F. Lin and H. Worman, Structural organization of the human gene encoding nuclear lamin A and nuclear lamin C, J Biol Chem, vol.268, pp.16321-16326, 1993.

J. Broers, F. Ramaekers, G. Bonne, R. Yaou, and C. Hutchison, Nuclear Lamins: Laminopathies and Their Role in Premature Ageing, Physiological Reviews, vol.86, issue.3, pp.967-1008, 2006.
DOI : 10.1152/physrev.00047.2005

URL : http://physrev.physiology.org/content/physrev/86/3/967.full.pdf

T. Dechat, K. Pfleghaar, K. Sengupta, T. Shimi, and D. Shumaker, Nuclear lamins: major factors in the structural organization and function of the nucleus and chromatin, Genes & Development, vol.22, issue.7, pp.832-853, 2008.
DOI : 10.1101/gad.1652708

M. Prokocimer, M. Davidovich, M. Nissim-rafinia, N. Wiesel-motiuk, and D. Bar, Nuclear lamins: key regulators of nuclear structure and activities, Journal of Cellular and Molecular Medicine, vol.17, issue.6, pp.1059-1085, 2009.
DOI : 10.1002/ajmg.a.31803

R. Lutz, M. Trujillo, K. Denham, L. Wenger, and M. Sinensky, Nucleoplasmic localization of prelamin A: implications for prenylation-dependent lamin A assembly into the nuclear lamina., Proceedings of the National Academy of Sciences, vol.89, issue.7, pp.3000-3004, 1992.
DOI : 10.1073/pnas.89.7.3000

M. Bergo, B. Gavino, R. J. Schmidt, W. Hong, and C. , Zmpste24 deficiency in mice causes spontaneous bone fractures, muscle weakness, and a prelamin A processing defect, Proceedings of the National Academy of Sciences, vol.102, issue.4, pp.13049-13054, 2002.
DOI : 10.1002/ajmg.1463

D. Corrigan, D. Kuszczak, A. Rusinol, D. Thewke, and C. Hrycyna, by recombinant Zmpste24, Biochemical Journal, vol.387, issue.1, pp.129-138, 2005.
DOI : 10.1042/BJ20041359

A. Pendas, Z. Zhou, J. Cadinanos, J. Freije, and J. Wang, Defective prelamin A processing and muscular and adipocyte alterations in Zmpste24 metalloproteinase???deficient mice, Nature Genetics, vol.31, pp.94-99, 2002.
DOI : 10.1038/ng871

V. Andres and J. Gonzalez, Role of A-type lamins in signaling, transcription, and chromatin organization, The Journal of Cell Biology, vol.270, issue.7, pp.945-957, 2009.
DOI : 10.1016/j.bbrc.2005.10.020

G. Bonne, D. Barletta, M. Varnous, S. Becane, H. Hammouda et al., Mutations in the gene encoding lamin A/C cause autosomal dominant Emery-Dreifuss muscular dystrophy, Nature Genetics, vol.21, issue.3, pp.285-288, 1999.
DOI : 10.1038/6799

D. Fatkin, C. Macrae, T. Sasaki, M. Wolff, and M. Porcu, Missense Mutations in the Rod Domain of the Lamin A/C Gene as Causes of Dilated Cardiomyopathy and Conduction-System Disease, New England Journal of Medicine, vol.341, issue.23, pp.1715-1724, 1999.
DOI : 10.1056/NEJM199912023412302

A. Muchir, G. Bonne, A. Van-der-kooi, M. Van-meegen, and F. Baas, Identification of mutations in the gene encoding lamins A/C in autosomal dominant limb girdle muscular dystrophy with atrioventricular conduction disturbances (LGMD1B), Human Molecular Genetics, vol.9, issue.9, pp.1453-1459, 2000.
DOI : 10.1093/hmg/9.9.1453

D. Sandre-giovannoli, A. Chaouch, M. Kozlov, S. Vallat, J. Tazir et al., 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, The American Journal of Human Genetics, vol.70, issue.3, pp.726-736, 2002.
DOI : 10.1086/339274

S. Shackleton, D. Lloyd, S. Jackson, R. Evans, and M. Niermeijer, LMNA, encoding lamin A/C, is mutated in partial lipodystrophy, Nature Genetics, vol.112, issue.2, pp.153-156, 2000.
DOI : 10.1046/j.1523-1747.1999.00613.x

A. Decaudain, M. Vantyghem, B. Guerci, A. Hecart, and M. Auclair, Mutations in Patients with Severe Metabolic Syndrome, The Journal of Clinical Endocrinology & Metabolism, vol.92, issue.12, pp.4835-4844, 2007.
DOI : 10.1210/jc.2007-0654

A. Dutour, P. Roll, B. Gaborit, S. Courrier, and M. Alessi, High prevalence of laminopathies among patients with metabolic syndrome, Human Molecular Genetics, vol.20, issue.19, pp.3779-3786, 2011.
DOI : 10.1093/hmg/ddr294

M. Eriksson, W. Brown, L. Gordon, M. Glynn, and J. Singer, Recurrent de novo point mutations in lamin A cause Hutchinson???Gilford progeria syndrome, Nature, vol.66, issue.6937, pp.293-298, 2003.
DOI : 10.1086/302836

C. Navarro, P. Cau, and N. Levy, Molecular bases of progeroid syndromes, Human Molecular Genetics, vol.15, issue.suppl_2, pp.151-161, 2006.
DOI : 10.1093/hmg/ddl214

R. Hennekam, Hutchinson???Gilford progeria syndrome: Review of the phenotype, American Journal of Medical Genetics Part A, vol.81, issue.23, pp.2603-2624, 2006.
DOI : 10.1016/S1388-1981(02)00342-6

C. Navarro, D. Sandre-giovannoli, A. Bernard, R. Boccaccio, I. Boyer et al., Lamin A and ZMPSTE24 (FACE-1) defects cause nuclear disorganization and identify restrictive dermopathy as a lethal neonatal laminopathy, Human Molecular Genetics, vol.13, issue.20, pp.2493-2503, 2004.
DOI : 10.1093/hmg/ddh265

URL : https://hal.archives-ouvertes.fr/hal-01668977

C. Navarro, J. Cadinanos, D. Sandre-giovannoli, A. Bernard, R. Courrier et al., Loss of ZMPSTE24 (FACE-1) causes autosomal recessive restrictive dermopathy and accumulation of Lamin A precursors, Human Molecular Genetics, vol.14, issue.11, pp.1503-1513, 2005.
DOI : 10.1093/hmg/ddi159

P. Scaffidi and T. Misteli, Lamin A-Dependent Nuclear Defects in Human Aging, Science, vol.312, issue.5776, pp.1059-1063, 2006.
DOI : 10.1126/science.1127168

C. Ragnauth, D. Warren, Y. Liu, R. Mcnair, and T. Tajsic, Prelamin A Acts to Accelerate Smooth Muscle Cell Senescence and Is a Novel Biomarker of Human Vascular Aging, Circulation, vol.121, issue.20, pp.2200-2210, 2010.
DOI : 10.1161/CIRCULATIONAHA.109.902056

S. Deeks and A. Phillips, HIV infection, antiretroviral treatment, ageing, and non-AIDS related morbidity, BMJ, vol.338, issue.jan26 2, p.3172, 2009.
DOI : 10.1136/bmj.a3172

S. Deeks, HIV Infection, Inflammation, Immunosenescence, and Aging, Annual Review of Medicine, vol.62, issue.1, pp.141-155, 2011.
DOI : 10.1146/annurev-med-042909-093756

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3759035/pdf

A. Garg, Lipodystrophies: Genetic and Acquired Body Fat Disorders, The Journal of Clinical Endocrinology & Metabolism, vol.96, issue.11, pp.3313-3325, 2011.
DOI : 10.1210/jc.2011-1159

G. Guaraldi, G. Orlando, S. Zona, M. Menozzi, and F. Carli, Premature Age-Related Comorbidities Among HIV-Infected Persons Compared With the General Population, Clinical Infectious Diseases, vol.53, issue.11, pp.1120-1126, 2011.
DOI : 10.1093/cid/cir627

URL : https://academic.oup.com/cid/article-pdf/53/11/1120/865121/cir627.pdf

E. Scott, O. Hare, and P. , Fate of the Inner Nuclear Membrane Protein Lamin B Receptor and Nuclear Lamins in Herpes Simplex Virus Type 1 Infection, Journal of Virology, vol.75, issue.18, pp.8818-8830, 2001.
DOI : 10.1128/JVI.75.18.8818-8830.2001

W. Muranyi, J. Haas, M. Wagner, G. Krohne, and U. Koszinowski, Cytomegalovirus Recruitment of Cellular Kinases to Dissolve the Nuclear Lamina, Science, vol.297, issue.5582, pp.854-857, 2002.
DOI : 10.1126/science.1071506

C. De-noronha, M. Sherman, H. Lin, M. Cavrois, and R. Moir, Dynamic Disruptions in Nuclear Envelope Architecture and Integrity Induced by HIV-1 Vpr, Science, vol.294, issue.5544, pp.1105-1108, 2001.
DOI : 10.1126/science.1063957

C. Coffinier, S. Hudon, E. Farber, S. Chang, and C. Hrycyna, HIV protease inhibitors block the zinc metalloproteinase ZMPSTE24 and lead to an accumulation of prelamin A in cells, Proceedings of the National Academy of Sciences, vol.22, issue.22, pp.13432-13437, 2007.
DOI : 10.1093/emboj/cdg571

C. Coffinier, S. Hudon, R. Lee, E. Farber, and C. Nobumori, 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, vol.67, issue.15, pp.9797-9804, 2008.
DOI : 10.2165/00003495-200767180-00010

Q. Liu, D. Kim, J. Syme, P. Luvalle, and B. Burke, Dynamics of Lamin-A Processing Following Precursor Accumulation, PLoS ONE, vol.272, issue.5, p.10874, 2010.
DOI : 10.1371/journal.pone.0010874.s004

C. Lefevre, M. Auclair, F. Boccara, J. Bastard, and J. Capeau, Premature Senescence of Vascular Cells Is Induced by HIV Protease Inhibitors: Implication of Prelamin A and Reversion by Statin, Arteriosclerosis, Thrombosis, and Vascular Biology, vol.30, issue.12, pp.2611-2620, 2010.
DOI : 10.1161/ATVBAHA.110.213603

M. Miranda, M. Chacon, F. Vidal, A. Megia, and C. Richart, LMNA Messenger RNA Expression in Highly Active Antiretroviral Therapy-Treated HIV-Positive Patients, JAIDS Journal of Acquired Immune Deficiency Syndromes, vol.46, issue.4, pp.384-389, 2007.
DOI : 10.1097/QAI.0b013e31815aba1b

URL : http://pdfs.journals.lww.com/jaids/2007/12010/LMNA_Messenger_RNA_Expression_in_Highly_Active.2.pdf?token=method|ExpireAbsolute;source|Journals;ttl|1506172169439;payload|mY8D3u1TCCsNvP5E421JYK6N6XICDamxByyYpaNzk7FKjTaa1Yz22MivkHZqjGP4kdS2v0J76WGAnHACH69s21Csk0OpQi3YbjEMdSoz2UhVybFqQxA7lKwSUlA502zQZr96TQRwhVlocEp/sJ586aVbcBFlltKNKo+tbuMfL73hiPqJliudqs17cHeLcLbV/CqjlP3IO0jGHlHQtJWcICDdAyGJMnpi6RlbEJaRheGeh5z5uvqz3FLHgPKVXJzdgN4iEYs15JjLUNJhcn8FAfBRzzRprlnqj3VHUky7nc4=;hash|9bEbHdtPWqVbtHugjUP0zQ==

M. Caron, M. Auclair, B. Donadille, V. Bereziat, and B. Guerci, 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 and Differentiation, vol.20, issue.10, pp.1759-1767, 2007.
DOI : 10.1006/excr.2000.4823

V. Bereziat, P. Cervera, L. Dour, C. Verpont, M. Dumont et al., LMNA Mutations Induce a Non-Inflammatory Fibrosis and a Brown Fat-Like Dystrophy of Enlarged Cervical Adipose Tissue, The American Journal of Pathology, vol.179, issue.5, pp.2443-2453, 2011.
DOI : 10.1016/j.ajpath.2011.07.049

S. Perrin, J. Cremer, P. Roll, O. Faucher, and A. Menard, HIV-1 Infection and First Line ART Induced Differential Responses in Mitochondria from Blood Lymphocytes and Monocytes: The ANRS EP45 ???Aging??? Study, PLoS ONE, vol.32, issue.7, p.41129, 2012.
DOI : 10.1371/journal.pone.0041129.s009

URL : https://hal.archives-ouvertes.fr/hal-01646453

U. Justesen, C. Pedersen, and N. Klitgaard, 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, Journal of Chromatography B, vol.783, issue.2, pp.491-500, 2003.
DOI : 10.1016/S1570-0232(02)00728-6

S. Colombo, N. Guignard, C. Marzolini, A. Telenti, and J. Biollaz, Determination of the new HIV-protease inhibitor atazanavir by liquid chromatography after solid-phase extraction, Journal of Chromatography B, vol.810, issue.1, pp.25-34, 2004.
DOI : 10.1016/S1570-0232(04)00580-X

C. Marzolini, A. Beguin, A. Telenti, A. Schreyer, and T. Buclin, Determination of lopinavir and nevirapine by high-performance liquid chromatography after solid-phase extraction: application for the assessment of their transplacental passage at delivery, Journal of Chromatography B, vol.774, issue.2, pp.127-140, 2002.
DOI : 10.1016/S1570-0232(02)00169-1

K. Marsh, E. Eiden, and E. Mcdonald, Determination of ritonavir, a new HIV protease inhibitor, in biological samples using reversed-phase high-performance liquid chromatography, Journal of Chromatography B: Biomedical Sciences and Applications, vol.704, issue.1-2, pp.307-313, 1997.
DOI : 10.1016/S0378-4347(97)00454-4

R. Van-heeswijk, R. Hoetelmans, R. Harms, P. Meenhorst, and J. Mulder, 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, Journal of Chromatography B: Biomedical Sciences and Applications, vol.719, issue.1-2, 1998.
DOI : 10.1016/S0378-4347(98)00392-2

S. Quaranta, C. Woloch, A. Paccou, M. Giocanti, and C. Solas, Validation of an Electrospray Ionization LC-MS/MS Method for Quantitative Analysis of Raltegravir, Etravirine, and 9 Other Antiretroviral Agents in Human Plasma Samples, Therapeutic Drug Monitoring, vol.31, issue.6, pp.695-702, 2009.
DOI : 10.1097/FTD.0b013e3181c05adf

I. Varela, S. Pereira, A. Ugalde, C. Navarro, and M. Suarez, Combined treatment with statins and aminobisphosphonates extends longevity in a mouse model of human premature aging, Nature Medicine, vol.154, issue.7, pp.767-772, 2008.
DOI : 10.1172/JCI200422087

URL : https://hal.archives-ouvertes.fr/inserm-00376425

M. Caron, M. Auclair, H. Sterlingot, M. Kornprobst, and J. Capeau, Some HIV protease inhibitors alter lamin A/C maturation and stability, SREBP-1 nuclear localization and adipocyte differentiation, AIDS, vol.17, issue.17, pp.2437-2444, 2003.
DOI : 10.1097/00002030-200311210-00005

URL : http://pdfs.journals.lww.com/aidsonline/2003/11210/Some_HIV_protease_inhibitors_alter_lamin_A_C.5.pdf?token=method|ExpireAbsolute;source|Journals;ttl|1504538561413;payload|mY8D3u1TCCsNvP5E421JYK6N6XICDamxByyYpaNzk7FKjTaa1Yz22MivkHZqjGP4kdS2v0J76WGAnHACH69s21Csk0OpQi3YbjEMdSoz2UhVybFqQxA7lKwSUlA502zQZr96TQRwhVlocEp/sJ586aVbcBFlltKNKo+tbuMfL73hiPqJliudqs17cHeLcLbV/CqjlP3IO0jGHlHQtJWcICDdAyGJMnpi6RlbEJaRheGeh5z5uvqz3FLHgPKVXJzdN2ziiHhY6wPF9tfA80junqMD2wtE6oUb5qXHb0AcZKI=;hash|wBK1qG/5HQ70n2XA2WzxXQ==

A. Ghosh, B. Chapsal, I. Weber, and H. Mitsuya, Design of HIV Protease Inhibitors Targeting Protein Backbone: An Effective Strategy for Combating Drug Resistance, Accounts of Chemical Research, vol.41, issue.1, pp.78-86, 2008.
DOI : 10.1021/ar7001232

S. Hudon, C. Coffinier, S. Michaelis, L. Fong, and S. Young, HIV-protease inhibitors block the enzymatic activity of purified Ste24p, HIVprotease inhibitors block the enzymatic activity of purified Ste24p, pp.365-368, 2008.
DOI : 10.1016/j.bbrc.2008.07.033

. Yeni, Prise en charge médicale des personnes infectées par le VIH Available: http://www.sante.gouv.fr/rapport-2010-sur-la- prise-en-charge-medicale-des-personnes-infectees-par, pp.978-980, 2010.

M. Arab-alameddine, L. Decosterd, T. Buclin, A. Telenti, and C. Csajka, Antiretroviral drug toxicity in relation to pharmacokinetics, metabolic profile and pharmacogenetics, Expert Opinion on Drug Metabolism & Toxicology, vol.9, issue.5, 2011.
DOI : 10.1038/clpt.2009.12

C. Bazzoli, V. Jullien, L. Tiec, C. Rey, E. Mentre et al., Intracellular Pharmacokinetics of Antiretroviral Drugs in HIV-Infected Patients, and their Correlation with Drug Action, Clinical Pharmacokinetics, vol.41, issue.11, pp.17-45, 2010.
DOI : 10.1111/j.1468-1293.2008.00513.x

URL : https://hal.archives-ouvertes.fr/inserm-00461125

J. Ford, S. Khoo, and D. Back, The intracellular pharmacology of antiretroviral protease inhibitors, Journal of Antimicrobial Chemotherapy, vol.54, issue.6, pp.982-990, 2004.
DOI : 10.1093/jac/dkh487

M. Boffito, D. Back, T. Blaschke, R. M. Bertz, and R. , Protein Binding in Antiretroviral Therapies, AIDS Research and Human Retroviruses, vol.19, issue.9, pp.825-835, 2003.
DOI : 10.1089/088922203769232629

O. Janneh, R. Hartkoorn, E. Jones, A. Owen, and S. Ward, Cultured CD4T cells and primary human lymphocytes express hOATPs: intracellular accumulation of saquinavir and lopinavir, British Journal of Pharmacology, vol.19, issue.Suppl 2, pp.875-883, 2008.
DOI : 10.1038/bjp.2008.320

R. Hartkoorn, W. Kwan, V. Shallcross, A. Chaikan, and N. Liptrott, HIV protease inhibitors are substrates for OATP1A2, OATP1B1 and OATP1B3 and lopinavir plasma concentrations are influenced by SLCO1B1 polymorphisms, Pharmacogenetics and Genomics, vol.20, issue.2, pp.112-120, 2010.
DOI : 10.1097/FPC.0b013e328335b02d

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4859410/pdf

O. Turriziani, N. Gianotti, F. Falasca, A. Boni, and A. Vestri, Expression levels of MDR1, MRP1, MRP4, and MRP5 in peripheral blood mononuclear cells from HIV infected patients failing antiretroviral therapy, Journal of Medical Virology, vol.35, issue.5, pp.766-771, 2008.
DOI : 10.1002/jmv.21152

V. Oldfield and G. Plosker, Lopinavir/Ritonavir, Drugs, vol.62, issue.2, pp.1275-1299, 2006.
DOI : 10.2165/00003495-200666090-00012

A. Mukhopadhyay, B. Wei, S. Zullo, L. Wood, and H. Weiner, In vitro evidence of inhibition of mitochondrial protease processing by HIV-1 protease inhibitors in yeast: a possible contribution to lipodystrophy syndrome, Mitochondrion, vol.1, issue.6, pp.511-518, 2002.
DOI : 10.1016/S1567-7249(02)00042-9

A. Lafeuillade, C. Solas, P. Halfon, S. Chadapaud, and G. Hittinger, Differences in the detection of three HIV-1 protease inhibitors in non-blood compartments: Clinical correlations, HIV Clinical Trials, vol.14, issue.1, pp.27-35, 2002.
DOI : 10.1086/319864

D. Mascio, M. Srinivasula, S. Bhattacharjee, A. Cheng, L. Martiniova et al., Antiretroviral Tissue Kinetics: In Vivo Imaging Using Positron Emission Tomography, Antimicrobial Agents and Chemotherapy, vol.53, issue.10, pp.4089-4095, 2009.
DOI : 10.1128/AAC.00419-09

J. Cohen, Tissue Says Blood Is Misleading, Confusing HIV Cure Efforts, Science, vol.334, issue.6063, p.1614, 2011.
DOI : 10.1126/science.334.6063.1614

K. Patterson, H. Prince, E. Kraft, A. Jenkins, and N. Shaheen, Penetration of Tenofovir and Emtricitabine in Mucosal Tissues: Implications for Prevention of HIV-1 Transmission, Science Translational Medicine, vol.40, issue.9, pp.112-114, 2011.
DOI : 10.1086/527387

M. Prot, L. Heripret, N. Cardot-leccia, C. Perrin, and M. Aouadi, Long-Term Treatment with Lopinavir-Ritonavir Induces a Reduction in Peripheral Adipose Depots in Mice, Antimicrobial Agents and Chemotherapy, vol.50, issue.12, pp.3998-4004, 2006.
DOI : 10.1128/AAC.00625-06

J. Bastard, M. Caron, H. Vidal, J. V. Auclair, and M. , Association between altered expression of adipogenic factor SREBP1 in lipoatrophic adipose tissue from HIV-1-infected patients and abnormal adipocyte differentiation and insulin resistance, The Lancet, vol.359, issue.9311, pp.1026-1031, 2002.
DOI : 10.1016/S0140-6736(02)08094-7

M. Caron, M. Auclair, C. Vigouroux, M. Glorian, and C. Forest, The HIV Protease Inhibitor Indinavir Impairs Sterol Regulatory Element-Binding Protein-1 Intranuclear Localization, Inhibits Preadipocyte Differentiation, and Induces Insulin Resistance, Diabetes, vol.50, issue.6, pp.1378-1388, 2001.
DOI : 10.2337/diabetes.50.6.1378

URL : http://diabetes.diabetesjournals.org/content/diabetes/50/6/1378.full.pdf

B. Kudlow, S. Jameson, and B. Kennedy, HIV protease inhibitors block adipocyte differentiation independently of lamin A/C, AIDS, vol.19, issue.15, pp.1565-1573, 2005.
DOI : 10.1097/01.aids.0000186827.91408.90

R. Boguslavsky, C. Stewart, and H. Worman, Nuclear lamin A inhibits adipocyte differentiation: implications for Dunnigan-type familial partial lipodystrophy, Human Molecular Genetics, vol.15, issue.4, pp.653-663, 2006.
DOI : 10.1093/hmg/ddi480

URL : https://academic.oup.com/hmg/article-pdf/15/4/653/1800967/ddi480.pdf