achieves a good accuracy in predicting human-B.anthracis PPI. Availability and implementation All code can be downloaded from ftp://ftp.sanbi.ac.za/machine_learning/. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Ibrahim Ahmed
- South African National Bioinformatics Institute, South African MRC Bioinformatics Unit
| | - Peter Witbooi
- Department of Mathematics and Applied Mathematics, University of the Western Cape, Bellville, South Africa
| | - Alan Christoffels
- South African National Bioinformatics Institute, South African MRC Bioinformatics Unit
| |
Collapse
|
10
|
Cafaro A, Tripiciano A, Picconi O, Sgadari C, Moretti S, Buttò S, Monini P, Ensoli B. Anti-Tat Immunity in HIV-1 Infection: Effects of Naturally Occurring and Vaccine-Induced Antibodies Against Tat on the Course of the Disease. Vaccines (Basel) 2019; 7:vaccines7030099. [PMID: 31454973 PMCID: PMC6789840 DOI: 10.3390/vaccines7030099] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 08/08/2019] [Accepted: 08/15/2019] [Indexed: 02/07/2023] Open
Abstract
HIV-1 Tat is an essential protein in the virus life cycle, which is required for virus gene expression and replication. Most Tat that is produced during infection is released extracellularly and it plays a key role in HIV pathogenesis, including residual disease upon combination antiretroviral therapy (cART). Here, we review epidemiological and experimental evidence showing that antibodies against HIV-1 Tat, infrequently occurring in natural infection, play a protective role against disease progression, and that vaccine targeting Tat can intensify cART. In fact, Tat vaccination of subjects on suppressive cART in Italy and South Africa promoted immune restoration, including CD4+ T-cell increase in low immunological responders, and a reduction of proviral DNA even after six years of cART, when both CD4+ T-cell gain and DNA decay have reached a plateau. Of note, DNA decay was predicted by the neutralization of Tat-mediated entry of Env into dendritic cells by anti-Tat antibodies, which were cross-clade binding and neutralizing. Anti-Tat cellular immunity also contributed to the DNA decay. Based on these data, we propose the Tat therapeutic vaccine as a pathogenesis-driven intervention that effectively intensifies cART and it may lead to a functional cure, providing new perspectives and opportunities also for prevention and virus eradication strategies.
Collapse
Affiliation(s)
- Aurelio Cafaro
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Rome 00161, Italy
| | - Antonella Tripiciano
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Rome 00161, Italy
| | - Orietta Picconi
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Rome 00161, Italy
| | - Cecilia Sgadari
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Rome 00161, Italy
| | - Sonia Moretti
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Rome 00161, Italy
| | - Stefano Buttò
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Rome 00161, Italy
| | - Paolo Monini
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Rome 00161, Italy
| | - Barbara Ensoli
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Rome 00161, Italy.
| |
Collapse
|
11
|
Roebuck KA, Saifuddin M. Regulation of HIV-1 transcription. Gene Expr 2018; 8:67-84. [PMID: 10551796 PMCID: PMC6157391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2023]
Abstract
Human immunodeficiency virus type-1 (HIV-1) is a highly pathogenic lentivirus that requires transcription of its provirus genome for completion of the viral life cycle and the production of progeny virions. Since the first genetic analysis of HIV-1 in 1985, much has been learned about the transcriptional regulation of the HIV-1 genome in infected cells. It has been demonstrated that HIV-1 transcription depends on a varied and complex interaction of host cell transcription factors with the viral long terminal repeat (LTR) promoter. The regulatory elements within the LTR interact with constitutive and inducible transcription factors to direct the assembly of a stable transcription complex that stimulates multiple rounds of transcription by RNA polymerase II (RNAPII). However, the majority of these transcripts terminate prematurely in the absence of the virally encoded trans-activator protein Tat, which stimulates HIV-1 transcription elongation by interacting with a stem-loop RNA element (TAR) formed at the extreme 5' end of all viral transcripts. The Tat-TAR interaction recruits a cellular kinase into the initiation-elongation complex that alters the elongation properties of RNAPII during its transit through TAR. This review summarizes our current knowledge and understanding of the regulation of HIV-1 transcription in infected cells and highlights the important contributions human lentivirus gene regulation has made to our general understanding of the transcription process.
Collapse
Affiliation(s)
- K A Roebuck
- Department of Immunology/Microbiology, Rush Presbyterian St. Luke's Medical Center, Chicago, IL 60612, USA.
| | | |
Collapse
|
12
|
Clark E, Nava B, Caputi M. Tat is a multifunctional viral protein that modulates cellular gene expression and functions. Oncotarget 2018; 8:27569-27581. [PMID: 28187438 PMCID: PMC5432358 DOI: 10.18632/oncotarget.15174] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 01/24/2017] [Indexed: 12/02/2022] Open
Abstract
The human immunodeficiency virus type I (HIV-1) has developed several strategies to condition the host environment to promote viral replication and spread. Viral proteins have evolved to perform multiple functions, aiding in the replication of the viral genome and modulating the cellular response to the infection. Tat is a small, versatile, viral protein that controls transcription of the HIV genome, regulates cellular gene expression and generates a permissive environment for viral replication by altering the immune response and facilitating viral spread to multiple tissues. Studies carried out utilizing biochemical, cellular, and genomic approaches show that the expression and activity of hundreds of genes and multiple molecular networks are modulated by Tat via multiple mechanisms.
Collapse
Affiliation(s)
- Evan Clark
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, USA
| | - Brenda Nava
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, USA
| | - Massimo Caputi
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, USA
| |
Collapse
|
13
|
Rahimian P, He JJ. Exosome-associated release, uptake, and neurotoxicity of HIV-1 Tat protein. J Neurovirol 2016; 22:774-788. [PMID: 27173397 PMCID: PMC5690550 DOI: 10.1007/s13365-016-0451-6] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 04/18/2016] [Accepted: 05/02/2016] [Indexed: 12/17/2022]
Abstract
HIV-1 Tat is an indispensible transactivator for HIV gene transcription and replication. It has been shown to exit cells as a free protein and enter neighboring cells or interact with surface receptors of neighboring cells to regulate gene expression and cell function. In this study, we report, for the first time, exosome-associated Tat release and uptake. Using a HIV-1 LTR-driven luciferase reporter-based cell assay and Western blotting or in combination with exosome inhibitor, OptiPrep gradient fractionation, and exosome depletion, we demonstrated significant presence of HIV-1 Tat in exosomes derived from Tat-expressing primary astrocytes, Tat-transfected U373.MG and 293T, and HIV-infected MT4. We further showed that exosome-associated Tat from Tat-expressing astrocytes was capable of causing neurite shortening and neuron death, further supporting that this new form of extracellular Tat is biologically active. Lastly, we constructed a Tat mutant deleted of its basic domain and determined the role of the basic domain in Tat trafficking into exosomes. Basic domain-deleted Tat exhibited no apparent effects on Tat trafficking into exosomes, while maintained its dominant-negative function in Tat-mediated LTR transactivation. Taken together, these results show a significant fraction of Tat is secreted and present in the form of exosomes and may contribute to the stability of extracellular Tat and broaden the spectrum of its target cells.
Collapse
Affiliation(s)
- Pejman Rahimian
- Department of Cell Biology and Immunology, Graduate School of Biomedical Sciences, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA
| | - Johnny J He
- Department of Cell Biology and Immunology, Graduate School of Biomedical Sciences, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA.
| |
Collapse
|
14
|
Ensoli B, Nchabeleng M, Ensoli F, Tripiciano A, Bellino S, Picconi O, Sgadari C, Longo O, Tavoschi L, Joffe D, Cafaro A, Francavilla V, Moretti S, Pavone Cossut MR, Collacchi B, Arancio A, Paniccia G, Casabianca A, Magnani M, Buttò S, Levendal E, Ndimande JV, Asia B, Pillay Y, Garaci E, Monini P. HIV-Tat immunization induces cross-clade neutralizing antibodies and CD4(+) T cell increases in antiretroviral-treated South African volunteers: a randomized phase II clinical trial. Retrovirology 2016; 13:34. [PMID: 27277839 PMCID: PMC4899930 DOI: 10.1186/s12977-016-0261-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 04/14/2016] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Although combined antiretroviral therapy (cART) has saved millions of lives, it is incapable of full immune reconstitution and virus eradication. The transactivator of transcription (Tat) protein is a key human immunodeficiency virus (HIV) virulence factor required for virus replication and transmission. Tat is expressed and released extracellularly by infected cells also under cART and in this form induces immune dysregulation, and promotes virus reactivation, entry and spreading. Of note, anti-Tat antibodies are rare in natural infection and, when present, correlate with asymptomatic state and reduced disease progression. This suggested that induction of anti-Tat antibodies represents a pathogenesis-driven intervention to block progression and to intensify cART. Indeed Tat-based vaccination was safe, immunogenic and capable of immune restoration in an open-label, randomized phase II clinical trial conducted in 168 cART-treated volunteers in Italy. To assess whether B-clade Tat immunization would be effective also in patients with different genetic background and infecting virus, a phase II trial was conducted in South Africa. METHODS The ISS T-003 was a 48-week randomised, double-blinded, placebo-controlled trial to evaluate immunogenicity (primary endpoint) and safety (secondary endpoint) of B-clade Tat (30 μg) given intradermally, three times at 4-week intervals, in 200 HIV-infected adults on effective cART (randomised 1:1) with CD4(+) T-cell counts ≥200 cells/µL. Study outcomes also included cross-clade anti-Tat antibodies, neutralization, CD4(+) T-cell counts and therapy compliance. RESULTS Immunization was safe and well-tolerated and induced durable, high titers anti-Tat B-clade antibodies in 97 % vaccinees. Anti-Tat antibodies were cross-clade (all vaccinees tested) and neutralized Tat-mediated entry of oligomeric B-clade and C-clade envelope in dendritic cells (24 participants tested). Anti-Tat antibody titers correlated positively with neutralization. Tat vaccination increased CD4(+) T-cell numbers (all participants tested), particularly when baseline levels were still low after years of therapy, and this had a positive correlation with HIV neutralization. Finally, in cART non-compliant patients (24 participants), vaccination contained viral load rebound and maintained CD4(+) T-cell numbers over study entry levels as compared to placebo. CONCLUSIONS The data indicate that Tat vaccination can restore the immune system and induces cross-clade neutralizing anti-Tat antibodies in patients with different genetic backgrounds and infecting viruses, supporting the conduct of phase III studies in South Africa. Trial registration ClinicalTrials.gov NCT01513135, 01/23/2012.
Collapse
Affiliation(s)
- Barbara Ensoli
- National AIDS Center, Istituto Superiore di Sanità, Rome, Italy.
| | | | - Fabrizio Ensoli
- Laboratory of Clinical Pathology and Microbiology, San Gallicano Institute, Istituti Fisioterapici Ospitalieri, Rome, Italy
| | - Antonella Tripiciano
- National AIDS Center, Istituto Superiore di Sanità, Rome, Italy.,Laboratory of Clinical Pathology and Microbiology, San Gallicano Institute, Istituti Fisioterapici Ospitalieri, Rome, Italy
| | - Stefania Bellino
- National AIDS Center, Istituto Superiore di Sanità, Rome, Italy.,National Center for Epidemiology, Surveillance and Health Promotion, Istituto Superiore di Sanità, Rome, Italy
| | - Orietta Picconi
- National AIDS Center, Istituto Superiore di Sanità, Rome, Italy
| | - Cecilia Sgadari
- National AIDS Center, Istituto Superiore di Sanità, Rome, Italy
| | - Olimpia Longo
- National AIDS Center, Istituto Superiore di Sanità, Rome, Italy.,Italian Medicines Agency, Rome, Italy
| | - Lara Tavoschi
- Head Office, National AIDS Center, Istituto Superiore di Sanità, Cape Town, South Africa.,European Center for Disease Prevention and Control, Stockholm, Sweden
| | - Daniel Joffe
- Head Office, National AIDS Center, Istituto Superiore di Sanità, Cape Town, South Africa
| | - Aurelio Cafaro
- National AIDS Center, Istituto Superiore di Sanità, Rome, Italy
| | - Vittorio Francavilla
- National AIDS Center, Istituto Superiore di Sanità, Rome, Italy.,Laboratory of Clinical Pathology and Microbiology, San Gallicano Institute, Istituti Fisioterapici Ospitalieri, Rome, Italy
| | - Sonia Moretti
- National AIDS Center, Istituto Superiore di Sanità, Rome, Italy
| | | | | | - Angela Arancio
- National AIDS Center, Istituto Superiore di Sanità, Rome, Italy.,Laboratory of Clinical Pathology and Microbiology, San Gallicano Institute, Istituti Fisioterapici Ospitalieri, Rome, Italy
| | - Giovanni Paniccia
- National AIDS Center, Istituto Superiore di Sanità, Rome, Italy.,Laboratory of Clinical Pathology and Microbiology, San Gallicano Institute, Istituti Fisioterapici Ospitalieri, Rome, Italy
| | - Anna Casabianca
- Department of Biomolecular Science, University of Urbino, Urbino, Italy
| | - Mauro Magnani
- Department of Biomolecular Science, University of Urbino, Urbino, Italy
| | - Stefano Buttò
- National AIDS Center, Istituto Superiore di Sanità, Rome, Italy
| | - Elise Levendal
- South African Medical Research Council, Cape Town, South Africa.,Health Systems Trust, Cape Town, South Africa
| | | | - Bennett Asia
- National Department of Health, Pretoria, South Africa
| | - Yogan Pillay
- National Department of Health, Pretoria, South Africa
| | - Enrico Garaci
- Istituto Superiore di Sanità, Rome, Italy.,University of Tor Vergata, Rome, Italy
| | - Paolo Monini
- Head Office, National AIDS Center, Istituto Superiore di Sanità, Cape Town, South Africa
| | | |
Collapse
|
15
|
Abstract
A virus protein called Tat plays a dual role in HIV infection by regulating the expression of genes belonging to the virus and genes belonging to the host cells.
Collapse
Affiliation(s)
- Matjaz Barboric
- Department of Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Koh Fujinaga
- Department of Medicine, University of California, San Francisco, San Francisco, United States
| |
Collapse
|
16
|
Reeder JE, Kwak YT, McNamara RP, Forst CV, D'Orso I. HIV Tat controls RNA Polymerase II and the epigenetic landscape to transcriptionally reprogram target immune cells. eLife 2015; 4. [PMID: 26488441 PMCID: PMC4733046 DOI: 10.7554/elife.08955] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 10/20/2015] [Indexed: 12/22/2022] Open
Abstract
HIV encodes Tat, a small protein that facilitates viral transcription by binding an RNA structure (trans-activating RNA [TAR]) formed on nascent viral pre-messenger RNAs. Besides this well-characterized mechanism, Tat appears to modulate cellular transcription, but the target genes and molecular mechanisms remain poorly understood. We report here that Tat uses unexpected regulatory mechanisms to reprogram target immune cells to promote viral replication and rewire pathways beneficial for the virus. Tat functions through master transcriptional regulators bound at promoters and enhancers, rather than through cellular ‘TAR-like’ motifs, to both activate and repress gene sets sharing common functional annotations. Despite the complexity of transcriptional regulatory mechanisms in the cell, Tat precisely controls RNA polymerase II recruitment and pause release to fine-tune the initiation and elongation steps in target genes. We propose that a virus with a limited coding capacity has optimized its genome by evolving a small but ‘multitasking’ protein to simultaneously control viral and cellular transcription. DOI:http://dx.doi.org/10.7554/eLife.08955.001 The human immunodeficiency virus (HIV) reproduces and spreads throughout the body by hijacking human immune cells and causing them to copy the virus’s genetic information. As the virus multiplies, it also causes the death of the immune system cells that help the human body recognize and eliminate viruses. This allows the virus to multiply unchecked. Studies of the genetic material of HIV – which is in the form of single-stranded RNA molecules and contains only a handful of genes – have begun to reveal how the virus can wreak such havoc to the human immune system. A small protein encoded by the virus, called Tat, boosts the expression of HIV genes in infected immune cells by binding to a structure that forms on newly synthesized viral RNAs. Recent evidence suggests that HIV also changes the expression of human genes to make immune cells more hospitable to the virus. However, it was not known exactly which specific genes are targeted, or how the virus alters their expression. Now, Reeder, Kwak et al. reveal how the Tat protein alters the expression of more than 400 human genes. Rather than bind to the same structure seen in newly forming HIV RNAs, Tat turns on or off the expression of its human target genes by interacting with proteins that regulate human gene expression. In doing so, Tat is able to precisely control the activity of an enzyme called RNA Polymerase II that is necessary for the early steps of gene expression. Tat’s multitasking ability – boosting HIV gene expression at the same time as reprogramming human gene expression – helps explain how a virus with so little genetic material of its own can perform such a wide range of activities in infected cells. The work of Reeder, Kwak et al. suggests that Tat reshapes the human genome to position target genes in ways that allow them to be efficiently turned on or off. Future studies will further reveal how Tat accomplishes this genome remodeling during different stages of infection. In addition, further research is also necessary to look closely into the sets of genes targeted by Tat to find patterns of genes that work together to alter cell behavior, and investigate how these new behaviors allow HIV to thrive. DOI:http://dx.doi.org/10.7554/eLife.08955.002
Collapse
Affiliation(s)
- Jonathan E Reeder
- Department of Biological Sciences, University of Texas at Dallas, Richardson, United States.,Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Youn-Tae Kwak
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Ryan P McNamara
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Christian V Forst
- Department of Genetics and Genomic Sciences, Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Iván D'Orso
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, United States
| |
Collapse
|
17
|
Virological Mechanisms in the Coinfection between HIV and HCV. Mediators Inflamm 2015; 2015:320532. [PMID: 26494946 PMCID: PMC4606210 DOI: 10.1155/2015/320532] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 09/01/2015] [Accepted: 09/07/2015] [Indexed: 12/14/2022] Open
Abstract
Due to shared transmission routes, coinfection with Hepatitis C Virus (HCV) is common in patients infected by Human Immunodeficiency Virus (HIV). The immune-pathogenesis of liver disease in HIV/HCV coinfected patients is a multifactorial process. Several studies demonstrated that HIV worsens the course of HCV infection, increasing the risk of cirrhosis and hepatocellular carcinoma. Also, HCV might increase immunological defects due to HIV and risk of comorbidities. A specific cross-talk among HIV and HCV proteins in coinfected patients modulates the natural history, the immune responses, and the life cycle of both viruses. These effects are mediated by immune mechanisms and by a cross-talk between the two viruses which could interfere with host defense mechanisms. In this review, we focus on some virological/immunological mechanisms of the pathogenetic interactions between HIV and HCV in the human host.
Collapse
|
18
|
Cafaro A, Tripiciano A, Sgadari C, Bellino S, Picconi O, Longo O, Francavilla V, Buttò S, Titti F, Monini P, Ensoli F, Ensoli B. Development of a novel AIDS vaccine: the HIV-1 transactivator of transcription protein vaccine. Expert Opin Biol Ther 2015; 15 Suppl 1:S13-29. [PMID: 26096836 DOI: 10.1517/14712598.2015.1021328] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Classical approaches aimed at targeting the HIV-1 envelope as well as other structural viral proteins have largely failed. The HIV-1 transactivator of transcription (Tat) is a key HIV virulence factor, which plays pivotal roles in virus gene expression, replication, transmission and disease progression. Notably, anti-Tat Abs are uncommon in natural infection and, when present, correlate with the asymptomatic state and lead to lower or no disease progression. Hence, targeting Tat represents a pathogenesis-driven intervention. AREAS COVERED Here, we review the rationale and the translational development of a therapeutic vaccine targeting the Tat protein. Preclinical and Phase I studies, Phase II trials with Tat in anti-Tat Ab-negative, virologically suppressed highly active antiretroviral therapy-treated subjects in Italy and South Africa were conducted. The results indicate that Tat-induced immune responses are necessary to restore immune homeostasis, to block the replenishment and to reduce the size of the viral reservoir. Additionally, they may help in establishing key parameters for highly active antiretroviral therapy intensification and a functional cure. EXPERT OPINION We propose the therapeutic setting as the most feasible to speed up the testing and comparison of preventative vaccine candidates, as the distinction lies in the use of the vaccine in uninfected versus infected subjects and not in the vaccine formulation.
Collapse
Affiliation(s)
- Aurelio Cafaro
- Istituto Superiore di Sanità, National AIDS Center , Rome , Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Ensoli F, Cafaro A, Casabianca A, Tripiciano A, Bellino S, Longo O, Francavilla V, Picconi O, Sgadari C, Moretti S, Cossut MRP, Arancio A, Orlandi C, Sernicola L, Maggiorella MT, Paniccia G, Mussini C, Lazzarin A, Sighinolfi L, Palamara G, Gori A, Angarano G, Di Pietro M, Galli M, Mercurio VS, Castelli F, Di Perri G, Monini P, Magnani M, Garaci E, Ensoli B. HIV-1 Tat immunization restores immune homeostasis and attacks the HAART-resistant blood HIV DNA: results of a randomized phase II exploratory clinical trial. Retrovirology 2015; 12:33. [PMID: 25924841 PMCID: PMC4414440 DOI: 10.1186/s12977-015-0151-y] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 02/11/2015] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The phase II multicenter, randomized, open label, therapeutic trial (ISS T-002, Clinicaltrials.gov NCT00751595) was aimed at evaluating the immunogenicity and the safety of the biologically active HIV-1 Tat protein administered at 7.5 or 30 μg, given 3 or 5 times monthly, and at exploring immunological and virological disease biomarkers. The study duration was 48 weeks, however, vaccinees were followed until the last enrolled subject reached the 48 weeks. Reported are final data up to 144 weeks of follow-up. The ISS T-002 trial was conducted in 11 clinical centers in Italy on 168 HIV positive subjects under Highly Active Antiretroviral Therapy (HAART), anti-Tat Antibody (Ab) negative at baseline, with plasma viremia <50 copies/mL in the last 6 months prior to enrollment, and CD4(+) T-cell number ≥200 cells/μL. Subjects from a parallel observational study (ISS OBS T-002, Clinicaltrials.gov NCT0102455) enrolled at the same clinical sites with the same criteria constituted an external reference group to explore biomarkers of disease. RESULTS The vaccine was safe and well tolerated and induced anti-Tat Abs in most patients (79%), with the highest frequency and durability in the Tat 30 μg groups (89%) particularly when given 3 times (92%). Vaccination promoted a durable and significant restoration of T, B, natural killer (NK) cells, and CD4(+) and CD8(+) central memory subsets. Moreover, a significant reduction of blood proviral DNA was seen after week 72, particularly under PI-based regimens and with Tat 30 μg given 3 times (30 μg, 3x), reaching a predicted 70% decay after 3 years from vaccination with a half-life of 88 weeks. This decay was significantly associated with anti-Tat IgM and IgG Abs and neutralization of Tat-mediated entry of oligomeric Env in dendritic cells, which predicted HIV-1 DNA decay. Finally, the 30 μg, 3x group was the only one showing significant increases of NK cells and CD38(+)HLA-DR(+)/CD8(+) T cells, a phenotype associated with increased killing activity in elite controllers. CONCLUSIONS Anti-Tat immune responses are needed to restore immune homeostasis and effective anti-viral responses capable of attacking the virus reservoir. Thus, Tat immunization represents a promising pathogenesis-driven intervention to intensify HAART efficacy.
Collapse
Affiliation(s)
- Fabrizio Ensoli
- Pathology and Microbiology, San Gallicano Institute, Istituti Fisioterapici Ospitalieri, Rome, Italy.
| | - Aurelio Cafaro
- National AIDS Center, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome, 00161, Italy.
| | - Anna Casabianca
- Department of Biomolecular Science, University of Urbino, Urbino, Italy.
| | - Antonella Tripiciano
- Pathology and Microbiology, San Gallicano Institute, Istituti Fisioterapici Ospitalieri, Rome, Italy. .,National AIDS Center, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome, 00161, Italy.
| | - Stefania Bellino
- National AIDS Center, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome, 00161, Italy.
| | - Olimpia Longo
- National AIDS Center, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome, 00161, Italy.
| | - Vittorio Francavilla
- Pathology and Microbiology, San Gallicano Institute, Istituti Fisioterapici Ospitalieri, Rome, Italy. .,National AIDS Center, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome, 00161, Italy.
| | - Orietta Picconi
- National AIDS Center, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome, 00161, Italy.
| | - Cecilia Sgadari
- National AIDS Center, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome, 00161, Italy.
| | - Sonia Moretti
- National AIDS Center, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome, 00161, Italy.
| | - Maria R Pavone Cossut
- National AIDS Center, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome, 00161, Italy.
| | - Angela Arancio
- Pathology and Microbiology, San Gallicano Institute, Istituti Fisioterapici Ospitalieri, Rome, Italy. .,National AIDS Center, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome, 00161, Italy.
| | - Chiara Orlandi
- Department of Biomolecular Science, University of Urbino, Urbino, Italy.
| | - Leonardo Sernicola
- National AIDS Center, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome, 00161, Italy.
| | - Maria T Maggiorella
- National AIDS Center, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome, 00161, Italy.
| | - Giovanni Paniccia
- Pathology and Microbiology, San Gallicano Institute, Istituti Fisioterapici Ospitalieri, Rome, Italy. .,National AIDS Center, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome, 00161, Italy.
| | - Cristina Mussini
- Division of Infectious Diseases, University Policlinic of Modena, Modena, Italy.
| | - Adriano Lazzarin
- Division of Infectious Diseases, S. Raffaele Hospital, Milan, Italy.
| | - Laura Sighinolfi
- Unit of Infectious Diseases, University Hospital of Ferrara, Ferrara, Italy.
| | - Guido Palamara
- Department of Infectious Dermatology, San Gallicano Hospital, Rome, Italy.
| | - Andrea Gori
- Division of Infectious Diseases, San Gerardo Hospital, Monza, Italy.
| | - Gioacchino Angarano
- Division of Infectious Diseases, University of Bari, Policlinic Hospital, Bari, Italy.
| | - Massimo Di Pietro
- Unit of Infectious Diseases, S.M. Annunziata Hospital, Florence, Italy.
| | - Massimo Galli
- Institute of Tropical and Infectious Diseases, L. Sacco Hospital, University of Milan, Milan, Italy.
| | - Vito S Mercurio
- Department of Infectious Diseases, S. Maria Goretti Hospital, Latina, Italy.
| | - Francesco Castelli
- Division of Tropical and Infectious Diseases, Spedali Civili, Brescia, Italy.
| | - Giovanni Di Perri
- Clinic of Infectious Diseases, Amedeo di Savoia Hospital, Turin, Italy.
| | - Paolo Monini
- National AIDS Center, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome, 00161, Italy.
| | - Mauro Magnani
- Department of Biomolecular Science, University of Urbino, Urbino, Italy.
| | - Enrico Garaci
- Istituto Superiore di Sanità, Rome, Italy, present address University of Tor Vergata, Rome, 00173, Italy.
| | - Barbara Ensoli
- National AIDS Center, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome, 00161, Italy.
| |
Collapse
|
20
|
Kumar A, Herbein G. The macrophage: a therapeutic target in HIV-1 infection. MOLECULAR AND CELLULAR THERAPIES 2014; 2:10. [PMID: 26056579 PMCID: PMC4452058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2013] [Accepted: 01/27/2014] [Indexed: 11/21/2023]
Abstract
Human immunodeficiency virus (HIV) is still a serious global health concern responsible for more than 25 million deaths in last three decades. More than 34 million people are living with HIV infection. Macrophages and CD4+ T cells are the principal targets of HIV-1. The pathogenesis of HIV-1 takes different routes in macrophages and CD4+ T cells. Macrophages are resistant to the cytopathic effect of HIV-1 and produce virus for longer periods of time. In addition, macrophages being present in every organ system thus can disseminate virus to the different anatomical sites leading to the formation of viral sanctuaries. Complete cure of HIV-1 needs better understanding of viral pathogenesis in these reservoirs and implementation of knowledge into robust therapeutic products. In this review we will focus on the unique relationship between HIV-1 and macrophages. Furthermore, we will describe how successful antiretroviral therapy (ART) is in suppressing HIV and novel molecular and cellular strategies against HIV-1 in macrophages.
Collapse
Affiliation(s)
- Amit Kumar
- />Department of Virology, UPRES EA4266 Pathogens & Inflammation, University of Franche-Comte, SFR FED 4234, F-25030 Besançon, France
| | - Georges Herbein
- />Department of Virology, UPRES EA4266 Pathogens & Inflammation, University of Franche-Comte, SFR FED 4234, F-25030 Besançon, France
- />Department of Virology, Hôpital Saint-Jacques, CHRU Besançon, 2 place Saint-Jacques, F-25030 Besançon cedex, France
| |
Collapse
|
21
|
Kumar A, Herbein G. The macrophage: a therapeutic target in HIV-1 infection. MOLECULAR AND CELLULAR THERAPIES 2014; 2:10. [PMID: 26056579 PMCID: PMC4452058 DOI: 10.1186/2052-8426-2-10] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2013] [Accepted: 01/27/2014] [Indexed: 12/21/2022]
Abstract
Human immunodeficiency virus (HIV) is still a serious global health concern responsible for more than 25 million deaths in last three decades. More than 34 million people are living with HIV infection. Macrophages and CD4+ T cells are the principal targets of HIV-1. The pathogenesis of HIV-1 takes different routes in macrophages and CD4+ T cells. Macrophages are resistant to the cytopathic effect of HIV-1 and produce virus for longer periods of time. In addition, macrophages being present in every organ system thus can disseminate virus to the different anatomical sites leading to the formation of viral sanctuaries. Complete cure of HIV-1 needs better understanding of viral pathogenesis in these reservoirs and implementation of knowledge into robust therapeutic products. In this review we will focus on the unique relationship between HIV-1 and macrophages. Furthermore, we will describe how successful antiretroviral therapy (ART) is in suppressing HIV and novel molecular and cellular strategies against HIV-1 in macrophages.
Collapse
Affiliation(s)
- Amit Kumar
- Department of Virology, UPRES EA4266 Pathogens & Inflammation, University of Franche-Comte, SFR FED 4234, F-25030 Besançon, France
| | - Georges Herbein
- Department of Virology, UPRES EA4266 Pathogens & Inflammation, University of Franche-Comte, SFR FED 4234, F-25030 Besançon, France ; Department of Virology, Hôpital Saint-Jacques, CHRU Besançon, 2 place Saint-Jacques, F-25030 Besançon cedex, France
| |
Collapse
|
22
|
Ronsard L, Lata S, Singh J, Ramachandran VG, Das S, Banerjea AC. Molecular and genetic characterization of natural HIV-1 Tat Exon-1 variants from North India and their functional implications. PLoS One 2014; 9:e85452. [PMID: 24465566 PMCID: PMC3900424 DOI: 10.1371/journal.pone.0085452] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Accepted: 11/27/2013] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Designing an ideal vaccine against HIV-1 has been difficult due to enormous genetic variability as a result of high replication rate and lack of proofreading activity of reverse transcriptase leading to emergence of genetic variants and recombinants. Tat transactivates HIV-1 LTR, resulting in a remarkable increase in viral gene expression, and plays a vital role in pathogenesis. The aim of this study was to characterize the genetic variations of Tat exon-1 from HIV-1 infected patients from North India. METHODS Genomic DNA was isolated from PBMCs and Tat exon-1 was PCR amplified with specific primers followed by cloning, sequencing and sequence analyses using bioinformatic tools for predicting HIV-1 subtypes, recombination events, conservation of domains and phosphorylation sites, and LTR transactivation by luciferase assay. RESULTS Phylogenetic analysis of Tat exon-1 variants (n = 120) revealed sequence similarity with South African Tat C sequences and distinct geographical relationships were observed for B/C recombinants. Bootscan analysis of our variants showed 90% homology to Tat C and 10% to B/C recombinants with a precise breakpoint. Natural substitutions were observed with high allelic frequencies which may be beneficial for virus. High amino acid conservation was observed in Tat among Anti Retroviral Therapy (ART) recipients. Barring few changes, most of the functional domains, predicted motifs and phosphorylation sites were well conserved in most of Tat variants. dN/dS analysis revealed purifying selection, implying the importance of functional conservation of Tat exon-1. Our Indian Tat C variants and B/C recombinants showed differential LTR transactivation. CONCLUSIONS The possible role of Tat exon-1 variants in shaping the current HIV-1 epidemic in North India was highlighted. Natural substitutions across conserved functional domains were observed and provided evidence for the emergence of B/C recombinants within the ORF of Tat exon-1. These events are likely to have implications for viral pathogenesis and vaccine formulations.
Collapse
Affiliation(s)
- Larance Ronsard
- Virology Laboratory, National Institute of Immunology, New Delhi, India
- Department of Microbiology, University College of Medical Sciences and Guru Teg Bahadur Hospital, Delhi, India
| | - Sneh Lata
- Department of Microbiology, University College of Medical Sciences and Guru Teg Bahadur Hospital, Delhi, India
| | - Jyotsna Singh
- Virology Laboratory, National Institute of Immunology, New Delhi, India
- Department of Microbiology, University College of Medical Sciences and Guru Teg Bahadur Hospital, Delhi, India
| | | | - Shukla Das
- Department of Microbiology, University College of Medical Sciences and Guru Teg Bahadur Hospital, Delhi, India
| | - Akhil C. Banerjea
- Virology Laboratory, National Institute of Immunology, New Delhi, India
- * E-mail: ,
| |
Collapse
|
23
|
HIV-1 Tat protein induces viral internalization through Env-mediated interactions in dose-dependent manner. AIDS 2013; 27:2355-64. [PMID: 23842123 DOI: 10.1097/01.aids.0000432452.83604.59] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
OBJECTIVE To study the dose-dependent manner of HIV-1 Tat-induced effects on viral replication, internalization and spread, and to directly observe these effects on soluble Env immunogens and virus-like particles. DESIGN In order to determine the manner through which Tat affects viral replication, we incubated cells, virions and soluble Env spikes with Tat at different concentrations, and directly visualized the effects of such incubation. METHODS Cell-based infectivity assays were carried out to assay Tat dose-dependency of viral infectivity. Transmission electron microscopy of virus-like particles and soluble gp140 immunogens incubated with Tat at various concentrations was performed to directly observe Tat-induced effects. RESULTS Treating virus with exogenous Tat increased infectivity in a dose-dependent manner. In the presence of anti-Tat antibodies, virus replication and spread were repressed, postulating Tat contributions to disease progression. When CXCR4 coreceptors were blocked, Tat treatment overcame the inhibition relative to absence of Tat treatment. Similarly, syncytium formation between chronically infected and uninfected target cells was also increased by exogenous Tat treatment. Inhibiting the CD4 receptor for virus entry abolished syncytium formation and Tat treatment was unable to overcome CD4 dependency. We show that Tat reduces virus infectivity at higher Tat concentrations through Env interactions resulting in viral aggregation. CONCLUSION Treating virions or chronically infected cells with exogenous Tat could enhance virus infectivity and spread through coreceptor tropism switch or through another undetermined mechanism. The aggregation potential of Tat suggests a mechanism of negative-feedback regulation of viral replication, providing another regulative function to control viral replication.
Collapse
|
24
|
Abbas W, Herbein G. T-Cell Signaling in HIV-1 Infection. Open Virol J 2013; 7:57-71. [PMID: 23986795 PMCID: PMC3751038 DOI: 10.2174/1874357920130621001] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2013] [Revised: 05/31/2013] [Accepted: 06/04/2013] [Indexed: 12/20/2022] Open
Abstract
HIV exploits the T-cell signaling network to gain access to downstream cellular components, which serves as effective tools to break the cellular barriers. Multiple host factors and their interaction with viral proteins contribute to the complexity of HIV-1 pathogenesis and disease progression. HIV-1 proteins gp120, Nef, Tat and Vpr alter the T-cell signaling pathways by activating multiple transcription factors including NF-ĸB, Sp1 and AP-1. HIV-1 evades the immune system by developing a multi-pronged strategy. Additionally, HIV-1 encoded proteins influence the apoptosis in the host cell favoring or blocking T-cell apoptosis. Thus, T-cell signaling hijacked by viral proteins accounts for both viral persistence and immune suppression during HIV-1 infection. Here, we summarize past and present studies on HIV-1 T-cell signaling with special focus on the possible role of T cells in facilitating viral infection and pathogenesis
Collapse
Affiliation(s)
- Wasim Abbas
- Department of Virology, Pathogens & Inflammation Laboratory, UPRES EA4266, SFR FED 4234, University of Franche-Comte, CHRU Besançon, F-25030 Besançon, France
| | | |
Collapse
|
25
|
Abbas W, Herbein G. Plasma membrane signaling in HIV-1 infection. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2013; 1838:1132-42. [PMID: 23806647 DOI: 10.1016/j.bbamem.2013.06.020] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Revised: 06/12/2013] [Accepted: 06/16/2013] [Indexed: 10/26/2022]
Abstract
Plasma membrane is a multifunctional structure that acts as the initial barrier against infection by intracellular pathogens. The productive HIV-1 infection depends upon the initial interaction of virus and host plasma membrane. Immune cells such as CD4+ T cells and macrophages contain essential cell surface receptors and molecules such as CD4, CXCR4, CCR5 and lipid raft components that facilitate HIV-1 entry. From plasma membrane HIV-1 activates signaling pathways that prepare the grounds for viral replication. Through viral proteins HIV-1 hijacks host plasma membrane receptors such as Fas, TNFRs and DR4/DR5, which results in immune evasion and apoptosis both in infected and uninfected bystander cells. These events are hallmark in HIV-1 pathogenesis that leads towards AIDS. The interplay between HIV-1 and plasma membrane signaling has much to offer in terms of viral fitness and pathogenicity, and a better understanding of this interplay may lead to development of new therapeutic approaches. This article is part of a Special Issue entitled: Viral Membrane Proteins - Channels for Cellular Networking.
Collapse
Affiliation(s)
- Wasim Abbas
- Department of Virology, EA 4266 "Pathogens & Inflammation", SFR FED4234, University of Franche-Comte, CHRU Besançon, F-25030 Besançon, France.
| | - Georges Herbein
- Department of Virology, EA 4266 "Pathogens & Inflammation", SFR FED4234, University of Franche-Comte, CHRU Besançon, F-25030 Besançon, France.
| |
Collapse
|
26
|
Monini P, Cafaro A, Srivastava IK, Moretti S, Sharma VA, Andreini C, Chiozzini C, Ferrantelli F, Cossut MRP, Tripiciano A, Nappi F, Longo O, Bellino S, Picconi O, Fanales-Belasio E, Borsetti A, Toschi E, Schiavoni I, Bacigalupo I, Kan E, Sernicola L, Maggiorella MT, Montin K, Porcu M, Leone P, Leone P, Collacchi B, Palladino C, Ridolfi B, Falchi M, Macchia I, Ulmer JB, Buttò S, Sgadari C, Magnani M, Federico MPM, Titti F, Banci L, Dallocchio F, Rappuoli R, Ensoli F, Barnett SW, Garaci E, Ensoli B. HIV-1 tat promotes integrin-mediated HIV transmission to dendritic cells by binding Env spikes and competes neutralization by anti-HIV antibodies. PLoS One 2012; 7:e48781. [PMID: 23152803 PMCID: PMC3496724 DOI: 10.1371/journal.pone.0048781] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Accepted: 10/01/2012] [Indexed: 12/31/2022] Open
Abstract
Use of Env in HIV vaccine development has been disappointing. Here we show that, in the presence of a biologically active Tat subunit vaccine, a trimeric Env protein prevents in monkeys virus spread from the portal of entry to regional lymph nodes. This appears to be due to specific interactions between Tat and Env spikes that form a novel virus entry complex favoring R5 or X4 virus entry and productive infection of dendritic cells (DCs) via an integrin-mediated pathway. These Tat effects do not require Tat-transactivation activity and are blocked by anti-integrin antibodies (Abs). Productive DC infection promoted by Tat is associated with a highly efficient virus transmission to T cells. In the Tat/Env complex the cysteine-rich region of Tat engages the Env V3 loop, whereas the Tat RGD sequence remains free and directs the virus to integrins present on DCs. V2 loop deletion, which unshields the CCR5 binding region of Env, increases Tat/Env complex stability. Of note, binding of Tat to Env abolishes neutralization of Env entry or infection of DCs by anti-HIV sera lacking anti-Tat Abs, which are seldom present in natural infection. This is reversed, and neutralization further enhanced, by HIV sera containing anti-Tat Abs such as those from asymptomatic or Tat-vaccinated patients, or by sera from the Tat/Env vaccinated monkeys. Thus, both anti-Tat and anti-Env Abs are required for efficient HIV neutralization. These data suggest that the Tat/Env interaction increases HIV acquisition and spreading, as a mechanism evolved by the virus to escape anti-Env neutralizing Abs. This may explain the low effectiveness of Env-based vaccines, which are also unlikely to elicit Abs against new Env epitopes exposed by the Tat/Env interaction. As Tat also binds Envs from different clades, new vaccine strategies should exploit the Tat/Env interaction for both preventative and therapeutic interventions.
Collapse
MESH Headings
- AIDS Vaccines/administration & dosage
- AIDS Vaccines/immunology
- Animals
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/metabolism
- Binding Sites
- Dendritic Cells/immunology
- Dendritic Cells/virology
- HIV Antibodies/immunology
- HIV Antibodies/metabolism
- HIV Envelope Protein gp120/immunology
- HIV Envelope Protein gp120/metabolism
- HIV Infections/immunology
- HIV Infections/prevention & control
- HIV Infections/transmission
- HIV Infections/virology
- HIV-1/immunology
- HIV-1/metabolism
- Humans
- Integrins/immunology
- Integrins/metabolism
- Macaca fascicularis
- Male
- Molecular Docking Simulation
- Neutralization Tests
- Oligopeptides/metabolism
- Protein Binding
- Protein Interaction Domains and Motifs/immunology
- Receptors, CCR5/metabolism
- Receptors, CXCR4/metabolism
- Recombinant Proteins/immunology
- Recombinant Proteins/isolation & purification
- Recombinant Proteins/metabolism
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- T-Lymphocytes/virology
- Vaccines, Virus-Like Particle/administration & dosage
- Vaccines, Virus-Like Particle/immunology
- Virus Internalization
- Virus Replication
- env Gene Products, Human Immunodeficiency Virus/chemistry
- env Gene Products, Human Immunodeficiency Virus/immunology
- env Gene Products, Human Immunodeficiency Virus/metabolism
- tat Gene Products, Human Immunodeficiency Virus/chemistry
- tat Gene Products, Human Immunodeficiency Virus/immunology
- tat Gene Products, Human Immunodeficiency Virus/metabolism
Collapse
Affiliation(s)
- Paolo Monini
- National AIDS Center, Istituto Superiore di Sanità, Rome, Italy
| | - Aurelio Cafaro
- National AIDS Center, Istituto Superiore di Sanità, Rome, Italy
| | - Indresh K. Srivastava
- Novartis Vaccines & Diagnostics, Inc., Cambridge, Massachusetts, United States of America
| | - Sonia Moretti
- National AIDS Center, Istituto Superiore di Sanità, Rome, Italy
| | - Victoria A. Sharma
- Novartis Vaccines & Diagnostics, Inc., Cambridge, Massachusetts, United States of America
| | | | | | | | | | - Antonella Tripiciano
- National AIDS Center, Istituto Superiore di Sanità, Rome, Italy
- San Gallicano Hospital, Rome, Italy
| | - Filomena Nappi
- National AIDS Center, Istituto Superiore di Sanità, Rome, Italy
| | - Olimpia Longo
- National AIDS Center, Istituto Superiore di Sanità, Rome, Italy
| | | | - Orietta Picconi
- National AIDS Center, Istituto Superiore di Sanità, Rome, Italy
| | | | | | - Elena Toschi
- National AIDS Center, Istituto Superiore di Sanità, Rome, Italy
| | | | | | - Elaine Kan
- Novartis Vaccines & Diagnostics, Inc., Cambridge, Massachusetts, United States of America
| | | | | | - Katy Montin
- Department of Biochemistry, University of Ferrara, Ferrara, Italy
| | - Marco Porcu
- CERM, University of Florence, Florence, Italy
| | - Patrizia Leone
- National AIDS Center, Istituto Superiore di Sanità, Rome, Italy
| | | | | | | | - Barbara Ridolfi
- National AIDS Center, Istituto Superiore di Sanità, Rome, Italy
| | - Mario Falchi
- National AIDS Center, Istituto Superiore di Sanità, Rome, Italy
| | - Iole Macchia
- National AIDS Center, Istituto Superiore di Sanità, Rome, Italy
| | - Jeffrey B. Ulmer
- Novartis Vaccines & Diagnostics, Inc., Cambridge, Massachusetts, United States of America
| | - Stefano Buttò
- National AIDS Center, Istituto Superiore di Sanità, Rome, Italy
| | - Cecilia Sgadari
- National AIDS Center, Istituto Superiore di Sanità, Rome, Italy
| | - Mauro Magnani
- Department of Biomolecular Science, University of Urbino, Urbino, Italy
| | | | - Fausto Titti
- National AIDS Center, Istituto Superiore di Sanità, Rome, Italy
| | - Lucia Banci
- CERM, University of Florence, Florence, Italy
| | | | | | | | - Susan W. Barnett
- Novartis Vaccines & Diagnostics, Inc., Cambridge, Massachusetts, United States of America
| | - Enrico Garaci
- National AIDS Center, Istituto Superiore di Sanità, Rome, Italy
| | - Barbara Ensoli
- National AIDS Center, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
27
|
The deleterious influence of tenofovir-based therapies on the progression of atherosclerosis in HIV-infected patients. Mediators Inflamm 2012; 2012:372305. [PMID: 22645407 PMCID: PMC3356719 DOI: 10.1155/2012/372305] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2011] [Revised: 01/30/2012] [Accepted: 02/16/2012] [Indexed: 02/02/2023] Open
Abstract
We investigated the potential differential effects of antiretroviral therapies on unbalanced chemokine homeostasis and on the progression of atherosclerosis in HIV-infected patients. A two-year prospective study was performed in 67 consecutive HIV-infected patients initiating antiretroviral therapy with abacavir/lamivudine or tenofovir/emtricitabine. Circulating levels of inflammatory biomarkers, progression of subclinical atherosclerosis and expression levels of selected chemokines genes in circulating leukocytes were assessed. Control subjects showed significantly lower plasma concentrations of CRP, tPA, IL-6, and MCP-1 than HIV-infected patients at a baseline. After two years of followup, the observed decreases in plasma inflammatory biomarker levels were only significant for MCP-1, tPA, and IL-6. The decrease in plasma MCP-1 concentration was associated with the progression of atherosclerosis, and this effect was negligible only in patients receiving TDF-based therapy. Multivariate analysis confirmed that treatment with TDF was positively and significantly associated with a higher likelihood of subclinical atherosclerosis progression. However, the expression levels of selected genes in blood cells only showed associations with the viral load and total and HDL-cholesterol levels. Current antiretroviral treatments may partially attenuate the influence of HIV infection on certain inflammatory pathways, though patients receiving TDF therapy must be carefully monitored with respect to the presence and/or progression of atherosclerosis.
Collapse
|
28
|
Qu J, Zhang Q, Li Y, Liu W, Chen L, Zhu Y, Wu J. The Tat protein of human immunodeficiency virus-1 enhances hepatitis C virus replication through interferon gamma-inducible protein-10. BMC Immunol 2012; 13:15. [PMID: 22471703 PMCID: PMC3350415 DOI: 10.1186/1471-2172-13-15] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2011] [Accepted: 04/03/2012] [Indexed: 01/28/2023] Open
Abstract
Background Co-infection with human immunodeficiency virus-1 (HIV-1) and hepatitis C virus (HCV) is associated with faster progression of liver disease and an increase in HCV persistence. However, the mechanism by which HIV-1 accelerates the progression of HCV liver disease remains unknown. Results HIV-1/HCV co-infection is associated with increased expression of interferon gamma-induced protein-10 (IP-10) mRNA in peripheral blood mononuclear cells (PBMCs). HCV RNA levels were higher in PBMCs of patients with HIV-1/HCV co-infection than in patients with HCV mono-infection. HIV-1 Tat and IP-10 activated HCV replication in a time-dependent manner, and HIV-1 Tat induced IP-10 production. In addition, the effect of HIV-1 Tat on HCV replication was blocked by anti-IP-10 monoclonal antibody, demonstrating that the effect of HIV-1 Tat on HCV replication depends on IP-10. Taken together, these results suggest that HIV-1 Tat protein activates HCV replication by upregulating IP-10 production. Conclusions HIV-1/HCV co-infection is associated with increased expression of IP-10 mRNA and replication of HCV RNA. Furthermore, both HIV-1 Tat and IP-10 activate HCV replication. HIV-1 Tat activates HCV replication by upregulating IP-10 production. These results expand our understanding of HIV-1 in HCV replication and the mechanism involved in the regulation of HCV replication mediated by HIV-1 during co-infection.
Collapse
Affiliation(s)
- Jing Qu
- State Key Laboratory of Virology, College of Life Sciences, and Chinese-French Liver Disease Research Institute at Zhongnan Hospital, Wuhan University, Wuhan 430072, PR China
| | | | | | | | | | | | | |
Collapse
|
29
|
Mediouni S, Watkins JD, Pierres M, Bole A, Loret EP, Baillat G. A monoclonal antibody directed against a conformational epitope of the HIV-1 trans-activator (Tat) protein neutralizes cross-clade. J Biol Chem 2012; 287:11942-50. [PMID: 22362765 DOI: 10.1074/jbc.m111.319863] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The identification of a neutralizing mAb against extracellular HIV-1 transactivator of transcription (Tat) is important for the development of an efficient HIV-1 treatment. Tat plays an essential role in HIV-1 pathogenesis, not only for HIV-1 replication but also as an extracellular toxin able to disrupt the immune system. We showed previously that immunization of rabbits with Tat Oyi, a variant cloned from an African woman who did not develop AIDS following HIV-1 infection, raised antibodies able to recognize different Tat variants. We carried out mice immunization with Tat Oyi and selected a mAb named 7G12, which had the capacity to cross-recognize heterologous Tat variants by a common three-dimensional epitope. These results highlighted that Tat variants were able to acquire a structure, in contrast to a number of studies showing Tat as an unfolded protein. mAb 7G12 also had the capacity to neutralize the biological activities of these Tat variants by blocking the cellular uptake of extracellular Tat. This is the first study using Tat Oyi to produce a mAb able to neutralize effectively activities of extracellular Tats from different HIV-1 subtypes. This mAb has an important potential in therapeutic passive immunization and could help HIV-1 infected patients to restore their immunity.
Collapse
Affiliation(s)
- Sonia Mediouni
- Equipe de Recherche Technologique 2011, Université de la Méditerranée, Faculté de Pharmacie, 27 BD Jean Moulin, 13385 Marseille Cedex 5, France
| | | | | | | | | | | |
Collapse
|
30
|
Ensoli B, Bellino S, Tripiciano A, Longo O, Francavilla V, Marcotullio S, Cafaro A, Picconi O, Paniccia G, Scoglio A, Arancio A, Ariola C, Ruiz Alvarez MJ, Campagna M, Scaramuzzi D, Iori C, Esposito R, Mussini C, Ghinelli F, Sighinolfi L, Palamara G, Latini A, Angarano G, Ladisa N, Soscia F, Mercurio VS, Lazzarin A, Tambussi G, Visintini R, Mazzotta F, Di Pietro M, Galli M, Rusconi S, Carosi G, Torti C, Di Perri G, Bonora S, Ensoli F, Garaci E. Therapeutic immunization with HIV-1 Tat reduces immune activation and loss of regulatory T-cells and improves immune function in subjects on HAART. PLoS One 2010; 5:e13540. [PMID: 21085635 PMCID: PMC2978690 DOI: 10.1371/journal.pone.0013540] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2010] [Accepted: 09/28/2010] [Indexed: 11/18/2022] Open
Abstract
Although HAART suppresses HIV replication, it is often unable to restore immune homeostasis. Consequently, non-AIDS-defining diseases are increasingly seen in treated individuals. This is attributed to persistent virus expression in reservoirs and to cell activation. Of note, in CD4+ T cells and monocyte-macrophages of virologically-suppressed individuals, there is continued expression of multi-spliced transcripts encoding HIV regulatory proteins. Among them, Tat is essential for virus gene expression and replication, either in primary infection or for virus reactivation during HAART, when Tat is expressed, released extracellularly and exerts, on both the virus and the immune system, effects that contribute to disease maintenance. Here we report results of an ad hoc exploratory interim analysis (up to 48 weeks) on 87 virologically-suppressed HAART-treated individuals enrolled in a phase II randomized open-label multicentric clinical trial of therapeutic immunization with Tat (ISS T-002). Eighty-eight virologically-suppressed HAART-treated individuals, enrolled in a parallel prospective observational study at the same sites (ISS OBS T-002), served for intergroup comparison. Immunization with Tat was safe, induced durable immune responses, and modified the pattern of CD4+ and CD8+ cellular activation (CD38 and HLA-DR) together with reduction of biochemical activation markers and persistent increases of regulatory T cells. This was accompanied by a progressive increment of CD4+ T cells and B cells with reduction of CD8+ T cells and NK cells, which were independent from the type of antiretroviral regimen. Increase in central and effector memory and reduction in terminally-differentiated effector memory CD4+ and CD8+ T cells were accompanied by increases of CD4+ and CD8+ T cell responses against Env and recall antigens. Of note, more immune-compromised individuals experienced greater therapeutic effects. In contrast, these changes were opposite, absent or partial in the OBS population. These findings support the use of Tat immunization to intensify HAART efficacy and to restore immune homeostasis.
Collapse
Affiliation(s)
- Barbara Ensoli
- National AIDS Center, Istituto Superiore di Sanità, Rome, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Tansi FL, Blanchard V, Berger M, Tauber R, Reutter W, Fan H. Interaction of human dipeptidyl peptidase IV and human immunodeficiency virus type-1 transcription transactivator in Sf9 cells. Virol J 2010; 7:267. [PMID: 20942971 PMCID: PMC2967539 DOI: 10.1186/1743-422x-7-267] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2010] [Accepted: 10/13/2010] [Indexed: 12/14/2022] Open
Abstract
Background Dipeptidyl peptidase IV (DPPIV) also known as the T cell activation marker CD26 is a multifunctional protein which is involved in various biological processes. The association of human-DPPIV with components of the human immunodeficiency virus type-1 (HIV1) is well documented and raised some discussions. Several reports implicated the interaction of human-DPPIV with the HIV1 transcription transactivator protein (HIV1-Tat) and the inhibition of the dipeptidyl peptidase activity of DPPIV by the HIV1-Tat protein. Furthermore, enzyme kinetic data implied another binding site for the HIV1-Tat other than the active centre of DPPIV. However, the biological significance of this interaction of the HIV1-Tat protein and human-DPPIV has not been studied, yet. Therefore, we focused on the interaction of HIV1-Tat protein with DPPIV and investigated the subsequent biological consequences of this interaction in Spodoptera frugiperda cells, using the BAC-TO-BAC baculovirus system. Results The HIV1-Tat protein (Tat-BRU) co-localized and co-immunoprecipitated with human-DPPIV protein, following co-expression in the baculovirus-driven Sf9 cell expression system. Furthermore, tyrosine phosphorylation of DPPIV protein was up-regulated in Tat/DPPIV-co-expressing cells after 72 h culturing and also in DPPIV-expressing Sf9 cells after application of purified recombinant Tat protein. As opposed to the expression of Tat alone, serine phosphorylation of the Tat protein was decreased when co-expressed with human-DPPIV protein. Conclusions We show for the first time that human-DPPIV and HIV1-Tat co-immunoprecipitate. Furthermore, our findings indicate that the interaction of HIV1-Tat and human-DPPIV may be involved in signalling platforms that regulate the biological function of both human-DPPIV and HIV1-Tat.
Collapse
Affiliation(s)
- Felista L Tansi
- Institut für Biochemie und Molekularbiologie, Charité-Universitätsmedizin Berlin, Arnimallee 22 Berlin-Dahlem, Germany
| | | | | | | | | | | |
Collapse
|
32
|
Cafaro A, Macchia I, Maggiorella MT, Titti F, Ensoli B. Innovative approaches to develop prophylactic and therapeutic vaccines against HIV/AIDS. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 655:189-242. [PMID: 20047043 DOI: 10.1007/978-1-4419-1132-2_14] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The acquired immunodeficiency syndrome (AIDS) emerged in the human population in the summer of 1981. According to the latest United Nations estimates, worldwide over 33 million people are infected with human immunodeficiency virus (HIV) and the prevalence rates continue to rise globally. To control the alarming spread of HIV, an urgent need exists for developing a safe and effective vaccine that prevents individuals from becoming infected or progressing to disease. To be effective, an HIV/AIDS vaccine should induce broad and long-lasting humoral and cellular immune responses, at both mucosal and systemic level. However, the nature of protective immune responses remains largely elusive and this represents one of the major roadblocks preventing the development of an effective vaccine. Here we summarize our present understanding of the factors responsible for resistance to infection or control of progression to disease in human and monkey that may be relevant to vaccine development and briefly review recent approaches which are currently being tested in clinical trials. Finally, the rationale and the current status of novel strategies based on nonstructural HIV-1 proteins, such as Tat, Nef and Rev, used alone or in combination with modified structural HIV-1 Env proteins are discussed.
Collapse
Affiliation(s)
- Aurelio Cafaro
- National AIDS Center, Istituto Superiore di Sanità, V.le Regina Elena 299, 00161, Rome, Italy
| | | | | | | | | |
Collapse
|
33
|
Akhtar LN, Qin H, Muldowney MT, Yanagisawa LL, Kutsch O, Clements JE, Benveniste EN. Suppressor of cytokine signaling 3 inhibits antiviral IFN-beta signaling to enhance HIV-1 replication in macrophages. THE JOURNAL OF IMMUNOLOGY 2010; 185:2393-404. [PMID: 20631305 DOI: 10.4049/jimmunol.0903563] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
HIV-1 replication within macrophages of the CNS often results in cognitive and motor impairment, which is known as HIV-associated dementia (HAD) in its most severe form. IFN-beta suppresses viral replication within these cells during early CNS infection, but the effect is transient. HIV-1 eventually overcomes this protective innate immune response to resume replication through an unknown mechanism, initiating the progression toward HAD. In this article, we show that Suppressor of Cytokine Signaling (SOCS)3, a molecular inhibitor of IFN signaling, may allow HIV-1 to evade innate immunity within the CNS. We found that SOCS3 is elevated in an in vivo SIV/macaque model of HAD and that the pattern of expression correlates with recurrence of viral replication and onset of CNS disease. In vitro, the HIV-1 regulatory protein transactivator of transcription induces SOCS3 in human and murine macrophages in a NF-kappaB-dependent manner. SOCS3 expression attenuates the response of macrophages to IFN-beta at proximal levels of pathway activation and downstream antiviral gene expression and consequently overcomes the inhibitory effect of IFN-beta on HIV-1 replication. These studies indicate that SOCS3 expression, induced by stimuli present in the HIV-1-infected brain, such as transactivator of transcription, inhibits antiviral IFN-beta signaling to enhance HIV-1 replication in macrophages. This consequence of SOCS3 expression in vitro, supported by a correlation with increased viral load and onset of CNS disease in vivo, suggests that SOCS3 may allow HIV-1 to evade the protective innate immune response within the CNS, allowing the recurrence of viral replication and, ultimately, promoting progression toward HAD.
Collapse
|
34
|
Rayne F, Debaisieux S, Yezid H, Lin YL, Mettling C, Konate K, Chazal N, Arold ST, Pugnière M, Sanchez F, Bonhoure A, Briant L, Loret E, Roy C, Beaumelle B. Phosphatidylinositol-(4,5)-bisphosphate enables efficient secretion of HIV-1 Tat by infected T-cells. EMBO J 2010; 29:1348-62. [PMID: 20224549 DOI: 10.1038/emboj.2010.32] [Citation(s) in RCA: 136] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2009] [Accepted: 02/17/2010] [Indexed: 11/09/2022] Open
Abstract
Human immunodeficiency virus type 1 (HIV-1) transcription relies on its transactivating Tat protein. Although devoid of a signal sequence, Tat is released by infected cells and secreted Tat can affect uninfected cells, thereby contributing to HIV-1 pathogenesis. The mechanism and the efficiency of Tat export remained to be documented. Here, we show that, in HIV-1-infected primary CD4(+) T-cells that are the main targets of the virus, Tat accumulates at the plasma membrane because of its specific binding to phosphatidylinositol-4,5-bisphosphate (PI(4,5)P(2)). This interaction is driven by a specific motif of the Tat basic domain that recognizes a single PI(4,5)P(2) molecule and is stabilized by membrane insertion of Tat tryptophan side chain. This original recognition mechanism enables binding to membrane-embedded PI(4,5)P(2) only, but with an unusually high affinity that allows Tat to perturb the PI(4,5)P(2)-mediated recruitment of cellular proteins. Tat-PI(4,5)P(2) interaction is strictly required for Tat secretion, a process that is very efficient, as approximately 2/3 of Tat are exported by HIV-1-infected cells during their lifespan. The function of extracellular Tat in HIV-1 infection might thus be more significant than earlier thought.
Collapse
Affiliation(s)
- Fabienne Rayne
- CPBS, UMR 5236 CNRS, Case 100, Université Montpellier 2, Montpellier, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Nappi F, Chiozzini C, Bordignon V, Borsetti A, Bellino S, Cippitelli M, Barillari G, Caputo A, Tyagi M, Giacca M, Ensoli B. Immobilized HIV-1 Tat protein promotes gene transfer via a transactivation-independent mechanism which requires binding of Tat to viral particles. J Gene Med 2010; 11:955-65. [PMID: 19653251 DOI: 10.1002/jgm.1381] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Retroviral transduction of cells is improved upon virus adsorption onto immobilized fibronectin (FN) fragments. Because HIV-1 Tat possesses the same functional domains that lead to increased transduction efficiency in FN by colocalization of bound virus and cells, we hypothesized that Tat could enhance gene transfer by a similar mechanism. METHODS Single-cycle replication retro- or lentivirus carrying green fluorescent protein or cloramphenicol acetyltransferase as reporter genes were added to wells coated with Tat or Tat peptides. Wells were extensively washed to remove unbound virus and levels of transduction were detected by measuring reporter gene expression. Virus adsorption to immobilized Tat was measured using a p24 antigen capture assay. RESULTS Immobilized Tat efficiently binds retro- and lentiviral particles and mediates virus transmission at virus input doses that were otherwise unable to transduce susceptible cells. Virus adsorption to Tat is not mediated by envelope glycoprotein (Env) because immobilized Tat binds and retains vesicular stomatitis virus G (VSV-G) pseudotypes as well as envelope-free particles. HIV-1 Env or VSV-G are required for Tat-assisted transduction, which is abrogated by an antibody blocking the HIV-1 Env-CD4 interaction. Tat-assisted transduction is mediated by the cysteine-rich region of Tat, which is known to be essential for Tat transactivation activity. However, Tat transactivation is not required for Tat-assisted transduction, as indicated by the enhancement of transduction by transactivation-silent Tat mutants. CONCLUSIONS Immobilized Tat promotes virus transduction by a transactiva- tion-independent mechanism, which requires binding of virus to Tat. Recombinant Tat or Tat fragments provide a new method to increase efficiency of retro- and lentiviral based gene transfer and gene therapy.
Collapse
Affiliation(s)
- Filomena Nappi
- National AIDS Center, Istituto Superiore Sanità, Rome, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Campbell GR, Loret EP, Spector SA. HIV-1 clade B Tat, but not clade C Tat, increases X4 HIV-1 entry into resting but not activated CD4+ T cells. J Biol Chem 2010; 285:1681-91. [PMID: 19917610 PMCID: PMC2804326 DOI: 10.1074/jbc.m109.049957] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2009] [Revised: 11/13/2009] [Indexed: 11/06/2022] Open
Abstract
CXCR4-using human immunodeficiency virus, type 1 (HIV-1) variants emerge late in the course of infection in >40% of individuals infected with clade B HIV-1 but are described less commonly with clade C isolates. Tat is secreted by HIV-1-infected cells where it acts on both uninfected bystander cells and infected cells. In this study, we show that clade B Tat, but not clade C Tat, increases CXCR4 surface expression on resting CD4+ T cells through a CCR2b-dependent mechanism that does not involve de novo protein synthesis. The expression of plectin, a cytolinker protein that plays an important role as a scaffolding platform for proteins involved in cellular signaling including CXCR4 signaling and trafficking, was found to be significantly increased following B Tat but not C Tat treatment. Knockdown of plectin using RNA interference showed that plectin is essential for the B Tat-induced translocation of CXCR4 to the surface of resting CD4+ T cells. The increased surface CXCR4 expression following B Tat treatment led to increased function of CXCR4 including increased chemoattraction toward CXCR4-using-gp120. Moreover, increased CXCR4 surface expression rendered resting CD4+ T cells more permissive to X4 but not R5 HIV-1 infection. However, neither B Tat nor C Tat was able to up-regulate surface expression of CXCR4 on activated CD4+ T cells, and both proteins inhibited the infection of activated CD4+ T cells with X4 but not R5 HIV-1. Thus, B Tat, but not C Tat, has the capacity to render resting, but not activated, CD4+ T cells more susceptible to X4 HIV-1 infection.
Collapse
Affiliation(s)
- Grant R. Campbell
- From the Department of Pediatrics, Division of Infectious Diseases, University of California San Diego, La Jolla, California 92093-0672
| | - Erwann P. Loret
- INSERM U911, Faculté de Pharmacie, Université de la Méditerranée, 13385 Marseille Cedex 5, France
| | - Stephen A. Spector
- From the Department of Pediatrics, Division of Infectious Diseases, University of California San Diego, La Jolla, California 92093-0672
- Rady Children's Hospital, San Diego, California 92123, and
| |
Collapse
|
37
|
Role of Tat protein in HIV neuropathogenesis. Neurotox Res 2009; 16:205-20. [PMID: 19526283 DOI: 10.1007/s12640-009-9047-8] [Citation(s) in RCA: 105] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2009] [Revised: 03/09/2009] [Accepted: 03/09/2009] [Indexed: 12/13/2022]
Abstract
The Tat protein of the human immunodeficiency virus (HIV) has been implicated in the pathophysiology of the neurocognitive deficits associated with HIV infection. This is the earliest protein to be produced by the proviral DNA in the infected cell. The protein not only drives the regulatory regions of the virus but may also be actively released from the cell and then interact with the cell surface receptors of other uninfected cells in the brain leading to cellular dysfunction. It may also be taken up by these cells and can then activate a number of host genes. The Tat protein is highly potent and has the unique ability to travel along neuronal pathways. Importantly, its production is not impacted by the use of antiretroviral drugs once the proviral DNA has been formed. This article reviews the pleomorphic actions of Tat protein and the evidence supporting its central role in the neuropathogenesis of the HIV infection.
Collapse
|
38
|
Gandhi N, Saiyed Z, Thangavel S, Rodriguez J, Rao K, Nair MP. Differential effects of HIV type 1 clade B and clade C Tat protein on expression of proinflammatory and antiinflammatory cytokines by primary monocytes. AIDS Res Hum Retroviruses 2009; 25:691-9. [PMID: 19621989 DOI: 10.1089/aid.2008.0299] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The existence of multiple subtypes of HIV-1 worldwide has created new challenges to control HIV-1 infection and associated neuropathogenesis. Previous studies indicate a difference in neuropathogenic manifestations of HIV-1-associated neuroAIDS between clade B- and clade C-infected subjects with clade B being more neuropathogenic than clade C. However, the exact mechanism underlying the differences in the neuropathogenesis by both the subtypes remains elusive. Development of neuroAIDS is associated with a complex interplay between proinflammatory and antiinflammatory cytokines and chemokines. In the current study, we hypothesize that HIV-1 clade B and C Tat protein exert differential effects on human primary monocytes leading to differences in gene and protein expression of cytokines implicated in neuroAIDS. Primary human monocytes were treated with clade B and clade C Tat protein and quantitative real time PCR was performed to determine gene expression of proinflammatory cytokines (IL-6 and TNF-alpha) and antiinflammatory cytokines (IL-4 and IL-10). Further, cytokine secretion was measured in culture supernatants by ELISA, whereas intracellular cytokine expression was detected by flow cytometry. Results indicate that monocytes treated with Tat B showed significant upregulation of proinflammatory cytokines, IL-6 and TNF-alpha, as compared to Tat C-treated cultures. However, expression of antiinflammatory molecules and IL-4 and IL-10 was found to be higher in Tat C-treated compared to Tat B-treated cultures. Thus, our result shows for the first time that Tat B and Tat C differentially modulate expression of neuropathogenic molecules that may be correlated with the differences in neuroAIDS manifestation induced by clade-specific infections.
Collapse
Affiliation(s)
- Nimisha Gandhi
- Department of Immunology, College of Medicine, Florida International University, Miami, Florida 33199
| | - Zainulabedin Saiyed
- Department of Immunology, College of Medicine, Florida International University, Miami, Florida 33199
| | - Samikkannu Thangavel
- Department of Immunology, College of Medicine, Florida International University, Miami, Florida 33199
| | - Jose Rodriguez
- Department of Microbiology and Immunology, Universidad Central del Caribe, School of Medicine, Bayamon, Puerto Rico 00960
| | - K.V.K. Rao
- Department of Immunology, College of Medicine, Florida International University, Miami, Florida 33199
| | - Madhavan P.N. Nair
- Department of Immunology, College of Medicine, Florida International University, Miami, Florida 33199
| |
Collapse
|
39
|
Florese RH, Demberg T, Xiao P, Kuller L, Larsen K, Summers LE, Venzon D, Cafaro A, Ensoli B, Robert-Guroff M. Contribution of nonneutralizing vaccine-elicited antibody activities to improved protective efficacy in rhesus macaques immunized with Tat/Env compared with multigenic vaccines. THE JOURNAL OF IMMUNOLOGY 2009; 182:3718-27. [PMID: 19265150 DOI: 10.4049/jimmunol.0803115] [Citation(s) in RCA: 116] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Previously, chronic-phase protection against SHIV(89.6P) challenge was significantly greater in macaques primed with replicating adenovirus type 5 host range mutant (Ad5hr) recombinants encoding HIVtat and env and boosted with Tat and Env protein compared with macaques primed with multigenic adenovirus recombinants (HIVtat, HIVenv, SIVgag, SIVnef) and boosted with Tat, Env, and Nef proteins. The greater protection was correlated with Tat- and Env-binding Abs. Because the macaques lacked SHIV(89.6P)-neutralizing activity prechallenge, we investigated whether Ab-dependent cellular cytotoxicity (ADCC) and Ab-dependent cell-mediated viral inhibition (ADCVI) might exert a protective effect. We clearly show that Tat can serve as an ADCC target, although the Tat-specific activity elicited did not correlate with better protection. However, Env-specific ADCC activity was consistently higher in the Tat/Env group, with sustained cell killing postchallenge exhibited at higher levels (p < 0.00001) for a longer duration (p = 0.0002) compared with the multigenic group. ADCVI was similarly higher in the Tat/Env group and significantly correlated with reduced acute-phase viremia at wk 2 and 4 postchallenge (p = 0.046 and 0.011, respectively). Viral-specific IgG and IgA Abs in mucosal secretions were elicited but did not influence the outcome of the i.v. SHIV(89.6P) challenge. The higher ADCC and ADCVI activities seen in the Tat/Env group provide a plausible mechanism responsible for the greater chronic-phase protection. Because Tat is known to enhance cell-mediated immunity to coadministered Ags, further studies should explore its impact on Ab induction so that it may be optimally incorporated into HIV vaccine regimens.
Collapse
|
40
|
Follicular dendritic cells and human immunodeficiency virus type 1 transcription in CD4+ T cells. J Virol 2008; 83:150-8. [PMID: 18971284 DOI: 10.1128/jvi.01652-08] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
HIV replication occurs throughout the natural course of infection in secondary lymphoid tissues and in particular within the germinal centers (GCs), where follicular dendritic cells (FDCs) are adjacent to CD4(+) T cells. Because FDCs provide signaling that increases lymphocyte activation, we postulated that FDCs could increase human immunodeficiency virus (HIV) replication. We cultured HIV-infected CD4(+) T cells alone or with FDCs and measured subsequent virus expression using HIV-p24 production and reverse transcription-PCR analyses. When cultured with FDCs, infected CD4(+) T cells produced almost fourfold more HIV than when cultured alone, and the rate of virus transcription was doubled. Both FDCs and their supernatant increased HIV transcription and resulted in nuclear translocation of NF-kappaB and phosphorylated c-Jun in infected cells. FDCs produced soluble tumor necrosis factor alpha (TNF-alpha) ex vivo, and the addition of a blocking soluble TNF receptor ablated FDC-mediated HIV transcription. Furthermore, TNF-alpha was found highly expressed within GCs, and ex vivo GC CD4(+) T cells supported greater levels of HIV-1 replication than other CD4(+) T cells. These data indicated that FDCs increase HIV transcription and production by a soluble TNF-alpha-mediated mechanism. This FDC-mediated effect may account, at least in part, for the presence of persistent HIV replication in GCs. Therefore, in addition to providing an important reservoir of infectious virus, FDCs increase HIV production, contributing to a tissue microenvironment that is highly conducive to HIV transmission and expression.
Collapse
|
41
|
Lapidot A, Berchanski A, Borkow G. Insight into the mechanisms of aminoglycoside derivatives interaction with HIV-1 entry steps and viral gene transcription. FEBS J 2008; 275:5236-57. [PMID: 18803669 DOI: 10.1111/j.1742-4658.2008.06657.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
In recent years, based on peptide models of HIV-1 RNA binding, NMR structures of Tat-responsive element-ligand complexes and aminoglycoside-RNA interactions, and HIV-1 Tat structure, we have designed and synthesized aminoglycoside-arginine conjugates (AACs) and aminoglycoside poly-arginine conjugates (APACs), to serve as Tat mimetics. These novel molecules inhibit HIV-1 infectivity with 50% effective concentration values in the low micromolar range, the most potent compounds being the hexa-arginine-neomycin B and nona-D-arginine-neomycin conjugates. Importantly, these compounds, in addition to acting as Tat antagonists, inhibit HIV-1 infectivity by blocking several steps in HIV-1 cell entry. The AACs and APACs inhibit HIV-1 cell entry by interacting with gp120 at the CD4-binding site, by interacting with CXCR4 at the binding site of the CXCR4 mAb 12G5, and apparently by interacting with transient structures of the ectodomain of gp41. In the current review, we discuss the mechanisms of anti-HIV-1 activities of these AACs, APACs and other aminoglycoside derivatives in detail. Targeting several key processes in the viral life cycle by the same compound not only may increase its antiviral efficacy, but more importantly, may reduce the capacity of the virus to develop resistance to the compound. AACs and APACs may thus serve as leading compounds for the development of multitargeting novel HIV-1 inhibitors.
Collapse
Affiliation(s)
- Aviva Lapidot
- Department of Organic Chemistry, The Weizmann Institute of Science, Rehovot, Israel.
| | | | | |
Collapse
|
42
|
Hidalgo-Estévez AM, Punzón C, Sanchez-Duffhues G, Muñoz E, Fresno M. HIV-1-Tat potentiates CXCL12/stromal cell-derived factor 1-induced downregulation of membrane CXCR4 in T lymphocytes through protein kinase C zeta. Mol Immunol 2008; 46:106-15. [PMID: 18760839 DOI: 10.1016/j.molimm.2008.07.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2008] [Revised: 07/04/2008] [Accepted: 07/08/2008] [Indexed: 12/25/2022]
Abstract
We have investigated the role of intracellular HIV-1 Tat on CXCR4 expression on T cells. We found that stable or doxycycline-regulated expression of HIV-1 Tat on Jurkat T cells results in lower cell surface expression of CXCR4, but not of other chemokine receptors. This effect was not due to an alteration in CXCR4 transcription, and total CXCR4 levels remained unaltered. Rather, when cells were treated with CXCL12/Stromal Cell-Derived Factor 1, a faster downmodulation of CXCR4 was observed although resurfacing was unaffected. Similar effect was seen in peripheral human T cells transiently transfected with Tat. At the molecular level Tat did not alter cellular levels of G-coupled receptor kinases 2 and 6 and beta-arrestin, proteins involved in CXCR4 downregulation. Neither Tat significantly affected phosphatidylinositol 3-kinase activation in response to CXCL12. Interestingly, in Jurkat cell clones stably expressing both Protein kinase (PK)-Czeta and HIV-1 Tat, CXCL12 induced a faster CXCR4 internalization than in cells only expressing HIV-1 Tat. In contrast in Jurkat cell stably expressing a dominant negative PKCzeta, Tat enhancement of CXCR4 internalization was abrogated. Thus, our results show a new function of HIV-1 Tat, its ability to regulate CXCR4 expression via PKCzeta. The significance of those results is discussed.
Collapse
Affiliation(s)
- Alicia M Hidalgo-Estévez
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Universidad Autónoma de Madrid, c/Nicolás Cabrera, 1. Campus Universitario de Cantoblanco, 28049 Madrid, Spain
| | | | | | | | | |
Collapse
|
43
|
Leghmari K, Contreras X, Moureau C, Bahraoui E. HIV-1 Tat protein induces TNF-alpha and IL-10 production by human macrophages: differential implication of PKC-betaII and -delta isozymes and MAP kinases ERK1/2 and p38. Cell Immunol 2008; 254:46-55. [PMID: 18692180 DOI: 10.1016/j.cellimm.2008.06.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2008] [Revised: 06/09/2008] [Accepted: 06/26/2008] [Indexed: 01/06/2023]
Abstract
In this study, we demonstrate that HIV-1 Tat protein is able to induce IL-10 and TNF-alpha in human macrophages. We show that N-terminal Tat 1-45 fragment initiates the PKC pathway by acting at the membrane. Inhibition of PKC pathway, by chemical inhibitors or after PMA treatment, abolishes both IL-10 and TNF-alpha production. Among the eight PKC isoforms present in macrophages, we show that only PKC-betaIotaIota and -delta are activated by Tat or Tat 1-45 in human macrophages. However, their selective inhibition affects only IL-10 production. Downstream of PKC, Tat activates the MAP kinases p38 and ERK1/2 and the transcription factor NF-kappaB. Using chemical inhibitors we show that (i) both ERK1/2 MAP kinase and NF-kappaB transcription factor play an important role in IL-10 and TNF-alpha production, in macrophages stimulated by Tat. However, p38 MAP kinase seems to be involved only in IL-10 and not TNF-alpha production.
Collapse
Affiliation(s)
- Kaoutar Leghmari
- Laboratoire d'Immuno-Virologie des Lentivirus des Primates, Université Paul Sabatier 118, Toulouse, France
| | | | | | | |
Collapse
|
44
|
Kitaura H, Ohara N, Kobayashi K, Yamada T. TNF-α-mediated multiplication of human immunodeficiency virus in chronically infected monocytoid cells by mycobacterial infection. APMIS 2008. [DOI: 10.1111/j.1600-0463.2001.907807.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
45
|
Dyer MD, Murali TM, Sobral BW. The landscape of human proteins interacting with viruses and other pathogens. PLoS Pathog 2008; 4:e32. [PMID: 18282095 PMCID: PMC2242834 DOI: 10.1371/journal.ppat.0040032] [Citation(s) in RCA: 244] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2007] [Accepted: 01/04/2008] [Indexed: 12/28/2022] Open
Abstract
Infectious diseases result in millions of deaths each year. Mechanisms of infection have been studied in detail for many pathogens. However, many questions are relatively unexplored. What are the properties of human proteins that interact with pathogens? Do pathogens interact with certain functional classes of human proteins? Which infection mechanisms and pathways are commonly triggered by multiple pathogens? In this paper, to our knowledge, we provide the first study of the landscape of human proteins interacting with pathogens. We integrate human-pathogen protein-protein interactions (PPIs) for 190 pathogen strains from seven public databases. Nearly all of the 10,477 human-pathogen PPIs are for viral systems (98.3%), with the majority belonging to the human-HIV system (77.9%). We find that both viral and bacterial pathogens tend to interact with hubs (proteins with many interacting partners) and bottlenecks (proteins that are central to many paths in the network) in the human PPI network. We construct separate sets of human proteins interacting with bacterial pathogens, viral pathogens, and those interacting with multiple bacteria and with multiple viruses. Gene Ontology functions enriched in these sets reveal a number of processes, such as cell cycle regulation, nuclear transport, and immune response that participate in interactions with different pathogens. Our results provide the first global view of strategies used by pathogens to subvert human cellular processes and infect human cells. Supplementary data accompanying this paper is available at http://staff.vbi.vt.edu/dyermd/publications/dyer2008a.html.
Collapse
Affiliation(s)
- Matthew D Dyer
- Genetics, Bioinformatics, and Computational Biology Program, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, United States of America
- Virginia Bioinformatics Institute, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, United States of America
| | - T. M Murali
- Department of Computer Science, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, United States of America
- * To whom correspondence should be addressed. E-mail: (TMM), (BWS)
| | - Bruno W Sobral
- Virginia Bioinformatics Institute, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, United States of America
- * To whom correspondence should be addressed. E-mail: (TMM), (BWS)
| |
Collapse
|
46
|
Florese RH, Wiseman RW, Venzon D, Karl JA, Demberg T, Larsen K, Flanary L, Kalyanaraman VS, Pal R, Titti F, Patterson LJ, Heath MJ, O'Connor DH, Cafaro A, Ensoli B, Robert-Guroff M. Comparative study of Tat vaccine regimens in Mauritian cynomolgus and Indian rhesus macaques: influence of Mauritian MHC haplotypes on susceptibility/resistance to SHIV(89.6P) infection. Vaccine 2008; 26:3312-21. [PMID: 18486283 DOI: 10.1016/j.vaccine.2008.03.100] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2007] [Revised: 03/24/2008] [Accepted: 03/31/2008] [Indexed: 11/15/2022]
Abstract
Protection afforded by HIV Tat-based vaccines has differed in Indian rhesus and Mauritian cynomolgus macaques. We evaluated native Tat and Ad-HIVtat priming/Tat-boosting regimens in both species. Both vaccines were immunogenic. Only the Ad-tat regimen modestly reduced acute viremia in rhesus macaques after SHIV(89.6P) challenge. Confounding variables uncovered in Mauritian macaques included significant associations of susceptibility to infection with MHC class IB and class II H2 and H5 haplotypes, and resistance to infection with class IB haplotypes H3 and H6. Although protection here was limited, Tat-based vaccines incorporating other HIV components have shown greater efficacy. Combination strategies should be further explored.
Collapse
Affiliation(s)
- Ruth H Florese
- Vaccine Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Li JCB, Lau ASY. A role for mitogen-activated protein kinase and Ets-1 in the induction of interleukin-10 transcription by human immunodeficiency virus-1 Tat. Immunology 2007; 121:337-48. [PMID: 17376198 PMCID: PMC2265950 DOI: 10.1111/j.1365-2567.2007.02580.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
The human immunodeficiency virus (HIV) Tat protein has multiple regulatory roles, including trans-activation of the HIV genome and regulation of immune signalling processes, including kinase activation and cytokine expression. We recently demonstrated that HIV-1 Tat induces the expression of interleukin (IL)-10 via p38 mitogen-activated protein kinase (MAPK) activation. We further delineated that the Tat-responsive element of the IL-10 promoter was located within 625 to 595 bp upstream from the transcription start site. Using electrophoretic mobility shift assays, the transcription factors Ets-1 and Sp-1 were shown to bind to the IL-10 promoter to activate transcription of the gene. Furthermore, sequential deletional mutations of the Ets-1- and Sp-1-binding sites in the -625/-595 region reduced the DNA binding and transcription activity of the IL-10 promoter. Our results also showed that both the Tat-induced and Ets-1-regulated IL-10 promoter-driven luciferase activity can be abrogated by inhibitors of the p38 MAPK activity. In conclusion, the coordinated activities of p38 MAPK and the transcription factors, Ets-1 and Sp-1, may play an important role in the HIV-1 Tat-induced IL-10 transcription.
Collapse
Affiliation(s)
- James C B Li
- Department of Paediatrics and Adolescent Medicine and Bio-Screening Unit, Li Ka Shing Faculty of Medicine, Teh University of Hong Kong, Hong Kong, China
| | | |
Collapse
|
48
|
Titti F, Cafaro A, Ferrantelli F, Tripiciano A, Moretti S, Caputo A, Gavioli R, Ensoli F, Robert-Guroff M, Barnett S, Ensoli B. Problems and emerging approaches in HIV/AIDS vaccine development. Expert Opin Emerg Drugs 2007; 12:23-48. [PMID: 17355212 DOI: 10.1517/14728214.12.1.23] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
According to recent estimates, 39.5 million people have been infected with HIV and 2.9 million have already died. The effect of HIV infection on individuals and communities is socially and economically devastating. Although antiretroviral drugs have had a dramatically beneficial impact on HIV-infected individuals who have access to treatment, it has had a negligible impact on the global epidemic. Therefore, the need for an efficacious HIV/AIDS vaccine remains the highest priority of the world HIV/AIDS agenda. The generation of a vaccine against HIV/AIDS has turned out to be extremely challenging, as indicated by > 20 years of unsuccessful attempts. This review discusses the major challenges in the field and key experimental evidence providing a rationale for the use of non-structural HIV proteins, such as Rev, Tat and Nef, either in the native form or expressed by viral vectors such as a replicating adeno-vector. These non-structural proteins alone or in combination with modified structural HIV-1 Env proteins represent a novel strategy for both preventative and therapeutic HIV/AIDS vaccine development.
Collapse
Affiliation(s)
- Fausto Titti
- Istituto Superiore di Sanità, National AIDS Center, V.le Regina Elena 299, Rome 00161, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Devadas K, Boykins RA, Hewlett IK, Wood OL, Clouse KA, Yamada KM, Dhawan S. Antibodies against a multiple-peptide conjugate comprising chemically modified human immunodeficiency virus type-1 functional Tat peptides inhibit infection. Peptides 2007; 28:496-504. [PMID: 17188401 DOI: 10.1016/j.peptides.2006.11.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2006] [Revised: 11/14/2006] [Accepted: 11/15/2006] [Indexed: 11/21/2022]
Abstract
We demonstrated recently that selective side-chain modification of functional cysteine-rich (Tat(21-40)) and arginine-rich (Tat(53-68)) domains of the HIV-1 Tat protein blocks pathogenic activities of these peptides while retaining their immunological characteristics. In the present study, we have synthesized a multiple-peptide conjugate system comprising modified Tat(21-40) and Tat(53-68) peptides (HIV-1-Tat-MPC). Immunization of mice with this highly homogeneous 10.7 kDa HIV-1-Tat-MPC synthetic construct induced an effective immune response in mice. The antibodies generated against HIV-1-Tat-MPC efficiently suppressed Tat-induced viral replication and significantly reduced HIV-associated cytopathic effects in human monocytes. These results indicate that epitope-specific antibodies directed against functional sites of Tat protein using non-pathogenic peptides inhibit HIV pathogenesis. The HIV-1-Tat-MPC, therefore, has potential for the development of a safe, effective, and economical therapeutic vaccine to reduce the progression of HIV infection.
Collapse
Affiliation(s)
- Krishnakumar Devadas
- Immunopathogenesis Section, Laboratory of Molecular Virology, Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, MD 20892, United States
| | | | | | | | | | | | | |
Collapse
|
50
|
Demberg T, Florese RH, Heath MJ, Larsen K, Kalisz I, Kalyanaraman VS, Lee EM, Pal R, Venzon D, Grant R, Patterson LJ, Korioth-Schmitz B, Buzby A, Dombagoda D, Montefiori DC, Letvin NL, Cafaro A, Ensoli B, Robert-Guroff M. A replication-competent adenovirus-human immunodeficiency virus (Ad-HIV) tat and Ad-HIV env priming/Tat and envelope protein boosting regimen elicits enhanced protective efficacy against simian/human immunodeficiency virus SHIV89.6P challenge in rhesus macaques. J Virol 2007; 81:3414-27. [PMID: 17229693 PMCID: PMC1866031 DOI: 10.1128/jvi.02453-06] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
We previously demonstrated that replication-competent adenovirus (Ad)-simian immunodeficiency virus (SIV) recombinant prime/protein boost regimens elicit potent immunogenicity and strong, durable protection of rhesus macaques against SIV(mac251). Additionally, native Tat vaccines have conferred strong protection against simian/human immunodeficiency virus SHIV(89.6P) challenge of cynomolgus monkeys, while native, inactivated, or vectored Tat vaccines have failed to elicit similar protective efficacy in rhesus macaques. Here we asked if priming rhesus macaques with replicating Ad-human immunodeficiency virus (HIV) tat and boosting with the Tat protein would elicit protection against SHIV(89.6P). We also evaluated a Tat/Env regimen, adding an Ad-HIV env recombinant and envelope protein boost to test whether envelope antibodies would augment acute-phase protection. Further, expecting cellular immunity to enhance chronic viremia control, we tested a multigenic group: Ad-HIV tat, -HIV env, -SIV gag, and -SIV nef recombinants and Tat, Env, and Nef proteins. All regimens were immunogenic. A hierarchy was observed in enzyme-linked immunospot responses (with the strongest response for Env, followed by Gag, followed by Nef, followed by Tat) and antibody titers (with the highest titer for Env, followed by Tat, followed by Nef, followed by Gag). Following intravenous SHIV(89.6P) challenge, all macaques became infected. Compared to controls, no protection was seen in the Tat-only group, confirming previous reports for rhesus macaques. However, the multigenic group blunted acute viremia by approximately 1 log (P = 0.017), and both the multigenic and Tat/Env groups reduced chronic viremia by 3 and 4 logs, respectively, compared to controls (multigenic, P = 0.0003; Tat/Env, P < 0.0001). The strikingly greater reduction in the Tat/Env group than in the multigenic group (P = 0.014) was correlated with Tat and Env binding antibodies. Since prechallenge anti-Env antibodies lacked SHIV(89.6P)-neutralizing activity, other functional anti-Env and anti-Tat activities are under investigation, as is a possible synergy between the Tat and Env immunogens.
Collapse
Affiliation(s)
- Thorsten Demberg
- Vaccine Branch, National Cancer Institute/NIH, 41 Medlars Drive, Building 41, Bethesda, MD 20892-5065, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|