151
|
Li W, Lu L, Li W, Jiang S. Small-molecule HIV-1 entry inhibitors targeting gp120 and gp41: a patent review (2010-2015). Expert Opin Ther Pat 2017; 27:707-719. [PMID: 28076686 DOI: 10.1080/13543776.2017.1281249] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION It is essential to discover and develop small-molecule HIV-1 entry inhibitors with suitable pharmaceutical properties. Areas covered: We review the development of small-molecule HIV-1 entry inhibitors as evidenced in patents, patent applications, and related research articles published between 2010 and 2015. Expert opinion: HIV-1 Env gp120 and gp41 are important targets for development of HIV-1 entry inhibitors. The Phe43 pocket in gp120 and the highly conserved hydrophobic pocket on gp41 NHR-trimer are important targets for identification of HIV-1 attachment and fusion inhibitors, respectively. Compounds that bind to Phe43 pocket can block viral gp120 binding to CD4 on T cells, thus inhibiting HIV-1 attachment. However, most compounds targeting Phe43 pocket identified so far are HIV-1 entry agonists with the ability to enhance infectivity of HIV-1 in CD4-negative cells. Therefore, it is essential to identify HIV-1 entry antagonist-based HIV-1 attachment/entry inhibitors. Compounds binding to the gp41 hydrophobic pocket may inhibit CHR binding to the gp41 NHR trimer, thus blocking six-helix bundle formation and gp41-mediated virus-cell fusion. However, most lead compounds targeting this pocket have low potency, possibly because the pocket is too big or too deep. Therefore, it is necessary to identify other pockets in gp41 for developing HIV-1 fusion/entry inhibitors.
Collapse
Affiliation(s)
- Wen Li
- a Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, School of Basic Medical Science , Fudan University , Shanghai , China
| | - Lu Lu
- a Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, School of Basic Medical Science , Fudan University , Shanghai , China
| | - Weihua Li
- b Key Laboratory of Reproduction Regulation of NPFPC, SIPPR, IRD , Fudan University , Shanghai , China
| | - Shibo Jiang
- a Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, School of Basic Medical Science , Fudan University , Shanghai , China.,b Key Laboratory of Reproduction Regulation of NPFPC, SIPPR, IRD , Fudan University , Shanghai , China.,c Lindsley F. Kimball Research Institute , New York Blood Center , New York , NY , USA
| |
Collapse
|
152
|
Martini R, Esposito F, Corona A, Ferrarese R, Ceresola ER, Visconti L, Tintori C, Barbieri A, Calcaterra A, Iovine V, Canducci F, Tramontano E, Botta M. Natural Product Kuwanon-L Inhibits HIV-1 Replication through Multiple Target Binding. Chembiochem 2017; 18:374-377. [PMID: 27992102 DOI: 10.1002/cbic.201600592] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Indexed: 12/30/2022]
Abstract
In recent years many advances have been made in the fight against HIV-1 infection. However, the lack of a vaccine, together with the increasing resistance to the highly active anti-retroviral therapy (HAART), make HIV-1 infection still a serious global emergency. Thus, new compounds with original modes of action are continuously required, and natural products have ever been a very interesting class of pharmacologically active molecules. Some of them have been used since ancient times against viral infections. Here we present a work in which we suggest that kuwanon-L, a natural product active as an HIV-1 integrase (IN) inhibitor, might exert its overall antiviral activity through binding to multiple viral targets. Specific enzymatic tests, together with a time-of-addition (TOA) experiment, support our hypothesis of binding both to IN and to reverse transcriptase (RT). Overall, this compound can be considered an attractive lead for the development of new classes of antiviral agents able to overcome the problem of resistance, due to its ability to exert its action by binding simultaneously to multiple viral targets.
Collapse
Affiliation(s)
- Riccardo Martini
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100, Siena, Italy
| | - Francesca Esposito
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria di Monserrato, SS 554, 09042, Monserrato, Cagliari, Italy
| | - Angela Corona
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria di Monserrato, SS 554, 09042, Monserrato, Cagliari, Italy
| | - Roberto Ferrarese
- Laboratory of Microbiology, San Raffaele Hospital, IRCCS, Via Olgettina 58, 20132, Milan, Italy
| | - Elisa Rita Ceresola
- Department of Biotechnology and Life Sciences, University of Insubria, Via Ravasi 2, 21100, Varese, Italy
| | - Laura Visconti
- Laboratory of Microbiology, San Raffaele Hospital, IRCCS, Via Olgettina 58, 20132, Milan, Italy
| | - Cristina Tintori
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100, Siena, Italy
| | - Alessandro Barbieri
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100, Siena, Italy
| | - Andrea Calcaterra
- Department of Chemistry and Technology of Drugs, University of Rome "Sapienza", Piazzale Aldo Moro 5, 00185, Roma, Italy
| | - Valentina Iovine
- Department of Chemistry and Technology of Drugs, University of Rome "Sapienza", Piazzale Aldo Moro 5, 00185, Roma, Italy
| | - Filippo Canducci
- Laboratory of Microbiology, San Raffaele Hospital, IRCCS, Via Olgettina 58, 20132, Milan, Italy.,Department of Biotechnology and Life Sciences, University of Insubria, Via Ravasi 2, 21100, Varese, Italy
| | - Enzo Tramontano
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria di Monserrato, SS 554, 09042, Monserrato, Cagliari, Italy
| | - Maurizio Botta
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100, Siena, Italy.,Lead Discovery Siena s.r.l., Via Vittorio Alfieri 31, 53019, Castelnuovo Berardenga, Italy
| |
Collapse
|
153
|
Sevenich A, Liu GQ, Arduengo AJ, Gupton BF, Opatz T. Asymmetric One-Pot Synthesis of (3R,3aS,6aR)-Hexahydrofuro[2,3-b]furan-3-ol: A Key Component of Current HIV Protease Inhibitors. J Org Chem 2017; 82:1218-1223. [DOI: 10.1021/acs.joc.6b02588] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Adrian Sevenich
- Institute
of Organic Chemistry, Johannes Gutenberg University, Duesbergweg 10−14, 55128 Mainz, Germany
| | - Gong-Qing Liu
- Institute
of Organic Chemistry, Johannes Gutenberg University, Duesbergweg 10−14, 55128 Mainz, Germany
| | - Anthony J. Arduengo
- Department
of Chemistry, The University of Alabama, Tuscaloosa, Alabama 35487, United States
| | - B. Frank Gupton
- Department
of Chemistry, Virginia Commonwealth University, Richmond, Virginia 23284, United States
| | - Till Opatz
- Institute
of Organic Chemistry, Johannes Gutenberg University, Duesbergweg 10−14, 55128 Mainz, Germany
| |
Collapse
|
154
|
Gosselin A, Wiche Salinas TR, Planas D, Wacleche VS, Zhang Y, Fromentin R, Chomont N, Cohen ÉA, Shacklett B, Mehraj V, Ghali MP, Routy JP, Ancuta P. HIV persists in CCR6+CD4+ T cells from colon and blood during antiretroviral therapy. AIDS 2017; 31:35-48. [PMID: 27835617 PMCID: PMC5131694 DOI: 10.1097/qad.0000000000001309] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 09/07/2016] [Accepted: 10/18/2016] [Indexed: 12/19/2022]
Abstract
OBJECTIVES The objective of this article is to investigate the contribution of colon and blood CD4 T-cell subsets expressing the chemokine receptor CCR6 to HIV persistence during antiretroviral therapy. DESIGN Matched sigmoid biopsies and blood samples (n = 13) as well as leukapheresis (n = 20) were collected from chronically HIV-infected individuals receiving antiretroviral therapy. Subsets of CD4 T cells with distinct differentiation/polarization profiles were identified using surface markers as follows: memory (TM, CD45RA), central memory (TCM; CD45RACCR7), effector (TEM/TM; CD45RACCR7), Th17 (CCR6CCR4), Th1Th17 (CCR6CXCR3), Th1 (CCR6CXCR3), and Th2 (CCR6CCR4). METHODS We used polychromatic flow cytometry for cell sorting, nested real-time PCR for HIV DNA quantification, ELISA and flow cytometry for HIV p24 quantification. HIV reactivation was induced by TCR triggering in the presence/absence of all-trans retinoic acid. RESULTS Compared with blood, the frequency of CCR6 TM was higher in the colon. In both colon and blood compartments, CCR6 TM were significantly enriched in HIV DNA when compared with their CCR6 counterparts (n = 13). In blood, integrated HIV DNA levels were significantly enriched in CCR6 versus CCR6 TCM of four of five individuals and CCR6 versus CCR6 TEM of three of five individuals. Among blood TCM, Th17 and Th1Th17 contributed the most to the pool of cells harboring integrated HIV DNA despite their reduced frequency compared with Th2, which were infected the least. HIV reactivation was induced by TCR triggering and/or retinoic acid exposure at higher levels in CCR6 versus CCR6 TM, TCM, and TEM. CONCLUSION CCR6 is a marker for colon and blood CD4 T cells enriched for replication-competent HIV DNA. Novel eradication strategies should target HIV persistence in CCR6CD4 T cells from various anatomic sites.
Collapse
Affiliation(s)
| | - Tomas Raul Wiche Salinas
- CHUM-Research Centre
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal
| | - Delphine Planas
- CHUM-Research Centre
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal
| | - Vanessa S. Wacleche
- CHUM-Research Centre
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal
| | - Yuwei Zhang
- CHUM-Research Centre
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal
| | | | - Nicolas Chomont
- CHUM-Research Centre
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal
| | - Éric A. Cohen
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal
- Institut de Recherche Clinique de Montréal, Montréal, Québec, Canada
| | | | - Vikram Mehraj
- Chronic Viral Illness Service and Research Institute
| | | | - Jean-Pierre Routy
- Chronic Viral Illness Service and Research Institute
- Division of Hematology, McGill University Health Centre, Montreal, Québec, Canada
| | - Petronela Ancuta
- CHUM-Research Centre
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal
| |
Collapse
|
155
|
Retroviruses and Retroviral Infections. Infect Dis (Lond) 2017. [DOI: 10.1016/b978-0-7020-6285-8.00174-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
156
|
Structure-activity relationship studies on a Trp dendrimer with dual activities against HIV and enterovirus A71. Modifications on the amino acid. Antiviral Res 2016; 139:32-40. [PMID: 28017762 DOI: 10.1016/j.antiviral.2016.12.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 12/13/2016] [Accepted: 12/19/2016] [Indexed: 11/20/2022]
Abstract
We have recently described a new class of dendrimers with tryptophan (Trp) on the surface that show dual antiviral activities against HIV and EV71 enterovirus. The prototype compound of this family is a pentaerythritol derivative with 12 Trps on the periphery. Here we complete the structure-activity relationship studies of this family to identify key features that might be significant for the antiviral activity. With this aim, novel dendrimers containing different amino acids (aromatic and non-aromatic), tryptamine (a "decarboxylated" analogue of Trp) and N-methyl Trp on the periphery have been prepared. Dendrimer with N-Methyl Trp was the most active against HIV-1 and HIV-2 while dendrimer with tyrosine was endowed with the most potent antiviral activity against EV71. This tyrosine dendrimer proved to inhibit a large panel of EV71 clinical isolates (belonging to different clusters) in the low nanomolar/high picomolar range. In addition, a new synthetic procedure (convergent approach) has been developed for the synthesis of the prototype and some other dendrimers. This convergent approach proved more efficient (higher yields, easier purification) than the divergent approach previously reported.
Collapse
|
157
|
Kaushik A, Vabbina PK, Atluri V, Shah P, Vashist A, Jayant RD, Yandart A, Nair M. Electrochemical monitoring-on-chip (E-MoC) of HIV-infection in presence of cocaine and therapeutics. Biosens Bioelectron 2016; 86:426-431. [PMID: 27419908 PMCID: PMC5028277 DOI: 10.1016/j.bios.2016.06.086] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 06/27/2016] [Accepted: 06/29/2016] [Indexed: 10/21/2022]
Abstract
Electrochemical monitoring-on-chip (E-MoC)-based approach for rapid assessment of human immunodeficiency virus (HIV)-infection in the presence of cocaine (Coc) and specific drugs namely i.e., tenofovir (Tef), rimcazole (RA) is demonstrated here, for the first time, using electrochemical impedance spectroscopy (EIS). An in-vitro primary human astrocytes (HA) model was developed using a cultureware chip (CC, used for E-MoC) for HIV-infection, Coc exposure and treatment with anti-HIV drug i.e., Tef, and Coc antagonist i.e., RA. The charge transfer resistance (Rct) value of each CC well varies with respect to infection and treatment demonstrated highly responsive sensitivity of developed chip. The results of E-MoC, a proof-of-the concept, suggested that HIV-infection progression due to Coc ingestion and therapeutic effects of highly specific drugs are measurable on the basis of cell electrophysiology. Though, this work needs various molecular biology-based optimizations to promote this technology as an analytical tool for the rapid assessment of HIV-infection in a patient to manage HIV diseases for timely diagnosis. The presented study is based on using CNS cells and efforts are being made to perform this method using peripheral cells such as monocytes derived dendritic cells.
Collapse
Affiliation(s)
- Ajeet Kaushik
- Center for Personalized Nanomedicine, Institute of NeuroImmune Pharmacology, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA.
| | - Phani Kiran Vabbina
- Department of Electrical and Computer Engineering, Florida International University, Miami, FL, USA
| | - Venkata Atluri
- Center for Personalized Nanomedicine, Institute of NeuroImmune Pharmacology, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Pratikkumar Shah
- Department of Electrical and Computer Engineering, Florida International University, Miami, FL, USA
| | - Arti Vashist
- Center for Personalized Nanomedicine, Institute of NeuroImmune Pharmacology, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Rahul Dev Jayant
- Center for Personalized Nanomedicine, Institute of NeuroImmune Pharmacology, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Adriana Yandart
- Center for Personalized Nanomedicine, Institute of NeuroImmune Pharmacology, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Madhavan Nair
- Center for Personalized Nanomedicine, Institute of NeuroImmune Pharmacology, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA.
| |
Collapse
|
158
|
Poorghasem R, Saberi RS, Shayan M, Mehrgardi MA, Kiani A. Closed Bipolar Electrochemistry for the Detection of Human Immunodeficiency Virus Short Oligonucleotide. Electrochim Acta 2016. [DOI: 10.1016/j.electacta.2016.11.127] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
|
159
|
Symonds G, Bartlett JS, Kiem HP, Tsie M, Breton L. Cell-Delivered Entry Inhibitors for HIV-1: CCR5 Downregulation and Blocking Virus/Membrane Fusion in Defending the Host Cell Population. AIDS Patient Care STDS 2016; 30:545-550. [PMID: 27905841 DOI: 10.1089/apc.2016.0245] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
HIV-1 infection requires the presence of the CD4 receptor on the target cell surface and a coreceptor, predominantly CC-chemokine receptor 5 (CCR5). It has been shown that individuals who are homozygous for a defective CCR5 gene are protected from HIV-1 infection. A novel self-inactivating lentiviral vector LVsh5/C46 (Cal-1) has been engineered to block HIV-1 infection with two viral entry inhibitors, conferring resistance to HIV-1 infection from both CCR5 and CXCR4 tropic strains. Cal-1 encodes a short hairpin RNA (sh5) to downregulate CCR5 and C46, an HIV-1 fusion inhibitor. Gene therapy by Cal-1 is aimed at transducing CD4+ T cells and CD34+ hematopoietic stem/progenitor cells in an autologous transplant setting. Pre-clinical safety and efficacy studies in vitro and in vivo (humanized mouse model and nonhuman primates) have shown that Cal-1 is safe with no indication of any toxicity risk and acts to decrease viral load and increase CD4 counts. Two clinical trials are underway using Cal-1: a phase I/II study to assess safety and feasibility in an adult HIV-1-positive population not on antiretroviral therapy (ART); and a second Fred Hutchinson Investigator Initiated phase I study to assess safety and feasibility in adults with HIV-1-associated non-Hodgkin or Hodgkin lymphoma.
Collapse
|
160
|
Lin Z, Cantone J, Lu H, Nowicka-Sans B, Protack T, Yuan T, Yang H, Liu Z, Drexler D, Regueiro-Ren A, Meanwell NA, Cockett M, Krystal M, Lataillade M, Dicker IB. Mechanistic Studies and Modeling Reveal the Origin of Differential Inhibition of Gag Polymorphic Viruses by HIV-1 Maturation Inhibitors. PLoS Pathog 2016; 12:e1005990. [PMID: 27893830 PMCID: PMC5125710 DOI: 10.1371/journal.ppat.1005990] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 10/11/2016] [Indexed: 12/27/2022] Open
Abstract
HIV-1 maturation inhibitors (MIs) disrupt the final step in the HIV-1 protease-mediated cleavage of the Gag polyprotein between capsid p24 capsid (CA) and spacer peptide 1 (SP1), leading to the production of infectious virus. BMS-955176 is a second generation MI with improved antiviral activity toward polymorphic Gag variants compared to a first generation MI bevirimat (BVM). The underlying mechanistic reasons for the differences in polymorphic coverage were studied using antiviral assays, an LC/MS assay that quantitatively characterizes CA/SP1 cleavage kinetics of virus like particles (VLPs) and a radiolabel binding assay to determine VLP/MI affinities and dissociation kinetics. Antiviral assay data indicates that BVM does not achieve 100% inhibition of certain polymorphs, even at saturating concentrations. This results in the breakthrough of infectious virus (partial antagonism) regardless of BVM concentration. Reduced maximal percent inhibition (MPI) values for BVM correlated with elevated EC50 values, while rates of HIV-1 protease cleavage at CA/SP1 correlated inversely with the ability of BVM to inhibit HIV-1 Gag polymorphic viruses: genotypes with more rapid CA/SP1 cleavage kinetics were less sensitive to BVM. In vitro inhibition of wild type VLP CA/SP1 cleavage by BVM was not maintained at longer cleavage times. BMS-955176 exhibited greatly improved MPI against polymorphic Gag viruses, binds to Gag polymorphs with higher affinity/longer dissociation half-lives and exhibits greater time-independent inhibition of CA/SP1 cleavage compared to BVM. Virological (MPI) and biochemical (CA/SP1 cleavage rates, MI-specific Gag affinities) data were used to create an integrated semi-quantitative model that quantifies CA/SP1 cleavage rates as a function of both MI and Gag polymorph. The model outputs are in accord with in vitro antiviral observations and correlate with observed in vivo MI efficacies. Overall, these findings may be useful to further understand antiviral profiles and clinical responses of MIs at a basic level, potentially facilitating further improvements to MI potency and coverage.
Collapse
Affiliation(s)
- Zeyu Lin
- Departments of Virology, Bristol-Myers Squibb Research & Development, Wallingford, Connecticut, United States of America
| | - Joseph Cantone
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Research & Development, Wallingford, Connecticut, United States of America
| | - Hao Lu
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Research & Development, Wallingford, Connecticut, United States of America
| | - Beata Nowicka-Sans
- Departments of Virology, Bristol-Myers Squibb Research & Development, Wallingford, Connecticut, United States of America
| | - Tricia Protack
- Departments of Virology, Bristol-Myers Squibb Research & Development, Wallingford, Connecticut, United States of America
| | - Tian Yuan
- Discovery Chemistry Platforms, Princeton, New Jersey, United States of America
| | - Hong Yang
- Discovery Chemistry Platforms, Princeton, New Jersey, United States of America
| | - Zheng Liu
- Discovery Chemistry, Bristol-Myers Squibb Research & Development, Wallingford, Connecticut, United States of America
| | - Dieter Drexler
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Research & Development, Wallingford, Connecticut, United States of America
| | - Alicia Regueiro-Ren
- Discovery Chemistry, Bristol-Myers Squibb Research & Development, Wallingford, Connecticut, United States of America
| | - Nicholas A. Meanwell
- Discovery Chemistry, Bristol-Myers Squibb Research & Development, Wallingford, Connecticut, United States of America
| | - Mark Cockett
- Departments of Virology, Bristol-Myers Squibb Research & Development, Wallingford, Connecticut, United States of America
| | - Mark Krystal
- Departments of Virology, Bristol-Myers Squibb Research & Development, Wallingford, Connecticut, United States of America
| | - Max Lataillade
- Global Clinical Development, Bristol-Myers Squibb Research & Development, Wallingford, Connecticut, United States of America
| | - Ira B. Dicker
- Departments of Virology, Bristol-Myers Squibb Research & Development, Wallingford, Connecticut, United States of America
- * E-mail: ,
| |
Collapse
|
161
|
Fuchs SP, Desrosiers RC. Promise and problems associated with the use of recombinant AAV for the delivery of anti-HIV antibodies. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2016; 3:16068. [PMID: 28197421 PMCID: PMC5289440 DOI: 10.1038/mtm.2016.68] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 09/11/2016] [Indexed: 02/07/2023]
Abstract
Attempts to elicit antibodies with potent neutralizing activity against a broad range of human immunodeficiency virus (HIV) isolates have so far proven unsuccessful. Long-term delivery of monoclonal antibodies (mAbs) with such activity is a creative alternative that circumvents the need for an immune response and has the potential for creating a long-lasting sterilizing barrier against HIV. This approach is made possible by an incredible array of potent broadly neutralizing antibodies (bnAbs) that have been identified over the last several years. Recombinant adeno-associated virus (rAAV) vectors are ideally suited for long-term delivery for a variety of reasons. The only products made from rAAV are derived from the transgenes that are put into it; as long as those products are not viewed as foreign, expression from muscle tissue may continue for decades. Thus, use of rAAV to achieve long-term delivery of anti-HIV mAbs with potent neutralizing activity against a broad range of HIV-1 isolates is emerging as a promising concept for the prevention or treatment of HIV-1 infection in humans. Experiments in mice and monkeys that have demonstrated protective efficacy against AIDS virus infection have raised hopes for the promise of this approach. However, all published experiments in monkeys have encountered unwanted immune responses to the AAV-delivered antibody, and these immune responses appear to limit the levels of delivered antibody that can be achieved. In this review, we highlight the promise of rAAV-mediated antibody delivery for the prevention or treatment of HIV infection in humans, but we also discuss the obstacles that will need to be understood and solved in order for the promise of this approach to be realized.
Collapse
Affiliation(s)
- Sebastian P Fuchs
- Department of Pathology, Miller School of Medicine, University of Miami, Miami, Florida, USA; Institut für Klinische und Molekulare Virologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Ronald C Desrosiers
- Department of Pathology, Miller School of Medicine, University of Miami , Miami, Florida, USA
| |
Collapse
|
162
|
Rossi FW, Prevete N, Rivellese F, Lobasso A, Napolitano F, Granata F, Selleri C, de Paulis A. HIV-1 Nef promotes migration and chemokine synthesis of human basophils and mast cells through the interaction with CXCR4. Clin Mol Allergy 2016; 14:15. [PMID: 27822141 PMCID: PMC5088669 DOI: 10.1186/s12948-016-0052-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 10/25/2016] [Indexed: 01/03/2023] Open
Abstract
Background The Nef protein can be detected in plasma of HIV-1-infected patients and plays a role in the pathogenesis of HIV-1. Nef produced during the early stages of infection is fundamental in creating the ideal environment for viral replication, e.g. by reducing the ability of infected cells to induce an immune response. Aim Based on previous experience showing that both Tat and gp41 of HIV-1 are potent chemotactic factors for basophils and mast cells, and gp120 is a powerful stimulus for the release of histamine and cytokines (IL-4 and IL-13) from basophils, in this study we aimed to verify if the HIV Nef protein can exert some effects on basophils and mast cells purified from healthy volunteers through the interaction with the CXCL12 receptor, CXCR4. Methods Basophils purified from peripheral blood cells of 30 healthy volunteers and mast cells obtained from lung tissue of ten healthy volunteers were tested by flow cytometric analysis, chemotaxis and chemokine production by ELISA assays. Results Nef is a potent chemoattractant for basophils and lung mast cells obtained from healthy, HIV-1 and HIV-2 seronegative individuals. Incubation of basophils and mast cells with Nef induces the release of chemokines (CXCL8/IL-8 and CCL3/MIP-1α). The chemotactic activity of Nef on basophils and mast cells is mediated by the interaction with CXCR4 receptors, being blocked by preincubation of FcεRI+ cells with an anti-CXCR4 Ab. Stimulation with Nef or CXCL12/SDF-1α, a CXCR4 ligand, desensitizes basophils to a subsequent challenge with an autologous or heterologous stimulus. Conclusions These results indicate that Nef, a HIV-1-encoded α-chemokine homolog protein, plays a direct role in basophils and mast cell recruitment and activation at sites of HIV-1 replication, by promoting directional migration of human FcεRI+ cells and the release of chemokines from these cells. Together with our previous results, these data suggest that FcεRI+ cells contribute to the dysregulation of the immune system in HIV-1 infection.
Collapse
Affiliation(s)
- Francesca Wanda Rossi
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy
| | - Nella Prevete
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy
| | - Felice Rivellese
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy ; Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Antonio Lobasso
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy
| | - Filomena Napolitano
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy
| | - Francescopaolo Granata
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy
| | - Carmine Selleri
- Hematology Branch, Department of Medicine, University of Salerno, Salerno, Italy
| | - Amato de Paulis
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy
| |
Collapse
|
163
|
Kirtane AR, Langer R, Traverso G. Past, Present, and Future Drug Delivery Systems for Antiretrovirals. J Pharm Sci 2016; 105:3471-3482. [PMID: 27771050 DOI: 10.1016/j.xphs.2016.09.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 09/06/2016] [Accepted: 09/15/2016] [Indexed: 10/20/2022]
Abstract
The human immunodeficiency virus has infected millions of people and the epidemic continues to grow rapidly in some parts of the world. Antiretroviral (ARV) therapy has provided improved treatment and prolonged the life expectancy of patients. Moreover, there is growing interest in using ARVs to protect against new infections. Hence, ARVs have emerged as our primary strategy in combating the virus. Unfortunately, several challenges limit the optimal performance of these drugs. First, ARVs often require life-long use and complex dosing regimens. This results in low patient adherence and periods of lapsed treatment manifesting in drug resistance. This has prompted the development of alternate dosage forms such as vaginal rings and long-acting injectables that stand to improve patient adherence. Another problem central to therapeutic failure is the inadequate penetration of drugs into infected tissues. This can lead to incomplete treatment, development of resistance, and viral rebound. Several strategies have been developed to improve drug penetration into these drug-free sanctuaries. These include encapsulation of drugs in nanoparticles, use of pharmacokinetic enhancers, and cell-based drug delivery platforms. In this review, we discuss issues surrounding ARV therapy and their impact on drug efficacy. We also describe various drug delivery-based approaches developed to overcome these issues.
Collapse
Affiliation(s)
- Ameya R Kirtane
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139; The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Robert Langer
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139; The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139; Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139.
| | - Giovanni Traverso
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139; The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139; Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115.
| |
Collapse
|
164
|
Pleiotropic properties of ASK1. Biochim Biophys Acta Gen Subj 2016; 1861:3030-3038. [PMID: 27693599 DOI: 10.1016/j.bbagen.2016.09.028] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 09/16/2016] [Accepted: 09/27/2016] [Indexed: 12/22/2022]
Abstract
BACKGROUND Apoptosis signal-regulating kinase 1 (ASK1), also known as mitogen-activated protein kinase kinase kinase 5 (MAP3K5), has the potential to induce cellular apoptosis under various physiological conditions. It has long been suggested that ASK1 is highly sensitive to oxidative stress and contributes substantially to apoptosis. However, recent studies have indicated that ASK1 has pleiotropic roles in living organisms through other mechanisms in addition to apoptosis. SCOPE OF THE REVIEW This review describes the physiological functions of ASK1 in living organisms, focusing on the regulatory mechanisms of ASK1 activity and its importance in the pathogenesis of various diseases. We also highlight recent works published within the past few years. MAJOR CONCLUSIONS ASK1 forms a high-molecular-mass complex within the cell, designated as the ASK1 signalosome. Soon after the discovery of ASK1, several regulatory components of the ASK1 signalosome have been revealed, including thioredoxin (Trx), tumor-necrosis factor α receptor-associated factors (TRAFs) and 14-3-3s. In parallel with the precise analyses unveiling the molecular basis of ASK1 regulation, the physiological or pathophysiological significance of ASK1 in diverse organs has been elucidated. In addition to the generation of global knockout mice or tissue-specific knockout mice, ASK1-specific inhibitors have illuminated the biological roles of ASK1. GENERAL SIGNIFICANCE The multi-faceted features of the function of ASK1 have been discovered over the past two decades, revealing that ASK1 is a crucial molecule for maintaining cellular homeostasis, especially under conditions of stress. Based on the results that ASK1 deficiency provides beneficial effects for several diseases, modulating ASK1 activity is a promising method to ameliorate a subset of diseases.
Collapse
|
165
|
Identification of interaction between HIV-1 glycoprotein 41 and integrase. Virol Sin 2016; 31:415-424. [PMID: 27681265 DOI: 10.1007/s12250-016-3820-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 08/30/2016] [Indexed: 10/20/2022] Open
Abstract
Human immunodeficiency virus-1 (HIV-1) encodes 15 viral proteins. Protein-protein interactions play a large role in the function of these proteins. In this study, we attempted to identify novel interactions between the HIV-1 proteins to better understand the role played by viral protein-protein interactions in the life cycle of HIV-1. Genes encoding the 15 viral proteins from the HIV-1 strain AD8 were inserted into the plasmids of a yeast two-hybrid system. By screening 120 pairs of proteins, interactions between seven pairs were found. This led to the discovery of an interaction between the HIV-1 proteins integrase (IN) and glycoprotein 41 (gp41), which was confirmed by both co-immunoprecipitation (Co-IP) assays and fluorescence resonance energy transfer (FRET) imaging in live cells. In addition, it was found that the amino acids at positions 76-100 of gp41 are required for it to bind to IN. Deletion of this region from gp41 prevented its interaction with IN and reduced the production of HIV-1 in 293T cells. This study provides new information on HIV-1 protein-protein interactions which improves the understanding of the biological functions of gp41 and IN during the virus life cycle.
Collapse
|
166
|
Editorial: Biomedical Engineering Approaches for HIV/AIDS Prophylaxis, Diagnostics and Therapy. Adv Drug Deliv Rev 2016; 103:1-4. [PMID: 27378403 DOI: 10.1016/j.addr.2016.06.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
167
|
Deeks SG, Lewin SR, Ross AL, Ananworanich J, Benkirane M, Cannon P, Chomont N, Douek D, Lifson JD, Lo YR, Kuritzkes D, Margolis D, Mellors J, Persaud D, Tucker JD, Barre-Sinoussi F, Alter G, Auerbach J, Autran B, Barouch DH, Behrens G, Cavazzana M, Chen Z, Cohen ÉA, Corbelli GM, Eholié S, Eyal N, Fidler S, Garcia L, Grossman C, Henderson G, Henrich TJ, Jefferys R, Kiem HP, McCune J, Moodley K, Newman PA, Nijhuis M, Nsubuga MS, Ott M, Palmer S, Richman D, Saez-Cirion A, Sharp M, Siliciano J, Silvestri G, Singh J, Spire B, Taylor J, Tolstrup M, Valente S, van Lunzen J, Walensky R, Wilson I, Zack J. International AIDS Society global scientific strategy: towards an HIV cure 2016. Nat Med 2016; 22:839-50. [PMID: 27400264 PMCID: PMC5322797 DOI: 10.1038/nm.4108] [Citation(s) in RCA: 364] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 04/12/2016] [Indexed: 02/07/2023]
Abstract
Antiretroviral therapy is not curative. Given the challenges in providing lifelong therapy to a global population of more than 35 million people living with HIV, there is intense interest in developing a cure for HIV infection. The International AIDS Society convened a group of international experts to develop a scientific strategy for research towards an HIV cure. This Perspective summarizes the group's strategy.
Collapse
Affiliation(s)
- Steven G Deeks
- Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Sharon R Lewin
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne and Royal Melbourne Hospital, Melbourne, Australia
- Department of Infectious Diseases, Alfred Hospital and Monash University, Melbourne, Australia
| | - Anna Laura Ross
- International and Scientific Relations Office, ANRS, Paris, France
| | - Jintanat Ananworanich
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
| | - Monsef Benkirane
- Molecular Virology Lab, Institute of Human Genetics, CNRS UPR 1142, Université de Montpellier, Montpellier, France
| | - Paula Cannon
- Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Nicolas Chomont
- CRCHUM and Department of Microbiology, Infectiology, and Immunology, Université de Montréal, Faculty of Medicine, Montréal, Quebec, Canada
| | - Daniel Douek
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Jeffrey D Lifson
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory, Frederick, Maryland, USA
| | - Ying-Ru Lo
- World Health Organization Regional Office for the Western Pacific, Manila, Philippines
| | | | - David Margolis
- University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, USA
| | - John Mellors
- Division of Infectious Diseases, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Deborah Persaud
- Johns Hopkins University School of Medicine &Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Joseph D Tucker
- University of North Carolina-Project China, Guangzhou, China
| | | | - Galit Alter
- Ragon Institute of MGH, MIT and Harvard, Boston, Massachusetts, USA
| | - Judith Auerbach
- Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Brigitte Autran
- Sorbonne Universités, UPMC Univ Paris 06, CIMI-Paris, France
- Inserm U1135, CIMI-Paris, Paris, France
- AP-HP, Hôpital Pitié-Salpêtrière, Département d'Immunologie, Paris, France
| | - Dan H Barouch
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
- Ragon Institute of MGH, MIT and Harvard, Boston, Massachusetts, USA
| | - Georg Behrens
- Clinic for Immunology and Rhematology, Hannover Medical School, Hanover, Germany
| | - Marina Cavazzana
- Centre d'Investigation Clinique en biothérapie, Hôpital Necker-Enfants Malades, Paris, France
| | - Zhiwei Chen
- AIDS Institute, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Éric A Cohen
- Institut de Recherches Cliniques de Montréal, Université de Montréal, Montréal, Quebec, Canada
| | | | - Serge Eholié
- Programme PAC-CI, Centre Hospitalier Universitaire de Treichville, Abidjan, Côte d'Ivoire
| | - Nir Eyal
- Harvard T. H. Chan School of Public Health, Department of Global Health and Population, Boston, Massachusetts, USA
| | - Sarah Fidler
- Department of Medicine, Imperial College London, London, United Kingdom
| | | | - Cynthia Grossman
- National Institute of Mental Health, NIH, Bethesda, Maryland, USA
| | - Gail Henderson
- University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, USA
| | - Timothy J Henrich
- Department of Medicine, University of California, San Francisco, San Francisco, California, USA
- Brigham &Women's Hospital, Boston, Massachusetts, USA
| | | | - Hans-Peter Kiem
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Joseph McCune
- Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Keymanthri Moodley
- Centre for Medical Ethics and Law, Department of Medicine, Stellenbosch University, Western Cape, South Africa
| | - Peter A Newman
- Factor-Inwentash Faculty of Social Work, University of Toronto, Toronto, Ontario, Canada
| | - Monique Nijhuis
- Department of Medical Microbiology, Virology, University Medical Center Utrecht, Utrecht, Netherlands
| | | | - Melanie Ott
- Gladstone Institutes, University of California, San Francisco, San Francisco, California, USA
| | - Sarah Palmer
- Westmead Millennium Institute for Medical Research, University of Sydney, Sydney, Australia
| | - Douglas Richman
- Virginia San Diego Healthcare System and University of California, San Diego, San Diego, California, USA
| | | | - Matthew Sharp
- Independent HIV Education and Advocacy Consultant, San Francisco, California, USA
| | - Janet Siliciano
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Guido Silvestri
- Yerkes National Primate Research Centre, Emory University, Atlanta, Georgia, USA
| | - Jerome Singh
- Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | | | - Jeffrey Taylor
- CARE Collaboratory Community Advisory Board, Palm Springs, California, USA
| | - Martin Tolstrup
- Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Susana Valente
- Department of Immunology and Microbial Sciences, The Scripps Research Institute, Jupiter, Florida, USA
| | | | - Rochelle Walensky
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ira Wilson
- Department of Health Services, Policy &Practice, Brown University School of Public Health, Providence, Rhode Island, USA
| | - Jerome Zack
- David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| |
Collapse
|
168
|
Nowicka-Sans B, Protack T, Lin Z, Li Z, Zhang S, Sun Y, Samanta H, Terry B, Liu Z, Chen Y, Sin N, Sit SY, Swidorski JJ, Chen J, Venables BL, Healy M, Meanwell NA, Cockett M, Hanumegowda U, Regueiro-Ren A, Krystal M, Dicker IB. Identification and Characterization of BMS-955176, a Second-Generation HIV-1 Maturation Inhibitor with Improved Potency, Antiviral Spectrum, and Gag Polymorphic Coverage. Antimicrob Agents Chemother 2016; 60:3956-69. [PMID: 27090171 PMCID: PMC4914680 DOI: 10.1128/aac.02560-15] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2015] [Accepted: 12/03/2015] [Indexed: 12/26/2022] Open
Abstract
BMS-955176 is a second-generation human immunodeficiency virus type 1 (HIV-1) maturation inhibitor (MI). A first-generation MI, bevirimat, showed clinical efficacy in early-phase studies, but ∼50% of subjects had viruses with reduced susceptibility associated with naturally occurring polymorphisms in Gag near the site of MI action. MI potency was optimized using a panel of engineered reporter viruses containing site-directed polymorphic changes in Gag that reduce susceptibility to bevirimat (including V362I, V370A/M/Δ, and T371A/Δ), leading incrementally to the identification of BMS-955176. BMS-955176 exhibits potent activity (50% effective concentration [EC50], 3.9 ± 3.4 nM [mean ± standard deviation]) toward a library (n = 87) of gag/pr recombinant viruses representing 96.5% of subtype B polymorphic Gag diversity near the CA/SP1 cleavage site. BMS-955176 exhibited a median EC50 of 21 nM toward a library of subtype B clinical isolates assayed in peripheral blood mononuclear cells (PBMCs). Potent activity was maintained against a panel of reverse transcriptase, protease, and integrase inhibitor-resistant viruses, with EC50s similar to those for the wild-type virus. A 5.4-fold reduction in EC50 occurred in the presence of 40% human serum plus 27 mg/ml of human serum albumin (HSA), which corresponded well to an in vitro measurement of 86% human serum binding. Time-of-addition and pseudotype reporter virus studies confirm a mechanism of action for the compound that occurs late in the virus replication cycle. BMS-955176 inhibits HIV-1 protease cleavage at the CA/SP1 junction within Gag in virus-like particles (VLPs) and in HIV-1-infected cells, and it binds reversibly and with high affinity to assembled Gag in purified HIV-1 VLPs. Finally, in vitro combination studies showed no antagonistic interactions with representative antiretrovirals (ARVs) of other mechanistic classes. In conclusion, BMS-955176 is a second-generation MI with potent in vitro anti-HIV-1 activity and a greatly improved preclinical profile compared to that of bevirimat.
Collapse
Affiliation(s)
- Beata Nowicka-Sans
- Bristol-Myers Squibb, Research and Development, Department of Virology, Wallingford, Connecticut, USA
| | - Tricia Protack
- Bristol-Myers Squibb, Research and Development, Department of Virology, Wallingford, Connecticut, USA
| | - Zeyu Lin
- Bristol-Myers Squibb, Research and Development, Department of Virology, Wallingford, Connecticut, USA
| | - Zhufang Li
- Bristol-Myers Squibb, Research and Development, Department of Virology, Wallingford, Connecticut, USA
| | - Sharon Zhang
- Bristol-Myers Squibb, Research and Development, Department of Virology, Wallingford, Connecticut, USA
| | - Yongnian Sun
- Bristol-Myers Squibb, Research and Development, Department of Virology, Wallingford, Connecticut, USA
| | - Himadri Samanta
- Bristol-Myers Squibb, Research and Development, Department of Virology, Wallingford, Connecticut, USA
| | - Brian Terry
- Bristol-Myers Squibb, Research and Development, Department of Virology, Wallingford, Connecticut, USA
| | - Zheng Liu
- Bristol-Myers Squibb, Research and Development, Department of Discovery Chemistry, Wallingford, Connecticut, USA
| | - Yan Chen
- Bristol-Myers Squibb, Research and Development, Department of Discovery Chemistry, Wallingford, Connecticut, USA
| | - Ny Sin
- Bristol-Myers Squibb, Research and Development, Department of Discovery Chemistry, Wallingford, Connecticut, USA
| | - Sing-Yuen Sit
- Bristol-Myers Squibb, Research and Development, Department of Discovery Chemistry, Wallingford, Connecticut, USA
| | - Jacob J Swidorski
- Bristol-Myers Squibb, Research and Development, Department of Discovery Chemistry, Wallingford, Connecticut, USA
| | - Jie Chen
- Bristol-Myers Squibb, Research and Development, Department of Discovery Chemistry, Wallingford, Connecticut, USA
| | - Brian L Venables
- Bristol-Myers Squibb, Research and Development, Department of Discovery Chemistry, Wallingford, Connecticut, USA
| | - Matthew Healy
- Bristol-Myers Squibb, Research and Development, Department of Genomics, Wallingford, Connecticut, USA
| | - Nicholas A Meanwell
- Bristol-Myers Squibb, Research and Development, Department of Discovery Chemistry, Wallingford, Connecticut, USA
| | - Mark Cockett
- Bristol-Myers Squibb, Research and Development, Department of Virology, Wallingford, Connecticut, USA
| | - Umesh Hanumegowda
- Bristol-Myers Squibb, Research and Development, Department of Preclinical Optimization, Wallingford, Connecticut, USA
| | - Alicia Regueiro-Ren
- Bristol-Myers Squibb, Research and Development, Department of Discovery Chemistry, Wallingford, Connecticut, USA
| | - Mark Krystal
- Bristol-Myers Squibb, Research and Development, Department of Virology, Wallingford, Connecticut, USA
| | - Ira B Dicker
- Bristol-Myers Squibb, Research and Development, Department of Virology, Wallingford, Connecticut, USA
| |
Collapse
|
169
|
Goldenberg NM, Steinberg BE, Rutka JT, Chen R, Cabral V, Rosenblum ND, Kapus A, Lee WL. Research projects in the Surgeon-Scientist and Clinician-Investigator programs at the University of Toronto (1987-2016): a cohort study. CMAJ Open 2016; 4:E444-E447. [PMID: 27730108 PMCID: PMC5047840 DOI: 10.9778/cmajo.20160062] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Physicians have traditionally been at the forefront of medical research, bringing clinical questions to the laboratory and returning with ideas for treatment. However, we have anecdotally observed a decline in the popularity of basic science research among trainees. We hypothesized that fewer resident physicians have been pursuing basic science research training over time. METHODS We examined records from residents in the Surgeon-Scientist and Clinician-Investigator programs at the University of Toronto (1987-2016). Research by residents was categorized independently by 2 raters as basic science, clinical epidemiology or education-related based on the title of the project, the name of the supervisor and Pubmed searches. The study population was divided into quintiles of time, and the proportion pursuing basic science training in each quintile was calculated. RESULTS Agreement between the raters was 100%; the categorization of the research topic remained unclear in 9 cases. The proportion of trainees pursuing basic science training dropped by 60% from 1987 to 2016 (p = 0.005). INTERPRETATION Significantly fewer residents in the Surgeon-Scientist and Clinician-Investigator Programs at the University of Toronto are pursuing training in the basic sciences as compared with previous years.
Collapse
Affiliation(s)
- Neil M Goldenberg
- Departments of Anesthesia (Goldenberg, Steinberg); Surgery (Rutka, Cabral, Kapus); Medicine (Chen, Lee); Pediatrics (Rosenblum), University of Toronto; Keenan Research Centre for Biomedical Science (Kapus, Lee), Li Ka Shing Knowledge Institute at St. Michael's Hospital; Interdepartmental Division of Critical Care Medicine (Lee), University of Toronto, Toronto, Ont
| | - Benjamin E Steinberg
- Departments of Anesthesia (Goldenberg, Steinberg); Surgery (Rutka, Cabral, Kapus); Medicine (Chen, Lee); Pediatrics (Rosenblum), University of Toronto; Keenan Research Centre for Biomedical Science (Kapus, Lee), Li Ka Shing Knowledge Institute at St. Michael's Hospital; Interdepartmental Division of Critical Care Medicine (Lee), University of Toronto, Toronto, Ont
| | - James T Rutka
- Departments of Anesthesia (Goldenberg, Steinberg); Surgery (Rutka, Cabral, Kapus); Medicine (Chen, Lee); Pediatrics (Rosenblum), University of Toronto; Keenan Research Centre for Biomedical Science (Kapus, Lee), Li Ka Shing Knowledge Institute at St. Michael's Hospital; Interdepartmental Division of Critical Care Medicine (Lee), University of Toronto, Toronto, Ont
| | - Robert Chen
- Departments of Anesthesia (Goldenberg, Steinberg); Surgery (Rutka, Cabral, Kapus); Medicine (Chen, Lee); Pediatrics (Rosenblum), University of Toronto; Keenan Research Centre for Biomedical Science (Kapus, Lee), Li Ka Shing Knowledge Institute at St. Michael's Hospital; Interdepartmental Division of Critical Care Medicine (Lee), University of Toronto, Toronto, Ont
| | - Val Cabral
- Departments of Anesthesia (Goldenberg, Steinberg); Surgery (Rutka, Cabral, Kapus); Medicine (Chen, Lee); Pediatrics (Rosenblum), University of Toronto; Keenan Research Centre for Biomedical Science (Kapus, Lee), Li Ka Shing Knowledge Institute at St. Michael's Hospital; Interdepartmental Division of Critical Care Medicine (Lee), University of Toronto, Toronto, Ont
| | - Norman D Rosenblum
- Departments of Anesthesia (Goldenberg, Steinberg); Surgery (Rutka, Cabral, Kapus); Medicine (Chen, Lee); Pediatrics (Rosenblum), University of Toronto; Keenan Research Centre for Biomedical Science (Kapus, Lee), Li Ka Shing Knowledge Institute at St. Michael's Hospital; Interdepartmental Division of Critical Care Medicine (Lee), University of Toronto, Toronto, Ont
| | - Andras Kapus
- Departments of Anesthesia (Goldenberg, Steinberg); Surgery (Rutka, Cabral, Kapus); Medicine (Chen, Lee); Pediatrics (Rosenblum), University of Toronto; Keenan Research Centre for Biomedical Science (Kapus, Lee), Li Ka Shing Knowledge Institute at St. Michael's Hospital; Interdepartmental Division of Critical Care Medicine (Lee), University of Toronto, Toronto, Ont
| | - Warren L Lee
- Departments of Anesthesia (Goldenberg, Steinberg); Surgery (Rutka, Cabral, Kapus); Medicine (Chen, Lee); Pediatrics (Rosenblum), University of Toronto; Keenan Research Centre for Biomedical Science (Kapus, Lee), Li Ka Shing Knowledge Institute at St. Michael's Hospital; Interdepartmental Division of Critical Care Medicine (Lee), University of Toronto, Toronto, Ont
| |
Collapse
|
170
|
Spanevello F, Calistri A, Del Vecchio C, Mantelli B, Frasson C, Basso G, Palù G, Cavazzana M, Parolin C. Development of Lentiviral Vectors Simultaneously Expressing Multiple siRNAs Against CCR5, vif and tat/rev Genes for an HIV-1 Gene Therapy Approach. MOLECULAR THERAPY. NUCLEIC ACIDS 2016; 5:e312. [PMID: 27093170 PMCID: PMC5014525 DOI: 10.1038/mtna.2016.24] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 03/08/2016] [Indexed: 02/08/2023]
Abstract
Gene therapy holds considerable promise for the functional cure of HIV-1 infection and, in this context, RNA interference (RNAi)-based approaches represent powerful strategies. Stable expression of small interfering RNAs (siRNAs) targeting HIV genes or cellular cofactors has the potential to render HIV-1 susceptible cells resistant to infection. To inhibit different steps of virus life cycle, self-inactivating lentiviral vectors expressing multiple siRNAs targeting the CCR5 cellular gene as well as vif and tat/rev viral transcripts, under the control of different RNA polymerase III promoters (U6, 7SK, H1) were developed. The use of a single RNA polymerase III promoter driving the expression of a sequence giving rise to three siRNAs directed against the selected targets (e-shRNA) was also investigated. Luciferase assay and inhibition of HIV-1 replication in human Jurkat T-cell line were adopted to select the best combination of promoter/siRNA. The efficacy of selected developed combinatorial vectors in interfering with viral replication was evaluated in human primary CD4(+) T lymphocytes. We identified two effective anti-HIV combinatorial vectors that conferred protection against R5- and X4- tropic viruses. Overall, our results showed that the antiviral effect is influenced by different factors, including the promoter used to express the RNAi molecules and the selected cassette combination. These findings contribute to gain further insights in the design of RNAi-based gene therapy approaches against HIV-1 for clinical application.
Collapse
Affiliation(s)
| | - Arianna Calistri
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | | | - Barbara Mantelli
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Chiara Frasson
- Oncohematology Laboratory, Department of Women's and Children's Health, University of Padova, Padova, Italy
- Istituto di Ricerca Pediatrica Città della Speranza (IRP), Padova, Italy
| | - Giuseppe Basso
- Oncohematology Laboratory, Department of Women's and Children's Health, University of Padova, Padova, Italy
| | - Giorgio Palù
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Marina Cavazzana
- Biotherapy Department, Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique–Hôpitaux de Paris, INSERM, Paris, France
- Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, Paris, France
- INSERM UMR1163, Laboratory of Human Lymphohematopoiesis, Paris, France
| | - Cristina Parolin
- Department of Molecular Medicine, University of Padova, Padova, Italy
| |
Collapse
|
171
|
Price AA, Grakoui A, Weiss DS. Harnessing the Prokaryotic Adaptive Immune System as a Eukaryotic Antiviral Defense. Trends Microbiol 2016; 24:294-306. [PMID: 26852268 PMCID: PMC4808413 DOI: 10.1016/j.tim.2016.01.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 01/12/2016] [Accepted: 01/13/2016] [Indexed: 02/07/2023]
Abstract
Clustered, regularly interspaced, short palindromic repeats - CRISPR-associated (CRISPR-Cas) systems - are sequence-specific RNA-directed endonuclease complexes that bind and cleave nucleic acids. These systems evolved within prokaryotes as adaptive immune defenses to target and degrade nucleic acids derived from bacteriophages and other foreign genetic elements. The antiviral function of these systems has now been exploited to combat eukaryotic viruses throughout the viral life cycle. Here we discuss current advances in CRISPR-Cas9 technology as a eukaryotic antiviral defense.
Collapse
Affiliation(s)
- Aryn A Price
- Department of Microbiology and Immunology, Microbiology and Molecular Genetics Program, Emory University, Atlanta, GA 30329, USA; Emory Vaccine Center, Emory University, Atlanta, GA 30329, USA; Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Arash Grakoui
- Emory Vaccine Center, Emory University, Atlanta, GA 30329, USA; Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA; Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia 30329, USA.
| | - David S Weiss
- Emory Vaccine Center, Emory University, Atlanta, GA 30329, USA; Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA; Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia 30329, USA.
| |
Collapse
|
172
|
Design, synthesis and activity evaluation of novel peptide fusion inhibitors targeting HIV-1 gp41. Bioorg Med Chem 2016; 24:201-6. [DOI: 10.1016/j.bmc.2015.12.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Revised: 11/05/2015] [Accepted: 12/02/2015] [Indexed: 11/19/2022]
|
173
|
Munawwar A, Singh S. Human Herpesviruses as Copathogens of HIV Infection, Their Role in HIV Transmission, and Disease Progression. J Lab Physicians 2016; 8:5-18. [PMID: 27013807 PMCID: PMC4785766 DOI: 10.4103/0974-2727.176228] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Of eight human herpesviruses (HHVs), often, only herpes simplex virus types 1 (HSV-1) and 2 (HSV-2) find mention in medical literature as both of these viruses are commonly associated with genital lesions and oral ulcers, commonly known as cold sores. However, role of human herpesviruses as copathogens and in aggravation and in the transmission of other human diseases, especially the Acquired immunodeficiency syndrome (HIV/AIDS) has only very recently been recognized. Therefore, screening and treating subclinical HHV infections may offer slowing of HIV infection, disease progression, and its transmission. Beside HSV-1 and HSV-2, HHV-3 a causative agent of herpes zoster remained one of the first manifestations of HIV disease before the era of highly active antiretroviral therapy (HAART). HHV-5 also known as human Cytomegalovirus infection remains a significant risk factor for HIV-associated mortality and morbidity even in HAART era. It is proposed that Cytomegalovirus viremia could be a better predictor of HIV disease progression than CD4+ T-lymphocyte count. The role of HHV-4 or Epstein-Burr virus and HHV-6, HHV-7, and HHV-8 is still being investigated in HIV disease progression. This review provides insight into the current understanding about these 8 HHVs, their co-pathogenesis, and role in HIV/AIDS disease progression. The review also covers recent literature in favor and against administering anti-HHV treatment along with HAART for slower AIDS progression and interrupted sexual transmission.
Collapse
Affiliation(s)
- Arshi Munawwar
- Department of Laboratory Medicine, Division of Clinical Microbiology and Molecular Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Sarman Singh
- Department of Laboratory Medicine, Division of Clinical Microbiology and Molecular Medicine, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
174
|
The α7-nicotinic receptor is upregulated in immune cells from HIV-seropositive women: consequences to the cholinergic anti-inflammatory response. Clin Transl Immunology 2015; 4:e53. [PMID: 26719799 PMCID: PMC4685439 DOI: 10.1038/cti.2015.31] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Revised: 10/08/2015] [Accepted: 10/12/2015] [Indexed: 01/23/2023] Open
Abstract
Antiretroviral therapy partially restores the immune system and markedly increases life expectancy of HIV-infected patients. However, antiretroviral therapy does not restore full health. These patients suffer from poorly understood chronic inflammation that causes a number of AIDS and non-AIDS complications. Here we show that chronic inflammation in HIV+ patients may be due to the disruption of the cholinergic anti-inflammatory pathway by HIV envelope protein gp120IIIB. Our results demonstrate that HIV gp120IIIB induces α7 nicotinic acetylcholine receptor (α7) upregulation and a paradoxical proinflammatory phenotype in macrophages, as activation of the upregulated α7 is no longer capable of inhibiting the release of proinflammatory cytokines. Our results demonstrate that disruption of the cholinergic-mediated anti-inflammatory response can result from an HIV protein. Collectively, these findings suggest that HIV tampering with a natural strategy to control inflammation could contribute to a crucial, unresolved problem of HIV infection: chronic inflammation.
Collapse
|
175
|
Tryptophan dendrimers that inhibit HIV replication, prevent virus entry and bind to the HIV envelope glycoproteins gp120 and gp41. Eur J Med Chem 2015; 106:34-43. [DOI: 10.1016/j.ejmech.2015.10.031] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 10/13/2015] [Accepted: 10/15/2015] [Indexed: 11/17/2022]
|
176
|
Mocquet V, Durand S, Jalinot P. How Retroviruses Escape the Nonsense-Mediated mRNA Decay. AIDS Res Hum Retroviruses 2015; 31:948-58. [PMID: 26066561 DOI: 10.1089/aid.2014.0326] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Many posttranscriptional processes are known to regulate gene expression and some of them can act as an antiviral barrier. The nonsense-mediated mRNA decay (NMD) was first identified as an mRNA quality control pathway that triggers rapid decay of mRNA containing premature stop codons due to mutations. NMD is now considered as a general posttranscriptional regulation pathway controlling the expression of a large set of cellular genes. In addition to premature stop codons, many other features including alternative splicing, 5' uORF, long 3' UTR, selenocystein codons, and frameshift are able to promote NMD. Interestingly, many viral mRNAs exhibit some of these features suggesting that virus expression and replication might be sensitive to NMD. Several studies, including recent ones, have shown that this is the case for retroviruses; however, it also appears that retroviruses have developed strategies to overcome NMD in order to protect their genome and ensure a true expression of their genes. As a consequence of NMD inhibition, these viruses also affect the expression of host genes that are prone to NMD, and therefore can potentially trigger pathological effects on infected cells. Here, we review recent studies supporting this newly uncovered function of the NMD pathway as a defense barrier that viruses must overcome in order to replicate.
Collapse
Affiliation(s)
- Vincent Mocquet
- Laboratoire de Biologie Moléculaire de la Cellule, Unité Mixte de Recherche 5239, Centre National de la Recherche Scientifique , Ecole Normale Supérieure, Lyon, France
| | - Sebastien Durand
- Laboratoire de Biologie Moléculaire de la Cellule, Unité Mixte de Recherche 5239, Centre National de la Recherche Scientifique , Ecole Normale Supérieure, Lyon, France
| | - Pierre Jalinot
- Laboratoire de Biologie Moléculaire de la Cellule, Unité Mixte de Recherche 5239, Centre National de la Recherche Scientifique , Ecole Normale Supérieure, Lyon, France
| |
Collapse
|
177
|
Katsarou K, Rao ALN, Tsagris M, Kalantidis K. Infectious long non-coding RNAs. Biochimie 2015; 117:37-47. [PMID: 25986218 DOI: 10.1016/j.biochi.2015.05.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 05/07/2015] [Indexed: 02/06/2023]
Abstract
Long non protein coding RNAs (lncRNAs) constitute a large category of the RNA world, able to regulate different biological processes. In this review we are focusing on infectious lncRNAs, their classification, pathogenesis and impact on the infected organisms. Here they are presented in two separate groups: 'dependent lncRNAs' (comprising satellites RNA, Hepatitis D virus and lncRNAs of viral origin) which need a helper virus and 'independent lncRNAs' (viroids) that can self-replicate. Even though these lncRNA do not encode any protein, their structure and/or sequence comprise all the necessary information to drive specific interactions with host factors and regulate several cellular functions. These new data that have emerged during the last few years concerning lncRNAs modify the way we understand molecular biology's 'central dogma' and give new perspectives for applications and potential therapeutic strategies.
Collapse
Affiliation(s)
- Konstantina Katsarou
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, Heraklion, Crete, Greece
| | - A L N Rao
- Department of Plant Pathology and Microbiology, University of California, Riverside, CA, 92521-01222, USA
| | - Mina Tsagris
- Department of Biology, University of Crete, Heraklion, Crete, Greece
| | - Kriton Kalantidis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, Heraklion, Crete, Greece; Department of Biology, University of Crete, Heraklion, Crete, Greece.
| |
Collapse
|
178
|
Wu B, Miskolci V, Sato H, Tutucci E, Kenworthy CA, Donnelly SK, Yoon YJ, Cox D, Singer RH, Hodgson L. Synonymous modification results in high-fidelity gene expression of repetitive protein and nucleotide sequences. Genes Dev 2015; 29:876-86. [PMID: 25877922 PMCID: PMC4403262 DOI: 10.1101/gad.259358.115] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 03/18/2015] [Indexed: 01/30/2023]
Abstract
Repetitive nucleotide or amino acid sequences are often engineered into probes and biosensors to achieve functional readouts and robust signal amplification, but these repeated sequences are notoriously prone to aberrant deletion and degradation. Wu et al. developed an approach to solve this problem by modifying the nucleotide sequences of the target mRNA to make them nonrepetitive but still functional (“synonymous”). Using the synonymous modification to FRET biosensors, they achieved correct expression of full-length sensors and found that the biological interpretations of the sensor are significantly different when a correct, full-length biosensor is expressed. Repetitive nucleotide or amino acid sequences are often engineered into probes and biosensors to achieve functional readouts and robust signal amplification. However, these repeated sequences are notoriously prone to aberrant deletion and degradation, impacting the ability to correctly detect and interpret biological functions. Here, we introduce a facile and generalizable approach to solve this often unappreciated problem by modifying the nucleotide sequences of the target mRNA to make them nonrepetitive but still functional (“synonymous”). We first demonstrated the procedure by designing a cassette of synonymous MS2 RNA motifs and tandem coat proteins for RNA imaging and showed a dramatic improvement in signal and reproducibility in single-RNA detection in live cells. The same approach was extended to enhancing the stability of engineered fluorescent biosensors containing a fluorescent resonance energy transfer (FRET) pair of fluorescent proteins on which a great majority of systems thus far in the field are based. Using the synonymous modification to FRET biosensors, we achieved correct expression of full-length sensors, eliminating the aberrant truncation products that often were assumed to be due to nonspecific proteolytic cleavages. Importantly, the biological interpretations of the sensor are significantly different when a correct, full-length biosensor is expressed. Thus, we show here a useful and generally applicable method to maintain the integrity of expressed genes, critical for the correct interpretation of probe readouts.
Collapse
Affiliation(s)
- Bin Wu
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, New York 10461, USA; Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | - Veronika Miskolci
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | - Hanae Sato
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | - Evelina Tutucci
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | - Charles A Kenworthy
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | - Sara K Donnelly
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, New York 10461, USA; Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | - Young J Yoon
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | - Dianne Cox
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, New York 10461, USA; Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | - Robert H Singer
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, New York 10461, USA; Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | - Louis Hodgson
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, New York 10461, USA; Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| |
Collapse
|
179
|
The nanoscale organization of signaling domains at the plasma membrane. CURRENT TOPICS IN MEMBRANES 2015; 75:125-65. [PMID: 26015282 DOI: 10.1016/bs.ctm.2015.03.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
In this chapter, we present an overview of the role of the nanoscale organization of signaling domains in regulating key cellular processes. In particular, we illustrate the importance of protein and lipid nanodomains as triggers and mediators of cell signaling. As particular examples, we summarize the state of the art of understanding the role of nanodomains in the mounting of an immune response, cellular adhesion, intercellular communication, and cell proliferation. Thus, this chapter underlines the essential role the nanoscale organization of key signaling proteins and lipid domains. We will also see how nanodomains play an important role in the lifecycle of many pathogens relevant to human disease and therefore illustrate how these structures may become future therapeutic targets.
Collapse
|
180
|
Okamoto M, Miyazawa T, Morikawa S, Ono F, Nakamura S, Sato E, Yoshida T, Yoshikawa R, Sakai K, Mizutani T, Nagata N, Takano JI, Okabayashi S, Hamano M, Fujimoto K, Nakaya T, Iida T, Horii T, Miyabe-Nishiwaki T, Watanabe A, Kaneko A, Saito A, Matsui A, Hayakawa T, Suzuki J, Akari H, Matsuzawa T, Hirai H. Emergence of infectious malignant thrombocytopenia in Japanese macaques (Macaca fuscata) by SRV-4 after transmission to a novel host. Sci Rep 2015; 5:8850. [PMID: 25743183 PMCID: PMC4351523 DOI: 10.1038/srep08850] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 01/28/2015] [Indexed: 11/14/2022] Open
Abstract
We discovered a lethal hemorrhagic syndrome arising from severe thrombocytopenia in Japanese macaques kept at the Primate Research Institute, Kyoto University. Extensive investigation identified that simian retrovirus type 4 (SRV-4) was the causative agent of the disease. SRV-4 had previously been isolated only from cynomolgus macaques in which it is usually asymptomatic. We consider that the SRV-4 crossed the so-called species barrier between cynomolgus and Japanese macaques, leading to extremely severe acute symptoms in the latter. Infectious agents that cross the species barrier occasionally amplify in virulence, which is not observed in the original hosts. In such cases, the new hosts are usually distantly related to the original hosts. However, Japanese macaques are closely related to cynomolgus macaques, and can even hybridize when given the opportunity. This lethal outbreak of a novel pathogen in Japanese macaques highlights the need to modify our expectations about virulence with regards crossing species barriers.
Collapse
Affiliation(s)
- Munehiro Okamoto
- Center for Human Evolution Modeling Research, Primate Research Institute, Kyoto University, Inuyama, Aichi 484-8506, Japan
| | - Takayuki Miyazawa
- Laboratory of Signal Transduction, Department of Cell Biology, Institute for Virus Research, Kyoto University, Kyoto 606-8507, Japan
| | - Shigeru Morikawa
- National Institute of Infectious Diseases, Toyama 1-23-1, Shinjuku-ku, Tokyo 162-8640, Japan
| | - Fumiko Ono
- The Corporation for Production and Research of Laboratory Primates, 1-16-2 Sakura, Tsukuba, Ibaraki 305-0003, Japan
| | - Shota Nakamura
- Department of Infection Metagenomics, Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Eiji Sato
- Center for Human Evolution Modeling Research, Primate Research Institute, Kyoto University, Inuyama, Aichi 484-8506, Japan
| | - Tomoyuki Yoshida
- Center for Human Evolution Modeling Research, Primate Research Institute, Kyoto University, Inuyama, Aichi 484-8506, Japan
| | - Rokusuke Yoshikawa
- Laboratory of Signal Transduction, Department of Cell Biology, Institute for Virus Research, Kyoto University, Kyoto 606-8507, Japan
| | - Kouji Sakai
- National Institute of Infectious Diseases, Toyama 1-23-1, Shinjuku-ku, Tokyo 162-8640, Japan
| | - Tetsuya Mizutani
- National Institute of Infectious Diseases, Toyama 1-23-1, Shinjuku-ku, Tokyo 162-8640, Japan
| | - Noriyo Nagata
- National Institute of Infectious Diseases, Toyama 1-23-1, Shinjuku-ku, Tokyo 162-8640, Japan
| | - Jun-ichiro Takano
- 1] The Corporation for Production and Research of Laboratory Primates, 1-16-2 Sakura, Tsukuba, Ibaraki 305-0003, Japan [2] Tsukuba Primate Research Center, National Institute of Biomedical Innovation, 1-1 Hachimandai, Tsukuba, Ibaraki 305-0843, Japan
| | - Sachi Okabayashi
- The Corporation for Production and Research of Laboratory Primates, 1-16-2 Sakura, Tsukuba, Ibaraki 305-0003, Japan
| | - Masataka Hamano
- The Corporation for Production and Research of Laboratory Primates, 1-16-2 Sakura, Tsukuba, Ibaraki 305-0003, Japan
| | - Koji Fujimoto
- The Corporation for Production and Research of Laboratory Primates, 1-16-2 Sakura, Tsukuba, Ibaraki 305-0003, Japan
| | - Takaaki Nakaya
- 1] Department of Infection Metagenomics, Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan [2] Department of Infectious Diseases, Kyoto Prefectural University of Medicine, 465 Kawaramachi-hirokoji, Kamigyo-ku, Kyoto, Japan
| | - Tetsuya Iida
- Department of Infection Metagenomics, Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Toshihiro Horii
- 1] Department of Infection Metagenomics, Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan [2] Department of Molecular Protozoology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Takako Miyabe-Nishiwaki
- Center for Human Evolution Modeling Research, Primate Research Institute, Kyoto University, Inuyama, Aichi 484-8506, Japan
| | - Akino Watanabe
- Center for Human Evolution Modeling Research, Primate Research Institute, Kyoto University, Inuyama, Aichi 484-8506, Japan
| | - Akihisa Kaneko
- Center for Human Evolution Modeling Research, Primate Research Institute, Kyoto University, Inuyama, Aichi 484-8506, Japan
| | - Akatsuki Saito
- Center for Human Evolution Modeling Research, Primate Research Institute, Kyoto University, Inuyama, Aichi 484-8506, Japan
| | - Atsushi Matsui
- Center for Human Evolution Modeling Research, Primate Research Institute, Kyoto University, Inuyama, Aichi 484-8506, Japan
| | - Toshiyuki Hayakawa
- Center for Human Evolution Modeling Research, Primate Research Institute, Kyoto University, Inuyama, Aichi 484-8506, Japan
| | - Juri Suzuki
- Center for Human Evolution Modeling Research, Primate Research Institute, Kyoto University, Inuyama, Aichi 484-8506, Japan
| | - Hirofumi Akari
- Center for Human Evolution Modeling Research, Primate Research Institute, Kyoto University, Inuyama, Aichi 484-8506, Japan
| | - Tetsuro Matsuzawa
- Department of Brain and Behavioral Sciences, Primate Research Institute, Kyoto University, Inuyama, Aichi 484-8506, Japan
| | - Hirohisa Hirai
- Department of Molecular Biology, Primate Research Institute, Kyoto University, Inuyama, Aichi 484-8506, Japan
| |
Collapse
|
181
|
Okazaki S, Mizuhara T, Shimura K, Murayama H, Ohno H, Oishi S, Matsuoka M, Fujii N. Identification of anti-HIV agents with a novel benzo[4,5]isothiazolo[2,3-a]pyrimidine scaffold. Bioorg Med Chem 2015; 23:1447-52. [PMID: 25744188 DOI: 10.1016/j.bmc.2015.02.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Revised: 02/06/2015] [Accepted: 02/10/2015] [Indexed: 12/29/2022]
Abstract
3,4-Dihydro-2H-benzo[4,5]isothiazolo[2,3-a]pyrimidine is a newly identified antiviral agent against human immunodeficiency virus type 1 (HIV-1) infection, derived from 3,4-dihydro-2H,6H-pyrimido[1,2-c][1,3]benzothiazin-6-imine (PD 404182). The introduction of the hydrophobic 8-aryl substituent on the benzene substructure improved its anti-HIV activity, resulting in the identification of 6-fold more potent analogs. In addition, it was demonstrated that these isothiazolopyrimidine derivatives exert anti-HIV effects at an early stage of viral infection.
Collapse
Affiliation(s)
- Shiho Okazaki
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Tsukasa Mizuhara
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Kazuya Shimura
- Institute for Virus Research, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | - Hiroto Murayama
- Institute for Virus Research, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | - Hiroaki Ohno
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Shinya Oishi
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan.
| | - Masao Matsuoka
- Institute for Virus Research, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | - Nobutaka Fujii
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan.
| |
Collapse
|
182
|
Steinbrenner H, Al-Quraishy S, Dkhil MA, Wunderlich F, Sies H. Dietary selenium in adjuvant therapy of viral and bacterial infections. Adv Nutr 2015; 6:73-82. [PMID: 25593145 PMCID: PMC4288282 DOI: 10.3945/an.114.007575] [Citation(s) in RCA: 173] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Viral and bacterial infections are often associated with deficiencies in macronutrients and micronutrients, including the essential trace element selenium. In selenium deficiency, benign strains of Coxsackie and influenza viruses can mutate to highly pathogenic strains. Dietary supplementation to provide adequate or supranutritional selenium supply has been proposed to confer health benefits for patients suffering from some viral diseases, most notably with respect to HIV and influenza A virus (IAV) infections. In addition, selenium-containing multimicronutrient supplements improved several clinical and lifestyle variables in patients coinfected with HIV and Mycobacterium tuberculosis. Selenium status may affect the function of cells of both adaptive and innate immunity. Supranutritional selenium promotes proliferation and favors differentiation of naive CD4-positive T lymphocytes toward T helper 1 cells, thus supporting the acute cellular immune response, whereas excessive activation of the immune system and ensuing host tissue damage are counteracted through directing macrophages toward the M2 phenotype. This review provides an up-to-date overview on selenium in infectious diseases caused by viruses (e.g., HIV, IAV, hepatitis C virus, poliovirus, West Nile virus) and bacteria (e.g., M. tuberculosis, Helicobacter pylori). Data from epidemiologic studies and intervention trials, with selenium alone or in combination with other micronutrients, and animal experiments are discussed against the background of dietary selenium requirements to alter immune functions.
Collapse
Affiliation(s)
| | - Saleh Al-Quraishy
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia; and
| | - Mohamed A Dkhil
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia; and Department of Zoology and Entomology, Faculty of Science, Helwan University, Cairo, Egypt
| | - Frank Wunderlich
- Department of Biology, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Helmut Sies
- Institute of Biochemistry and Molecular Biology I and Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia; and
| |
Collapse
|
183
|
Colson P, Ravaux I, Tamalet C, Glazunova O, Baptiste E, Chabriere E, Wiedemann A, Lacabaratz C, Chefrour M, Picard C, Stein A, Levy Y, Raoult D. HIV infection en route to endogenization: two cases. Clin Microbiol Infect 2014; 20:1280-8. [PMID: 25366539 PMCID: PMC4360783 DOI: 10.1111/1469-0691.12807] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Revised: 10/28/2014] [Accepted: 10/28/2014] [Indexed: 12/30/2022]
Abstract
The long-term spontaneous evolution of humans and the human immunodeficiency virus (HIV) is not well characterized; many vertebrate species, including humans, exhibit remnants of other retroviruses in their genomes that question such possible endogenization of HIV. We investigated two HIV-infected patients with no HIV-related disease and no detection with routine tests of plasma HIV RNA or cell-associated HIV DNA. We used Sanger and deep sequencing to retrieve HIV DNA sequences integrated in the human genome and tested the host humoral and cellular immune responses. We noticed that viruses from both patients were inactivated by the high prevalence of the transformation of tryptophan codons into stop codons (25% overall (3-100% per gene) and 24% overall (0-50% per gene)). In contrast, the humoral and/or cellular responses were strong for one patient and moderate for the other, indicating that a productive infection occurred at one stage of the infection. We speculate that the stimulation of APOBEC, the enzyme group that exchanges G for A in viral nucleic acids and is usually inhibited by the HIV protein Vif, has been amplified and made effective from the initial stage of the infection. Furthermore, we propose that a cure for HIV may occur through HIV endogenization in humans, as observed for many other retroviruses in mammals, rather than clearance of all traces of HIV from human cells, which defines viral eradication.
Collapse
Affiliation(s)
- P Colson
- Unité de Recherche sur les Maladies Infectieuses et Tropicales Emergentes (URMITE), UM63 CNRS 7278 IRD 198 INSERM U1095, Aix-Marseille Université, Marseille, France; Fondation Institut Hospitalo-Universitaire (IHU) MéiterranéInfection, Pôle des Maladies Infectieuses et Tropicales Clinique et Biologique, Fédération de Bactériologie-Hygiène-Virologie, Centre Hospitalo-Universitaire Timone, Assistance Publique-Hôpitaux de Marseille, Marseille, France
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
184
|
de Goede AL, Vulto AG, Osterhaus ADME, Gruters RA. Understanding HIV infection for the design of a therapeutic vaccine. Part II: Vaccination strategies for HIV. ANNALES PHARMACEUTIQUES FRANÇAISES 2014; 73:169-79. [PMID: 25528627 DOI: 10.1016/j.pharma.2014.11.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Accepted: 11/07/2014] [Indexed: 02/07/2023]
Abstract
HIV infection leads to a gradual loss CD4(+) T lymphocytes comprising immune competence and progression to AIDS. Effective treatment with combined antiretroviral drugs (cART) decreases viral load below detectable levels but is not able to eliminate the virus from the body. The success of cART is frustrated by the requirement of expensive lifelong adherence, accumulating drug toxicities and chronic immune activation resulting in increased risk of several non-AIDS disorders, even when viral replication is suppressed. Therefore, there is a strong need for therapeutic strategies as an alternative to cART. Immunotherapy, or therapeutic vaccination, aims to increase existing immune responses against HIV or induce de novo immune responses. These immune responses should provide a functional cure by controlling viral replication and preventing disease progression in the absence of cART. The key difficulty in the development of an HIV vaccine is our ignorance of the immune responses that control of viral replication, and thus how these responses can be elicited and how they can be monitored. Part one of this review provides an extensive overview of the (patho-) physiology of HIV infection. It describes the structure and replication cycle of HIV, the epidemiology and pathogenesis of HIV infection and the innate and adaptive immune responses against HIV. Part two of this review discusses therapeutic options for HIV. Prevention modalities and antiretroviral therapy are briefly touched upon, after which an extensive overview on vaccination strategies for HIV is provided, including the choice of immunogens and delivery strategies.
Collapse
Affiliation(s)
- A L de Goede
- Department of Viroscience, Erasmus MC, 's-Gravendijkwal 230, PO box 2040, 3000 CA Rotterdam, The Netherlands; Department of Hospital Pharmacy, Erasmus MC, 's-Gravendijkwal 230, PO box 2040, 3000 CA Rotterdam, The Netherlands.
| | - A G Vulto
- Department of Hospital Pharmacy, Erasmus MC, 's-Gravendijkwal 230, PO box 2040, 3000 CA Rotterdam, The Netherlands
| | - A D M E Osterhaus
- Department of Viroscience, Erasmus MC, 's-Gravendijkwal 230, PO box 2040, 3000 CA Rotterdam, The Netherlands
| | - R A Gruters
- Department of Viroscience, Erasmus MC, 's-Gravendijkwal 230, PO box 2040, 3000 CA Rotterdam, The Netherlands
| |
Collapse
|
185
|
de Goede AL, Vulto AG, Osterhaus ADME, Gruters RA. Understanding HIV infection for the design of a therapeutic vaccine. Part I: Epidemiology and pathogenesis of HIV infection. ANNALES PHARMACEUTIQUES FRANÇAISES 2014; 73:87-99. [PMID: 25496723 DOI: 10.1016/j.pharma.2014.11.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Revised: 11/01/2014] [Accepted: 11/07/2014] [Indexed: 02/07/2023]
Abstract
HIV infection leads to a gradual loss CD4+ T lymphocytes comprising immune competence and progression to AIDS. Effective treatment with combined antiretroviral drugs (cART) decreases viral load below detectable levels but is not able to eliminate the virus from the body. The success of cART is frustrated by the requirement of expensive life-long adherence, accumulating drug toxicities and chronic immune activation resulting in increased risk of several non-AIDS disorders, even when viral replication is suppressed. Therefore there is a strong need for therapeutic strategies as an alternative to cART. Immunotherapy, or therapeutic vaccination, aims to increase existing immune responses against HIV or induce de novo immune responses. These immune responses should provide a functional cure by controlling viral replication and preventing disease progression in the absence of cART. The key difficulty in the development of an HIV vaccine is our ignorance of the immune responses that control of viral replication, and thus how these responses can be elicited and how they can be monitored. Part one of this review provides an extensive overview of the (patho-) physiology of HIV infection. It describes the structure and replication cycle of HIV, the epidemiology and pathogenesis of HIV infection and the innate and adaptive immune responses against HIV. Part two of this review discusses therapeutic options for HIV. Prevention modalities and antiretroviral therapy are briefly touched upon, after which an extensive overview on vaccination strategies for HIV is provided, including the choice of immunogens and delivery strategies.
Collapse
Affiliation(s)
- A L de Goede
- Department of Viroscience, Erasmus MC 's-Gravendijkwal 230, 2040, 3000 CA Rotterdam, The Netherlands; Department of Hospital Pharmacy, Erasmus MC 's-Gravendijkwal 230, 2040, 3000 CA Rotterdam, The Netherlands.
| | - A G Vulto
- Department of Hospital Pharmacy, Erasmus MC 's-Gravendijkwal 230, 2040, 3000 CA Rotterdam, The Netherlands
| | - A D M E Osterhaus
- Department of Viroscience, Erasmus MC 's-Gravendijkwal 230, 2040, 3000 CA Rotterdam, The Netherlands
| | - R A Gruters
- Department of Viroscience, Erasmus MC 's-Gravendijkwal 230, 2040, 3000 CA Rotterdam, The Netherlands
| |
Collapse
|
186
|
Boons E, Li G, Vanstreels E, Vercruysse T, Pannecouque C, Vandamme AM, Daelemans D. A stably expressed llama single-domain intrabody targeting Rev displays broad-spectrum anti-HIV activity. Antiviral Res 2014; 112:91-102. [DOI: 10.1016/j.antiviral.2014.10.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Revised: 10/10/2014] [Accepted: 10/15/2014] [Indexed: 01/24/2023]
|
187
|
Target silencing of components of the conserved oligomeric Golgi complex impairs HIV-1 replication. Virus Res 2014; 192:92-102. [PMID: 25179963 DOI: 10.1016/j.virusres.2014.08.015] [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] [Received: 02/03/2014] [Revised: 08/21/2014] [Accepted: 08/22/2014] [Indexed: 12/23/2022]
Abstract
All viruses require host cell factors to replicate. A large number of host factors have been identified that participate at numerous points of the human immunodeficiency virus 1 (HIV-1) life cycle. Recent evidence supports a role for components of the trans-Golgi network (TGN) in mediating early steps in the HIV-1 life cycle. The conserved oligomeric Golgi (COG) complex is a heteroctamer complex that functions in coat protein complex I (COPI)-mediated intra-Golgi retrograde trafficking and plays an important role in the maintenance of Golgi structure and integrity as well as glycosylation enzyme homeostasis. The targeted silencing of components of lobe B of the COG complex, namely COG5, COG6, COG7 and COG8, inhibited HIV-1 replication. This inhibition of HIV-1 replication preceded late reverse transcription (RT) but did not affect viral fusion. Silencing of the COG interacting protein the t-SNARE syntaxin 5, showed a similar defect in late RT product formation, strengthening the role of the TGN in HIV replication.
Collapse
|
188
|
Co-expression of foreign proteins tethered to HIV-1 envelope glycoprotein on the cell surface by introducing an intervening second membrane-spanning domain. PLoS One 2014; 9:e96790. [PMID: 24804933 PMCID: PMC4013048 DOI: 10.1371/journal.pone.0096790] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 04/11/2014] [Indexed: 11/19/2022] Open
Abstract
The envelope glycoprotein (Env) of human immunodeficiency virus type I (HIV-1) mediates membrane fusion. To analyze the mechanism of HIV-1 Env-mediated membrane fusion, it is desirable to determine the expression level of Env on the cell surface. However, the quantification of Env by immunological staining is often hampered by the diversity of HIV-1 Env and limited availability of universal antibodies that recognize different Envs with equal efficiency. To overcome this problem, here we linked a tag protein called HaloTag at the C-terminus of HIV-1 Env. To relocate HaloTag to the cell surface, we introduced a second membrane-spanning domain (MSD) between Env and HaloTag. The MSD of transmembrane protease serine 11D, a type II transmembrane protein, successfully relocated HaloTag to the cell surface. The surface level of Env can be estimated indirectly by staining HaloTag with a specific membrane-impermeable fluorescent ligand. This tagging did not compromise the fusogenicity of Env drastically. Furthermore, fusogenicity of Env was preserved even after the labeling with the ligands. We have also found that an additional foreign peptide or protein such as C34 or neutralizing single-chain variable fragment (scFv) can be linked to the C-terminus of the HaloTag protein. Using these constructs, we were able to determine the required length of C34 and critical residues of neutralizing scFv for blocking membrane fusion, respectively.
Collapse
|
189
|
Calistri A, Munegato D, Carli I, Parolin C, Palù G. The ubiquitin-conjugating system: multiple roles in viral replication and infection. Cells 2014; 3:386-417. [PMID: 24805990 PMCID: PMC4092849 DOI: 10.3390/cells3020386] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2014] [Revised: 04/23/2014] [Accepted: 04/24/2014] [Indexed: 12/17/2022] Open
Abstract
Through the combined action of ubiquitinating and deubiquitinating enzymes, conjugation of ubiquitin to a target protein acts as a reversible post-translational modification functionally similar to phosphorylation. Indeed, ubiquitination is more and more recognized as a central process for the fine regulation of many cellular pathways. Due to their nature as obligate intracellular parasites, viruses rely on the most conserved host cell machineries for their own replication. Thus, it is not surprising that members from almost every viral family are challenged by ubiquitin mediated mechanisms in different steps of their life cycle and have evolved in order to by-pass or exploit the cellular ubiquitin conjugating system to maximize their chance to establish a successful infection. In this review we will present several examples of the complex interplay that links viruses and the ubiquitin conjugation machinery, with a special focus on the mechanisms evolved by the human immunodeficiency virus to escape from cellular restriction factors and to exit from infected cells.
Collapse
Affiliation(s)
- Arianna Calistri
- Department of Molecular Medicine, University of Padova, via Gabelli 63, Padova 35121, Italy.
| | - Denis Munegato
- Department of Molecular Medicine, University of Padova, via Gabelli 63, Padova 35121, Italy.
| | - Ilaria Carli
- Department of Molecular Medicine, University of Padova, via Gabelli 63, Padova 35121, Italy.
| | - Cristina Parolin
- Department of Molecular Medicine, University of Padova, via Gabelli 63, Padova 35121, Italy.
| | - Giorgio Palù
- Department of Molecular Medicine, University of Padova, via Gabelli 63, Padova 35121, Italy.
| |
Collapse
|
190
|
Wang N, Li Y, Niu W, Sun M, Cerny R, Li Q, Guo J. Construction of a live-attenuated HIV-1 vaccine through genetic code expansion. Angew Chem Int Ed Engl 2014; 53:4867-71. [PMID: 24715496 PMCID: PMC4984542 DOI: 10.1002/anie.201402092] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Indexed: 11/10/2022]
Abstract
A safe and effective vaccine against human immunodeficiency virus type 1 (HIV-1) is urgently needed to combat the worldwide AIDS pandemic, but still remains elusive. The fact that uncontrolled replication of an attenuated vaccine can lead to regaining of its virulence creates safety concerns precluding many vaccines from clinical application. We introduce a novel approach to control HIV-1 replication, which entails the manipulation of essential HIV-1 protein biosynthesis through unnatural amino acid (UAA*)-mediated suppression of genome-encoded blank codon. We successfully demonstrate that HIV-1 replication can be precisely turned on and off in vitro.
Collapse
Affiliation(s)
- Nanxi Wang
- Department of Chemistry, University of Nebraska–Lincoln, Lincoln, NE 68588 (USA)
| | - Yue Li
- Nebraska Center for Virology & School of Biological Sciences, University of Nebraska–Lincoln, Lincoln, NE 68588 (USA)
| | - Wei Niu
- Department of Chemistry, University of Nebraska–Lincoln, Lincoln, NE 68588 (USA)
| | - Ming Sun
- Nebraska Center for Virology & School of Biological Sciences, University of Nebraska–Lincoln, Lincoln, NE 68588 (USA)
| | - Ronald Cerny
- Department of Chemistry, University of Nebraska–Lincoln, Lincoln, NE 68588 (USA)
| | - Qingsheng Li
- Department of Chemistry, University of Nebraska–Lincoln, Lincoln, NE 68588 (USA)
| | - Jiantao Guo
- Department of Chemistry, University of Nebraska–Lincoln, Lincoln, NE 68588 (USA)
| |
Collapse
|
191
|
Wang N, Li Y, Niu W, Sun M, Cerny R, Li Q, Guo J. Construction of a Live-Attenuated HIV-1 Vaccine through Genetic Code Expansion. Angew Chem Int Ed Engl 2014. [DOI: 10.1002/ange.201402092] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|