D. R. Green and J. C. Reed, Mitochondria and apoptosis, Science, vol.281, pp.1309-1312, 1998.

C. Brenner and G. Kroemer, Apoptosis. Mitochondria -the death signal integrators, Science, vol.289, pp.1150-1151, 2000.
URL : https://hal.archives-ouvertes.fr/hal-00354243

S. Desagher and J. C. Martinou, Mitochondria as the central control point of apoptosis, Trends Cell Biol, vol.10, pp.369-377, 2000.

G. Kroemer, L. Galluzzi, and C. Brenner, Mitochondrial membrane permeabilization in cell death, Physiol Rev, vol.87, pp.99-163, 2007.
URL : https://hal.archives-ouvertes.fr/hal-00172652

M. R. Duchen and G. Szabadkai, Roles of mitochondria in human disease, Essays Biochem, vol.47, pp.115-137, 2010.

F. Ichas, L. Jouaville, and J. Mazat, Mitochondria are excitable organelles capable of generating and conveying electrical and calcium signals, Cell, vol.89, pp.1145-1153, 1997.

R. Rizzuto and T. Pozzan, Microdomains of intracellular Ca 2+ : molecular determinants and functional consequences, Physiol Rev, vol.86, pp.369-408, 2006.

H. M. Viola and L. C. Hool, Cross-talk between L-type Ca2+ channels and mitochondria, Clin Exp Pharmacol Physiol, vol.37, pp.229-235, 2010.

T. S. Luongo, J. P. Lambert, A. Yuan, X. Zhang, P. Gross et al., The mitochondrial calcium uniporter matches energetic supply with cardiac workload during stress and modulates permeability transition, Cell Rep, vol.12, pp.23-34, 2015.

J. Q. Kwong, X. Lu, R. N. Correll, J. A. Schwanekamp, R. J. Vagnozzi et al., The mitochondrial calcium uniporter selectively matches metabolic output to acute contractile stress in the heart, Cell Rep, vol.12, pp.15-22, 2015.

E. J. Griffiths and A. P. Halestrap, Mitochondrial non-specific pores remain closed during cardiac ischaemia, but open upon reperfusion, Biochem J, vol.307, pp.93-98, 1995.

A. P. Halestrap and A. P. Richardson, The mitochondrial permeability transition: a current perspective on its identity and role in ischaemia/reperfusion injury, J Mol Cell Cardiol, 2015.

M. Boerma, Experimental radiation-induced heart disease: past, present, and future, Radiat Res, vol.178, pp.1-6, 2012.

J. Q. Kwong and J. D. Molkentin, Physiological and pathological roles of the mitochondrial permeability transition pore in the heart, Cell Metab, vol.21, pp.206-214, 2015.

C. Piot, P. Croisille, P. Staat, H. Thibault, G. Rioufol et al., Effect of cyclosporine on reperfusion injury in acute myocardial infarction, N Engl J Med, vol.359, pp.473-481, 2008.
URL : https://hal.archives-ouvertes.fr/hal-00443408

A. Guellich, H. Mehel, and R. Fischmeister, Cyclic AMP synthesis and hydrolysis in the normal and failing heart, Pflugers Archiv, vol.466, pp.1163-1175, 2014.

J. De-rooij, F. J. Zwartkruis, M. H. Verheijen, R. H. Cool, S. M. Nijman et al., Epac is a Rap1 guanine-nucleotide-exchange factor directly activated by cyclic AMP, Nature, vol.396, pp.474-477, 1998.

H. Kawasaki, G. M. Springett, S. Toki, J. J. Canales, P. Harlan et al., A Rap guanine nucleotide exchange factor enriched highly in the basal ganglia, Proc Natl Acad Sci, vol.95, pp.13278-13283, 1998.

M. Schmidt, F. J. Dekker, and H. Maarsingh, Exchange protein directly activated by cAMP (epac): a multidomain cAMP mediator in the regulation of diverse biological functions, Pharmacol Rev, vol.65, pp.670-709, 2013.

M. Metrich, M. Berthouze, E. Morel, B. Crozatier, A. M. Gomez et al., Role of the cAMPbinding protein Epac in cardiovascular physiology and pathophysiology, Pflugers Archiv, vol.459, pp.535-546, 2010.

H. Chen, C. Wild, X. Zhou, N. Ye, X. Cheng et al., Recent advances in the discovery of small molecules targeting exchange proteins directly activated by cAMP (EPAC), J Med Chem, vol.57, pp.3651-3665, 2014.

E. Parnell, T. M. Palmer, and S. J. Yarwood, The future of EPAC-targeted therapies: agonism versus antagonism, Trends Pharmacol Sci, vol.36, pp.203-214, 2015.

T. N. Litvin, M. Kamenetsky, A. Zarifyan, J. Buck, and L. R. Levin, Kinetic properties of 'soluble' adenylyl cyclase. Synergism between calcium and bicarbonate, J Biol Chem, vol.278, pp.15922-15926, 2003.

B. S. Jaiswal and M. Conti, Calcium regulation of the soluble adenylyl cyclase expressed in mammalian spermatozoa, Proc Natl Acad Sci, vol.100, pp.10676-10681, 2003.

A. Spät, D. Katona, A. Rajki, D. Benedetto, G. Pozzan et al., Calcium-dependent mitochondrial cAMP production enhances aldosterone secretion, Mol Cell Endocrinol, vol.412, pp.196-204, 2015.

M. Kamenetsky, S. Middelhaufe, E. M. Bank, L. R. Levin, J. Buck et al., Molecular details of cAMP generation in mammalian cells: a tale of two systems, J Mol Biol, vol.362, pp.623-639, 2006.

R. Acin-perez, E. Salazar, M. Kamenetsky, J. Buck, L. R. Levin et al., Cyclic AMP produced inside mitochondria regulates oxidative phosphorylation, Cell Metab, vol.9, pp.265-276, 2009.

J. H. Zippin, L. R. Levin, and J. Buck, CO(2)/HCO(3)( ? )-responsive soluble adenylyl cyclase as a putative metabolic sensor, Trends Endocrinol Metab, vol.12, pp.366-370, 2001.

D. Benedetto, G. Scalzotto, E. Mongillo, M. Pozzan, and T. , Mitochondrial Ca(2+) uptake induces cyclic AMP generation in the matrix and modulates organelle ATP levels, Cell Metab, vol.17, pp.965-975, 2013.

R. Acin-perez, M. Russwurm, K. Gunnewig, M. Gertz, G. Zoidl et al., A phosphodiesterase 2A isoform localized to mitochondria regulates respiration, J Biol Chem, vol.286, pp.30423-30432, 2011.

S. Kumar, J. P. Flacke, S. Kostin, A. Appukuttan, H. P. Reusch et al., SLC4A7 sodium bicarbonate co-transporter controls mitochondrial apoptosis in ischaemic coronary endothelial cells, Cardiovasc Res, vol.89, pp.392-400, 2011.

S. Kumar, S. Kostin, J. P. Flacke, H. P. Reusch, and Y. Ladilov, Soluble adenylyl cyclase controls mitochondria-dependent apoptosis in coronary endothelial cells, J Biol Chem, vol.284, pp.14760-14768, 2009.

R. Acin-perez, E. Salazar, S. Brosel, H. Yang, E. A. Schon et al., Modulation of mitochondrial protein phosphorylation by soluble adenylyl cyclase ameliorates cytochrome oxidase defects, EMBO Mol Med, vol.1, pp.392-406, 2009.

S. Neubauer, The failing heart -an engine out of fuel, N Engl J Med, vol.356, pp.1140-1151, 2007.

K. Lefkimmiatis, D. Leronni, and A. M. Hofer, The inner and outer compartments of mitochondria are sites of distinct cAMP/PKA signaling dynamics, J Cell Biol, vol.202, pp.453-462, 2013.

J. Klarenbeek, J. Goedhart, A. Van-batenburg, D. Groenewald, and K. Jalink, Fourth-generation epac-based FRET sensors for cAMP feature exceptional brightness, photostability and dynamic range: characterization of dedicated sensors for FLIM, for ratiometry and with high affinity, PLoS One, vol.10, p.122513, 2015.

Z. Wang, C. Nicolas, R. Fischmeister, and C. Brenner, Enzymatic assays for probing mitochondrial apoptosis, Methods Mol Biol, vol.1265, pp.407-414, 2015.

J. Buck, M. L. Sinclair, L. Schapal, M. J. Cann, and L. R. Levin, Cytosolic adenylyl cyclase defines a unique signaling molecule in mammals, Proc Natl Acad Sci, vol.96, pp.79-84, 1999.

C. Brenner and M. Moulin, Physiological roles of the permeability transition pore, Circ Res, vol.111, pp.1237-1247, 2012.

A. S. Belzacq-casagrande, C. Martel, C. Pertuiset, A. Borgne-sanchez, E. Jacotot et al., Pharmacological screening and enzymatic assays for apoptosis, Front Biosci, vol.14, pp.3550-3562, 2009.
URL : https://hal.archives-ouvertes.fr/hal-00354311

A. M. Sardanelli, Z. Technikova-dobrova, S. C. Scacco, F. Speranza, and S. Papa, Characterization of proteins phosphorylated by the cAMP-dependent protein kinase of bovine heart mitochondria, FEBS Lett, vol.377, pp.470-474, 1995.

A. Carlucci, L. Lignitto, and A. Feliciello, Control of mitochondria dynamics and oxidative metabolism by cAMP, AKAPs and the proteasome, Trends Cell Biol, vol.18, pp.604-613, 2008.

H. Chen, C. Ding, C. Wild, H. Liu, T. Wang et al., Efficient synthesis of ESI-09, a novel non-cyclic nucleotide EPAC antagonist, Tetrahedron Lett, vol.54, pp.1546-1549, 2013.

D. Courilleau, P. Bouyssou, R. Fischmeister, F. Lezoualc'h, and J. P. Blondeau, The (R)-enantiomer of CE3F4 is a preferential inhibitor of human exchange protein directly activated by cyclic AMP isoform 1 (Epac1), Biochem Biophys Res Commun, vol.440, pp.443-448, 2013.
URL : https://hal.archives-ouvertes.fr/inserm-01011788

F. Joubert, J. R. Wilding, D. Fortin, V. Domergue-dupont, M. Novotova et al., Local energetic regulation of sarcoplasmic and myosin ATPase is differently impaired in rats with heart failure, J Physiol, vol.586, pp.5181-5192, 2008.
URL : https://hal.archives-ouvertes.fr/inserm-00325088

M. Marcil, A. Ascah, J. Matas, S. Bélanger, C. Deschepper et al., Compensated volume overload increases the vulnerability of heart mitochondria without affecting their functions in the absence of stress, J Mol Cell Cardiol, vol.41, pp.998-1009, 2006.

J. H. Zippin, Y. Chen, P. Nahirney, M. Kamenetsky, M. S. Wuttke et al., Compartmentalization of bicarbonate-sensitive adenylyl cyclase in distinct signaling microdomains, FASEB J, vol.17, pp.82-84, 2003.

S. Sulimovici and B. Lunenfeld, Effect of gonadotrophins on adenylate cyclase of the outer and inner membrane subfractions of rat testis mitochondria, FEBS Lett, vol.41, pp.345-347, 1974.

A. S. Fine, R. W. Egnor, E. Forrester, and S. S. Stahl, Adenylate cyclase localization in unfixed specimens of rat oral mucosa and isolated mitochondria, J Histochem Cytochem, vol.30, pp.1171-1178, 1982.

S. Papa, D. De-rasmo, S. Scacco, A. Signorile, Z. Technikova-dobrova et al., Mammalian complex I: a regulable and vulnerable pacemaker in mitochondrial respiratory function, Biochim Biophys Acta, vol.1777, pp.719-728, 2008.

R. Acin-perez, D. L. Gatti, Y. Bai, and G. Manfredi, Protein phosphorylation and prevention of cytochrome oxidase inhibition by ATP: coupled mechanisms of energy metabolism regulation, Cell Metab, vol.13, pp.712-719, 2011.

J. Qiao, F. C. Mei, V. L. Popov, L. A. Vergara, and X. Cheng, Cell cycle-dependent subcellular localization of exchange factor directly activated by cAMP, J Biol Chem, vol.277, pp.26581-26586, 2002.

L. Pereira, H. Rehmann, D. H. Lao, J. R. Erickson, J. Bossuyt et al., Novel Epac fluorescent ligand reveals distinct Epac1 vs. Epac2 distribution and function in cardiomyocytes, Proc Natl Acad Sci, vol.112, pp.3991-3996, 2015.

L. Galluzzi, J. M. Bravo-san-pedro, I. Vitale, S. A. Aaronson, J. M. Abrams et al., Essential versus accessory aspects of cell death: recommendations of the NCCD 2015, Cell Death Differ, vol.22, pp.58-73, 2015.

G. Kung, K. Konstantinidis, and R. N. Kitsis, Programmed necrosis, not apoptosis, in the heart, Circ Res, vol.108, pp.1017-1036, 2011.

C. Kilkenny, W. Browne, I. C. Cuthill, M. Emerson, and D. G. Altman, Animal research: reporting in vivo experiments: the ARRIVE guidelines, Br J Pharmacol, vol.160, pp.1577-1579, 2010.

F. Hubert, M. Belacel-ouari, B. Manoury, K. Zhai, V. Domergue-dupont et al., Alteration of vascular reactivity in heart failure: role of phosphodiesterases 3 and 4, Br J Pharmacol, vol.171, pp.5361-5375, 2014.

Y. Chen, M. J. Cann, T. N. Litvin, V. Iourgenko, M. L. Sinclair et al., Soluble adenylyl cyclase as an evolutionarily conserved bicarbonate sensor, Science, vol.289, pp.625-628, 2000.

E. Morel, A. Marcantoni, M. Gastineau, R. Birkedal, F. Rochais et al., cAMP-binding protein Epac induces cardiomyocyte hypertrophy, Circ Res, vol.97, pp.1296-1304, 2005.

F. Rochais, G. Vandecasteele, F. Lefebvre, C. Lugnier, H. Lum et al., Negative feedback exerted by cAMP-dependent protein kinase and cAMP phosphodiesterase on subsarcolemmal cAMP signals in intact cardiac myocytes: an in vivo study using adenovirusmediated expression of CNG channels, J Biol Chem, vol.279, pp.52095-52105, 2004.

C. Brenner and G. Kroemer, Apoptosis. Mitochondria--the death signal integrators, Science, vol.289, issue.5482, pp.1150-1151, 2000.
URL : https://hal.archives-ouvertes.fr/hal-00354243

S. E. Weinberg and N. S. Chandel, Targeting mitochondria metabolism for cancer therapy, Nature chemical biology, vol.2015, issue.1, pp.9-15

G. Vandecasteele, G. Szabadkai, and R. Rizzuto, Mitochondrial calcium homeostasis: mechanisms and molecules, IUBMB life, vol.52, issue.3-5, pp.213-219, 2001.

R. Acin-perez, M. Russwurm, K. Gunnewig, M. Gertz, G. Zoidl et al., A phosphodiesterase 2A isoform localized to mitochondria regulates respiration, The Journal of biological chemistry, vol.286, issue.35, pp.30423-30432, 2011.

S. Kumar, J. P. Flacke, S. Kostin, A. Appukuttan, H. P. Reusch et al., SLC4A7 sodium bicarbonate co-transporter controls mitochondrial apoptosis in ischaemic coronary endothelial cells, Cardiovascular research, vol.89, issue.2, pp.392-400, 2011.

S. Kumar, S. Kostin, J. P. Flacke, H. P. Reusch, and Y. Ladilov, Soluble adenylyl cyclase controls mitochondria-dependent apoptosis in coronary endothelial cells, The Journal of biological chemistry, vol.284, issue.22, pp.14760-14768, 2009.

R. Acin-perez, E. Salazar, S. Brosel, H. Yang, E. A. Schon et al., Modulation of mitochondrial protein phosphorylation by soluble adenylyl cyclase ameliorates cytochrome oxidase defects, EMBO molecular medicine, vol.1, issue.8-9, pp.392-406, 2009.

Z. Wang, D. Liu, A. Varin, V. Nicolas, D. Courilleau et al., A cardiac mitochondrial cAMP signaling pathway regulates calcium accumulation, permeability transition and cell death, Cell death & disease, vol.7, p.2198, 2016.

A. T. Bender and J. A. Beavo, Cyclic nucleotide phosphodiesterases: molecular regulation to clinical use, Pharmacological reviews, vol.58, issue.3, pp.488-520, 2006.

S. Sharma, K. Kumar, R. Deshmukh, and P. L. Sharma, Phosphodiesterases: Regulators of cyclic nucleotide signals and novel molecular target for movement disorders, European journal of pharmacology, vol.714, issue.1-3, pp.486-497, 2013.

M. Conti and J. Beavo, Biochemistry and physiology of cyclic nucleotide phosphodiesterases: essential components in cyclic nucleotide signaling. Annual review of biochemistry, vol.76, pp.481-511, 2007.

K. Omori and J. Kotera, Overview of PDEs and their regulation, Circulation research, vol.100, issue.3, pp.309-327, 2007.

S. Middelhaufe, L. Garzia, U. M. Ohndorf, B. Kachholz, M. Zollo et al., Domain mapping on the human metastasis regulator protein h-Prune reveals a C-terminal dimerization domain, The Biochemical journal, vol.407, issue.2, pp.199-205, 2007.
URL : https://hal.archives-ouvertes.fr/hal-00478779

D. Garzia, L. Andre, A. Carotenuto, P. Aglio, V. Guardiola et al., Prune cAMP phosphodiesterase binds nm23-H1 and promotes cancer metastasis, Cancer cell, vol.5, issue.2, pp.137-149, 2004.

R. Acin-perez, E. Salazar, M. Kamenetsky, J. Buck, L. R. Levin et al., Cyclic AMP produced inside mitochondria regulates oxidative phosphorylation, Cell metabolism, vol.9, issue.3, pp.265-276, 2009.

J. K. Millar, B. S. Pickard, S. Mackie, R. James, S. Christie et al., DISC1 and PDE4B are interacting genetic factors in schizophrenia that regulate cAMP signaling, Science, vol.310, issue.5751, pp.1187-1191, 2005.

:. Inserm-umr-s-769, L. Labex, and F. Châtenay-malabry, , vol.2

, Buffer H+ Bovine serum albumin (BSA): weigh 102

, Mitochondria membrane potential and swelling

, Buffer S: weigh 68.46g sucrose, 2.09g 3-(N-morpholino)propanesulfonic acid (MOPS), 3.8mg EGTA

, Succinate stock solution: weigh 1.35g succinate; buffer: 250mM sucrose, 15mM Potassium chloride (KCl), p.1

, EGTA, 5mM Magnesium chloride (MgCl2), p.30

, Reconstitute Mito-ID® Extracellular O 2 Sensor kit

, Loading buffer (5×): Tris 300mM, Glycerol 50%, sodium dodecylsulfate (SDS)

, 5%, dithiothreitol (DTT) 50mM

, -15% precast gel (BioRad Mini-PROTEAN TGX)

, Migration buffer (10×): weigh 30g Tris Base, p.144

, DTT (10×)

. Membrane,

, Trans-blot Turbo transfer system

, Phosphate buffered saline (PBS)-Tween (10×)

, Antibody anti-cytochrome c (BD Pharmingen)

, Ultra-sensitive enhanced chemiluminescent (ECL) substrate, e.g. Supersignal West Femto Maximum Sensitivity Substrate

, Gel imaging system, e.g. Chemidoc (BioRad), p.169

, Block the membrane with 5% of milk in PBS-Tween

, Wash the membrane with PBS-Tween for 6×5min

, Wash the membrane with PBS-Tween for 6×5min

J. C. Martinou, Apoptosis -Key to the mitochondrial gate, Nature, vol.399, pp.411-412, 1999.

C. Brenner and G. Kroemer, Apoptosis. Mitochondria--the death signal integrators, Science, vol.289, pp.1150-1151, 2000.
URL : https://hal.archives-ouvertes.fr/hal-00354243

G. Kroemer, L. Galluzzi, and C. Brenner, Mitochondrial membrane permeabilization in cell death
URL : https://hal.archives-ouvertes.fr/hal-00172652

, Physiol Rev, vol.87, pp.99-163, 2007.

N. Zamzami, P. Marchetti, M. Castedo, D. Decaudin, A. Macho et al.,

, J. Exp. Med, vol.182, pp.367-377, 1995.

V. Gogvadze, J. D. Robertson, B. Zhivotovsky, and S. Orrenius, Cytochrome c release occurs via Ca2+-dependent and Ca2+-independent mechanisms that are regulated by Bax, J Biol Chem, vol.276, pp.19066-19071, 1996.

X. Liu, C. N. Kim, J. Yang, R. Jemmerson, and X. Wang, Induction of apoptic program in cell-free extracts: requirement for dATP and cytochrome C, Cell, vol.86, pp.147-157, 1996.

M. Crompton, O. Mcguinness, and W. Nazareth, The involvement of cyclosporin A binding proteins in regulating and uncoupling mitochondrial energy transduction, Biochim Biophys Acta, vol.1101, pp.214-217, 1992.

A. Belzacq-casagrande, C. Martel, C. Pertuiset, A. Borgne-sanchez, E. Jacotot et al., Pharmacological screening and enzymatic assays for apoptosis, Front Biosci, vol.14, pp.3550-3562, 2009.
URL : https://hal.archives-ouvertes.fr/hal-00354311

M. Porceddu, N. Buron, C. Roussel, G. Labbe, B. Fromenty et al., Prediction of Liver Injury Induced by Chemicals in Human with a Multiparametric Assay on Isolated Mouse Liver Mitochondria, Toxicol Sci, 2012.
URL : https://hal.archives-ouvertes.fr/hal-00866136

C. Martel, M. Allouche, D. D. Esposti, E. Fanelli, C. Boursier et al.,

, 71 5. VDAC phosphorylation in lipid-induced liver pathology, VDAC, its post-translational modifications and their implication

.. .. Acknowledgments,

.. .. References, 74 1. Introduction The existence of channels in the mitochondrial outer membrane (OM) has been evidenced concomitantly by electrophysiology and electron microscopy in the, 1970.

. Shinohara, isoforms, i.e. VDAC1, 2, and 3 in mammals (Colombini and Mannella, 2000.

, Abbreviations: ACSL, long-chain acyl-CoA synthetase

, ANT, adenine nucleotide translocase; BN-PAGE, blue-native polyacrylamide gel electrophoresis

, CL, cardiolipin; CPT1a, carnitine palmitoyltransferase 1a

. Cypd,

. Ko,

I. Im and . Membrane,

L. Lpc,

. Lpe,

. Pc, ;. Pg, and . Ptp,

. Tnf-?,

, E-mail address: catherine, VDAC, voltage-dependent anion channel. ? Corresponding author at: INSERM UMR-S 769

,

A. Agafonov, E. Gritsenko, K. Belosludtsev, A. Kovalev, O. Gateau-roesch et al., A permeability transition in liposomes induced by the formation of Ca 2+ /palmitic acid complexes, Biochim. Biophys. Acta, vol.1609, pp.153-160, 2003.

A. V. Agafonov, E. N. Gritsenko, E. A. Shlyapnikova, D. P. Kharakoz, N. V. Belosludtseva et al., Ca 2+ -induced phase separation in the membrane of palmitate-containing liposomes and its possible relation to membrane permeabilization, J. Membr. Biol, vol.215, pp.57-68, 2007.

A. Bitar, F. Roosens, N. Smeyers, M. Vauterin, M. Van-boxtel et al., Sequence analysis, transcriptional and posttranscriptional regulation of the rice VDAC family, Biochim. Biophys. Acta, vol.1625, pp.43-51, 2003.

K. Anflous, D. D. Armstrong, and W. J. Craigen, Altered mitochondrial sensitivity for ADP and maintenance of creatine-stimulated respiration in oxidative striated muscles from VDAC1-deficient mice, J. Biol. Chem, vol.276, pp.1954-1960, 2001.

B. Antonsson, F. Conti, A. Ciavatta, S. Montessuit, S. Lewis et al., Inhibition of Bax channel-forming activity by Bcl-2, Science, vol.277, pp.370-372, 1997.

B. Antonsson, S. Montessuit, S. Lauper, R. Eskes, and J. C. Martinou, Bax oligomerization is required for channel-forming activity in liposomes and to trigger cytochrome c release from mitochondria, Biochem. J, vol.345, pp.271-278, 2000.

N. Arbel, D. Ben-hail, and V. Shoshan-barmatz, Mediation of the antiapoptotic activity of Bcl-XL protein upon interaction with VDAC1 protein, J. Biol. Chem, vol.287, pp.23152-23161, 2012.

E. Arcelay, A. M. Salicioni, E. Wertheimer, and P. E. Visconti, Identification of proteins undergoing tyrosine phosphorylation during mouse sperm capacitation, Int. J. Dev. Biol, vol.52, pp.463-472, 2008.

C. P. Baines, C. X. Song, Y. T. Zheng, G. W. Wang, J. Zhang et al., Protein kinase Cepsilon interacts with and inhibits the permeability transition pore in cardiac mitochondria, Circ. Res, vol.92, pp.873-880, 2003.

C. P. Baines, R. A. Kaiser, T. Sheiko, W. J. Craigen, and J. D. Molkentin, Voltage-dependent anion channels are dispensable for mitochondrial-dependent cell death, Nat. Cell Biol, vol.9, pp.550-555, 2007.

M. A. Baker, J. D. Ly, and A. Lawen, Characterization of VDAC1 as a plasma membrane NADH-oxidoreductase, Biofactors, vol.21, pp.215-221, 2004.

B. A. Ballif, G. R. Carey, S. R. Sunyaev, and S. P. Gygi, Large-scale identification and evolution indexing of tyrosine phosphorylation sites from murine brain, J. Proteome Res, vol.7, pp.311-318, 2008.

J. Banerjee and S. Ghosh, Phosphorylation of rat brain mitochondrial voltagedependent anion as a potential tool to control leakage of cytochrome c, J. Neurochem, vol.98, pp.670-676, 2006.

G. Basanez, J. C. Sharpe, J. Galanis, T. B. Brandt, J. M. Hardwick et al., Baxtype apoptotic proteins porate pure lipid bilayers through a mechanism sensitive to intrinsic monolayer curvature, J. Biol. Chem, vol.277, pp.49360-49365, 2002.

G. Bathori, G. Csordas, C. Garcia-perez, E. Davies, and G. Hajnoczky, Ca 2+ and voltage-dependent anion-selective channel (VDAC), J. Biol. Chem, vol.281, pp.17347-17358, 2006.

M. Bayrhuber, T. Meins, M. Habeck, S. Becker, K. Giller et al., Structure of the human voltagedependent anion channel, Proc. Natl. Acad. Sci. U. S. A, vol.105, pp.15370-15375, 2008.

K. N. Belosludtsev, N. E. Saris, N. V. Belosludtseva, A. S. Trudovishnikov, L. D. Lukyanova et al., Physiological aspects of the mitochondrial cyclosporin Ainsensitive palmitate/Ca 2+ -induced pore: tissue specificity, age profile and dependence on the animal's adaptation to hypoxia, J. Bioenerg. Biomembr, vol.41, pp.395-401, 2009.

A. S. Belzacq-casagrande, C. Martel, C. Pertuiset, A. Borgne-sanchez, E. Jacotot et al., Pharmacological screening and enzymatic assays for apoptosis, Front. Biosci. (Landmark Ed.), vol.14, pp.3550-3562, 2009.
URL : https://hal.archives-ouvertes.fr/hal-00354311

A. K. Bera and S. Ghosh, Dual mode of gating of voltage-dependent anion channel as revealed by phosphorylation, J. Struct. Biol, vol.135, pp.67-72, 2001.

V. Betaneli, E. P. Petrov, and P. Schwille, The role of lipids in VDAC oligomerization, Biophys. J, vol.102, pp.523-531, 2012.

G. Beutner, A. Ruck, B. Riede, W. Welte, and D. Brdiczka, Complexes between kinases, mitochondrial porin and adenylate translocator in rat brain resemble the permeability transition pore, FEBS Lett, vol.396, pp.189-195, 1996.

W. V. Bienvenut, D. Sumpton, A. Martinez, S. Lilla, C. Espagne et al., Comparative large scale characterization of plant versus mammal proteins reveals similar and idiosyncratic N-alpha-acetylation features, Mol. Cell. Proteomics, vol.11, 2012.

C. Brenner and M. Moulin, Physiological roles of the permeability transition pore, Circ. Res, vol.111, pp.1237-1247, 2012.

A. M. Campbell and S. H. Chan, The voltage dependent anion channel affects mitochondrial cholesterol distribution and function, Arch. Biochem. Biophys, vol.466, pp.203-210, 2007.

A. M. Campbell and S. H. Chan, Mitochondrial membrane cholesterol, the voltage dependent anion channel (VDAC), and the Warburg effect, J. Bioenerg. Biomembr, vol.40, pp.193-197, 2008.

M. Carre, N. Andre, G. Carles, H. Borghi, L. Brichese et al., Tubulin is an inherent component of mitochondrial membranes that interacts with the voltagedependent anion channel, J. Biol. Chem, vol.277, pp.33664-33669, 2002.

Y. Chen, W. J. Craigen, and D. J. Riley, Nek1 regulates cell death and mitochondrial membrane permeability through phosphorylation of VDAC1, Cell Cycle, vol.8, pp.257-267, 2009.

E. H. Cheng, T. V. Sheiko, J. K. Fisher, W. J. Craigen, and S. J. Korsmeyer, VDAC2 inhibits BAK activation and mitochondrial apoptosis, Science, vol.301, pp.513-517, 2003.

C. Choudhary, C. Kumar, F. Gnad, M. L. Nielsen, M. Rehman et al., Lysine acetylation targets protein complexes and co-regulates major cellular functions, Science, vol.325, pp.834-840, 2009.

M. Colombini and C. A. Mannella, VDAC, the early days, Biochim. Biophys. Acta, vol.1818, pp.1438-1443, 2012.

M. Cuadrado-tejedor, M. Vilarino, F. Cabodevilla, J. Del-rio, D. Frechilla et al., , 2011.

, Alzheimer's disease transgenic mice: an insight into the pathogenic effects of amyloid-beta, J. Alzheimers Dis, vol.23, pp.195-206

S. Das, R. Wong, N. Rajapakse, E. Murphy, and C. Steenbergen, Glycogen synthase kinase 3 inhibition slows mitochondrial adenine nucleotide transport and regulates voltage-dependent anion channel phosphorylation, Circ. Res, vol.103, pp.983-991, 2008.

M. A. De-pablo, S. A. Susin, E. Jacotot, N. Larochette, P. Costantini et al., Palmitate induces apoptosis via a direct effect on mitochondria, Apoptosis, vol.4, pp.81-87, 1999.

V. De-pinto, R. Benz, and F. Palmieri, Interaction of non-classical detergents with the mitochondrial porin. A new purification procedure and characterization of the pore-forming unit, Eur. J. Biochem, vol.183, pp.179-187, 1989.

D. De-stefani, A. Raffaello, E. Teardo, I. Szabo, and R. Rizzuto, A forty-kilodalton protein of the inner membrane is the mitochondrial calcium uniporter, Nature, vol.476, pp.336-340, 2011.

A. M. Distler, J. Kerner, and C. L. Hoppel, Post-translational modifications of rat liver mitochondrial outer membrane proteins identified by mass spectrometry, Biochim. Biophys. Acta, vol.1774, pp.628-636, 2007.

R. F. Epand, J. C. Martinou, M. Fornallaz-mulhauser, D. W. Hughes, and R. M. Epand, The apoptotic protein tBid promotes leakage by altering membrane curvature, J. Biol. Chem, vol.277, pp.32632-32639, 2002.

M. D. Esposti, J. T. Erler, J. A. Hickman, and C. Dive, Bid, a widely expressed proapoptotic protein of the Bcl-2 family, displays lipid transfer activity, Mol. Cell. Biol, vol.21, pp.7268-7276, 2001.

B. Faustin, R. Rossignol, A. Deniaud, C. Rocher, S. Claverol et al., The respiratory-dependent assembly of ANT1 differentially regulates Bax and Ca 2+ mediated cytochrome c release, Front. Biosci. (Elite Ed.), vol.3, pp.395-409, 2011.

S. Fu, S. M. Watkins, and G. S. Hotamisligil, The role of endoplasmic reticulum in hepatic lipid homeostasis and stress signaling, Cell Metab, vol.15, pp.623-634, 2012.

S. Fulda, L. Galluzzi, and G. Kroemer, Targeting mitochondria for cancer therapy, Nat. Rev. Drug Discov, vol.9, pp.447-464, 2010.

T. Furuno, T. Kanno, K. Arita, M. Asami, T. Utsumi et al., Roles of long chain fatty acids and carnitine in mitochondrial membrane permeability transition, Biochem. Pharmacol, vol.62, pp.1037-1046, 2001.

L. Galluzzi and G. Kroemer, Mitochondrial apoptosis without VDAC, Nat. Cell Biol, vol.9, pp.487-489, 2007.

S. Gauci, A. O. Helbig, M. Slijper, J. Krijgsveld, A. J. Heck et al., Lys-N and trypsin cover complementary parts of the phosphoproteome in a refined SCXbased approach, Anal. Chem, vol.81, pp.4493-4501, 2009.

C. L. Gentile, M. A. Frye, and M. J. Pagliassotti, Fatty acids and the endoplasmic reticulum in nonalcoholic fatty liver disease, Biofactors, vol.37, pp.8-16, 2011.

M. Gonzalez-gronow, R. Ray, F. Wang, and S. V. Pizzo, The voltage-dependent anion channel (VDAC) binds tissue-type plasminogen activator and promotes activation of plasminogen on the cell surface, J. Biol. Chem, vol.288, pp.498-509, 2013.

D. J. Granville and R. A. Gottlieb, The mitochondrial voltage-dependent anion channel (VDAC) as a therapeutic target for initiating cell death, Curr. Med. Chem, vol.10, pp.1527-1533, 2003.

P. A. Gurnev, T. K. Rostovtseva, and S. M. Bezrukov, Tubulin-blocked state of VDAC studied by polymer and ATP partitioning, FEBS Lett, vol.585, pp.2363-2366, 2011.

P. A. Gurnev, M. Queralt-martin, V. M. Aguilella, T. K. Rostovtseva, and S. M. Bezrukov, Probing tubulin-blocked state of VDAC by varying membrane surface charge, Biophys. J, vol.102, pp.2070-2076, 2012.

A. P. Halestrap and C. Brenner, The adenine nucleotide translocase: a central component of the mitochondrial permeability transition pore and key player in cell death, Curr. Med. Chem, vol.10, pp.1507-1525, 2003.

J. L. Herrera, C. Fernandez, M. Diaz, D. Cury, and R. Marin, Estradiol and tamoxifen differentially regulate a plasmalemmal voltage-dependent anion channel involved in amyloid-beta induced neurotoxicity, Steroids, vol.76, pp.840-844, 2011.

S. Hiller, R. G. Garces, T. J. Malia, V. Y. Orekhov, M. Colombini et al., Solution structure of the integral human membrane protein VDAC-1 in detergent micelles, Science, vol.321, pp.1206-1210, 2008.

H. Huang, X. Hu, C. O. Eno, G. Zhao, C. Li et al., An interaction between Bcl-X L and the voltage-dependent anion channel (VDAC) promotes mitochondrial Ca 2+ uptake, J. Biol. Chem, vol.288, pp.19870-19881, 2013.

V. Jancsik, M. Linden, L. Dorbani, A. Rendon, and B. D. Nelson, Studies on the relationship between the inner and outer membranes of rat liver mitochondria as determined by subfractionation with digitonin, Arch. Biochem. Biophys, vol.264, pp.295-301, 1988.

S. Javadov, V. Rajapurohitam, A. Kilic, A. Zeidan, A. Choi et al., Antihypertrophic effect of NHE-1 inhibition involves GSK-3beta-dependent attenuation of mitochondrial dysfunction, J. Mol. Cell. Cardiol, vol.46, pp.998-1007, 2009.

V. L. Johnsen, D. D. Belke, C. C. Hughey, D. S. Hittel, R. T. Hepple et al., Enhanced cardiac protein glycosylation (O-GlcNAc) of selected mitochondrial proteins in rats artificially selected for low running capacity, Physiol. Genomics, vol.45, pp.17-25, 2013.

H. Kayser, H. D. Kratzin, F. P. Thinnes, H. Gotz, W. E. Schmidt et al., Identification of human porins. II. Characterization and primary structure of a 31-lDa porin from human B lymphocytes (porin 31HL), Biol. Chem. Hoppe Seyler, vol.370, pp.1265-1278, 1989.

N. Keinan, D. Tyomkin, and V. Shoshan-barmatz, Oligomerization of the mitochondrial protein voltage-dependent anion channel is coupled to the induction of apoptosis, 2010.

, Mol. Cell. Biol, vol.30, pp.5698-5709

J. Kerner, K. Lee, B. Tandler, and C. L. Hoppel, VDAC proteomics: post-translation modifications, Biochim. Biophys. Acta, vol.1818, pp.1520-1525, 2012.

V. Koshkin, F. F. Dai, C. A. Robson-doucette, C. B. Chan, and M. B. Wheeler, Limited mitochondrial permeabilization is an early manifestation of palmitate-induced lipotoxicity in pancreatic beta-cells, J. Biol. Chem, vol.283, pp.7936-7948, 2008.

G. S. Krasnov, A. A. Dmitriev, V. A. Lakunina, A. A. Kirpiy, and A. V. Kudryavtseva, Targeting VDAC-bound hexokinase II: a promising approach for concomitant anti-cancer therapy, Expert Opin. Ther. Targets, vol.17, pp.1221-1233, 2013.

A. Krauskopf, O. Eriksson, W. J. Craigen, M. A. Forte, and P. Bernardi, Properties of the permeability transition in VDAC1(?/?) mitochondria, Biochim. Biophys. Acta, vol.1757, pp.590-595, 2006.

T. Kuwana, M. R. Mackey, G. Perkins, M. H. Ellisman, M. Latterich et al., Bid, Bax, and lipids cooperate to form supramolecular openings in the outer mitochondrial membrane, Cell, vol.111, pp.331-342, 2002.

Y. W. Lam, Y. Yuan, J. Isaac, C. V. Babu, J. Meller et al., Comprehensive identification and modified-site mapping of S-nitrosylated targets in prostate epithelial cells, Biochim. Biophys. Acta, vol.5, pp.540-545, 2010.

M. Lazarou, D. Stojanovski, A. E. Frazier, A. Kotevski, G. Dewson et al., Inhibition of Bak activation by VDAC2 is dependent on the Bak transmembrane anchor, J. Biol. Chem, vol.285, pp.36876-36883, 2010.

L. Bras, M. Clement, M. V. Pervaiz, S. Brenner, and C. , Reactive oxygen species and the mitochondrial signaling pathway of cell death, Histol. Histopathol, vol.20, pp.205-219, 2005.
URL : https://hal.archives-ouvertes.fr/hal-00101347

J. Lee, Y. Xu, Y. Chen, R. Sprung, S. C. Kim et al., Mitochondrial phosphoproteome revealed by an improved IMAC method and MS/MS/MS, Mol. Cell. Proteomics, vol.6, pp.669-676, 2007.

K. Lee, J. Kerner, and C. L. Hoppel, Mitochondrial carnitine palmitoyltransferase 1a (CPT1a) is part of an outer membrane fatty acid transfer complex, J. Biol. Chem, vol.286, pp.25655-25662, 2011.

K. Lefkimmiatis, D. Leronni, and A. M. Hofer, The inner and outer compartments of mitochondria are sites of distinct cAMP/PKA signaling dynamics, J. Cell Biol, vol.202, pp.453-462, 2013.

J. J. Lemasters and E. Holmuhamedov, Voltage-dependent anion channel (VDAC) as mitochondrial governator-thinking outside the box, Biochim. Biophys. Acta, vol.1762, pp.181-190, 2006.

S. Liberatori, B. Canas, C. Tani, L. Bini, G. Buonocore et al., Proteomic approach to the identification of voltage-dependent anion channel protein isoforms in guinea pig brain synaptosomes, Proteomics, vol.4, pp.1335-1340, 2004.

J. Liu, M. B. Rone, and V. Papadopoulos, Protein-protein interactions mediate mitochondrial cholesterol transport and steroid biosynthesis, J. Biol. Chem, vol.281, pp.38879-38893, 2006.

H. Low, F. L. Crane, and D. J. Morre, Putting together a plasma membrane NADH oxidase: a tale of three laboratories, Int. J. Biochem. Cell Biol, vol.44, pp.1834-1838, 2012.

M. Lutter, M. Fang, X. Luo, M. Nishijima, X. Xie et al., Cardiolipin provides specificity for targeting of tBid to mitochondria, Nat. Cell Biol, vol.2, pp.754-761, 2000.

S. Ma, C. Hockings, K. Anwari, T. Kratina, S. Fennell et al., Assembly of the Bak apoptotic pore: a critical role for the Bak protein alpha6 helix in the multimerization of homodimers during apoptosis, J. Biol. Chem, vol.288, pp.26027-26038, 2013.

C. Martel, / Mitochondrion, vol.19, pp.69-77, 2014.

M. Madesh and G. Hajnoczky, VDAC-dependent permeabilization of the outer mitochondrial membrane by superoxide induces rapid and massive cytochrome c release, 2001.

, J. Cell Biol, vol.155, pp.1003-1015

E. N. Maldonado and J. J. Lemasters, Warburg revisited: regulation of mitochondrial metabolism by voltage-dependent anion channels in cancer cells, J. Pharmacol. Exp. Ther, vol.342, pp.637-641, 2012.

T. J. Malia and G. Wagner, NMR structural investigation of the mitochondrial outer membrane protein VDAC and its interaction with antiapoptotic Bcl-X L, Biochemistry, vol.46, pp.514-525, 2007.

D. Marsh, Protein modulation of lipids, and vice-versa, in membranes, Biochim. Biophys. Acta, vol.1778, pp.1545-1575, 2008.

C. Martel, M. Allouche, D. D. Esposti, E. Fanelli, C. Boursier et al., Glycogen synthase kinase 3-mediated voltage-dependent anion channel phosphorylation controls outer mitochondrial membrane permeability during lipid accumulation, Hepatology, vol.57, pp.93-102, 2013.
URL : https://hal.archives-ouvertes.fr/hal-00734262

I. Marzo, C. Brenner, N. Zamzami, S. A. Susin, G. Beutner et al., The permeability transition pore complex: a target for apoptosis regulation by caspases and bcl-2-related proteins, J. Exp. Med, vol.187, pp.1261-1271, 1998.

C. M. Matsko, O. C. Hunter, H. Rabinowich, M. T. Lotze, and A. A. Amoscato, Mitochondrial lipid alterations during Fas-and radiation-induced apoptosis, Biochem. Biophys. Res. Commun, vol.287, pp.1112-1120, 2001.

S. R. Maurya and R. Mahalakshmi, Modulation of human mitochondrial voltagedependent anion channel 2 (hVDAC-2) structural stability by cysteine-assisted barrel-lipid interactions, J. Biol. Chem, vol.288, pp.25584-25592, 2013.

K. S. Mccommis and C. P. Baines, The role of VDAC in cell death: friend or foe?, Biochim. Biophys. Acta, vol.1818, pp.1444-1450, 2012.

T. J. Mcintosh and S. A. Simon, Roles of bilayer material properties in function and distribution of membrane proteins, Annu. Rev. Biophys. Biomol. Struct, vol.35, pp.177-198, 2006.

A. Messina, F. Guarino, M. Oliva, L. P. Van-den-heuvel, J. Smeitink et al., Characterization of the human porin isoform 1 (HVDAC1) gene by amplification on the whole human genome: A tool for porin deficiency analysis, Biochem. Biophys. Res. Commun, vol.270, pp.787-792, 2000.

A. Messina, S. Reina, F. Guarino, and V. De-pinto, VDAC isoforms in mammals, Biochim. Biophys. Acta, vol.1818, pp.1466-1476, 2012.

R. P. Munton, R. Tweedie-cullen, M. Livingstone-zatchej, F. Weinandy, M. Waidelich et al., Qualitative and quantitative analyses of protein phosphorylation in naive and stimulated mouse synaptosomal preparations, Mol. Cell. Proteomics, vol.6, pp.283-293, 2007.

J. V. Olsen, B. Blagoev, F. Gnad, B. Macek, C. Kumar et al., Global, in vivo, and site-specific phosphorylation dynamics in signaling networks, Cell, vol.127, pp.635-648, 2006.

F. S. Oppermann, K. Grundner-culemann, C. Kumar, O. J. Gruss, P. V. Jallepalli et al., Combination of chemical genetics and phosphoproteomics for kinase signaling analysis enables confident identification of cellular downstream targets, Mol. Cell. Proteomics, vol.11, issue.O111, pp.443-455, 2007.

D. B. Ostrander, G. C. Sparagna, A. A. Amoscato, J. B. Mcmillin, and W. Dowhan, Decreased cardiolipin synthesis corresponds with cytochrome c release in palmitate-induced cardiomyocyte apoptosis, J. Biol. Chem, vol.276, pp.38061-38067, 2001.

M. Ott, V. Gogvadze, S. Orrenius, and B. Zhivotovsky, Mitochondria, oxidative stress and cell death, Apoptosis, vol.12, pp.913-922, 2007.

E. Oyanagi, H. Yano, M. Uchida, K. Utsumi, and J. Sasaki, Protective action of L-carnitine on cardiac mitochondrial function and structure against fatty acid stress, Biochem. Biophys. Res. Commun, vol.412, pp.61-67, 2011.

J. G. Pastorino, N. Shulga, and J. B. Hoek, Mitochondrial binding of hexokinase II inhibits Bax-induced cytochrome c release and apoptosis, J. Biol. Chem, vol.277, pp.7610-7618, 2002.

J. G. Pastorino, J. B. Hoek, and N. Shulga, Activation of glycogen synthase kinase 3beta disrupts the binding of hexokinase II to mitochondria by phosphorylating voltagedependent anion channel and potentiates chemotherapy-induced cytotoxicity, Cancer Res, vol.65, pp.10545-10554, 2005.

M. B. Paumen, Y. Ishida, M. Muramatsu, M. Yamamoto, and T. Honjo, Inhibition of carnitine palmitoyltransferase I augments sphingolipid synthesis and palmitate-induced apoptosis, J. Biol. Chem, vol.272, pp.3324-3329, 1997.

B. Popp, A. Schmid, and R. Benz, Role of sterols in the functional reconstitution of water-soluble mitochondrial porins from different organisms, Biochemistry, vol.34, pp.3352-3361, 1995.

A. Raghavan, T. Sheiko, B. H. Graham, and W. J. Craigen, Voltage-dependant anion channels: novel insights into isoform function through genetic models, Biochim. Biophys. Acta, vol.1818, pp.1477-1485, 2012.

P. H. Reddy, Amyloid beta-induced glycogen synthase kinase 3beta phosphorylated VDAC1 in Alzheimer's disease: implications for synaptic dysfunction and neuronal damage, Biochim. Biophys. Acta, vol.1832, pp.1913-1921, 2013.

D. Ren, H. Kim, H. C. Tu, T. D. Westergard, J. K. Fisher et al., The VDAC2-BAK rheostat controls thymocyte survival, Sci. Signal, vol.2, p.48, 2009.

C. Rogers, B. Davis, P. D. Neufer, M. P. Murphy, E. J. Anderson et al., A transient increase in lipid peroxidation primes preadipocytes for delayed mitochondrial inner membrane permeabilization and ATP depletion during prolonged exposure to fatty acids. Free Radic, Biol. Med, vol.67, pp.330-341, 2014.

M. B. Rone, J. Fan, and V. Papadopoulos, Cholesterol transport in steroid biosynthesis: role of protein-protein interactions and implications in disease states, Biochim. Biophys. Acta, vol.1791, pp.646-658, 2009.

M. B. Rone, A. S. Midzak, L. Issop, G. Rammouz, S. Jagannathan et al., Identification of a dynamic mitochondrial protein complex driving cholesterol import, trafficking, and metabolism to steroid hormones, Mol. Endocrinol, vol.26, pp.1868-1882, 2012.

C. Rosano, Molecular model of hexokinase binding to the outer mitochondrial membrane porin (VDAC1): implication for the design of new cancer therapies, vol.11, pp.513-519, 2011.

T. K. Rostovtseva and S. M. Bezrukov, VDAC inhibition by tubulin and its physiological implications, Biochim. Biophys. Acta, vol.1818, pp.1526-1535, 2012.

T. K. Rostovtseva, B. Antonsson, M. Suzuki, R. J. Youle, M. Colombini et al., Bid, but not Bax, regulates VDAC channels, J. Biol. Chem, vol.279, pp.13575-13583, 2004.

T. K. Rostovtseva, N. Kazemi, M. Weinrich, and S. M. Bezrukov, Voltage gating of VDAC is regulated by nonlamellar lipids of mitochondrial membranes, J. Biol. Chem, vol.281, pp.37496-37506, 2006.

T. K. Rostovtseva, K. L. Sheldon, E. Hassanzadeh, C. Monge, V. Saks et al., Tubulin binding blocks mitochondrial voltage-dependent anion channel and regulates respiration, Proc. Natl. Acad. Sci. U. S. A, vol.105, pp.18746-18751, 2008.

T. K. Rostovtseva, P. A. Gurnev, M. Y. Chen, and S. M. Bezrukov, Membrane lipid composition regulates tubulin interaction with mitochondrial voltage-dependent anion channel, J. Biol. Chem, vol.287, pp.29589-29598, 2012.

X. Roucou, S. Montessuit, B. Antonsson, and J. C. Martinou, Bax oligomerization in mitochondrial membranes requires tBid (caspase-8-cleaved Bid) and a mitochondrial protein, Biochem. J, vol.368, pp.915-921, 2002.

M. J. Sampson, R. S. Lovell, and W. J. Craigen, The murine voltage-dependent anion channel gene family. Conserved structure and function, J. Biol. Chem, vol.272, pp.18966-18973, 1997.

M. J. Sampson, W. K. Decker, A. L. Beaudet, W. Ruitenbeek, D. Armstrong et al., Immotile sperm and infertility in mice lacking mitochondrial voltage-dependent anion channel type 3, J. Biol. Chem, vol.276, pp.39206-39212, 2001.

H. Schwertz, J. M. Carter, M. Abdudureheman, M. Russ, U. Buerke et al., Myocardial ischemia/ reperfusion causes VDAC phosphorylation which is reduced by cardioprotection with a p38 MAP kinase inhibitor, Proteomics, vol.7, pp.4579-4588, 2007.

K. L. Sheldon, E. N. Maldonado, J. J. Lemasters, T. K. Rostovtseva, and S. M. Bezrukov, Phosphorylation of voltage-dependent anion channel by serine/threonine kinases governs its interaction with tubulin, PLoS One, vol.6, p.25539, 2011.

S. Shimizu, M. Narita, and Y. Tsujimoto, Bcl-2 family proteins regulate the release of apoptogenic cytochrome c by the mitochondrial channel VDAC, Nature, vol.399, pp.483-487, 1999.

S. Shimizu, T. Ide, T. Yanagida, and Y. Tsujimoto, Electrophysiological study of a novel large pore formed by Bax and the voltage-dependent anion channel that is permeable to cytochrome c, J. Biol. Chem, vol.275, pp.12321-12325, 2000.

Y. Shinohara, T. Ishida, M. Hino, N. Yamazaki, Y. Baba et al., Characterization of porin isoforms expressed in tumor cells, Eur. J. Biochem, vol.267, pp.6067-6073, 2000.

V. Shoshan-barmatz and D. Ben-hail, VDAC, a multi-functional mitochondrial protein as a pharmacological target, Mitochondrion, vol.12, pp.24-34, 2012.

V. Shoshan-barmatz and M. Golan, Mitochondrial VDAC1: function in cell life and death and a target for cancer therapy, Curr. Med. Chem, vol.19, pp.714-735, 2012.

V. Shoshan-barmatz, N. Keinan, and H. Zaid, Uncovering the role of VDAC in the regulation of cell life and death, J. Bioenerg. Biomembr, vol.40, pp.183-191, 2008.

V. Shoshan-barmatz, M. Zakar, K. Rosenthal, and S. Abu-hamad, Key regions of VDAC1 functioning in apoptosis induction and regulation by hexokinase, Biochim. Biophys. Acta, vol.1787, pp.421-430, 2009.

V. Shoshan-barmatz, V. De-pinto, M. Zweckstetter, Z. Raviv, N. Keinan et al., VDAC, a multi-functional mitochondrial protein regulating cell life and death, Mol. Aspects Med, vol.31, pp.227-285, 2010.

V. Shoshan-barmatz, D. Mizrachi, and N. Keinan, Oligomerization of the mitochondrial protein VDAC1: from structure to function and cancer therapy, Prog. Mol. Biol. Transl. Sci, vol.117, pp.303-334, 2013.

A. W. Smith, Lipid-protein interactions in biological membranes: a dynamic perspective, Biochim. Biophys. Acta, vol.1818, pp.172-177, 2012.

E. M. Sol, S. A. Wagner, B. T. Weinert, A. Kumar, H. S. Kim et al., Proteomic investigations of lysine acetylation identify diverse substrates of mitochondrial deacetylase sirt3, PLoS One, vol.7, p.50545, 2012.

G. C. Sparagna, D. L. Hickson-bick, L. M. Buja, and J. B. Mcmillin, A metabolic role for mitochondria in palmitate-induced cardiac myocyte apoptosis, Am. J. Physiol. Heart Circ. Physiol, vol.279, pp.2124-2132, 2000.

L. Sun, S. Shukair, T. J. Naik, F. Moazed, and H. Ardehali, Glucose phosphorylation and mitochondrial binding are required for the protective effects of hexokinases I and II, Mol. Cell. Biol, vol.28, pp.1007-1017, 2008.

I. Szabo and M. Zoratti, The mitochondrial permeability transition pore may comprise VDAC molecules. I. Binary structure and voltage dependence of the pore, FEBS Lett, vol.330, pp.201-205, 1993.

W. Tan and M. Colombini, VDAC closure increases calcium ion flux, Biochim. Biophys. Acta, vol.1768, pp.2510-2515, 2007.

F. P. Thinnes and G. Burckhardt, On a fully closed state of native human type-1 VDAC enriched in Nonidet P40, Mol. Genet. Metab, vol.107, pp.632-633, 2012.

F. P. Thinnes, H. Gotz, H. Kayser, R. Benz, W. E. Schmidt et al., Identification of human porins. I. Purification of a porin from human B-lymphocytes (porin 31HL) and the topochemical proof of its expression on the plasmalemma of the progenitor cell, Biol. Chem. Hoppe Seyler, vol.370, pp.1253-1264, 1989.

R. Ujwal, D. Cascio, J. P. Colletier, S. Faham, J. Zhang et al., The crystal structure of mouse VDAC1 at 2.3 Å resolution reveals mechanistic insights into metabolite gating, Proc. Natl. Acad. Sci. U. S. A, vol.105, pp.17742-17747, 2008.

P. Van-damme, M. Lasa, B. Polevoda, C. Gazquez, A. Elosegui-artola et al., , 2012.

C. Martel, / Mitochondrion, vol.19, pp.69-77, 2014.

, analyses and functional insights of the N-terminal acetyltransferase NatB, Proc. Natl. Acad. Sci. U. S. A, vol.109, pp.12449-12454

G. Meer, D. R. Voelker, and G. W. Feigenson, Membrane lipids: where they are and how they behave, Nat. Rev. Mol. Cell Biol, vol.9, pp.112-124, 2008.

M. G. Vander-heiden, X. X. Li, E. Gottleib, R. B. Hill, C. B. Thompson et al., Bcl-X L promotes the open configuration of the voltage-dependent anion channel and metabolite passage through the outer mitochondrial membrane, J. Biol. Chem, vol.276, pp.19414-19419, 2001.

L. Vela, O. Gonzalo, J. Naval, and I. Marzo, Direct interaction of Bax and Bak proteins with Bcl-2 homology domain 3 (BH3)-only proteins in living cells revealed by fluorescence complementation, J. Biol. Chem, vol.288, pp.4935-4946, 2013.

H. Vitrac, M. Bogdanov, P. Heacock, and W. Dowhan, Lipids and topological rules of membrane protein assembly: balance between long and short range lipid-protein interactions, J. Biol. Chem, vol.286, pp.15182-15194, 2011.

Z. Wang, Y. Ge, H. Bao, L. Dworkin, A. Peng et al., Redox-sensitive glycogen synthase kinase 3beta-directed control of mitochondrial permeability transition: rheostatic regulation of acute kidney injury. Free Radic, Biol. Med, vol.65, pp.849-858, 2013.

E. J. Weeber, M. Levy, M. J. Sampson, K. Anflous, D. L. Armstrong et al., The role of mitochondrial porins and the permeability transition pore in learning and synaptic plasticity, J. Biol. Chem, vol.277, pp.18891-18897, 2002.

M. R. Wieckowski and L. Wojtczak, Fatty acid-induced uncoupling of oxidative phosphorylation is partly due to opening of the mitochondrial permeability transition pore, FEBS Lett, vol.423, pp.339-342, 1998.

K. Woodfield, A. Ruck, D. Brdiczka, and A. P. Halestrap, Direct demonstration of a specific interaction between cyclophilin-D and the adenine nucleotide translocase confirms their role in the mitochondrial permeability transition, Biochem. J, vol.336, pp.287-290, 1998.

S. Wu, M. J. Sampson, W. K. Decker, and W. J. Craigen, Each mammalian mitochondrial outer membrane porin protein is dispensable: effects on cellular respiration, Biochim. Biophys. Acta, vol.1452, pp.68-78, 1999.

T. Yamamoto, A. Yamada, M. Watanabe, Y. Yoshimura, N. Yamazaki et al., VDAC1, having a shorter Nterminus than VDAC2 but showing the same migration in an SDS-polyacrylamide gel, is the predominant form expressed in mitochondria of various tissues, J. Proteome Res, vol.5, pp.3336-3344, 2006.

C. H. Yi, H. Pan, J. Seebacher, I. H. Jang, S. G. Hyberts et al., Metabolic regulation of protein N-alpha-acetylation by Bcl-XL promotes cell survival, Cell, vol.146, pp.607-620, 2011.

S. Yuan, Y. Fu, X. Wang, H. Shi, Y. Huang et al., Voltagedependent anion channel 1 is involved in endostatin-induced endothelial cell apoptosis, FASEB J, vol.22, pp.2809-2820, 2008.

H. Zaid, S. Abu-hamad, A. Israelson, I. Nathan, and V. Shoshan-barmatz, The voltagedependent anion channel-1 modulates apoptotic cell death, Cell Death Differ, vol.12, pp.751-760, 2005.

C. Martel, / Mitochondrion, vol.19, pp.69-77, 2014.

. Inserm-u848, G. Institut, and . Roussy, Université Paris-Sud 11, PR1, 39 rue Camille Desmoulins

. Inserm-u-996,

, Metabolomics Platform, Institut Gustave Roussy

, Keywords: apoptosis; chemotherapy

, DTT, dithiothreitol; IRE1, ER to nucleus signaling 1; MMP, mitochondrial membrane permeabilization

, MS, mass spectrometry; NSLC, non-small lung cancer

, PDI, protein disulfide isomerase; PERK, protein kinase RNA-like endoplasmic reticulum kinase; GRP78, 78 kDa glucose-regulated protein

. Dcm, Cell Death and Differentiation, vol.21, pp.685-695, 2014.

M. Promega and . Wi, ) with the following parameters: Swiss-Prot 57.7 database, Homo sapiens taxonomy, one missed cleavage by trypsin, fixed carbamido-SDS-PAGE and immunoblotted with the following antibodies: ATF6 (Abcam, & 2014 Macmillan Publishers Limited All rights reserved 1350-9047/14 trypsin

. Bax, BD Biosciences

, Caspase, vol.9

, Cell Signaling, p.78

, HSP70

, Abcam, p.1

, Abcam, p.3

S. Pdia4-(abcam and . Cruz,

, Santa Cruz and Abcam, p.6

, PERK (Abcam); and RIPK1 (BioVision)

, Cell viability analysis. The number of surviving cells was determined by the WST-1 assay

, Cell death analysis. A549 and A2780 WT and resistant cells were analyzed for cell death as previously described

. Briefly, . Tmrm, S. Invitrogen, and . Aubin, France) flow cytometry (FACSCalibur flow cytometer, BD Biosciences). Finally, early and late apoptosis were analyzed using Annexin-PE/ 7AAD (BD Pharmingen

(. Assay and . Promega,

, Villeurbanne, France) by using insulin as substrate to evaluate PDI activity in A549 whole extracts and ER fraction. Clonogenic assay. Cells were harvested, washed with PBS and plated for 2-3 weeks at 37 1C. Then, colonies were stained with 0.25% of crystal violet and counted using GS800 calibrated densitometer (Bio-Rad) and PDQuest software (Bio-Rad). [Ca 2 þ ] measurements in the ER and cytoplasm. For steady state, PDI activity assay. ProteoStat PDI assay kit was used following the manufacturer's instruction

, fitted with a computer-controlled motorized heated stage (37 1C, Applied Scientific Instrumentation, with a  40 Hamamatsu C10600-10B CCD camera, vol.3, p.substrate

, la Recherche Médicale (FRM

, Institut National du Cancer

L. Labex,

, LabEx Immuno-Oncologie

, Fondation de France

, Fondation Bettencourt-Schueller

, AXA Chair for Longevity Research; and Cancéropôle Ile-de-France and Paris Alliance of Cancer Research Institutes (PACRI)

G. Kroemer, L. Galluzzi, and C. Brenner, Mitochondrial membrane permeabilization in cell death, Physiol Rev, vol.87, pp.99-163, 2007.
URL : https://hal.archives-ouvertes.fr/hal-00172652

D. Hanahan and R. A. Weinberg, The hallmarks of cancer, Cell, vol.100, pp.57-70, 2000.

A. Rebillard, X. Tekpli, O. Meurette, O. Sergent, G. Lemoigne-muller et al., Cisplatin-induced apoptosis involves membrane fluidification via inhibition of NHE1 in human colon cancer cells, Cancer Res, vol.67, pp.7865-7874, 2007.
URL : https://hal.archives-ouvertes.fr/hal-00690301

L. Galluzzi, L. Senovilla, I. Vitale, J. Michels, I. Martins et al., Molecular mechanisms of cisplatin resistance, Oncogene, vol.31, pp.1869-1883, 2012.

B. Köberle, M. Tomicic, S. Usanova, and B. Kaina, Cisplatin resistance: preclinical findings and clinical implications, Biochim Biophys Acta, vol.1806, pp.172-182, 2010.

Y. Ma and L. M. Hendershot, The role of the unfolded protein response in tumour development: friend or foe?, Nat Rev, vol.4, pp.966-977, 2004.

K. Zhang and R. J. Kaufman, Signaling the unfolded protein response from the endoplasmic reticulum, J Biol Chem, vol.279, pp.25935-25938, 2004.

D. Ron and P. Walter, Signal integration in the endoplasmic reticulum unfolded protein response, Nat Rev Mol Cell Biol, vol.8, pp.519-529, 2007.

P. Hersey and X. D. Zhang, Adaptation to ER stress as a driver of malignancy and resistance to therapy in human melanoma, Pigment Cell Melanoma Res, vol.21, pp.358-367, 2008.

I. Kim, W. Xu, and J. Reed, Cell death and endoplasmic reticulum stress: disease relevance and therapeutic opportunities, Nat Rev Drug Discov, vol.7, pp.1013-1030, 2008.

A. Deniaud, O. Sharaf-el-dein, E. Maillier, D. Poncet, G. Kroemer et al., Endoplasmic reticulum stress induces calcium-dependent permeability transition, mitochondrial outer membrane permeabilization and apoptosis, Oncogene, vol.27, pp.285-299, 2008.
URL : https://hal.archives-ouvertes.fr/hal-00321149

A. S. Lee, GRP78 induction in cancer: therapeutic and prognostic implications, Cancer Res, vol.67, pp.3496-3499, 2007.

W. Han, J. Xie, L. Li, Z. Liu, and X. Hu, Necrostatin-1 reverts shikonin-induced necroptosis to apoptosis, Apoptosis, vol.14, pp.674-686, 2009.

Y. Kim, G. Haidl, M. Schaefer, U. Egner, and J. Herr, Compartmentalization of a unique ADP/ATP carrier protein SFEC (Sperm Flagellar Energy Carrier, AAC4) with glycolytic enzymes in the fibrous sheath of the human sperm flagellar principal piece, Dev Biol, vol.302, pp.463-476, 2007.

D. T. Rutkowski, S. M. Arnold, C. N. Miller, J. Wu, J. Li et al., Adaptation to ER stress is mediated by differential stabilities of pro-survival and pro-apoptotic mRNAs and proteins, PLoS Biol, vol.4, p.374, 2006.

Z. Yao, A. Jones, E. Fassone, M. Sweeney, M. Lebiedzinska et al., PGC-1b mediates adaptive chemoresistance associated with mitochondrial DNA mutations, Oncogene, vol.32, pp.2592-2600, 2013.

L. Galluzzi, I. Vitale, J. M. Abrams, E. S. Alnemri, E. H. Baehrecke et al., Molecular definitions of cell death subroutines: recommendations of the Nomenclature Committee on Cell Death, Cell Death Differ, vol.19, pp.107-120, 2012.

N. Tajeddine, L. Galluzzi, O. Kepp, E. Hangen, E. Morselli et al., Hierarchical involvement of Bak, VDAC1 and Bax in cisplatin-induced cell death, Oncogene, vol.27, pp.4221-4232, 2008.

C. A. Belfi, S. Chatterjee, D. M. Gosky, S. J. Berger, and N. A. Berger, Increased sensitivity of human colon cancer cells to DNA cross-linking agents after GRP78 up-regulation, Biochem Biophys Res Commun, vol.257, pp.361-368, 1999.

M. Konopleva, S. Zhao, W. Hu, S. Jiang, V. Snell et al., The anti-apoptotic genes Bcl-X(L) and Bcl-2 are over-expressed and contribute to chemoresistance of nonproliferating leukaemic CD34 þ cells, Br J Haematol, vol.118, pp.521-534, 2002.

L. Ravagnan, S. Gurbuxani, S. A. Susin, C. Maisse, E. Daugas et al., Heat-shock protein 70 antagonizes apoptosis-inducing factor, Nat Cell Biol, vol.3, pp.839-843, 2001.
URL : https://hal.archives-ouvertes.fr/hal-01606079

J. Michels, I. Vitale, L. Senovilla, D. Enot, P. Garcia et al., Synergistic interaction between cisplatin and PARP inhibitors in non-small cell lung cancer, Cell Cycle, vol.12, pp.877-883, 2013.

A. Mandic, J. Hansson, S. Linder, and M. C. Shoshan, Cisplatin induces endoplasmic reticulum stress and nucleus-independent apoptotic signaling, J Biol Chem, vol.278, pp.9100-9106, 2003.

M. Chami, B. Oulès, G. Szabadkai, R. Tacine, R. Rizzuto et al., Role of SERCA1 truncated isoform in the proapoptotic calcium transfer from ER to mitochondria during ER stress, Mol Cell, vol.32, pp.641-651, 2008.

A. E. Palmer, J. C. Reed, J. C. Tsien, and R. Y. , Bcl-2-mediated alterations in endoplasmic reticulum Ca2 þ analyzed with an improved genetically encoded fluorescent sensor, Proc Natl Acad Sci, vol.101, pp.17404-17409, 2004.

J. Vicencio, C. Ortiz, A. Criollo, A. Jones, O. Kepp et al., The inositol 1,4,5-trisphosphate receptor regulates autophagy through its interaction with Beclin 1, Cell Death Differ, vol.16, pp.1006-1017, 2009.
URL : https://hal.archives-ouvertes.fr/hal-00378189

G. Inesi and Y. Sagara, Thapsigargin, a high affinity and global inhibitor of intracellular Ca2 þ transport ATPases, Arch Biochem Biophys, vol.298, pp.313-317, 1992.

M. Schwaller, B. Wilkinson, and H. F. Gilbert, Reduction-reoxidation cycles contribute to catalysis of disulfide isomerization by protein-disulfide isomerase, J Biol Chem, vol.278, pp.7154-7159, 2003.

R. A. Roth, Bacitracin: an inhibitor of the insulin degrading activity of glutathione-insulin transhydrogenase, Biochem Biophys Res Commun, vol.98, pp.431-438, 1981.

P. Lovat, M. Corazzari, J. Armstrong, S. Martin, V. Pagliarini et al., Increasing melanoma cell death using inhibitors of protein disulfide isomerases to abrogate survival responses to endoplasmic reticulum stress, Cancer Res, vol.68, pp.5363-5369, 2008.

L. Ellgaard and E. M. Frickel, Calnexin, calreticulin, and ERp57: teammates in glycoprotein folding, Cell Biochem Biophys, vol.39, pp.223-247, 2003.

Z. M. Bian, S. G. Elner, and V. M. Elner, Dual involvement of caspase-4 in inflammatory and ER stress-induced apoptotic responses in human retinal pigment epithelial cells, Invest Ophthalmol Vis Sci, vol.50, pp.6006-6014, 2009.

J. Hitomi, T. Katayama, Y. Eguchi, T. Kudo, M. Taniguchi et al., Involvement of caspase-4 in endoplasmic reticulum stress-induced apoptosis and Abeta-induced cell death, J Cell Biol, vol.165, pp.347-356, 2004.

P. Vandenabeele, W. Declercq, F. Van-herreweghe, V. Berghe, and T. , The role of the kinases RIP1 and RIP3 in TNF-induced necrosis, Sci Signal, vol.3, p.4, 2010.

J. Michels, I. Vitale, L. Galluzzi, J. Adam, K. Olaussen et al., Cisplatin resistance associated with PARP hyperactivation, Cancer Res, vol.73, pp.2271-2280, 2013.

S. Xu, A. Butkevich, R. Yamada, Y. Zhou, B. Debnath et al., Discovery of an orally active small-molecule irreversible inhibitor of protein disulfide isomerase for ovarian cancer treatment, Proc Natl Acad Sci, vol.109, pp.16348-16353, 2012.

C. Turano, S. Coppari, F. Altieri, and A. Ferraro, Proteins of the PDI family: unpredicted non-ER locations and functions, J Cell Physiol, vol.193, pp.154-163, 2002.

R. Vanderwaal, D. Spitz, C. Griffith, R. Higashikubo, R. Roti et al., Evidence that protein disulfide isomerase (PDI) is involved in DNA-nuclear matrix anchoring, J Cell Biochem, vol.85, pp.689-702, 2002.

Y. Honjo, H. Ito, T. Horibe, R. Takahashi, and K. Kawakami, Protein disulfide isomerase immunopositive glial cytoplasmic inclusions in patients with multiple system atrophy, Int J Neurosci, vol.121, pp.543-550, 2011.

Y. Honjo, H. Ito, T. Horibe, R. Takahashi, and K. Kawakami, Protein disulfide isomerase-immunopositive inclusions in patients with Alzheimer disease, Brain Res, vol.1349, pp.90-96, 2010.

A. Walker, M. Farg, C. Bye, C. Mclean, M. Horne et al., Protein disulphide isomerase protects against protein aggregation and is S-nitrosylated in amyotrophic lateral sclerosis, Brain Res, vol.133, pp.105-116, 2010.

J. Uys, Y. Xiong, and D. Townsend, Nitrosative stress-induced S-glutathionylation of protein disulfide isomerase, Methods Enzymol, vol.490, pp.321-332, 2011.

P. Bernardoni, B. Fazi, A. Costanzi, R. Nardacci, C. Montagna et al., Reticulon1-C modulates protein disulphide isomerase function, Cell Death Dis, vol.4, p.581, 2013.

T. Nakamura and S. Lipton, Redox modulation by S-nitrosylation contributes to protein misfolding, mitochondrial dynamics, and neuronal synaptic damage in neurodegenerative diseases, Cell Death Differ, vol.18, pp.1478-1486, 2011.

N. Krynetskaia, M. Phadke, S. Adhav, and E. Krynetskiy, Chromatin-associated proteins HMGB1/ 2 and PDIA3 trigger cellular response to chemotherapy-induced DNA damage, Mol Cancer Ther, vol.4, pp.864-872, 2009.

C. Martel, M. Allouche, D. D. Esposti, E. Fanelli, C. Boursier et al., GSK3-mediated VDAC phosphorylation controls outer mitochondrial membrane permeability during lipid accumulation, Hepatology, vol.57, pp.93-102, 2013.

L. Bras, M. Borgne-sanchez, A. Touat, Z. , S. El-dein et al., Chemosensitization by knock-down of adenine nucleotide translocase-2, Cancer Res, vol.66, pp.9143-9152, 2006.
URL : https://hal.archives-ouvertes.fr/hal-00172574

Y. Gu, W. Di, D. Kelsell, and D. Zicha, Quantitative fluorescence resonance energy transfer (FRET) measurement with acceptor photobleaching and spectral unmixing, J Microsc, vol.215, pp.162-173, 2004.

, Supplementary Information accompanies this paper on Cell Death and Differentiation website