1
|
Schemelev AN, Davydenko VS, Ostankova YV, Reingardt DE, Serikova EN, Zueva EB, Totolian AA. Involvement of Human Cellular Proteins and Structures in Realization of the HIV Life Cycle: A Comprehensive Review, 2024. Viruses 2024; 16:1682. [PMID: 39599797 PMCID: PMC11599013 DOI: 10.3390/v16111682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 10/25/2024] [Accepted: 10/26/2024] [Indexed: 11/29/2024] Open
Abstract
Human immunodeficiency virus (HIV) continues to be a global health challenge, with over 38 million people infected by the end of 2022. HIV-1, the predominant strain, primarily targets and depletes CD4+ T cells, leading to immunodeficiency and subsequent vulnerability to opportunistic infections. Despite the progress made in antiretroviral therapy (ART), drug resistance and treatment-related toxicity necessitate novel therapeutic strategies. This review delves into the intricate interplay between HIV-1 and host cellular proteins throughout the viral life cycle, highlighting key host factors that facilitate viral entry, replication, integration, and immune evasion. A focus is placed on actual findings regarding the preintegration complex, nuclear import, and the role of cellular cofactors such as FEZ1, BICD2, and NPC components in viral transport and genome integration. Additionally, the mechanisms of immune evasion via HIV-1 proteins Nef and Vpu, and their interaction with host MHC molecules and interferon signaling pathways, are explored. By examining these host-virus interactions, this review underscores the importance of host-targeted therapies in complementing ART, with a particular emphasis on the potential of genetic research and host protein stability in developing innovative treatments for HIV/AIDS.
Collapse
Affiliation(s)
- Alexandr N. Schemelev
- St. Petersburg Pasteur Institute, St. Petersburg 197101, Russia; (V.S.D.); (Y.V.O.); (D.E.R.); (E.N.S.); (E.B.Z.); (A.A.T.)
| | | | | | | | | | | | | |
Collapse
|
2
|
Complex Relationships between HIV-1 Integrase and Its Cellular Partners. Int J Mol Sci 2022; 23:ijms232012341. [PMID: 36293197 PMCID: PMC9603942 DOI: 10.3390/ijms232012341] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/09/2022] [Accepted: 10/11/2022] [Indexed: 11/06/2022] Open
Abstract
RNA viruses, in pursuit of genome miniaturization, tend to employ cellular proteins to facilitate their replication. HIV-1, one of the most well-studied retroviruses, is not an exception. There is numerous evidence that the exploitation of cellular machinery relies on nucleic acid-protein and protein-protein interactions. Apart from Vpr, Vif, and Nef proteins that are known to regulate cellular functioning via interaction with cell components, another viral protein, integrase, appears to be crucial for proper virus-cell dialog at different stages of the viral life cycle. The goal of this review is to summarize and systematize existing data on known cellular partners of HIV-1 integrase and their role in the HIV-1 life cycle.
Collapse
|
3
|
Sui H, Hao M, Chang W, Imamichi T. The Role of Ku70 as a Cytosolic DNA Sensor in Innate Immunity and Beyond. Front Cell Infect Microbiol 2021; 11:761983. [PMID: 34746031 PMCID: PMC8566972 DOI: 10.3389/fcimb.2021.761983] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 10/06/2021] [Indexed: 12/24/2022] Open
Abstract
Human Ku70 is a well-known endogenous nuclear protein involved in the non-homologous end joining pathway to repair double-stranded breaks in DNA. However, Ku70 has been studied in multiple contexts and grown into a multifunctional protein. In addition to the extensive functional study of Ku70 in DNA repair process, many studies have emphasized the role of Ku70 in various other cellular processes, including apoptosis, aging, and HIV replication. In this review, we focus on discussing the role of Ku70 in inducing interferons and proinflammatory cytokines as a cytosolic DNA sensor. We explored the unique structure of Ku70 binding with DNA; illustrated, with evidence, how Ku70, as a nuclear protein, responds to extracellular DNA stimulation; and summarized the mechanisms of the Ku70-involved innate immune response pathway. Finally, we discussed several new strategies to modulate Ku70-mediated innate immune response and highlighted some potential physiological insights based on the role of Ku70 in innate immunity.
Collapse
Affiliation(s)
- Hongyan Sui
- Laboratory of Human Retrovirology and Immunoinformatics, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | | | | | - Tomozumi Imamichi
- Laboratory of Human Retrovirology and Immunoinformatics, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| |
Collapse
|
4
|
Hristova DB, Lauer KB, Ferguson BJ. Viral interactions with non-homologous end-joining: a game of hide-and-seek. J Gen Virol 2020; 101:1133-1144. [PMID: 32735206 PMCID: PMC7879558 DOI: 10.1099/jgv.0.001478] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 07/14/2020] [Indexed: 02/06/2023] Open
Abstract
There are extensive interactions between viruses and the host DNA damage response (DDR) machinery. The outcome of these interactions includes not only direct effects on viral nucleic acids and genome replication, but also the activation of host stress response signalling pathways that can have further, indirect effects on viral life cycles. The non-homologous end-joining (NHEJ) pathway is responsible for the rapid and imprecise repair of DNA double-stranded breaks in the nucleus that would otherwise be highly toxic. Whilst directly repairing DNA, components of the NHEJ machinery, in particular the DNA-dependent protein kinase (DNA-PK), can activate a raft of downstream signalling events that activate antiviral, cell cycle checkpoint and apoptosis pathways. This combination of possible outcomes results in NHEJ being pro- or antiviral depending on the infection. In this review we will describe the broad range of interactions between NHEJ components and viruses and their consequences for both host and pathogen.
Collapse
Affiliation(s)
- Dayana B. Hristova
- Department of Pathology, Division of Immunology, University of Cambridge, Cambridge, UK
| | - Katharina B. Lauer
- Department of Pathology, Division of Immunology, University of Cambridge, Cambridge, UK
- Present address: ELIXIR Hub, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Brian J. Ferguson
- Department of Pathology, Division of Immunology, University of Cambridge, Cambridge, UK
| |
Collapse
|
5
|
Bai L, Hirose T, Assi W, Wada S, Takeshima SN, Aida Y. Bovine Leukemia Virus Infection Affects Host Gene Expression Associated with DNA Mismatch Repair. Pathogens 2020; 9:pathogens9110909. [PMID: 33143351 PMCID: PMC7694100 DOI: 10.3390/pathogens9110909] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 10/28/2020] [Accepted: 10/29/2020] [Indexed: 01/17/2023] Open
Abstract
Bovine leukemia virus (BLV) causes enzootic bovine leukosis, a malignant form of B-cell lymphoma, and is closely related to human T-cell leukemia viruses. We investigated whether BLV infection affects host genes associated with DNA mismatch repair (MMR). Next-generation sequencing of blood samples from five calves experimentally infected with BLV revealed the highest expression levels of seven MMR genes (EXO1, UNG, PCNA, MSH2, MSH3, MSH6, and PMS2) at the point of peak proviral loads (PVLs). Furthermore, MMR gene expression was only upregulated in cattle with higher PVLs. In particular, the expression levels of MSH2, MSH3, and UNG positively correlated with PVL in vivo. The expression levels of all seven MMR genes in pig kidney-15 cells and the levels of PMS2 and EXO1 in HeLa cells also increased tendencies after transient transfection with a BLV infectious clone. Moreover, MMR gene expression levels were significantly higher in BLV-expressing cell lines compared with those in the respective parental cell lines. Expression levels of MSH2 and EXO1 in BLV-infected cattle with lymphoma were significantly lower and higher, respectively, compared with those in infected cattle in vivo. These results reveal that BLV infection affects MMR gene expression, offering new candidate markers for lymphoma diagnosis.
Collapse
Affiliation(s)
- Lanlan Bai
- Photonics Control Technology Team, RIKEN Center for Advanced Photonics, Wako 351-0198, Japan; (L.B.); (W.A.); (S.W.); (S.-n.T.)
- Viral Infectious Diseases Unit, RIKEN, Wako 351-0198, Japan;
| | - Tomoya Hirose
- Viral Infectious Diseases Unit, RIKEN, Wako 351-0198, Japan;
- Laboratory of Viral Infectious Diseases, Department of Medical Genome Sciences, Graduate School of Frontier Science, The University of Tokyo, Tokyo 113-8657, Japan
| | - Wlaa Assi
- Photonics Control Technology Team, RIKEN Center for Advanced Photonics, Wako 351-0198, Japan; (L.B.); (W.A.); (S.W.); (S.-n.T.)
- Viral Infectious Diseases Unit, RIKEN, Wako 351-0198, Japan;
- Laboratory of Viral Infectious Diseases, Department of Medical Genome Sciences, Graduate School of Frontier Science, The University of Tokyo, Tokyo 113-8657, Japan
| | - Satoshi Wada
- Photonics Control Technology Team, RIKEN Center for Advanced Photonics, Wako 351-0198, Japan; (L.B.); (W.A.); (S.W.); (S.-n.T.)
| | - Shin-nosuke Takeshima
- Photonics Control Technology Team, RIKEN Center for Advanced Photonics, Wako 351-0198, Japan; (L.B.); (W.A.); (S.W.); (S.-n.T.)
- Viral Infectious Diseases Unit, RIKEN, Wako 351-0198, Japan;
- Department of Food and Nutrition, Faculty of Human Life, Jumonji University, Niiza 352-0017, Japan
| | - Yoko Aida
- Viral Infectious Diseases Unit, RIKEN, Wako 351-0198, Japan;
- Laboratory of Viral Infectious Diseases, Department of Medical Genome Sciences, Graduate School of Frontier Science, The University of Tokyo, Tokyo 113-8657, Japan
- Nakamura Laboratory, Baton Zone Program, RIKEN Cluster for Science, Technology and Innovation Hub, Wako 351-0198, Japan
- Correspondence: ; Tel.: +81-48-462-4418
| |
Collapse
|
6
|
Anisenko A, Kan M, Shadrina O, Brattseva A, Gottikh M. Phosphorylation Targets of DNA-PK and Their Role in HIV-1 Replication. Cells 2020; 9:E1907. [PMID: 32824372 PMCID: PMC7464883 DOI: 10.3390/cells9081907] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 08/13/2020] [Accepted: 08/14/2020] [Indexed: 02/07/2023] Open
Abstract
The DNA dependent protein kinase (DNA-PK) is a trimeric nuclear complex consisting of a large protein kinase and the Ku heterodimer. The kinase activity of DNA-PK is required for efficient repair of DNA double-strand breaks (DSB) by non-homologous end joining (NHEJ). We also showed that the kinase activity of DNA-PK is essential for post-integrational DNA repair in the case of HIV-1 infection. Besides, DNA-PK is known to participate in such cellular processes as protection of mammalian telomeres, transcription, and some others where the need for its phosphorylating activity is not clearly elucidated. We carried out a systematic search and analysis of DNA-PK targets described in the literature and identified 67 unique DNA-PK targets phosphorylated in response to various in vitro and/or in vivo stimuli. A functional enrichment analysis of DNA-PK targets and determination of protein-protein associations among them were performed. For 27 proteins from these 67 DNA-PK targets, their participation in the HIV-1 life cycle was demonstrated. This information may be useful for studying the functioning of DNA-PK in various cellular processes, as well as in various stages of HIV-1 replication.
Collapse
Affiliation(s)
- Andrey Anisenko
- Chemistry Department and Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119234, Russia; (O.S.); (M.G.)
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow 119234, Russia;; (M.K.); (A.B.)
| | - Marina Kan
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow 119234, Russia;; (M.K.); (A.B.)
| | - Olga Shadrina
- Chemistry Department and Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119234, Russia; (O.S.); (M.G.)
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow 119234, Russia;; (M.K.); (A.B.)
| | - Anna Brattseva
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow 119234, Russia;; (M.K.); (A.B.)
| | - Marina Gottikh
- Chemistry Department and Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119234, Russia; (O.S.); (M.G.)
| |
Collapse
|
7
|
Shadrina O, Garanina I, Korolev S, Zatsepin T, Van Assche J, Daouad F, Wallet C, Rohr O, Gottikh M. Analysis of RNA binding properties of human Ku protein reveals its interactions with 7SK snRNA and protein components of 7SK snRNP complex. Biochimie 2020; 171-172:110-123. [PMID: 32105815 DOI: 10.1016/j.biochi.2020.02.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 02/21/2020] [Indexed: 12/21/2022]
Abstract
Human Ku heterodimeric protein composed of Ku70 and Ku80 subunits plays an important role in the non-homologous end-joining DNA repair pathway as a sensor of double strand DNA breaks. Ku is also involved in numerous cellular processes, and in some of them it acts in an RNA-dependent manner. However, RNA binding properties of the human Ku have not been well studied. Here we have analyzed interactions of a recombinant Ku heterodimer with a set of RNAs of various structure as well as eCLIP (enhanced crosslinking and immunoprecipitation) data for human Ku70. As a result, we have proposed a consensus RNA structure preferable for the Ku binding that is a hairpin possessing a bulge just near GpG sequence-containing terminal loop. 7SK snRNA is a scaffold for a ribonucleoprotein complex (7SK snRNP), which is known to participate in transcription regulation. We have shown that the recombinant Ku specifically binds a G-rich loop of hairpin 1 within 7SK snRNA. Moreover, Ku protein has been co-precipitated from HEK 293T cells with endogenous 7SK snRNA and such proteins included in 7SK snRNP as HEXIM1, Cdk9 and CTIP2. Ku and Cdk9 binding is found to be RNA-independent, meanwhile HEXIM1 and Ku co-precipitation depended on the presence of intact 7SK snRNA. The latter result has been confirmed using recombinant HEXIM1 and Ku proteins. Colocalization of Ku and CTIP2 was additionally confirmed by confocal microscopy. These results allow us to propose human Ku as a new component of the 7SK snRNP complex.
Collapse
Affiliation(s)
- Olga Shadrina
- Chemistry Department, Lomonosov Moscow State University, Moscow, 199991, Russia; Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992, Moscow, Russia.
| | - Irina Garanina
- Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, 119435, Russia
| | - Sergey Korolev
- Chemistry Department, Lomonosov Moscow State University, Moscow, 199991, Russia; Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992, Moscow, Russia
| | - Timofei Zatsepin
- Chemistry Department, Lomonosov Moscow State University, Moscow, 199991, Russia; Skolkovo Institute of Science and Technology, Skolkovo, 121205, Russia
| | - Jeanne Van Assche
- Université de Strasbourg, EA7292, FMTS, IUT Louis Pasteur, Schiltigheim, France
| | - Fadoua Daouad
- Université de Strasbourg, EA7292, FMTS, IUT Louis Pasteur, Schiltigheim, France
| | - Clementine Wallet
- Université de Strasbourg, EA7292, FMTS, IUT Louis Pasteur, Schiltigheim, France
| | - Olivier Rohr
- Université de Strasbourg, EA7292, FMTS, IUT Louis Pasteur, Schiltigheim, France
| | - Marina Gottikh
- Chemistry Department, Lomonosov Moscow State University, Moscow, 199991, Russia; Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992, Moscow, Russia
| |
Collapse
|
8
|
Knyazhanskaya E, Anisenko A, Shadrina O, Kalinina A, Zatsepin T, Zalevsky A, Mazurov D, Gottikh M. NHEJ pathway is involved in post-integrational DNA repair due to Ku70 binding to HIV-1 integrase. Retrovirology 2019; 16:30. [PMID: 31690330 PMCID: PMC6833283 DOI: 10.1186/s12977-019-0492-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 10/23/2019] [Indexed: 12/12/2022] Open
Abstract
Background HIV-1 integration results in genomic DNA gaps that are repaired by cellular DNA repair pathways. This step of the lentiviral life cycle remains poorly understood despite its crucial importance for successful replication. We and others reported that Ku70 protein of the non-homologous end joining pathway (NHEJ) directly binds HIV-1 integrase (IN). Here, we studied the importance of this interaction for post-integrational gap repair and the recruitment of NHEJ factors in this process. Results We engineered HIV-based pseudovirus with mutant IN defective in Ku70 binding and generated heterozygous Ku70, Ku80 and DNA-PKcs human knockout (KO) cells using CRISPR/Cas9. KO of either of these proteins or inhibition of DNA-PKcs catalytic activity substantially decreased the infectivity of HIV-1 with native IN but not with the mutant one. We used a recently developed qPCR assay for the measurement of gap repair efficiency to show that HIV-1 with mutant IN was defective in DNA post-integrational repair, whereas the wild type virus displayed such a defect only when NHEJ system was disrupted in any way. This effect was present in CRISPR/Cas9 modified 293T cells, in Jurkat and CEM lymphoid lines and in primary human PBMCs. Conclusions Our data provide evidence that IN recruits DNA-PK to the site of HIV-1 post-integrational repair due to Ku70 binding—a novel finding that explains the involvement of DNA-PK despite the absence of free double stranded DNA breaks. In addition, our data clearly indicate the importance of interactions between HIV-1 IN and Ku70 in HIV-1 replication at the post-integrational repair step.
Collapse
Affiliation(s)
- Ekaterina Knyazhanskaya
- Chemistry Department, Lomonosov Moscow State University, Moscow, 199234, Russia. .,Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119234, Russia. .,Department of Biochemistry and Molecular Biology, Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, TX, 77555, USA.
| | - Andrey Anisenko
- Chemistry Department, Lomonosov Moscow State University, Moscow, 199234, Russia. .,Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119234, Russia.
| | - Olga Shadrina
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, 119234, Russia
| | - Anastasia Kalinina
- Federal State Budgetary Institution « N.N. Blokhin National Medical Research Center of Oncology » of the Ministry of Health of the Russian Federation, Moscow, 115478, Russia
| | - Timofei Zatsepin
- Chemistry Department, Lomonosov Moscow State University, Moscow, 199234, Russia.,Skolkovo Institute of Science and Technology, Skolkovo, 121205, Russia
| | - Arthur Zalevsky
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, 119234, Russia
| | - Dmitriy Mazurov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, RAS, Moscow, 119334, Russia.,NRC Institute of Immunology FMBA of Russia, Moscow, 115478, Russia
| | - Marina Gottikh
- Chemistry Department, Lomonosov Moscow State University, Moscow, 199234, Russia.,Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
| |
Collapse
|
9
|
|
10
|
Targeting the DNA-PK complex: Its rationale use in cancer and HIV-1 infection. Biochem Pharmacol 2018; 160:80-91. [PMID: 30529192 DOI: 10.1016/j.bcp.2018.12.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 12/04/2018] [Indexed: 02/07/2023]
Abstract
The DNA-PK complex is the major component of the predominant mechanism of DSB repair in humans. In addition, this complex is involved in many other processes such as DNA recombination, genome maintenance, apoptosis and transcription regulation. Several studies have linked the decrease of the DNA-PK activity with cancer initiation, due to defects in the repair. On another hand, higher DNA-PK expression and activity have been observed in various other tumor cells and have been linked with a decrease of the efficiency of anti-tumor drugs. It has also been shown that DNA-PK is critical for the integration of the HIV-1 DNA into the cell host genome and promotes replication and transcription of the virus. Targeting this complex makes therefore sense to treat these two pathologies. However, according to the status of HIV-1 replication (active versus latent replication) or to the tumor grade cells (initiation versus metastasis), the way to target this DNA-PK complex might be rather different. In this review, we discuss the importance of DNA-PK complex in two major pathologies i.e. HIV-1 infection and cancer, and the rationale use of therapies aiming to target this complex.
Collapse
|
11
|
Characterization of HIV-1 integrase interaction with human Ku70 protein and initial implications for drug targeting. Sci Rep 2017; 7:5649. [PMID: 28717247 PMCID: PMC5514147 DOI: 10.1038/s41598-017-05659-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 06/01/2017] [Indexed: 11/27/2022] Open
Abstract
Human Ku70/Ku80 protein is known to influence HIV-1 replication. One of the possible reasons may be the protection of integrase from proteasomal degradation by Ku70 subunit. We demonstrated that recombinant HIV-1 integrase and Ku70 form a stable complex, while no interaction of Ku70 with integrase from prototype foamy virus was observed. By analyzing protein subdomains we determined two binding sites in the structure of both Ku70 and integrase: the 51–160 a.a. region of integrase interacts with residues 251–438 of Ku70, whereas Ku70 N-terminal domain (1–250 a.a.) contacts an α6-helix in the 200–220 a.a. integrase region. Single substitutions within integrase (E212A or L213A) block the interaction with Ku70 thus indicating that the binding site formed by the 200–220 a.a. integrase region is crucial for complex formation. E212A/L213A substitutions decreased the integrase capacity to bind Ku70 in HEK293T cells. A conjugate of 2′-ОMe-GGUUUUUGUGU oligonucleotide with eosin is shown by molecular modeling to shield integrase residues E212/L213 and is effective in blocking complex formation of Ku70 with integrase what makes the complex between α6-helix and Ku70(1–250) a possible target for drug development.
Collapse
|
12
|
Knyazhanskaya ES, Shadrina OA, Anisenko AN, Gottikh MB. Role of DNA-dependent protein kinase in the HIV-1 replication cycle. Mol Biol 2016. [DOI: 10.1134/s0026893316040075] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
13
|
Activation of the DNA Damage Response by RNA Viruses. Biomolecules 2016; 6:2. [PMID: 26751489 PMCID: PMC4808796 DOI: 10.3390/biom6010002] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 11/17/2015] [Accepted: 11/24/2015] [Indexed: 12/11/2022] Open
Abstract
RNA viruses are a genetically diverse group of pathogens that are responsible for some of the most prevalent and lethal human diseases. Numerous viruses introduce DNA damage and genetic instability in host cells during their lifecycles and some species also manipulate components of the DNA damage response (DDR), a complex and sophisticated series of cellular pathways that have evolved to detect and repair DNA lesions. Activation and manipulation of the DDR by DNA viruses has been extensively studied. It is apparent, however, that many RNA viruses can also induce significant DNA damage, even in cases where viral replication takes place exclusively in the cytoplasm. DNA damage can contribute to the pathogenesis of RNA viruses through the triggering of apoptosis, stimulation of inflammatory immune responses and the introduction of deleterious mutations that can increase the risk of tumorigenesis. In addition, activation of DDR pathways can contribute positively to replication of viral RNA genomes. Elucidation of the interactions between RNA viruses and the DDR has provided important insights into modulation of host cell functions by these pathogens. This review summarises the current literature regarding activation and manipulation of the DDR by several medically important RNA viruses.
Collapse
|
14
|
Shadrina OA, Knyazhanskaya ES, Korolev S, Gottikh MB. Host Proteins Ku and HMGA1 As Participants of HIV-1 Transcription. Acta Naturae 2016; 8:34-47. [PMID: 27099783 PMCID: PMC4837570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Human immunodeficiency virus type 1 is known to use the transcriptional machinery of the host cell for viral gene transcription, and the only viral protein that partakes in this process is Tat, the viral trans-activator of transcription. During acute infection, the binding of Tat to the hairpin at the beginning of the transcribed viral RNA recruits the PTEFb complex, which in turn hyperphosphorylates RNA-polymerase II and stimulates transcription elongation. Along with acute infection, HIV-1 can also lead to latent infection that is characterized by a low level of viral transcription. During the maintenance and reversal of latency, there are no detectable amounts of Tat protein in the cell and the mechanism of transcription activation in the absence of Tat protein remains unclear. The latency maintenance is also a problematic question. It seems evident that cellular proteins with a yet unknown nature or role regulate both transcriptional repression in the latent phase and its activation during transition into the lytic phase. The present review discusses the role of cellular proteins Ku and HMGA1 in the initiation of transcription elongation of the HIV-1 provirus. The review presents data regarding Ku-mediated HIV-1 transcription and its dependence on the promoter structure and the shape of viral DNA. We also describe the differential influence of the HMGA1 protein on the induced and basal transcription of HIV-1. Finally, we offer possible mechanisms for Ku and HMGA1 proteins in the proviral transcription regulation.
Collapse
Affiliation(s)
- O. A. Shadrina
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Leninskie Gory, Moscow, 119991, Russia
| | - E. S. Knyazhanskaya
- Chemistry Department, Lomonosov Moscow State University, Leninskie Gory, Moscow, 119991, Russia
| | - S.P. Korolev
- Chemistry Department, Lomonosov Moscow State University, Leninskie Gory, Moscow, 119991, Russia
| | - M. B. Gottikh
- Belozersky Institute of Physical-Chemical Biology, Lomonosov Moscow State University, Leninskie Gory, Moscow, Russia; 119991
| |
Collapse
|
15
|
Stake M, Singh D, Singh G, Marcela Hernandez J, Kaddis Maldonado R, Parent LJ, Boris-Lawrie K. HIV-1 and two avian retroviral 5' untranslated regions bind orthologous human and chicken RNA binding proteins. Virology 2015; 486:307-20. [PMID: 26584240 DOI: 10.1016/j.virol.2015.06.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 05/31/2015] [Accepted: 06/01/2015] [Indexed: 01/12/2023]
Abstract
Essential host cofactors in retrovirus replication bind cis-acting sequences in the 5'untranslated region (UTR). Although host RBPs are crucial to all aspects of virus biology, elucidating their roles in replication remains a challenge to the field. Here RNA affinity-coupled-proteomics generated a comprehensive, unbiased inventory of human and avian RNA binding proteins (RBPs) co-isolating with 5'UTRs of HIV-1, spleen necrosis virus and Rous sarcoma virus. Applying stringent biochemical and statistical criteria, we identified 185 RBP; 122 were previously implicated in retrovirus biology and 63 are new to the 5'UTR proteome. RNA electrophoretic mobility assays investigated paralogs present in the common ancestor of vertebrates and one hnRNP was identified as a central node to the biological process-anchored networks of HIV-1, SNV, and RSV 5' UTR-proteomes. This comprehensive view of the host constituents of retroviral RNPs is broadly applicable to investigation of viral replication and antiviral response in both human and avian cell lineages.
Collapse
Affiliation(s)
- Matthew Stake
- Department of Medicine, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033, USA.
| | - Deepali Singh
- School of Biotechnology, Gautam Buddha University, Greater Noida, 201312, India.
| | - Gatikrushna Singh
- Department Veterinary & Biomedical Sciences, University of Minnesota, 205 VSB, 1971 Commonwealth Avenue, Saint Paul, MN 55108.
| | - J Marcela Hernandez
- Department of Veterinary Biosciences, Center for Retrovirus Research, Center for RNA Biology, Comprehensive Cancer Center, Ohio State University, Columbus, OH, USA.
| | - Rebecca Kaddis Maldonado
- Department of Medicine, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033, USA.
| | - Leslie J Parent
- Department of Medicine, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033, USA; Department Microbiology & Immunology, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033, USA.
| | - Kathleen Boris-Lawrie
- Department Veterinary & Biomedical Sciences, University of Minnesota, 205 VSB, 1971 Commonwealth Avenue, Saint Paul, MN 55108.
| |
Collapse
|
16
|
AAV-mediated in vivo functional selection of tissue-protective factors against ischaemia. Nat Commun 2015; 6:7388. [PMID: 26066847 PMCID: PMC4477044 DOI: 10.1038/ncomms8388] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 05/05/2015] [Indexed: 02/08/2023] Open
Abstract
Functional screening of expression libraries in vivo would offer the possibility of identifying novel biotherapeutics without a priori knowledge of their biochemical function. Here we describe a procedure for the functional selection of tissue-protective factors based on the in vivo delivery of arrayed cDNA libraries from the mouse secretome using adeno-associated virus (AAV) vectors. Application of this technique, which we call FunSel, in the context of acute ischaemia, revealed that the peptide ghrelin protects skeletal muscle and heart from ischaemic damage. When delivered to the heart using an AAV9 vector, ghrelin markedly reduces infarct size and preserves cardiac function over time. This protective activity associates with the capacity of ghrelin to sustain autophagy and remove dysfunctional mitochondria after myocardial infarction. Our findings describe an innovative tool to identify biological therapeutics and reveal a novel role of ghrelin as an inducer of myoprotective autophagy. Cell-based screening assays allow functional testing of chemicals but do not mimic the in vivo situation well. Here, the authors report a method for the discovery of secreted cytoprotective factors in mice and use it to demonstrate that the hormone ghrelin protects cardiac muscle from ischaemic damage.
Collapse
|
17
|
Abstract
Ribozymes are structured RNA molecules that act as catalysts in different biological reactions. From simple genome cleaving activities in satellite RNAs to more complex functions in cellular protein synthesis and gene regulation, ribozymes play important roles in all forms of life. Several naturally existing ribozymes have been modified for use as therapeutics in different conditions, with HIV-1 infection being one of the most studied. This chapter summarizes data from different preclinical and clinical studies conducted to evaluate the potential of ribozymes to be used in HIV-1 therapies. The different ribozyme motifs that have been modified, as well as their target sites and expression strategies, are described. RNA conjugations used to enhance the antiviral effect of ribozymes are also presented and the results from clinical trials conducted to date are summarized. Studies on anti-HIV-1 ribozymes have provided valuable information on the optimal expression strategies and clinical protocols for RNA gene therapy and remain competitive candidates for future therapy.
Collapse
|
18
|
Che P, Tang H, Li Q. The interaction between claudin-1 and dengue viral prM/M protein for its entry. Virology 2013; 446:303-13. [PMID: 24074594 DOI: 10.1016/j.virol.2013.08.009] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Revised: 07/18/2013] [Accepted: 08/09/2013] [Indexed: 02/04/2023]
Abstract
Dengue disease is becoming a huge public health concern around the world as more than one-third of the world's population living in areas at risk of infection. In an effort to assess host factors interacting with dengue virus, we identified claudin-1, a major tight junction component, as an essential cell surface protein for dengue virus entry. When claudin-1 was knocked down in Huh 7.5 cells via shRNA, the amount of dengue virus entering host cells was reduced. Consequently, the progeny virus productions were decreased and dengue virus-induced CPE was prevented. Furthermore, restoring the expression of claudin-1 in the knockdown cells facilitated dengue virus entry. The interaction between claudin-1 and dengue viral prM protein was further demonstrated using the pull-down assay. Deletion of the extracellular loop 1 (ECL1) of claudin-1 abolished such interaction, so did point mutations C54A, C64A and I32M on ECL1. These results suggest that the interaction between viral protein prM and host protein claudin-1 was essential for dengue entry. Since host and viral factors involved in virus entry are promising therapeutic targets, determining the essential role of claudin-1 could lead to the discovery of entry inhibitors with attractive therapeutic potential against dengue disease.
Collapse
Affiliation(s)
- Pulin Che
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, United States; University of Alabama at Birmingham, Department of Medicine, Division of Infectious Diseases, BBRB 562, 845 19th Street South, Birmingham, AL 35294, United States
| | | | | |
Collapse
|
19
|
Manic G, Maurin-Marlin A, Laurent F, Vitale I, Thierry S, Delelis O, Dessen P, Vincendeau M, Leib-Mösch C, Hazan U, Mouscadet JF, Bury-Moné S. Impact of the Ku complex on HIV-1 expression and latency. PLoS One 2013; 8:e69691. [PMID: 23922776 PMCID: PMC3726783 DOI: 10.1371/journal.pone.0069691] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Accepted: 06/17/2013] [Indexed: 01/20/2023] Open
Abstract
Ku, a cellular complex required for human cell survival and involved in double strand break DNA repair and multiple other cellular processes, may modulate retroviral multiplication, although the precise mechanism through which it acts is still controversial. Recently, Ku was identified as a possible anti-human immunodeficiency virus type 1 (HIV-1) target in human cells, in two global approaches. Here we investigated the role of Ku on the HIV-1 replication cycle by analyzing the expression level of a panel of non-replicative lentiviral vectors expressing the green fluorescent protein in human colorectal carcinoma HCT 116 cells, stably or transiently depleted of Ku. We found that in this cellular model the depletion of Ku did not affect the efficiency of (pre-)integrative steps but decreased the early HIV-1 expression by acting at the transcriptional level. This negative effect was specific of the HIV-1 promoter, required the obligatory step of viral DNA integration and was reversed by transient depletion of p53. We also provided evidence on a direct binding of Ku to HIV-1 LTR in transduced cells. Ku not only promotes the early transcription from the HIV-1 promoter, but also limits the constitution of viral latency. Moreover, in the presence of a normal level of Ku, HIV-1 expression was gradually lost over time, likely due to the counter-selection of HIV-1-expressing cells. On the contrary, the reactivation of transgene expression from HIV-1 by means of trichostatin A- or tumor necrosis factor α-administration was enhanced under condition of Ku haplodepletion, suggesting a phenomenon of provirus latency. These observations plead in favor of the hypothesis that Ku has an impact on HIV-1 expression and latency at early- and mid-time after integration.
Collapse
Affiliation(s)
- Gwenola Manic
- Laboratoire de Biologie et Pharmacologie Appliquée, Centre national de la recherche scientifique-UMR8113, Ecole Normale Supérieure de Cachan, Cachan, France
| | - Aurélie Maurin-Marlin
- Laboratoire de Biologie et Pharmacologie Appliquée, Centre national de la recherche scientifique-UMR8113, Ecole Normale Supérieure de Cachan, Cachan, France
| | - Fanny Laurent
- Laboratoire de Biologie et Pharmacologie Appliquée, Centre national de la recherche scientifique-UMR8113, Ecole Normale Supérieure de Cachan, Cachan, France
| | - Ilio Vitale
- Regina Elena National Cancer Institute, Rome, Italy
- National Institute of Health, Rome, Italy
| | - Sylvain Thierry
- Laboratoire de Biologie et Pharmacologie Appliquée, Centre national de la recherche scientifique-UMR8113, Ecole Normale Supérieure de Cachan, Cachan, France
| | - Olivier Delelis
- Laboratoire de Biologie et Pharmacologie Appliquée, Centre national de la recherche scientifique-UMR8113, Ecole Normale Supérieure de Cachan, Cachan, France
| | - Philippe Dessen
- Institut Gustave Roussy, Villejuif, France
- Institut National de la Santé et de la Recherche Médicale-U985, Villejuif, France
| | - Michelle Vincendeau
- Institute of Virology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Research Unit Cellular Signal Integration, Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Christine Leib-Mösch
- Institute of Virology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Department of Hematology and Oncology, Mannheim Medical Center, University of Heidelberg, Mannheim, Germany
| | - Uriel Hazan
- Laboratoire de Biologie et Pharmacologie Appliquée, Centre national de la recherche scientifique-UMR8113, Ecole Normale Supérieure de Cachan, Cachan, France
| | - Jean-François Mouscadet
- Laboratoire de Biologie et Pharmacologie Appliquée, Centre national de la recherche scientifique-UMR8113, Ecole Normale Supérieure de Cachan, Cachan, France
| | - Stéphanie Bury-Moné
- Laboratoire de Biologie et Pharmacologie Appliquée, Centre national de la recherche scientifique-UMR8113, Ecole Normale Supérieure de Cachan, Cachan, France
- * E-mail:
| |
Collapse
|
20
|
Jarboui MA, Bidoia C, Woods E, Roe B, Wynne K, Elia G, Hall WW, Gautier VW. Nucleolar protein trafficking in response to HIV-1 Tat: rewiring the nucleolus. PLoS One 2012; 7:e48702. [PMID: 23166591 PMCID: PMC3499507 DOI: 10.1371/journal.pone.0048702] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Accepted: 10/03/2012] [Indexed: 12/20/2022] Open
Abstract
The trans-activator Tat protein is a viral regulatory protein essential for HIV-1 replication. Tat trafficks to the nucleoplasm and the nucleolus. The nucleolus, a highly dynamic and structured membrane-less sub-nuclear compartment, is the site of rRNA and ribosome biogenesis and is involved in numerous cellular functions including transcriptional regulation, cell cycle control and viral infection. Importantly, transient nucleolar trafficking of both Tat and HIV-1 viral transcripts are critical in HIV-1 replication, however, the role(s) of the nucleolus in HIV-1 replication remains unclear. To better understand how the interaction of Tat with the nucleolar machinery contributes to HIV-1 pathogenesis, we investigated the quantitative changes in the composition of the nucleolar proteome of Jurkat T-cells stably expressing HIV-1 Tat fused to a TAP tag. Using an organellar proteomic approach based on mass spectrometry, coupled with Stable Isotope Labelling in Cell culture (SILAC), we quantified 520 proteins, including 49 proteins showing significant changes in abundance in Jurkat T-cell nucleolus upon Tat expression. Numerous proteins exhibiting a fold change were well characterised Tat interactors and/or known to be critical for HIV-1 replication. This suggests that the spatial control and subcellular compartimentaliation of these cellular cofactors by Tat provide an additional layer of control for regulating cellular machinery involved in HIV-1 pathogenesis. Pathway analysis and network reconstruction revealed that Tat expression specifically resulted in the nucleolar enrichment of proteins collectively participating in ribosomal biogenesis, protein homeostasis, metabolic pathways including glycolytic, pentose phosphate, nucleotides and amino acids biosynthetic pathways, stress response, T-cell signaling pathways and genome integrity. We present here the first differential profiling of the nucleolar proteome of T-cells expressing HIV-1 Tat. We discuss how these proteins collectively participate in interconnected networks converging to adapt the nucleolus dynamic activities, which favor host biosynthetic activities and may contribute to create a cellular environment supporting robust HIV-1 production.
Collapse
Affiliation(s)
- Mohamed Ali Jarboui
- Centre for Research in Infectious Diseases (CRID), School of Medicine and Medical Science (SMMS), University College Dublin (UCD), Dublin, Ireland
| | - Carlo Bidoia
- Centre for Research in Infectious Diseases (CRID), School of Medicine and Medical Science (SMMS), University College Dublin (UCD), Dublin, Ireland
| | - Elena Woods
- Centre for Research in Infectious Diseases (CRID), School of Medicine and Medical Science (SMMS), University College Dublin (UCD), Dublin, Ireland
| | - Barbara Roe
- Centre for Research in Infectious Diseases (CRID), School of Medicine and Medical Science (SMMS), University College Dublin (UCD), Dublin, Ireland
| | - Kieran Wynne
- Mass Spectrometry Resource (MSR), Conway Institute for Biomolecular and Biomedical Research, University College Dublin (UCD), Dublin, Ireland
| | - Giuliano Elia
- Mass Spectrometry Resource (MSR), Conway Institute for Biomolecular and Biomedical Research, University College Dublin (UCD), Dublin, Ireland
| | - William W. Hall
- Centre for Research in Infectious Diseases (CRID), School of Medicine and Medical Science (SMMS), University College Dublin (UCD), Dublin, Ireland
| | - Virginie W. Gautier
- Centre for Research in Infectious Diseases (CRID), School of Medicine and Medical Science (SMMS), University College Dublin (UCD), Dublin, Ireland
| |
Collapse
|
21
|
Abstract
Hammerhead ribozymes have been extensively used as RNA-inactivating agents for therapy as well as forward genomics. A ribozyme can be designed so as to specifically pair with virtually any target RNA, and cleave the phosphodiester backbone at a specified location, thereby functionally inactivating the RNA. Two major factors that determine whether ribozymes will be effective for posttranscriptional gene silencing are colocalization of the ribozyme and the target RNAs, and the choice of an appropriate target site on the mRNA. Complex secondary structures and the ability to bind to some of the cellular proteins mandate that some RNA sequences could stearically occlude binding of RNA-based antivirals like ribozymes to these sites. The use of ribozyme libraries in cell culture factors in these interactions to select for target sites on the RNA, which are more accessible to RNA-based antivirals like ribozymes or siRNA. This chapter provides a useful guide toward using ribozyme libraries to screen for effective target sites on mRNA.
Collapse
|
22
|
Wu X, Robotham JM, Lee E, Dalton S, Kneteman NM, Gilbert DM, Tang H. Productive hepatitis C virus infection of stem cell-derived hepatocytes reveals a critical transition to viral permissiveness during differentiation. PLoS Pathog 2012; 8:e1002617. [PMID: 22496645 PMCID: PMC3320597 DOI: 10.1371/journal.ppat.1002617] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Accepted: 02/17/2012] [Indexed: 01/14/2023] Open
Abstract
Primary human hepatocytes isolated from patient biopsies represent the most physiologically relevant cell culture model for hepatitis C virus (HCV) infection, but these primary cells are not readily accessible, display individual variability, and are largely refractory to genetic manipulation. Hepatocyte-like cells differentiated from pluripotent stem cells provide an attractive alternative as they not only overcome these shortcomings but can also provide an unlimited source of noncancer cells for both research and cell therapy. Despite its promise, the permissiveness to HCV infection of differentiated human hepatocyte-like cells (DHHs) has not been explored. Here we report a novel infection model based on DHHs derived from human embryonic (hESCs) and induced pluripotent stem cells (iPSCs). DHHs generated in chemically defined media under feeder-free conditions were subjected to infection by both HCV derived in cell culture (HCVcc) and patient-derived virus (HCVser). Pluripotent stem cells and definitive endoderm were not permissive for HCV infection whereas hepatic progenitor cells were persistently infected and secreted infectious particles into culture medium. Permissiveness to infection was correlated with induction of the liver-specific microRNA-122 and modulation of cellular factors that affect HCV replication. RNA interference directed toward essential cellular cofactors in stem cells resulted in HCV-resistant hepatocyte-like cells after differentiation. The ability to infect cultured cells directly with HCV patient serum, to study defined stages of viral permissiveness, and to produce genetically modified cells with desired phenotypes all have broad significance for host-pathogen interactions and cell therapy. Physiologically relevant cell-culture models for infection with hepatitis C virus (HCV) are scarce, and infection by viruses derived from patient serum has been inefficient. Differentiated human hepatocyte-like cells derived from pluripotent stem cells demonstrate hepatic functions but have not been explored for HCV infection studies. Here we report a novel infection model based on these hepatocyte-like cells. Stem cells and definitive endoderm successfully resisted HCV infection, whereas hepatic progenitor cells derived from the stem cells were productively infected by both human- and cell-culture-derived HCV. We determined the point of transition from resistance to susceptibility and, by comparative gene profiling, identified the host factors that were correlated with susceptibility. Genetic modification of human embryonic stem cells, coupled with hepatic differentiation, generated hepatocyte-like cells that were resistant to HCV infection. Our study establishes a new noncancerous and renewable cell-culture system for HCV infection, permits direct infection of cells by patient sera in vitro, identifies a defined transition to HCV susceptibility during hepatocyte differentiation, and demonstrates the feasibility of generating virus-resistant human hepatocyte-like cells in vitro.
Collapse
Affiliation(s)
- Xianfang Wu
- Department of Biological Science, Florida State University, Tallahassee, Florida, United States of America
| | - Jason M. Robotham
- Department of Biological Science, Florida State University, Tallahassee, Florida, United States of America
| | - Emily Lee
- Department of Biological Science, Florida State University, Tallahassee, Florida, United States of America
| | - Stephen Dalton
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, United States of America
| | - Norman M. Kneteman
- Division of Transplantation, Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - David M. Gilbert
- Department of Biological Science, Florida State University, Tallahassee, Florida, United States of America
| | - Hengli Tang
- Department of Biological Science, Florida State University, Tallahassee, Florida, United States of America
- * E-mail:
| |
Collapse
|
23
|
Espeseth AS, Fishel R, Hazuda D, Huang Q, Xu M, Yoder K, Zhou H. siRNA screening of a targeted library of DNA repair factors in HIV infection reveals a role for base excision repair in HIV integration. PLoS One 2011; 6:e17612. [PMID: 21448273 PMCID: PMC3063162 DOI: 10.1371/journal.pone.0017612] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2010] [Accepted: 01/31/2011] [Indexed: 12/19/2022] Open
Abstract
Host DNA repair enzymes have long been assumed to play a role in HIV replication, and many different DNA repair factors have been associated with HIV. In order to identify DNA repair pathways required for HIV infection, we conducted a targeted siRNA screen using 232 siRNA pools for genes associated with DNA repair. Mapping the genes targeted by effective siRNA pools to well-defined DNA repair pathways revealed that many of the siRNAs targeting enzymes associated with the short patch base excision repair (BER) pathway reduced HIV infection. For six siRNA pools targeting BER enzymes, the negative effect of mRNA knockdown was rescued by expression of the corresponding cDNA, validating the importance of the gene in HIV replication. Additionally, mouse embryo fibroblasts (MEFs) lacking expression of specific BER enzymes had decreased transduction by HIV-based retroviral vectors. Examining the role BER enzymes play in HIV infection suggests a role for the BER pathway in HIV integration.
Collapse
Affiliation(s)
- Amy S. Espeseth
- Department of Antiviral Research, Merck
Research Laboratories, West Point, Pennsylvania, United States of
America
| | - Rick Fishel
- Department of Molecular Virology, Immunology,
and Medical Genetics, The Ohio State University Medical Center, Columbus, Ohio,
United States of America
| | - Daria Hazuda
- Department of Antiviral Research, Merck
Research Laboratories, West Point, Pennsylvania, United States of
America
| | - Qian Huang
- Department of Antiviral Research, Merck
Research Laboratories, West Point, Pennsylvania, United States of
America
| | - Min Xu
- Department of Antiviral Research, Merck
Research Laboratories, West Point, Pennsylvania, United States of
America
| | - Kristine Yoder
- Department of Molecular Virology, Immunology,
and Medical Genetics, The Ohio State University Medical Center, Columbus, Ohio,
United States of America
| | - Honglin Zhou
- Department of Antiviral Research, Merck
Research Laboratories, West Point, Pennsylvania, United States of
America
| |
Collapse
|
24
|
Kongruttanachok N, Phuangphairoj C, Thongnak A, Ponyeam W, Rattanatanyong P, Pornthanakasem W, Mutirangura A. Replication independent DNA double-strand break retention may prevent genomic instability. Mol Cancer 2010; 9:70. [PMID: 20356374 PMCID: PMC2867818 DOI: 10.1186/1476-4598-9-70] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2009] [Accepted: 03/31/2010] [Indexed: 01/16/2023] Open
Abstract
Background Global hypomethylation and genomic instability are cardinal features of cancers. Recently, we established a method for the detection of DNA methylation levels at sites close to endogenous DNA double strand breaks (EDSBs), and found that those sites have a higher level of methylation than the rest of the genome. Interestingly, the most significant differences between EDSBs and genomes were observed when cells were cultured in the absence of serum. DNA methylation levels on each genomic location are different. Therefore, there are more replication-independent EDSBs (RIND-EDSBs) located in methylated genomic regions. Moreover, methylated and unmethylated RIND-EDSBs are differentially processed. Euchromatins respond rapidly to DSBs induced by irradiation with the phosphorylation of H2AX, γ-H2AX, and these initiate the DSB repair process. During G0, most DSBs are repaired by non-homologous end-joining repair (NHEJ), mediated by at least two distinct pathways; the Ku-mediated and the ataxia telangiectasia-mutated (ATM)-mediated. The ATM-mediated pathway is more precise. Here we explored how cells process methylated RIND-EDSBs and if RIND-EDSBs play a role in global hypomethylation-induced genomic instability. Results We observed a significant number of methylated RIND-EDSBs that are retained within deacetylated chromatin and free from an immediate cellular response to DSBs, the γ-H2AX. When cells were treated with tricostatin A (TSA) and the histones became hyperacetylated, the amount of γ-H2AX-bound DNA increased and the retained RIND-EDSBs were rapidly repaired. When NHEJ was simultaneously inhibited in TSA-treated cells, more EDSBs were detected. Without TSA, a sporadic increase in unmethylated RIND-EDSBs could be observed when Ku-mediated NHEJ was inhibited. Finally, a remarkable increase in RIND-EDSB methylation levels was observed when cells were depleted of ATM, but not of Ku86 and RAD51. Conclusions Methylated RIND-EDSBs are retained in non-acetylated heterochromatin because there is a prolonged time lag between RIND-EDSB production and repair. The rapid cellular responses to DSBs may be blocked by compact heterochromatin structure which then allows these breaks to be repaired by a more precise ATM-dependent pathway. In contrast, Ku-mediated NHEJ can repair euchromatin-associated EDSBs. Consequently, spontaneous mutations in hypomethylated genome are produced at faster rates because unmethylated EDSBs are unable to avoid the more error-prone NHEJ mechanisms.
Collapse
Affiliation(s)
- Narisorn Kongruttanachok
- Center of Excellence in Molecular Genetics of Cancer and Human Diseases, Department of Anatomy, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | | | | | | | | | | | | |
Collapse
|
25
|
Kenworthy R, Lambert D, Yang F, Wang N, Chen Z, Zhu H, Zhu F, Liu C, Li K, Tang H. Short-hairpin RNAs delivered by lentiviral vector transduction trigger RIG-I-mediated IFN activation. Nucleic Acids Res 2009; 37:6587-99. [PMID: 19729514 PMCID: PMC2770676 DOI: 10.1093/nar/gkp714] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2009] [Revised: 08/13/2009] [Accepted: 08/14/2009] [Indexed: 12/25/2022] Open
Abstract
Activation of the type I interferon (IFN) pathway by small interfering RNA (siRNA) is a major contributor to the off-target effects of RNA interference in mammalian cells. While IFN induction complicates gene function studies, immunostimulation by siRNAs may be beneficial in certain therapeutic settings. Various forms of siRNA, meeting different compositional and structural requirements, have been reported to trigger IFN activation. The consensus is that intracellularly expressed short-hairpin RNAs (shRNAs) are less prone to IFN activation because they are not detected by the cell-surface receptors. In particular, lentiviral vector-mediated transduction of shRNAs has been reported to avoid IFN response. Here we identify a shRNA that potently activates the IFN pathway in human cells in a sequence- and 5'-triphosphate-dependent manner. In addition to suppressing its intended mRNA target, expression of the shRNA results in dimerization of interferon regulatory factor-3, activation of IFN promoters and secretion of biologically active IFNs into the extracellular medium. Delivery by lentiviral vector transduction did not avoid IFN activation by this and another, unrelated shRNA. We also demonstrated that retinoic-acid-inducible gene I, and not melanoma differentiation associated gene 5 or toll-like receptor 3, is the cytoplasmic sensor for intracellularly expressed shRNAs that trigger IFN activation.
Collapse
Affiliation(s)
- Rachael Kenworthy
- Department of Biological Science, Florida State University, Tallahassee, FL 32306-4295, Department of Molecular Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555 and Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Diana Lambert
- Department of Biological Science, Florida State University, Tallahassee, FL 32306-4295, Department of Molecular Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555 and Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Feng Yang
- Department of Biological Science, Florida State University, Tallahassee, FL 32306-4295, Department of Molecular Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555 and Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Nan Wang
- Department of Biological Science, Florida State University, Tallahassee, FL 32306-4295, Department of Molecular Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555 and Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Zihong Chen
- Department of Biological Science, Florida State University, Tallahassee, FL 32306-4295, Department of Molecular Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555 and Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Haizhen Zhu
- Department of Biological Science, Florida State University, Tallahassee, FL 32306-4295, Department of Molecular Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555 and Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Fanxiu Zhu
- Department of Biological Science, Florida State University, Tallahassee, FL 32306-4295, Department of Molecular Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555 and Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Chen Liu
- Department of Biological Science, Florida State University, Tallahassee, FL 32306-4295, Department of Molecular Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555 and Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Kui Li
- Department of Biological Science, Florida State University, Tallahassee, FL 32306-4295, Department of Molecular Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555 and Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Hengli Tang
- Department of Biological Science, Florida State University, Tallahassee, FL 32306-4295, Department of Molecular Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555 and Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|
26
|
Qing M, Yang F, Zhang B, Zou G, Robida JM, Yuan Z, Tang H, Shi PY. Cyclosporine inhibits flavivirus replication through blocking the interaction between host cyclophilins and viral NS5 protein. Antimicrob Agents Chemother 2009; 53:3226-35. [PMID: 19451286 PMCID: PMC2715601 DOI: 10.1128/aac.00189-09] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2009] [Revised: 03/27/2009] [Accepted: 05/12/2009] [Indexed: 02/07/2023] Open
Abstract
Although flaviviruses cause significant human diseases, no effective therapy is currently available. Host factors essential for viral replication are potential targets for antiviral development. Here we report that cyclophilins (CyPs), a family of cellular peptidyl-prolyl isomerases (PPIases), play a role in flavivirus replication. Huh-7.5 cells with knockdown of different isoforms of CyP were less efficient than parental cells in supporting flavivirus replication, including West Nile virus (WNV), dengue virus, and yellow fever virus. The low viral replication in CyP A (CyPA) knockdown cells could be rescued by trans supplying of a wild-type CyPA but not by trans supplying of a mutant CyPA (defective in the PPIase activity), indicating that the isomerase activity of CyPA is critical for viral replication. Immunoprecipitation and biochemical pulldown analyses showed that CyPA interacts with WNV genomic RNA and viral NS5 protein in the replication complex. Furthermore, antiviral experiments demonstrated that cyclosporine (Cs; an 11-amino-acid cyclic peptide known to block the PPIase activity of CyPA) inhibits flavivirus replication in cell culture at nontoxic concentrations. Time-of-addition and transient replicon results indicated that Cs inhibits flavivirus at the step of viral RNA synthesis. Biochemical analysis showed that Cs directly blocks the interaction between CyPA and WNV NS5 protein. Our results suggest that host CyPA is a component of flavivirus replication complex and could be targeted for potential antiviral development.
Collapse
Affiliation(s)
- Min Qing
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Unwalla HJ, Li H, Li SY, Abad D, Rossi JJ. Use of a U16 snoRNA-containing ribozyme library to identify ribozyme targets in HIV-1. Mol Ther 2008; 16:1113-9. [PMID: 18388915 PMCID: PMC2775071 DOI: 10.1038/mt.2008.54] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Hammerhead ribozymes have been shown to silence human immunodeficiency virus-1 (HIV-1) gene expression by site-specific cleavage of viral mRNA. The two major factors that determine whether ribozymes will be effective for post-transcriptional gene silencing are colocalization of the ribozyme and the target RNAs, and the choice of an appropriate target site on the mRNA. An effective screening strategy for potential targets on the viral genome is the use of ribozyme libraries in cell culture. Capitalizing on previous findings that HIV-1 and ribozymes can be colocalized in the nucleolus, we created a novel hammerhead ribozyme library by inserting hammerhead ribozymes with fully randomized stems 1 and 2 into the body of the U16 small nucleolar RNA (snoRNA). Following three rounds of cotransfection with an HIV-1 proviral DNA harboring the herpes simplex virus thymidine kinase (HSV-TK) gene, we selected for gancyclovir-resistant cells and identified a ribozyme sequence that could potentially target both the U5 and gag genes of HIV-1 regions on the HIV-1 genome through partial homologies with these targets. When the ribozymes were converted to full complementarity with the targets, they provided potent inhibition of HIV-1 replication in cell culture. These results provide a novel approach for identifying ribozyme targets in HIV-1.
Collapse
Affiliation(s)
- Hoshang J Unwalla
- Division of Molecular Biology, Beckman Research Institute of The City of Hope, Duarte, California 91010, USA
| | | | | | | | | |
Collapse
|
28
|
Cyclophilin A is an essential cofactor for hepatitis C virus infection and the principal mediator of cyclosporine resistance in vitro. J Virol 2008; 82:5269-78. [PMID: 18385230 DOI: 10.1128/jvi.02614-07] [Citation(s) in RCA: 185] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Cyclosporine (CsA) and its derivatives potently suppress hepatitis C virus (HCV) replication. Recently, CsA-resistant HCV replicons have been identified in vitro. We examined the dependence of the wild-type and CsA-resistant replicons on various cyclophilins for replication. A strong correlation between CsA resistance and reduced dependency on cyclophilin A (CyPA) for replication was identified. Silencing of CyPB or CyPC expression had no significant effect on replication, whereas various forms of small interfering RNA (siRNA) directed at CyPA inhibited HCV replication of wild-type but not CsA-resistant replicons. The efficiency of a particular siRNA in suppressing CyPA expression was correlated with its potency in inhibiting HCV replication, and expression of an siRNA-resistant CyPA cDNA rescued replication. In addition, an anti-CyPA antibody blocked replication of the wild-type but not the resistant replicon in an in vitro replication assay. Depletion of CyPA alone in the CsA-resistant replicon cells eliminated CsA resistance, indicating that CyPA is the chief mediator of the observed CsA resistance. The dependency on CyPA for replication was observed for both genotype (GT) 1a and 1b replicons as well as a GT 2a infectious virus. An interaction between CyPA and HCV RNA as well as the viral polymerase that is sensitive to CsA treatment in wild-type but not in resistant replicons was detected. These findings reveal the molecular mechanism of CsA resistance and identify CyPA as a critical cellular cofactor for HCV replication and infection.
Collapse
|
29
|
Smith JA, Wang FX, Zhang H, Wu KJ, Williams KJ, Daniel R. Evidence that the Nijmegen breakage syndrome protein, an early sensor of double-strand DNA breaks (DSB), is involved in HIV-1 post-integration repair by recruiting the ataxia telangiectasia-mutated kinase in a process similar to, but distinct from, cellular DSB repair. Virol J 2008; 5:11. [PMID: 18211700 PMCID: PMC2262065 DOI: 10.1186/1743-422x-5-11] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2007] [Accepted: 01/22/2008] [Indexed: 01/30/2023] Open
Abstract
Retroviral transduction involves integrase-dependent linkage of viral and host DNA that leaves an intermediate that requires post-integration repair (PIR). We and others proposed that PIR hijacks the host cell double-strand DNA break (DSB) repair pathways. Nevertheless, the geometry of retroviral DNA integration differs considerably from that of DSB repair and so the precise role of host-cell mechanisms in PIR remains unclear. In the current study, we found that the Nijmegen breakage syndrome 1 protein (NBS1), an early sensor of DSBs, associates with HIV-1 DNA, recruits the ataxia telangiectasia-mutated (ATM) kinase, promotes stable retroviral transduction, mediates efficient integration of viral DNA and blocks integrase-dependent apoptosis that can arise from unrepaired viral-host DNA linkages. Moreover, we demonstrate that the ATM kinase, recruited by NBS1, is itself required for efficient retroviral transduction. Surprisingly, recruitment of the ATR kinase, which in the context of DSB requires both NBS1 and ATM, proceeds independently of these two proteins. A model is proposed emphasizing similarities and differences between PIR and DSB repair. Differences between the pathways may eventually allow strategies to block PIR while still allowing DSB repair.
Collapse
Affiliation(s)
- Johanna A Smith
- Division of Infectious Diseases - Center for Human Virology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA.
| | | | | | | | | | | |
Collapse
|
30
|
Liu Z, Kenworthy R, Green C, Tang H. Molecular determinants of nucleolar translocation of RNA helicase A. Exp Cell Res 2007; 313:3743-54. [PMID: 17822697 DOI: 10.1016/j.yexcr.2007.07.037] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2007] [Revised: 07/11/2007] [Accepted: 07/27/2007] [Indexed: 11/30/2022]
Abstract
RNA helicase A (RHA) is a member of the DEAH-box family of DNA/RNA helicases involved in multiple cellular processes and the life cycles of many viruses. The subcellular localization of RHA is dynamic despite its steady-state concentration in the nucleoplasm. We have previously shown that it shuttles rapidly between the nucleus and the cytoplasm by virtue of a bidirectional nuclear transport domain (NTD) located in its carboxyl terminus. Here, we investigate the molecular determinants for its translocation within the nucleus and, more specifically, its redistribution from the nucleoplasm to nucleolus or the perinucleolar region. We found that low temperature treatment, transcription inhibition or replication of hepatitis C virus caused the intranuclear redistribution of the protein, suggesting that RHA shuttles between the nucleolus and nucleoplasm and becomes trapped in the nucleolus or the perinucleolar region upon blockade of transport to the nucleoplasm. Both the NTD and ATPase activity were essential for RHA's transport to the nucleolus or perinucleolar region. One of the double-stranded RNA binding domains (dsRBD II) was also required for this nucleolar translocation (NoT) phenotype. RNA interference studies revealed that RHA is essential for survival of cultured hepatoma cells and the ATPase activity appears to be important for this critical role.
Collapse
Affiliation(s)
- Zhe Liu
- Department of Biological Science, Florida State University, Tallahassee, FL 32306-4370, USA
| | | | | | | |
Collapse
|
31
|
Robida JM, Nelson HB, Liu Z, Tang H. Characterization of hepatitis C virus subgenomic replicon resistance to cyclosporine in vitro. J Virol 2007; 81:5829-40. [PMID: 17376913 PMCID: PMC1900250 DOI: 10.1128/jvi.02524-06] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2006] [Accepted: 03/11/2007] [Indexed: 12/17/2022] Open
Abstract
Treatment of hepatitis C virus (HCV) infection has been met with less than satisfactory results due primarily to its resistance to and significant side effects from alpha interferon (IFN-alpha). New classes of safe and broadly acting treatments are urgently needed. Cyclosporine (CsA), an immunosuppressive and anti-inflammatory drug for organ transplant patients, has recently been shown to be highly effective in suppressing HCV replication through a mechanism that is distinct from the IFN pathway. Here we report the selection and characterization of HCV replicon cells that are resistant to CsA treatment in vitro, taking advantage of our ability to sort live cells that are actively replicating HCV RNA in the presence of drug treatments. This resistance is specific to CsA as the replicon cells most resistant to CsA were still sensitive to IFN-alpha and a polymerase inhibitor. We demonstrate that the resistant phenotype is not a result of general enhanced replication and, furthermore, that mutations in the coding region of HCV NS5B contribute to the resistance. Interestingly, a point mutation (I432V) isolated from the most resistant replicon was able to rescue a lethal mutation (P540A) in NS5B that disrupts its interaction with its cofactor, cyclophilin B (CypB), even though the I432V mutation is located outside of the reported CypB binding site (amino acids 520 to 591). Our results demonstrate that CsA exerts selective pressure on the HCV genome, leading to the emergence of resistance-conferring mutations in the viral genome despite acting upon a cellular protein.
Collapse
Affiliation(s)
- John M Robida
- Department of Biological Science, Florida State University, Tallahassee, FL 32306-4370, USA
| | | | | | | |
Collapse
|
32
|
Shi L, Qiu D, Zhao G, Corthesy B, Lees-Miller S, Reeves WH, Kao PN. Dynamic binding of Ku80, Ku70 and NF90 to the IL-2 promoter in vivo in activated T-cells. Nucleic Acids Res 2007; 35:2302-10. [PMID: 17389650 PMCID: PMC1874627 DOI: 10.1093/nar/gkm117] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2006] [Revised: 02/07/2007] [Accepted: 02/08/2007] [Indexed: 01/07/2023] Open
Abstract
IL-2 gene expression in activated T-cells is initiated by chromatin remodeling at the IL-2 proximal promoter and conversion of a transcriptional repressor into a potent transcriptional activator. A purine-box regulator complex was purified from activated Jurkat T-cell nuclei based on sequence-specific DNA binding to the antigen receptor response element (ARRE)/nuclear factor of activated T-cells (NF-AT) target DNA sequence in the proximal IL-2 promoter. ARRE DNA-binding subunits were identified as NF90, NF45 and systemic lupus erythematosis autoantigens, Ku80 and Ku70. Monoclonal antibodies to Ku80, Ku70 and NF90 specifically inhibit constitutive and inducible ARRE DNA-binding activity in Jurkat T-cells. Ku80, Ku70 and NF90 bind specifically to the IL-2 gene promoter in vivo, as demonstrated by chromatin immunoprecipitation. Activation of Jurkat T-cells and mouse primary spleen cells induces binding of Ku80 and NF90 to the IL-2 promoter in vivo, and decreases binding of Ku70 to the IL-2 promoter in vivo, and these dynamic changes are inhibited by immunosuppressants cyclosporin A and triptolide. Dynamic changes in binding of Ku80, Ku70 and NF90 to the IL-2 proximal promoter in vivo correlate with chromatin remodeling and transcriptional initiation in activated T-cells.
Collapse
Affiliation(s)
- Lingfang Shi
- Pulmonary and Critical Care Medicine, Stanford University Medical Center, Stanford, CA 94305-5236, USA, Immunology and Allergy, Internal Medicine Department, University of Lausanne, CH-1011, Lausanne, Switzerland, Department of Biochemistry & Molecular Biology, University of Calgary, 3330 Hospital Drive NW, Calgary, Alta., Canada T2N 4N1 and Division of Rheumatology and Clinical Immunology, University of Florida, Gainesville, FL 32610-0211, USA
| | - Daoming Qiu
- Pulmonary and Critical Care Medicine, Stanford University Medical Center, Stanford, CA 94305-5236, USA, Immunology and Allergy, Internal Medicine Department, University of Lausanne, CH-1011, Lausanne, Switzerland, Department of Biochemistry & Molecular Biology, University of Calgary, 3330 Hospital Drive NW, Calgary, Alta., Canada T2N 4N1 and Division of Rheumatology and Clinical Immunology, University of Florida, Gainesville, FL 32610-0211, USA
| | - Guohua Zhao
- Pulmonary and Critical Care Medicine, Stanford University Medical Center, Stanford, CA 94305-5236, USA, Immunology and Allergy, Internal Medicine Department, University of Lausanne, CH-1011, Lausanne, Switzerland, Department of Biochemistry & Molecular Biology, University of Calgary, 3330 Hospital Drive NW, Calgary, Alta., Canada T2N 4N1 and Division of Rheumatology and Clinical Immunology, University of Florida, Gainesville, FL 32610-0211, USA
| | - Blaise Corthesy
- Pulmonary and Critical Care Medicine, Stanford University Medical Center, Stanford, CA 94305-5236, USA, Immunology and Allergy, Internal Medicine Department, University of Lausanne, CH-1011, Lausanne, Switzerland, Department of Biochemistry & Molecular Biology, University of Calgary, 3330 Hospital Drive NW, Calgary, Alta., Canada T2N 4N1 and Division of Rheumatology and Clinical Immunology, University of Florida, Gainesville, FL 32610-0211, USA
| | - Susan Lees-Miller
- Pulmonary and Critical Care Medicine, Stanford University Medical Center, Stanford, CA 94305-5236, USA, Immunology and Allergy, Internal Medicine Department, University of Lausanne, CH-1011, Lausanne, Switzerland, Department of Biochemistry & Molecular Biology, University of Calgary, 3330 Hospital Drive NW, Calgary, Alta., Canada T2N 4N1 and Division of Rheumatology and Clinical Immunology, University of Florida, Gainesville, FL 32610-0211, USA
| | - Westley H. Reeves
- Pulmonary and Critical Care Medicine, Stanford University Medical Center, Stanford, CA 94305-5236, USA, Immunology and Allergy, Internal Medicine Department, University of Lausanne, CH-1011, Lausanne, Switzerland, Department of Biochemistry & Molecular Biology, University of Calgary, 3330 Hospital Drive NW, Calgary, Alta., Canada T2N 4N1 and Division of Rheumatology and Clinical Immunology, University of Florida, Gainesville, FL 32610-0211, USA
| | - Peter N. Kao
- Pulmonary and Critical Care Medicine, Stanford University Medical Center, Stanford, CA 94305-5236, USA, Immunology and Allergy, Internal Medicine Department, University of Lausanne, CH-1011, Lausanne, Switzerland, Department of Biochemistry & Molecular Biology, University of Calgary, 3330 Hospital Drive NW, Calgary, Alta., Canada T2N 4N1 and Division of Rheumatology and Clinical Immunology, University of Florida, Gainesville, FL 32610-0211, USA
| |
Collapse
|
33
|
Ultrasensitive monitoring of ribozyme cleavage product using molecular-beacon-ligation system. ACTA ACUST UNITED AC 2007. [DOI: 10.1007/s11434-007-0074-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
34
|
Nguyen DG, Wolff KC, Yin H, Caldwell JS, Kuhen KL. "UnPAKing" human immunodeficiency virus (HIV) replication: using small interfering RNA screening to identify novel cofactors and elucidate the role of group I PAKs in HIV infection. J Virol 2007; 80:130-7. [PMID: 16352537 PMCID: PMC1317519 DOI: 10.1128/jvi.80.1.130-137.2006] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
In order to identify novel proviral host factors involved in human immunodeficiency virus (HIV) infection, we performed a screen of a small interfering RNA (siRNA) library targeting 5,000 genes with the highest potential for being targets for therapeutics. Many siRNAs in the library against known host factors, such as TSG101, furin, and CXCR4, were identified as inhibitors by the screen and thus served as internal validation. In addition, many novel factors whose knockdown inhibited infection were identified, including Pak3, a member of the serine/threonine group I PAK kinases. The HIV accessory factor Nef has been shown to associate with a PAK kinase, leading to enhanced viral production; however, the exact identity of the kinase has remained controversial. Prompted by the Pak3 screen hit, we further investigated the involvement of group I PAK kinases in HIV using siRNA. Contrary to the current literature, Pak1 depletion strongly inhibited HIV infection in multiple cell systems and decreased levels of integrated provirus, while Pak2 depletion showed no effect. Overexpression of a constitutively active Pak1 mutant also enhanced HIV infection, further supporting its role as the dominant PAK involved.
Collapse
Affiliation(s)
- Deborah G Nguyen
- Genomics Institute of the Novartis Research Foundation, San Diego, CA 92121, USA.
| | | | | | | | | |
Collapse
|
35
|
Ke N, Zhou D, Chatterton JE, Liu G, Chionis J, Zhang J, Tsugawa L, Lynn R, Yu D, Meyhack B, Wong-Staal F, Li QX. A new inducible RNAi xenograft model for assessing the staged tumor response to mTOR silencing. Exp Cell Res 2006; 312:2726-34. [PMID: 16765945 DOI: 10.1016/j.yexcr.2006.05.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2006] [Revised: 04/28/2006] [Accepted: 05/01/2006] [Indexed: 12/20/2022]
Abstract
Human xenograft tumor models are widely used for efficacy evaluation of potential cancer targets. siRNA is usually stably introduced into tumor cells prior to transplantation. However, silencing of the cancer therapeutic target usually results in reduced cell growth/survival in vitro and/or failure to establish tumors in vivo, thus hindering tumor response-based efficacy evaluation. The present study explored a new tumor response model based on regulated RNAi, which is more relevant from a clinical standpoint. As a proof of principle, an inducible lentiviral RNAi vector was used to silence the known cancer therapeutic target mTOR upon induction with Doxycycline (DOX). The responses to DOX-induced mTOR silencing were tested both in vitro and in vivo for prostate cancer PC3 models. Significant reduction in cancer cell survival was observed due to cell cycle arrest and apoptosis when mTOR silencing was induced in vitro. mTOR silencing also caused tumor regression for the early-staged PC3 tumors (100% tumor regressed and 45% became tumor-free). The advanced-staged tumors also demonstrated significant responses (100% regressed). Therefore, our results demonstrate the powerful utility of this new inducible xenograft tumor model for efficacy evaluation of cancer targets, and it provides a direct in vivo efficacy validation of mTOR as a cancer therapeutic target.
Collapse
Affiliation(s)
- Ning Ke
- Immusol, Inc., 10790 Roselle Street, San Diego, CA 92121, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Smith JA, Daniel R. Following the path of the virus: the exploitation of host DNA repair mechanisms by retroviruses. ACS Chem Biol 2006; 1:217-26. [PMID: 17163676 DOI: 10.1021/cb600131q] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Numerous host cellular cofactors are involved in the life cycle of retroviruses. Importantly, DNA repair machinery of infected cells is activated by retroviruses and retroviral vectors during the process of integration and host cell DNA repair proteins are employed to create a fully integrated provirus. The full delineation of these repair mechanisms that are triggered by retroviruses also has implications outside of the field of retrovirology. It will undoubtedly be of interest to developers of gene therapy and will also further facilitate our understanding of DNA repair and cancer. This review gives a brief summary of the accomplishments in the field of DNA repair and retroviral integration and the opportunities that this area of science provides with regards to the elucidation of repair mechanisms, in the context of retroviral infection.
Collapse
Affiliation(s)
- Johanna A Smith
- Division of Infectious Diseases--Center for Human Virology, Thomas Jefferson University, 1020 Locust Street, Philadelphia, Pennsylvania 19107, USA
| | | |
Collapse
|
37
|
Eberhardy SR, Goncalves J, Coelho S, Segal DJ, Berkhout B, Barbas CF. Inhibition of human immunodeficiency virus type 1 replication with artificial transcription factors targeting the highly conserved primer-binding site. J Virol 2006; 80:2873-83. [PMID: 16501096 PMCID: PMC1395442 DOI: 10.1128/jvi.80.6.2873-2883.2006] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The human immunodeficiency virus type 1 (HIV-1) primer-binding site (PBS) is a highly conserved region in the HIV genome and represents an attractive target for the development of new anti-HIV therapies. In this study, we designed four artificial zinc finger transcription factors to bind at or adjacent to the PBS and repress transcription from the HIV-1 long terminal repeat (LTR). These proteins bound to the LTR in vivo, as demonstrated by the chromatin immunoprecipitation assay. In transient reporter assays, three of the four proteins repressed transcription of a reporter driven by the HIV-1 LTR. Only one of these proteins, however, designated KRAB-PBS2, was able to prevent virus production when transduced into primary lymphocytes. We observed >90% inhibition of viral replication over the course of several weeks compared to untransduced cells, and no significant cytotoxicity was observed. Long-term exposure of HIV-1 to KRAB-PBS2 induced mutations in the HIV-1 PBS that reduced the effectiveness of the repressor, but these mutations also resulted in decreased rates of viral replication. These results show that KRAB-PBS2 has the potential to be used in antiviral therapy for AIDS patients and might complement other gene-based strategies.
Collapse
Affiliation(s)
- Scott R Eberhardy
- The Skaggs Institute for Chemical Biology, La Jolla, California 92037, USA
| | | | | | | | | | | |
Collapse
|
38
|
Abstract
Many viruses, with distinct replication strategies, activate DNA-damage response pathways, including the lentivirus human immunodeficiency virus (HIV) and the DNA viruses Epstein-Barr virus (EBV), herpes simplex virus 1, adenovirus and SV40. DNA-damage response pathways involving DNA-dependent protein kinase, ataxia-telengiectasia mutated (ATM) and 'ataxia-telengiectasia and Rad3-related' (ATR) have all been implicated. This review focuses on the effects of HIV and EBV replication on DNA repair pathways. It has been suggested that activation of cellular DNA repair and recombination enzymes is beneficial for viral replication, as illustrated by the ability of suppressors of the ATM and ATR family to inhibit HIV replication. However, activation of DNA-damage response pathways can also promote apoptosis. Viruses can tailor the cellular response by suppressing downstream signalling from DNA-damage sensors, as exemplified by EBV. New small-molecule inhibitors of the DNA-damage response pathways could therefore be of value to treat viral infections.
Collapse
Affiliation(s)
- Alison Sinclair
- School of Life Sciences, University of Sussex, Brighton, BN1 9QG, UK
| | - Sarah Yarranton
- School of Life Sciences, University of Sussex, Brighton, BN1 9QG, UK. Tel: +44 (0)1273 678 194; Fax: +44 1273 678 433;
| | - Celine Schelcher
- School of Life Sciences, University of Sussex, Brighton, BN1 9QG, UK. Tel: +44 (0)1273 678 194; Fax +44 1273 678 433;
| |
Collapse
|
39
|
Nelson HB, Tang H. Effect of cell growth on hepatitis C virus (HCV) replication and a mechanism of cell confluence-based inhibition of HCV RNA and protein expression. J Virol 2006; 80:1181-90. [PMID: 16414995 PMCID: PMC1346944 DOI: 10.1128/jvi.80.3.1181-1190.2006] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
An intimate relationship between hepatitis C virus (HCV) replication and the physiological state of the host liver cells has been reported. In particular, a highly reproducible and reversible inhibitory effect of high cell density on HCV replication was observed: high levels of HCV RNA and protein can be detected in actively growing cells but decline sharply when the replicon cells reach confluence. Arrested cell growth of confluent cells has been proposed to be responsible for the inhibitory effect. Indeed, other means of arresting cell growth have also been shown to inhibit HCV replication. Here, we report a detailed study of the effect of cell growth and confluence on HCV replication using a flow cytometry-based assay that is not biased against cytostasis and reduced cell number. Although we readily reproduced the inhibitory effect of cell confluence on HCV replication, we found no evidence of inhibition by serum starvation, which arrested cell growth as expected. In addition, we observed no inhibitory effect by agents that perturb the cell cycle. Instead, our results suggest that the reduced intracellular pools of nucleosides account for the suppression of HCV expression in confluent cells, possibly through the shutoff of the de novo nucleoside biosynthetic pathway when cells become confluent. Adding exogenous uridine and cytidine to the culture medium restored HCV replication and expression in confluent cells. These results suggest that cell growth arrest is not sufficient for HCV replicon inhibition and reveal a mechanism for HCV RNA inhibition by cell confluence.
Collapse
Affiliation(s)
- Heather B Nelson
- Bio Unit I, Chieftan Way, Department of Biological Science, Florida State University, Tallahassee, FL 32306-4370, USA
| | | |
Collapse
|
40
|
Fanning GC, Symonds G. Gene-expressed RNA as a therapeutic: issues to consider, using ribozymes and small hairpin RNA as specific examples. Handb Exp Pharmacol 2006:289-303. [PMID: 16594621 DOI: 10.1007/3-540-27262-3_14] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
In recent years there has been a greater appreciation of both the role of RNA in intracellular gene regulation and the potential to use RNA in therapeutic modalities. In the latter case, RNA can be used as a therapeutic target or a drug. The chapters in this volume cover the varied and potent actions of RNA as antisense, ribozymes, aptamers, microRNA and small hairpin RNA in gene regulation, as well as their use as potential therapeutics for metabolic and infectious diseases. Our group has been involved in the development of anti-HIV gene expression constructs to treat HIV. In this chapter, we address the relevant scientific and some of the commercial issues in the use of RNA as a therapeutic. Specifically, the chapter discusses delivery, expression, potency, toxicity and commercial development using, as examples, hammerhead ribozymes and small hairpin RNA.
Collapse
Affiliation(s)
- G C Fanning
- Johnson Johnson Research, The Australian Technology Park, Strawberry Hills, Locked Bag 4555, 2012 Sydney NSW, Australia
| | | |
Collapse
|
41
|
Matsumoto S, Akashi H, Taira K. Screening and determination of gene function using randomized ribozyme and siRNA libraries. Handb Exp Pharmacol 2006:197-221. [PMID: 16594617 DOI: 10.1007/3-540-27262-3_10] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Rapid progress in the sequencing of the genomes of model organisms, such as the mouse, rat, nematode, fly, and Arabidopsis, as well as the human genome, has provided abundant sequence information, but functions of long stretches of these genomes remain to be determined. RNA-based technologies hold promise as tools that allow us to identify the specific functions of portions of these genomes. In particular, catalytic RNAs, known also as ribozymes, can be engineered for optimization of their activities in the intracellular environment. The introduction of a library of active ribozymes into cells, with subsequent screening for phenotypic changes, can be used for the rapid identification ofa gene function. Ribozyme technology complements another RNA-based tool for the determination of gene function, which is based on libraries of small interfering RNAs (siRNAs).
Collapse
Affiliation(s)
- S Matsumoto
- Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, 7-3-1 Hongo, 113-8656 Tokyo, Japan
| | | | | |
Collapse
|
42
|
Abstract
Catalytic RNAs, also known as ribozymes, can be engineered to optimize their activities in the intracellular environment. The introduction of a library of active ribozymes into cells, and the subsequent screening for phenotypic changes, allows the rapid identification of gene function. For the determination of gene function, ribozyme technology complements another RNA-based tool that is based on libraries of small interfering RNAs.
Collapse
Affiliation(s)
- Hideo Akashi
- Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Tokyo 113-8656, Japan
| | | | | |
Collapse
|
43
|
|
44
|
Nunnari G, Argyris E, Fang J, Mehlman KE, Pomerantz RJ, Daniel R. Inhibition of HIV-1 replication by caffeine and caffeine-related methylxanthines. Virology 2005; 335:177-84. [PMID: 15840517 DOI: 10.1016/j.virol.2005.02.015] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2004] [Revised: 02/15/2005] [Accepted: 02/23/2005] [Indexed: 01/06/2023]
Abstract
Human immunodeficiency virus type I (HIV-1) DNA integration is an essential step of viral replication. We have suggested recently that this stage of HIV-1 life-cycle triggers a cellular DNA damage response and requires cellular DNA repair proteins for its completion. These include DNA-PK (DNA-dependent protein kinase), ATR (ataxia telangiectasia and Rad3-related), and, at least in some circumstances, ATM (ataxia telangiectasia mutated). Host cell proteins may constitute an attractive target for anti-HIV-1 therapeutics, since development of drug resistance against compounds targeting these cellular cofactor proteins is unlikely. In this study, we show that an inhibitor of ATR and ATM kinases, caffeine, can suppress replication of infectious HIV-1 strains, and provide evidence that caffeine exerts its inhibitory effect at the integration step of the HIV-1 life-cycle. We also demonstrate that caffeine-related methylxanthines including the clinically used compound, theophylline, act at the same step of the HIV-1 life-cycle as caffeine and efficiently inhibit HIV-1 replication in primary human cells. These data reveal the feasibility of therapeutic approaches targeting host cell proteins and further support the hypothesis that ATR and ATM proteins are involved in retroviral DNA integration.
Collapse
Affiliation(s)
- Giuseppe Nunnari
- The Dorrance H. Hamilton Laboratories, Center for Human Virology and Biodefense, Division of Infectious Diseases and Environmental Medicine, Jefferson Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | | | | | | | | | | |
Collapse
|
45
|
Sano M, Kato Y, Taira K. Functional gene-discovery systems based on libraries of hammerhead and hairpin ribozymes and short hairpin RNAs. MOLECULAR BIOSYSTEMS 2005; 1:27-35. [PMID: 16880960 DOI: 10.1039/b503235k] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Abundant information about the nucleotide sequence of the human genome has become readily available and it is now necessary to develop methods for the identification of genes that are involved in important cellular, developmental and disease-related processes. Identification methods based on the activities of hammerhead and hairpin ribozymes and of short hairpin RNAs (shRNAs), whose target specificities are coupled with loss-of-function phenotypes, have received increasing attention as possible tools for the rapid identification of key genes involved in such processes. We describe here recent advances that have been made with libraries of ribozymes and shRNAs and compare the advantages of the different types of library. The use of such libraries has already revealed new details of several important physiological phenomena.
Collapse
Affiliation(s)
- Masayuki Sano
- Gene Function Research Center, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba Science City, Japan
| | | | | |
Collapse
|