HIV-1 infection and first line ART induced differential responses in mitochondria from blood lymphocytes and monocytes: the ANRS EP45 "Aging" study

Sophie Perrin, Jonathan Cremer, Patrice Roll, Olivia Faucher, Amélie Ménard, Jacques Reynes, Pierre Dellamonica, Alissa Naqvi, Joëlle Micallef, Elisabeth Jouve, Catherine Tamalet, Caroline Solas, Christel Pissier, Isabelle Arnoux, Corine Nicolino-Brunet, Léon Espinosa, Nicolas Lévy, Elise Kaspi, Andrée Robaglia-Schlupp, Isabelle Poizot-Martin, Pierre Cau, Sophie Perrin, Jonathan Cremer, Patrice Roll, Olivia Faucher, Amélie Ménard, Jacques Reynes, Pierre Dellamonica, Alissa Naqvi, Joëlle Micallef, Elisabeth Jouve, Catherine Tamalet, Caroline Solas, Christel Pissier, Isabelle Arnoux, Corine Nicolino-Brunet, Léon Espinosa, Nicolas Lévy, Elise Kaspi, Andrée Robaglia-Schlupp, Isabelle Poizot-Martin, Pierre Cau

Abstract

Background: The ANRS EP45 "Aging" study investigates the cellular mechanisms involved in the accelerated aging of HIV-1 infected and treated patients. The data reported focus on mitochondria, organelles known to be involved in cell senescence.

Methods: 49 HIV-1 infected patients untreated with antiretroviral therapy, together with 49 seronegative age- and sex-matched control subjects and 81 HIV-1 infected and treated patients, were recruited by 3 AIDS centres (Marseille, Montpellier, Nice; France; https://ichgcp.net/clinical-trials-registry/NCT01038999" title="See in ClinicalTrials.gov">NCT01038999). In more than 88% of treated patients, the viral load was <40 copies/ml and the CD4+ cell count was >500/mm(3). ROS (reactive oxygen species) production and ΔΨm (inner membrane potential) were measured by flow cytometry in blood lymphocytes and monocytes (functional parameters). Three mitochondrial network quantitative morphological parameters were computed using confocal microscopy and image analysis. Three PBMC mitochondrial proteins (porin and subunits 2 and 4 of cytochrome C oxidase encoded by mtDNA or nuclear DNA, respectively) were analysed by western blotting.

Results: Quantitative changes in PBMC mitochondrial proteins were not induced by either HIV-1 infection or ART. Discriminant analysis integrating functional (ROS production and ΔΨm) or morphological (network volume density, fragmentation and branching) parameters revealed HIV-1 infection and ART differential effects according to cell type. First line ART tended to rescue lymphocyte mitochondrial parameters altered by viral infection, but induced slight changes in monocytes. No statistical difference was found between the effects of three ART regimens on mitochondrial parameters. Correlations between functional parameters and viral load confirmed the damaging effects of HIV-1 in lymphocyte mitochondria.

Conclusions: In patients considered to be clinically stable, mitochondria exhibited functional and morphological modifications in PBMCs resulting from either direct or indirect effects of HIV-1 infection (lymphocytes), or from first line ART (monocytes). Together with other tissue impairments, these changes may contribute to global aging.

Conflict of interest statement

Competing Interests: GlaxoSmithKline and Boehringen Mannheim grants were received. This does not alter the authors’ adherence to all PLoS ONE policies on sharing data and materials. The funding was not related to employment, consultancy, patents and product in development or marketed products.

Figures

Figure 1. Flow cytometric analysis of ROS…
Figure 1. Flow cytometric analysis of ROS production revealed two subpopulations of both CD14+ monocytes and lymphocytes.
(A to C) Monocytes. (A) CD14+/SSC monocyte gating. (B) Representative zebra plot based on side scatter and ROS production showing low- and high-ROS producing cell subpopulations. (C) Histogram illustrating ROS production as a function of cell count (low-ROS production, gray; high-ROS production, red). ROS production levels (MFI ± SD) are indicated as percentages relative to CD14+ monocytes. The percentages of high-ROS monocytes were not statistically different between the groups: Control: 75.8±21.7%; ART naive: 70.7±19.4%; 2NRTI+1PI/r: 71.1±21.1%; 2NRTI+1NNRTI: 71.0±22.8%; 3NRTI: 66.2±23.7%. (D to F) Lymphocytes. (D) FSC/SSC lymphocyte gating. (E) Representative zebra plot based on side scatter and ROS production showing low- and high-ROS producing cell subpopulations. (F) Histogram illustrating ROS production as a function of cell count (low-ROS production, gray; high-ROS production, red). ROS production levels (MFI ± SD) are indicated as percentages relative to FSC/SSC-selected lymphocytes. The percentages of high-ROS lymphocytes were not statistically different between the groups: Control: 55.4±12.7%; ART naive: 48.0±14.1%; 2NRTI+1PI/r: 55.1±13.8%; 2NRTI+1NNRTI: 50.1±14.1%; 3NRTI: 47.6±15.0%. ROS production by the low- and high-ROS subpopulations was statistically different in both HIV and ART patients.
Figure 2. Determination of mitochondrial inner membrane…
Figure 2. Determination of mitochondrial inner membrane potential (ΔΨm) in lymphocytes and monocytes.
JC-1 is a cationic carbocyanine probe that exibits a potential-dependent accumulation in mitochondria as either a monomer at low concentrations (green fluorescence) or as aggregates at higher concentrations (red fluorescence) . Therefore, the red/green fluorescence intensity ratio illustrates ΔΨm. (A to C) Monocytes. (A) CD14+/SSC monocyte gating. (B) Representative zebra plot based on side scatter and ΔΨm. (C) Representative zebra plot based on side scatter and ΔΨm after inhibition of ΔΨm with CCCP (control experiment). (D to F) Lymphocytes. (D) FSC/SSC lymphocyte gating. (E) Representative zebra plot based on side scatter and ΔΨm. Two lymphocyte subpopulations were distinguished (low-ΔΨm, bottom right quadrants; high-ΔΨm, top right quadrants). The top and bottom right quadrants indicate the ΔΨm mean values (cell percentages). The percentages of low-ΔΨm lymphocytes were: Control: 14.7±8.2%; ART naive: 12.56±4.4% (statistically different from control); 2NRTI+1PI/r: 17.0±8.4%; 2NRTI+1NNRTI: 15.3±7.3%; 3NRTI: 16.3±5.1%. (F) Representative zebra plot based on side scatter and ΔΨm after inhibition of ΔΨm with CCCP (control experiment).
Figure 3. Mitochondrial functional parameters: differential effects…
Figure 3. Mitochondrial functional parameters: differential effects of HIV-1 infection and ART on lymphocytes and monocytes.
(A, E) Discriminant analysis using ROS and ΔΨm parameters. The five cohort populations are delineated by their 95% confidence circles around the means. The perpendicular axes describe the combined variance of the parameters analysed (lymphocytes: 77.26%, monocytes: 92.25%). The contributions of each parameter to the variances on the X and Y axes are shown in the insets. (A) Lymphocytes. ART naive patients are statistically different to control subjects. ART partially rescues mitochondrial parameter changes induced by HIV infection. Inset: The 8 mitochondrial parameters used were: a: Basal ROS production by high-ROS lymphocytes (see Figure 3B). b: PMA/Basal ratio of ROS production by high-ROS lymphocytes (see Figure 3C). cHigh: ΔΨm of high-ΔΨm lymphocytes. cLow: ΔΨm of low-ΔΨm lymphocytes (see Figure 3D). cLow%: Percentage of low-ΔΨm lymphocytes. dHigh: Basal to CCCP ΔΨm ratio of high-ΔΨm lymphocytes. dLow: Basal to CCCP ΔΨm ratio of low-ΔΨm lymphocytes. dLow%: CCCP to Basal ratio of low-ΔΨm lymphocyte percentage. (E) Monocytes. ART naive patients are close to control subjects whereas ART combinations are more dispersed. Inset: The 4 mitochondrial parameters used were: a: Basal ROS production by high-ROS monocytes (see Figure 3F). b: PMA to Basal ratio of ROS production by high-ROS monocytes (see Figure 3G). c: ΔΨm of monocytes (see Figure 3H). d: Basal to CCCP ΔΨm ratio of monocytes. (B to D; F to H) Box plots (XLSTAT) displaying 1st quartile (Q1), median, mean (displayed by ‘+’), 3rd quartile (Q3) together with both lower and upper limits were calculated as follows: lower limit = Xi such that {Xi – [Q1–1.5 (Q3– Q1)]} is the minimum and Xi ≥ Q1–1.5 (Q3– Q1); upper limit = Yi such that { Yi – [Q3+1.5 (Q3– Q1)]} is the minimum and Yi ≤ Q3+1.5 (Q3– Q1). *p<0,05, **p<0,01.
Figure 4. Correlation between mitochondrial functional parameters…
Figure 4. Correlation between mitochondrial functional parameters and viral load in cells from ART naive patients.
(A) ROS production by high-ROS lymphocytes (r = 0.355, p = 0.029, n = 38). (B) ΔΨm of high-ΔΨm lymphocytes (r = −0.417, p = 0.007, n = 40). (C) Percentage of low-ΔΨm lymphocytes (r = 0.457, p = 0.003, n = 40). (D) ROS production by high-ROS monocytes (r = 0.164, p = 0.288, n = 44) (E) ΔΨm of monocytes (r = −0.455, p = 0.003, n = 40).
Figure 5. Mitochondrial morphological parameters: differential effects…
Figure 5. Mitochondrial morphological parameters: differential effects of HIV-1 infection and ART on lymphocytes and monocytes.
(A, E) Discriminant analysis using three mitochondrial morphological parameters. The five cohort populations are delineated by their 95% confidence circles around the means. The two perpendicular axes describe the combined variance of the parameters analysed (lymphocytes: 90.66%, monocytes: 98.73%). The contributions of each parameter to the variances on the X and Y axes are shown in the insets. Inset: The 3 mitochondrial parameters used were: Vv%: Volume density (lymphocytes: see Figure 5B; monocytes: see Figure 5F). Frag: Fragmentation (lymphocytes: see Figure 5C; monocytes: see Figure 5G). Bran: Branching (lymphocytes: see Figure 5D; monocytes: see Figure 5H). (A) Lymphocytes ART naive patients and control subjects are statistically different. Mitochondrial changes induced by HIV infection are partially reduced by ART. (E) Monocytes ART naive patients exhibit no variations compared to control subjects, while significant differences are observed for the three ART groups. (B to D; F to H) Box plots: *p<0,05, **p<0,01.

References

    1. Guaraldi G, Orlando G, Zona S, Menozzi M, Carli F, et al. Premature Age-Related Comorbidities Among HIV-Infected Persons Compared With the General Population. Clin Infect Dis. 2011;53:1120–1126.
    1. Hamanaka RB, Chandel NS. Mitochondrial reactive oxygen species regulate cellular signaling and dictate biological outcomes. Trends in Biochemical Sciences. 2010;35:505–513.
    1. Scott I. The role of mitochondria in the mammalian antiviral defense system. Mitochondrion. 2010;10:316–320.
    1. Wallace DC. A mitochondrial paradigm of metabolic and degenerative diseases, aging, and cancer: a dawn for evolutionary medicine. Annu Rev Genet. 2005;39:359–407.
    1. Fu W, Sanders-Beer BE, Katz KS, Maglott DR, Pruitt KD, et al. Human immunodeficiency virus type 1, human protein interaction database at NCBI. Nucleic Acids Res. 2009;37:D417–422.
    1. Narayanan A, Kehn-Hall K, Bailey C, Kashanchi F. Analysis of the roles of HIV-derived microRNAs. Expert Opin Biol Ther. 2011;11:17–29.
    1. Pinti M, Salomoni P, Cossarizza A. Anti-HIV drugs and the mitochondria. Biochimica et Biophysica Acta (BBA) - Bioenergetics. 2006;1757:700–707.
    1. Cote HC. Mechanisms of antiretroviral therapy-induced mitochondrial dysfunction. Curr Opin HIV AIDS. 2007;2:253–260.
    1. Cummins NW, Badley AD. Mechanisms of HIV-associated lymphocyte apoptosis: 2010. Cell Death Dis. 2010;1:e99.
    1. Ma YS, Wu SB, Lee WY, Cheng JS, Wei YH. Response to the increase of oxidative stress and mutation of mitochondrial DNA in aging. Biochim Biophys Acta. 2009;1790:1021–1029.
    1. Forman HJ, Maiorino M, Ursini F. Signaling functions of reactive oxygen species. Biochemistry. 2010;49:835–842.
    1. Poyton RO, Ball KA, Castello PR. Mitochondrial generation of free radicals and hypoxic signaling. Trends in Endocrinology & Metabolism. 2009;20:332–340.
    1. Mammucari C, Rizzuto R. Signaling pathways in mitochondrial dysfunction and aging. Mech Ageing Dev. 2010;131:536–543.
    1. Benard G, Karbowski M. Mitochondrial fusion and division: Regulation and role in cell viability. Semin Cell Dev Biol. 2009;20:365–374.
    1. Campello S, Scorrano L. Mitochondrial shape changes: orchestrating cell pathophysiology. EMBO Rep. 2010;11:678–684.
    1. Malena A, Loro E, Di Re M, Holt IJ, Vergani L. Inhibition of mitochondrial fission favours mutant over wild-type mitochondrial DNA. Hum Mol Genet. 2009;18:3407–3416.
    1. Jezek P, Plecita-Hlavata L. Mitochondrial reticulum network dynamics in relation to oxidative stress, redox regulation, and hypoxia. Int J Biochem Cell Biol. 2009;41:1790–1804.
    1. McCoy MK, Cookson MR. DJ-1 regulation of mitochondrial function and autophagy through oxidative stress. Autophagy. 2011;7:5.
    1. Koshiba T, Yasukawa K, Yanagi Y, Kawabata S. Mitochondrial membrane potential is required for MAVS-mediated antiviral signaling. Sci Signal. 2011;4:ra7.
    1. Wang C, Liu X, Wei B. Mitochondrion: an emerging platform critical for host antiviral signaling. Expert Opin Ther Targets. 2011;15:647–665.
    1. Solis M, Nakhaei P, Jalalirad M, Lacoste J, Douville R, et al. RIG-I Mediated Antiviral Signalling is Inhibited in HIV-1 Infection by a Protease-Mediated Sequestration of RIG-I. J Virol. 2011;85:1224–1236.
    1. Klase Z, Winograd R, Davis J, Carpio L, Hildreth R, et al. HIV-1 TAR miRNA protects against apoptosis by altering cellular gene expression. Retrovirology. 2009;6:18.
    1. Circu ML, Aw TY. Reactive oxygen species, cellular redox systems, and apoptosis. Free Radic Biol Med. 2010;48:749–762.
    1. Karamchand L, Dawood H, Chuturgoon AA. Lymphocyte mitochondrial depolarization and apoptosis in HIV-1-infected HAART patients. J Acquir Immune Defic Syndr. 2008;48:381–388.
    1. Perfettini JL, Roumier T, Kroemer G. Mitochondrial fusion and fission in the control of apoptosis. Trends Cell Biol. 2005;15:179–183.
    1. Koczor CA, Lewis W. Nucleoside reverse transcriptase inhibitor toxicity and mitochondrial DNA. Expert Opin Drug Metab Toxicol. 2010;6:1493–1504.
    1. Payne BA, Wilson IJ, Hateley CA, Horvath R, Santibanez-Koref M, et al. Mitochondrial aging is accelerated by anti-retroviral therapy through the clonal expansion of mtDNA mutations. Nat Genet. 2011;43:806–810.
    1. Cossarizza A, Salvioli S. Analysis of Mitochondrial Membrane Potential with the Sensitive Fluorescent Probe JC-1. ISBN: 1-890473-03-0. Purdue cytometry CD ROOM Vol 4. 1998. Available . Accessed: 13 March 2012.
    1. Huberty CJ, Olejnik S editors. Applied MANOVA and discriminant analysis. 2006. Second edition ed. Hoboken (NJ): Wiley-Interscience. 488 p.
    1. Deeks SG. HIV infection, inflammation, immunosenescence, and aging. Annu Rev Med. 2011;62:141–155.
    1. Margineantu DH, Emerson CB, Diaz D, Hockenbery DM. Hsp90 inhibition decreases mitochondrial protein turnover. PLoS One. 2007;2:e1066.
    1. Narendra DP, Youle RJ. Targeting Mitochondrial Dysfunction: Role for PINK1 and Parkin in Mitochondrial Quality Control. Antioxid Redox Signal. 2011;14:1929–1938.
    1. Azzu V, Brand MD. Degradation of an intramitochondrial protein by the cytosolic proteasome. J Cell Sci. 2010;123:578–585.
    1. Livnat-Levanon N, Glickman MH. Ubiquitin-proteasome system and mitochondria - reciprocity. Biochim Biophys Acta. 2011;1809:80–87.
    1. Ciccosanti F, Corazzari M, Soldani F, Matarrese P, Pagliarini V, et al. Proteomic analysis identifies prohibitin down-regulation as a crucial event in the mitochondrial damage observed in HIV-infected patients. Antivir Ther. 2010;15:377–390.
    1. Acin-Perez R, Bayona-Bafaluy MP, Fernandez-Silva P, Moreno-Loshuertos R, Perez-Martos A, et al. Respiratory complex III is required to maintain complex I in mammalian mitochondria. Mol Cell. 2004;13:805–815.
    1. Diaz F, Fukui H, Garcia S, Moraes CT. Cytochrome c oxidase is required for the assembly/stability of respiratory complex I in mouse fibroblasts. Mol Cell Biol. 2006;26:4872–4881.
    1. Fitzgerald-Bocarsly P, Dai J, Singh S. Plasmacytoid dendritic cells and type I IFN: 50 years of convergent history. Cytokine Growth Factor Rev. 2008;19:3–19.
    1. Zhao C, Zhang H, Wong WC, Sem X, Han H, et al. Identification of novel functional differences in monocyte subsets using proteomic and transcriptomic methods. J Proteome Res. 2009;8:4028–4038.
    1. Zhao C, Tan YC, Wong WC, Sem X, Zhang H, et al. The CD14(+/low)CD16(+) monocyte subset is more susceptible to spontaneous and oxidant-induced apoptosis than the CD14(+)CD16(-) subset. Cell Death Dis. 2010;1:e95.
    1. Trachootham D, Lu W, Ogasawara MA, Nilsa RD, Huang P. Redox regulation of cell survival. Antioxid Redox Signal. 2008;10:1343–1374.
    1. Lugli E, Troiano L, Ferraresi R, Roat E, Prada N, et al. Characterization of cells with different mitochondrial membrane potential during apoptosis. Cytometry A. 2005;68:28–35.
    1. Mandas A, Iorio EL, Congiu MG, Balestrieri C, Mereu A, et al. Oxidative imbalance in HIV-1 infected patients treated with antiretroviral therapy. J Biomed Biotechnol. 2009;2009:749575.
    1. Sternfeld T, Schmid M, Tischleder A, Mudra S, Schlamp A, et al. The influence of HIV infection and antiretroviral therapy on the mitochondrial membrane potential of peripheral mononuclear cells. Antivir Ther. 2007;12:769–778.
    1. Sternfeld T, Tischleder A, Schuster M, Bogner JR. Mitochondrial membrane potential and apoptosis of blood mononuclear cells in untreated HIV-1 infected patients. HIV Med. 2009;10:512–519.
    1. Romani B, Engelbrecht S. Human immunodeficiency virus type 1 Vpr: functions and molecular interactions. J Gen Virol. 2009;90:1795–1805.
    1. Fausther-Bovendo H, Vieillard V, Sagan S, Bismuth G, Debre P. HIV gp41 engages gC1qR on CD4+ T cells to induce the expression of an NK ligand through the PIP3/H2O2 pathway. PLoS Pathog. 2010;6:e1000975.
    1. Jolly C, Sattentau QJ. Regulated secretion from CD4+ T cells. Trends Immunol. 2007;28:474–481.
    1. Herbein G, Gras G, Khan KA, Abbas W. Macrophage signaling in HIV-1 infection. Retrovirology. 2010;7:34.
    1. Klatt NR, Silvestri G. CD4+ T Cells and HIV: A Paradoxical Pas de Deux. Sci Transl Med. 2012;4:123ps124.
    1. Muratori C, Cavallin LE, Kratzel K, Tinari A, De Milito A, et al. Massive secretion by T cells is caused by HIV Nef in infected cells and by Nef transfer to bystander cells. Cell Host Microbe. 2009;6:218–230.
    1. Lenassi M, Cagney G, Liao M, Vaupotic T, Bartholomeeusen K, et al. HIV Nef is secreted in exosomes and triggers apoptosis in bystander CD4+ T cells. Traffic. 2010;11:110–122.
    1. Sun G, Rossi JJ. MicroRNAs and their potential involvement in HIV infection. Trends Pharmacol Sci. 2011;32:675–681.
    1. Xu W, Santini PA, Sullivan JS, He B, Shan M, et al. HIV-1 evades virus-specific IgG2 and IgA responses by targeting systemic and intestinal B cells via long-range intercellular conduits. Nat Immunol. 2009;10:1008–1017.
    1. Lee HC, Wei YH. Mitochondrial biogenesis and mitochondrial DNA maintenance of mammalian cells under oxidative stress. Int J Biochem Cell Biol. 2005;37:822–834.
    1. Castanier C, Garcin D, Vazquez A, Arnoult D. Mitochondrial dynamics regulate the RIG-I-like receptor antiviral pathway. EMBO reports. 2010;11:133–138.
    1. Berg RK, Melchjorsen J, Rintahaka J, Diget E, Soby S, et al. Genomic HIV RNA induces innate immune responses through RIG-I-dependent sensing of secondary-structured RNA. PLoS One. 2012;7:e29291.
    1. Elbim C, Pillet S, Prevost MH, Preira A, Girard PM, et al. Redox and activation status of monocytes from human immunodeficiency virus-infected patients: relationship with viral load. J Virol. 1999;73:4561–4566.
    1. Kadiu I, Wang T, Schlautman JD, Dubrovsky L, Ciborowski P, et al. HIV-1 transforms the monocyte plasma membrane proteome. Cellular Immunology. 2009;258:44–58.
    1. Robinson JM. Phagocytic leukocytes and reactive oxygen species. Histochem Cell Biol. 2009;131:465–469.
    1. Reiss M, Roos D. Differences in oxygen metabolism of phagocytosing monocytes and neutrophils. J Clin Invest. 1978;61:480–488.
    1. Nakamura H, Masutani H, Yodoi J. Redox imbalance and its control in HIV infection. Antioxid Redox Signal. 2002;4:455–464.
    1. Stehbens WE. Oxidative stress in viral hepatitis and AIDS. Exp Mol Pathol. 2004;77:121–132.
    1. Jones SP, Qazi N, Morelese J, Lebrecht D, Sutinen J, et al. Assessment of adipokine expression and mitochondrial toxicity in HIV patients with lipoatrophy on stavudine- and zidovudine-containing regimens. J Acquir Immune Defic Syndr. 2005;40:565–572.
    1. McComsey GA, Kang M, Ross AC, Lebrecht D, Livingston E, et al. Increased mtDNA levels without change in mitochondrial enzymes in peripheral blood mononuclear cells of infants born to HIV-infected mothers on antiretroviral therapy. HIV Clin Trials. 2008;9:126–136.
    1. Cossarizza A, Riva A, Pinti M, Ammannato S, Fedeli P, et al. Increased mitochondrial DNA content in peripheral blood lymphocytes from HIV-infected patients with lipodystrophy. Antivir Ther. 2003;8:315–321.
    1. Montaner JS, Cote HC, Harris M, Hogg RS, Yip B, et al. Mitochondrial toxicity in the era of HAART: evaluating venous lactate and peripheral blood mitochondrial DNA in HIV-infected patients taking antiretroviral therapy. J Acquir Immune Defic Syndr. 2003;34:S85–90.
    1. Vidal F, Domingo JC, Guallar J, Saumoy M, Cordobilla B, et al. In vitro cytotoxicity and mitochondrial toxicity of tenofovir alone and in combination with other antiretrovirals in human renal proximal tubule cells. Antimicrob Agents Chemother. 2006;50:3824–3832.
    1. Maagaard A, Kvale D. Mitochondrial toxicity in HIV-infected patients both off and on antiretroviral treatment: a continuum or distinct underlying mechanisms? Journal of Antimicrobial Chemotherapy. 2009;64:901–909.
    1. Morse CG, Voss JG, Rakocevic G, McLaughlin M, Vinton CL, et al. HIV Infection and Antiretroviral Therapy Have Divergent Effects on Mitochondria in Adipose Tissue. J Infect Dis. 2012;205:1778–1787.
    1. Mukhopadhyay A, Wei B, Zullo SJ, Wood LV, Weiner H. In vitro evidence of inhibition of mitochondrial protease processing by HIV-1 protease inhibitors in yeast: a possible contribution to lipodystrophy syndrome. Mitochondrion. 2002;1:511–518.
    1. Pajonk F, Himmelsbach J, Riess K, Sommer A, McBride WH. The human immunodeficiency virus (HIV)-1 protease inhibitor saquinavir inhibits proteasome function and causes apoptosis and radiosensitization in non-HIV-associated human cancer cells. Cancer Res. 2002;62:5230–5235.
    1. Piccinini M, Rinaudo MT, Chiapello N, Ricotti E, Baldovino S, et al. The human 26S proteasome is a target of antiretroviral agents. Aids. 2002;16:693–700.
    1. Groener JB, Seybold U, Vollbrecht T, Bogner JR. Short Communication: Decrease in Mitochondrial Transmembrane Potential in Peripheral Blood Mononuclear Cells of HIV-Uninfected Subjects Undergoing HIV Postexposure Prophylaxis. AIDS Res Hum Retroviruses. 2011;27:969–972.
    1. Lopez S, Negredo E, Garrabou G, Puig J, Ruiz L, et al. Longitudinal study on mitochondrial effects of didanosine-tenofovir combination. AIDS Res Hum Retroviruses. 2006;22:33–39.
    1. Maggiolo F, Roat E, Pinti M, Nasi M, Gibellini L, et al. Mitochondrial changes during D-drug-containing once-daily therapy in HIV-positive treatment-naive patients. Antivir Ther. 2010;15:51–59.
    1. Einsiedel L, Cherry CL, Sheeran FL, Friedhuber A, Wesselingh SL, et al. Mitochondrial dysfunction in CD4+ lymphocytes from stavudine-treated HIV patients. Mitochondrion. 2010;10:534–539.
    1. Petrovas C, Mueller YM, Dimitriou ID, Altork SR, Banerjee A, et al. Increased mitochondrial mass characterizes the survival defect of HIV-specific CD8(+) T cells. Blood. 2007;109:2505–2513.
    1. Caron M, Auclair M, Lagathu C, Lombes A, Walker UA, et al. The HIV-1 nucleoside reverse transcriptase inhibitors stavudine and zidovudine alter adipocyte functions in vitro. Aids. 2004;18:2127–2136.
    1. Apostolova N, Gomez-Sucerquia LJ, Moran A, Alvarez A, Blas-Garcia A, et al. Enhanced oxidative stress and increased mitochondrial mass during efavirenz-induced apoptosis in human hepatic cells. Br J Pharmacol. 2010;160:2069–2084.
    1. Gerschenson M, Kim C, Berzins B, Taiwo B, Libutti DE, et al. Mitochondrial function, morphology and metabolic parameters improve after switching from stavudine to a tenofovir-containing regimen. Journal of Antimicrobial Chemotherapy. 2009;63:1244–1250.
    1. Curran A, Ribera E. From old to new nucleoside reverse transcriptase inhibitors: changes in body fat composition, metabolic parameters and mitochondrial toxicity after the switch from thymidine analogs to tenofovir or abacavir. Expert Opin Drug Saf. 2011;10:389–406.
    1. Martin AM, Almeida CA, Cameron P, Purcell AW, Nolan D, et al. Immune responses to abacavir in antigen-presenting cells from hypersensitive patients. Aids. 2007;21:1233–1244.
    1. Danaher RJ, Kaetzel CS, Greenberg RN, Wang C, Bruno ME, et al. HIV protease inhibitors alter innate immune response signaling to double-stranded RNA in oral epithelial cells: implications for immune reconstitution inflammatory syndrome? Aids. 2010;24:2587–2590.
    1. Coleman CM, Wu L. HIV interactions with monocytes and dendritic cells: viral latency and reservoirs. Retrovirology. 2009;6:51.
    1. Le Douce V, Herbein G, Rohr O, Schwartz C. Molecular mechanisms of HIV-1 persistence in the monocyte-macrophage lineage. Retrovirology. 2010;7:32.
    1. Bergamaschi A, Pancino G. Host hindrance to HIV-1 replication in monocytes and macrophages. Retrovirology. 2010;7:31.
    1. Kaushik R, Zhu X, Stranska R, Wu Y, Stevenson M. A cellular restriction dictates the permissivity of nondividing monocytes/macrophages to lentivirus and gammaretrovirus infection. Cell Host Microbe. 2009;6:68–80.
    1. Laguette N, Sobhian B, Casartelli N, Ringeard M, Chable-Bessia C, et al. SAMHD1 is the dendritic- and myeloid-cell-specific HIV-1 restriction factor counteracted by Vpx. Nature. 2011;474:654–657.
    1. Whitelaw DM. Observations on human monocyte kinetics after pulse labeling. Cell Tissue Kinet. 1972;5:311–317.
    1. Carter CC, McNamara LA, Onafuwa-Nuga A, Shackleton M, Riddell Jt, et al. HIV-1 utilizes the CXCR4 chemokine receptor to infect multipotent hematopoietic stem and progenitor cells. Cell Host Microbe. 2011;9:223–234.
    1. Alexaki A, Wigdahl B. HIV-1 infection of bone marrow hematopoietic progenitor cells and their role in trafficking and viral dissemination. PLoS Pathog. 2008;4:e1000215.
    1. Onafuwa-Nuga A, McNamara LA, Collins KL. Towards a cure for HIV: the identification and characterization of HIV reservoirs in optimally treated people. Cell Res. 2010;20:1185–1187.
    1. Garrabou G, López S, Morén C, Martínez E, Fontdevila J, et al. Mitochondrial damage in adipose tissue of untreated HIV-infected patients. Aids. 2011;25:165–170.
    1. Gavegnano C, Schinazi RF. Antiretroviral therapy in macrophages: implication for HIV eradication. Antivir Chem Chemother. 2009;20:63–78.
    1. Liu X, Ma Q, Zhang F. Therapeutic drug monitoring in highly active antiretroviral therapy. Expert Opin Drug Saf. 2010;9:743–758.
    1. Wei YH, Wu SB, Ma YS, Lee HC. Respiratory function decline and DNA mutation in mitochondria, oxidative stress and altered gene expression during aging. Chang Gung Med J. 2009;32:113–132.

Source: PubMed

3
Abonner