Abrupt and altered cell-type specific DNA methylation profiles in blood during acute HIV infection persists despite prompt initiation of ART

Michael J Corley, Carlo Sacdalan, Alina P S Pang, Nitiya Chomchey, Nisakorn Ratnaratorn, Victor Valcour, Eugene Kroon, Kyu S Cho, Andrew C Belden, Donn Colby, Merlin Robb, Denise Hsu, Serena Spudich, Robert Paul, Sandhya Vasan, Lishomwa C Ndhlovu, SEARCH010/RV254 and SEARCH013/RV304 study groups, Michael J Corley, Carlo Sacdalan, Alina P S Pang, Nitiya Chomchey, Nisakorn Ratnaratorn, Victor Valcour, Eugene Kroon, Kyu S Cho, Andrew C Belden, Donn Colby, Merlin Robb, Denise Hsu, Serena Spudich, Robert Paul, Sandhya Vasan, Lishomwa C Ndhlovu, SEARCH010/RV254 and SEARCH013/RV304 study groups

Abstract

HIV-1 disrupts the host epigenetic landscape with consequences for disease pathogenesis, viral persistence, and HIV-associated comorbidities. Here, we examined how soon after infection HIV-associated epigenetic changes may occur in blood and whether early initiation of antiretroviral therapy (ART) impacts epigenetic modifications. We profiled longitudinal genome-wide DNA methylation in monocytes and CD4+ T lymphocytes from 22 participants in the RV254/SEARCH010 acute HIV infection (AHI) cohort that diagnoses infection within weeks after estimated exposure and immediately initiates ART. We identified monocytes harbored 22,697 differentially methylated CpGs associated with AHI compared to 294 in CD4+ T lymphocytes. ART minimally restored less than 1% of these changes in monocytes and had no effect upon T cells. Monocyte DNA methylation patterns associated with viral load, CD4 count, CD4/CD8 ratio, and longitudinal clinical phenotypes. Our findings suggest HIV-1 rapidly embeds an epigenetic memory not mitigated by ART and support determining epigenetic signatures in precision HIV medicine. Trial Registration: NCT00782808 and NCT00796146.

Conflict of interest statement

I have read the journal’s policy and the authors of this manuscript have the following competing interests: LCN has served as an advisory board member for Abbvie, Cytodyn and ViiV for work unrelated to this project. All other authors declare no competing interests.

Figures

Fig 1. Monocyte cell type-specific differentially methylated…
Fig 1. Monocyte cell type-specific differentially methylated loci associated with acute HIV infection.
(a) Diagram of experimental design. Created with BioRender.com (b) Manhattan plot of differentially methylated loci associated with AHI in monocytes displayed across chromosomes. P values transformed using -log10(P) (c) Volcano plot displaying HUGO gene symbols and related CpG ID (cg#) for top hypomethylated and hypermethylated sites. Difference in DNA methylation displayed as delta beta values plotted against P values transformed using -log10(P). NS: non-significant. (d-q) Plots demonstrating changes in DNA methylation in AHI Fiebig I (red), Fiebig II (blue) Fiebig III-V (purple) compared to HIV- (green) at single CpGs sites related to specific genes or intergenic regions of the genome displayed in bold italic above CpG ID (cg#). Significance determined comparing HIV- vs. AHI and using FDR adjusted P-values <0.05.
Fig 2. Associations of monocyte cell type-specific…
Fig 2. Associations of monocyte cell type-specific DNA methylation with viral load, CD4 count, and CD4/CD8 ratio.
Scatter plots showing the correlation between (a-e) plasma viral load, (f-g) pre-ART CD4 count, and (h-o) pre-ART CD4/CD8 ratio and DNA methylation levels at single CpGs sites related to specific annotated protein-coding genes displayed in bold italic above CpG ID (cg#) in monocyte cells during AHI at entry. P-values were calculated with the Spearman correlation test.
Fig 3. Cell-type independent differentially methylated loci…
Fig 3. Cell-type independent differentially methylated loci associated with acute HIV infection.
(a) Diagram of experimental design displaying comparison utilized to identify DML in CD4+ T cells associated with AHI. Created with BioRender.com (b) Volcano plot displaying gene symbol and CpG ID (cg#) for top hypomethylated and hypermethylated sites associated with AHI in CD4+ T lymphocytes. Difference in DNA methylation displayed as delta beta values plotted against P values transformed using -log10(P). (c) Venn diagram displaying the number of overlapping DML and cell-type specific DML identified in CD4+ T cells and monocyte cells (d-s) Plots demonstrating changes in DNA methylation in AHI Fiebig I (red dots), Fiebig II (blue dots) Fiebig III-V (purple dots) participants compared to HIV- (green dots) at single CpGs sites related to annotated protein-coding genes displayed in bold italic above CpG ID (cg#) for CD4+ T lymphocytes and monocytes. Significance determined comparing HIV- vs. AHI and using FDR adjusted P-values <0.05.
Fig 4. Early initiation of ART during…
Fig 4. Early initiation of ART during acute infection minimally impacts monocyte cell type-specific differentially methylated loci associated with acute HIV infection.
(a) Diagram of longitudinal experimental design utilized to identify DML-related to ART treatment in purified monocytes. Created with BioRender.com (b) Venn diagram displaying the overlap of DML between DNA methylation sites identified in monocytes related to AHI and DNA methylation sites identified in monocytes that changed before and after early initiation of ART during AHI. (c) Heatmap showing the unsupervised clustering of DML at annotated protein-coding genes related to interferon at pre-ART (aqua color) and post-ART (light blue) timepoints for AHI participants staged at Fiebig I (red color), II (blue color), or III-V (purple color). Dendrogram shown above. Colors in the heatmap indicate CpG methylation levels (blue to red: low to high methylation levels). (d-o) Plots displaying longitudinal DNA methylation levels of AHI participants at Pre-ART and Post-ART timepoints by Fiebig stage. Red dots represent individuals in Fiebig I. Significance determined comparing repeated measures comparison of Pre-ART vs. Post-ART and using FDR adjusted P-values <0.05.
Fig 5. Differentially methylated loci identified in…
Fig 5. Differentially methylated loci identified in monocyte cells that persist in acute and chronic HIV infection despite ART.
(a-l) Plots of durable differentially methylated loci in in AHI Fiebig I (red), Fiebig II (blue) Fiebig III-V (purple) at Pre-ART and Post-ART time points compared to HIV- (green) at single CpGs sites related to specific genes or intergenic regions of the genome displayed in bold italic above CpG ID (cg#). Validation of these same durable differentially methylated loci in monocytes displayed for chronic HIV infection at Pre-ART (aqua) and Post-ART (light blue) along with the Berlin patient (star). Significance determined comparing HIV- vs. AHI pre-ART, AHI post-ART, CHI pre-ART, and CHI post-ART and using FDR adjusted P-values

Fig 6. Early initiation of ART during…

Fig 6. Early initiation of ART during acute infection impacts interferon-related gene programs in monocytes.

Fig 6. Early initiation of ART during acute infection impacts interferon-related gene programs in monocytes.
(a) Volcano plot showing gene symbol for top differentially expressed genes in monocytes comparing pre-ART and post-ART time points. Log fold change plotted against transformed -log10 P value. (b-m) Plots showing differentially expressed interferon-related genes in monocyte cells by Fiebig stage comparing pre-ART and post-ART timepoints. Fiebig I displayed in red, Fiebig II displayed in blue, and Fiebig III-V displayed in purple.

Fig 7. Monocyte gene expression associates with…

Fig 7. Monocyte gene expression associates with DNA methylation levels and clinical parameters during acute…

Fig 7. Monocyte gene expression associates with DNA methylation levels and clinical parameters during acute HIV infection.
(a-d) Correlation matrix plots showing the correlation coefficient for associations between gene expression levels, CD4 count, CD4/CD8 ratio, viral load, and DNA methylation levels at specific genomic loci. Significant correlations displayed as solid colored boxes (red displayed for negative correlations and blue for positive correlations). Correlation coefficient displayed.

Fig 8. DNA methylation related to the…

Fig 8. DNA methylation related to the IRF7 gene in monocytes relates to CD4 fold…

Fig 8. DNA methylation related to the IRF7 gene in monocytes relates to CD4 fold change at 96 weeks following the initiation of ART during acute HIV infection.
(a-d) Line plots of longitudinal CD4 count in participants by Fiebig stage from week 0 (pre-ART) through post-ART time points out to week 96. (e) Correlation plot displaying relationship between IRF7 DNA methylation levels in monocyte cells assessed at baseline and CD4 fold change calculated from week 0 to week 96. Fiebig stage of participant displayed: Fiebig I displayed in red, Fiebig II displayed in blue, and Fiebig III-V displayed in purple.

Fig 9. DNA methylation features associated with…

Fig 9. DNA methylation features associated with AHI in monocytes and CD4 + T lymphocytes…

Fig 9. DNA methylation features associated with AHI in monocytes and CD4+ T lymphocytes predict favorable clinical phenotypes and neurocognitive performance following ART.
(a-b) Feature importance plots of gradient boosting machine learning and logistic regression models with interaction features for predicting favorable clinical phenotype at week 96 post-ART utilizing cell-type specific DNA methylation measures and clinical parameters (estimated days of infection, viral load, CD4 count, CD8 count, and CD4/CD8 ratio) during viral establishment in AHI prior to ART initiation. (c) Receiver operating characteristic curve showing performance of classification models (area under curve AUC) in discovery AHI DNA methylation feature set. (e-f) Feature importance plots of gradient boosting machine learning and logistic regression models with interaction features for predicting neurocognitive performance trajectory at week 96 post-ART. (g) Receiver operating characteristic curve showing performance of classification models.
All figures (9)
Fig 6. Early initiation of ART during…
Fig 6. Early initiation of ART during acute infection impacts interferon-related gene programs in monocytes.
(a) Volcano plot showing gene symbol for top differentially expressed genes in monocytes comparing pre-ART and post-ART time points. Log fold change plotted against transformed -log10 P value. (b-m) Plots showing differentially expressed interferon-related genes in monocyte cells by Fiebig stage comparing pre-ART and post-ART timepoints. Fiebig I displayed in red, Fiebig II displayed in blue, and Fiebig III-V displayed in purple.
Fig 7. Monocyte gene expression associates with…
Fig 7. Monocyte gene expression associates with DNA methylation levels and clinical parameters during acute HIV infection.
(a-d) Correlation matrix plots showing the correlation coefficient for associations between gene expression levels, CD4 count, CD4/CD8 ratio, viral load, and DNA methylation levels at specific genomic loci. Significant correlations displayed as solid colored boxes (red displayed for negative correlations and blue for positive correlations). Correlation coefficient displayed.
Fig 8. DNA methylation related to the…
Fig 8. DNA methylation related to the IRF7 gene in monocytes relates to CD4 fold change at 96 weeks following the initiation of ART during acute HIV infection.
(a-d) Line plots of longitudinal CD4 count in participants by Fiebig stage from week 0 (pre-ART) through post-ART time points out to week 96. (e) Correlation plot displaying relationship between IRF7 DNA methylation levels in monocyte cells assessed at baseline and CD4 fold change calculated from week 0 to week 96. Fiebig stage of participant displayed: Fiebig I displayed in red, Fiebig II displayed in blue, and Fiebig III-V displayed in purple.
Fig 9. DNA methylation features associated with…
Fig 9. DNA methylation features associated with AHI in monocytes and CD4+ T lymphocytes predict favorable clinical phenotypes and neurocognitive performance following ART.
(a-b) Feature importance plots of gradient boosting machine learning and logistic regression models with interaction features for predicting favorable clinical phenotype at week 96 post-ART utilizing cell-type specific DNA methylation measures and clinical parameters (estimated days of infection, viral load, CD4 count, CD8 count, and CD4/CD8 ratio) during viral establishment in AHI prior to ART initiation. (c) Receiver operating characteristic curve showing performance of classification models (area under curve AUC) in discovery AHI DNA methylation feature set. (e-f) Feature importance plots of gradient boosting machine learning and logistic regression models with interaction features for predicting neurocognitive performance trajectory at week 96 post-ART. (g) Receiver operating characteristic curve showing performance of classification models.

References

    1. Youngblood B, Reich NO. The early expressed HIV-1 genes regulate DNMT1 expression. Epigenetics. 2008;3: 149–156. doi: 10.4161/epi.3.3.6372
    1. Johnson JS, Lucas SY, Amon LM, Skelton S, Nazitto R, Carbonetti S, et al.. Reshaping of the Dendritic Cell Chromatin Landscape and Interferon Pathways during HIV Infection. Cell Host Microbe. 2018;23: 366–381.e9. doi: 10.1016/j.chom.2018.01.012
    1. Einkauf KB, Lee GQ, Gao C, Sharaf R, Sun X, Hua S, et al.. Intact HIV-1 proviruses accumulate at distinct chromosomal positions during prolonged antiretroviral therapy. J Clin Invest. 2019. doi: 10.1172/JCI124291
    1. Lucic B, Chen H-C, 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. 2019;10: 4059. doi: 10.1038/s41467-019-12046-3
    1. Liu R, Yeh Y-HJ, Varabyou A, Collora JA, Sherrill-Mix S, Talbot CC Jr, et al.. Single-cell transcriptional landscapes reveal HIV-1-driven aberrant host gene transcription as a potential therapeutic target. Sci Transl Med. 2020;12. doi: 10.1126/scitranslmed.aaz0802
    1. Morales-Nebreda L, McLafferty FS, Singer BD. DNA methylation as a transcriptional regulator of the immune system. Transl Res. 2019;204: 1–18. doi: 10.1016/j.trsl.2018.08.001
    1. Zhang X, Justice AC, Hu Y, Wang Z, Zhao H, Wang G, et al.. Epigenome-wide differential DNA methylation between HIV-infected and uninfected individuals. Epigenetics. 2016;11: 750–760. doi: 10.1080/15592294.2016.1221569
    1. Zhang X, Hu Y, Aouizerat BE, Peng G, Marconi VC, Corley MJ, et al.. Machine learning selected smoking-associated DNA methylation signatures that predict HIV prognosis and mortality. Clin Epigenetics. 2018;10: 155. doi: 10.1186/s13148-018-0591-z
    1. Corley MJ, Dye C, D’Antoni ML, Byron MM, Yo KL-A, Lum-Jones A, et al.. Comparative DNA Methylation Profiling Reveals an Immunoepigenetic Signature of HIV-related Cognitive Impairment. Sci Rep. 2016;6: 33310. doi: 10.1038/srep33310
    1. Dye CK, Corley MJ, Li D, Khadka VS, Mitchell BI, Sultana R, et al.. Comparative DNA methylomic analyses reveal potential origins of novel epigenetic biomarkers of insulin resistance in monocytes from virally suppressed HIV-infected adults. Clin Epigenetics. 2019;11: 95. doi: 10.1186/s13148-019-0694-1
    1. Zhang X, Hu Y, Justice AC, Li B, Wang Z, Zhao H, et al.. DNA methylation signatures of illicit drug injection and hepatitis C are associated with HIV frailty. Nat Commun. 2017;8: 2243. doi: 10.1038/s41467-017-02326-1
    1. Boulias K, Lieberman J, Greer EL. An Epigenetic Clock Measures Accelerated Aging in Treated HIV Infection. Molecular cell. 2016. pp. 153–155. doi: 10.1016/j.molcel.2016.04.008
    1. Horvath S, Levine AJ. HIV-1 Infection Accelerates Age According to the Epigenetic Clock. J Infect Dis. 2015;212: 1563–1573. doi: 10.1093/infdis/jiv277
    1. Oriol-Tordera B, Berdasco M, Llano A, Mothe B, Gálvez C, Martinez-Picado J, et al.. Methylation regulation of Antiviral host factors, Interferon Stimulated Genes (ISGs) and T-cell responses associated with natural HIV control. PLoS Pathog. 2020;16: e1008678. doi: 10.1371/journal.ppat.1008678
    1. Roadmap Epigenomics Consortium, Kundaje A, Meuleman W, Ernst J, Bilenky M, Yen A, et al.. Integrative analysis of 111 reference human epigenomes. Nature. 2015;518: 317–330. doi: 10.1038/nature14248
    1. Ananworanich J, Chomont N, Eller LA, Kroon E, Tovanabutra S, Bose M, et al.. HIV DNA Set Point is Rapidly Established in Acute HIV Infection and Dramatically Reduced by Early ART. EBioMedicine. 2016;11: 68–72. doi: 10.1016/j.ebiom.2016.07.024
    1. Colby DJ, Trautmann L, Pinyakorn S, Leyre L, Pagliuzza A, Kroon E, et al.. Rapid HIV RNA rebound after antiretroviral treatment interruption in persons durably suppressed in Fiebig I acute HIV infection. Nat Med. 2018;24: 923–926. doi: 10.1038/s41591-018-0026-6
    1. Sacdalan C, Crowell T, Colby D, Kroon E, Chan P, Pinyakorn S, et al.. Brief Report: Safety of Frequent Blood Sampling in Research Participants in an Acute HIV Infection Cohort in Thailand. J Acquir Immune Defic Syndr. 2017;76: 98–101.
    1. Horvath S. DNA methylation age of human tissues and cell types. Genome Biol. 2013;14: R115. doi: 10.1186/gb-2013-14-10-r115
    1. Gross AM, Jaeger PA, Kreisberg JF, Licon K, Jepsen KL, Khosroheidari M, et al.. Methylome-wide Analysis of Chronic HIV Infection Reveals Five-Year Increase in Biological Age and Epigenetic Targeting of HLA. Mol Cell. 2016;62: 157–168. doi: 10.1016/j.molcel.2016.03.019
    1. Esteban-Cantos A, Rodríguez-Centeno J, Barruz P, Alejos B, Saiz-Medrano G, Nevado J, et al.. Epigenetic age acceleration changes 2 years after antiretroviral therapy initiation in adults with HIV: a substudy of the NEAT001/ANRS143 randomised trial. Lancet HIV. 2021;8: e197–e205. doi: 10.1016/S2352-3018(21)00006-0
    1. Shin J-S, Greer AM. The role of FcεRI expressed in dendritic cells and monocytes. Cell Mol Life Sci. 2015;72: 2349–2360. doi: 10.1007/s00018-015-1870-x
    1. Handoko R, Colby DJ, Kroon E, Sacdalan C, de Souza M, Pinyakorn S, et al.. Determinants of suboptimal CD4+ T cell recovery after antiretroviral therapy initiation in a prospective cohort of acute HIV-1 infection. J Int AIDS Soc. 2020;23: e25585. doi: 10.1002/jia2.25585
    1. Sirois M, Robitaille L, Allary R, Shah M, Woelk CH, Estaquier J, et al.. TRAF6 and IRF7 control HIV replication in macrophages. PLoS One. 2011;6: e28125. doi: 10.1371/journal.pone.0028125
    1. Hernández-Walias F, Ruiz-de-León MJ, Rosado-Sánchez I, Vázquez E, Leal M, Moreno S, et al.. New signatures of poor CD4 cell recovery after suppressive antiretroviral therapy in HIV-1-infected individuals: involvement of miR-192, IL-6, sCD14 and miR-144. Sci Rep. 2020;10: 2937. doi: 10.1038/s41598-020-60073-8
    1. Chan P, Kerr SJ, Kroon E, Colby D, Sacdalan C, Hellmuth J, et al.. Cognitive trajectories after treatment in acute HIV infection. AIDS. 2021. doi: 10.1097/QAD.0000000000002831
    1. D’Antoni ML, Byron MM, Chan P, Sailasuta N, Sacdalan C, Sithinamsuwan P, et al.. Normalization of Soluble CD163 Levels After Institution of Antiretroviral Therapy During Acute HIV Infection Tracks with Fewer Neurological Abnormalities. J Infect Dis. 2018;218: 1453–1463. doi: 10.1093/infdis/jiy337
    1. Kore I, Ananworanich J, Valcour V, Fletcher JLK, Chalermchai T, Paul R, et al.. Neuropsychological Impairment in Acute HIV and the Effect of Immediate Antiretroviral Therapy. J Acquir Immune Defic Syndr. 2015;70: 393–399. doi: 10.1097/QAI.0000000000000746
    1. Grinsztejn B, Hosseinipour MC, Ribaudo HJ, Swindells S, Eron J, Chen YQ, et al.. Effects of early versus delayed initiation of antiretroviral treatment on clinical outcomes of HIV-1 infection: results from the phase 3 HPTN 052 randomised controlled trial. Lancet Infect Dis. 2014;14: 281–290. doi: 10.1016/S1473-3099(13)70692-3
    1. Fidler S, Olson AD, Bucher HC, Fox J, Thornhill J, Morrison C, et al.. Virological Blips and Predictors of Post Treatment Viral Control After Stopping ART Started in Primary HIV Infection. J Acquir Immune Defic Syndr. 2017;74: 126–133. doi: 10.1097/QAI.0000000000001220
    1. Thornhill J, Inshaw J, Oomeer S, Kaleebu P, Cooper D, Ramjee G, et al.. Enhanced normalisation of CD4/CD8 ratio with early antiretroviral therapy in primary HIV infection. J Int AIDS Soc. 2014;17: 19480. doi: 10.7448/IAS.17.4.19480
    1. Noel N, Lerolle N, Lécuroux C, Goujard C, Venet A, Saez-Cirion A, et al.. Immunologic and Virologic Progression in HIV Controllers: The Role of Viral “Blips” and Immune Activation in the ANRS CO21 CODEX Study. PLoS One. 2015;10: e0131922. doi: 10.1371/journal.pone.0131922
    1. Young J, Rickenbach M, Calmy A, Bernasconi E, Staehelin C, Schmid P, et al.. Transient detectable viremia and the risk of viral rebound in patients from the Swiss HIV Cohort Study. BMC Infect Dis. 2015;15: 382. doi: 10.1186/s12879-015-1120-8
    1. Hoare J, Stein DJ, Heany SJ, Fouche J-P, Phillips N, Er S, et al.. Accelerated epigenetic aging in adolescents living with HIV is associated with altered development of brain structures. J Neurovirol. 2021. doi: 10.1007/s13365-021-00947-3
    1. Ananworanich J, Eller LA, Pinyakorn S, Kroon E, Sriplenchan S, Fletcher JL, et al.. Viral kinetics in untreated versus treated acute HIV infection in prospective cohort studies in Thailand. J Int AIDS Soc. 2017;20: 21652. doi: 10.7448/IAS.20.1.21652
    1. Crowell TA, Phanuphak N, Pinyakorn S, Kroon E, Fletcher JLK, Colby D, et al.. Virologic failure is uncommon after treatment initiation during acute HIV infection. AIDS. 2016;30: 1943–1950. doi: 10.1097/QAD.0000000000001148
    1. Sereti I, Krebs SJ, Phanuphak N, Fletcher JL, Slike B, Pinyakorn S, et al.. Persistent, Albeit Reduced, Chronic Inflammation in Persons Starting Antiretroviral Therapy in Acute HIV Infection. Clin Infect Dis. 2017;64: 124–131. doi: 10.1093/cid/ciw683
    1. Moron-Lopez S, Urrea V, Dalmau J, Lopez M, Puertas MC, Ouchi D, et al.. The genome-wide methylation profile of CD4+ T cells from HIV-infected individuals identifies distinct patterns associated with disease progression. Clin Infect Dis. 2020. doi: 10.1093/cid/ciaa1047
    1. Rempel H, Sun B, Calosing C, Abadjian L, Monto A, Pulliam L. Monocyte activation in HIV/HCV coinfection correlates with cognitive impairment. PLoS One. 2013;8: e55776. doi: 10.1371/journal.pone.0055776
    1. Wie S-H, Du P, Luong TQ, Rought SE, Beliakova-Bethell N, Lozach J, et al.. HIV downregulates interferon-stimulated genes in primary macrophages. J Interferon Cytokine Res. 2013;33: 90–95. doi: 10.1089/jir.2012.0052
    1. Tilton JC, Johnson AJ, Luskin MR, Manion MM, Yang J, Adelsberger JW, et al.. Diminished production of monocyte proinflammatory cytokines during human immunodeficiency virus viremia is mediated by type I interferons. J Virol. 2006;80: 11486–11497. doi: 10.1128/JVI.00324-06
    1. Rempel H, Sun B, Calosing C, Pillai SK, Pulliam L. Interferon-alpha drives monocyte gene expression in chronic unsuppressed HIV-1 infection. AIDS. 2010;24: 1415–1423. doi: 10.1097/QAD.0b013e32833ac623
    1. Pulliam L, Rempel H, Sun B, Abadjian L, Calosing C, Meyerhoff DJ. A peripheral monocyte interferon phenotype in HIV infection correlates with a decrease in magnetic resonance spectroscopy metabolite concentrations. AIDS. 2011;25: 1721–1726. doi: 10.1097/QAD.0b013e328349f022
    1. Patro SC, Pal S, Bi Y, Lynn K, Mounzer KC, Kostman JR, et al.. Shift in monocyte apoptosis with increasing viral load and change in apoptosis-related ISG/Bcl2 family gene expression in chronically HIV-1-infected subjects. J Virol. 2015;89: 799–810. doi: 10.1128/JVI.02382-14
    1. Utay NS, Douek DC. Interferons and HIV Infection: The Good, the Bad, and the Ugly. Pathog Immun. 2016;1: 107–116. doi: 10.20411/pai.v1i1.125
    1. Sandler NG, Bosinger SE, Estes JD, Zhu RTR, Tharp GK, Boritz E, et al.. Type I interferon responses in rhesus macaques prevent SIV infection and slow disease progression. Nature. 2014;511: 601–605. doi: 10.1038/nature13554
    1. Liu XS, Wu H, Ji X, Stelzer Y, Wu X, Czauderna S, et al.. Editing DNA Methylation in the Mammalian Genome. Cell. 2016;167: 233–247.e17. doi: 10.1016/j.cell.2016.08.056
    1. Mlambo T, Nitsch S, Hildenbeutel M, Romito M, Müller M, Bossen C, et al.. Designer epigenome modifiers enable robust and sustained gene silencing in clinically relevant human cells. Nucleic Acids Res. 2018;46: 4456–4468. doi: 10.1093/nar/gky171
    1. Fourati S, Ribeiro SP, Blasco Tavares Pereira Lopes F, Talla A, Lefebvre F, Cameron M, et al.. Integrated systems approach defines the antiviral pathways conferring protection by the RV144 HIV vaccine. Nat Commun. 2019;10: 863. doi: 10.1038/s41467-019-08854-2
    1. Lu R, Pitha PM. Monocyte differentiation to macrophage requires interferon regulatory factor 7. J Biol Chem. 2001;276: 45491–45496. doi: 10.1074/jbc.C100421200
    1. McMichael AJ, Borrow P, Tomaras GD, Goonetilleke N, Haynes BF. The immune response during acute HIV-1 infection: clues for vaccine development. Nat Rev Immunol. 2010;10: 11–23. doi: 10.1038/nri2674
    1. Zheng SC, Breeze CE, Beck S, Dong D, Zhu T, Ma L, et al.. EpiDISH web server: Epigenetic Dissection of Intra-Sample-Heterogeneity with online GUI. Bioinformatics. 2019. doi: 10.1093/bioinformatics/btz833
    1. Teschendorff AE, Breeze CE, Zheng SC, Beck S. A comparison of reference-based algorithms for correcting cell-type heterogeneity in Epigenome-Wide Association Studies. BMC Bioinformatics. 2017;18: 105. doi: 10.1186/s12859-017-1511-5
    1. Salas LA, Koestler DC, Butler RA, Hansen HM, Wiencke JK, Kelsey KT, et al.. An optimized library for reference-based deconvolution of whole-blood biospecimens assayed using the Illumina HumanMethylationEPIC BeadArray. Genome Biol. 2018;19: 64. doi: 10.1186/s13059-018-1448-7
    1. Clark SJ, Smallwood SA, Lee HJ, Krueger F, Reik W, Kelsey G. Genome-wide base-resolution mapping of DNA methylation in single cells using single-cell bisulfite sequencing (scBS-seq). Nat Protoc. 2017;12: 534–547. doi: 10.1038/nprot.2016.187
    1. Le T, Wright EJ, Smith DM, He W, Catano G, Okulicz JF, et al.. Enhanced CD4+ T-cell recovery with earlier HIV-1 antiretroviral therapy. N Engl J Med. 2013;368: 218–230. doi: 10.1056/NEJMoa1110187
    1. Kassutto S, Maghsoudi K, Johnston MN, Robbins GK, Burgett NC, Sax PE, et al.. Longitudinal analysis of clinical markers following antiretroviral therapy initiated during acute or early HIV type 1 infection. Clin Infect Dis. 2006;42: 1024–1031. doi: 10.1086/500410
    1. Marchetti G, Bellistrì GM, Borghi E, Tincati C, Ferramosca S, La Francesca M, et al.. Microbial translocation is associated with sustained failure in CD4+ T-cell reconstitution in HIV-infected patients on long-term highly active antiretroviral therapy. AIDS. 2008;22: 2035–2038. doi: 10.1097/QAD.0b013e3283112d29
    1. Jiang W, Lederman MM, Hunt P, Sieg SF, Haley K, Rodriguez B, et al.. Plasma levels of bacterial DNA correlate with immune activation and the magnitude of immune restoration in persons with antiretroviral-treated HIV infection. J Infect Dis. 2009;199: 1177–1185. doi: 10.1086/597476
    1. Jähner D, Jaenisch R. Retrovirus-induced de novo methylation of flanking host sequences correlates with gene inactivity. Nature. 1985;315: 594–597. doi: 10.1038/315594a0
    1. Folks TM, Kessler SW, Orenstein JM, Justement JS, Jaffe ES, Fauci AS. Infection and replication of HIV-1 in purified progenitor cells of normal human bone marrow. Science. 1988;242: 919–922. doi: 10.1126/science.2460922
    1. Stanley SK, Kessler SW, Justement JS, Schnittman SM, Greenhouse JJ, Brown CC, et al.. CD34+ bone marrow cells are infected with HIV in a subset of seropositive individuals. J Immunol. 1992;149: 689–697.
    1. Fantuzzi L, Canini I, Belardelli F, Gessani S. HIV-1 gp120 stimulates the production of beta-chemokines in human peripheral blood monocytes through a CD4-independent mechanism. J Immunol. 2001;166: 5381–5387. doi: 10.4049/jimmunol.166.9.5381
    1. 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. N Engl J Med. 2009;360: 692–698. doi: 10.1056/NEJMoa0802905
    1. Ananworanich J, Sacdalan CP, Pinyakorn S, Chomont N, de Souza M, Luekasemsuk T, et al.. Virological and immunological characteristics of HIV-infected individuals at the earliest stage of infection. J Virus Erad. 2016;2: 43–48.
    1. D’Elia L, Satz P, Uchiyama CL, White T. Color trails 1 and 2. Odessa, FL: Psychological Assessment Resources. 1989.
    1. Heaps J, Valcour V, Chalermchai T, Paul R, Rattanamanee S, Siangphoe U, et al.. Development of normative neuropsychological performance in Thailand for the assessment of HIV-associated neurocognitive disorders. J Clin Exp Neuropsychol. 2013;35: 1–8. doi: 10.1080/13803395.2012.733682
    1. Tian Y, Morris TJ, Webster AP, Yang Z, Beck S, Feber A, et al.. ChAMP: updated methylation analysis pipeline for Illumina BeadChips. Bioinformatics. 2017;33: 3982–3984. doi: 10.1093/bioinformatics/btx513
    1. Smyth GK. Limma: linear models for microarray data. Gentleman RCarey VDudoit SIrizarry RHuber W Bioinformatics and computational biology solutions using R and Bioconductor. New York: Springer; 2005.
    1. Hansen KD. IlluminaHumanMethylationEPICanno.ilm10b4.hg19: Annotation for Illumina’s EPIC methylation arrays. 2017. Available:
    1. LiLin-Yin. CMplot: Circle Manhattan Plot. 2020. Available:
    1. Blighe K, Rana S, and Lewis M. EnhancedVolcano: Publication-ready volcano plots with enhanced colouring and labeling. 2018. Available:
    1. Kolde R. pheatmap: Pretty Heatmaps. 2019.
    1. Li M, Zou D, Li Z, Gao R, Sang J, Zhang Y, et al.. EWAS Atlas: a curated knowledgebase of epigenome-wide association studies. Nucleic Acids Res. 2019;47: D983–D988. doi: 10.1093/nar/gky1027
    1. Phipson B, Maksimovic J, Oshlack A. missMethyl: an R package for analyzing data from Illumina’s HumanMethylation450 platform. Bioinformatics. 2016;32: 286–288. doi: 10.1093/bioinformatics/btv560
    1. Lu AT, Quach A, Wilson JG, Reiner AP, Aviv A, Raj K, et al.. DNA methylation GrimAge strongly predicts lifespan and healthspan. Aging. 2019;11: 303–327. doi: 10.18632/aging.101684
    1. Robinson MD, McCarthy DJ, Smyth GK. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 2010;26: 139–140. doi: 10.1093/bioinformatics/btp616
    1. Smyth GK. Linear models and empirical bayes methods for assessing differential expression in microarray experiments. Stat Appl Genet Mol Biol. 2004;3: Article3. doi: 10.2202/1544-6115.1027
    1. Virtanen P, Gommers R, Oliphant TE, Haberland M, Reddy T, Cournapeau D, et al.. SciPy 1.0: fundamental algorithms for scientific computing in Python. Nat Methods. 2020;17: 261–272. doi: 10.1038/s41592-019-0686-2

Source: PubMed

3
Subscribe