Role of BRD4 and MLL1 in HIV transcription and reactivation

Pellaers Eline
Persbericht

De weg naar een block-and-lock genezingsstrategie van HIV

1. Noodzaak voor een genezing van HIV

De wereldgezondheidsorganisatie constateerde dat in 2020 37.7 miljoen mensen leefden met het humaan immunodeficiëntie virus (HIV) virus. Dit duidt aan dat de impact van HIV op de volksgezondheid nog steeds significant is. Echter, de vooruitgang in de behandeling van HIV resulteerde in een toegenomen levensverwachting van geïnfecteerde patiënten. Momenteel worden HIV-patiënten behandeld met combinatie antiretrovirale therapie, die de virale lading vermindert maar de ziekte niet geneest. De persistentie van geïntegreerd viraal DNA in een transcriptioneel stille modus in cellulaire reservoirs is de belangrijkste barrière voor genezing van HIV infectie. Mijn onderzoeksgroep focust op een block-and-lock genezingsstrategie met als doel een cellulair reservoir te genereren resistent tegen reactivatie na onderbreking van de behandeling. Zo werden LEDGINs ontwikkeld, kleine moleculen die de interactie tussen het viraal integrase en lens epithelium-derived-growth-factor (LEDGF/p75) inhiberen en zo de virale integratie inhiberen en het residuele provirus omleiden naar regio's resistent tegen reactivatie. Helaas zijn LEDGINs onvoldoende om de transcriptie van HIV volledig te blokkeren. Daarom is diepgaand onderzoek nodig naar andere co-factoren die belangrijk zijn in de regulatie van HIV expressie, zoals bromodomain-containing-protein 4 (BRD4) en mixed-lineage-leukemia-1 (MLL1), om het potentieel voor een block-and-lock genezingsstrategie te verhogen. De branched-DNA (bDNA) techniek laat toe om het het viraal DNA en RNA niveau per cel te analyseren. Bijgevolg is deze techniek een elegante methode om het effect van een inhibitor op de transcriptionele toestand van het provirus te bestuderen, om zo nieuwe doelwitten te identificeren voor een block-and-lock genezingsstrategie.

2. De rol van BRD4 en enhancers in residuele transcriptie van HIV

2.1. JQ1 promoot basale transcriptie en reactivatie van HIV

JQ1 is een bekende BRD4 remmer, gebruikt in de shock-and-kill strategie, die gericht is op het uitroeien van het latente reservoir na reactivatie. Verschillende studies toonden aan dat JQ1 de HIV replicatie stimuleert. In deze studie toonde de bDNA techniek dat JQ1 de basale transcriptie en de tumor necrosis factor-α (TNF-α) geïnduceerde reactivatie van HIV bevorderde op ‘single cell’ niveau met een optimum bij 1 μM JQ1. Deze optimale concentratie van JQ1 accentueert een complexe rol van BRD4 in de transcriptionele regulatie van HIV.

Figuur 1

Figuur 1. Het aantal vRNA spots per geïnfecteerde cel, na de bDNA analyse, zijn geplot voor telkens 72 cellen voor elke conditie van de niet gereactiveerde (oranje spots) en gereactiveerde (blauwe spots) cellen. Elke stip representeert het aantal vRNA spots per single cell en de balk representeert de mediaan. Statistische significantie werd berekend met een Krushkall-Wallis test (ns=non-significant, *p<0.05, **p<0.01, ****p<0.0001) tussen de niet gereactiveerde en de geractiveerde cellen en tussen de cellen behandeld met JQ1 in vergelijking met de controle (0 µM JQ1).

2.2. ZL0580 heeft geen effect op de replicatie van HIV

Niu et al. rapporteerde, naast het gebruik van BRD4-inhibitors in de shock-and-kill strategie, onlangs de eerste BRD4-inhibitor die HIV replicatie onderdrukt, ZL0580. Helaas belemmerde de hoge toxiciteit en het gebrek aan effect op HIV expressie in deze studie het potentieel van ZL0580 in een block-and-lock genezingsstrategie.  

Figuur 2

Figuur 2. Het aantal vRNA spots per geïnfecteerde cel, na de bDNA analyse, zijn geplot voor telkens 73 cellen voor elke conditie van de niet gereactiveerde (oranje spots) en gereactiveerde (blauwe spots) cellen. Elke stip representeert het aantal vRNA spots per single cell en de balk representeert de mediaan. Statistische significantie werd berekend met een Krushkall-Wallis test (ns=non-significant, *p<0.05) tussen de niet gereactiveerde en de geractiveerde cellen en tussen de cellen behandeld met ZL0580 in vergelijking met de controle (0 µM ZL0580).

2.3. JQ1 interefereert met de reactivatie van LEDGIN-geheroriënteerd provirus

LEDGINs inhiberen de HIV transcriptie maar residuele vRNA expressie blijft aanwezig. Het is het eerder aangetoond dat LEDGINs geen invloed hebben op integratie in de nabijheid van enhancer-regio’s, DNA elementen die transcriptie versterken. Omdat deze enhancer-regio’s wel afhankelijk zijn van BRD4, werd onderzocht of JQ1-gemedieerde inhibitie van BRD4 de residuele vRNA expressie na LEDGIN-gemedieerde retargeting zou inhiberen. In tegenstelling, JQ1 had geen inhiberend effect op residuele HIV replicatie. Interessant is dat deze studie wel aantoonde dat JQ1 de TNF-α geïnduceerde reactivatie van LEDGIN-geheroriënteerd provirus antagoniseert. Desondanks stimuleert JQ1 nog steeds de TNF-α geïnduceerde reactivatie van de cellen die niet behandeld zijn met LEDGINs of met een lage concentratie van LEDGINs met een optimum concentratie van JQ1. Het blijft onduidelijk waarom JQ1 de TNF-α geïnduceerde reactivatie van LEDGIN-geheroriënteerd provirus verhindert en verder onderzoek is nodig om dit te verklaren.

Figuur 3

Figuur 3. De relative fold reactivation geïnduceerd door TNF-α (totaal aantal vRNA spots van TNF-α gereactiveerde cellen/ totaal aantal vRNA spots van niet gereactiveerde cellen) is gerepresenteerd voor cellen behandeld met verschillende concentraties LEDGIN en JQ1. De concentratie LEDGIN is aangeduid met verschillende kleuren, terwijl de concentratie JQ1 gedemonstreerd staat in de x-as.

3. MLL1 depletie inhibeert HIV replicatie

Tenslotte rapporteerden Gao et al. een rol van MLL1 in reactivatie van HIV. Daarom werd het effect van MLL1 depletie op HIV replicatie verder onderzocht. Hiervoor werden infectie-experimenten opgezet waar de HIV replicatie vergeleken werd tussen cellen getransduceerd met lentivirale vectoren en cellen met een depletie voor MLL1 door middel van specifieke miRNA-gebaseerde lentivirale vectoren. De resultaten bewezen dat MLL1 depletie sterk de HIV-infectiviteit vermindert. Er blijven echter verschillende vragen over het moleculaire mechanisme achter deze bevindingen en verder onderzoek is nodig om MLL1 als een doelwit in een block-and-lock genezingsstrategie te identificeren.

Figuur 4

Figuur 4. De relatieve luciferase counts, genormaliseerd voor de proteïne expressie (bepaald met de BCA assay) zijn geplot voor het wildtype en de cellen met een MLL1 depletie voor verschillende virus diluties. Elke kleur geeft een verschillende virus dilutie weer en de balken representeren de standaarddeviatie bepaald via technische duplicaten.

4. Conclusie

Dit onderzoek beklemtoont dat BRD4 en MLL1 cruciale determinanten zijn van de virale transcriptie van HIV. Een aanhoudende inspanning in academisch onderzoek naar de rol van deze co-factoren in de transcriptionele regulatie van HIV kan deuren openen naar een block-and-lock genezingsstrategie voor HIV.

Bibliografie

 

  1. Weber J. The pathogenesis of HIV-1 infection. Br Med Bull. 2001;58:61–72.

  2. Fanales-Belasio E, Raimondo M, Suligoi B, Buttò S. HIV virology and pathogenetic mechanisms of infection: a brief overview Emanuele. 2011;46(1):5–14.

  3. Maartens G, Celum C, Lewin SR. HIV infection: Epidemiology, pathogenesis, treatment, and prevention. Lancet [Internet]. 2014;384(9939):258–71. Available from: http://dx.doi.org/10.1016/S0140-6736(14)60164-1

  4. nd Kelsey C. MartinMhatre V. Ho, Ji-Ann Lee A. Conformational Changes in HIV-1 gp41 in the Course of HIV-1 Envelope Glycoprotein-Mediated Fusion and Inactivation†. Bone [Internet]. 2012;23(1):1–7. Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3624763/pdf/nihms412728.pdf

  5. Atta MG, De Seigneux S, Lucas GM. Clinical pharmacology in HIV therapy. Clin J Am Soc Nephrol. 2019;14(3):435–44.

  6. Ferguson MR, Rojo DR, Von Lindern JJ, O’Brien WA. HIV-1 replication cycle. Clin Lab Med. 2002;22(3):611–35.

  7. Arhel N. Revisiting HIV-1 uncoating. 2010;25–8.

  8. Craigie R, Bushman FD. HIV DNA integration. Cold Spring Harb Perspect Med. 2012;2(7):1–18.

  9. Freed E. HIV-1 assembly, release and maturation Eric. Nat Rev Microbiol. 2015;13(8):484–96.

  10. Sundquist WI, Kräusslich H. HIV-1 Assembly, Budding, and Maturation. Cold Spring HarbPerspect Med. 2012;2:1–24.

  11. Deeks SG, Lewin SR, Ross AL, Ananworanich J, Benkirane M, Cannon P, et al. International AIDS Society global scientific strategy: Towards an HIV cure 2016. Nat Med. 2016;22(8):839–50.

  12. Chun T, Carruth L, Finzi D, Shen X, Digiuseppet JA, Taylor H, et al. Quantification of latent tissue reservoirs and total body viral load in HIV-1 infection. 1997;141(1995):80–3.

  13. Castro-Gonzalez S, Colomer-Lluch M, Serra-Moreno R. Barriers for HIV Cure: The Latent Reservoir. AIDS Res Hum Retroviruses. 2018;34(9):739–59.

  14. Debyser Z, Vansant G, Bruggemans A, Janssens J, Christ F. Insight in HIV integration site selection provides a block-and-lock strategy for a functional cure of HIV infection. Viruses. 2019;11(1):1–12.

  15. Finzi D, Blankson J, Siliciano JD, Margolick JB, Chadwick K. Latent infection of CD4+ T cells provides a mechanism for lifelong persistence of HIV-1, even in patients on effective combination therapy. 1999;5(5):512–7.

  16. García M, Buzón MJ, Benito JM, Rallón N. Peering into the HIV reservoir. Rev Med Virol. 2018;28(4):1–13.

  17. Churchill MJ, Deeks SG, Margolis DM, Siliciano RF. HIV reservoirs : what , where and how to target them. Nat Publ Gr. 2016;14(January):55–60.

  18. Chomont N, El-Far M, Ancuta P, Trautmann L, A Procopio F, Yassine-Diab B, et al. HIV reservoir size and persistence are driven by T cell survival and homeostatic proliferation. Nat Med. 2009;15(8):893–900.

  19. Maldarelli F, Wu X, Su L, Simonetti FR, Shao W, Hill S, et al. Specific HIV integration sites are linked to clonal expansion and persistence of infected cells. Science (80). 2014;345(6193):179–83.

  20. Lorenzo-Redondo R, Fryer HR, Bedford T, Kim EY, Archer J, Kosakovsky Pond SL, et al. Persistent HIV-1 replication maintains the tissue reservoir during therapy. Nature. 2016;530(7588):51–6.

  21. Fletcher C V., Staskus K, Wietgrefe SW, Rothenberger M, Reilly C, Chipman JG, et al. Persistent HIV-1 replication is associated with lower antiretroviral drug concentrations in lymphatic tissues. Proc Natl Acad Sci U S A. 2014;111(6):2307–12.

  22. van Zyl G, Bale MJ, Kearney MF. HIV evolution and diversity in ART-treated patients. Retrovirology [Internet]. 2018;15(1):1–12. Available from: https://doi.org/10.1186/s12977- 018-0395-4

  23. Symons J, Cameron PU, Lewin SR, Hospital M, Hospital A. HIV integration sites and implications for maintenance of the reservoir. 2019;13(2):152–9.

  24. Janssens J, Bruggemans A, Christ F, Debyser Z. Towards a Functional Cure of HIV-1: Insight Into the Chromatin Landscape of the Provirus. Front Microbiol. 2021;12(March).

  25. Lusic M, Siliciano RF. Nuclear landscape of HIV-1 infection and integration. Nat Rev Microbiol. 2017;15(2):69–82.

  26. Debyser Z, Christ F, De Rijck J, Gijsbers R. Host factors for retroviral integration site selection. Trends Biochem Sci [Internet]. 2015;40(2):108–16. Available from: http://dx.doi.org/10.1016/j.tibs.2014.12.001

  27. 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(7):1726–38.

  28. Engelman AN, Singh PK. Cellular and molecular mechanisms of HIV-1 integration targeting. Cell Mol Life Sci [Internet]. 2018;75(14):2491–507. Available from: https://doi.org/10.1007/s00018-018-2772-5

  29. Jiang C, Lian X, Gao C, Sun X, Einkauf KB, Chevalier JM, et al. Distinct viral reservoirs in individuals with spontaneous control of HIV-1. Nature [Internet]. 2020;585:261–7. Available from: http://www.ncbi.nlm.nih.gov/pubmed/32848246

  30. Gonzalo-Gil E, Ikediobi U, Sutton RE. Mechanisms of virologic control and clinical characteristics of HIV+ elite/viremic controllers. Yale J Biol Med. 2017;90(2):245–59.

  31. Lobritz MA, Lassen KG, Arts EJ. HIV-1 replicative fitness in elite controllers Michael. Curr Opin HIV AIDS. 2011;6(3):214–20.

  32. Miura T, Brockman MA, Brumme CJ, Brumme ZL, Carlson JM, Pereyra F, et al. Genetic Characterization of Human Immunodeficiency Virus Type 1 in Elite Controllers: Lack of Gross Genetic Defects or Common Amino Acid Changes. J Virol. 2008;82(17):8422–30.

  33. Cutter A, J. H. A Brief Review of Nucleosome Structure. FEBS Lett. 2016;589(2000):2914– 22.

  34. Marini B, Kertesz-Farkas A, Ali H, Lucic B, Lisek K, Manganaro L, et al. Nuclear architecture dictates HIV-1 integration site selection. Nature. 2015;521(7551):227–31.

  35. An Integrated Overview of HIV-1 Latency. 2013;155(3):519–29.

  36. Dahabieh MS, Battivelli E, Verdin E. Understanding HIV latency: The road to an HIV cure. Annu Rev Med. 2015;66:407–21.

  37. Verdin E. Transcriptional activation and chromatin remodeling of the HIV-I promoter in response to histone acetylation. Chemtracts. 1997;10(10):773–8.

  38. Bednarik DP, Cook JA, Pitha PM. Inactivation of the HIV LTR by DNA CpG methylation: Evidence for a role in latency. EMBO J. 1990;9(4):1157–64.

  39. Blokken J, De Rijck J, Christ F, Debyser Z. Protein–protein and protein–chromatin interactions,of LEDGF/p75 as novel drug targets. Drug Discov Today Technol [Internet]. 2017;24:25–31. Available from: http://dx.doi.org/10.1016/j.ddtec.2017.11.002

  40. Cherepanov P. LEDGF/p75 interacts with divergent lentiviral integrases and modulates their enzymatic activity in vitro. Nucleic Acids Res. 2007;35(1):113–24.

  41. Cherepanov P, Devroe E, Silver PA, Engelman A. Identification of an evolutionarily conserved domain in human lens epithelium-derived growth factor/transcriptional co-activator p75 (LEDGF/p75) that binds HIV-1 integrase. J Biol Chem. 2004;279(47):48883–92.

  42. Tesina P, Cermáková K, Horejší M, Procházková K, Fábry M, Sharma S, et al. Multiple cellular proteins interact with LEDGF/p75 through a conserved unstructured consensus motif. Nat Commun. 2015;6.

  43. Christ F, Debyser Z. The LEDGF/p75 integrase interaction, a novel target for anti-HIV therapy. Virology [Internet]. 2013;435(1):102–9. Available from: http://dx.doi.org/10.1016/j.virol.2012.09.033

  44. Bartholomeeusen K, De Rijck J, Busschots K, Desender L, Gijsbers R, Emiliani S, et al. Differential Interaction of HIV-1 Integrase and JPO2 with the C Terminus of LEDGF/p75. J Mol Biol. 2007;372(2):407–21.

  45. Maertens GN, Cherepanov P, Engelman A. Transcriptional co-activator p75 binds and tethers the Myc-interacting protein JPO2 to chromatin. J Cell Sci. 2006;119(12):2563–71.

  46. Yokoyama A, Cleary M. Menin critically links MLL proteins with LEDGF on cancer- associated target genes Akihiko. 2008;14(1):36–46.

  47. Sharma S, Cermáková K, De Rijck J, Demeulemeester J, Fábry M, Ashkar S El, et al. Affinity switching of the LEDGF/p75 IBD interactome is governed by kinase-dependent phosphorylation. Proc Natl Acad Sci U S A. 2018;115(30):E7053–62.

  48. Gérard A, Ségéral E, Naughtin M, Abdouni A, Charmeteau B, Cheynier R, et al. The Integrase Cofactor LEDGF/p75 Associates with Iws1 and Spt6 for Postintegration Silencing of HIV-1 Gene Expression in Latently Infected Cells. Cell Host Microbe. 2015;17(1):107–17.

  49. Wang H, Jurado KA, Wu X, Shun MC, Li X, Ferris AL, et al. HRP2 determines the efficiency and specificity of HIV-1 integration in LEDGF/p75 knockout cells but does not contribute to the antiviral activity of a potent LEDGF/p75-binding site integrase inhibitor. Nucleic Acids Res. 2012;40(22):11518–30.

  50. Engelman A, Cherepanov P. The lentiviral integrase binding protein LEDGF/p75 and HIV-1 replication. PLoS Pathog. 2008;4(3).

  51. Cherepanov P, Maertens G, Proost P, Devreese B, Van Beeumen J, Engelborghs Y, et al. HIV- 1 integrase forms stable tetramers and associates with LEDGF/p75 protein in human cells. J Biol Chem. 2003;278(1):372–81.

  52. Llano M, Delgado S, Vanegas M, Poeschla EM. Lens epithelium-derived growth factor/p75 prevents proteasomal degradation of HIV-1 integrase. J Biol Chem. 2004;279(53):55570–7.

  53. Turlure F, Maertens G, Rahman S, Cherepanov P, Engelman A. A tripartite DNA-binding element, comprised of the nuclear localization signal and two AT-hook motifs, mediates the association of LEDGF/p75 with chromatin in vivo. Nucleic Acids Res. 2006;34(5):1653–65.

  54. Vandekerckhove L, Christ F, Van Maele B, De Rijck J, Gijsbers R, Van den Haute C, et al. Transient and Stable Knockdown of the Integrase Cofactor LEDGF/p75 Reveals Its Role in the Replication Cycle of Human Immunodeficiency Virus. J Virol. 2006;80(4):1886–96.

  55. De Rijck J, Vandekerckhove L, Gijsbers R, Hombrouck A, Hendrix J, Vercammen J, et al. Overexpression of the Lens Epithelium-Derived Growth Factor/p75 Integrase Binding Domain Inhibits Human Immunodeficiency Virus Replication. J Virol. 2006;80(23):11498–509.

  56. Christ F, Voet A, Marchand A, Nicolet S, Desimmie BA, Marchand D, et al. Rational design of small-molecule inhibitors of the LEDGF/p75-integrase interaction and HIV replication. Nat Chem Biol. 2010 Jun;6(6):442–8.

  57. Vansant G, Bruggemans A, Janssens J, Debyser Z. Block-and-lock strategies to cure HIV infection. Viruses. 2020;12(1):1–17.

  58. Desimmie BA, Schrijvers R, Demeulemeester J, Borrenberghs D, Weydert C, Thys W, et al. LEDGINs inhibit late stage HIV-1 replication by modulating integrase multimerization in the virions. Retrovirology. 2020;17(1):1–16.

  59. Christ F, Shaw S, Demeulemeester J, Desimmie BA, Marchan A, Butler S, et al. Small- molecule inhibitors of the LEDGF/p75 binding site of integrase block HIV replication and modulate integrase multimerization. Antimicrob Agents Chemother. 2012;56(8):4365–74.

  60. Kessl JJ, Li M, Ignatov M, Shkriabai N, Eidahl JO, Feng L, et al. FRET analysis reveals distinct conformations of in tetramers in the presence of viral DNA or LEDGF/p75. Nucleic Acids Res. 2011;39(20):9009–22.

  61. Vranckx LS, Demeulemeester J, Saleh S, Boll A, Vansant G, Schrijvers R, et al. LEDGIN- mediated Inhibition of Integrase-LEDGF/p75 Interaction Reduces Reactivation of Residual Latent HIV. EBioMedicine [Internet]. 2016;8:248–64. Available from: http://dx.doi.org/10.1016/j.ebiom.2016.04.039

  62. Vansant G, Chen HC, Zorita E, Trejbalová K, Miklík D, Filion G, et al. The chromatin landscape at the HIV-1 provirus integration site determines viral expression. Nucleic Acids Res. 2020;48(14):7801–17.

  63. Lori M. Millner, Mark W. Linder and RVJ. Direct Visualization of HIV-1 Replication Intermediates shows that Viral Capsid and CPSF6 Modulate HIV-1 Intra-nuclear Invasion and Integration. cell rep. 2015;13(8):1717–31.

  64. Achuthan V, Perreira JM, Sowd GA, Puray-chavez M, Mcdougall WM, Paulucci-holthauzen A, et al. Capsid-CPSF6 interaction licenses nuclear HIV-1 trafficking to sites of viral DNA integration Vasudevan. 2019;24(3):392–404.

  65. Rasheedi S, Shun MC, Serrao E, Sowd GA, Qian J, Hao C, et al. The Cleavage and polyadenylation specificity factor 6 (CPSF6) subunit of the capsid-recruited pre-messenger RNA cleavage factor I (CFIm) complex mediates HIV-1 integration into genes. J Biol Chem. 2016;291(22):11809–19.

  66. Sowd GA, Serrao E, Wang H, Wang W, Fadel HJ, Poeschla EM, et al. A critical role for alternative polyadenylation factor CPSF6 in targeting HIV-1 integration to transcriptionally active chromatin. Proc Natl Acad Sci U S A. 2016;113(8):E1054–63.

  67. Schaller T, Ocwieja KE, Rasaiyaah J, Price AJ, Brady TL, Roth SL, et al. HIV-1 capsid- cyclophilin interactions determine nuclear import pathway, integration targeting and replication efficiency. PLoS Pathog. 2011;7(12).

  68. Lee KE, Ambrose Z, Martin TD, Oztop I, Mulky A, Julias JG, et al. Flexible Use of Nuclear Import Pathways by HIV-1. Cell Host Microbe [Internet]. 2010;7(3):221–33. Available from: http://dx.doi.org/10.1016/j.chom.2010.02.007

  69. Fricke T, Valle-Casuso JC, White TE, Brandariz-Nuñez A, Bosche WJ, Reszka N, et al. The ability of TNPO3-depleted cells to inhibit HIV-1 infection requires CPSF6. Retrovirology. 2013;10(1):1–14.

  70. World Health Organization (WHO): HIV/AIDS.(2020). Retrieved November 14 2020, from: https://www.who.int/health-topics/hiv-aids#tab=tab_1.

  71. Volberding PA, Deeks SG. Antiretroviral therapy and management of HIV infection. Lancet [Internet]. 2010;376(9734):49–62. Available from: http://dx.doi.org/10.1016/S0140- 6736(10)60676-9

  72. De Silva Feelixge HS, Stone D, Pietz HL, Roychoudhury P, Greninger AL, Schiffer JT, et al. Detection of treatment-resistant infectious HIV after genome-directed antiviral endonuclease therapy. Antiviral Res [Internet]. 2016;126:90–8. Available from: http://dx.doi.org/10.1016/j.antiviral.2015.12.007

  73. Harrigan PR, Hogg RS, Dong WWY, Yip B, Wynhoven B, Woodward J, et al. Predictors of HIV drug-resistance mutations in a large antiretroviral-naive cohort initiating triple antiretroviral therapy. J Infect Dis. 2005;191(3):339–47.

  74. Meresse M, March L, Kouanfack C, Bonono RC, Boyer S, Laborde-Balen G, et al. Patterns of adherence to antiretroviral therapy and HIV drug resistance over time in the Stratall ANRS 12110/ESTHER trial in Cameroon. HIV Med. 2014;15(8):478–87.

  75. H ADC, Chevalier P. HOSPITAL ECONOMIC BURDEN OF HIV AND ITS COMPLICATIONS IN BELGIUM. 5th Symp Belgian Res AIDS HIV Consort. 2016;(2014):2016.

  76. Church JA. Evidence for the cure of HIV infection by CCR5δ32/Δ32 stem cell transplantation. Pediatrics. 2011;128(SUPPL. 3):2791–9.

  77. Hütter G, Nowak D, Mossner M, Ganepola S, Müßig A, Allers K, et al. Long-Term Control of HIV by CCR5 Delta32/ Delta32 Stem-Cell Transplantation. new Engl J Med Br [Internet]. 2009;360(7):692–7. Available from: http://www.nejm.org/doi/abs/10.1056/NEJMoa0802905

  78. Bassett E, Clark RF. Shift of HIV Tropism in Stem-Cell Transplantation with CCR5 Delta32 Mutation. N Engl J Med. 2014;371(9):880–2.

  79. Henrich TJ, Hu Z, Li JZ, Sciaranghella G, Busch MP, Keating SM, et al. Long-term reduction in peripheral blood HIV type 1 reservoirs following reduced-intensity conditioning allogeneic stem cell transplantation. J Infect Dis. 2013;207(11):1694–702.

  80. Gupta RK, Abdul-Jawad S, McCoy LE, Mok HP, Peppa D, Salgado M, et al. HIV-1 remission following CCR5Δ32/Δ32 haematopoietic stem-cell transplantation. Nature [Internet]. 2019;568(7751):244–8. Available from: http://dx.doi.org/10.1038/s41586-019-1027-4

  81. Liu Z, Chen S, Jin X, Wang Q, Yang K, Li C, et al. Genome editing of the HIV co-receptors CCR5 and CXCR4 by CRISPR-Cas9 protects CD4+ T cells from HIV-1 infection. Cell Biosci. 2017;7(1):1–15.

  82. Allen AG, Chung C-H, Atkins A, Dampier W, Khalili K, Nonnemacher MR, et al. Gene Editing of HIV-1 Co-receptors to Prevent and/or Cure Virus Infection. Front Microbiol. 2018;9(December):1–14.

  83. Peterson C.W., Kiem HP. (2017) Cell and Gene Therapy for HIV Cure. In: Silvestri G., Lichterfeld M. (Eds) HIV-1 Latency. Current Topics in Microbiology and Immunology, vol 417. Springer, Cham. pp211-248.

  84. Burton DR, Hangartner L. Broadly Neutralizing Antibodies to HIV and Their Role in Vaccine Design. Annu Rev Immunol. 2016;34:635–59.

  85. Haynes BF, Burton DR, Mascola JR. Multiple roles for HIV broadly neutralizing antibodies. Sci Transl Med. 2019;11(516):1–4.

  86. Ollerton MT, Berger EA, Connick E, Burton GF. HIV-1-Specific Chimeric Antigen Receptor T Cells Fail To Recognize and Eliminate the Follicular Dendritic Cell HIV Reservoir In Vitro. J Virol. 2020;94(10):1–12.

  87. Wagner TA. Quarter Century of Anti-HIV CAR T Cells. Curr HIV/AIDS Rep. 2018;15(2):147–54.

  88. Qi J, Ding C, Jiang X, Gao Y. Advances in Developing CAR T-Cell Therapy for HIV Cure. Front Immunol. 2020;11(March):1–13.

  89. Abner E, Jordan A. HIV “shock and kill” therapy: In need of revision. Antiviral Res. 2019;166(November 2018):19–34.

  90. Davenport MP, Khoury DS, Cromer D, Lewin SR, Kelleher AD, Kent SJ. Functional cure of HIV: the scale of the challenge. Nat Rev Immunol [Internet]. 2019;19(1):45–54. Available from: http://dx.doi.org/10.1038/s41577-018-0085-4

  91. Mediouni S, Chinthalapudi K, Ekka MK, Usui I, Jablonski JA, Clementz MA, et al. Didehydro-cortistatin a inhibits HIV-1 by specifically binding to the unstructured basic region of tat. MBio. 2019;10(1):1–19.

  92. Jean MJ, Hayashi T, Huang H, Brennan J, Simpson S, Purmal A, et al. Curaxin CBL0100 blocks HIV-1 replication and reactivation through inhibition of viral transcriptional elongation. Front Microbiol. 2017;8(OCT):1–13.

  93. Trivedi J, Parveen A, Rozy F, Mitra A, Bal C, Mitra D, et al. Discovery of 2-isoxazol-3-yl- acetamide analogues as heat shock protein 90 (HSP90) inhibitors with significant anti-HIV activity. Eur J Med Chem [Internet]. 2019;183:111699. Available from: https://doi.org/10.1016/j.ejmech.2019.111699

  94. Govers G, Poesen J, Goossens D. Novel mechanisms to inhibit HIV reservoir seeding using Jak inhibitors Christina. Manag soil Qual challenges Mod Agric. 2009;199–217.

  95. Taylor HE, Calantone N, Lichon D, Hudson H, Clerc I, Campbell EM, et al. mTOR Overcomes Multiple Metabolic Restrictions to Enable HIV-1 Reverse Transcription and Intracellular Transport. Cell Rep [Internet]. 2020;31(12):107810. Available from: https://doi.org/10.1016/j.celrep.2020.107810

  96. Vansant G, Vranckx LS, Zurnic I, Van Looveren D, Van De Velde P, Nobles C, et al. Impact of LEDGIN treatment during virus production on residual HIV-1 transcription. Retrovirology [Internet]. 2019;16(1):1–17. Available from: https://doi.org/10.1186/s12977-019-0472-3

  97. Wu M, Shen J. From Super-Enhancer Non-coding RNA to Immune Checkpoint: Frameworks to Functions. Front Oncol. 2019;9(November):1–16.

  98. Andersson R, Sandelin A. Determinants of enhancer and promoter activities of regulatory elements. Nat Rev Genet [Internet]. 2020;21(2):71–87. Available from: http://dx.doi.org/10.1038/s41576-019-0173-8

  99. Lucic B, Chen HC, Kuzman M, Zorita E, Wegner J, Minneker V, et al. Spatially clustered loci with multiple enhancers are frequent targets of HIV-1 integration. Nat Commun [Internet]. 2019;10(1):1–12. Available from: http://dx.doi.org/10.1038/s41467-019-12046-3

  100. Chen HC, Martinez JP, Zorita E, Meyerhans A, Filion GJ. Position effects influence HIV latency reversal. Nat Struct Mol Biol. 2017;24(1):47–54.

  101. Taniguchi Y. The bromodomain and extra-terminal domain (BET) family: Functional anatomy of BET paralogous proteins. Int J Mol Sci. 2016;17(11):1–24.

  102. Rahman S, Sowa ME, Ottinger M, Smith JA, Shi Y, Harper JW, et al. The Brd4 Extraterminal Domain Confers Transcription Activation Independent of pTEFb by Recruiting Multiple Proteins, Including NSD3. Mol Cell Biol. 2011;31(13):2641–52.

  103. Li Z, Guo J, Wu Y, Zhou Q. The BET bromodomain inhibitor JQ1 activates HIV latency through antagonizing Brd4 inhibition of Tat-transactivation. Nucleic Acids Res. 2013;41(1):277–87.

  104. Lovén J, Hoke HA, Lin CY, Lau A, Orlando DA, Vakoc CR, et al. Selective Inhibition of Tumor Oncogenes by Disruption of Super- Enhancers. Cell. 2013;153(2):320–34.

  105. Liu Z, Wang P, Chen H, Wold EA, Tian B, Brasier AR, et al. Drug Discovery Targeting Bromodomain-Containing Protein 4. J Med Chem. 2017;60(11):4533–58.

  106. Niu Q, Liu Z, Alamer E, Fan X, Chen H, Endsley J, et al. Structure-guided drug design identifies a BRD4-selective small molecule that suppresses HIV. J Clin Invest. 2019;129(8):3361–73.

  107. Bartholomeeusen K, Xiang Y, Fujinaga K, Peterlin BM. Bromodomain and extra-terminal (BET) bromodomain inhibition activate transcription via transient release of Positive Transcription Elongation Factor b (P-TEFb) from 7SK small nuclear ribonucleoprotein. J Biol Chem. 2012;287(43):36609–16.

  108. Li Z, Mbonye U, Feng Z, Wang X, Gao X, Karn J, et al. The KAT5-Acetyl-Histone4-Brd4 axis silences HIV-1 transcription and promotes viral latency. PLoS Pathog. 2018;14(4):1–24.

  109. Liedtke M, Cleary ML. Therapeutic targeting of MLL. Blood. 2009;113(24):6061–8.

  110. Ciuffi A, Llano M, Poeschla E, Hoffmann C, Leipzig J, Shinn P, et al. A role for LEDGF/p75in targeting HIV DNA integration. Nat Med. 2005;11(12):1287–9.

  111. Gao R, Bao J, Yan H, Xie L, Qin W, Ning H, et al. Competition between PAF1 and MLL1/COMPASS confers the opposing function of LEDGF/p75 in HIV latency and proviral reactivation. Sci Adv. 2020;6.

  112. Zeng Y, Wagner EJ, Cullen BR. Both natural and designed micro RNAs can inhibit the expression of cognate mRNAs when expressed in human cells. Mol Cell. 2002;9(6):1327–33.

  113. Reichelt WN, Waldschitz D, Herwig C, Neutsch L. Bioprocess monitoring: minimizing sample matrix effects for total protein quantification with bicinchoninic acid assay. J Ind Microbiol Biotechnol. 2016;43(9):1271–80.

  114. Pannecouque C, Daelemans D, De Clercq E. Tetrazolium-based colorimetric assay for the detection of HIV replication inhibitors: Revisited 20 years later. Nat Protoc. 2008;3(3):427–34.

  115. Puray-Chavez M, Tedbury PR, Huber AD, Ukah OB, Yapo V, Liu D, et al. Multiplex single- cell visualization of nucleic acids and protein during HIV infection. Nat Commun. 2017;8(1):1–11.

  116. Zhu J, Gaiha GD, John SP, Pertel T, Chin CR, Gao G, et al. Reactivation of Latent HIV-1 by Inhibition of BRD4. Cell Rep. 2012;2(4):807–16.

  117. Boehm D, Calvanese V, Dar RD, Xing S, Schroeder S, Martins L, et al. BET bromodomain-targeting compounds reactivate HIV from latency via a Tat-independent mechanism. Cell Cycle. 2013;12(3):452–62.

  118. Bisgrove DA, Mahmoudi T, Henklein P, Verdin E. Conserved P-TEFb-interacting domain of BRD4 inhibits HIV transcription. Proc Natl Acad Sci U S A. 2007;104(34):13690–5.

  119. McNamara LA, Ganesh JA, Collins KL. Latent HIV-1 Infection Occurs in Multiple Subsets of Hematopoietic Progenitor Cells and Is Reversed by NF- B Activation. J Virol. 2012;86(17):9337–50.

  120. Necrosis T, Fetal H, Cells G, Tornatore CS, Traub R, Drew PD, et al. Stimulation of HIV type 1 gene expression and induction of NF-kappa B (p50/p65)-binding activity in tumor necrosis factor alpha-treated human fetal glial cells. 1994;10(10):1207–11.

  121. Alamer E, Zhong C, Hajnik R, Soong L, Hu H. Modulation of BRD4 in HIV epigenetic regulation: implications for finding an HIV cure. Retrovirology [Internet]. 2021;18(1):1–9. Available from: https://doi.org/10.1186/s12977-020-00547-9

  122. Ott M, Geyer M, Zhou Q. The control of HIV transcription: Keeping RNA polymerase II on track. Cell Host Microbe. 2011;10(5):426–35.

  123. Yang Z, Yik JHN, Chen R, He N, Moon KJ, Ozato K, et al. Recruitment of P-TEFb for stimulation of transcriptional elongation by the bromodomain protein Brd4. Mol Cell. 2005;19(4):535–45.

  124. Moon KJ, Mochizuki K, Zhou M, Jeong HS, Brady JN, 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(4):523–34.

  125. Ai N, Hu X, Ding F, Yu B, Wang H, Lu X, et al. Signal-induced Brd4 release from chromatin is essential for its role transition from chromatin targeting to transcriptional regulation. Nucleic Acids Res. 2011;39(22):9592–604.

  126. Zou Z, Huang B, Wu X, Zhang H, Qi J, Bradner J, et al. Brd4 maintains constitutively active NF-κB in cancer cells by binding to acetylated RelA. Oncogene. 2014;33(18):2395–404.

  127. Klein K. Bromodomain protein inhibition: A novel therapeutic strategy in rheumatic diseases. RMD Open. 2018;4(2):1–10.

  128. Brown JD, Lin CY, Duan Q, Griffin G, Paranal RM, Bair S, et al. NF-kB directs dynamic super enhancer formation in inflammation and atherogenesis Jonathan. Mol Cell. 2014;56(2):219–31.

  129. Karn J. The molecular biology of HIV latency: Breaking and restoring the Tat-dependent transcriptional circuit. Curr Opin HIV AIDS. 2011;6(1):4–11.

  130. Abraham RT, Weiss A. Jurkat T cells and development of the T-cell receptor signalling paradigm 01 2017/5/1. 2017;4(April 2004):1–8.

  131. Roderick JE, Tesell J, Shultz LD, Brehm MA, Greiner DL, Harris MH, et al. c-Myc inhibition prevents leukemia initiation in mice and impairs the growth of relapsed and induction failure pediatric T-ALL cells. Blood. 2014;123(7):1040–50.

  132. Follini E, Marchesini M, Roti G. Strategies to overcome resistance mechanisms in t-cell acute lymphoblastic leukemia. Int J Mol Sci. 2019;20(12):1–28.

  133. Knoechel B, Roderick JE, Williamson KE, Zhu J, Lohr JG, Cotton MJ, et al. An epigenetic mechanism of resistance to targeted therapy in T cell acute lymphoblastic leukemia. Nat Genet. 2014;46(4):364–70.

  134. Loosveld M, Castellano R, Gon S, Goubard A, Crouzet T, Pouyet L, et al. Therapeutic Targeting of c-Myc in T-Cell Acute Lymphoblastic Leukemia (T-ALL). Oncotarget. 2014;5(10):3168–72.

  135. Chung C-H, Mele A, Allen A, Costello R, Dampier W, Nonnemacher Mi, et al. Integrated Human Immunodeficiency Virus Type 1 Sequence in J-Lat 10.6. Am Soc Microbiol. 2014;9(18):1–3.

  136. Creyghton MP, Cheng AW, Welstead GG, Kooistra T, Carey BW, Steine EJ, et al. Histone H3K27ac separates active from poised enhancers and predicts developmental state. Proc Natl Acad Sci U S A. 2010;107(50):21931–6.

  137. Long M, Tao S, Vega D, Jiang T, Wen Q, Sophia L. A unique chromatin signature uncovers early developmental enhancers in humans. Nature. 2011;470(7333):279–83.

  138. Raisner R, Kharbanda S, Jin L, Jeng E, Chan E, Merchant M, et al. Enhancer Activity Requires CBP/P300 Bromodomain-Dependent Histone H3K27 Acetylation. Cell Rep [Internet]. 2018;24(7):1722–9. Available from: https://doi.org/10.1016/j.celrep.2018.07.041

  139. Mousseau G, Valente ST. Didehydro-Cortistatin A: a new player in HIV-therapy? Expert Rev Anti Infect Ther. 2016;14(2):145–8.

  140. Winters AC, Bernt KM. MLL-rearranged leukemias- An update on science and clinical approaches. Front Pediatr. 2017;5(February):11–3.

  141. Hsieh JJ-D, Ernst P, Erdjument-Bromage H, Tempst P, Korsmeyer SJ. Proteolytic Cleavage of MLL Generates a Complex of N- and C-Terminal Fragments That Confers Protein Stability and Subnuclear Localization. Mol Cell Biol. 2003;23(1):186–94.

  142. Rabalski AJ, Gyenis L, Litchfield DW. Molecular pathways: Emergence of protein kinase CK2 (CSNK2) as a potential target to inhibit survival and DNA damage response and repair pathways in cancer cells. Clin Cancer Res. 2016;22(12):2840–7.

Universiteit of Hogeschool
Master in de geneesmiddelenontwikkeling
Publicatiejaar
2021
Promotor(en)
Prof. Zeger Debyser
Kernwoorden
Share this on: