HIV protease inhibitors do not cause the accumulation of prelamin A in PBMCs from patients receiving first line therapy: the ANRS EP45 "aging" study

Sophie Perrin, Jonathan Cremer, Olivia Faucher, Jacques Reynes, Pierre Dellamonica, Joëlle Micallef, Caroline Solas, Bruno Lacarelle, Charlotte Stretti, Elise Kaspi, Andrée Robaglia-Schlupp, Corinne Nicolino-Brunet, Catherine Tamalet, Nicolas Lévy, Isabelle Poizot-Martin, Pierre Cau, Patrice Roll, Sophie Perrin, Jonathan Cremer, Olivia Faucher, Jacques Reynes, Pierre Dellamonica, Joëlle Micallef, Caroline Solas, Bruno Lacarelle, Charlotte Stretti, Elise Kaspi, Andrée Robaglia-Schlupp, Corinne Nicolino-Brunet, Catherine Tamalet, Nicolas Lévy, Isabelle Poizot-Martin, Pierre Cau, Patrice Roll

Abstract

Background: The ANRS EP45 "Aging" study investigates the cellular mechanisms involved in the accelerated aging of HIV-1 infected and treated patients. The present report focuses on lamin A processing, a pathway known to be altered in systemic genetic progeroid syndromes.

Methods: 35 HIV-1 infected patients being treated with first line antiretroviral therapy (ART, mean duration at inclusion: 2.7±1.3 years) containing boosted protease inhibitors (PI/r) (comprising lopinavir/ritonavir in 65% of patients) were recruited together with 49 seronegative age- and sex-matched control subjects (https://ichgcp.net/clinical-trials-registry/NCT01038999" title="See in ClinicalTrials.gov">NCT01038999). In more than 88% of patients, the viral load was <40 copies/ml and the CD4+ cell count was >500/mm³. Prelamin A processing in peripheral blood mononuclear cells (PBMCs) from patients and controls was analysed by western blotting at inclusion. PBMCs from patients were also investigated at 12 and 24 months after enrolment in the study. PBMCs from healthy controls were also incubated with boosted lopinavir in culture medium containing various concentrations of proteins (4 to 80 g/L).

Results: Lamin A precursor was not observed in cohort patient PBMC regardless of the PI/r used, the dose and the plasma concentration. Prelamin A was detected in PBMC incubated in culture medium containing a low protein concentration (4 g/L) but not in plasma (60-80 g/L) or in medium supplemented with BSA (40 g/L), both of which contain a high protein concentration.

Conclusions: Prelamin A processing abnormalities were not observed in PBMCs from patients under the PI/r first line regimen. Therefore, PI/r do not appear to contribute to lamin A-related aging in PBMCs. In cultured PBMCs from healthy donors, prelamin A processing abnormalities were only observed when the protein concentration in the culture medium was low, thus increasing the amount of PI available to enter cells. ClinicalTrials.gov NCT01038999 https://ichgcp.net/clinical-trials-registry/NCT01038999.

Conflict of interest statement

Competing Interests: The authors have the following interests. PC was a recipient of grants from GlaxoSmithKline and from Boehringer Mannheim. There are no patents, products in development or marketed products to declare. This does not alter the authors' adherence to all the PLOS ONE policies on sharing data and materials, as detailed online in the guide for authors.

Figures

Figure 1. Lack of prelamin A in…
Figure 1. Lack of prelamin A in PBMCs from compliant patients receiving the 2NRTI+1PI/r regimen.
(A) Plasma concentrations of lopinavir, atazanavir and ritonavir from ANRS EP45 “Aging” patients reported at M0 (circles), M12 (square) and M24 (triangle). Colors indicate the durations between last drug uptake and venipuncture (Cmax, red; Cmin, green, Cintermediate, brown) and PI therapeutic concentration range (Cmax, red; Cmin, green). The majority of patients receiving lopinavir treatment were assayed at Cmin and remained within the therapeutic range. Samples from patients being treated with the atazanavir regimen were mainly assayed at mid-dose. (B) Representative western blots of PBMCs from patients receiving lopinavir or atazanavir treatment using three different lamin A/C-specific antibodies and one prelamin A-specific antibody. No prelamin A was detected in PBMCs from patients. Control PBMCs from healthy controls incubated with ZoPra were used as positive control cells. PI plasma concentrations in the patients shown: lopinavir 10.7 µM (M0), 12.3 µM (M12) and 9.3 µM (M24); atazanavir 1.5 µM (M0), 0.7 µM (M12) and 0 µM (M24).
Figure 2. The protein concentration of PI…
Figure 2. The protein concentration of PI incubation medium influences the effect of PI on prelamin A processing in PBMC.
PBMC were incubated for 24 hours with increasing concentrations of lopinavir (0, 2, 20, 40, 200 µM) diluted in plasma (total protein concentration: 60–80 g/L), in RPMI culture medium supplemented with 10% FBS and 2 mM L-glutamine (total protein concentration: 4 g/L), or in the RPMI culture medium supplemented with BSA (total protein concentration: 40 g/L). (A) Percentage of viable cells in plasma (white), culture medium (black), and BSA supplemented culture medium (gray) (error bars = SD, n ≥3, at least 200 cells counted in each experiment). **p<0.01. No viability changes were observed when PBMC were incubated in plasma containing lopinavir. A decrease in cell viability was apparent when PBMC were incubated in culture medium containing 20 µM or 40 µM PI. Only a slight decrease in viability was observed in cells incubated in BSA supplemented culture medium containing 200 µM PI. (B) Western blotting experiments. Both plasma and BSA supplemented culture medium containing 2 to 40 µM lopinavir had no effect on prelamin A maturation in PBMC. Prelamin A (anti-lamin A/C H110) was detected (*) in culture medium containing 20 µM and 40 µM lopinavir, and in BSA supplemented culture medium containing 200 µM lopinavir. Fibroblasts were cultured in the presence or absence of either 20 µM lopinavir (farnesylated prelamin A positive control) or 60 µM ZoPra (unfarnesylated prelamin A positive control) for 72 hours. GAPDH was used as total cellular protein loading control. (A) Western blot comparing the three lamin A/C antibodies used (N18, sc6215; H110, sc20681; Jol2, MAB3211). All antibodies recognized both lamin A and lamin C. Different amounts of farnesylated prelamin A were detected by N18 and H110, as shown by the ratio of prelamin A reported to the total prelamin A+lamin A signal. In the same conditions, Jol2 did not detect prelamin A. (B) Western blot comparing the two prelamin A antibodies tested (sc6214, ANT0045). The sc6214 antibody exhibited a higher affinity for both farnesylated and unfarnesylated prelamin A than the ANT0045 antibody. (C) Prelamin A, lamin A and lamin C protein domains and antibody epitopes. Lamin A/C N18 (sc6215, green); lamin A/C H110 (sc20681, blue); lamin A/C Jol2 (MAB3211, purple); prelamin A sc6214 (pink); prelamin A ANT0045 (orange).

References

    1. Lin F, Worman HJ (1993) Structural organization of the human gene encoding nuclear lamin A and nuclear lamin C. J Biol Chem. 268: 16321–16326.
    1. Broers JL, Ramaekers FC, Bonne G, Yaou RB, Hutchison CJ (2006) Nuclear lamins: laminopathies and their role in premature ageing. Physiol Rev 86: 967–1008.
    1. Dechat T, Pfleghaar K, Sengupta K, Shimi T, Shumaker DK, et al. (2008) Nuclear lamins: major factors in the structural organization and function of the nucleus and chromatin. Genes Dev 22: 832–853.
    1. Prokocimer M, Davidovich M, Nissim-Rafinia M, Wiesel-Motiuk N, Bar DZ, et al. (2009) Nuclear lamins: key regulators of nuclear structure and activities. J Cell Mol Med 13: 1059–1085.
    1. Lutz RJ, Trujillo MA, Denham KS, Wenger L, Sinensky M (1992) Nucleoplasmic localization of prelamin A: implications for prenylation-dependent lamin A assembly into the nuclear lamina. Proc Natl Acad Sci U S A 89: 3000–3004.
    1. Bergo MO, Gavino B, Ross J, Schmidt WK, Hong C, et al. (2002) Zmpste24 deficiency in mice causes spontaneous bone fractures, muscle weakness, and a prelamin A processing defect. Proc Natl Acad Sci U S A 99: 13049–13054.
    1. Corrigan DP, Kuszczak D, Rusinol AE, Thewke DP, Hrycyna CA, et al. (2005) Prelamin A endoproteolytic processing in vitro by recombinant Zmpste24. Biochem J 387: 129–138.
    1. Pendas AM, Zhou Z, Cadinanos J, Freije JM, Wang J, et al. (2002) Defective prelamin A processing and muscular and adipocyte alterations in Zmpste24 metalloproteinase-deficient mice. Nat Genet 31: 94–99.
    1. Andres V, Gonzalez JM (2009) Role of A-type lamins in signaling, transcription, and chromatin organization. Journal of Cell Biology 187: 945–957.
    1. Bonne G, Di Barletta MR, Varnous S, Becane HM, Hammouda EH, et al. (1999) Mutations in the gene encoding lamin A/C cause autosomal dominant Emery-Dreifuss muscular dystrophy. Nat Genet 21: 285–288.
    1. Fatkin D, MacRae C, Sasaki T, Wolff MR, Porcu M, et al. (1999) Missense mutations in the rod domain of the lamin A/C gene as causes of dilated cardiomyopathy and conduction-system disease. N Engl J Med 341: 1715–1724.
    1. Muchir A, Bonne G, van der Kooi AJ, van Meegen M, Baas F, et al. (2000) Identification of mutations in the gene encoding lamins A/C in autosomal dominant limb girdle muscular dystrophy with atrioventricular conduction disturbances (LGMD1B). Hum Mol Genet 9: 1453–1459.
    1. De Sandre-Giovannoli A, Chaouch M, Kozlov S, Vallat JM, Tazir M, et al. (2002) Homozygous defects in LMNA, encoding lamin A/C nuclear-envelope proteins, cause autosomal recessive axonal neuropathy in human (Charcot-Marie-Tooth disorder type 2) and mouse. Am J Hum Genet 70: 726–736.
    1. Shackleton S, Lloyd DJ, Jackson SN, Evans R, Niermeijer MF, et al. (2000) LMNA, encoding lamin A/C, is mutated in partial lipodystrophy. Nat Genet 24: 153–156.
    1. Decaudain A, Vantyghem MC, Guerci B, Hecart AC, Auclair M, et al. (2007) New metabolic phenotypes in laminopathies: LMNA mutations in patients with severe metabolic syndrome. Journal of Clinical Endocrinology and Metabolism 92: 4835–4844.
    1. Dutour A, Roll P, Gaborit B, Courrier S, Alessi MC, et al. (2011) High prevalence of laminopathies among patients with metabolic syndrome. Hum Mol Genet 20: 3779–3786.
    1. Eriksson M, Brown WT, Gordon LB, Glynn MW, Singer J, et al. (2003) Recurrent de novo point mutations in lamin A cause Hutchinson-Gilford progeria syndrome. Nature 423: 293–298.
    1. Navarro CL, Cau P, Levy N (2006) Molecular bases of progeroid syndromes. Hum Mol Genet 15 Suppl 2R151–161.
    1. Hennekam RC (2006) Hutchinson-Gilford progeria syndrome: review of the phenotype. Am J Med Genet A 140: 2603–2624.
    1. Navarro CL, De Sandre-Giovannoli A, Bernard R, Boccaccio I, Boyer A, et al. (2004) Lamin A and ZMPSTE24 (FACE-1) defects cause nuclear disorganization and identify restrictive dermopathy as a lethal neonatal laminopathy. Hum Mol Genet 13: 2493–2503.
    1. Navarro CL, Cadinanos J, De Sandre-Giovannoli A, Bernard R, Courrier S, et al. (2005) Loss of ZMPSTE24 (FACE-1) causes autosomal recessive restrictive dermopathy and accumulation of Lamin A precursors. Hum Mol Genet 14: 1503–1513.
    1. Scaffidi P, Misteli T (2006) Lamin A-dependent nuclear defects in human aging. Science 312: 1059–1063.
    1. Ragnauth CD, Warren DT, Liu Y, McNair R, Tajsic T, et al. (2010) Prelamin A acts to accelerate smooth muscle cell senescence and is a novel biomarker of human vascular aging. Circulation 121: 2200–2210.
    1. Deeks SG, Phillips AN (2009) HIV infection, antiretroviral treatment, ageing, and non-AIDS related morbidity. Bmj 338: a3172.
    1. Deeks SG (2011) HIV infection, inflammation, immunosenescence, and aging. Annu Rev Med 62: 141–155.
    1. Garg A (2011) Clinical review#: Lipodystrophies: genetic and acquired body fat disorders. J Clin Endocrinol Metab 96: 3313–3325.
    1. Guaraldi G, Orlando G, Zona S, Menozzi M, Carli F, et al. (2011) Premature age-related comorbidities among HIV-infected persons compared with the general population. Clin Infect Dis 53: 1120–1126.
    1. Scott ES, O'Hare P (2001) Fate of the inner nuclear membrane protein lamin B receptor and nuclear lamins in herpes simplex virus type 1 infection. J Virol 75: 8818–8830.
    1. Muranyi W, Haas J, Wagner M, Krohne G, Koszinowski UH (2002) Cytomegalovirus recruitment of cellular kinases to dissolve the nuclear lamina. Science 297: 854–857.
    1. de Noronha CM, Sherman MP, Lin HW, Cavrois MV, Moir RD, et al. (2001) Dynamic disruptions in nuclear envelope architecture and integrity induced by HIV-1 Vpr. Science 294: 1105–1108.
    1. Coffinier C, Hudon SE, Farber EA, Chang SY, Hrycyna CA, et al. (2007) HIV protease inhibitors block the zinc metalloproteinase ZMPSTE24 and lead to an accumulation of prelamin A in cells. Proc Natl Acad Sci U S A 104: 13432–13437.
    1. Coffinier C, Hudon SE, Lee R, Farber EA, Nobumori C, et al. (2008) A potent HIV protease inhibitor, darunavir, does not inhibit ZMPSTE24 or lead to an accumulation of farnesyl-prelamin A in cells. Journal of Biological Chemistry 283: 9797–9804.
    1. Liu Q, Kim DI, Syme J, LuValle P, Burke B, et al. (2010) Dynamics of lamin-A processing following precursor accumulation. PLoS ONE 5: e10874.
    1. Lefevre C, Auclair M, Boccara F, Bastard JP, Capeau J, et al. (2010) Premature senescence of vascular cells is induced by HIV protease inhibitors: implication of prelamin A and reversion by statin. Arteriosclerosis, Thrombosis, and Vascular Biology 30: 2611–2620.
    1. Miranda M, Chacon MR, Vidal F, Megia A, Richart C, et al. (2007) LMNA messenger RNA expression in highly active antiretroviral therapy-treated HIV-positive patients. J Acquir Immune Defic Syndr 46: 384–389.
    1. Caron M, Auclair M, Donadille B, Bereziat V, Guerci B, et al. (2007) Human lipodystrophies linked to mutations in A-type lamins and to HIV protease inhibitor therapy are both associated with prelamin A accumulation, oxidative stress and premature cellular senescence. Cell Death Differ 14: 1759–1767.
    1. Bereziat V, Cervera P, Le Dour C, Verpont MC, Dumont S, et al. (2011) LMNA mutations induce a non-inflammatory fibrosis and a brown fat-like dystrophy of enlarged cervical adipose tissue. Am J Pathol 179: 2443–2453.
    1. Perrin S, Cremer J, Roll P, Faucher O, Menard A, et al. (2012) HIV-1 Infection and First Line ART Induced Differential Responses in Mitochondria from Blood Lymphocytes and Monocytes: The ANRS EP45 “Aging” Study. PLoS One 7: e41129.
    1. Justesen US, Pedersen C, Klitgaard NA (2003) Simultaneous quantitative determination of the HIV protease inhibitors indinavir, amprenavir, ritonavir, lopinavir, saquinavir, nelfinavir and the nelfinavir active metabolite M8 in plasma by liquid chromatography. J Chromatogr B Analyt Technol Biomed Life Sci 783: 491–500.
    1. Colombo S, Guignard N, Marzolini C, Telenti A, Biollaz J, et al. (2004) Determination of the new HIV-protease inhibitor atazanavir by liquid chromatography after solid-phase extraction. J Chromatogr B Analyt Technol Biomed Life Sci 810: 25–34.
    1. Marzolini C, Beguin A, Telenti A, Schreyer A, Buclin T, et al. (2002) Determination of lopinavir and nevirapine by high-performance liquid chromatography after solid-phase extraction: application for the assessment of their transplacental passage at delivery. J Chromatogr B Analyt Technol Biomed Life Sci 774: 127–140.
    1. Marsh KC, Eiden E, McDonald E (1997) Determination of ritonavir, a new HIV protease inhibitor, in biological samples using reversed-phase high-performance liquid chromatography. J Chromatogr B Biomed Sci Appl 704: 307–313.
    1. van Heeswijk RP, Hoetelmans RM, Harms R, Meenhorst PL, Mulder JW, et al. (1998) Simultaneous quantitative determination of the HIV protease inhibitors amprenavir, indinavir, nelfinavir, ritonavir and saquinavir in human plasma by ion-pair high-performance liquid chromatography with ultraviolet detection. J Chromatogr B Biomed Sci Appl 719: 159–168.
    1. Quaranta S, Woloch C, Paccou A, Giocanti M, Solas C, et al. (2009) Validation of an electrospray ionization LC-MS/MS method for quantitative analysis of raltegravir, etravirine, and 9 other antiretroviral agents in human plasma samples. Ther Drug Monit 31: 695–702.
    1. Varela I, Pereira S, Ugalde AP, Navarro CL, Suarez MF, et al. (2008) Combined treatment with statins and aminobisphosphonates extends longevity in a mouse model of human premature aging. Nat Med 14: 767–772.
    1. Caron M, Auclair M, Sterlingot H, Kornprobst M, Capeau J (2003) Some HIV protease inhibitors alter lamin A/C maturation and stability, SREBP-1 nuclear localization and adipocyte differentiation. Aids 17: 2437–2444.
    1. Ghosh AK, Chapsal BD, Weber IT, Mitsuya H (2008) Design of HIV protease inhibitors targeting protein backbone: an effective strategy for combating drug resistance. Acc Chem Res 41: 78–86.
    1. Hudon SE, Coffinier C, Michaelis S, Fong LG, Young SG, et al. (2008) HIV-protease inhibitors block the enzymatic activity of purified Ste24p. Biochemical and Biophysical Research Communications 374: 365–368.
    1. Yeni (2010) Prise en charge médicale des personnes infectées par le VIH. ISBN : 978–2-11–008038–7. Available: . Accessed: 2012 Jun 21.
    1. Arab-Alameddine M, Decosterd LA, Buclin T, Telenti A, Csajka C (2011) Antiretroviral drug toxicity in relation to pharmacokinetics, metabolic profile and pharmacogenetics. Expert Opin Drug Metab Toxicol.
    1. Bazzoli C, Jullien V, Le Tiec C, Rey E, Mentre F, et al. (2010) Intracellular Pharmacokinetics of Antiretroviral Drugs in HIV-Infected Patients, and their Correlation with Drug Action. Clin Pharmacokinet 49: 17–45.
    1. Ford J, Khoo SH, Back DJ (2004) The intracellular pharmacology of antiretroviral protease inhibitors. J Antimicrob Chemother 54: 982–990.
    1. Boffito M, Back DJ, Blaschke TF, Rowland M, Bertz RJ, et al. (2003) Protein binding in antiretroviral therapies. AIDS Res Hum Retroviruses 19: 825–835.
    1. Janneh O, Hartkoorn RC, Jones E, Owen A, Ward SA, et al. (2008) Cultured CD4T cells and primary human lymphocytes express hOATPs: intracellular accumulation of saquinavir and lopinavir. Br J Pharmacol 155: 875–883.
    1. Hartkoorn RC, Kwan WS, Shallcross V, Chaikan A, Liptrott N, et al. (2010) HIV protease inhibitors are substrates for OATP1A2, OATP1B1 and OATP1B3 and lopinavir plasma concentrations are influenced by SLCO1B1 polymorphisms. Pharmacogenet Genomics 20: 112–120.
    1. Turriziani O, Gianotti N, Falasca F, Boni A, Vestri AR, et al. (2008) Expression levels of MDR1, MRP1, MRP4, and MRP5 in peripheral blood mononuclear cells from HIV infected patients failing antiretroviral therapy. J Med Virol 80: 766–771.
    1. Oldfield V, Plosker GL (2006) Lopinavir/ritonavir: a review of its use in the management of HIV infection. Drugs 66: 1275–1299.
    1. Mukhopadhyay A, Wei B, Zullo SJ, Wood LV, Weiner H (2002) In vitro evidence of inhibition of mitochondrial protease processing by HIV-1 protease inhibitors in yeast: a possible contribution to lipodystrophy syndrome. Mitochondrion 1: 511–518.
    1. Lafeuillade A, Solas C, Halfon P, Chadapaud S, Hittinger G, et al. (2002) Differences in the detection of three HIV-1 protease inhibitors in non-blood compartments: clinical correlations. HIV Clin Trials 3: 27–35.
    1. Di Mascio M, Srinivasula S, Bhattacharjee A, Cheng L, Martiniova L, et al. (2009) Antiretroviral Tissue Kinetics: In Vivo Imaging Using Positron Emission Tomography. Antimicrob Agents Chemother 53: 4089–4095.
    1. Cohen J (2011) HIV/AIDS research. Tissue says blood is misleading, confusing HIV cure efforts. Science 334: 1614.
    1. Patterson KB, Prince HA, Kraft E, Jenkins AJ, Shaheen NJ, et al. (2011) Penetration of Tenofovir and Emtricitabine in Mucosal Tissues: Implications for Prevention of HIV-1 Transmission. Science Translational Medicine 3: 112re114–112re114.
    1. Prot M, Heripret L, Cardot-Leccia N, Perrin C, Aouadi M, et al. (2006) Long-term treatment with lopinavir-ritonavir induces a reduction in peripheral adipose depots in mice. Antimicrob Agents Chemother 50: 3998–4004.
    1. Bastard JP, Caron M, Vidal H, Jan V, Auclair M, et al. (2002) Association between altered expression of adipogenic factor SREBP1 in lipoatrophic adipose tissue from HIV-1-infected patients and abnormal adipocyte differentiation and insulin resistance. Lancet 359: 1026–1031.
    1. Caron M, Auclair M, Vigouroux C, Glorian M, Forest C, et al. (2001) The HIV protease inhibitor indinavir impairs sterol regulatory element-binding protein-1 intranuclear localization, inhibits preadipocyte differentiation, and induces insulin resistance. Diabetes 50: 1378–1388.
    1. Kudlow BA, Jameson SA, Kennedy BK (2005) HIV protease inhibitors block adipocyte differentiation independently of lamin A/C. Aids 19: 1565–1573.
    1. Boguslavsky RL, Stewart CL, Worman HJ (2006) Nuclear lamin A inhibits adipocyte differentiation: implications for Dunnigan-type familial partial lipodystrophy. Hum Mol Genet 15: 653–663.

Source: PubMed

3
구독하다