HIV-1 Proviral Transcription and Latency in the New Era
Abstract
:1. The Latent Reservoir in the Spotlight
1.1. Timeline, Size, Stability, and Composition of the Latent Reservoir
1.2. Routes of Latency Establishment
2. Contribution of Nuclear Topology Shaping the Integration Landscape, Transcriptional Regulation, and Proviral Fate
2.1. Importance of CD4+ T Cell Nuclear Topology for HIV Integration
2.2. CD4+ T Cell Nucleus Sub-Compartments and Regulatory Elements
2.3. Integration Site Effects on HIV Proviral Transcription and Fate
2.4. Conclusions and Future Directions
3. The HIV Transcriptional Program and Proviral Fate
3.1. Phases of the HIV Transcriptional Program
3.2. Feedback Loop and Theories of Latency Establishment due to Dysfunctions in the HIV Transcriptional Program
3.3. Conclusions and Future Directions
4. HIV Proviral Transcriptional Regulation in Homeostatic Conditions
4.1. Cis-Elements in the HIV Proviral Genome
4.2. Transcription Factors Acting on cis-Elements in the HIV Proviral Genome
4.3. Pol II Pausing at the HIV Proviral Genome and Elongation Factors
4.4. Conclusions and Future Directions
5. HIV Proviral Transcriptional Regulation During CD4+ T Cell Stimulation
5.1. P-TEFb/7SK snRNP Complex
5.2. Super Elongation Complex (SEC)
5.3. BRD4
5.4. KAP1
5.5. Conclusions and Future Directions
6. HIV Proviral Nucleosome Positioning and Epigenomics
6.1. Chromatin-Remodeling Complexes and Associated Factors
6.2. Histone Chaperones and Chromatin Reassembly Factors
6.3. Proviral Chromatin Acetylation/Deacetylation and Associated Factors
6.4. Proviral Chromatin Methylation/Demethylation and Associated Factors
6.5. Proviral Genome DNA Methylation
6.6. Conclusions and Future Directions
7. Disease Relevance and Current Therapeutic Challenges
8. The Ephemeral Nature of Ideas and Considerations for Future Research
Funding
Acknowledgments
Conflicts of Interest
References
- Perelson, A.S.; Essunger, P.; Cao, Y.; Vesanen, M.; Hurley, A.; Saksela, K.; Markowitz, M.; Ho, D.D. Decay characteristics of HIV-1-infected compartments during combination therapy. Nature 1997, 387, 188–191. [Google Scholar] [CrossRef] [PubMed]
- Sengupta, S.; Siliciano, R.F. Targeting the Latent Reservoir for HIV-1. Immunity 2018, 48, 872–895. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Davey, R.T.; Bhat, N.; Yoder, C.; Chun, T.W.; Metcalf, J.A.; Dewar, R.; Natarajan, V.; Lempicki, R.A.; Adelsberger, J.W.; Miller, K.D.; et al. HIV-1 and T cell dynamics after interruption of highly active antiretroviral therapy (HAART) in patients with a history of sustained viral suppression. Proc. Natl. Acad. Sci. USA 1999, 96, 15109–15114. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chun, T.W.; Carruth, L.; Finzi, D.; Shen, X.; DiGiuseppe, J.A.; Taylor, H.; Hermankova, M.; Chadwick, K.; Margolick, J.; Quinn, T.C.; et al. Quantification of latent tissue reservoirs and total body viral load in HIV-1 infection. Nature 1997, 387, 183–188. [Google Scholar] [CrossRef]
- Chun, T.W.; Finzi, D.; Margolick, J.; Chadwick, K.; Schwartz, D.; Siliciano, R.F. In vivo fate of HIV-1-infected T cells: Quantitative analysis of the transition to stable latency. Nat. Med. 1995, 1, 1284–1290. [Google Scholar] [CrossRef]
- Chun, T.W.; Engel, D.; Berrey, M.M.; Shea, T.; Corey, L.; Fauci, A.S. Early establishment of a pool of latently infected, resting CD4+ T cells during primary HIV-1 infection. Proc. Natl. Acad. Sci. USA 1998, 95, 8869–8873. [Google Scholar] [CrossRef] [Green Version]
- Siliciano, J.D.; Kajdas, J.; Finzi, D.; Quinn, T.C.; Chadwick, K.; Margolick, J.B.; Kovacs, C.; Gange, S.J.; Siliciano, R.F. Long-term follow-up studies confirm the stability of the latent reservoir for HIV-1 in resting CD4+ T cells. Nat. Med. 2003, 9, 727–728. [Google Scholar] [CrossRef]
- Ho, Y.C.; Shan, L.; Hosmane, N.N.; Wang, J.; Laskey, S.B.; Rosenbloom, D.I.S.; Lai, J.; Blankson, J.N.; Siliciano, J.D.; Siliciano, R.F. XReplication-competent noninduced proviruses in the latent reservoir increase barrier to HIV-1 cure. Cell 2013, 155, 540. [Google Scholar] [CrossRef] [Green Version]
- Bruner, K.M.; Murray, A.J.; Pollack, R.A.; Soliman, M.G.; Laskey, S.B.; Capoferri, A.A.; Lai, J.; Strain, M.C.; Lada, S.M.; Hoh, R.; et al. Defective proviruses rapidly accumulate during acute HIV-1 infection. Nat. Med. 2016, 22, 1043–1049. [Google Scholar] [CrossRef] [Green Version]
- Imamichia, H.; Dewar, R.L.; Adelsberger, J.W.; Rehm, C.A.; O’doherty, U.; Paxinos, E.E.; Fauci, A.S.; Lane, H.C. Defective HIV-1 proviruses produce novel proteincoding RNA species in HIV-infected patients on combination antiretroviral therapy. Proc. Natl. Acad. Sci. USA 2016, 113, 8783–8788. [Google Scholar] [CrossRef] [Green Version]
- Hiener, B.; Horsburgh, B.A.; Eden, J.S.; Barton, K.; Schlub, T.E.; Lee, E.; von Stockenstrom, S.; Odevall, L.; Milush, J.M.; Liegler, T.; et al. Identification of Genetically Intact HIV-1 Proviruses in Specific CD4+ T Cells from Effectively Treated Participants. Cell Rep. 2017, 21, 813–822. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, G.Q.; Orlova-Fink, N.; Einkauf, K.; Chowdhury, F.Z.; Sun, X.; Harrington, S.; Kuo, H.H.; Hua, S.; Chen, H.R.; Ouyang, Z.; et al. Clonal expansion of genome-intact HIV-1 in functionally polarized Th1 CD4+ T cells. J. Clin. Investig. 2017, 127, 2689–2696. [Google Scholar] [CrossRef] [PubMed]
- Pollack, R.A.; Jones, R.B.; Pertea, M.; Bruner, K.M.; Martin, A.R.; Thomas, A.S.; Capoferri, A.A.; Beg, S.A.; Huang, S.H.; Karandish, S.; et al. Defective HIV-1 Proviruses Are Expressed and Can Be Recognized by Cytotoxic T Lymphocytes, which Shape the Proviral Landscape. Cell Host Microbe 2017, 21, 494–506. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Siliciano, J.D.; Siliciano, R.F. Enhanced culture assay for detection and quantitation of latently infected, resting CD4+ T-cells carrying replication-competent virus in HIV-1-infected individuals. Methods Mol. Biol. 2005, 304, 3–15. [Google Scholar] [CrossRef] [PubMed]
- Laird, G.M.; Rosenbloom, D.I.S.; Lai, J.; Siliciano, R.F.; Siliciano, J.D. Measuring the frequency of latent HIV-1 in resting CD4+ T cells using a limiting dilution coculture assay. Methods Mol. Biol. 2016, 1354, 239–253. [Google Scholar] [CrossRef]
- Finzi, D.; Blankson, J.; Siliciano, J.D.; Margolick, J.B.; Chadwick, K.; Pierson, T.; Smith, K.; Lisziewicz, J.; Lori, F.; Flexner, C.; et al. Latent infection of CD4+ T cells provides a mechanism for lifelong persistence of HIV-1, even in patients on effective combination therapy. Nat. Med. 1999, 5, 512–517. [Google Scholar] [CrossRef]
- Strain, M.C.; Günthard, H.F.; Havlir, D.V.; Ignacio, C.C.; Smith, D.M.; Leigh-Brown, A.J.; Macaranas, T.R.; Lam, R.Y.; Daly, O.A.; Fischer, M.; et al. Heterogeneous clearance rates of long-lived lymphocytes infected with HIV: Intrinsic stability predicts lifelong persistence. Proc. Natl. Acad. Sci. USA 2003, 100, 4819–4824. [Google Scholar] [CrossRef] [Green Version]
- Crooks, A.M.; Bateson, R.; Cope, A.B.; Dahl, N.P.; Griggs, M.K.; Kuruc, J.D.; Gay, C.L.; Eron, J.J.; Margolis, D.M.; Bosch, R.J.; et al. Precise Quantitation of the Latent HIV-1 Reservoir: Implications for Eradication Strategies. J. Infect. Dis. 2015, 212, 1361–1365. [Google Scholar] [CrossRef]
- Bruner, K.M.; Wang, Z.; Simonetti, F.R.; Bender, A.M.; Kwon, K.J.; Sengupta, S.; Fray, E.J.; Beg, S.A.; Antar, A.A.R.; Jenike, K.M.; et al. A quantitative approach for measuring the reservoir of latent HIV-1 proviruses. Nature 2019, 566, 120–125. [Google Scholar] [CrossRef]
- Hosmane, N.N.; Kwon, K.J.; Bruner, K.M.; Capoferri, A.A.; Beg, S.; Rosenbloom, D.I.S.; Keele, B.F.; Ho, Y.C.; Siliciano, J.D.; Siliciano, R.F. Proliferation of latently infected CD4+ T cells carrying replication-competent HIV-1: Potential role in latent reservoir dynamics. J. Exp. Med. 2017, 214, 959–972. [Google Scholar] [CrossRef]
- Cohn, L.B.; Silva, I.T.; Oliveira, T.Y.; Rosales, R.A.; Parrish, E.H.; Learn, G.H.; Hahn, B.H.; Czartoski, J.L.; McElrath, M.J.; Lehmann, C.; et al. HIV-1 integration landscape during latent and active infection. Cell 2015, 160, 420–432. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Garcia-Broncano, P.; Maddali, S.; Einkauf, K.B.; Jiang, C.; Gao, C.; Chevalier, J.; Chowdhury, F.Z.; Maswabi, K.; Ajibola, G.; Moyo, S.; et al. Early antiretroviral therapy in neonates with HIV-1 infection restricts viral reservoir size and induces a distinct innate immune profile. Sci. Transl. Med. 2019, 11. [Google Scholar] [CrossRef] [PubMed]
- Einkauf, K.B.; Lee, G.Q.; Gao, C.; Sharaf, R.; Sun, X.; Hua, S.; Chen, S.M.; Jiang, C.; Lian, X.; Chowdhury, F.Z.; et al. Intact HIV-1 proviruses accumulate at distinct chromosomal positions during prolonged antiretroviral therapy. J. Clin. Investig. 2019, 129, 988–998. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Abrahams, M.R.; Joseph, S.B.; Garrett, N.; Tyers, L.; Moeser, M.; Archin, N.; Council, O.D.; Matten, D.; Zhou, S.; Doolabh, D.; et al. The replication-competent HIV-1 latent reservoir is primarily established near the time of therapy initiation. Sci. Transl. Med. 2019, 11. [Google Scholar] [CrossRef] [PubMed]
- Maldarelli, F.; Wu, X.; Su, L.; Simonetti, F.R.; Shao, W.; Hill, S.; Spindler, J.; Ferris, A.L.; Mellors, J.W.; Kearney, M.F.; et al. HIV latency. Specific HIV integration sites are linked to clonal expansion and persistence of infected cells. Science 2014, 345, 179–183. [Google Scholar] [CrossRef] [PubMed]
- Simonetti, F.R.; Sobolewski, M.D.; Fyne, E.; Shao, W.; Spindler, J.; Hattori, J.; Anderson, E.M.; Watters, S.A.; Hill, S.; Wu, X.; et al. Clonally expanded CD4+ T cells can produce infectious HIV-1 in vivo. Proc. Natl. Acad. Sci. USA 2016, 113, 1883–1888. [Google Scholar] [CrossRef] [Green Version]
- Bui, J.K.; Sobolewski, M.D.; Keele, B.F.; Spindler, J.; Musick, A.; Wiegand, A.; Luke, B.T.; Shao, W.; Hughes, S.H.; Coffin, J.M.; et al. Proviruses with identical sequences comprise a large fraction of the replication-competent HIV reservoir. PLoS Pathog. 2017, 13, e1006283. [Google Scholar] [CrossRef]
- Lorenzi, J.C.C.; Cohen, Y.Z.; Cohn, L.B.; Kreider, E.F.; Barton, J.P.; Learn, G.H.; Oliveira, T.; Lavine, C.L.; Horwitz, J.A.; Settler, A.; et al. Paired quantitative and qualitative assessment of the replication-competent HIV-1 reservoir and comparison with integrated proviral DNA. Proc. Natl. Acad. Sci. USA 2016, 113, E7908–E7916. [Google Scholar] [CrossRef] [Green Version]
- Chomont, N.; El-Far, M.; Ancuta, P.; Trautmann, L.; Procopio, F.A.; Yassine-Diab, B.; Boucher, G.; Boulassel, M.R.; Ghattas, G.; Brenchley, J.M.; et al. HIV reservoir size and persistence are driven by T cell survival and homeostatic proliferation. Nat. Med. 2009, 15, 893–900. [Google Scholar] [CrossRef]
- Wang, Z.; Gurule, E.E.; Brennan, T.P.; Gerold, J.M.; Kwon, K.J.; Hosmane, N.N.; Kumar, M.R.; Beg, S.A.; Capoferri, A.A.; Ray, S.C.; et al. Expanded cellular clones carrying replication-competent HIV-1 persist, wax, and wane. Proc. Natl. Acad. Sci. USA 2018, 115, E2575–E2584. [Google Scholar] [CrossRef] [Green Version]
- Zerbato, J.M.; Serrao, E.; Lenzi, G.; Kim, B.; Ambrose, Z.; Watkins, S.C.; Engelman, A.N.; Sluis-Cremer, N. Establishment and Reversal of HIV-1 Latency in Naive and Central Memory CD4+ T Cells In Vitro. J. Virol. 2016, 90, 8059–8073. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dobrowolski, C.; Valadkhan, S.; Graham, A.C.; Shukla, M.; Ciuffi, A.; Telenti, A.; Karn, J. Entry of Polarized Effector Cells into Quiescence Forces HIV Latency. mBio 2019, 10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Agosto, L.M.; Henderson, A.J. CD4(+) T Cell Subsets and Pathways to HIV Latency. Aids. Res. Hum. Retrovir. 2018, 34, 780–789. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Agosto, L.M.; Herring, M.B.; Mothes, W.; Henderson, A.J. HIV-1-Infected CD4+ T Cells Facilitate Latent Infection of Resting CD4+ T Cells through Cell-Cell Contact. Cell Rep. 2018, 24, 2088–2100. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shan, L.; Deng, K.; Gao, H.; Xing, S.; Capoferri, A.A.; Durand, C.M.; Rabi, S.A.; Laird, G.M.; Kim, M.; Hosmane, N.N.; et al. Transcriptional Reprogramming during Effector-to-Memory Transition Renders CD4(+) T Cells Permissive for Latent HIV-1 Infection. Immunity 2017, 47, 766–775.e3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Julg, B.; Barouch, D.H. HIV-1 Latency by Transition. Immunity 2017, 47, 611–612. [Google Scholar] [CrossRef]
- Chavez, L.; Calvanese, V.; Verdin, E. HIV Latency Is Established Directly and Early in Both Resting and Activated Primary CD4 T Cells. PLoS Pathog. 2015, 11, e1004955. [Google Scholar] [CrossRef] [Green Version]
- Lenasi, T.; Contreras, X.; Peterlin, B.M. Transcriptional interference antagonizes proviral gene expression to promote HIV latency. Cell Host Microbe 2008, 4, 123–133. [Google Scholar] [CrossRef] [Green Version]
- Gallastegui, E.; Millan-Zambrano, G.; Terme, J.-M.; Chavez, S.; Jordan, A. Chromatin Reassembly Factors Are Involved in Transcriptional Interference Promoting HIV Latency. J. Virol. 2011, 85, 3187–3202. [Google Scholar] [CrossRef] [Green Version]
- Verdin, E.; Paras, P.; Van Lint, C. Chromatin disruption in the promoter of human immunodeficiency virus type 1 during transcriptional activation. EMBO J. 1993, 12, 3249–3259. [Google Scholar] [CrossRef]
- Van Lint, C.; Emiliani, S.; Ott, M.; Verdin, E. Transcriptional activation and chromatin remodeling of the HIV-1 promoter in response to histone acetylation. EMBO J. 1996, 15, 1112–1120. [Google Scholar] [CrossRef] [PubMed]
- Kauder, S.E.; Bosque, A.; Lindqvist, A.; Planelles, V.; Verdin, E. Epigenetic regulation of HIV-1 latency by cytosine methylation. PLoS Pathog. 2009, 5, e1000495. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Coull, J.J.; Romerio, F.; Sun, J.-M.; Volker, J.L.; Galvin, K.M.; Davie, J.R.; Shi, Y.; Hansen, U.; Margolis, D.M. The Human Factors YY1 and LSF Repress the Human Immunodeficiency Virus Type 1 Long Terminal Repeat via Recruitment of Histone Deacetylase 1. J. Virol. 2000, 74, 6790–6799. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hsia, S.-C.V.; Shi, Y.-B. Chromatin Disruption and Histone Acetylation in Regulation of the Human Immunodeficiency Virus Type 1 Long Terminal Repeat by Thyroid Hormone Receptor. Mol. Cell. Biol. 2002, 22, 4043–4052. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chan, J.K.; Greene, W.C. NF-kappaB/Rel: Agonist and antagonist roles in HIV-1 latency. Curr. Opin. HIV Aids 2011, 6, 12–18. [Google Scholar] [CrossRef] [PubMed]
- Williams, S.A.; Chen, L.F.; Kwon, H.; Ruiz-Jarabo, C.M.; Verdin, E.; Greene, W.C. NF-κB p50 promotes HIV latency through HDAC recruitment and repression of transcriptional initiation. EMBO J. 2006, 25, 139–149. [Google Scholar] [CrossRef] [Green Version]
- Razooky, B.S.; Pai, A.; Aull, K.; Rouzine, I.M.; Weinberger, L.S. A hardwired HIV latency program. Cell 2015, 160, 990–1001. [Google Scholar] [CrossRef] [Green Version]
- Fauci, A.S.; Redfield, R.R.; Sigounas, G.; Weahkee, M.D.; Giroir, B.P. Ending the HIV Epidemic: A Plan for the United States. JAMA 2019, 321, 844–845. [Google Scholar] [CrossRef] [Green Version]
- Mbonye, U.; Karn, J. Control of HIV latency by epigenetic and non-epigenetic mechanisms. Curr. Hiv Res. 2011, 9, 554–567. [Google Scholar]
- Transcriptional control of HIV latency: Cellular signaling pathways, epigenetics, happenstance and the hope for a cure. Virology 2014, 454–455, 328–339. [CrossRef] [Green Version]
- Mbonye, U.; Karn, J. The Molecular Basis for Human Immunodeficiency Virus Latency. Annu. Rev. Virol. 2017, 4, 261–285. [Google Scholar] [CrossRef] [PubMed]
- Hughes, S.H.; Coffin, J.M. What Integration Sites Tell Us about HIV Persistence. Cell Host Microbe 2016, 19, 588–598. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Michieletto, D.; Lusic, M.; Marenduzzo, D.; Orlandini, E. Physical principles of retroviral integration in the human genome. Nat. Commun. 2019, 10, 1–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Han, Y.; Lin, Y.B.; An, W.; Xu, J.; Yang, H.C.; O’Connell, K.; Dordai, D.; Boeke, J.D.; Siliciano, J.D.; Siliciano, R.F. Orientation-Dependent Regulation of Integrated HIV-1 Expression by Host Gene Transcriptional Readthrough. Cell Host Microbe 2008, 4, 134–146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schröder, A.R.W.; Shinn, P.; Chen, H.; Berry, C.; Ecker, J.R.; Bushman, F. HIV-1 Integration in the Human Genome Favors Active Genes and Local Hotspots. Cell 2002, 110, 521–529. [Google Scholar] [CrossRef] [Green Version]
- Han, Y.; Lassen, K.; Monie, D.; Sedaghat, A.R.; Shimoji, S.; Liu, X.; Pierson, T.C.; Margolick, J.B.; Siliciano, R.F.; Siliciano, J.D. Resting CD4+ T Cells from Human Immunodeficiency Virus Type 1 (HIV-1)-Infected Individuals Carry Integrated HIV-1 Genomes within Actively Transcribed Host Genes. J. Virol. 2004, 78, 6122–6133. [Google Scholar] [CrossRef] [Green Version]
- Marini, B.; Kertesz-Farkas, A.; Ali, H.; Lucic, B.; Lisek, K.; Manganaro, L.; Pongor, S.; Luzzati, R.; Recchia, A.; Mavilio, F.; et al. Nuclear architecture dictates HIV-1 integration site selection. Nature 2015, 521, 227–231. [Google Scholar] [CrossRef]
- Lucic, B.; Chen, H.C.; Kuzman, M.; Zorita, E.; Wegner, J.; Minneker, V.; Wang, W.; Fronza, R.; Laufs, S.; Schmidt, M.; et al. Spatially clustered loci with multiple enhancers are frequent targets of HIV-1 integration. Nat. Commun. 2019, 10, 1–12. [Google Scholar] [CrossRef] [Green Version]
- Wong, R.W.; Mamede, J.I.; Hope, T.J. Impact of Nucleoporin-Mediated Chromatin Localization and Nuclear Architecture on HIV Integration Site Selection. J. Virol. 2015, 89, 9702–9705. [Google Scholar] [CrossRef] [Green Version]
- Kane, M.; Rebensburg, S.V.; Takata, M.A.; Zang, T.M.; Yamashita, M.; Kvaratskhelia, M.; Bieniasz, P.D. Nuclear pore heterogeneity influences HIV-1 infection and the antiviral activity of MX2. eLlife 2018, 7. [Google Scholar] [CrossRef]
- Lusic, M.; Siliciano, R.F. Nuclear landscape of HIV-1 infection and integration. Nat. Rev. Microbiol. 2017, 15, 69–82. [Google Scholar] [CrossRef] [PubMed]
- Anderson, E.M.; Maldarelli, F. The role of integration and clonal expansion in HIV infection: Live long and prosper. Retrovirology 2018, 15, 71. [Google Scholar] [CrossRef] [PubMed]
- Dekker, J.; Mirny, L. The 3D Genome as Moderator of Chromosomal Communication. Cell 2016, 164, 1110–1121. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yu, M.; Ren, B. The Three-Dimensional Organization of Mammalian Genomes. Annu. Rev. Cell Dev. Biol. 2017, 33, 265–289. [Google Scholar] [CrossRef] [PubMed]
- Meaburn, K.J.; Misteli, T. Cell biology: Chromosome territories. Nature 2007, 445, 379–781. [Google Scholar] [CrossRef] [PubMed]
- Lieberman-Aiden, E.; van Berkum, N.L.; Williams, L.; Imakaev, M.; Ragoczy, T.; Telling, A.; Amit, I.; Lajoie, B.R.; Sabo, P.J.; Dorschner, M.O.; et al. Comprehensive mapping of long-range interactions reveals folding principles of the human genome. Science 2009, 326, 289–293. [Google Scholar] [CrossRef] [PubMed]
- Rao, S.S.P.; Huntley, M.H.; Durand, N.C.; Stamenova, E.K.; Bochkov, I.D.; Robinson, J.T.; Sanborn, A.L.; Machol, I.; Omer, A.D.; Lander, E.S.; et al. A 3D map of the human genome at kilobase resolution reveals principles of chromatin looping. Cell 2014, 159, 1665–1680. [Google Scholar] [CrossRef] [Green Version]
- Whyte, W.A.; Orlando, D.A.; Hnisz, D.; Abraham, B.J.; Lin, C.Y.; Kagey, M.H.; Rahl, P.B.; Lee, T.I.; Young, R.A. Master transcription factors and mediator establish super-enhancers at key cell identity genes. Cell 2013, 153, 307–319. [Google Scholar] [CrossRef] [Green Version]
- Dixon, J.R.; Selvaraj, S.; Yue, F.; Kim, A.; Li, Y.; Shen, Y.; Hu, M.; Liu, J.S.; Ren, B. Topological domains in mammalian genomes identified by analysis of chromatin interactions. Nature 2012, 485, 376–380. [Google Scholar] [CrossRef] [Green Version]
- Bukrinsky, M.I.; Stanwick, T.L.; Dempsey, M.P.; Stevenson, M. Quiescent T lymphocytes as an inducible virus reservoir in HIV-1 infection. Science 1991, 254, 423–427. [Google Scholar] [CrossRef]
- Jordan, A.; Defechereux, P.; Verdin, E. The site of HIV-1 integration in the human genome determines basal transcriptional activity and response to Tat transactivation. EMBO J. 2001, 20, 1726–1738. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sherrill-Mix, S.; Lewinski, M.K.; Famiglietti, M.; Bosque, A.; Malani, N.; Ocwieja, K.E.; Berry, C.C.; Looney, D.; Shan, L.; Agosto, L.M.; et al. HIV latency and integration site placement in five cell-based models. Retrovirology 2013, 10, 90. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wagner, T.A.; McLaughlin, S.; Garg, K.; Cheung, C.Y.K.; Larsen, B.B.; Styrchak, S.; Huang, H.C.; Edlefsen, P.T.; Mullins, J.I.; Frenkel, L.M. Proliferation of cells with HIV integrated into cancer genes contributes to persistent infection. Science 2014, 345, 570–573. [Google Scholar] [CrossRef] [PubMed]
- Chuong, E.B.; Elde, N.C.; Feschotte, C. Regulatory activities of transposable elements: From conflicts to benefits. Nat. Rev. Genet. 2017, 18, 71–86. [Google Scholar] [CrossRef] [Green Version]
- Ruess, H.; Lee, J.; Guzman, C.; Malladi, V.; D’Orso, I. An integrated genomics approach towards deciphering human genome codes shaping HIV-1 proviral transcription and fate. bioRxiv 2020. [Google Scholar] [CrossRef]
- Emiliani, S.; Fischle, W.; Ott, M.; Van Lint, C.; Amella, C.A.; Verdin, E. Mutations in the tat Gene Are Responsible for Human Immunodeficiency Virus Type 1 Postintegration Latency in the U1 Cell Line. J. Virol. 1998, 72, 1666–1670. [Google Scholar] [CrossRef] [Green Version]
- Emiliani, S.; Van Lint, C.; Fischle, W.; Paras, P.; Ott, M.; Brady, J.; Verdin, E. A point mutation in the HIV-1 Tat responsive element is associated with postintegration latency. Proc. Natl. Acad. Sci. USA 1996, 93, 6377–6381. [Google Scholar] [CrossRef] [Green Version]
- Feinberg, M.B.; Baltimore, D.; Frankel, A.D. The role of Tat in the human immunodeficiency virus life cycle indicates a primary effect on transcriptional elongation. Proc. Natl. Acad. Sci. USA 1991, 88, 4045–4049. [Google Scholar] [CrossRef] [Green Version]
- Nabel, G.; Baltimore, D. An inducible transcription factor activates expression of human immunodeficiency virus in T cells. Nature 1987, 326, 711–713. [Google Scholar] [CrossRef]
- Pache, L.; Dutra, M.S.; Spivak, A.M.; Marlett, J.M.; Murry, J.P.; Hwang, Y.; Maestre, A.M.; Manganaro, L.; Vamos, M.; Teriete, P.; et al. BIRC2/cIAP1 Is a Negative Regulator of HIV-1 Transcription and Can Be Targeted by Smac Mimetics to Promote Reversal of Viral Latency. Cell Host Microbe 2015, 18, 345–353. [Google Scholar] [CrossRef] [Green Version]
- Kinoshita, S.; Su, L.; Amano, M.; Timmerman, L.A.; Kaneshima, H.; Nolan, G.P. The T cell activation factor NF-ATc positively regulates HIV-1 replication and gene expression in T cells. Immunity 1997, 6, 235–244. [Google Scholar] [CrossRef] [Green Version]
- Morton, E.L.; Forst, C.V.; Zheng, Y.; DePaula-Silva, A.B.; Ramirez, N.G.P.; Planelles, V.; D’Orso, I. Transcriptional Circuit Fragility Influences HIV Proviral Fate. Cell Rep. 2019, 27, 154–171.e9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Weinberger, L.S.; Burnett, J.C.; Toettcher, J.E.; Arkin, A.P.; Schaffer, D.V. Stochastic Gene Expression in a Lentiviral Positive-Feedback Loop: HIV-1 Tat Fluctuations Drive Phenotypic Diversity. Cell 2005, 122, 169–182. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Singh, A.; Razooky, B.; Cox, C.D.; Simpson, M.L.; Weinberger, L.S. Transcriptional Bursting from the HIV-1 Promoter Is a Significant Source of Stochastic Noise in HIV-1 Gene Expression. Biophys. J. 2010, 98, L32–L34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Razooky, B.S.; Weinberger, L.S. Mapping the architecture of the HIV-1 Tat circuit: A decision-making circuit that lacks bistability and exploits stochastic noise. Methods 2011, 53, 68–77. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rouzine, I.M.; Razooky, B.S.; Weinberger, L.S. Stochastic variability in HIV affects viral eradication. Proc. Natl. Acad. Sci. USA 2014, 111, 13251–13252. [Google Scholar] [CrossRef] [Green Version]
- Jordan, A.; Bisgrove, D.; Verdin, E. HIV reproducibly establishes a latent infection after acute infection of T cells in vitro. EMBO J. 2003, 22, 1868–1877. [Google Scholar] [CrossRef] [Green Version]
- Lassen, K.G.; Hebbeler, A.M.; Bhattacharyya, D.; Lobritz, M.A.; Greene, W.C. A Flexible Model of HIV-1 Latency Permitting Evaluation of Many Primary CD4 T-Cell Reservoirs. PLoS ONE 2012, 7, e30176. [Google Scholar] [CrossRef] [Green Version]
- Cillo, A.R.; Sobolewski, M.D.; Bosch, R.J.; Fyne, E.; Piatak, M.; Coffin, J.M.; Mellors, J.W. Quantification of HIV-1 latency reversal in resting CD4+ T cells from patients on suppressive antiretroviral therapy. Proc. Natl. Acad. Sci. USA 2014, 111, 7078–7083. [Google Scholar] [CrossRef] [Green Version]
- Golumbeanu, M.; Cristinelli, S.; Rato, S.; Munoz, M.; Cavassini, M.; Beerenwinkel, N.; Ciuffi, A. Single-Cell RNA-Seq Reveals Transcriptional Heterogeneity in Latent and Reactivated HIV-Infected Cells. Cell Rep. 2018, 23, 942–950. [Google Scholar] [CrossRef] [Green Version]
- Siliciano, R.F.; Greene, W.C. HIV latency. Cold Spring Harb. Perspect. Med. 2011, 1, a007096. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Klaver, B.; Berkhout, B. Comparison of 5′ and 3′ long terminal repeat promoter function in human immunodeficiency virus. J. Virol. 1994, 68, 3830–3840. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cullen, B.R.; Lomedico, P.T.; Ju, G. Transcriptional interference in avian retroviruses—implications for the promoter insertion model of leukaemogenesis. Nature 1984, 307, 241–245. [Google Scholar] [CrossRef] [PubMed]
- Wu, Y. HIV-1 gene expression: Lessons from provirus and non-integrated DNA. Retrovirology 2004, 1, 13. [Google Scholar] [CrossRef] [Green Version]
- Delannoy, A.; Poirier, M.; Bell, B. Cat and Mouse: HIV Transcription in Latency, Immune Evasion and Cure/Remission Strategies. Viruses 2019, 11, 269. [Google Scholar] [CrossRef] [Green Version]
- Taube, R.; Peterlin, M. Lost in transcription: Molecular mechanisms that control HIV latency. Viruses 2013, 5, 902–927. [Google Scholar] [CrossRef] [Green Version]
- Pereira, L.A. A compilation of cellular transcription factor interactions with the HIV-1 LTR promoter. Nucleic Acids Res. 2000, 28, 663–668. [Google Scholar] [CrossRef]
- Mousseau, G.; Valente, S.T. Role of Host Factors on the Regulation of Tat-Mediated HIV-1 Transcription. Curr. Pharm. Des. 2017, 23, 4079–4090. [Google Scholar] [CrossRef] [Green Version]
- Jones, K.; Kadonaga, J.; Luciw, P.; Tjian, R. Activation of the AIDS retrovirus promoter by the cellular transcription factor, Sp1. Science 1986, 232, 755–759. [Google Scholar] [CrossRef]
- Garcia, J.A.; Harrich, D.; Soultanakis, E.; Wu, F.; Mitsuyasu, R.; Gaynor, R.B. Human immunodeficiency virus type 1 LTR TATA and TAR region sequences required for transcriptional regulation. EMBO J. 1989, 8, 765–778. [Google Scholar] [CrossRef]
- Rittner, K.; Churcher, M.J.; Gait, M.J.; Karn, J. The Human Immunodeficiency Virus Long Terminal Repeat Includes a Specialised Initiator Element which is Required for Tat-responsive Transcription. J. Mol. Biol. 1995, 248, 562–580. [Google Scholar] [CrossRef] [PubMed]
- Xiao, H.; Lis, J.T.; Jeang, K.T. Promoter activity of Tat at steps subsequent to TATA-binding protein recruitment. Mol. Cell. Biol. 1997, 17, 6898–6905. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Perkins, N.D.; Edwards, N.L.; Duckett, C.S.; Agranoff, A.B.; Schmid, R.M.; Nabel, G.J. A cooperative interaction between NF-kappa B and Sp1 is required for HIV-1 enhancer activation. EMBO J. 1993, 12, 3551–3558. [Google Scholar] [CrossRef] [PubMed]
- Perkins, N.D.; Agranoff, A.B.; Pascal, E.; Nabel, G.J. An interaction between the DNA-binding domains of RelA(p65) and Sp1 mediates human immunodeficiency virus gene activation. Mol. Cell. Biol. 1994, 14, 6570–6583. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sif, S.; Gilmore, T.D. Interaction of the v-Rel oncoprotein with cellular transcription factor Sp1. J. Virol. 1994, 68, 7131–7138. [Google Scholar] [CrossRef] [Green Version]
- Bosque, A.; Planelles, V. Induction of HIV-1 latency and reactivation in primary memory CD4+ T cells. Blood 2009, 113, 58–65. [Google Scholar] [CrossRef] [Green Version]
- Cron, R.Q.; Bartz, S.R.; Clausell, A.; Bort, S.J.; Klebanoff, S.J.; Lewis, D.B. NFAT1 Enhances HIV-1 Gene Expression in Primary Human CD4 T Cells. Clin. Immunol. 2000, 94, 179–191. [Google Scholar] [CrossRef]
- Kim, Y.K.; Mbonye, U.; Hokello, J.; Karn, J. T-Cell Receptor Signaling Enhances Transcriptional Elongation from Latent HIV Proviruses by Activating P-TEFb through an ERK-Dependent Pathway. J. Mol. Biol. 2011, 410, 896–916. [Google Scholar] [CrossRef] [Green Version]
- Chen, B.K.; Feinberg, M.B.; Baltimore, D. The kappaB sites in the human immunodeficiency virus type 1 long terminal repeat enhance virus replication yet are not absolutely required for viral growth. J. Virol. 1997, 71, 5495–5504. [Google Scholar] [CrossRef] [Green Version]
- Alcamí, J.; Laín de Lera, T.; Folgueira, L.; Pedraza, M.A.; Jacqué, J.M.; Bachelerie, F.; Noriega, A.R.; Hay, R.T.; Harrich, D.; Gaynor, R.B. Absolute dependence on kappa B responsive elements for initiation and Tat-mediated amplification of HIV transcription in blood CD4 T lymphocytes. EMBO J. 1995, 14, 1552–1560. [Google Scholar] [CrossRef]
- Tyagi, M.; Pearson, R.J.; Karn, J. Establishment of HIV Latency in Primary CD4+ Cells Is due to Epigenetic Transcriptional Silencing and P-TEFb Restriction. J. Virol. 2010, 84, 6425–6437. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Brooks, D.G.; Arlen, P.A.; Gao, L.; Kitchen, C.M.R.; Zack, J.A. Identification of T cell-signaling pathways that stimulate latent HIV in primary cells. Proc. Natl. Acad. Sci. USA 2003, 100, 12955–12960. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Coiras, M.; López-Huertas, M.; Rullas, J.; Mittelbrunn, M.; Alcamí, J. Basal shuttle of NF-κB/IκBα in resting T lymphocytes regulates HIV-1 LTR dependent expression. Retrovirology 2007, 4, 56. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ne, E.; Palstra, R.-J.; Mahmoudi, T. Transcription: Insights From the HIV-1 Promoter. Int. Rev. Cell Mol. Biol. 2018, 335, 191–243. [Google Scholar] [CrossRef] [PubMed]
- Ruelas, D.S.; Greene, W.C. An Integrated Overview of HIV-1 Latency. Cell 2013, 155, 519–529. [Google Scholar] [CrossRef] [Green Version]
- Karin, M.; Liu, Z.; Zandi, E. AP-1 function and regulation. Curr. Opin. Cell Biol. 1997, 9, 240–246. [Google Scholar] [CrossRef]
- Yang, X.; Chen, Y.; Gabuzda, D. ERK MAP Kinase Links Cytokine Signals to Activation of Latent HIV-1 Infection by Stimulating a Cooperative Interaction of AP-1 and NF-κB. J. Biol. Chem. 1999, 274, 27981–27988. [Google Scholar] [CrossRef] [Green Version]
- Duverger, A.; Wolschendorf, F.; Zhang, M.; Wagner, F.; Hatcher, B.; Jones, J.; Cron, R.Q.; van der Sluis, R.M.; Jeeninga, R.E.; Berkhout, B.; et al. An AP-1 Binding Site in the Enhancer/Core Element of the HIV-1 Promoter Controls the Ability of HIV-1 To Establish Latent Infection. J. Virol. 2013, 87, 2264–2277. [Google Scholar] [CrossRef] [Green Version]
- El Kharroubi, A.; Piras, G.; Zensen, R.; Martin, M.A. Transcriptional Activation of the Integrated Chromatin-Associated Human Immunodeficiency Virus Type 1 Promoter. Mol. Cell. Biol. 1998, 18, 2535–2544. [Google Scholar] [CrossRef] [Green Version]
- Olson, A.; Basukala, B.; Wong, W.W.; Henderson, A.J. Targeting HIV-1 proviral transcription. Curr. Opin. Virol. 2019, 38, 89–96. [Google Scholar] [CrossRef]
- Muse, G.W.; Gilchrist, D.A.; Nechaev, S.; Shah, R.; Parker, J.S.; Grissom, S.F.; Zeitlinger, J.; Adelman, K. RNA polymerase is poised for activation across the genome. Nat. Genet. 2007, 39, 1507–1511. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, F.X.; Smith, E.R.; Shilatifard, A. Born to run: Control of transcription elongation by RNA polymerase II. Nat. Rev. Mol. Cell Biol. 2018, 19, 464–478. [Google Scholar] [CrossRef] [PubMed]
- Jadlowsky, J.K.; Wong, J.Y.; Graham, A.C.; Dobrowolski, C.; Devor, R.L.; Adams, M.D.; Fujinaga, K.; Karn, J. Negative Elongation Factor Is Required for the Maintenance of Proviral Latency but Does Not Induce Promoter-Proximal Pausing of RNA Polymerase II on the HIV Long Terminal Repeat. Mol. Cell. Biol. 2014, 34, 1911–1928. [Google Scholar] [CrossRef] [Green Version]
- Zhang, Z.; Klatt, A.; Gilmour, D.S.; Henderson, A.J. Negative elongation factor NELF represses human immunodeficiency virus transcription by pausing the RNA polymerase II complex. J. Biol. Chem. 2007, 282, 16981–16988. [Google Scholar] [CrossRef] [Green Version]
- Kao, S.Y.; Calman, A.F.; Luciw, P.A.; Peterlin, B.M. Anti-termination of transcription within the long terminal repeat of HIV-1 by tat gene product. Nature 1987, 330, 489–493. [Google Scholar] [CrossRef] [PubMed]
- Wada, T.; Takagi, T.; Yamaguchi, Y.; Ferdous, A.; Imai, T.; Hirose, S.; Sugimoto, S.; Yano, K.; Hartzog, G.A.; Winston, F.; et al. DSIF, a novel transcription elongation factor that regulates RNA polymerase II processivity, is composed of human Spt4 and Spt5 homologs. Genes Dev. 1998, 12, 343–356. [Google Scholar] [CrossRef]
- Yamaguchi, Y.; Wada, T.; Watanabe, D.; Takagi, T.; Hasegawa, J.; Handa, H. Structure and Function of the Human Transcription Elongation Factor DSIF. J. Biol. Chem. 1999, 274, 8085–8092. [Google Scholar] [CrossRef] [Green Version]
- Yamaguchi, Y.; Takagi, T.; Wada, T.; Yano, K.; Furuya, A.; Sugimoto, S.; Hasegawa, J.; Handa, H. NELF, a Multisubunit Complex Containing RD, Cooperates with DSIF to Repress RNA Polymerase II Elongation. Cell 1999, 97, 41–51. [Google Scholar] [CrossRef] [Green Version]
- Rao, J.N.; Neumann, L.; Wenzel, S.; Schweimer, K.; Rösch, P.; Wöhrl, B.M. Structural studies on the RNA-recognition motif of NELF E, a cellular negative transcription elongation factor involved in the regulation of HIV transcription. Biochem. J. 2006, 400, 449–456. [Google Scholar] [CrossRef]
- Pagano, J.M.; Kwak, H.; Waters, C.T.; Sprouse, R.O.; White, B.S.; Ozer, A.; Szeto, K.; Shalloway, D.; Craighead, H.G.; Lis, J.T. Defining NELF-E RNA Binding in HIV-1 and Promoter-Proximal Pause Regions. PLoS Genet. 2014, 10, e1004090. [Google Scholar] [CrossRef] [Green Version]
- Narita, T.; Yamaguchi, Y.; Yano, K.; Sugimoto, S.; Chanarat, S.; Wada, T.; Kim, D.-K.; Hasegawa, J.; Omori, M.; Inukai, N.; et al. Human Transcription Elongation Factor NELF: Identification of Novel Subunits and Reconstitution of the Functionally Active Complex. Mol. Cell. Biol. 2003, 23, 1863–1873. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gressel, S.; Schwalb, B.; Decker, T.M.; Qin, W.; Leonhardt, H.; Eick, D.; Cramer, P. CDK9-dependent RNA polymerase II pausing controls transcription initiation. eLife 2017, 6. [Google Scholar] [CrossRef] [PubMed]
- Shao, W.; Zeitlinger, J. Paused RNA polymerase II inhibits new transcriptional initiation. Nat. Genet. 2017, 49, 1045–1051. [Google Scholar] [CrossRef]
- Sathyan, K.M.; McKenna, B.D.; Anderson, W.D.; Duarte, F.M.; Core, L.; Guertin, M.J. An improved auxin-inducible degron system preserves native protein levels and enables rapid and specific protein depletion. Genes Dev. 2019, 33, 1441–1455. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nabet, B.; Roberts, J.M.; Buckley, D.L.; Paulk, J.; Dastjerdi, S.; Yang, A.; Leggett, A.L.; Erb, M.A.; Lawlor, M.A.; Souza, A.; et al. The dTAG system for immediate and target-specific protein degradation. Nat. Chem. Biol. 2018, 14, 431–441. [Google Scholar] [CrossRef] [PubMed]
- Burslem, G.M.; Crews, C.M. Proteolysis-Targeting Chimeras as Therapeutics and Tools for Biological Discovery. Cell 2020, 181, 102–114. [Google Scholar] [CrossRef] [PubMed]
- Aoi, Y.; Smith, E.R.; Shah, A.P.; Rendleman, E.J.; Marshall, S.A.; Woodfin, A.R.; Chen, F.X.; Shiekhattar, R.; Shilatifard, A. NELF Regulates a Promoter-Proximal Step Distinct from RNA Pol II Pause-Release. Mol. Cell 2020, 78, 261–274.e5. [Google Scholar] [CrossRef]
- Hartzog, G.A.; Basrai, M.A.; Ricupero-Hovasse, S.L.; Hieter, P.; Winston, F. Identification and analysis of a functional human homolog of the SPT4 gene of Saccharomyces cerevisiae. Mol. Cell. Biol. 1996, 16, 2848–2856. [Google Scholar] [CrossRef] [Green Version]
- Glover-Cutter, K.; Kim, S.; Espinosa, J.; Bentley, D.L. RNA polymerase II pauses and associates with pre-mRNA processing factors at both ends of genes. Nat. Struct. Mol. Biol. 2008, 15, 71–78. [Google Scholar] [CrossRef] [Green Version]
- Gilchrist, D.A.; Dos Santos, G.; Fargo, D.C.; Xie, B.; Gao, Y.; Li, L.; Adelman, K. Pausing of RNA Polymerase II Disrupts DNA-Specified Nucleosome Organization to Enable Precise Gene Regulation. Cell 2010, 143, 540–551. [Google Scholar] [CrossRef] [Green Version]
- Missra, A.; Gilmour, D.S. Interactions between DSIF (DRB sensitivity inducing factor), NELF (negative elongation factor), and the Drosophila RNA polymerase II transcription elongation complex. Proc. Natl. Acad. Sci. USA 2010, 107, 11301–11306. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wen, Y.; Shatkin, A.J. Transcription elongation factor hSPT5 stimulates mRNA capping. Genes Dev. 1999, 13, 1774–1779. [Google Scholar] [CrossRef] [PubMed]
- Marciniak, R.A.; Sharp, P.A. HIV-1 Tat protein promotes formation of more-processive elongation complexes. EMBO J. 1991, 10, 4189–4196. [Google Scholar] [CrossRef] [PubMed]
- Karn, J. The molecular biology of HIV latency: Breaking and restoring the Tat-dependent transcriptional circuit. Curr. Opin. HIV Aids 2011, 6, 4–11. [Google Scholar] [CrossRef] [PubMed]
- Mancebo, H.S.Y.; Lee, G.; Flygare, J.; Tomassini, J.; Luu, P.; Zhu, Y.; Peng, J.; Blau, C.; Hazuda, D.; Price, D.; et al. P-TEFb kinase is required for HIV Tat transcriptional activation in vivo and in vitro. Genes Dev. 1997, 11, 2633–2644. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhu, Y.; Pe’ery, T.; Peng, J.; Ramanathan, Y.; Marshall, N.; Marshall, T.; Amendt, B.; Mathews, M.B.; Price, D.H. Transcription elongation factor P-TEFb is required for HIV-1 Tat transactivation in vitro. Genes Dev. 1997, 11, 2622–2632. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wei, P.; Garber, M.E.; Fang, S.M.; Fischer, W.H.; Jones, K.A. A novel CDK9-associated C-type cyclin interacts directly with HIV-1 Tat and mediates its high-affinity, loop-specific binding to TAR RNA. Cell 1998, 92, 451–462. [Google Scholar] [CrossRef] [Green Version]
- Fujinaga, K.; Irwin, D.; Huang, Y.; Taube, R.; Kurosu, T.; Peterlin, B.M. Dynamics of Human Immunodeficiency Virus Transcription: P-TEFb Phosphorylates RD and Dissociates Negative Effectors from the Transactivation Response Element. Mol. Cell. Biol. 2004, 24, 787–795. [Google Scholar] [CrossRef] [Green Version]
- Yamada, T.; Yamaguchi, Y.; Inukai, N.; Okamoto, S.; Mura, T.; Handa, H. P-TEFb-Mediated Phosphorylation of hSpt5 C-Terminal Repeats Is Critical for Processive Transcription Elongation. Mol. Cell 2006, 21, 227–237. [Google Scholar] [CrossRef]
- Bourgeois, C.F.; Kim, Y.K.; Churcher, M.J.; West, M.J.; Karn, J. Spt5 Cooperates with Human Immunodeficiency Virus Type 1 Tat by Preventing Premature RNA Release at Terminator Sequences. Mol. Cell. Biol. 2002, 22, 1079–1093. [Google Scholar] [CrossRef] [Green Version]
- Marshall, N.F.; Peng, J.; Xie, Z.; Price, D.H. Control of RNA Polymerase II Elongation Potential by a Novel Carboxyl-terminal Domain Kinase. J. Biol. Chem. 1996, 271, 27176–27183. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ramanathan, Y.; Rajpara, S.M.; Reza, S.M.; Lees, E.; Shuman, S.; Mathews, M.B.; Pe’ery, T. Three RNA Polymerase II Carboxyl-terminal Domain Kinases Display Distinct Substrate Preferences. J. Biol. Chem. 2001, 276, 10913–10920. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Komarnitsky, P.; Cho, E.J.; Buratowski, S. Different phosphorylated forms of RNA polymerase II and associated mRNA processing factors during transcription. Genes Dev. 2000, 14, 2452–2460. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Peterlin, B.M.; Price, D.H. Controlling the Elongation Phase of Transcription with P-TEFb. Mol. Cell 2006, 23, 297–305. [Google Scholar] [CrossRef] [PubMed]
- Zhou, Q.; Li, T.; Price, D.H. RNA polymerase II elongation control. Annu. Rev. Biochem. 2012, 81, 119–143. [Google Scholar] [CrossRef] [Green Version]
- Nguyen, V.T.; Kiss, T.; Michels, A.A.; Bensaude, O. 7SK small nuclear RNA binds to and inhibits the activity of CDK9/cyclin T complexes. Nature 2001, 414, 322–325. [Google Scholar] [CrossRef]
- Yang, Z.; Zhu, Q.; Luo, K.; Zhou, Q. The 7SK small nuclear RNA inhibits the CDK9/cyclin T1 kinase to control transcription. Nature 2001, 414, 317–322. [Google Scholar] [CrossRef]
- Chen, R.; Yang, Z.; Zhou, Q. Phosphorylated Positive Transcription Elongation Factor b (P-TEFb) Is Tagged for Inhibition through Association with 7SK snRNA. J. Biol. Chem. 2004, 279, 4153–4160. [Google Scholar] [CrossRef] [Green Version]
- Li, Q.; Price, J.P.; Byers, S.A.; Cheng, D.; Peng, J.; Price, D.H. Analysis of the Large Inactive P-TEFb Complex Indicates That It Contains One 7SK Molecule, a Dimer of HEXIM1 or HEXIM2, and Two P-TEFb Molecules Containing Cdk9 Phosphorylated at Threonine 186. J. Biol. Chem. 2005, 280, 28819–28826. [Google Scholar] [CrossRef] [Green Version]
- Xue, Y.; Yang, Z.; Chen, R.; Zhou, Q. A capping-independent function of MePCE in stabilizing 7SK snRNA and facilitating the assembly of 7SK snRNP. Nucleic Acids Res. 2010, 38, 360–369. [Google Scholar] [CrossRef] [Green Version]
- Krueger, B.J.; Jeronimo, C.; Roy, B.B.; Bouchard, A.; Barrandon, C.; Byers, S.A.; Searcey, C.E.; Cooper, J.J.; Bensaude, O.; Cohen, E.A.; et al. LARP7 is a stable component of the 7SK snRNP while P-TEFb, HEXIM1 and hnRNP A1 are reversibly associated. Nucleic Acids Res. 2008, 36, 2219–2229. [Google Scholar] [CrossRef] [Green Version]
- Jeronimo, C.; Forget, D.; Bouchard, A.; Li, Q.; Chua, G.; Poitras, C.; Thérien, C.; Bergeron, D.; Bourassa, S.; Greenblatt, J.; et al. Systematic Analysis of the Protein Interaction Network for the Human Transcription Machinery Reveals the Identity of the 7SK Capping Enzyme. Mol. Cell 2007, 27, 262–274. [Google Scholar] [CrossRef] [Green Version]
- He, N.; Jahchan, N.S.; Hong, E.; Li, Q.; Bayfield, M.A.; Maraia, R.J.; Luo, K.; Zhou, Q. A La-Related Protein Modulates 7SK snRNP Integrity to Suppress P-TEFb-Dependent Transcriptional Elongation and Tumorigenesis. Mol. Cell 2008, 29, 588–599. [Google Scholar] [CrossRef] [PubMed]
- Bacon, C.W.; D’Orso, I. CDK9: A signaling hub for transcriptional control. Transcription 2019, 10, 57–75. [Google Scholar] [CrossRef] [PubMed]
- McNamara, R.P.; McCann, J.L.; Gudipaty, S.A.; D’Orso, I. Transcription factors mediate the enzymatic disassembly of promoter-bound 7SK snRNP to locally recruit P-TEFb for transcription elongation. Cell Rep. 2013, 5, 1256–1268. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Faust, T.B.; Li, Y.; Bacon, C.W.; Jang, G.M.; Weiss, A.; Jayaraman, B.; Newton, B.W.; Krogan, N.J.; D’Orso, I.; Frankel, A.D. The HIV-1 Tat protein recruits a ubiquitin ligase to reorganize the 7SK snRNP for transcriptional activation. eLife 2018, 7. [Google Scholar] [CrossRef] [PubMed]
- D’Orso, I.; Frankel, A.D. RNA-mediated displacement of an inhibitory snRNP complex activates transcription elongation. Nat. Struct. Mol. Biol. 2010, 17, 815–821. [Google Scholar] [CrossRef] [Green Version]
- D’Orso, I.; Jang, G.M.; Pastuszak, A.W.; Faust, T.B.; Quezada, E.; Booth, D.S.; Frankel, A.D. Transition Step during Assembly of HIV Tat:P-TEFb Transcription Complexes and Transfer to TAR RNA. Mol. Cell. Biol. 2012, 32, 4780–4793. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Barboric, M.; Yik, J.H.N.; Czudnochowski, N.; Yang, Z.; Chen, R.; Contreras, X.; Geyer, M.; Matija Peterlin, B.; Zhou, Q. Tat competes with HEXIM1 to increase the active pool of P-TEFb for HIV-1 transcription. Nucleic Acids Res. 2007, 35, 2003–2012. [Google Scholar] [CrossRef] [PubMed]
- Sedore, S.C.; Byers, S.A.; Biglione, S.; Price, J.P.; Maury, W.J.; Price, D.H. Manipulation of P-TEFb control machinery by HIV: Recruitment of P-TEFb from the large form by Tat and binding of HEXIM1 to TAR. Nucleic Acids Res. 2007, 35, 4347–4358. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Garber, M.E.; Mayall, T.P.; Suess, E.M.; Meisenhelder, J.; Thompson, N.E.; Jones, K.A. CDK9 Autophosphorylation Regulates High-Affinity Binding of the Human Immunodeficiency Virus Type 1 Tat-P-TEFb Complex to TAR RNA. Mol. Cell. Biol. 2000, 20, 6958–6969. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jang, M.K.; Mochizuki, K.; Zhou, M.; Jeong, H.-S.; Brady, J.N.; Ozato, K. The Bromodomain Protein Brd4 Is a Positive Regulatory Component of P-TEFb and Stimulates RNA Polymerase II-Dependent Transcription. Mol. Cell 2005, 19, 523–534. [Google Scholar] [CrossRef] [PubMed]
- Yang, Z.; Yik, J.H.N.; Chen, R.; He, N.; Moon, K.J.; Ozato, K.; Zhou, Q. Recruitment of P-TEFb for stimulation of transcriptional elongation by the bromodomain protein Brd4. Mol. Cell 2005, 19, 535–545. [Google Scholar] [CrossRef] [PubMed]
- He, N.; Liu, M.; Hsu, J.; Xue, Y.; Chou, S.; Burlingame, A.; Krogan, N.J.; Alber, T.; Zhou, Q. HIV-1 Tat and Host AFF4 Recruit Two Transcription Elongation Factors into a Bifunctional Complex for Coordinated Activation of HIV-1 Transcription. Mol. Cell 2010, 38, 428–438. [Google Scholar] [CrossRef] [Green Version]
- Sobhian, B.; Laguette, N.; Yatim, A.; Nakamura, M.; Levy, Y.; Kiernan, R.; Benkirane, M. HIV-1 Tat Assembles a Multifunctional Transcription Elongation Complex and Stably Associates with the 7SK snRNP. Mol. Cell 2010, 38, 439–451. [Google Scholar] [CrossRef] [Green Version]
- McNamara, R.P.; Reeder, J.E.; McMillan, E.A.; Bacon, C.W.; McCann, J.L.; D’Orso, I. KAP1 Recruitment of the 7SK snRNP Complex to Promoters Enables Transcription Elongation by RNA Polymerase II. Mol. Cell 2016, 61, 39–53. [Google Scholar] [CrossRef] [Green Version]
- Lin, C.; Smith, E.R.; Takahashi, H.; Lai, K.C.; Martin-Brown, S.; Florens, L.; Washburn, M.P.; Conaway, J.W.; Conaway, R.C.; Shilatifard, A. AFF4, a Component of the ELL/P-TEFb Elongation Complex and a Shared Subunit of MLL Chimeras, Can Link Transcription Elongation to Leukemia. Mol. Cell 2010, 37, 429–437. [Google Scholar] [CrossRef] [Green Version]
- Chou, S.; Upton, H.; Bao, K.; Schulze-Gahmen, U.; Samelson, A.J.; He, N.; Nowak, A.; Lu, H.; Krogan, N.J.; Zhou, Q.; et al. HIV-1 Tat recruits transcription elongation factors dispersed along a flexible AFF4 scaffold. Proc. Natl. Acad. Sci. USA 2013, 110, E123–E131. [Google Scholar] [CrossRef] [Green Version]
- Lu, H.; Li, Z.; Xue, Y.; Schulze-Gahmen, U.; Johnson, J.R.; Krogan, N.J.; Alber, T.; Zhou, Q. AFF1 is a ubiquitous P-TEFb partner to enable Tat extraction of P-TEFb from 7SK snRNP and formation of SECs for HIV transactivation. Proc. Natl. Acad. Sci. USA 2014, 111, E15–E24. [Google Scholar] [CrossRef] [Green Version]
- Luo, Z.; Lin, C.; Guest, E.; Garrett, A.S.; Mohaghegh, N.; Swanson, S.; Marshall, S.; Florens, L.; Washburn, M.P.; Shilatifard, A. The super elongation complex family of RNA polymerase II elongation factors: Gene target specificity and transcriptional output. Mol. Cell Biol. 2012, 32, 2608–2617. [Google Scholar] [CrossRef] [Green Version]
- Li, Z.; Lu, H.; Zhou, Q. A Minor Subset of Super Elongation Complexes Plays a Predominant Role in Reversing HIV-1 Latency. Mol. Cell. Biol. 2016, 36, 1194–1205. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Filippakopoulos, P.; Qi, J.; Picaud, S.; Shen, Y.; Smith, W.B.; Fedorov, O.; Morse, E.M.; Keates, T.; Hickman, T.T.; Felletar, I.; et al. Selective inhibition of BET bromodomains. Nature 2010, 468, 1067–1073. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dey, A.; Chitsaz, F.; Abbasi, A.; Misteli, T.; Ozato, K. The double bromodomain protein Brd4 binds to acetylated chromatin during interphase and mitosis. Proc. Natl. Acad. Sci. USA 2003, 100, 8758–8763. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Boehm, D.; Calvanese, V.; Dar, R.D.; Xing, S.; Schroeder, S.; Martins, L.; Aull, K.; Li, P.-C.; Planelles, V.; Bradner, J.E.; et al. BET bromodomain-targeting compounds reactivate HIV from latency via a Tat-independent mechanism. Cell Cycle 2013, 12, 452–462. [Google Scholar] [CrossRef]
- Banerjee, C.; Archin, N.; Michaels, D.; Belkina, A.C.; Denis, G.V.; Bradner, J.; Sebastiani, P.; Margolis, D.M.; Montano, M. BET bromodomain inhibition as a novel strategy for reactivation of HIV-1. J. Leukoc. Biol. 2012, 92, 1147–1154. [Google Scholar] [CrossRef]
- Kanno, T.; Kanno, Y.; LeRoy, G.; Campos, E.; Sun, H.-W.; Brooks, S.R.; Vahedi, G.; Heightman, T.D.; Garcia, B.A.; Reinberg, D.; et al. BRD4 assists elongation of both coding and enhancer RNAs by interacting with acetylated histones. Nat. Struct. Mol. Biol. 2014, 21, 1047–1057. [Google Scholar] [CrossRef] [Green Version]
- Winter, G.E.; Mayer, A.; Buckley, D.L.; Erb, M.A.; Roderick, J.E.; Vittori, S.; Reyes, J.M.; di Iulio, J.; Souza, A.; Ott, C.J.; et al. BET Bromodomain Proteins Function as Master Transcription Elongation Factors Independent of CDK9 Recruitment. Mol. Cell 2017, 67, 5–18. [Google Scholar] [CrossRef] [Green Version]
- Devaiah, B.N.; Lewis, B.A.; Cherman, N.; Hewitt, M.C.; Albrecht, B.K.; Robey, P.G.; Ozato, K.; Sims, R.J.; Singer, D.S. BRD4 is an atypical kinase that phosphorylates Serine2 of the RNA Polymerase II carboxy-terminal domain. Proc. Natl. Acad. Sci. USA 2012, 109, 6927–6932. [Google Scholar] [CrossRef] [Green Version]
- Liu, W.; Ma, Q.; Wong, K.; Li, W.; Ohgi, K.; Zhang, J.; Aggarwal, A.K.; Rosenfeld, M.G. Brd4 and JMJD6-Associated Anti-Pause Enhancers in Regulation of Transcriptional Pause Release. Cell 2013, 155, 1581–1595. [Google Scholar] [CrossRef] [Green Version]
- D’Orso, I. 7SKiing on chromatin: Move globally, act locally. RNA Biol. 2016, 13, 545–553. [Google Scholar] [CrossRef] [Green Version]
- Wolf, D.; Goff, S.P. TRIM28 mediates primer binding site-targeted silencing of murine leukemia virus in embryonic cells. Cell 2007, 131, 46–57. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rowe, H.M.; Jakobsson, J.; Mesnard, D.; Rougemont, J.; Reynard, S.; Aktas, T.; Maillard, P.V.; Layard-Liesching, H.; Verp, S.; Marquis, J.; et al. KAP1 controls endogenous retroviruses in embryonic stem cells. Nature 2010, 463, 237–240. [Google Scholar] [CrossRef] [PubMed]
- Bacon, C.W.; Challa, A.; Hyder, U.; Shukla, A.; Borkar, A.N.; Bayo, J.; Liu, J.; Wu, S.Y.; Chiang, C.-M.; Kutateladze, T.G.; et al. A chromatin reader couples steps RNA polymerase II transcription to sustain oncogenic programs. Mol. Cell 2020, 78, 1–19. [Google Scholar] [CrossRef] [PubMed]
- Jiang, C.; Pugh, B.F. Nucleosome positioning and gene regulation: Advances through genomics. Nat. Rev. Genet. 2009, 10, 161–172. [Google Scholar] [CrossRef] [Green Version]
- Steger, D.J.; Workman, J.L. Stable co-occupancy of transcription factors and histones at the HIV-1 enhancer. EMBO J. 1997, 16, 2463–2472. [Google Scholar] [CrossRef] [Green Version]
- Rafati, H.; Parra, M.; Hakre, S.; Moshkin, Y.; Verdin, E.; Mahmoudi, T. Repressive LTR Nucleosome Positioning by the BAF Complex Is Required for HIV Latency. PLoS Biol. 2011, 9, e1001206. [Google Scholar] [CrossRef] [Green Version]
- Verdin, E.; Becker, N.; Bex, F.; Droogmans, L.; Burny, A. Identification and characterization of an enhancer in the coding region of the genome of human immunodeficiency virus type 1. Proc. Natl. Acad. Sci. USA 1990, 87, 4874–4878. [Google Scholar] [CrossRef] [Green Version]
- Bushnell, D.A.; Cramer, P.; Kornberg, R.D. Structural basis of transcription: -Amanitin-RNA polymerase II cocrystal at 2.8 A resolution. Proc. Natl. Acad. Sci. USA 2002, 99, 1218–1222. [Google Scholar] [CrossRef] [Green Version]
- Wang, W.; Xue, Y.; Zhou, S.; Kuo, A.; Cairns, B.R.; Crabtree, G.R. Diversity and specialization of mammalian SWI/SNF complexes. Genes Dev. 1996, 10, 2117–2130. [Google Scholar] [CrossRef] [Green Version]
- Kadoch, C.; Crabtree, G.R. Mammalian SWI/SNF chromatin remodeling complexes and cancer: Mechanistic insights gained from human genomics. Sci. Adv. 2015, 1, e1500447. [Google Scholar] [CrossRef] [Green Version]
- Hodges, C.; Kirkland, J.G.; Crabtree, G.R. The Many Roles of BAF (mSWI/SNF) and PBAF Complexes in Cancer. Cold Spring Harb. Perspect. Med. 2016, 6, a026930. [Google Scholar] [CrossRef] [Green Version]
- Mizutani, T.; Ishizaka, A.; Tomizawa, M.; Okazaki, T.; Yamamichi, N.; Kawana-Tachikawa, A.; Iwamoto, A.; Iba, H. Loss of the Brm-Type SWI/SNF Chromatin Remodeling Complex Is a Strong Barrier to the Tat-Independent Transcriptional Elongation of Human Immunodeficiency Virus Type 1 Transcripts. J. Virol. 2009, 83, 11569–11580. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sodroski, J.; Rosen, C.; Wong-Staal, F.; Salahuddin, S.; Popovic, M.; Arya, S.; Gallo, R.; Haseltine, W. Trans-acting transcriptional regulation of human T-cell leukemia virus type III long terminal repeat. Science 1985, 227, 171–173. [Google Scholar] [CrossRef]
- Sodroski, J.; Rosen, C.; Haseltine, W. Trans-acting transcriptional activation of the long terminal repeat of human T lymphotropic viruses in infected cells. Science 1984, 225, 381–385. [Google Scholar] [CrossRef] [PubMed]
- Tréand, C.; Du Chéné, I.; Brès, V.; Kiernan, R.; Benarous, R.; Benkirane, M.; Emiliani, S. Requirement for SWI/SNF chromatin-remodeling complex in Tat-mediated activation of the HIV-1 promoter. EMBO J. 2006, 25, 1690–1699. [Google Scholar] [CrossRef] [Green Version]
- Mahmoudi, T.; Parra, M.; Vries, R.G.J.; Kauder, S.E.; Verrijzer, C.P.; Ott, M.; Verdin, E. The SWI/SNF chromatin-remodeling complex is a cofactor for Tat transactivation of the HIV promoter. J. Biol. Chem. 2006, 281, 19960–19968. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Agbottah, E.; Deng, L.; Dannenberg, L.O.; Pumfery, A.; Kashanchi, F. Effect of SWI/SNF chromatin remodeling complex on HIV-1 Tat activated transcription. Retrovirology 2006, 3, 48. [Google Scholar] [CrossRef] [Green Version]
- Stoszko, M.; De Crignis, E.; Rokx, C.; Khalid, M.M.; Lungu, C.; Palstra, R.-J.; Kan, T.W.; Boucher, C.; Verbon, A.; Dykhuizen, E.C.; et al. Small Molecule Inhibitors of BAF.; A Promising Family of Compounds in HIV-1 Latency Reversal. EBioMedicine 2016, 3, 108–121. [Google Scholar] [CrossRef] [Green Version]
- Conrad, R.J.; Fozouni, P.; Thomas, S.; Sy, H.; Zhang, Q.; Zhou, M.-M.; Ott, M. The Short Isoform of BRD4 Promotes HIV-1 Latency by Engaging Repressive SWI/SNF Chromatin-Remodeling Complexes. Mol. Cell 2017, 67, 1001–1012.e6. [Google Scholar] [CrossRef] [Green Version]
- Ott, M.; Schnölzer, M.; Garnica, J.; Fischle, W.; Emiliani, S.; Rackwitz, H.-R.; Verdin, E. Acetylation of the HIV-1 Tat protein by p300 is important for its transcriptional activity. Curr. Biol. 1999, 9, 1489–1493. [Google Scholar] [CrossRef] [Green Version]
- Kiernan, R.E. HIV-1 Tat transcriptional activity is regulated by acetylation. EMBO J. 1999, 18, 6106–6118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jäger, S.; Cimermancic, P.; Gulbahce, N.; Johnson, J.R.; McGovern, K.E.; Clarke, S.C.; Shales, M.; Mercenne, G.; Pache, L.; Li, K.; et al. Global landscape of HIV–human protein complexes. Nature 2012, 481, 365–370. [Google Scholar] [CrossRef] [PubMed]
- Benkirane, M.; Chun, R.F.; Xiao, H.; Ogryzko, V.V.; Howard, B.H.; Nakatani, Y.; Jeang, K.-T. Activation of integrated provirus requires histone acetyl- transferase. p300 and P/CAF are coactivators for HIV-1 Tat. J. Biol. Chem. 1998, 273, 24898–24905. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Col, E.; Caron, C.; Seigneurin-Berny, D.; Gracia, J.; Favier, A.; Khochbin, S. The Histone Acetyltransferase, hGCN5, Interacts with and Acetylates the HIV Transactivator, Tat. J. Biol. Chem. 2001, 276, 28179–28184. [Google Scholar] [CrossRef] [Green Version]
- Hottiger, M.O.; Nabel, G.J. Interaction of human immunodeficiency virus type 1 Tat with the transcriptional coactivators p300 and CREB binding protein. J. Virol. 1998, 72, 8252–8256. [Google Scholar] [CrossRef] [Green Version]
- Marzio, G.; Tyagi, M.; Gutierrez, M.I.; Giacca, M. HIV-1 Tat transactivator recruits p300 and CREB-binding protein histone acetyltransferases to the viral promoter. Proc. Natl. Acad. Sci. USA 1998, 95, 13519–13524. [Google Scholar] [CrossRef] [Green Version]
- Vardabasso, C.; Manganaro, L.; Lusic, M.; Marcello, A.; Giacca, M. The histone chaperone protein Nucleosome Assembly Protein-1 (hNAP-1) binds HIV-1 Tat and promotes viral transcription. Retrovirology 2008, 5, 8. [Google Scholar] [CrossRef] [Green Version]
- Gautier, V.W.; Gu, L.; O’Donoghue, N.; Pennington, S.; Sheehy, N.; Hall, W.W. In vitro nuclear interactome of the HIV-1 Tat protein. Retrovirology 2009, 6, 47. [Google Scholar] [CrossRef] [Green Version]
- Vanti, M.; Gallastegui, E.; Respaldiza, I.; Rodríguez-Gil, A.; Gómez-Herreros, F.; Jimeno-González, S.; Jordan, A.; Chávez, S. Yeast Genetic Analysis Reveals the Involvement of Chromatin Reassembly Factors in Repressing HIV-1 Basal Transcription. PLoS Genet. 2009, 5, e1000339. [Google Scholar] [CrossRef]
- Belotserkovskaya, R.; Oh, S.; Bondarenko, V.A.; Orphanides, G.; Studitsky, V.M.; Reinberg, D. FACT facilitates transcription-dependent nucleosome alteration. Science 2003, 301, 1090–1093. [Google Scholar] [CrossRef]
- Orphanides, G.; LeRoy, G.; Chang, C.-H.; Luse, D.S.; Reinberg, D. FACT, a Factor that Facilitates Transcript Elongation through Nucleosomes. Cell 1998, 92, 105–116. [Google Scholar] [CrossRef] [Green Version]
- Easley, R.; Carpio, L.; Dannenberg, L.; Choi, S.; Alani, D.; Van Duyne, R.; Guendel, I.; Klase, Z.; Agbottah, E.; Kehn-Hall, K.; et al. Transcription through the HIV-1 nucleosomes: Effects of the PBAF complex in Tat activated transcription. Virology 2010, 405, 322–333. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Huang, H.; Santoso, N.; Power, D.; Simpson, S.; Dieringer, M.; Miao, H.; Gurova, K.; Giam, C.-Z.; Elledge, S.J.; Zhu, J. FACT Proteins, SUPT16H and SSRP1, Are Transcriptional Suppressors of HIV-1 and HTLV-1 That Facilitate Viral Latency. J. Biol. Chem. 2015, 290, 27297–27310. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kaplan, C.D. Transcription Elongation Factors Repress Transcription Initiation from Cryptic Sites. Science 2003, 301, 1096–1099. [Google Scholar] [CrossRef]
- Anderson, J.; Lowary, P.; Widom, J. Effects of histone acetylation on the equilibrium accessibility of nucleosomal DNA target sites. J. Mol. Biol. 2001, 307, 977–985. [Google Scholar] [CrossRef]
- Allis, C.D.; Jenuwein, T. The molecular hallmarks of epigenetic control. Nat. Rev. Genet. 2016, 17, 487–500. [Google Scholar] [CrossRef]
- Lusic, M. Regulation of HIV-1 gene expression by histone acetylation and factor recruitment at the LTR promoter. EMBO J. 2003, 22, 6550–6561. [Google Scholar] [CrossRef] [Green Version]
- Ylisastigui, L.; Archin, N.M.; Lehrman, G.; Bosch, R.J.; Margolis, D.M. Coaxing HIV-1 from resting CD4 T cells: Histone deacetylase inhibition allows latent viral expression. AIDS 2004, 18, 1101–1108. [Google Scholar] [CrossRef]
- Archin, N.M.; Bateson, R.; Tripathy, M.K.; Crooks, A.M.; Yang, K.-H.; Dahl, N.P.; Kearney, M.F.; Anderson, E.M.; Coffin, J.M.; Strain, M.C.; et al. HIV-1 Expression Within Resting CD4+ T Cells After Multiple Doses of Vorinostat. J. Infect. Dis. 2014, 210, 728–735. [Google Scholar] [CrossRef]
- Wei, D.G.; Chiang, V.; Fyne, E.; Balakrishnan, M.; Barnes, T.; Graupe, M.; Hesselgesser, J.; Irrinki, A.; Murry, J.P.; Stepan, G.; et al. Histone Deacetylase Inhibitor Romidepsin Induces HIV Expression in CD4 T Cells from Patients on Suppressive Antiretroviral Therapy at Concentrations Achieved by Clinical Dosing. PLoS Pathog. 2014, 10, e1004071. [Google Scholar] [CrossRef] [Green Version]
- Shirakawa, K.; Chavez, L.; Hakre, S.; Calvanese, V.; Verdin, E. Reactivation of latent HIV by histone deacetylase inhibitors. Trends Microbiol. 2013, 21, 277–285. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Seto, E.; Yoshida, M. Erasers of Histone Acetylation: The Histone Deacetylase Enzymes. Cold Spring Harb. Perspect. Biol. 2014, 6, a018713. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- He, G.; Margolis, D.M. Counterregulation of Chromatin Deacetylation and Histone Deacetylase Occupancy at the Integrated Promoter of Human Immunodeficiency Virus Type 1 (HIV-1) by the HIV-1 Repressor YY1 and HIV-1 Activator Tat. Mol. Cell. Biol. 2002, 22, 2965–2973. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Margolis, D.M.; Garcia, J.V.; Hazuda, D.J.; Haynes, B.F. Latency reversal and viral clearance to cure HIV-1. Science 2016, 353. [Google Scholar] [CrossRef] [PubMed]
- Kim, Y.; Anderson, J.L.; Lewin, S.R. Getting the “Kill” into “Shock and Kill”: Strategies to Eliminate Latent HIV. Cell Host Microbe 2018, 23, 14–26. [Google Scholar] [CrossRef] [PubMed]
- Bullen, C.K.; Laird, G.M.; Durand, C.M.; Siliciano, J.D.; Siliciano, R.F. New ex vivo approaches distinguish effective and ineffective single agents for reversing HIV-1 latency in vivo. Nat. Med. 2014, 20, 425–429. [Google Scholar] [CrossRef] [Green Version]
- Rice, J.C.; Briggs, S.D.; Ueberheide, B.; Barber, C.M.; Shabanowitz, J.; Hunt, D.F.; Shinkai, Y.; Allis, C.D. Histone Methyltransferases Direct Different Degrees of Methylation to Define Distinct Chromatin Domains. Mol. Cell 2003, 12, 1591–1598. [Google Scholar] [CrossRef]
- Pearson, R.; Kim, Y.K.; Hokello, J.; Lassen, K.; Friedman, J.; Tyagi, M.; Karn, J. Epigenetic Silencing of Human Immunodeficiency Virus (HIV) Transcription by Formation of Restrictive Chromatin Structures at the Viral Long Terminal Repeat Drives the Progressive Entry of HIV into Latency. J. Virol. 2008, 82, 12291–12303. [Google Scholar] [CrossRef] [Green Version]
- Chéné, I.D.; Basyuk, E.; Lin, Y.L.; Triboulet, R.; Knezevich, A.; Chable-Bessia, C.; Mettling, C.; Baillat, V.; Reynes, J.; Corbeau, P.; et al. Suv39H1 and HP1γ are responsible for chromatin-mediated HIV-1 transcriptional silencing and post-integration latency. EMBO J. 2007, 26, 424–435. [Google Scholar] [CrossRef]
- Imai, K.; Togami, H.; Okamoto, T. Involvement of Histone H3 Lysine 9 (H3K9) Methyltransferase G9a in the Maintenance of HIV-1 Latency and Its Reactivation by BIX01294. J. Biol. Chem. 2010, 285, 16538–16545. [Google Scholar] [CrossRef] [Green Version]
- Van Duyne, R.; Easley, R.; Wu, W.; Berro, R.; Pedati, C.; Klase, Z.; Kehn-Hall, K.; Flynn, E.K.; Symer, D.E.; Kashanchi, F. Lysine methylation of HIV-1 Tat regulates transcriptional activity of the viral LTR. Retrovirology 2008, 5, 40. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lachner, M.; O’Carroll, D.; Rea, S.; Mechtler, K.; Jenuwein, T. Methylation of histone H3 lysine 9 creates a binding site for HP1 proteins. Nature 2001, 410, 116–120. [Google Scholar] [CrossRef] [PubMed]
- Maison, C.; Almouzni, G. HP1 and the dynamics of heterochromatin maintenance. Nat. Rev. Mol. Cell Biol. 2004, 5, 296–305. [Google Scholar] [CrossRef] [PubMed]
- Minc, E.; Courvalin, J.-C.; Buendia, B. HP1γ associates with euchromatin and heterochromatin in mammalian nuclei and chromosomes. Cytogenet. Genome Res. 2000, 90, 279–284. [Google Scholar] [CrossRef] [PubMed]
- Vakoc, C.R.; Mandat, S.A.; Olenchock, B.A.; Blobel, G.A. Histone H3 Lysine 9 Methylation and HP1γ Are Associated with Transcription Elongation through Mammalian Chromatin. Mol. Cell 2005, 19, 381–391. [Google Scholar] [CrossRef]
- Kwon, S.H.; Florens, L.; Swanson, S.K.; Washburn, M.P.; Abmayr, S.M.; Workman, J.L. Heterochromatin protein 1 (HP1) connects the FACT histone chaperone complex to the phosphorylated CTD of RNA polymerase II. Genes Dev. 2010, 24, 2133–2145. [Google Scholar] [CrossRef] [Green Version]
- Stewart, M.D.; Li, J.; Wong, J. Relationship between Histone H3 Lysine 9 Methylation, Transcription Repression, and Heterochromatin Protein 1 Recruitment. Mol. Cell. Biol. 2005, 25, 2525–2538. [Google Scholar] [CrossRef] [Green Version]
- Mateescu, B.; Bourachot, B.; Rachez, C.; Ogryzko, V.; Muchardt, C. Regulation of an inducible promoter by an HP1β-HP1γ switch. EMBO Rep. 2008, 9, 267–272. [Google Scholar] [CrossRef] [Green Version]
- Zhang, T.; Cooper, S.; Brockdorff, N. The interplay of histone modifications – writers that read. EMBO Rep. 2015, 16, 1467–1481. [Google Scholar] [CrossRef]
- Boyer, L.A.; Plath, K.; Zeitlinger, J.; Brambrink, T.; Medeiros, L.A.; Lee, T.I.; Levine, S.S.; Wernig, M.; Tajonar, A.; Ray, M.K.; et al. Polycomb complexes repress developmental regulators in murine embryonic stem cells. Nature 2006, 441, 349–353. [Google Scholar] [CrossRef]
- Lee, T.I.; Jenner, R.G.; Boyer, L.A.; Guenther, M.G.; Levine, S.S.; Kumar, R.M.; Chevalier, B.; Johnstone, S.E.; Cole, M.F.; Isono, K.; et al. Control of Developmental Regulators by Polycomb in Human Embryonic Stem Cells. Cell 2006, 125, 301–313. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Friedman, J.; Cho, W.-K.; Chu, C.K.; Keedy, K.S.; Archin, N.M.; Margolis, D.M.; Karn, J. Epigenetic Silencing of HIV-1 by the Histone H3 Lysine 27 Methyltransferase Enhancer of Zeste 2. J. Virol. 2011, 85, 9078–9089. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nguyen, K.; Das, B.; Dobrowolski, C.; Karn, J. Multiple Histone Lysine Methyltransferases Are Required for the Establishment and Maintenance of HIV-1 Latency. mBio 2017, 8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Boehm, D.; Jeng, M.; Camus, G.; Gramatica, A.; Schwarzer, R.; Johnson, J.R.; Hull, P.A.; Montano, M.; Sakane, N.; Pagans, S.; et al. SMYD2-Mediated Histone Methylation Contributes to HIV-1 Latency. Cell Host Microbe 2017, 21, 569–579.e6. [Google Scholar] [CrossRef] [Green Version]
- Blazkova, J.; Trejbalova, K.; Gondois-Rey, F.; Halfon, P.; Philibert, P.; Guiguen, A.; Verdin, E.; Olive, D.; Van Lint, C.; Hejnar, J.; et al. CpG Methylation Controls Reactivation of HIV from Latency. PLoS Pathog. 2009, 5, e1000554. [Google Scholar] [CrossRef] [Green Version]
- Blazkova, J.; Murray, D.; Justement, J.S.; Funk, E.K.; Nelson, A.K.; Moir, S.; Chun, T.-W.; Fauci, A.S. Paucity of HIV DNA Methylation in Latently Infected, Resting CD4+ T Cells from Infected Individuals Receiving Antiretroviral Therapy. J. Virol. 2012, 86, 5390–5392. [Google Scholar] [CrossRef] [Green Version]
- Weber, S.; Weiser, B.; Kemal, K.S.; Burger, H.; Ramirez, C.M.; Korn, K.; Anastos, K.; Kaul, R.; Kovacs, C.; Doerfler, W. Epigenetic analysis of HIV-1 proviral genomes from infected individuals: Predominance of unmethylated CpG’s. Virology 2014, 449, 181–189. [Google Scholar] [CrossRef] [Green Version]
- Pott, S. Simultaneous measurement of chromatin accessibility, DNA methylation, and nucleosome phasing in single cells. eLife 2017, 6. [Google Scholar] [CrossRef]
- Kelly, T.K.; Liu, Y.; Lay, F.D.; Liang, G.; Berman, B.P.; Jones, P.A. Genome-wide mapping of nucleosome positioning and DNA methylation within individual DNA molecules. Genome Res. 2012, 22, 2497–2506. [Google Scholar] [CrossRef] [Green Version]
- Rotem, A.; Ram, O.; Shoresh, N.; Sperling, R.A.; Goren, A.; Weitz, D.A.; Bernstein, B.E. Single-cell ChIP-seq reveals cell subpopulations defined by chromatin state. Nat. Biotechnol. 2015, 33, 1165–1172. [Google Scholar] [CrossRef]
- Mezger, A.; Klemm, S.; Mann, I.; Brower, K.; Mir, A.; Bostick, M.; Farmer, A.; Fordyce, P.; Linnarsson, S.; Greenleaf, W. High-throughput chromatin accessibility profiling at single-cell resolution. Nat. Commun. 2018, 9, 3647. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nagano, T.; Lubling, Y.; Stevens, T.J.; Schoenfelder, S.; Yaffe, E.; Dean, W.; Laue, E.D.; Tanay, A.; Fraser, P. Single-cell Hi-C reveals cell-to-cell variability in chromosome structure. Nature 2013, 502, 59–64. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kok, Y.L.; Ciuffi, A.; Metzner, K.J. Unravelling HIV-1 Latency, One Cell at a Time. Trends Microbiol. 2017, 25, 932–941. [Google Scholar] [CrossRef] [PubMed]
- Wong, J.K.; Hezareh, M.; Günthard, H.F.; Havlir, D.V.; Ignacio, C.C.; Spina, C.A.; Richman, D.D. Recovery of replication-competent HIV despite prolonged suppression of plasma viremia. Science 1997, 278, 1291–1295. [Google Scholar] [CrossRef] [PubMed]
- Cohn, L.B.; Chomont, N.; Deeks, S.G. The Biology of the HIV-1 Latent Reservoir and Implications for Cure Strategies. Cell Host Microbe 2020, 27, 519–530. [Google Scholar] [CrossRef] [PubMed]
- Gallo, R.C. HIV/AIDS Research for the Future. Cell Host Microbe 2020, 27, 499–501. [Google Scholar] [CrossRef]
- Archin, N.M.; Sung, J.M.; Garrido, C.; Soriano-Sarabia, N.; Margolis, D.M. Eradicating HIV-1 infection: Seeking to clear a persistent pathogen. Nat. Rev. Microbiol. 2014, 12, 750–764. [Google Scholar] [CrossRef] [Green Version]
- Margolis, D.M.; Archin, N.M.; Cohen, M.S.; Eron, J.J.; Ferrari, G.; Garcia, J.V.; Gay, C.L.; Goonetilleke, N.; Joseph, S.B.; Swanstrom, R.; et al. Curing HIV: Seeking to Target and Clear Persistent Infection. Cell 2020, 181, 189–206. [Google Scholar] [CrossRef]
- Bar, K.J.; Sneller, M.C.; Harrison, L.J.; Justement, J.S.; Overton, E.T.; Petrone, M.E.; Salantes, D.B.; Seamon, C.A.; Scheinfeld, B.; Kwan, R.W.; et al. Effect of HIV Antibody VRC01 on Viral Rebound after Treatment Interruption. N. Engl. J. Med. 2016, 375, 2037–2050. [Google Scholar] [CrossRef]
- Mendoza, P.; Gruell, H.; Nogueira, L.; Pai, J.A.; Butler, A.L.; Millard, K.; Lehmann, C.; Suárez, I.; Oliveira, T.Y.; Lorenzi, J.C.C.; et al. Combination therapy with anti-HIV-1 antibodies maintains viral suppression. Nature 2018, 561, 479–484. [Google Scholar] [CrossRef]
- Herzig, E.; Kim, K.C.; Packard, T.A.; Vardi, N.; Schwarzer, R.; Gramatica, A.; Deeks, S.G.; Williams, S.R.; Landgraf, K.; Killeen, N.; et al. Attacking Latent HIV with convertibleCAR-T Cells, a Highly Adaptable Killing Platform. Cell 2019, 179, 880–894.e10. [Google Scholar] [CrossRef] [PubMed]
- Kessing, C.F.; Nixon, C.C.; Li, C.; Tsai, P.; Takata, H.; Mousseau, G.; Ho, P.T.; Honeycutt, J.B.; Fallahi, M.; Trautmann, L.; et al. In Vivo Suppression of HIV Rebound by Didehydro-Cortistatin A, a “Block-and-Lock” Strategy for HIV-1 Treatment. Cell Rep 2017, 21, 600–611. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bosque, A.; Nilson, K.A.; Macedo, A.B.; Spivak, A.M.; Archin, N.M.; Van Wagoner, R.M.; Martins, L.J.; Novis, C.L.; Szaniawski, M.A.; Ireland, C.M.; et al. Benzotriazoles Reactivate Latent HIV-1 through Inactivation of STAT5 SUMOylation. Cell Rep. 2017, 18, 1324–1334. [Google Scholar] [CrossRef] [PubMed]
- Mousseau, G.; Clementz, M.A.; Bakeman, W.N.; Nagarsheth, N.; Cameron, M.; Shi, J.; Baran, P.; Fromentin, R.; Chomont, N.; Valente, S.T. An Analog of the Natural Steroidal Alkaloid Cortistatin A Potently Suppresses Tat-Dependent HIV Transcription. Cell Host Microbe 2012, 12, 97–108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mediouni, S.; Jablonski, J.; Paris, J.; Clementz, M.; Thenin-Houssier, S.; McLaughlin, J.; Valente, S. Didehydro-Cortistatin A Inhibits HIV-1 Tat Mediated Neuroinflammation and Prevents Potentiation of Cocaine Reward in Tat Transgenic Mice. Curr. HIV Res. 2015, 13, 64–79. [Google Scholar] [CrossRef] [Green Version]
- Marian, C.A.; Stoszko, M.; Wang, L.; Leighty, M.W.; de Crignis, E.; Maschinot, C.A.; Gatchalian, J.; Carter, B.C.; Chowdhury, B.; Hargreaves, D.C.; et al. Small Molecule Targeting of Specific BAF (mSWI/SNF) Complexes for HIV Latency Reversal. Cell Chem. Biol. 2018, 25, 1443–1455.e14. [Google Scholar] [CrossRef]
- Lindqvist, B.; Svensson Akusjärvi, S.; Sönnerborg, A.; Dimitriou, M.; Svensson, J.P. Chromatin maturation of the HIV-1 provirus in primary resting CD4+ T cells. PLoS Pathog. 2020, 16, e1008264. [Google Scholar] [CrossRef] [Green Version]
- Nixon, C.C.; Mavigner, M.; Sampey, G.C.; Brooks, A.D.; Spagnuolo, R.A.; Irlbeck, D.M.; Mattingly, C.; Ho, P.T.; Schoof, N.; Cammon, C.G.; et al. Systemic HIV and SIV latency reversal via non-canonical NF-κB signalling in vivo. Nature 2020, 578, 160–165. [Google Scholar] [CrossRef]
- McBrien, J.B.; Mavigner, M.; Franchitti, L.; Smith, S.A.; White, E.; Tharp, G.K.; Walum, H.; Busman-Sahay, K.; Aguilera-Sandoval, C.R.; Thayer, W.O.; et al. Robust and persistent reactivation of SIV and HIV by N-803 and depletion of CD8+ cells. Nature 2020, 578, 154–159. [Google Scholar] [CrossRef]
- Bialek, J.K.; Walther, T.; Hauber, J.; Lange, U.C. CRISPR-Cas9-based Genome Engineering to Generate Jurkat Reporter Models for HIV-1 Infection with Selected Proviral Integration Sites. J. Vis. Exp. 2018, 141, e58572. [Google Scholar] [CrossRef] [Green Version]
© 2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Shukla, A.; Ramirez, N.-G.P.; D’Orso, I. HIV-1 Proviral Transcription and Latency in the New Era. Viruses 2020, 12, 555. https://doi.org/10.3390/v12050555
Shukla A, Ramirez N-GP, D’Orso I. HIV-1 Proviral Transcription and Latency in the New Era. Viruses. 2020; 12(5):555. https://doi.org/10.3390/v12050555
Chicago/Turabian StyleShukla, Ashutosh, Nora-Guadalupe P. Ramirez, and Iván D’Orso. 2020. "HIV-1 Proviral Transcription and Latency in the New Era" Viruses 12, no. 5: 555. https://doi.org/10.3390/v12050555