B. Döring and E. Petzinger, Phase 0 and phase III transport in various organs: combined concept of phases in xenobiotic transport and metabolism, Drug Metab Rev, vol.46, issue.3, pp.261-82, 2014.

, Some non-heterocyclic polycyclic aromatic hydrocarbons and some related exposures, IARC Monographs, vol.92, 2010.

D. Hanahan and R. A. Weinberg, Hallmarks of cancer: the next generation, Cell, vol.144, issue.5, pp.646-74, 2011.

K. Hardonnière, L. Huc, O. Sergent, J. A. Holme, and D. Lagadic-gossmann, Environmental carcinogenesis and pH homeostasis: Not only a matter of dysregulated metabolism, Semin Cancer Biol, vol.43, pp.49-65, 2017.

S. Harguindey and S. J. Reshkin, The new pH-centric anticancer paradigm in Oncology and, Medicine

, Semin Cancer Biol, vol.43, pp.1-4, 2017.

C. H. Leem, D. Lagadic-gossmann, and V. Rd, Characterization of intracellular pH regulation in the guinea-pig ventricular myocyte, J Physiol, vol.517, pp.159-80, 1999.

D. Lagadic-gossmann, L. Huc, and V. Lecureur, Alterations of intracellular pH homeostasis in apoptosis: origins and roles, Cell Death Differ, vol.11, issue.9, pp.953-61, 2004.

D. Lagadic-gossmann, L. Huc, and W. Tekpli, Role for Na+/+ exchanger 1 (NHE1) in the control of apoptotic pathways, Cell Apoptotic Signaling Pathways, pp.83-114
URL : https://hal.archives-ouvertes.fr/hal-00699966

C. Stock and S. F. Pedersen, Roles of pH and the Na+/H+ exchanger NHE1 in cancer: From cell biology and animal models to an emerging translational perspective? Semin Cancer Biol, vol.43, pp.5-16, 2017.

P. Swietach, R. D. Vaughan-jones, A. L. Harris, and A. Hulikova, The chemistry, physiology and pathology of pH in cancer, Philos Trans R Soc Lond B Biol Sci, vol.369, p.20130099, 1638.

L. Counillon, Y. Bouret, I. Marchiq, and P. J. Na, +)/H(+) antiporter (NHE1) and lactate/H(+) symporters (MCTs) in pH homeostasis and cancer metabolism, Biochim Biophys Acta, issue.10, pp.2465-80, 1863.

J. Lacroix, M. Poët, and L. Counillon, The Na+/H+ exchanger NHE-1, a multi-controlled cellular integrator, Proton homeostasis in tumorigenesis and cell death, 2011.

J. Lacroix, M. Poët, L. Huc, V. Morello, N. Djerbi et al., Kinetic analysis of the regulation of the Na+/H+ exchanger NHE-1 by osmotic shocks, Biochemistry, vol.47, issue.51, pp.13674-85, 2008.
URL : https://hal.archives-ouvertes.fr/hal-00674405

X. Tekpli, L. Huc, J. Lacroix, M. Rissel, M. Poët et al., Regulation of Na+/H+ exchanger 1 allosteric balance by its localization in cholesterol-and caveolin-rich membrane microdomains, J Cell Physiol, vol.216, issue.1, pp.207-227, 2008.
URL : https://hal.archives-ouvertes.fr/hal-00696172

L. Huc, L. Sparfel, M. Rissel, M. T. Dimanche-boitrel, A. Guillouzo et al., Identification of Na+/H+ exchange as a new target for toxic polycyclic aromatic hydrocarbons, FASEB J, vol.18, issue.2, pp.344-350, 2004.

L. Huc, M. Rissel, A. Solhaug, X. Tekpli, M. Gorria et al., Multiple apoptotic pathways induced by p53-dependent acidification in benzo[a]pyrene-exposed hepatic F258 cells, J Cell Physiol, vol.208, issue.3, pp.527-564, 2006.
URL : https://hal.archives-ouvertes.fr/inserm-00130324

L. Huc, X. Tekpli, J. A. Holme, M. Rissel, A. Solhaug et al., Lagadic-Gossmann D. c-Jun NH2-terminal kinase-related Na+/H+ exchanger isoform 1 activation controls hexokinase II expression in benzo(a)pyrene-induced apoptosis, Cancer Res, vol.67, issue.4, pp.1696-705, 2007.

J. A. Holme, M. Gorria, V. M. Arlt, S. Ovrebø, A. Solhaug et al., Different mechanisms involved in apoptosis following exposure to benzo[a]pyrene in F258 and Hepa1c1c7 cells, Chem Biol Interact, vol.167, issue.1, pp.41-55, 2007.
URL : https://hal.archives-ouvertes.fr/hal-00690306

R. Barhoumi, Y. Mouneimne, E. Ramos, C. Morisseau, B. D. Hammock et al., Multiphoton spectral analysis of benzo[a]pyrene uptake and metabolism in a rat liver cell line, Toxicol Appl Pharmacol, vol.253, issue.1, pp.45-56, 2011.

M. Gorria, X. Tekpli, O. Sergent, L. Huc, F. Gaboriau et al., Membrane fluidity changes are associated with benzo[a]pyreneinduced apoptosis in F258 cells: protection by exogenous cholesterol, Ann N Y Acad Sci, vol.1090, pp.108-120, 2006.

X. Tekpli, L. Huc, O. Sergent, B. Dendelé, M. T. Dimanche-boitrel et al., NHE-1 relocation outside cholesterol-rich membrane microdomains is associated with its benzo[a]pyrene-related apoptotic function, Cell Physiol Biochem, vol.29, issue.5-6, pp.657-66, 2012.
URL : https://hal.archives-ouvertes.fr/inserm-00871487

S. Wakabayashi, T. Ikeda, T. Iwamoto, J. Pouysségur, and M. Shigekawa, Calmodulin-binding autoinhibitory domain controls "pH-sensing" in the Na+/H+ exchanger NHE1 through sequencespecific interaction, Biochemistry, vol.36, issue.42, pp.12854-61, 1997.

X. Li, D. Prins, M. Michalak, and L. Fliegel, Calmodulin-dependent binding to the NHE1 cytosolic tail mediates activation of the Na+/H+ exchanger by Ca2+ and endothelin, Am J Physiol Cell Physiol

, , vol.305, pp.1161-1170

X. Tekpli, M. Rissel, L. Huc, D. Catheline, O. Sergent et al., Membrane remodeling, an early event in benzo[a]pyreneinduced apoptosis, Toxicol Appl Pharmacol, vol.243, issue.1, pp.68-76, 2010.
URL : https://hal.archives-ouvertes.fr/hal-00729575

C. Aravena, A. R. Beltrán, M. Cornejo, V. Torres, E. S. Díaz et al., Potential role of sodium-proton exchangers in the low concentration arsenic trioxide-increased intracellular pH and cell proliferation, PLoS One, vol.7, issue.12, p.51451, 2012.

K. Hardonnière, E. Saunier, A. Lemarié, M. Fernier, I. Gallais et al., The environmental carcinogen benzo[a]pyrene induces a Warburg-like metabolic reprogramming dependent on NHE1 and associated with cell survival. Sci Rep, vol.6, p.30776, 2016.

J. Chiche, M. C. Brahimi-horn, and J. Pouysségur, Tumour hypoxia induces a metabolic shift causing acidosis: a common feature in cancer, J Cell Mol Med, vol.14, issue.4, pp.771-94, 2010.

L. Stüwe, M. Müller, A. Fabian, J. Waning, S. Mally et al., pH dependence of melanoma cell migration: protons extruded by NHE1 dominate protons of the bulk solution, J Physiol, vol.585, pp.351-60, 2007.

C. Pinheiro, A. Longatto-filho, J. Azevedo-silva, M. Casal, F. C. Schmitt et al., Role of monocarboxylate transporters in human cancers: state of the art, J Bioenerg Biomembr, vol.44, issue.1, pp.127-166, 2012.

C. S. Parkins, M. R. Stratford, M. F. Dennis, M. Stubbs, and D. J. Chaplin, The relationship between extracellular lactate and tumour pH in a murine tumour model of ischaemia-reperfusion, Br J Cancer, vol.75, issue.3, pp.319-342, 1997.

T. H. Dovmark, M. Saccomano, A. Hulikova, F. Alves, and P. Swietach, Connexin-43 channels are a pathway for discharging lactate from glycolytic pancreatic ductal adenocarcinoma cells, vol.36, pp.4538-4550, 2017.

X. Tekpli, E. Rivedal, M. Gorria, N. E. Landvik, M. Rissel et al., The B[a]P-increased intercellular communication via translocation of connexin-43 into gap junctions reduces apoptosis, Toxicol Appl Pharmacol, vol.242, issue.2, pp.231-271, 2010.
URL : https://hal.archives-ouvertes.fr/hal-00660130

M. C. Villa-abrille, E. Cingolani, H. E. Cingolani, and B. V. Alvarez, Silencing of cardiac mitochondrial NHE1 prevents mitochondrial permeability transition pore opening, Am J Physiol Heart Circ Physiol, vol.300, issue.4, pp.1237-51, 2011.

C. D. Garciarena, C. I. Caldiz, M. V. Correa, G. R. Schinella, S. M. Mosca et al., Na+/H+ exchanger-1 inhibitors decrease myocardial superoxide production via direct mitochondrial action, J Appl Physiol, vol.105, issue.6, pp.1706-1719, 1985.

G. Baffy, Uncoupling protein-2 and cancer. Mitochondrion, vol.10, pp.243-52, 2010.

G. Yu, J. Liu, K. Xu, and J. Dong, Uncoupling protein 2 mediates resistance to gemcitabine-induced apoptosis in hepatocellular carcinoma cell lines, Biosci Rep, vol.35, issue.4, p.231, 2015.

G. Baffy, Z. Derdak, and S. C. Robson, Mitochondrial recoupling: a novel therapeutic strategy for cancer?, Br J Cancer, vol.105, issue.4, pp.469-74, 2009.

K. Omidian, H. Rafiei, and B. Bandy, Polyphenol inhibition of benzo[a]pyrene-induced oxidative stress and neoplastic transformation in an in vitro model of carcinogenesis, Food Chem Toxicol

, , vol.106, pp.165-174

Z. Derdak, N. M. Mark, G. Beldi, S. C. Robson, J. R. Wands et al., The mitochondrial uncoupling protein-2 promotes chemoresistance in cancer cells, Cancer Res, vol.68, issue.8, pp.2813-2822, 2008.

M. Gorria, X. Tekpli, M. Rissel, O. Sergent, L. Huc et al., A new lactoferrin-and iron-dependent lysosomal death pathway is induced by benzo[a]pyrene in hepatic epithelial cells, Toxicol Appl Pharmacol, vol.228, issue.2, pp.212-236, 2008.
URL : https://hal.archives-ouvertes.fr/hal-00674390

B. Liu, J. Palmfeldt, L. Lin, A. Colaço, K. Clemmensen et al., STAT3 associates with vacuolar H+-ATPase and regulates cytosolic and lysosomal pH, Cell Res, vol.28, issue.10, pp.996-1012, 2018.

G. Ichim and S. W. Tait, A fate worse than death: apoptosis as an oncogenic process, Nat Rev Cancer, vol.16, issue.8, pp.539-587, 2016.

G. Kroemer, W. S. El-deiry, P. Golstein, M. E. Peter, D. Vaux et al., Nomenclature Committee on Cell Death. Classification of cell death: recommendations of the Nomenclature Committee on Cell Death, Cell Death Differ, vol.12, issue.2, pp.1463-1470, 2005.
URL : https://hal.archives-ouvertes.fr/hal-00407686

A. Solhaug, M. Refsnes, M. Låg, P. E. Schwarze, T. Husøy et al., Polycyclic aromatic hydrocarbons induce both apoptotic and anti-apoptotic signals in Hepa1c1c7 cells, Carcinogenesis, vol.25, issue.5, pp.809-828, 2004.

A. Collin, K. Hardonnière, M. Chevanne, J. Vuillemin, N. Podechard et al., Cooperative interaction of benzo[a]pyrene and ethanol on plasma membrane remodeling is responsible for enhanced oxidative stress and cell death in primary rat hepatocytes, Free Radic Biol Med, vol.72, pp.11-22, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01018147

S. P. Denker, D. C. Huang, J. Orlowski, H. Furthmayr, and D. L. Barber, Direct binding of the Na--H exchanger NHE1 to ERM proteins regulates the cortical cytoskeleton and cell shape independently of H(+) translocation, Mol Cell, vol.6, issue.6, pp.1425-1461, 2000.

M. Baumgartner, H. Patel, and D. L. Barber, Na(+)/H(+) exchanger NHE1 as plasma membrane scaffold in the assembly of signaling complexes, Am J Physiol Cell Physiol, vol.287, issue.4, pp.844-50, 2004.

M. E. Wiles, J. A. Dykens, and C. D. Wright, Regulation of polymorphonuclear leukocyte membrane fluidity: effect of cytoskeletal modification, J Leukoc Biol, vol.56, issue.2, pp.192-201, 1994.

C. Astarie, L. Quan-sang, K. H. David-dufilho, M. Devynck, and M. A. , Further investigation of platelet cytosolic alkalinization in essential hypertension, J Hypertens, vol.10, issue.8, pp.849-54, 1992.

C. Rauch, On the relationship between Lipinski's 2nd rule and cytosolic pH changes in doxorubicin resistance levels in cancer cells: a comparison to published data, Eur Biophys J, vol.38, issue.7, pp.829-875, 2009.

E. Proksch, pH in nature, humans and skin, J Dermatol, vol.45, issue.9, 2018.

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, issue.16, pp.7865-74, 2007.
URL : https://hal.archives-ouvertes.fr/hal-00690301

M. Aureli, N. Loberto, R. Bassi, A. Ferraretto, S. Perego et al., Plasma membrane-associated glycohydrolases activation by extracellular acidification due to proton exchangers, Neurochem Res, vol.37, issue.6, pp.1296-307, 2012.

M. Aureli, R. Bassi, A. Prinetti, E. Chiricozzi, B. Pappalardi et al., Ionizing radiations increase the activity of the cell surface glycohydrolases and the plasma membrane ceramide content, Glycoconj J, vol.29, issue.8-9, pp.585-97, 2012.

R. Valaperta, V. Chigorno, L. Basso, A. Prinetti, R. Bresciani et al., Plasma membrane production of ceramide from ganglioside GM3 in human fibroblasts, FASEB J, vol.20, issue.8, pp.1227-1236, 2006.

M. Gorria, L. Huc, O. Sergent, A. Rebillard, F. Gaboriau et al., Protective effect of monosialoganglioside GM1 against chemically induced apoptosis through targeting of mitochondrial function and iron transport, Biochem Pharmacol, vol.72, issue.10, pp.1343-53, 2006.
URL : https://hal.archives-ouvertes.fr/hal-00699820

D. R. Richardson and P. Ponka, The molecular mechanisms of the metabolism and transport of iron in normal and neoplastic cells, Biochim Biophys Acta, vol.1331, issue.1, pp.1-40, 1997.

N. Ben-dov and R. Korenstein, Proton-induced endocytosis is dependent on cell membrane fluidity, lipid-phase order and the membrane resting potential, Biochim Biophys Acta, issue.11, pp.2672-81, 1828.

B. Dendelé, X. Tekpli, O. Sergent, M. T. Dimanche-boitrel, J. A. Holme et al., Identification of the couple GSK3?/c-Myc as a new regulator of hexokinase II in benzo[a]pyrene-induced apoptosis, Toxicol In Vitro, vol.26, issue.1, pp.94-101, 2012.

J. G. Pastorino and J. B. Hoek, Hexokinase II: the integration of energy metabolism and control of apoptosis, Curr Med Chem, vol.10, issue.16, pp.1535-51, 2003.

W. S. Da-silva, A. Gómez-puyou, M. T. De-gómez-puyou, R. Moreno-sanchez, D. Felice et al.,

L. Meis, M. F. Oliveira, and A. Galina, Mitochondrial bound hexokinase activity as a preventive antioxidant defense: steady-state ADP formation as a regulatory mechanism of membrane potential and reactive oxygen species generation in mitochondria, J Biol Chem, vol.279, issue.38, pp.39846-55, 2004.

M. Broecker-preuss, N. Becher-boveleth, A. Bockisch, U. Dührsen, and S. Müller, Regulation of glucose uptake in lymphoma cell lines by c-Myc-and PI3K-dependent signaling pathways and impact of glycolytic pathways on cell viability, J Transl Med, vol.15, issue.1, p.158, 2017.

J. G. Pastorino and J. B. Hoek, Regulation of hexokinase binding to VDAC, J Bioenerg Biomembr, vol.40, issue.3, pp.171-82, 2008.

C. Zhong, L. Fan, F. Yao, J. Shi, W. Fang et al., HMGCR is necessary for the tumorigenecity of esophageal squamous cell carcinoma and is regulated by Myc, Tumour Biol, vol.35, issue.5, pp.4123-4132, 2014.

C. Naveenkumar, S. Raghunandhakumar, S. Asokkumar, and T. Devaki, Baicalein abrogates reactive oxygen species (ROS)-mediated mitochondrial dysfunction during experimental pulmonary carcinogenesis in vivo, Basic Clin Pharmacol Toxicol, vol.112, issue.4, pp.270-81, 2013.

T. Arif, A. Paul, Y. Krelin, A. Shteinfer-kuzmine, and V. Shoshan-barmatz, Mitochondrial VDAC1 Silencing Leads to Metabolic Rewiring and the Reprogramming of Tumour Cells into Advanced Differentiated States. Cancers (Basel), vol.10, 2018.

C. H. Quach, K. H. Jung, J. H. Lee, J. W. Park, S. H. Moon et al., Mild Alkalization Acutely Triggers the Warburg Effect by Enhancing Hexokinase Activity via Voltage-Dependent Anion Channel Binding, PLoS One, vol.11, issue.8, p.159529, 2016.

S. R. Amith, K. M. Vincent, J. M. Wilkinson, L. M. Postovit, and L. Fliegel, Defining the Na+/H+ exchanger NHE1 interactome in triple-negative breast cancer cells. Cell Signal, vol.29, pp.69-77, 2017.

R. Hendus-altenburger, E. Pedraz-cuesta, C. W. Olesen, E. Papaleo, J. A. Schnell et al., The human Na(+)/H(+) exchanger 1 is a membrane scaffold protein for extracellular signal-regulated kinase 2, BMC Biol, vol.14, p.31, 2016.

S. J. Reshkin, A. Bellizzi, S. Caldeira, V. Albarani, I. Malanchi et al., Na+/H+ exchanger-dependent intracellular alkalinization is an early event in malignant transformation and plays an essential role in the development of subsequent transformation-associated phenotypes, FASEB J, vol.14, issue.14, pp.2185-97, 2000.

R. B. Robey, J. Weisz, N. B. Kuemmerle, A. C. Salzberg, A. Berg et al., Metabolic reprogramming and dysregulated metabolism: cause, consequence and/or enabler of environmental carcinogenesis? Carcinogenesis, vol.36, pp.203-234, 2015.

A. Tameemi, W. Dale, T. P. , A. , R. Forsyth et al., Hypoxia-Modified Cancer Cell Metabolism. Front Cell Dev Biol, vol.7, p.4, 2019.

A. Rosenzweig, J. Blenis, and A. P. Gomes, Beyond the Warburg Effect: How Do Cancer Cells Regulate One-Carbon Metabolism? Front Cell Dev Biol, vol.6, p.90, 2018.

L. Sun, C. Suo, S. T. Li, H. Zhang, and P. Gao, Metabolic reprogramming for cancer cells and their microenvironment: Beyond the Warburg Effect, Biochim Biophys Acta Rev Cancer, 2018.

, , vol.1870, pp.51-66

J. F. Dumas, L. Brisson, S. Chevalier, K. Mahéo, G. Fromont et al., Metabolic reprogramming in cancer cells
URL : https://hal.archives-ouvertes.fr/hal-01907572

L. Wilde, M. Roche, M. Domingo-vidal, K. Tanson, N. Philp et al., Metabolic coupling and the Reverse Warburg Effect in cancer: Implications for novel biomarker and anticancer agent development, Semin Oncol, vol.44, issue.3, pp.198-203, 2017.

G. Gasparre, R. Rossignol, and P. Sonveaux, Mitochondria in cancer, Biochim Biophys Acta Bioenerg, issue.8, pp.553-555, 1858.

A. Lemarie, L. Huc, E. Pazarentzos, A. L. Mahul-mellier, and S. Grimm, Specific disintegration of complex II succinate:ubiquinone oxidoreductase links pH changes to oxidative stress for apoptosis induction, Cell Death Differ, vol.18, issue.2, pp.338-387, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00565452

K. Hardonnière and D. Lagadic-gossmann, ATPase inhibitory factor 1 (IF1): a novel player in pollutant-related diseases? Current Opinion in Toxicology, vol.8, pp.42-47, 2018.

E. Cabezon, P. J. Butler, M. J. Runswick, and J. E. Walker, Modulation of the oligomerization state of the bovine F1-ATPase inhibitor protein, IF1, by pH, J Biol Chem, vol.275, issue.33, pp.25460-25464, 2000.

K. Hardonnière, M. Fernier, I. Gallais, B. Mograbi, N. Podechard et al., Role for the ATPase inhibitory factor 1 in the environmental carcinogen-induced Warburg phenotype. Sci Rep, vol.7, p.195, 2017.

S. J. Reshkin, M. R. Greco, and R. A. Cardone, Role of pHi, and proton transporters in oncogene-driven neoplastic transformation, Philos Trans R Soc Lond B Biol Sci, vol.369, p.20130100, 1638.

H. Chen, L. S. Lee, G. Li, S. W. Tsao, and J. F. Chiu, Upregulation of glycolysis and oxidative phosphorylation in benzo[?]pyrene and arsenic-induced rat lung epithelial transformed cells, Oncotarget, vol.7, issue.26, pp.40674-40689, 2016.

S. Bortoli, E. Boutet-robinet, D. Lagadic-gossmann, and L. Huc, Nrf2 and AhR in metabolic reprogramming after contaminant exposure. Current Opinion in Toxicology, vol.8, pp.34-41, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01699380

J. Y. Guo, E. White, M. Autophagy, and C. , Cold Spring Harb Symp Quant Biol, vol.81, pp.73-78, 2016.

C. W. Yun and S. H. Lee, The Roles of Autophagy in Cancer, Int J Mol Sci, vol.19, issue.11, 2018.

E. White, Deconvoluting the context-dependent role for autophagy in cancer, Nat Rev Cancer, vol.12, issue.6, pp.401-411, 2012.

Y. Chun and J. Kim, Autophagy: An Essential Degradation Program for Cellular Homeostasis and Life. Cells, vol.7, 2018.

L. Galluzzi, T. Yamazaki, and G. Kroemer, Linking cellular stress responses to systemic homeostasis, Nat Rev Mol Cell Biol, vol.19, issue.11, pp.731-745, 2018.
URL : https://hal.archives-ouvertes.fr/hal-02022975

M. D. Rybstein, J. M. Bravo-san-pedro, G. Kroemer, and L. Galluzzi, The autophagic network and cancer, Nat Cell Biol, vol.20, issue.3, pp.243-251, 2018.

A. Scrivo, M. Bourdenx, O. Pampliega, and A. M. Cuervo, Selective autophagy as a potential therapeutic target for neurodegenerative disorders, Lancet Neurol, vol.17, issue.9, pp.30238-30240, 2018.

M. Abdellatif, S. Sedej, D. Carmona-gutierrez, F. Madeo, and G. Kroemer, Autophagy in Cardiovascular Aging, Circ Res, vol.123, issue.7, pp.803-824, 2018.

A. Belaid, P. D. Ndiaye, H. Filippakis, J. Roux, É. Röttinger et al., Autophagy : Moving Benchside Promises to Patient Bedsides, Curr Cancer Drug Targets, vol.15, issue.8, pp.684-702, 2015.

A. Belaid, B. Roméo, H. Filippakis, M. Meyer, I. Grosjean et al., Autophagy and cardiometabolic diseases: From molecular mechanisms to diseases, Drs Jun Ren, pp.255-275, 2018.

M. Pesonen and K. Vähäkangas, Autophagy in exposure to environmental chemicals, Toxicol Lett

, , pp.30009-30017, 2019.

A. Chargui, S. Zekri, G. Jacquillet, I. Rubera, M. Ilie et al., Cadmium-induced autophagy in rat kidney: an early biomarker of subtoxic exposure, Toxicol Sci, vol.121, issue.1, pp.31-42, 2011.

A. Lau, Y. Zheng, S. Tao, H. Wang, S. A. Whitman et al., Arsenic inhibits autophagic flux, activating the Nrf2-Keap1 pathway in a p62-dependent manner, Mol Cell Biol, vol.33, issue.12, pp.2436-2482, 2013.

R. A. González-polo, M. Niso-santano, M. A. Ortíz-ortíz, A. Gómez-martín, J. M. Morán et al., Inhibition of paraquat-induced autophagy accelerates the apoptotic cell death in neuroblastoma SH-SY5Y cells, Toxicol Sci, vol.97, issue.2, pp.448-58, 2007.

C. Quan, C. Wang, P. Duan, W. Huang, W. Chen et al., Bisphenol a induces autophagy and apoptosis concurrently involving the Akt/mTOR pathway in testes of pubertal SD rats, Environ Toxicol, vol.32, issue.8, pp.1977-1989, 2017.

X. Guo, Y. Dong, S. Yin, C. Zhao, Y. Huo et al., Patulin induces pro-survival functions via autophagy inhibition and p62 accumulation, Cell Death Dis, vol.4, p.822, 2013.

F. Fiorito, R. Ciarcia, G. E. Granato, G. Marfe, V. Iovane et al., 3,7,8-tetrachlorodibenzo-p-dioxin induced autophagy in a bovine kidney cell line, Toxicology, vol.2, issue.2-3, pp.258-70, 2011.

J. Zhao, C. Tang, X. Nie, H. Xi, S. Jiang et al., Autophagy potentially protects against 2,3,7,8-tetrachlorodibenzo-p-Dioxin induced apoptosis in SH-SY5Y cells, Environ Toxicol, vol.31, issue.9, pp.1068-79, 2016.

L. Yuan, J. Liu, H. Deng, and C. Gao, Benzo[a]pyrene Induces Autophagic and Pyroptotic Death Simultaneously in HL-7702 Human Normal Liver Cells, J Agric Food Chem, vol.65, issue.44, 2017.

D. N. Das, P. P. Naik, S. Mukhopadhyay, P. K. Panda, N. Sinha et al., Elimination of dysfunctional mitochondria through mitophagy suppresses benzo

, Free Radic Biol Med, vol.112, pp.452-463, 2017.

P. O. Seglen, B. Grinde, and A. E. Solheim, Inhibition of the lysosomal pathway of protein degradation in isolated rat hepatocytes by ammonia, methylamine, chloroquine and leupeptin, Eur J Biochem, vol.95, issue.2, pp.215-240, 1979.

M. Mauthe, I. Orhon, C. Rocchi, X. Zhou, M. Luhr et al., Chloroquine inhibits autophagic flux by decreasing autophagosome-lysosome fusion, Autophagy, vol.14, issue.8, pp.1435-1455, 2018.

K. Togashi, S. Wakatsuki, A. Furuno, S. Tokunaga, Y. Nagai et al., Na+/H+ exchangers induce autophagy in neurons and inhibit polyglutamine-induced aggregate formation. PLoS One, vol.8, 2013.

A. V. Berezhnov, M. P. Soutar, E. I. Fedotova, M. S. Frolova, H. Plun-favreau et al., Intracellular pH Modulates Autophagy and Mitophagy, J Biol Chem, vol.291, issue.16, pp.8701-8709, 2016.

R. E. Lawrence and R. Zoncu, The lysosome as a cellular centre for signalling, metabolism and quality control, Nat Cell Biol, vol.21, issue.2, pp.133-142, 2019.

M. Campanella, A. Seraphim, R. Abeti, E. Casswell, P. Echave et al., IF1, the endogenous regulator of the F(1)F(o)-ATPsynthase, defines mitochondrial volume fraction in HeLa cells by regulating autophagy, Biochim Biophys Acta, issue.5, pp.393-401, 1787.

D. J. Roberts, V. P. Tan-sah, E. Y. Ding, J. M. Smith, and S. Miyamoto, Hexokinase-II positively regulates glucose starvation-induced autophagy through TORC1 inhibition, Mol Cell, vol.53, issue.4, pp.521-554, 2014.

X. Zhang, J. L. Tian, Z. Wang, J. Yang, Y. Liu et al., Nitric oxide inhibits autophagy and promotes apoptosis in hepatocellular carcinoma, Cancer Sci, vol.110, issue.3, pp.1054-1063, 2019.

K. Hardonnière, L. Huc, N. Podechard, M. Fernier, X. Tekpli et al., Benzo[a]pyrene-induced nitric oxide production acts as a survival signal targeting mitochondrial membrane potential, Toxicol In Vitro, vol.29, issue.7, pp.1597-608, 2015.

D. Meurette, O. Jouan-lanhouët, S. Rebillard, A. Huc, L. Lagadicgossmann et al., The acidic extracellular pH : Origin, role in tumorigenesis and cancer therapy, Proton homeostasis in tumorigenesis and cell death, pp.105-118, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00682514

C. Corbet and O. Feron, Tumour acidosis: from the passenger to the driver's seat, Nat Rev Cancer, vol.17, issue.10, pp.577-593, 2017.

S. Peppicelli, E. Andreucci, J. Ruzzolini, A. Laurenzana, F. Margheri et al., The acidic microenvironment as a possible niche of dormant tumor cells, Cell Mol Life Sci, vol.74, issue.15, pp.2761-2771, 2017.

T. Morita, T. Nagaki, I. Fukuda, and K. Okumura, Clastogenicity of low pH to various cultured mammalian cells, Mutat Res, vol.268, issue.2, pp.297-305, 1992.

V. R. Jayanth, M. T. Bayne, and M. E. Varnes, Effects of extracellular and intracellular pH on repair of potentially lethal damage, chromosome aberrations and DNA double-strand breaks in irradiated plateau-phase A549 cells, Radiat Res, vol.139, issue.2, pp.152-62, 1994.

J. Yuan, L. Narayanan, S. Rockwell, and P. M. Glazer, Diminished DNA repair and elevated mutagenesis in mammalian cells exposed to hypoxia and low pH, Cancer Res, vol.60, issue.16, pp.4372-4378, 2000.

J. Massonneau, C. Ouellet, F. Lucien, C. M. Dubois, J. Tyler et al., Suboptimal extracellular pH values alter DNA damage response to induced double-strand breaks. FEBS Open Bio, vol.8, pp.416-425, 1935.

Q. Shi, L. Maas, C. Veith, F. J. Van-schooten, and R. W. Godschalk, Acidic cellular microenvironment modifies carcinogen-induced DNA damage and repair, Arch Toxicol, vol.91, issue.6, pp.2425-2441, 2017.

M. Zhao, Q. Liu, Y. Gong, X. Xu, C. Zhang et al., GSHdependent antioxidant defense contributes to the acclimation of colon cancer cells to acidic microenvironment, Cell Cycle, vol.15, issue.8, pp.1125-1158, 2016.

M. Logozzi, D. Mizzoni, D. F. Angelini, D. Raimo, R. Falchi et al., Microenvironmental pH and Exosome Levels Interplay in Human Cancer Cell Lines of Different Histotypes. Cancers (Basel), vol.10, 2018.

M. Tkach and C. Théry, Communication by Extracellular Vesicles: Where We Are and Where We Need to Go, Cell, vol.164, issue.6, pp.1226-1232, 2016.

M. Mathieu, L. Martin-jaular, G. Lavieu, and C. Théry, Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication, Nat Cell Biol, vol.21, issue.1, pp.9-17, 2019.

L. Zhang and D. Yu, Exosomes in cancer development, metastasis, and immunity, Biochim Biophys Acta Rev Cancer, issue.19, pp.30007-30008, 2019.

C. Federici, F. Petrucci, S. Caimi, A. Cesolini, M. Logozzi et al., Exosome release and low pH belong to a framework of resistance of human melanoma cells to cisplatin, PLoS One, vol.9, issue.2, p.88193, 2014.

I. Parolini, C. Federici, C. Raggi, L. Lugini, S. Palleschi et al., Microenvironmental pH is a key factor for exosome traffic in tumor cells, J Biol Chem, vol.284, issue.49, pp.34211-34233, 2009.

D. S. Harischandra, S. Ghaisas, D. Rokad, and A. G. Kanthasamy, Exosomes in Toxicology: Relevance to Chemical Exposure and Pathogenesis of Environmentally Linked Diseases, Toxicol Sci, vol.158, issue.1, pp.3-13, 2017.

K. Y. Neven, T. S. Nawrot, and V. Bollati, Extracellular Vesicles: How the External and Internal Environment Can Shape Cell-To-Cell Communication. Curr Environ Health Rep, vol.4, pp.30-37, 2017.

K. Glunde, S. E. Guggino, M. Solaiyappan, A. P. Pathak, Y. Ichikawa et al., Extracellular acidification alters lysosomal trafficking in human breast cancer cells, Neoplasia, vol.5, issue.6, pp.533-578, 2003.

E. K. Rofstad, B. Mathiesen, K. Kindem, and K. Galappathi, Acidic extracellular pH promotes experimental metastasis of human melanoma cells in athymic nude mice, Cancer Res, vol.66, issue.13, pp.6699-707, 2006.

G. Liu, J. Tang, B. T. Edmonds, J. Murray, S. Levin et al., F-actin sequesters elongation factor 1alpha from interaction with aminoacyl-tRNA in a pH-dependent reaction, J Cell Biol, vol.135, issue.4, pp.953-63, 1996.

D. Meng, D. D. Lv, X. Zhuang, H. Sun, L. Fan et al., Benzo[a]pyrene induces expression of matrix metalloproteinases and cell migration and invasion of vascular smooth muscle cells, Toxicol Lett, vol.184, issue.1, pp.44-53, 2009.

Q. Ba, J. Li, C. Huang, H. Qiu, J. Li et al., Effects of benzo[a]pyrene exposure on human hepatocellular carcinoma cell angiogenesis, metastasis, and NF-?B signaling. Environ Health Perspect, vol.123, pp.246-54, 2015.

M. A. Bumke, D. Neri, and G. Elia, Modulation of gene expression by extracellular pH variations in human fibroblasts: a transcriptomic and proteomic study, Proteomics, vol.3, issue.5, pp.675-88, 2003.

C. Stock, R. A. Cardone, G. Busco, H. Krähling, A. Schwab et al., Protons extruded by NHE1: digestive or glue?, Eur J Cell Biol, vol.87, issue.8-9, pp.591-600, 2008.

Q. Shi, X. Le, B. Wang, J. L. Abbruzzese, Q. Xiong et al., Regulation of vascular endothelial growth factor expression by acidosis in human cancer cells, Oncogene, vol.20, issue.28, pp.3751-3757, 2001.

F. Okajima, Regulation of inflammation by extracellular acidification and proton-sensing GPCRs, Cell Signal, vol.25, issue.11, 2013.

A. Riemann, A. Ihling, S. Reime, M. Gekle, and O. Thews, Impact of the Tumor Microenvironment on the Expression of Inflammatory Mediators in Cancer Cells, Adv Exp Med Biol, vol.923, pp.105-111, 2016.

A. Riemann, S. Reime, and O. Thews, Tumor Acidosis and Hypoxia Differently Modulate the Inflammatory Program: Measurements In Vitro and In Vivo, Neoplasia, vol.19, issue.12, pp.1033-1042, 2017.

V. Huber, C. Camisaschi, A. Berzi, S. Ferro, L. Lugini et al., Cancer acidity: An ultimate frontier of tumor immune escape and a novel target of immunomodulation. Semin Cancer Biol, vol.43, pp.74-89, 2017.

A. Lardner, The effects of extracellular pH on immune function, J Leukoc Biol, vol.69, issue.4, pp.522-552, 2001.

A. Riemann, S. Reime, and O. Thews, Hypoxia-Related Tumor Acidosis Affects MicroRNA Expression Pattern in Prostate and Breast Tumor Cells, Adv Exp Med Biol, vol.977, pp.119-124, 2017.

A. Riemann, S. Reime, and O. Thews, Acidic extracellular environment affects miRNA expression in tumors in vitro and in vivo, Int J Cancer, vol.144, issue.7, pp.1609-1618, 2019.

J. Song, M. Jun, M. R. Ahn, and O. Y. Kim, Involvement of miR-Let7A in inflammatory response and cell survival/apoptosis regulated by resveratrol in THP-1 macrophage, Nutr Res Pract, vol.10, issue.4, pp.377-84, 2016.

J. Eismann, M. Hirschfeld, T. Erbes, G. Rücker, M. Jäger et al., Hypoxia-and acidosis-driven aberrations of secreted microRNAs in endometrial cancer in vitro, Oncol Rep, vol.38, issue.2, pp.993-1004, 2017.

N. Shimokawa, M. Londoño, and N. Koibuchi, Gene expression and signaling pathways by extracellular acidification, Adv Exp Med Biol, vol.580, pp.351-360, 2006.

M. G. Ludwig, M. Vanek, D. Guerini, J. A. Gasser, C. E. Jones et al., Proton-sensing G-protein-coupled receptors, Nature, vol.425, issue.6953, pp.93-101, 2003.

K. Seuwen, M. G. Ludwig, and R. M. Wolf, Receptors for protons or lipid messengers or both?, J Recept Signal Transduct Res, vol.26, issue.5-6, pp.599-610, 2006.

C. Mogi, M. Tobo, H. Tomura, N. Murata, X. D. He et al., Involvement of proton-sensing TDAG8 in extracellular acidification-induced inhibition of proinflammatory cytokine production in peritoneal macrophages, J Immunol, vol.182, issue.5, pp.3243-51, 2009.

W. C. Huang, P. Swietach, R. D. Vaughan-jones, O. Ansorge, and M. D. Glitsch, Extracellular acidification elicits spatially and temporally distinct Ca2+ signals, Curr Biol, vol.18, issue.10, pp.781-785, 2008.

E. J. Sanderlin, N. R. Leffler, K. Lertpiriyapong, Q. Cai, H. Hong et al., GPR4 deficiency alleviates intestinal inflammation in a mouse model of acute experimental colitis, Biochim Biophys Acta Mol Basis Dis, vol.1863, issue.2, pp.569-584, 2017.

L. Sparfel, M. L. Pinel-marie, M. Boize, S. Koscielny, S. Desmots et al., Transcriptional signature of human macrophages exposed to the environmental contaminant benzo(a)pyrene, Toxicol Sci, vol.114, issue.2, pp.247-59, 2010.
URL : https://hal.archives-ouvertes.fr/hal-00657921

S. Z. Wiley, K. Sriram, C. Salmerón, and P. A. Insel, GPR68: An Emerging Drug Target in Cancer, Int J Mol Sci, vol.20, issue.3, 2019.