HIV-Sheltering Platelets From Immunological Non-Responders Induce a Dysfunctional Glycolytic CD4+ T-Cell Profile

Aiwei Zhu, Fernando Real, Jaja Zhu, Ségolène Greffe, Pierre de Truchis, Elisabeth Rouveix, Morgane Bomsel, Claude Capron, Aiwei Zhu, Fernando Real, Jaja Zhu, Ségolène Greffe, Pierre de Truchis, Elisabeth Rouveix, Morgane Bomsel, Claude Capron

Abstract

Immunological non-responders (InRs) are HIV-infected individuals in whom the administration of combination antiretroviral therapy (cART), although successful in suppressing viral replication, cannot properly reconstitute patient circulating CD4+ T-cell number to immunocompetent levels. The causes for this immunological failure remain elusive, and no therapeutic strategy is available to restore a proper CD4+ T-cell immune response in these individuals. We have recently demonstrated that platelets harboring infectious HIV are a hallmark of InR, and we now report on a causal connection between HIV-containing platelets and T-cell dysfunctions. We show here that in vivo, platelet-T-cell conjugates are more frequent among CD4+ T cells in InRs displaying HIV-containing platelets (<350 CD4+ T cells/μl blood for >1 year) as compared with healthy donors or immunological responders (IRs; >350 CD4+ T cells/μl). This contact between platelet containing HIV and T cell in the conjugates is not infectious for CD4+ T cells, as coculture of platelets from InRs containing HIV with healthy donor CD4+ T cells fails to propagate infection to CD4+ T cells. In contrast, when macrophages are the target of platelets containing HIV from InRs, macrophages become infected. Differential transcriptomic analyses comparing InR and IR CD4+ T cells reveal an upregulation of genes involved in both aerobic and anaerobic glycolysis in CD4+ T cells from InR vs. IR individuals. Accordingly, InR platelets containing HIV induce a dysfunctional increase in glycolysis-mediated energy production in CD4+ T cells as compared with T cells cocultured with IR platelets devoid of virus. In contrast, macrophage metabolism is not affected by platelet contact. Altogether, this brief report demonstrates a direct causal link between presence of HIV in platelets and T-cell dysfunctions typical of InR, contributing to devise a platelet-targeted therapy for improving immune reconstitution in these individuals.

Keywords: CD4+ T-cell metabolism; HIV-1; glycolysis; immunological failure; platelets; virus-containing platelets.

Conflict of interest statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2022 Zhu, Real, Zhu, Greffe, de Truchis, Rouveix, Bomsel and Capron.

Figures

Figure 1
Figure 1
Platelets from immunological non-responders (InRs) form more conjugates with CD4+ T cells in vivo but do not transfer infectious virus to CD4+ T cells in vitro. (A) Platelet–CD4+ T-cell conjugates are more frequent in the circulation of InRs in vivo. Flow cytometry analysis of CD4+ T cells (CD3+CD4+) conjugated with platelets (CD41/61high) circulating in the blood of combination antiretroviral therapy (cART)-treated patients virally suppressed. Left: Gating strategy to assess platelet–T-cell conjugates. Right: Percentage of platelet–CD4+ T cell (CD3+CD4+CD41/61high) conjugates among CD4+ T cells (CD3+CD4+) as detected in healthy donor (gray) and HIV-infected patient samples categorized as immunological responder (IR, orange) and InR (blue). Kruskal–Wallis, statistically relevant differences p < 0.05 (*). (B, C) Upon direct interaction, HIV-containing (HIV+) platelets propagate infection to macrophages (MΦ), not to CD4+ T cells. (B)In vitro experimental setup. Left: HIV+ platelets are directly incubated with activated CD4+ T cells from healthy donors for a 14-day viral outgrowth assay. Right: MΦ from heathy donors interacts overnight with HIV+ platelets. At day 7, MΦ culture supernatants are collected and tested for their infectious replicative virus content by addition of the same activated CD4+ T cells from healthy donors as in left for a 14-day viral outgrowth assay. (C) Viral production quantification: HIV-1 is quantified in coculture supernatants after 14 days of viral outgrowth in CD4+ T cells that were incubated directly with HIV+ platelets in the presence or absence of polybrene (left) or incubated with supernatants of MΦ infected upon interaction with HIV+ platelets in the presence or absence of abciximab (right). LOD, limit of detection for the technique. *Statistically significant.
Figure 2
Figure 2
Differential transcriptomic analyses of CD4+ T cells from immunological non-responder (InR) and immunological responder (IR) patients focusing on genes associated with metabolic pathways. (A) InR, IR, and healthy donor transcriptomes have distinct profiles. Uniform Manifold Approximation and Projection (UMAP) of transcriptomic data from the GSE106792 dataset based on CD4+ T cells InR, IR, and healthy donors grouping individuals according to blood CD4+ T-cell counts and assessed by the Gene Expression Omnibus (GEO) constructed at a probability of 0.99. (B) Volcano plot from the differentially expressed gene (DEG) analysis between InR and IR CD4+ T cells. In the y axis, significance is indicated by red dotted line threshold at -Log10p value >3 for both InR and IR datasets. In the x axis, significance is indicated by red dotted line thresholds at Log2 fold change >0.5 for InR and <-0.5 for IR DEGs. (C) Differential biological pathway analysis activated in InR vs. IR CD4+ T cells using g:Profiler. (D) Network of biological pathways enriched in CD4+ T cells from InR (blue nodes and connectors) or from IR (orange nodes and connectors) (E) HumanCyC metabolic map showing genes upregulated in CD4+ T cells from InR (blue font) and upregulated in CD4+ T cells from IR (orange font) implicated in glycolysis, pyruvate oxidation, and TCA cycle biochemical reactions.
Figure 3
Figure 3
Metabolic profiling of CD4+ T cells after interaction with immunological non-responder (InR), immunological responder (IR), or healthy donor platelets, sheltering or not HIV in platelets. (A) Oxygen consumption rate (OCR; upper) and extracellular acidification rate (ECAR; lower) obtained throughout 1 h of data acquisition from live CD4+ T cells that interacted with InR (blue), IR (orange), or healthy donor (gray) platelets. Filled or empty circles with the same color code indicate the presence (HIVpos) or absence (HIVneg) of HIV in platelets, respectively. During the time course of data acquisition, the oxidative phosphorylation (OXPHOS) inhibitor and mitochondrial respiratory chain blocker oligomycin and rotenone/antimycin were injected at time points indicated by red and purple lines, respectively. (B) Energy phenotype map of CD4+ T cells that interacted with InR (blue), IR (orange), or healthy donor (gray) platelets. Filled or empty circles with the same color code indicate the presence (HIVpos) or absence (HIVneg) of HIV in platelets, respectively. (C) ATP production rate from CD4+ T cells (left) or macrophages (right) that interacted with InR, IR, or healthy donor platelets. The contribution of either glycolysis or mitochondria respiration to ATP production is discriminated in the bar graph by orange and red, respectively. Asterisks indicate statistically relevant differences established using a threshold of p < 0.05 calculated with a Kruskal–Wallis test on glycolysis levels (orange). Mitochondrial ATP production does not differ between groups using the same statistical test and significance threshold. (D) ATP production rate from CD4+ T cells that interacted with healthy donor platelets, IR platelets without HIV (HIVneg) in platelets, IR platelets with HIV (HIVpos) in platelets, InR HIVneg platelets, and InR HIVpos platelets. The contribution of either glycolysis or mitochondria respiration to ATP production is discriminated in the bar graph by orange and red, respectively. Asterisks indicate statistically relevant differences established by threshold of p < 0.05 using ANOVA on glycolysis levels (orange). Mitochondrial ATP production does not differ between groups using the same statistical test and significance threshold.

References

    1. Kaufmann GR, Perrin L, Pantaleo G, Opravil M, Furrer H, Telenti A, et al. . CD4 T-Lymphocyte Recovery in Individuals With Advanced HIV-1 Infection Receiving Potent Antiretroviral Therapy for 4 Years: The Swiss HIV Cohort Study. Arch Intern Med (2003) 163(18):2187–95. doi: 10.1001/archinte.163.18.2187
    1. Baker JV, Peng G, Rapkin J, Krason D, Reilly C, Cavert WP, et al. . Poor Initial CD4+ Recovery With Antiretroviral Therapy Prolongs Immune Depletion and Increases Risk for AIDS and Non-AIDS Diseases. J Acquir Immune Defic Syndr (2008) 48(5):541–6. doi: 10.1097/QAI.0b013e31817bebb3
    1. Kelley CF, Kitchen CM, Hunt PW, Rodriguez B, Hecht FM, Kitahata M, et al. . Incomplete Peripheral CD4+ Cell Count Restoration in HIV-Infected Patients Receiving Long-Term Antiretroviral Treatment. Clin Infect Dis (2009) 48(6):787–94. doi: 10.1086/597093
    1. Chene G, Sterne JA, May M, Costagliola D, Ledergerber B, Phillips AN, et al. . Prognostic Importance of Initial Response in HIV-1 Infected Patients Starting Potent Antiretroviral Therapy: Analysis of Prospective Studies. Lancet (2003) 362(9385):679–86. doi: 10.1016/s0140-6736(03)14229-8
    1. Real F, Capron C, Sennepin A, Arrigucci R, Zhu A, Sannier G, et al. . Platelets From HIV-Infected Individuals on Antiretroviral Drug Therapy With Poor CD4(+) T Cell Recovery can Harbor Replication-Competent HIV Despite Viral Suppression. Sci Transl Med (2020) 12(535):1–11. doi: 10.1126/scitranslmed.aat6263
    1. Deeks SG, Kitchen CM, Liu L, Guo H, Gascon R, Narvaez AB, et al. . Immune Activation Set Point During Early HIV Infection Predicts Subsequent CD4+ T-Cell Changes Independent of Viral Load. Blood (2004) 104(4):942–7. doi: 10.1182/blood-2003-09-3333
    1. Hunt PW, Martin JN, Sinclair E, Bredt B, Hagos E, Lampiris H, et al. . T Cell Activation is Associated With Lower CD4+ T Cell Gains in Human Immunodeficiency Virus-Infected Patients With Sustained Viral Suppression During Antiretroviral Therapy. J Infect Dis (2003) 187(10):1534–43. doi: 10.1086/374786
    1. Marziali M, De Santis W, Carello R, Leti W, Esposito A, Isgro A, et al. . T-Cell Homeostasis Alteration in HIV-1 Infected Subjects With Low CD4 T-Cell Count Despite Undetectable Virus Load During HAART. AIDS (2006) 20(16):2033–41. doi: 10.1097/01.aids.0000247588.69438.fd
    1. Nunes-Alves C, Nobrega C, Behar SM, Correia-Neves M. Tolerance has Its Limits: How the Thymus Copes With Infection. Trends Immunol (2013) 34(10):502–10. doi: 10.1016/j.it.2013.06.004
    1. Wilson EM, Singh A, Hullsiek KH, Gibson D, Henry WK, Lichtenstein K, et al. . Monocyte-Activation Phenotypes Are Associated With Biomarkers of Inflammation and Coagulation in Chronic HIV Infection. J Infect Dis (2014) 210(9):1396–406. doi: 10.1093/infdis/jiu275
    1. Zamora C, Canto E, Nieto JC, Ortiz MA, Diaz-Torne C, Diaz-Lopez C, et al. . Functional Consequences of Platelet Binding to T Lymphocytes in Inflammation. J Leukoc Biol (2013) 94(3):521–9. doi: 10.1189/jlb.0213074
    1. Green SA, Smith M, Hasley RB, Stephany D, Harned A, Nagashima K, et al. . Activated Platelet-T-Cell Conjugates in Peripheral Blood of Patients With HIV Infection: Coupling Coagulation/Inflammation and T Cells. AIDS (2015) 29(11):1297–308. doi: 10.1097/QAD.0000000000000701
    1. Gerdes N, Zhu L, Ersoy M, Hermansson A, Hjemdahl P, Hu H, et al. . Platelets Regulate CD4(+) T-Cell Differentiation via Multiple Chemokines in Humans. Thromb Haemost (2011) 106(2):353–62. doi: 10.1160/TH11-01-0020
    1. Starossom SC, Veremeyko T, Yung AW, Dukhinova M, Au C, Lau AY, et al. . Platelets Play Differential Role During the Initiation and Progression of Autoimmune Neuroinflammation. Circ Res (2015) 117(9):779–92. doi: 10.1161/CIRCRESAHA.115.306847
    1. Shyer JA, Flavell RA, Bailis W. Metabolic Signaling in T Cells. Cell Res (2020) 30(8):649–59. doi: 10.1038/s41422-020-0379-5
    1. Mohammadi P, di Iulio J, Munoz M, Martinez R, Bartha I, Cavassini M, et al. . Dynamics of HIV Latency and Reactivation in a Primary CD4+ T Cell Model. PLoS Pathog (2014) 10(5):e1004156. doi: 10.1371/journal.ppat.1004156
    1. Younes SA, Talla A, Pereira Ribeiro S, Saidakova EV, Korolevskaya LB, Shmagel KV, et al. . Cycling CD4+ T Cells in HIV-Infected Immune Nonresponders Have Mitochondrial Dysfunction. J Clin Invest (2018) 128(11):5083–94. doi: 10.1172/JCI120245
    1. Reimand J, Isserlin R, Voisin V, Kucera M, Tannus-Lopes C, Rostamianfar A, et al. . Pathway Enrichment Analysis and Visualization of Omics Data Using G:Profiler, GSEA, Cytoscape and EnrichmentMap. Nat Protoc (2019) 14(2):482–517. doi: 10.1038/s41596-018-0103-9
    1. Raudvere U, Kolberg L, Kuzmin I, Arak T, Adler P, Peterson H, et al. . G:Profiler: A Web Server for Functional Enrichment Analysis and Conversions of Gene Lists (2019 Update). Nucleic Acids Res (2019) 47(W1):W191–W8. doi: 10.1093/nar/gkz369
    1. Romero P, Wagg J, Green ML, Kaiser D, Krummenacker M, Karp PD. Computational Prediction of Human Metabolic Pathways From the Complete Human Genome. Genome Biol (2005) 6(1):R2. doi: 10.1186/gb-2004-6-1-r2
    1. Mathis D, Shoelson SE. Immunometabolism: An Emerging Frontier. Nat Rev Immunol (2011) 11(2):81. doi: 10.1038/nri2922
    1. Pearce EL, Poffenberger MC, Chang CH, Jones RG. Fueling Immunity: Insights Into Metabolism and Lymphocyte Function. Science (2013) 342(6155):1242454. doi: 10.1126/science.1242454
    1. Waickman AT, Powell JD. mTOR, Metabolism, and the Regulation of T-Cell Differentiation and Function. Immunol Rev (2012) 249(1):43–58. doi: 10.1111/j.1600-065X.2012.01152.x
    1. Palmer CS, Henstridge DC, Yu D, Singh A, Balderson B, Duette G, et al. . Emerging Role and Characterization of Immunometabolism: Relevance to HIV Pathogenesis, Serious Non-AIDS Events, and a Cure. J Immunol (2016) 196(11):4437–44. doi: 10.4049/jimmunol.1600120
    1. Gerin I, Bury M, Baldin F, Graff J, Van Schaftingen E, Bommer GT. Phosphoglycolate has Profound Metabolic Effects But Most Likely No Role in a Metabolic DNA Response in Cancer Cell Lines. Biochem J (2019) 476(4):629–43. doi: 10.1042/BCJ20180435
    1. Chou JP, Ramirez CM, Wu JE, Effros RB. Accelerated Aging in HIV/AIDS: Novel Biomarkers of Senescent Human CD8+ T Cells. PLoS One (2013) 8(5):e64702. doi: 10.1371/journal.pone.0064702
    1. Singh PK, Plumb MR, Ferris AL, Iben JR, Wu X, Fadel HJ, et al. . LEDGF/p75 Interacts With mRNA Splicing Factors and Targets HIV-1 Integration to Highly Spliced Genes. Genes Dev (2015) 29(21):2287–97. doi: 10.1101/gad.267609.115
    1. Chesney J, Mitchell R, Benigni F, Bacher M, Spiegel L, Al-Abed Y, et al. . An Inducible Gene Product for 6-Phosphofructo-2-Kinase With an AU-Rich Instability Element: Role in Tumor Cell Glycolysis and the Warburg Effect. Proc Natl Acad Sci USA (1999) 96(6):3047–52. doi: 10.1073/pnas.96.6.3047
    1. Wong KKL, Liao JZ, Verheyen EM. A Positive Feedback Loop Between Myc and Aerobic Glycolysis Sustains Tumor Growth in a Drosophila Tumor Model. Elife (2019) 8:1–19. doi: 10.7554/eLife.46315
    1. Thejer BM, Adhikary PP, Kaur A, Teakel SL, Van Oosterum A, Seth I, et al. . PGRMC1 Phosphorylation Affects Cell Shape, Motility, Glycolysis, Mitochondrial Form and Function, and Tumor Growth. BMC Mol Cell Biol (2020) 21(1):24. doi: 10.1186/s12860-020-00256-3
    1. Teperino R, Amann S, Bayer M, McGee SL, Loipetzberger A, Connor T, et al. . Hedgehog Partial Agonism Drives Warburg-Like Metabolism in Muscle and Brown Fat. Cell (2012) 151(2):414–26. doi: 10.1016/j.cell.2012.09.021
    1. Maloveryan A, Finta C, Osterlund T, Kogerman P. A Possible Role of Mouse Fused (STK36) in Hedgehog Signaling and Gli Transcription Factor Regulation. J Cell Commun Signal (2007) 1(3-4):165–73. doi: 10.1007/s12079-007-0014-y
    1. Menk AV, Scharping NE, Moreci RS, Zeng X, Guy C, Salvatore S, et al. . Early TCR Signaling Induces Rapid Aerobic Glycolysis Enabling Distinct Acute T Cell Effector Functions. Cell Rep (2018) 22(6):1509–21. doi: 10.1016/j.celrep.2018.01.040
    1. Buck MD, O’Sullivan D, Pearce EL. T Cell Metabolism Drives Immunity. J Exp Med (2015) 212(9):1345–60. doi: 10.1084/jem.20151159
    1. Dore GJ, Li Y, McDonald A, Ree H, Kaldor JM, National HIVSC. Impact of Highly Active Antiretroviral Therapy on Individual AIDS-Defining Illness Incidence and Survival in Australia. J Acquir Immune Defic Syndr (2002) 29(4):388–95. doi: 10.1097/00126334-200204010-00010
    1. Lewden C, Chene G, Morlat P, Raffi F, Dupon M, Dellamonica P, et al. . HIV-Infected Adults With a CD4 Cell Count Greater Than 500 Cells/Mm3 on Long-Term Combination Antiretroviral Therapy Reach Same Mortality Rates as the General Population. J Acquir Immune Defic Syndr (2007) 46(1):72–7. doi: 10.1097/QAI.0b013e318134257a
    1. Goehringer F, Bonnet F, Salmon D, Cacoub P, Paye A, Chene G, et al. . Causes of Death in HIV-Infected Individuals With Immunovirologic Success in a National Prospective Survey. AIDS Res Hum Retroviruses (2017) 33(2):187–93. doi: 10.1089/aid.2016.0222
    1. Grulich AE, van Leeuwen MT, Falster MO, Vajdic CM. Incidence of Cancers in People With HIV/AIDS Compared With Immunosuppressed Transplant Recipients: A Meta-Analysis. Lancet (2007) 370(9581):59–67. doi: 10.1016/S0140-6736(07)61050-2
    1. Casper C. The Increasing Burden of HIV-Associated Malignancies in Resource-Limited Regions. Annu Rev Med (2011) 62:157–70. doi: 10.1146/annurev-med-050409-103711
    1. Hasse B, Ledergerber B, Furrer H, Battegay M, Hirschel B, Cavassini M, et al. . Morbidity and Aging in HIV-Infected Persons: The Swiss HIV Cohort Study. Clin Infect Dis (2011) 53(11):1130–9. doi: 10.1093/cid/cir626
    1. Marin B, Thiebaut R, Bucher HC, Rondeau V, Costagliola D, Dorrucci M, et al. . Non-AIDS-Defining Deaths and Immunodeficiency in the Era of Combination Antiretroviral Therapy. AIDS (2009) 23(13):1743–53. doi: 10.1097/QAD.0b013e32832e9b78
    1. Lundgren JD, Baxter J, Deeks SG, Lane HC. Biomarkers in HIV Disease. Curr Opin HIV AIDS (2010) 5(6):459–62. doi: 10.1097/COH.0b013e32833f2ed6
    1. Cenderello G, De Maria A. Discordant Responses to cART in HIV-1 Patients in the Era of High Potency Antiretroviral Drugs: Clinical Evaluation, Classification, Management Prospects. Expert Rev Anti Infect Ther (2016) 14(1):29–40. doi: 10.1586/14787210.2016.1106937
    1. Carvalho-Silva WHV, Andrade-Santos JL, Souto FO, Coelho AVC, Crovella S, Guimaraes RL. Immunological Recovery Failure in cART-Treated HIV-Positive Patients is Associated With Reduced Thymic Output and RTE CD4+ T Cell Death by Pyroptosis. J Leukoc Biol (2020) 107(1):85–94. doi: 10.1002/JLB.4A0919-235R
    1. Sarma J, Laan CA, Alam S, Jha A, Fox KA, Dransfield I. Increased Platelet Binding to Circulating Monocytes in Acute Coronary Syndromes. Circulation (2002) 105(18):2166–71. doi: 10.1161/01.CIR.0000015700.27754.6F
    1. Li N, Ji Q, Hjemdahl P. Platelet-Lymphocyte Conjugation Differs Between Lymphocyte Subpopulations. J Thromb Haemost (2006) 4(4):874–81. doi: 10.1111/j.1538-7836.2006.01817.x
    1. Hansson GK. Inflammation, Atherosclerosis, and Coronary Artery Disease. N Engl J Med (2005) 352(16):1685–95. doi: 10.1056/NEJMra043430
    1. Semple JW, Italiano JE, Jr, Freedman J. Platelets and the Immune Continuum. Nat Rev Immunol (2011) 11(4):264–74. doi: 10.1038/nri2956
    1. Knijff-Dutmer EA, Koerts J, Nieuwland R, Kalsbeek-Batenburg EM, van de Laar MA. Elevated Levels of Platelet Microparticles are Associated With Disease Activity in Rheumatoid Arthritis. Arthritis Rheum (2002) 46(6):1498–503. doi: 10.1002/art.10312
    1. Wagner DD, Burger PC. Platelets in Inflammation and Thrombosis. Arterioscler Thromb Vasc Biol (2003) 23(12):2131–7. doi: 10.1161/
    1. Burger PC, Wagner DD. Platelet P-Selectin Facilitates Atherosclerotic Lesion Development. Blood (2003) 101(7):2661–6. doi: 10.1182/blood-2002-07-2209
    1. Simpson SR, Singh MV, Dewhurst S, Schifitto G, Maggirwar SB. Platelets Function as an Acute Viral Reservoir During HIV-1 Infection by Harboring Virus and T-Cell Complex Formation. Blood Adv (2020) 4(18):4512–21. doi: 10.1182/bloodadvances.2020002420
    1. Soto-Heredero G, Gomez de Las Heras MM, Gabande-Rodriguez E, Oller J, Mittelbrunn M. Glycolysis - a Key Player in the Inflammatory Response. FEBS J (2020) 287(16):3350–69. doi: 10.1111/febs.15327
    1. Cohn LB, Chomont N, Deeks SG. The Biology of the HIV-1 Latent Reservoir and Implications for Cure Strategies. Cell Host Microbe (2020) 27(4):519–30. doi: 10.1016/j.chom.2020.03.014
    1. Sadallah S, Amicarella F, Eken C, Iezzi G, Schifferli JA. Ectosomes Released by Platelets Induce Differentiation of CD4+T Cells Into T Regulatory Cells. Thromb Haemost (2014) 112(6):1219–29. doi: 10.1160/TH14-03-0281
    1. Pandiyan P, Younes SA, Ribeiro SP, Talla A, McDonald D, Bhaskaran N, et al. . Mucosal Regulatory T Cells and T Helper 17 Cells in HIV-Associated Immune Activation. Front Immunol (2016) 7:228. doi: 10.3389/fimmu.2016.00228
    1. Piconi S, Trabattoni D, Gori A, Parisotto S, Magni C, Meraviglia P, et al. . Immune Activation, Apoptosis, and Treg Activity are Associated With Persistently Reduced CD4+ T-Cell Counts During Antiretroviral Therapy. AIDS (2010) 24(13):1991–2000. doi: 10.1097/QAD.0b013e32833c93ce
    1. D’Ambrosi S, Nilsson RJ, Wurdinger T. Platelets and Tumor-Associated RNA Transfer. Blood (2021) 137(23):3181–91. doi: 10.1182/blood.2019003978
    1. Rowley JW, Schwertz H, Weyrich AS. Platelet mRNA: The Meaning Behind the Message. Curr Opin Hematol (2012) 19(5):385–91. doi: 10.1097/MOH.0b013e328357010e
    1. Mann M, Mehta A, Zhao JL, Lee K, Marinov GK, Garcia-Flores Y, et al. . An NF-kappaB-microRNA Regulatory Network Tunes Macrophage Inflammatory Responses. Nat Commun (2017) 8(1):851. doi: 10.1038/s41467-017-00972-z
    1. Rodriguez-Munoz A, Martinez-Hernandez R, Ramos-Levi AM, Serrano-Somavilla A, Gonzalez-Amaro R, Sanchez-Madrid F, et al. . Circulating Microvesicles Regulate Treg and Th17 Differentiation in Human Autoimmune Thyroid Disorders. J Clin Endocrinol Metab (2015) 100(12):E1531–9. doi: 10.1210/jc.2015-3146
    1. Rowley JW, Chappaz S, Corduan A, Chong MM, Campbell R, Khoury A, et al. . Dicer1-Mediated miRNA Processing Shapes the mRNA Profile and Function of Murine Platelets. Blood (2016) 127(14):1743–51. doi: 10.1182/blood-2015-07-661371
    1. Du C, Liu C, Kang J, Zhao G, Ye Z, Huang S, et al. . MicroRNA miR-326 Regulates TH-17 Differentiation and is Associated With the Pathogenesis of Multiple Sclerosis. Nat Immunol (2009) 10(12):1252–9. doi: 10.1038/ni.1798
    1. Podshivalova K, Salomon DR. MicroRNA Regulation of T-Lymphocyte Immunity: Modulation of Molecular Networks Responsible for T-Cell Activation, Differentiation, and Development. Crit Rev Immunol (2013) 33(5):435–76. doi: 10.1615/CritRevImmunol.2013006858
    1. Ple H, Landry P, Benham A, Coarfa C, Gunaratne PH, Provost P. The Repertoire and Features of Human Platelet microRNAs. PLoS One (2012) 7(12):e50746. doi: 10.1371/journal.pone.0050746
    1. Fink L, Holschermann H, Kwapiszewska G, Muyal JP, Lengemann B, Bohle RM, et al. . Characterization of Platelet-Specific mRNA by Real-Time PCR After Laser-Assisted Microdissection. Thromb Haemost (2003) 90(4):749–56. doi: 10.1160/TH03-02-0095
    1. Valle-Casuso JC, Angin M, Volant S, Passaes C, Monceaux V, Mikhailova A, et al. . Cellular Metabolism Is a Major Determinant of HIV-1 Reservoir Seeding in CD4(+) T Cells and Offers an Opportunity to Tackle Infection. Cell Metab (2019) 29(3):611–26.e5. doi: 10.1016/j.cmet.2018.11.015
    1. Clerc I, Moussa DA, Vahlas Z, Tardito S, Oburoglu L, Hope TJ, et al. . Entry of Glucose- and Glutamine-Derived Carbons Into the Citric Acid Cycle Supports Early Steps of HIV-1 Infection in CD4 T Cells. Nat Metab (2019) 1(7):717–30. doi: 10.1038/s42255-019-0084-1
    1. Hegedus A, Kavanagh Williamson M, Huthoff H. HIV-1 Pathogenicity and Virion Production are Dependent on the Metabolic Phenotype of Activated CD4+ T Cells. Retrovirology (2014) 11:98. doi: 10.1186/s12977-014-0098-4
    1. Sen S, Kaminiski R, Deshmane S, Langford D, Khalili K, Amini S, et al. . Role of Hexokinase-1 in the Survival of HIV-1-Infected Macrophages. Cell Cycle (2015) 14(7):980–9. doi: 10.1080/15384101.2015.1006971
    1. Castellano P, Prevedel L, Valdebenito S, Eugenin EA. HIV Infection and Latency Induce a Unique Metabolic Signature in Human Macrophages. Sci Rep (2019) 9(1):3941. doi: 10.1038/s41598-019-39898-5
    1. Palmer CS, Ostrowski M, Gouillou M, Tsai L, Yu D, Zhou J, et al. . Increased Glucose Metabolic Activity is Associated With CD4+ T-Cell Activation and Depletion During Chronic HIV Infection. AIDS (2014) 28(3):297–309. doi: 10.1097/QAD.0000000000000128
    1. Korencak M, Byrne M, Richter E, Schultz BT, Juszczak P, Ake JA, et al. . Effect of HIV Infection and Antiretroviral Therapy on Immune Cellular Functions. JCI Insight (2019) 4(12):1–14. doi: 10.1172/jci.insight.126675
    1. Arnoult D, Petit F, Lelievre JD, Estaquier J. Mitochondria in HIV-1-Induced Apoptosis. Biochem Biophys Res Commun (2003) 304(3):561–74. doi: 10.1016/S0006-291X(03)00629-6

Source: PubMed

3
S'abonner