M. Alic, M. B. Mayfield, L. Akileswaran, and M. H. Gold, Homologous transformation of the lignin-degrading basidiomycete Phanerochaete chrysosporium, Curr Genet, vol.19, pp.491-494, 1991.

F. Besnard, G. Koutsovoulos, S. Dieudonné, M. Blaxter, and M. Félix, Toward Universal Forward Genetics: Using a Draft Genome Sequence of the Nematode Oscheius tipulae To Identify Mutations Affecting Vulva Development, Genetics, vol.206, pp.1747-1761, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01595198

M. Biasini, S. Bienert, A. Waterhouse, K. Arnold, and G. Studer, SWISS-MODEL: modelling protein tertiary and quaternary structure using evolutionary information, Nucleic Acids Res, vol.42, pp.252-258, 2014.

T. M. Diaz-guerra, E. Mellado, M. Cuenca-estrella, and J. L. Rodriguez-tudela, A Point Mutation in the 14?-Sterol Demethylase Gene cyp51A Contributes to Itraconazole Resistance in Aspergillus fumigatus, Antimicrobial Agents and Chemotherapy, vol.48, pp.1071-1071, 2004.

D. C. Eastwood, D. Floudas, M. Binder, A. Majcherczyk, and P. Schneider, The Plant Cell Wall-Decomposing Machinery Underlies the Functional Diversity of Forest Fungi, Science, vol.333, pp.762-765, 2011.
URL : https://hal.archives-ouvertes.fr/hal-02647602

D. Floudas, M. Binder, R. Riley, K. Barry, and R. A. Blanchette, The Paleozoic Origin of, 2012.
URL : https://hal.archives-ouvertes.fr/hal-01268324

, Enzymatic Lignin Decomposition Reconstructed from 31 Fungal Genomes, Science, vol.336, pp.1715-1719

S. A. Flowers, B. Colón, S. G. Whaley, M. A. Schuler, and P. D. Rogers, Contribution of Clinically Derived Mutations in ERG11 to Azole Resistance in Candida albicans, Antimicrobial Agents and Chemotherapy, vol.59, pp.450-460, 2015.

P. Gong, X. Guan, and E. Witter, A rapid method to extract ergosterol from soil by physical disruption, Applied Soil Ecology, vol.17, pp.285-289, 2001.

T. Y. Hargrove, Z. Wawrzak, D. C. Lamb, F. P. Guengerich, and G. I. Lepesheva, Structure-Functional Characterization of Cytochrome P450 Sterol 14?-Demethylase (CYP51B) from Aspergillus fumigatus and Molecular Basis for the Development of Antifungal Drugs, J. Biol. Chem, vol.290, pp.23916-23934, 2015.

J. Jeon, J. Choi, G. Lee, R. A. Dean, and Y. Lee, Experimental Evolution Reveals Genome-Wide Spectrum and Dynamics of Mutations in the Rice Blast Fungus, Magnaporthe oryzae, PLOS ONE, vol.8, p.65416, 2013.

P. Kersten and D. Cullen, Extracellular oxidative systems of the lignin-degrading Basidiomycete Phanerochaete chrysosporium, Fungal Genetics and Biology, vol.44, pp.77-87, 2007.

S. Wullschleger, R. Loewith, and M. N. Hall, TOR Signaling in Growth and Metabolism. Cell, vol.124, pp.471-484, 2006.

P. G. Bertram, J. H. Choi, J. Carvalho, T. Chan, A. W. Zheng et al., Convergence of TOR-Nitrogen and Snf1-Glucose Signaling Pathways onto Gln3, Molecular and Cellular Biology, vol.22, pp.1246-1252, 2002.

D. Stracka, S. Jozefczuk, F. Rudroff, U. Sauer, and M. N. Hall, Nitrogen Source Activates TOR (Target of Rapamycin) Complex 1 via Glutamine and Independently of Gtr/Rag Proteins, J Biol Chem, vol.289, pp.25010-25020, 2014.

J. L. Crespo, K. Daicho, T. Ushimaru, and M. N. Hall, The GATA Transcription Factors GLN3 and GAT1 Link TOR to Salt Stress in Saccharomyces cerevisiae, J Biol Chem, vol.276, pp.34441-34444, 2001.

B. J. Niles, A. C. Joslin, T. Fresques, and T. Powers, TOR Complex 2-Ypk1 signaling maintains sphingolipid homeostasis by sensing and regulating ROS accumulation, Cell Rep, vol.6, pp.541-552, 2014.

R. Weisman and M. Choder, The Fission Yeast TOR Homolog, tor1 +, Is Required for the Response to Starvation and Other Stresses via a Conserved Serine, J Biol Chem, vol.276, pp.7027-7032, 2001.

J. Heitman, N. R. Movva, and M. N. Hall, Targets for cell cycle arrest by the immunosuppressant rapamycin in yeast, Science, vol.253, pp.905-909, 1991.

R. Loewith, J. E. Wullschleger, S. Lorberg, A. Crespo, J. L. Bonenfant et al., Two TOR Complexes, Only One of which Is Rapamycin Sensitive, Have Distinct Roles in Cell Growth Control, Molecular Cell, vol.10, pp.457-468, 2002.

X. Zheng, D. Fiorentino, J. Chen, G. R. Crabtree, and S. L. Schreiber, TOR kinase domains are required for two distinct functions, only one of which is inhibited by rapamycin, Cell, vol.82, pp.90058-90064, 1995.

S. N. Sehgal, H. Baker, and C. Vézina, RAPAMYCIN (AY-22, 989), A NEW ANTIFUNGAL ANTIBIOTIC, J Antibiot, vol.28, pp.727-732, 1975.

M. C. Cruz, L. M. Cavallo, J. M. Görlach, G. Cox, J. R. Perfect et al., Rapamycin Antifungal Action Is Mediated via Conserved Complexes with FKBP12 and TOR Kinase Homologs in Cryptococcus neoformans, Molecular and Cellular Biology, vol.19, pp.4101-4112, 1999.

H. G. Meléndez, G. Billon-grand, M. Fèvre, and G. Mey, Role of the Botrytis cinerea FKBP12 ortholog in pathogenic development and in sulfur regulation, Fungal Genetics and Biology, vol.46, pp.308-320, 2009.

R. J. Bastidas, C. A. Shertz, S. C. Lee, J. Heitman, and M. E. Cardenas, Rapamycin Exerts Antifungal Activity In Vitro and In Vivo against Mucor circinelloides via FKBP12-Dependent Inhibition of Tor, Eukaryotic Cell, vol.11, pp.270-281, 2012.

F. Yu, Q. Gu, Y. Yun, Y. Yin, J. Xu et al., The TOR signaling pathway regulates vegetative development and virulence in Fusarium graminearum, New Phytologist, vol.203, pp.219-232, 2014.

K. Dementhon, M. Paoletti, B. Pinan-lucarré, N. Loubradou-bourges, M. Sabourin et al., Rapamycin Mimics the Incompatibility Reaction in the Fungus Podospora anserina, Eukaryot Cell, vol.2, pp.238-246, 2003.

C. A. Shertz, R. J. Bastidas, W. Li, J. Heitman, and M. E. Cardenas, Conservation, duplication, and loss of the Tor signaling pathway in the fungal kingdom, BMC Genomics, vol.11, p.510, 2010.

D. Chen, L. Shi, S. Yue, T. Zhang, S. Wang et al., The Slt2-MAPK pathway is involved in the mechanism by which target of rapamycin regulates cell wall components in Ganoderma lucidum, Fungal Genetics and Biology, vol.123, pp.70-77, 2019.

N. Valette, T. Perrot, R. Sormani, E. Gelhaye, and M. Morel-rouhier, Antifungal activities of wood extractives, Fungal Biology Reviews, vol.31, pp.113-123, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01595725

D. C. Eastwood, D. Floudas, M. Binder, A. Majcherczyk, P. Schneider et al., The Plant Cell Wall-Decomposing Machinery Underlies the Functional Diversity of Forest Fungi, Science, vol.333, pp.762-765, 2011.
URL : https://hal.archives-ouvertes.fr/hal-02647602

D. Floudas, M. Binder, R. Riley, K. Barry, R. A. Blanchette et al., The Paleozoic Origin of Enzymatic Lignin Decomposition Reconstructed from 31 Fungal Genomes, Science, vol.336, pp.1715-1719, 2012.
URL : https://hal.archives-ouvertes.fr/hal-01268324

M. Morel, E. Meux, Y. Mathieu, A. Thuillier, K. Chibani et al., Xenomic networks variability and adaptation traits in wood decaying fungi, Microbial Biotechnology, vol.6, pp.248-263, 2013.
URL : https://hal.archives-ouvertes.fr/hal-01268111

L. G. Nagy, R. Riley, P. J. Bergmann, K. Krizsán, F. M. Martin et al., Genetic Bases of Fungal White Rot Wood Decay Predicted by Phylogenomic Analysis of Correlated Gene-Phenotype Evolution, Mol Biol Evol, vol.34, pp.35-44, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01551070

D. Martinez, L. F. Larrondo, N. Putnam, M. Gelpke, K. Huang et al., Genome sequence of the lignocellulose degrading fungus Phanerochaete chrysosporium strain RP78, Nature Biotechnology, vol.22, pp.695-700, 2004.

M. Tien and T. K. Kirk, Lignin-Degrading Enzyme from the Hymenomycete Phanerochaete chrysosporium Burds, Science, vol.221, pp.661-663, 1983.

A. Joubert, B. Calmes, R. Berruyer, M. Pihet, J. Bouchara et al., Laser nephelometry applied in an automated microplate system to study filamentous fungus growth, BioTechniques, vol.48, pp.399-404, 2010.
URL : https://hal.archives-ouvertes.fr/hal-02662333

R. D. Finn, A. Bateman, J. Clements, P. Coggill, R. Y. Eberhardt et al., Pfam: the protein families database, Nucleic Acids Res, vol.42, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01294685

E. Harsay and R. Schekman, Avl9p, a Member of a Novel Protein Superfamily, Functions in the Late Secretory Pathway, Mol. Biol. Cell, vol.18, pp.1203-1219, 2007.

A. Joubert, B. Calmes, R. Berruyer, M. Pihet, J. Bouchara et al., Laser nephelometry applied in an automated microplate system to study filamentous fungus growth, BioTechniques, vol.48, pp.399-404, 2010.
URL : https://hal.archives-ouvertes.fr/hal-02662333

C. U. Köser, M. J. Ellington, and S. J. Peacock, Whole-genome sequencing to control antimicrobial resistance, Trends Genet, vol.30, pp.401-407, 2014.

T. P. Levine, R. D. Daniels, A. T. Gatta, L. H. Wong, and M. J. Hayes, The product of C9orf72, a gene strongly implicated in neurodegeneration, is structurally related to DENN Rab-GEFs, Bioinformatics, vol.29, pp.499-503, 2013.

H. Li, A statistical framework for SNP calling, mutation discovery, association mapping and population genetical parameter estimation from sequencing data, Bioinformatics, vol.27, pp.2987-2993, 2011.

,

H. Li and R. Durbin, Fast and accurate short read alignment with Burrows-Wheeler transform, Bioinformatics, vol.25, pp.1754-1760, 2009.

A. Linford, S. Yoshimura, R. N. Bastos, L. Langemeyer, A. Gerondopoulos et al., Rab14 and Its Exchange Factor FAM116 Link Endocytic Recycling and Adherens Junction Stability in Migrating Cells, Dev. Cell, vol.22, pp.952-966, 2012.

A. L. Marat, H. Dokainish, and P. S. Mcpherson, DENN Domain Proteins: Regulators of Rab GTPases, 2011.

, J. Biol. Chem, vol.286, pp.13791-13800

D. Martinez, L. F. Larrondo, N. Putnam, M. D. Gelpke, K. Huang et al.,

P. Ramaiya, J. C. Detter, F. Larimer, P. M. Coutinho, B. Henrissat et al., Genome sequence of the lignocellulose degrading fungus Phanerochaete chrysosporium strain RP78, Nat. Biotechnol, vol.22, pp.695-700, 2004.

Y. Mathieu, P. Prosper, M. Buée, S. Dumarçay, F. Favier et al.,

J. Jacquot, T. Lamant, E. Meux, S. Mathiot, C. Didierjean et al., Characterization of a Phanerochaete chrysosporium Glutathione Transferase Reveals a Novel Structural and Functional Class with Ligandin Properties, J. Biol. Chem, vol.287, pp.39001-39011, 2012.
URL : https://hal.archives-ouvertes.fr/hal-01268261

,

M. Morel, E. Meux, Y. Mathieu, A. Thuillier, K. Chibani et al., Xenomic networks variability and adaptation traits in wood decaying fungi, Microb. Biotechnol, vol.6, pp.248-263, 2013.
URL : https://hal.archives-ouvertes.fr/hal-01268111

K. J. Nordström, M. C. Albani, G. V. James, C. Gutjahr, B. Hartwig et al.,

G. Coupland and K. Schneeberger, Mutation identification by direct comparison of whole-genome sequencing data from mutant and wild-type individuals using k -mers, Nat. Biotechnol, vol.31, pp.325-330, 2013.

,

R. A. Ohm, R. Riley, A. Salamov, B. Min, I. Choi et al., Genomics of wood-degrading fungi, Biomass Degradation by Fungi, vol.72, pp.82-90, 2014.

,

T. Perrot, M. Schwartz, F. Saiag, G. Salzet, S. Dumarçay et al., , 2018.

, Chem. Eng, vol.6, pp.13078-13085

M. Royer, G. Herbette, V. Eparvier, J. Beauchêne, B. Thibaut et al., Secondary metabolites of Bagassa guianensis Aubl. wood: A study of the chemotaxonomy of the Moraceae family, Phytochemistry, vol.71, pp.1708-1713, 2010.
URL : https://hal.archives-ouvertes.fr/hal-00857200

K. Schneeberger, Using next-generation sequencing to isolate mutant genes from forward genetic screens, Nat. Rev. Genet, vol.15, pp.662-676, 2014.

K. Schneeberger and D. Weigel, Fast-forward genetics enabled by new sequencing technologies, Trends Plant Sci, vol.16, pp.282-288, 2011.

H. Sun and K. Schneeberger, SHOREmap v3.0: Fast and Accurate Identification of Causal Mutations from Forward Genetic Screens, Plant Functional Genomics: Methods and Protocols, pp.381-395, 2015.

A. Thuillier, K. Chibani, G. Belli, E. Herrero, S. Dumarçay et al.,

T. Dhalleine, R. Bchini, J. Jacquot, E. Gelhaye, and M. Morel-rouhier, Transcriptomic Responses of Phanerochaete chrysosporium to Oak Acetonic Extracts, Focus on a New Glutathione Transferase, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01268964

, Appl Env. Microbiol, vol.80, pp.6316-6327

M. Tien and T. K. Kirk, Lignin-Degrading Enzyme from the Hymenomycete Phanerochaete chrysosporium, Burds. Science, vol.221, pp.661-663, 1983.

S. Zuryn, S. L. Gras, K. Jamet, and S. Jarriault, A Strategy for Direct Mapping and Identification of REFERENCES AND BIBLIOGRAPHY 182, 2010.

C. Abdallah, E. Dumas-gaudot, J. Renaut, and K. Sergeant, Gel-Based and Gel-Free Quantitative Proteomics Approaches at a Glance, International Journal of Plant Genomics, 2012.
URL : https://hal.archives-ouvertes.fr/hal-01004759

A. Abdolrasouli, J. Rhodes, M. A. Beale, F. Hagen, T. R. Rogers et al., Genomic Context of Azole Resistance Mutations in Aspergillus fumigatus Determined Using Whole-Genome Sequencing, 2015.

S. S. Adav, E. S. Cheow, A. Ravindran, B. Dutta, and S. K. Sze, , 2012.

K. J. Affeldt, J. Carrig, M. Amare, and N. P. Keller, Global Survey of Canonical Aspergillus flavus G Protein-Coupled Receptors, vol.5, pp.1501-1515, 2014.

R. Ahlgren, G. Suske, M. R. Waterman, and J. Lund, Role of Sp1 in cAMP-dependent Transcriptional Regulation of the Bovine CYP11A Gene, J. Biol. Chem, vol.274, pp.19422-19428, 1999.

A. Alarco, R. , and M. , The bZip Transcription Factor Cap1p Is Involved in Multidrug Resistance and Oxidative Stress Response in Candida albicans, J. Bacteriol, vol.181, pp.700-708, 1999.

M. Alfaro, J. A. Oguiza, L. Ramírez, and A. G. Pisabarro, Comparative analysis of secretomes in basidiomycete fungi, J. Proteomics, vol.102, pp.28-43, 2014.

M. Alic, M. B. Mayfield, L. Akileswaran, and M. H. Gold, Homologous transformation of the lignin-degrading basidiomycete Phanerochaete chrysosporium, Curr. Genet, vol.19, pp.491-494, 1991.

A. Altmann, P. Weber, D. Bader, M. Preuß, E. B. Binder et al., A beginners guide to SNP calling from high-throughput DNA-sequencing data, Hum. Genet, vol.131, pp.1541-1554, 2012.

B. Alvarez and S. Moreno, Fission yeast Tor2 promotes cell growth and represses cell differentiation, J. Cell Sci, vol.119, pp.4475-4485, 2006.

J. S. Anouhe, F. B. Niamké, M. Faustin, D. Virieux, J. Pirat et al., The role of extractives in the natural durability of the heartwood of Dicorynia guianensis Amsh: new insights in antioxydant and antifungal properties, Ann. For. Sci, vol.75, p.15, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01779497

A. Anttila, A. M. Pirttilä, H. Häggman, A. Harju, M. Venäläinen et al., Condensed conifer tannins as antifungal agents in liquid culture, Holzforschung, vol.67, pp.825-832, 2013.

V. Arantes and B. Goodell, Current Understanding of Brown-Rot Fungal Biodegradation Mechanisms: A Review, Deterioration and Protection of Sustainable Biomaterials, pp.3-21, 2014.

A. Arbenz, A. , and L. , Chemical modification of tannins to elaborate aromatic biobased macromolecular architectures, Green Chem, vol.17, pp.2626-2646, 2015.

P. Arya, A. , and V. , Plant STAND P-loop NTPases: a current perspective of genome distribution, evolution, and function, Mol. Genet. Genomics, vol.293, pp.17-31, 2018.

Y. Bahn, C. Xue, A. Idnurm, J. C. Rutherford, J. Heitman et al., Sensing the environment: lessons from fungi, Nat. Rev. Microbiol, vol.5, pp.57-69, 2007.

E. Y. Basenko, J. A. Pulman, A. Shanmugasundram, O. S. Harb, K. Crouch et al., FungiDB: An Integrated Bioinformatic Resource for Fungi and Oomycetes, J. Fungi, vol.4, p.39, 2018.

R. J. Bastidas, C. A. Shertz, S. C. Lee, J. Heitman, and M. E. Cardenas, Rapamycin Exerts Antifungal Activity In Vitro and In Vivo against Mucor circinelloides via FKBP12-Dependent Inhibition of Tor, Eukaryot. Cell, vol.11, pp.270-281, 2012.

Ö. Bayram and G. H. Braus, Coordination of secondary metabolism and development in fungi: the velvet family of regulatory proteins, FEMS Microbiol. Rev, vol.36, pp.1-24, 2012.

T. Belt, T. Keplinger, T. Hänninen, and L. Rautkari, Cellular level distributions of Scots pine heartwood and knot heartwood extractives revealed by Raman spectroscopy imaging, Ind. Crops Prod, vol.108, pp.327-335, 2017.

G. Bendavit, T. Aboulkassim, K. Hilmi, S. Shah, and G. Batist, Nrf2 Transcription Factor Can Directly Regulate mTOR linking cytoprotective gene expression to a major metablolic regulator that generates redox activity, J. Biol. Chem, vol.291, pp.25476-25488, 2016.

P. G. Bertram, J. H. Choi, J. Carvalho, T. Chan, W. Ai et al., Convergence of TOR-Nitrogen and Snf1-Glucose Signaling Pathways onto Gln3, Mol. Cell. Biol, vol.22, pp.1246-1252, 2002.

R. Bertrand-teponno, S. Kusari, and M. Spiteller, Recent advances in research on lignans and neolignans, Nat. Prod. Rep, vol.33, pp.1044-1092, 2016.

F. Besnard, G. Koutsovoulos, S. Dieudonné, M. Blaxter, and M. Félix, Toward Universal Forward Genetics: Using a Draft Genome Sequence of the Nematode Oscheius tipulae To Identify Mutations Affecting Vulva Development, Genetics, vol.206, pp.1747-1761, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01595198

L. Bianco and G. Perrotta, Methodologies and Perspectives of Proteomics Applied to Filamentous Fungi: From Sample Preparation to Secretome Analysis, Int. J. Mol. Sci, vol.16, pp.5803-5829, 2015.

M. Biasini, S. Bienert, A. Waterhouse, K. Arnold, G. Studer et al., SWISS-MODEL: modelling protein tertiary and quaternary structure using evolutionary information, Nucleic Acids Res, vol.42, pp.252-258, 2014.

R. A. Blanchette, A review of microbial deterioration found in archaeological wood from different environments, Int. Biodeterior. Biodegrad, vol.46, pp.189-204, 2000.

M. Boeckstaens, B. André, and A. M. Marini, The yeast ammonium transport protein Mep2 and its positive regulator, the Npr1 kinase, play an important role in normal and pseudohyphal growth on various nitrogen media through retrieval of excreted ammonium, Mol. Microbiol, vol.64, pp.534-546, 2007.

K. A. Braun, S. Vaga, K. M. Dombek, F. Fang, S. Palmisano et al., , 2014.

, Phosphoproteomic analysis identifies proteins involved in transcription-coupled mRNA decay as targets of Snf1 signaling, Sci. Signal, vol.7, pp.64-64

C. Braunsdorf, D. Mailänder-sánchez, and M. Schaller, Fungal sensing of host environment, Cell. Microbiol, vol.18, pp.1188-1200, 2016.

D. Bridges, K. Fisher, S. N. Zolov, T. Xiong, K. Inoki et al., Rab5 proteins regulate activation and localization of target of rapamycin complex 1, J. Biol. Chem, vol.287, pp.20913-20921, 2012.

V. F. Brocco, J. B. Paes, L. G. Costa, . Da, S. Brazolin et al., Potential of teak heartwood extracts as a natural wood preservative, J. Clean. Prod, vol.142, pp.2093-2099, 2017.

G. Brosch, P. Loidl, and S. Graessle, Histone modifications and chromatin dynamics: a focus on filamentous fungi, FEMS Microbiol. Rev, vol.32, pp.409-439, 2008.

A. J. Brown, L. E. Cowen, A. Pietro, Q. , and J. , Stress Adaptation. Microbiol. Spectr, 2017.

N. A. Brown, P. F. De-gouvea, N. G. Krohn, M. Savoldi, and G. H. Goldman, Functional characterisation of the non-essential protein kinases and phosphatases regulating Aspergillus nidulans hydrolytic enzyme production, Biotechnol. Biofuels, vol.6, p.91, 2013.

N. A. Brown, L. N. Ries, and G. H. Goldman, How nutritional status signalling coordinates metabolism and lignocellulolytic enzyme secretion, Fungal Genet. Biol, vol.72, pp.48-63, 2014.

N. A. Brown, T. F. Reis, L. N. Ries, C. Caldana, J. Mah et al., G-protein coupled receptor-mediated nutrient sensing and developmental control in Aspergillus nidulans, Mol. Microbiol, vol.98, pp.420-439, 2015.

P. Cai, B. Wang, J. Ji, Y. Jiang, L. Wan et al., The Putative Cellodextrin Transporter-like Protein CLP1 Is Involved in Cellulase Induction in Neurospora crassa, J. Biol, 2015.

, Chem, vol.290, pp.788-796

M. D. Cameron, S. Timofeevski, and S. D. Aust, Enzymology of Phanerochaete chrysosporium with respect to the degradation of recalcitrant compounds and xenobiotics, Appl. Microbiol. Biotechnol, vol.54, pp.751-758, 2000.

S. M. Camps, B. E. Dutilh, M. C. Arendrup, A. J. Rijs, E. Snelders et al., Discovery of a hapE Mutation That Causes Azole Resistance in Aspergillus fumigatus through Whole Genome Sequencing and Sexual Crossing, PLOS ONE, vol.7, p.50034, 2012.

C. Casado, A. González, M. Platara, A. Ruiz, A. et al., The role of the protein kinase A pathway in the response to alkaline pH stress in yeast, Biochem. J, vol.438, pp.523-533, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00617329

P. A. Castro, . De, T. F. Reis, S. K. Dolan, A. O. Manfiolli et al., The Aspergillus fumigatus SchASCH9 kinase modulates SakAHOG1 MAP kinase activity and it is essential for virulence, Mol. Microbiol, vol.102, pp.642-671, 2016.

C. C. Celimene, J. A. Micales, L. Ferge, Y. , and R. A. , Efficacy of Pinosylvins against White-Rot and Brown-Rot Fungi, Holzforschung, vol.53, pp.491-497, 2005.

C. L. Céspedes, J. G. Avila, A. M. Garc?á, J. Becerra, C. Flores et al., Antifungal and Antibacterial Activities of Araucaria araucana, Z. Für Naturforschung C, vol.61, pp.35-43, 2014.

S. Chang, S. Wang, C. Wu, Y. Su, and Y. Kuo, , 2005.

, Ethyl Acetate Soluble Fraction of the Extractives of Taiwania (Taiwania cryptomerioides Hayata)

, Heartwood. Holzforschung, vol.53, pp.487-490

D. Chen, Y. Wang, X. Zhou, Y. Wang, and J. Xu, The Sch9 Kinase Regulates Conidium Size, Stress Responses, and Pathogenesis in Fusarium graminearum, PLOS ONE, vol.9, 2014.

D. Chen, L. Shi, S. Yue, T. Zhang, S. Wang et al., The Slt2-MAPK pathway is involved in the mechanism by which target of rapamycin regulates cell wall components in Ganoderma lucidum, Fungal Genet. Biol, vol.123, pp.70-77, 2019.

Y. Chen, T. Yeh, F. Chu, F. Hsu, C. et al., Proteomics Investigation Reveals Cell Death-Associated Proteins of Basidiomycete Fungus Trametes versicolor Treated with Ferruginol, J. Agric. Food Chem, vol.63, pp.85-91, 2015.

Y. Chen, C. Lin, P. Tsai, C. Yang, W. Hsieh et al., Rhb1 Regulates the Expression of Secreted Aspartic Protease 2 through the TOR Signaling Pathway in Candida albicans, Eukaryot. Cell, vol.11, pp.168-182, 2012.

Y. Chen, Y. Yeh, H. Chen, R. Chen, G. Lin et al., , 2019.

J. Choi, J. Chen, S. L. Schreiber, and J. Clardy, Structure of the FKBP12-Rapamycin Complex Interacting with Binding Domain of Human FRAP, Science, vol.273, pp.239-242, 1996.

J. L. Crespo, K. Daicho, T. Ushimaru, and M. N. Hall, The GATA Transcription Factors GLN3 and GAT1 Link TOR to Salt Stress in Saccharomyces cerevisiae, J. Biol. Chem, vol.276, pp.34441-34444, 2001.

M. C. Cruz, L. M. Cavallo, J. M. Görlach, G. Cox, J. R. Perfect et al., , 1999.

, Rapamycin Antifungal Action Is Mediated via Conserved Complexes with FKBP12 and TOR Kinase Homologs in Cryptococcus neoformans, Mol. Cell. Biol, vol.19, pp.4101-4112

T. H. De-koker, K. K. Nakasone, J. Haarhof, H. H. Burdsall, and B. J. Janse, Phylogenetic relationships of the genus Phanerochaete inferred from the internal transcribed spacer region, 2003.

, Mycol. Res, vol.107, pp.1032-1040

M. Delic, M. Valli, A. B. Graf, M. Pfeffer, D. Mattanovich et al., The secretory pathway: exploring yeast diversity, FEMS Microbiol. Rev, vol.37, pp.872-914, 2013.

K. Dementhon and S. J. Saupe, DNA-Binding Specificity of the IDI-4 Basic Leucine Zipper Factor of Podospora anserina Defined by Systematic Evolution of Ligands by Exponential Enrichment (SELEX), Eukaryot. Cell, vol.4, pp.476-483, 2005.

K. Dementhon, M. Paoletti, B. Pinan-lucarré, N. Loubradou-bourges, M. Sabourin et al., Rapamycin Mimics the Incompatibility Reaction in the Fungus Podospora anserina, Eukaryot. Cell, vol.2, pp.238-246, 2003.

K. Dementhon, S. J. Saupe, and C. Clavé, Characterization of IDI-4, a bZIP transcription factor inducing autophagy and cell death in the fungus Podospora anserina, Mol. Microbiol, vol.53, pp.1625-1640, 2004.

A. Deroy, F. Saiag, Z. Kebbi-benkeder, N. Touahri, A. Hecker et al., The GSTome Reflects the Chemical Environment of White-Rot Fungi, PLOS ONE, vol.10, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01269448

T. M. Diaz-guerra, E. Mellado, M. Cuenca-estrella, and J. L. Rodriguez-tudela, A Point Mutation in the 14?-Sterol Demethylase Gene cyp51A Contributes to Itraconazole Resistance in Aspergillus fumigatus, Antimicrob. Agents Chemother, vol.48, pp.1071-1071, 2004.

P. V. Dijck, N. A. Brown, G. H. Goldman, J. Rutherford, C. Xue et al., Nutrient Sensing at the Plasma Membrane of Fungal Cells, Microbiol. Spectr, vol.5, 2017.

L. Ding, W. Li, N. Li, L. Liang, X. Zhang et al., , 2019.

, Antioxidant responses to salinity stress in an invasive species, the red-eared slider (Trachemys scripta elegans) and involvement of a TOR-Nrf2 signaling pathway, Comp. Biochem. Physiol. Part C Toxicol. Pharmacol, vol.219, pp.59-67

H. Doddapaneni and J. S. Yadav, Microarray-based global differential expression profiling of P450 monooxygenases and regulatory proteins for signal transduction pathways in the white rot fungus Phanerochaete chrysosporium, Mol. Genet. Genomics, vol.274, pp.454-466, 2005.

H. C. Van-der-does and M. Rep, Adaptation to the Host Environment by Plant-Pathogenic Fungi, Annu. Rev. Phytopathol, vol.55, pp.427-450, 2017.

M. C. Donaton, I. Holsbeeks, O. Lagatie, G. V. Zeebroeck, M. Crauwels et al., The Gap1 general amino acid permease acts as an amino acid sensor for activation of protein kinase A targets in the yeast Saccharomyces cerevisiae, Mol. Microbiol, vol.50, pp.911-929, 2003.

W. Dyrka, M. Lamacchia, P. Durrens, B. Kobe, A. Daskalov et al., Diversity and Variability of NOD-Like Receptors in Fungi, Genome Biol. Evol, vol.6, pp.3137-3158, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01083450

D. C. Eastwood, D. Floudas, M. Binder, A. Majcherczyk, P. Schneider et al., The Plant Cell Wall-Decomposing Machinery Underlies the Functional Diversity of Forest Fungi, Science, vol.333, pp.762-765, 2011.
URL : https://hal.archives-ouvertes.fr/hal-02647602

B. Enjalbert, D. A. Smith, M. J. Cornell, I. Alam, S. Nicholls et al., Role of the Hog1 Stress-activated Protein Kinase in the Global Transcriptional Response to Stress in the Fungal Pathogen Candida albicans, Mol. Biol. Cell, vol.17, pp.1018-1032, 2005.
URL : https://hal.archives-ouvertes.fr/hal-01894209

E. Espagne, P. Balhadère, M. Penin, C. Barreau, and B. Turcq, HET-E and HET-D belong, 2002.

M. L. Falcone-ferreyra, S. Rius, and P. Casati, Flavonoids: biosynthesis, biological functions, and biotechnological applications, Front. Plant Sci, vol.3, 2012.

K. Falloon, P. R. Juvvadi, A. D. Richards, J. M. Vargas-muñiz, H. Renshaw et al., Characterization of the FKBP12-Encoding Genes in Aspergillus fumigatus, PLOS ONE, vol.10, 2015.

A. J. Fernández-gonzález, N. Valette, A. Kohler, S. Dumarçay, R. Sormani et al., Oak extractive-induced stress reveals the involvement of new enzymes in the early detoxification response of Phanerochaete chrysosporium, Environ. Microbiol, vol.20, pp.3890-3901, 2018.

R. D. Finn, A. Bateman, J. Clements, P. Coggill, R. Y. Eberhardt et al., Pfam: the protein families database, Nucleic Acids Res, vol.42, pp.222-230, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01294685

G. J. Fitzgibbon, I. Y. Morozov, M. G. Jones, and M. X. Caddick, Genetic Analysis of the TOR Pathway in Aspergillus nidulans, Eukaryot. Cell, vol.4, pp.1595-1598, 2005.

D. Floudas, M. Binder, R. Riley, K. Barry, R. A. Blanchette et al., The Paleozoic Origin of Enzymatic Lignin Decomposition Reconstructed from 31 Fungal Genomes, Science, vol.336, pp.1715-1719, 2012.
URL : https://hal.archives-ouvertes.fr/hal-01268324

S. A. Flowers, B. Colón, S. G. Whaley, M. A. Schuler, and P. D. Rogers, Contribution of Clinically Derived Mutations in ERG11 to Azole Resistance in Candida albicans, Antimicrob. Agents Chemother, vol.59, pp.450-460, 2015.

M. H. Fulekar, B. Pathak, J. Fulekar, T. E. Godambe, and Y. R. Goltapeh, Bioremediation of Organic Pollutants Using Phanerochaete chrysosporium, Fungi as Bioremediators, 2013.

R. Gacesa, W. C. Dunlap, and P. F. Long, Bioinformatics analyses provide insight into distant homology of the Keap1-Nrf2 pathway. Free Radic, Biol. Med, vol.88, pp.373-380, 2015.

I. García, M. Mathieu, I. Nikolaev, B. Felenbok, and C. Scazzocchio, Roles of the Aspergillus nidulans homologues of Tup1 and Ssn6 in chromatin structure and cell viability, FEMS Microbiol, 2008.

. Lett, , vol.289, pp.146-154

R. García-salcedo, T. Lubitz, G. Beltran, K. Elbing, Y. Tian et al., Glucose de-repression by yeast AMP-activated protein kinase SNF1 is controlled via at least two independent steps, FEBS J, vol.281, pp.1901-1917, 2014.

A. P. Gonçalves, J. Heller, A. Daskalov, A. Videira, and N. L. Glass, Regulated Forms of, Cell Death in Fungi. Front. Microbiol, vol.8, p.1837, 2017.

P. Gong, X. Guan, and E. Witter, A rapid method to extract ergosterol from soil by physical disruption, Appl. Soil Ecol, vol.17, pp.285-289, 2001.

A. González and M. N. Hall, Nutrient sensing and TOR signaling in yeast and mammals, 2017.

, EMBO J, vol.36, pp.397-408

A. González, M. Shimobayashi, T. Eisenberg, D. A. Merle, T. Pendl et al.,

, TORC1 Promotes Phosphorylation of Ribosomal Protein S6 via the AGC Kinase Ypk3 in Saccharomyces cerevisiae, PLoS ONE, vol.10

D. Hagiwara, Y. Asano, T. Yamashino, M. , and T. , Characterization of bZip-Type Transcription Factor AtfA with Reference to Stress Responses of Conidia of Aspergillus nidulans, 2008.

, Biosci. Biotechnol. Biochem, vol.72, pp.2756-2760

D. Hagiwara, Y. Asano, J. Marui, A. Yoshimi, T. Mizuno et al., Transcriptional profiling for Aspergillus nidulans HogA MAPK signaling pathway in response to fludioxonil and osmotic stress, Fungal Genet. Biol, vol.46, pp.868-878, 2009.

D. Hagiwara, K. Sakamoto, K. Abe, and K. Gomi, Signaling pathways for stress responses and adaptation in Aspergillus species: stress biology in the post-genomic era, Biosci. Biotechnol. Biochem, vol.80, pp.1667-1680, 2016.

K. E. Hammel, A. N. Kapich, K. A. Jensen, and Z. C. Ryan, Reactive oxygen species as agents of wood decay by fungi, Enzyme Microb. Technol, vol.30, pp.445-453, 2002.

T. Y. Hargrove, Z. Wawrzak, D. C. Lamb, F. P. Guengerich, and G. I. Lepesheva, Structure-Functional Characterization of Cytochrome P450, 2015.

, Aspergillus fumigatus and Molecular Basis for the Development of Antifungal Drugs, J. Biol. Chem, vol.290, pp.23916-23934

A. M. Harju and M. Venäläinen, Genetic Parameters Regarding the Resistance of Pinus sylvestris Heartwood to Decay Caused by Coniophora puteana, Scand. J. For. Res, vol.17, pp.199-205, 2002.

S. D. Harris, Hyphal branching in filamentous fungi, Dev. Biol, vol.451, pp.35-39, 2019.

E. Harsay and R. Schekman, Avl9p, a Member of a Novel Protein Superfamily, Functions in the Late Secretory Pathway, Mol. Biol. Cell, vol.18, pp.1203-1219, 2007.

A. Hatakka and K. E. Hammel, Fungal Biodegradation of Lignocelluloses, Industrial Applications, M. Hofrichter, pp.319-340, 2011.

B. M. Hayes, M. A. Anderson, A. Traven, N. L. Van-der-weerden, and M. R. Bleackley, , 2014.

, Activation of stress signalling pathways enhances tolerance of fungi to chemical fungicides and antifungal proteins, Cell. Mol. Life Sci, vol.71, pp.2651-2666

J. Heitman, N. R. Movva, and M. N. Hall, Targets for cell cycle arrest by the immunosuppressant rapamycin in yeast, Science, vol.253, pp.905-909, 1991.

S. Hirosue, M. Tazaki, N. Hiratsuka, S. Yanai, H. Kabumoto et al., Insight into functional diversity of cytochrome P450 in the white-rot basidiomycete Phanerochaete chrysosporium: Involvement of versatile monooxygenase, Biochem. Biophys. Res. Commun, vol.407, pp.118-123, 2011.

X. Huang and A. Madan, CAP3: A DNA Sequence Assembly Program, Genome Res, vol.9, pp.868-877, 1999.

L. B. Huberman, J. Liu, L. Qin, and N. L. Glass, Regulation of the lignocellulolytic response in filamentous fungi, Fungal Biol. Rev, vol.30, pp.101-111, 2016.

L. B. Huberman, S. T. Coradetti, and N. L. Glass, Network of nutrient-sensing pathways and a conserved kinase cascade integrate osmolarity and carbon sensing in Neurospora crassa, Proc. Natl. Acad. Sci, vol.114, pp.8665-8674, 2017.

S. Irmler, H. Rogniaux, D. Hess, and C. Pillonel, Induction of OS-2 phosphorylation in Neurospora crassa by treatment with phenylpyrrole fungicides and osmotic stress, Pestic. Biochem. Physiol, vol.84, pp.25-37, 2006.

A. Jacobs, J. Lundqvist, H. Stålbrand, F. Tjerneld, and O. Dahlman, Characterization of water-soluble hemicelluloses from spruce and aspen employing SEC/MALDI mass spectroscopy, Carbohydr. Res, vol.337, pp.711-717, 2002.

T. Y. James, M. Lee, L. T. Diepen, and . Van, A Single Mating-Type Locus Composed of Homeodomain Genes Promotes Nuclear Migration and Heterokaryosis in the White-Rot Fungus Phanerochaete chrysosporium, Eukaryot. Cell, vol.10, pp.249-261, 2011.

A. Javelle, M. Morel, B. Rodríguez-pastrana, B. Botton, B. André et al., Molecular characterization, function and regulation of ammonium transporters (Amt) and ammonium-metabolizing enzymes (GS, NADP-GDH) in the ectomycorrhizal fungus Hebeloma cylindrosporum, Mol. Microbiol, vol.47, pp.411-430, 2003.

J. Jeon, J. Choi, G. Lee, R. A. Dean, and Y. Lee, Experimental Evolution Reveals Genome-Wide Spectrum and Dynamics of Mutations in the Rice Blast Fungus, Magnaporthe oryzae, PLOS ONE, vol.8, p.65416, 2013.

A. Joubert, B. Calmes, R. Berruyer, M. Pihet, J. Bouchara et al.,

M. Jourdes, L. Pouységu, D. Deffieux, P. Teissedre, and S. Quideau, Natural Products: Phytochemistry, Botany and Metabolism of Alkaloids, Phenolics and, pp.1975-2010, 2013.

A. K. Kameshwar and W. Qin, Metadata Analysis of Phanerochaete chrysosporium Gene Expression Data Identified Common CAZymes Encoding Gene Expression Profiles Involved in Cellulose and Hemicellulose Degradation, Int. J. Biol. Sci, vol.13, pp.85-99, 2017.

N. Kasai, S. Ikushiro, S. Hirosue, A. Arisawa, H. Ichinose et al., Enzymatic properties of cytochrome P450 catalyzing 3?-hydroxylation of naringenin from the white-rot fungus Phanerochaete chrysosporium, Biochem. Biophys. Res. Commun, vol.387, pp.103-108, 2009.

N. Kasai, S. Ikushiro, S. Hirosue, A. Arisawa, H. Ichinose et al., Atypical kinetics of cytochromes P450 catalysing 3?-hydroxylation of flavone from the white-rot fungus Phanerochaete chrysosporium, J. Biochem. (Tokyo), vol.147, pp.117-125, 2010.

F. Kawamura, S. Ohara, and A. Nishida, Antifungal activity of constituents from the heartwood of Gmelina arborea: Part 1. Sensitive antifungal assay against Basidiomycetes, 2005.

, Holzforschung, vol.58, pp.189-192

P. Kersten and D. Cullen, Extracellular oxidative systems of the lignin-degrading, 2007.

, Basidiomycete Phanerochaete chrysosporium. Fungal Genet. Biol. FG B, vol.44, pp.77-87

P. Kersten and D. Cullen, Copper radical oxidases and related extracellular oxidoreductases of wood-decay Agaricomycetes, Fungal Genet. Biol, vol.72, pp.124-130, 2014.

K. Khanbabaee, R. , and T. Van, Tannins: Classification and Definition, Nat. Prod. Rep, vol.18, pp.641-649, 2001.

T. K. Kirk, Effects of a Brown-Rot Fungus, Lenzites trabea, on Lignin in Spruce Wood, Holzforsch. -Int. J. Biol. Chem. Phys. Technol. Wood, vol.29, pp.99-107, 1975.

G. T. Kirker, A. B. Blodgett, R. A. Arango, P. K. Lebow, and C. A. Clausen, The role of extractives in naturally durable wood species, Int. Biodeterior. Biodegrad, vol.82, pp.53-58, 2013.

H. Klasson, G. R. Fink, and P. O. Ljungdahl, Ssy1p and Ptr3p Are Plasma Membrane Components of a Yeast System That Senses Extracellular Amino Acids, Mol. Cell. Biol, vol.19, pp.5405-5416, 1999.

Y. Kodama, F. Omura, K. Takahashi, K. Shirahige, A. et al., , 2002.

, Genome-wide expression analysis of genes affected by amino acid sensor Ssy1p in Saccharomyces cerevisiae

, Curr. Genet, vol.41, pp.63-72

P. Korripally, C. G. Hunt, C. J. Houtman, D. C. Jones, P. J. Kitin et al., , 2015.

, Regulation of Gene Expression during the Onset of Ligninolytic Oxidation by Phanerochaete chrysosporium on Spruce Wood, Appl. Environ. Microbiol, vol.81, pp.7802-7812

C. U. Köser, M. J. Ellington, and S. J. Peacock, Whole-genome sequencing to control antimicrobial resistance, Trends Genet, vol.30, pp.401-407, 2014.

F. Krah, C. Bässler, C. Heibl, J. Soghigian, H. Schaefer et al., Evolutionary dynamics of host specialization in wood-decay fungi, BMC Evol. Biol, vol.18, p.119, 2018.

J. Krieger-elmar, L. Keehyoung, L. Jinwoo, R. Jooyoung, T. Srivatsan et al., Improving physical realism, stereochemistry, and side-chain accuracy in homology modeling: Four approaches that performed well in CASP8, Proteins Struct. Funct. Bioinforma, vol.77, pp.114-122, 2009.

U. Kües, D. R. Nelson, C. Liu, G. Yu, J. Zhang et al., Genome analysis of medicinal Ganoderma spp. with plant-pathogenic and saprotrophic life-styles, Phytochemistry, vol.114, pp.18-37, 2015.

T. Kulik, M. Bu?ko, A. Pszczó?kowska, J. Perkowski, and A. Okorski, Plant lignans inhibit growth and trichothecene biosynthesis in Fusarium graminearum, Lett. Appl. Microbiol, vol.59, pp.99-107, 2014.

P. Kulmburg, M. Mathieu, C. Dowzer, J. Kelly, and B. Felenbok, Specific binding sites in the alcR and alcA promoters of the ethanol regulon for the CREA repressor mediating carbon cataboiite repression in Aspergillus nidulans, Mol. Microbiol, vol.7, pp.847-857, 1993.

C. P. Kurtzman and J. Pi?kur, Taxonomy and phylogenetic diversity among the yeasts, Comparative Genomics: Using Fungi as Models, pp.29-46, 2015.

I. W. Kusuma and S. Tachibana, Antifungal secoabietane dialdehyde and bisabolane-type terpenoids from the heartwood of Cryptomeria japonica D. Don, Holzforschung, vol.62, pp.624-626, 2008.

N. Kusumoto, T. Ashitani, T. Murayama, K. Ogiyama, and K. Takahashi, , 2010.

, Abietane-Type Diterpenes from the Cones of Taxodium distichum Rich, J. Chem. Ecol, vol.36, pp.1381-1386

T. Laothanachareon, J. A. Tamayo-ramos, B. Nijsse, and P. J. Schaap, Forward Genetics by, 2018.

, Genome Sequencing Uncovers the Central Role of the Aspergillus niger goxB Locus in Hydrogen Peroxide Induced Glucose Oxidase Expression, Front. Microbiol, vol.9

H. Latva-mäenpää, T. Laakso, T. Sarjala, K. Wähälä, and P. Saranpää, Root neck of Norway spruce as a source of bioactive lignans and stilbenes, Holzforschung, vol.68, pp.1-7, 2013.

H. Lee, A. K. Lamichhane, H. M. Garraffo, K. J. Kwon-chung, C. et al., Involvement of PDK1, PKC and TOR signalling pathways in basal fluconazole tolerance in Cryptococcus neoformans, Mol. Microbiol, vol.84, pp.130-146, 2012.

H. V. Lee, S. B. Hamid, and S. K. Zain, Conversion of Lignocellulosic Biomass to Nanocellulose: Structure and Chemical Process, 2014.

E. Leistner, CHAPTER 11 -Occurrence and Biosynthesis of Quinones in Woody Plants, Biosynthesis and Biodegradation of Wood Components, pp.273-290, 1985.

S. Lekounougou, S. Mounguengui, S. Dumarçay, C. Rose, P. E. Courty et al., Initial stages of Fagus sylvatica wood colonization by the white-rot basidiomycete Trametes versicolor: Enzymatic characterization, Int. Biodeterior. Biodegrad, vol.61, pp.287-293, 2008.
URL : https://hal.archives-ouvertes.fr/hal-02666158

T. P. Levine, R. D. Daniels, A. T. Gatta, L. H. Wong, and M. J. Hayes, The product of C9orf72, a gene strongly implicated in neurodegeneration, 2013.

, Bioinformatics, vol.29, pp.499-503

H. Li, A statistical framework for SNP calling, mutation discovery, association mapping and population genetical parameter estimation from sequencing data, Bioinformatics, vol.27, pp.2987-2993, 2011.

H. Li and R. Durbin, Fast and accurate short read alignment with Burrows-Wheeler transform, Bioinformatics, vol.25, pp.1754-1760, 2009.

L. Li and K. A. Borkovich, GPR-4 Is a Predicted G-Protein-Coupled Receptor Required for Carbon Source-Dependent Asexual Growth and Development in Neurospora crassa, Eukaryot. Cell, vol.5, pp.1287-1300, 2006.

B. Li, F. A. Rotsaert, M. H. Gold, and V. Renganathan, Homologous Expression of, 2000.

, Recombinant Cellobiose Dehydrogenase in Phanerochaete chrysosporium, Biochem. Biophys. Res. Commun, vol.270, pp.141-146

B. Li, M. Carey, and J. L. Workman, The Role of Chromatin during Transcription, Cell, vol.128, pp.707-719, 2007.

L. Li, S. J. Wright, S. Krystofova, G. Park, and K. A. Borkovich, Heterotrimeric G Protein Signaling in Filamentous Fungi, Annu. Rev. Microbiol, vol.61, pp.423-452, 2007.

L. Li, T. Zhu, Y. Song, X. Luo, L. Feng et al., Functional Characterization of Target of Rapamycin Signaling in Verticillium dahliae, Front. Microbiol, vol.10, 2019.

A. Linford, S. Yoshimura, R. N. Bastos, L. Langemeyer, A. Gerondopoulos et al., Rab14 and Its Exchange Factor FAM116 Link Endocytic Recycling and Adherens Junction Stability in Migrating Cells, Dev. Cell, vol.22, pp.952-966, 2012.

X. Liu, Y. Hao, L. Jin, Z. Xu, T. A. Mcallister et al., Anti-Escherichia coli O157:H7 Properties of Purple Prairie Clover and Sainfoin Condensed Tannins, Molecules, vol.18, pp.2183-2199, 2013.

R. Loewith and M. N. Hall, Target of Rapamycin (TOR) in Nutrient Signaling and Growth Control, Genetics, vol.189, pp.1177-1201, 2011.

R. Loewith, E. Jacinto, S. Wullschleger, A. Lorberg, J. L. Crespo et al., Two TOR Complexes, Only One of which Is Rapamycin Sensitive, Have Distinct Roles in Cell Growth Control. Mol. Cell, vol.10, pp.457-468, 2002.

M. S. López-berges, N. Rispail, R. C. Prados-rosales, P. , and A. D. , A Nitrogen Response Pathway Regulates Virulence Functions in Fusarium oxysporum via the Protein Kinase TOR and the bZIP Protein MeaB, Plant Cell, vol.22, pp.2459-2475, 2010.

J. Lu, M. Venäläinen, R. Julkunen-tiitto, and A. M. Harju, Stilbene impregnation retards brown-rot decay of Scots pine sapwood, Holzforschung, vol.70, pp.261-266, 2015.

T. K. Lundell, M. R. Mäkelä, R. P. De-vries, and K. S. Hildén, Chapter Eleven -Genomics, Lifestyles and Future Prospects of Wood-Decay and Litter-Decomposing Basidiomycota, Advances in Botanical Research, F.M. Martin, pp.329-370, 2014.

H. Lyr, Detoxification of Heartwood Toxins and Chlorophenols by Higher Fungi, Nature, vol.195, pp.289-290, 1962.

B. Ma, M. B. Mayfield, and M. H. Gold, The Green Fluorescent Protein Gene Functions as a Reporter of Gene Expression in Phanerochaete chrysosporium, Appl Env. Microbiol, vol.67, pp.948-955, 2001.

J. Macdonald, H. Suzuki, and E. R. Master, Expression and regulation of genes encoding lignocellulose-degrading activity in the genus Phanerochaete, Appl. Microbiol. Biotechnol, vol.94, pp.339-351, 2012.

W. D. Macrae and G. H. Towers, Biological activities of lignans, Phytochemistry, vol.23, pp.1207-1220, 1984.

M. M. Maidan, J. M. Thevelein, and P. Van-dijck, Carbon source induced yeast-to-hypha transition in Candida albicans is dependent on the presence of amino acids and on the G-proteincoupled receptor Gpr1, Biochem. Soc. Trans, vol.33, pp.291-293, 2005.

T. Manavalan, A. Manavalan, and K. Heese, Characterization of Lignocellulolytic Enzymes from White-Rot Fungi, Curr. Microbiol, vol.70, pp.485-498, 2015.

A. L. Marat, H. Dokainish, and P. S. Mcpherson, DENN Domain Proteins: Regulators of Rab GTPases, J. Biol. Chem, vol.286, pp.13791-13800, 2011.

R. Mark, X. Lyu, J. J. Lee, R. Parra-saldívar, C. et al., Sustainable production of natural phenolics for functional food applications, J. Funct. Foods, vol.57, pp.233-254, 2019.

P. E. Marriott, L. D. Gómez, and S. J. Mcqueen-mason, Unlocking the potential of lignocellulosic biomass through plant science, New Phytol, vol.209, pp.1366-1381, 2016.

M. Marroquin-guzman and R. A. Wilson, GATA-Dependent Glutaminolysis Drives Appressorium Formation in Magnaporthe oryzae by Suppressing TOR Inhibition of cAMP/PKA Signaling, PLOS Pathog, vol.11, p.1004851, 2015.

J. F. Martín, . Van-den, M. A. Berg, E. Ver-loren-van-themaat, and P. Liras, Sensing and transduction of nutritional and chemical signals in filamentous fungi: Impact on cell development and secondary metabolites biosynthesis, Biotechnol. Adv, vol.37, p.107392, 2019.

A. T. Martínez, M. Speranza, F. J. Ruiz-dueñas, P. Ferreira, S. Camarero et al.,

A. Gutiérrez and J. C. Del-río, Biodegradation of lignocellulosics: microbial, chemical, and enzymatic aspects of the fungal attack of lignin, Int. Microbiol. Off. J. Span. Soc. Microbiol, vol.8, pp.195-204, 2005.

D. Martinez, L. F. Larrondo, N. Putnam, M. D. Gelpke, K. Huang et al., Genome sequence of the lignocellulose degrading fungus Phanerochaete chrysosporium strain RP78, Nat. Biotechnol, vol.22, pp.695-700, 2004.

C. Mascarenhas, L. C. Edwards-ingram, L. Zeef, D. Shenton, M. P. Ashe et al., , 2008.

, Gcn4 Is Required for the Response to Peroxide Stress in the Yeast Saccharomyces cerevisiae

, Biol. Cell, vol.19, pp.2995-3007

Y. Mathieu, P. Prosper, M. Buée, S. Dumarçay, F. Favier et al., Characterization of a Phanerochaete chrysosporium Glutathione Transferase Reveals a Novel Structural and Functional Class with Ligandin Properties, J. Biol. Chem, vol.287, pp.39001-39011, 2012.
URL : https://hal.archives-ouvertes.fr/hal-01268261

Y. Mathieu, P. Prosper, F. Favier, L. Harvengt, C. Didierjean et al., Diversification of Fungal Specific Class A Glutathione Transferases in Saprotrophic Fungi, PLOS ONE, vol.8, p.80298, 2013.
URL : https://hal.archives-ouvertes.fr/hal-01520994

S. W. Mccotter, L. C. Horianopoulos, and J. W. Kronstad, Regulation of the fungal secretome, Curr. Genet, vol.62, pp.533-545, 2016.

H. G. Meléndez, G. Billon-grand, M. Fèvre, and G. Mey, Role of the Botrytis cinerea FKBP12 ortholog in pathogenic development and in sulfur regulation, Fungal Genet. Biol, vol.46, pp.308-320, 2009.

A. E. Mendoza-martínez, N. Cano-domínguez, and J. Aguirre, , 2019.

E. Meux, M. Morel, T. Lamant, P. Gérardin, J. Jacquot et al., , 2013.

, New substrates and activity of Phanerochaete chrysosporium Omega glutathione transferases, Biochimie, vol.95, pp.336-346

J. Mierziak, K. Kostyn, and A. Kulma, Flavonoids as Important Molecules of Plant Interactions with the Environment, Molecules, vol.19, pp.16240-16265, 2014.

M. Minami, K. Suzuki, A. Shimizu, T. Hongo, T. Sakamoto et al.,

A. Kusaka, K. Iwama, I. , and T. , Changes in the Gene Expression of the White Rot Fungus Phanerochaete chrysosporium Due to the Addition of, Atropine. Biosci. Biotechnol, 2009.

, Biochem, vol.73, pp.1722-1731

B. C. Monk, T. M. Tomasiak, M. V. Keniya, F. U. Huschmann, J. D. Tyndall et al., Proc. Natl. Acad. Sci, vol.111, pp.3865-3870, 2014.

M. Morel, A. A. Ngadin, J. Jacquot, and E. Gelhaye, Chapter 6 Reactive Oxygen Species in Phanerochaete chrysosporium Relationship Between Extracellular Oxidative and Intracellular Antioxidant Systems, Advances in Botanical Research, pp.153-186, 2009.

M. Morel, A. A. Ngadin, M. Droux, J. Jacquot, and E. Gelhaye, The fungal glutathione S-transferase system, Evidence of new classes in the wood-degrading basidiomycete, 2009.
URL : https://hal.archives-ouvertes.fr/hal-02321274

, Phanerochaete chrysosporium. Cell. Mol. Life Sci. CMLS, vol.66, pp.3711-3725

M. Morel, E. Meux, Y. Mathieu, A. Thuillier, K. Chibani et al., Xenomic networks variability and adaptation traits in wood decaying fungi, Microb. Biotechnol, vol.6, pp.248-263, 2013.
URL : https://hal.archives-ouvertes.fr/hal-01268111

L. G. Nagy, R. Riley, P. J. Bergmann, K. Krizsán, F. M. Martin et al., Genetic Bases of Fungal White Rot Wood Decay Predicted by Phylogenomic Analysis of Correlated Gene-Phenotype Evolution, Mol. Biol. Evol, vol.34, pp.35-44, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01551070

K. Nakatsukasa, A. Kanada, M. Matsuzaki, S. D. Byrne, F. Okumura et al., The Nutrient Stress-induced Small GTPase Rab5 Contributes to the Activation of Vesicle Trafficking and Vacuolar Activity, J. Biol. Chem, vol.289, pp.20970-20978, 2014.

T. Nakazawa, M. Tsuzuki, T. Irie, M. Sakamoto, and Y. Honda, Marker recycling via 5-fluoroorotic acid and 5-fluorocytosine counter-selection in the white-rot agaricomycete Pleurotus ostreatus, Fungal Biol, vol.120, pp.1146-1155, 2016.

T. Nakazawa, A. Izuno, R. Kodera, Y. Miyazaki, M. Sakamoto et al., the white-rot agaricomycete Pleurotus ostreatus, Environ. Microbiol, vol.19, pp.261-272, 2017.

A. Nerg, J. Heijari, U. Noldt, H. Viitanen, M. Vuorinen et al., Significance of Wood Terpenoids in the Resistance of Scots Pine Provenances Against the Old House Borer, Hylotrupes bajulus, and Brown-Rot Fungus, Coniophora puteana, J. Chem. Ecol, vol.30, pp.125-141, 2004.

F. B. Niamké, N. Amusant, J. Charpentier, G. Chaix, Y. Baissac et al., Relationships between biochemical attributes (nonstructural carbohydrates and phenolics) and natural durability against fungi in dry teak wood (Tectona grandis L. f.), Ann. For. Sci, vol.68, pp.201-211, 2011.

F. B. Niamké, N. Amusant, D. Stien, G. Chaix, Y. Lozano et al., 4?,5?-Dihydroxy-epiisocatalponol, a new naphthoquinone from Tectona grandis L. f. heartwood, and fungicidal activity, Int. Biodeterior. Biodegrad, vol.74, pp.93-98, 2012.

B. J. Niles, A. C. Joslin, T. Fresques, and T. Powers, TOR Complex 2-Ypk1 signaling maintains sphingolipid homeostasis by sensing and regulating ROS accumulation, Cell Rep, vol.6, pp.541-552, 2014.

R. Noguchi, S. Banno, R. Ichikawa, F. Fukumori, A. Ichiishi et al., Identification of OS-2 MAP kinase-dependent genes induced in response to osmotic stress, antifungal agent fludioxonil, and heat shock in Neurospora crassa, 2007.

, Biol, vol.44, pp.208-218

K. J. Nordström, M. C. Albani, G. V. James, C. Gutjahr, B. Hartwig et al.,

G. Coupland and K. Schneeberger, Mutation identification by direct comparison of wholegenome sequencing data from mutant and wild-type individuals using k -mers, Nat. Biotechnol, vol.31, pp.325-330, 2013.

H. E. O'brien, J. L. Parrent, J. A. Jackson, J. Moncalvo, and R. Vilgalys, Fungal Community Analysis by Large-Scale Sequencing of Environmental Samples, Appl. Environ. Microbiol, vol.71, pp.5544-5550, 2005.

F. C. Odds, A. J. Brown, and N. A. Gow, Antifungal agents: mechanisms of action, Trends Microbiol, vol.11, pp.272-279, 2003.

S. Ogawa, Y. Matsuo, T. Tanaka, Y. Yazaki, S. Ogawa et al., Utilization of Flavonoid Compounds from Bark and Wood. III. Application in Health Foods, Molecules, vol.23, p.1860, 2018.

R. A. Ohm, R. Riley, A. Salamov, B. Min, I. Choi et al., Genomics of wooddegrading fungi, Fungal Genet. Biol, vol.72, pp.82-90, 2014.

W. Ohmura, S. Doi, M. Aoyama, and S. Ohara, Antifeedant activity of flavonoids and related compounds against the subterranean termite Coptotermes formosanus Shiraki, J. Wood Sci, vol.46, pp.149-153, 2000.

Y. Otsubo and M. Yamamoto, 12 -TOR and Sexual Development in Fission Yeast, The Enzymes, pp.229-250, 2010.

J. Pan, S. Chen, M. Yang, J. Wu, J. Sinkkonen et al., An update on lignans: natural products and synthesis, Nat. Prod. Rep, vol.26, pp.1251-1292, 2009.

M. Paoletti and C. Clavé, The Fungus-Specific HET Domain Mediates Programmed Cell Death in Podospora anserina, Eukaryot. Cell, vol.6, 2001.
URL : https://hal.archives-ouvertes.fr/hal-00173407

G. Park, J. A. Servin, G. E. Turner, L. Altamirano, H. V. Colot et al., Global Analysis of Serine-Threonine Protein Kinase Genes in Neurospora crassa, Eukaryot. Cell, vol.10, pp.1553-1564, 2011.

S. Paul, T. L. Doering, and W. S. Moye-rowley, Cryptococcus neoformans Yap1 is required for normal fluconazole and oxidative stress resistance, Fungal Genet. Biol, vol.74, pp.1-9, 2015.

H. Pereira, J. Graça, and J. C. Rodrigues, Wood chemistry in relation to quality, Wood Quality and Its Biological Basis, pp.53-56, 2009.

T. Perrot, M. Schwartz, F. Saiag, G. Salzet, S. Dumarçay et al., Fungal Glutathione Transferases as Tools to Explore the Chemical Diversity of Amazonian Wood Extractives, ACS Sustain. Chem. Eng, vol.6, pp.13078-13085, 2018.
URL : https://hal.archives-ouvertes.fr/hal-02492224

T. Pfirrmann, S. Heessen, D. J. Omnus, C. Andréasson, and P. O. Ljungdahl, The Prodomain of Ssy5 Protease Controls Receptor-Activated Proteolysis of Transcription Factor Stp1, Mol. Cell, 2010.

, Biol, vol.30, pp.3299-3309

S. P. Pietarinen, S. M. Willför, F. A. Vikström, and B. R. Holmbom, Aspen Knots, a Rich Source of Flavonoids, J. Wood Chem. Technol, vol.26, pp.245-258, 2006.

B. Pinan-lucarré, I. Iraqui, and C. Clavé, Podospora anserina target of rapamycin. Curr, 2006.

. Genet, , vol.50, pp.23-31

J. A. Polish, J. Kim, and M. Johnston, How the Rgt1 Transcription Factor of Saccharomyces cerevisiae Is Regulated by Glucose, Genetics, vol.169, pp.583-594, 2005.

B. Qian, X. Liu, J. Jia, Y. Cai, C. Chen et al., MoPpe1 partners with MoSap1 to mediate TOR and cell wall integrity signalling in growth and pathogenicity of the rice blast fungus Magnaporthe oryzae, Environ. Microbiol, vol.20, pp.3964-3979, 2018.

J. Ralph, C. Lapierre, and W. Boerjan, Lignin structure and its engineering, Curr. Opin. Biotechnol, vol.56, pp.240-249, 2019.
URL : https://hal.archives-ouvertes.fr/hal-02626144

R. Ramanujam, M. E. Calvert, P. Selvaraj, and N. I. Naqvi, The Late Endosomal HOPS Complex Anchors Active G-Protein Signaling Essential for Pathogenesis in Magnaporthe oryzae, PLOS Pathog, vol.9, p.1003527, 2013.

L. Ratnayake, K. K. Adhvaryu, E. Kafes, K. Motavaze, and P. Lakin-thomas, A component of the TOR (Target Of Rapamycin) nutrient-sensing pathway plays a role in circadian rhythmicity in Neurospora crassa, PLOS Genet, vol.14, p.1007457, 2018.

P. Ren, A. Rossettini, V. Chaturvedi, and S. D. Hanes, The Ess1 prolyl isomerase is dispensable for growth but required for virulence in Cryptococcus neoformans, Microbiology, vol.151, pp.1593-1605, 2005.

L. N. Ries, S. R. Beattie, E. A. Espeso, R. A. Cramer, and G. H. Goldman, Diverse Regulation of the CreA Carbon Catabolite Repressor in Aspergillus nidulans, Genetics, vol.203, pp.335-352, 2016.

R. Riley, A. A. Salamov, D. W. Brown, L. G. Nagy, D. Floudas et al., Extensive sampling of basidiomycete genomes demonstrates inadequacy of the white-rot/brown-rot paradigm for wood decay fungi, Proc. Natl. Acad. Sci, vol.111, pp.9923-9928, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01578657

A. R. Rodríguez, G. Koch, H. Richter, T. F. Fuentes, G. J. Silva et al., , 2019.

, Formation of heartwood, chemical composition of extractives and natural durability of plantation-grown teak wood from Mexico, Holzforschung, vol.73, pp.547-557

J. Rodríguez-romero, M. Hedtke, C. Kastner, S. Mueller, and R. Fischer, Fungi, hidden in soil or up in the air: light makes a difference, Annu. Rev. Microbiol, vol.64, pp.585-610, 2010.

F. M. Roelants, K. L. Leskoske, M. N. Martinez-marshall, M. N. Locke, and J. Thorner, The TORC2-Dependent Signaling Network in the Yeast Saccharomyces cerevisiae, Biomolecules, vol.7, p.66, 2017.

M. Rohlfs, M. Albert, N. P. Keller, and F. Kempken, Secondary chemicals protect mould from fungivory, Biol. Lett, vol.3, pp.523-525, 2007.

M. Royer, A. M. Rodrigues, G. Herbette, J. Beauchêne, M. Chevalier et al., Efficacy of Bagassa guianensis Aubl. extract against wood decay and human pathogenic fungi, Int. Biodeterior. Biodegrad, vol.70, pp.55-59, 2012.
URL : https://hal.archives-ouvertes.fr/hal-00694532

J. C. Rutherford, Y. Bahn, . Van-den, B. Berg, J. Heitman et al., Nutrient and Stress Sensing in Pathogenic Yeasts, Front. Microbiol, vol.10, 2019.

H. Saijo, H. Kofujita, K. Takahashi, A. , and T. , Antioxidant activity and mechanism of the abietane-type diterpene ferruginol, Nat. Prod. Res, vol.29, pp.1739-1743, 2015.

N. E. Sanchez, B. L. Harty, T. O'reilly-pol, S. D. Ackerman, A. L. Herbert et al., Whole Genome Sequencing-Based Mapping and Candidate Identification of Mutations from Fixed Zebrafish Tissue, G3 Genes Genomes Genet, vol.7, pp.3415-3425, 2017.

H. Sang, J. P. Hulvey, R. Green, H. Xu, J. Im et al., A Xenobiotic Detoxification Pathway through Transcriptional Regulation in Filamentous Fungi, vol.9, pp.457-475, 2018.

S. Saupe, B. Turcq, and J. Bégueret, A gene responsible for vegetative incompatibility in the fungus Podospora anserina encodes a protein with a GTP-binding motif and G? homologous domain, Gene, vol.162, pp.135-139, 1995.

A. Scalbert, Antimicrobial properties of tannins, Phytochemistry, vol.30, pp.3875-3883, 1991.
URL : https://hal.archives-ouvertes.fr/hal-02700780

A. Scalbert, Tannins in Woods and Their Contribution to Microbial Decay Prevention, Plant Polyphenols: Synthesis, Properties, pp.935-952, 1992.
URL : https://hal.archives-ouvertes.fr/hal-02843557

C. Schlötterer, R. Tobler, R. Kofler, and V. Nolte, Sequencing pools of individualsmining genome-wide polymorphism data without big funding, Nat. Rev. Genet, vol.15, pp.749-763, 2014.

K. Schneeberger, Using next-generation sequencing to isolate mutant genes from forward genetic screens, Nat. Rev. Genet, vol.15, pp.662-676, 2014.

K. Schneeberger and D. Weigel, Fast-forward genetics enabled by new sequencing technologies, Trends Plant Sci, vol.16, pp.282-288, 2011.

T. P. Schultz and D. D. Nicholas, Naturally durable heartwood: evidence for a proposed dual defensive function of the extractives, Phytochemistry, vol.54, pp.47-52, 2000.

T. P. Schultz and D. D. Nicholas, Development of environmentally-benign wood preservatives based on the combination of organic biocides with antioxidants and metal chelators, Phytochemistry, vol.61, pp.555-560, 2002.

T. P. Schultz, T. F. Hubbard, L. Jin, T. H. Fisher, and D. D. Nicholas, Role of stilbenes in the natural durability of wood: Fungicidal structure-activity relationships, Phytochemistry, vol.29, pp.1501-1507, 1990.

S. N. Sehgal, H. Baker, and C. Vézina, Rapamycin (AY-22,989), a new antifungal antibiotic, 1975.

, II. Fermentation, isolation and characterization, J. Antibiot. (Tokyo), vol.28, pp.727-732

N. Sekine, T. Ashitani, T. Murayama, S. Shibutani, S. Hattori et al., , 2009.

, Bioactivity of Latifolin and Its Derivatives against Termites and Fungi, J. Agric. Food Chem, vol.57, pp.5707-5712

K. K. Sharma and R. C. Kuhad, Genetic transformation of lignin degrading fungi facilitated by Agrobacterium tumefaciens, BMC Biotechnol, vol.10, p.67, 2010.

S. Shashkova, N. Welkenhuysen, and S. Hohmann, Molecular communication: crosstalk between the Snf1 and other signaling pathways, FEMS Yeast Res, vol.15, 2015.

G. Shen and A. Kong, Nrf2 plays an important role in coordinated regulation of Phase II drug metabolism enzymes and Phase III drug transporters, Biopharm. Drug Dispos, vol.30, pp.345-355, 2009.

T. Shen, X. Wang, and H. Lou, Natural stilbenes: an overview, Nat. Prod. Rep, vol.26, pp.916-935, 2009.

C. A. Shertz, R. J. Bastidas, W. Li, J. Heitman, and M. E. Cardenas, Conservation, duplication, and loss of the Tor signaling pathway in the fungal kingdom, BMC Genomics, vol.11, p.510, 2010.

C. Shin, S. Y. Kim, and W. Huh, TORC1 controls degradation of the transcription factor Stp1, a key effector of the SPS amino-acid-sensing pathway in Saccharomyces cerevisiae, J. Cell Sci, vol.122, pp.2089-2099, 2009.

C. Shnaiderman, I. Miyara, I. Kobiler, A. Sherman, and D. Prusky, Differential Activation of Ammonium Transporters During the Accumulation of Ammonia by Colletotrichum gloeosporioides and Its Effect on Appressoria Formation and Pathogenicity, Mol. Plant. Microbe Interact, vol.26, pp.345-355, 2013.

H. Simaan, S. Lev, and B. A. Horwitz, Oxidant-Sensing Pathways in the Responses of Fungal Pathogens to, Chemical Stress Signals. Front. Microbiol, vol.10, 2019.

B. Singh and R. A. Sharma, Plant terpenes: defense responses, phylogenetic analysis, regulation and clinical applications, 3 Biotech, vol.5, pp.129-151, 2015.

T. Singh and A. P. Singh, A review on natural products as wood protectant, 2012.

. Technol, , vol.46, pp.851-870

E. Sionov, Y. C. Chang, H. M. Garraffo, M. A. Dolan, M. A. Ghannoum et al., , 2012.

, Identification of a Cryptococcus neoformans Cytochrome P450 Lanosterol 14?-Demethylase

, Residue Critical for Differential Susceptibility between Fluconazole/Voriconazole and Itraconazole/Posaconazole, vol.56, pp.1162-1169

D. A. Smith, B. A. Morgan, Q. , and J. , Stress signalling to fungal stress-activated protein kinase pathways, FEMS Microbiol. Lett, vol.306, pp.1-8, 2010.

C. Snyman, L. Theron, and B. Divol, Understanding the regulation of extracellular protease gene expression in fungi: a key step towards their biotechnological applications, Appl. Microbiol. Biotechnol, vol.103, pp.5517-5532, 2019.

W. R. Souza, . De, E. R. Morais, N. G. Krohn, M. Savoldi et al., Identification of Metabolic Pathways Influenced by the G-Protein Coupled Receptors GprB and GprD in Aspergillus nidulans, PLOS ONE, vol.8, 2013.

C. M. Spencer, Y. Cai, R. Martin, S. H. Gaffney, P. N. Goulding et al., Polyphenol complexation-some thoughts and observations, Phytochemistry, vol.27, pp.2397-2409, 1988.

G. Steinberg, Hyphal Growth: a Tale of Motors, Lipids, and the Spitzenkörper, Eukaryot. Cell, vol.6, pp.351-360, 2007.

P. Stewart, J. Gaskell, and D. Cullen, A Homokaryotic Derivative of a Phanerochaete chrysosporium Strain and Its Use in Genomic Analysis of Repetitive Elements, Appl Env. Microbiol, vol.66, pp.1629-1633, 2000.

D. Stracka, S. Jozefczuk, F. Rudroff, U. Sauer, and M. N. Hall, Nitrogen Source Activates TOR (Target of Rapamycin) Complex 1 via Glutamine and Independently of Gtr/Rag Proteins, J. Biol. Chem, vol.289, pp.25010-25020, 2014.

H. Sun and K. Schneeberger, SHOREmap v3.0: Fast and Accurate Identification of Causal Mutations from Forward Genetic Screens, Plant Functional Genomics: Methods and Protocols, pp.381-395, 2015.

J. Sun and N. L. Glass, Identification of the CRE-1 Cellulolytic Regulon in Neurospora crassa, PLOS ONE, vol.6, 2011.

K. Syed and J. S. Yadav, P450 monooxygenases (P450ome) of the model white rot fungus Phanerochaete chrysosporium, Crit. Rev. Microbiol, vol.38, pp.339-363, 2012.

K. Syed, K. Shale, N. S. Pagadala, and J. Tuszynski, Systematic Identification and Evolutionary Analysis of Catalytically Versatile Cytochrome P450 Monooxygenase Families Enriched in Model Basidiomycete Fungi, PLOS ONE, vol.9, p.86683, 2014.

Z. Tan, M. Huang, A. Puga, and Y. Xia, A Critical Role For MAP Kinases in the Control of Ah Receptor Complex Activity, Toxicol. Sci, vol.82, pp.80-87, 2004.

A. H. Tayeb, E. Amini, S. Ghasemi, and M. Tajvidi, Cellulose Nanomaterials-Binding Properties and Applications: A Review, Molecules, vol.23, p.2684, 2018.

A. M. Taylor, B. L. Gartner, and J. J. Morrell, Heartwood Formation and Natural Durability-A Review, Wood Fiber Sci, vol.34, pp.587-611, 2007.

S. Teichert, M. Wottawa, B. Schönig, and B. Tudzynski, Role of the Fusarium fujikuroi TOR Kinase in Nitrogen Regulation and Secondary Metabolism, Eukaryot. Cell, vol.5, pp.1807-1819, 2006.

S. Teichert, J. C. Rutherford, M. Wottawa, J. Heitman, and B. Tudzynski, Impact of Ammonium Permeases MepA, MepB, and MepC on Nitrogen-Regulated Secondary Metabolism in Fusarium fujikuroi, Eukaryot. Cell, vol.7, pp.187-201, 2008.

J. M. Thevelein, R. Geladé, I. Holsbeeks, O. Lagatie, Y. Popova et al., Nutrient sensing systems for rapid activation of the protein kinase A pathway in yeast, Biochem. Soc. Trans, vol.33, pp.253-256, 2005.

D. Tholl, Biosynthesis and Biological Functions of Terpenoids in Plants, pp.63-106, 2015.

A. Thuillier, K. Chibani, G. Belli, E. Herrero, S. Dumarçay et al., Transcriptomic Responses of Phanerochaete chrysosporium to Oak Acetonic Extracts: Focus on a, New Glutathione Transferase. Appl Env. Microbiol, vol.80, pp.6316-6327, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01268964

M. Tien and T. K. Kirk, Lignin-Degrading Enzyme from the Hymenomycete Phanerochaete chrysosporium, Burds. Science, vol.221, pp.661-663, 1983.

M. Tien and T. K. Kirk, Lignin-degrading enzyme from Phanerochaete chrysosporium: Purification, characterization, and catalytic properties of a unique H2O2-requiring oxygenase, 1984.

, Proc. Natl. Acad. Sci, vol.81, pp.2280-2284

M. Tien and S. B. Myer, Selection and characterization of mutants of Phanerochaete chrysosporium exhibiting ligninolytic activity under nutrient-rich conditions, Appl. Environ, 1990.

. Microbiol, , vol.56, pp.2540-2544

M. A. Treitel and M. Carlson, Repression by SSN6-TUP1 is directed by MIG1, a repressor/activator protein, Proc. Natl. Acad. Sci, vol.92, pp.3132-3136, 1995.

J. Uehling, A. Deveau, and M. Paoletti, Do fungi have an innate immune response? An NLRbased comparison to plant and animal immune systems, PLOS Pathog, vol.13, 2017.
URL : https://hal.archives-ouvertes.fr/hal-02628840

T. Uemura, K. Kashiwagi, and K. Igarashi, Uptake of putrescine and spermidine by Gap1p on the plasma membrane in Saccharomyces cerevisiae, Biochem. Biophys. Res. Commun, vol.328, pp.1028-1033, 2005.

Y. Ukai, M. Kuroiwa, N. Kurihara, H. Naruse, T. Homma et al., , 2018.

, Contributions of yap1 Mutation and Subsequent atrF Upregulation to Voriconazole Resistance in Aspergillus flavus, Antimicrob. Agents Chemother, vol.62

J. Urban, A. Soulard, A. Huber, S. Lippman, D. Mukhopadhyay et al., Sch9 is a major target of TORC1 in Saccharomyces cerevisiae, Mol. Cell, vol.26, pp.663-674, 2007.

N. Valette, T. Perrot, R. Sormani, E. Gelhaye, and M. Morel-rouhier, Antifungal activities of wood extractives, Fungal Biol. Rev, vol.31, pp.113-123, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01595725

M. A. Van-der-nest, Å. Olson, M. Lind, H. Vélëz, K. Dalman et al., Distribution and evolution of het gene homologs in the basidiomycota, 2014.

, Biol, vol.64, pp.45-57

R. Vanholme, B. Demedts, K. Morreel, J. Ralph, and W. Boerjan, Lignin Biosynthesis and Structure, Plant Physiol, vol.153, pp.895-905, 2010.

R. Vasiliauskas, A. Menkis, R. D. Finlay, and J. Stenlid, Wood-decay fungi in fine living roots of conifer seedlings, New Phytol, vol.174, pp.441-446, 2007.

M. Venäläinen, A. M. Harju, P. Saranpää, P. Kainulainen, M. Tiitta et al., The concentration of phenolics in brown-rot decay resistant and susceptible Scots pine heartwood, Wood Sci. Technol, vol.38, pp.109-118, 2004.

S. Wang, J. Wu, L. Shyur, Y. Kuo, C. et al., Antioxidant Activity of, 2005.
URL : https://hal.archives-ouvertes.fr/hal-01003648

. Abietane-type, Diterpenes from Heartwood of Taiwania cryptomerioides Hayata, Holzforschung, vol.56, pp.487-492

H. Wariishi, K. Valli, and M. H. Gold, In vitro depolymerization of lignin by manganese peroxidase of Phanerochaete chrysosporium, Biochem. Biophys. Res. Commun, vol.176, pp.269-275, 1991.

A. Warrilow, C. Ugochukwu, D. Lamb, D. Kelly, K. et al., Expression and Characterization of CYP51, the Ancient Sterol 14-demethylase Activity for Cytochromes P450 (CYP), in the White-Rot Fungus Phanerochaete chrysosporium, Lipids, vol.43, p.1143, 2008.

R. Weisman and M. Choder, The fission yeast TOR homolog, tor1+, is required for the response to starvation and other stresses via a conserved serine, J. Biol. Chem, vol.276, pp.7027-7032, 2001.

H. A. Wiatrowski and M. Carlson, Yap1 Accumulates in the Nucleus in Response to Carbon Stress in Saccharomyces cerevisiae, Eukaryot. Cell, vol.2, pp.19-26, 2003.

S. Willför, J. Hemming, M. Reunanen, C. Eckerman, and B. Holmbom, Lignans and Lipophilic Extractives in Norway Spruce Knots and Stemwood, Holzforschung, vol.57, pp.27-36, 2005.

B. Winkel-shirley, Flavonoid Biosynthesis. A Colorful Model for Genetics, Cell Biology, and Biotechnology. Plant Physiol, vol.126, pp.485-493, 2001.

D. W. Wong, Structure and Action Mechanism of Ligninolytic Enzymes, Appl. Biochem. Biotechnol, vol.157, pp.174-209, 2009.

S. Wullschleger, R. Loewith, and M. N. Hall, TOR Signaling in Growth and Metabolism. Cell, vol.124, pp.471-484, 2006.

Q. Xiao, F. Ma, Y. Li, H. Yu, C. Li et al., Differential Proteomic Profiles of Pleurotus ostreatus in Response to Lignocellulosic Components Provide Insights into Divergent Adaptive Mechanisms, Front. Microbiol, vol.8, 2017.

Y. Xiong, S. T. Coradetti, X. Li, M. A. Gritsenko, T. Clauss et al.,

F. Yang, The proteome and phosphoproteome of Neurospora crassa in response to cellulose, sucrose and carbon starvation, Fungal Genet. Biol, vol.72, pp.21-33, 2014.

C. Xue, Y. Bahn, G. M. Cox, and J. Heitman, G Protein-coupled Receptor Gpr4 Senses Amino Acids and Activates the cAMP-PKA Pathway in Cryptococcus neoformans, Mol. Biol. Cell, vol.17, pp.667-679, 2005.

Y. Yazaki, Utilization of flavonoid compounds from bark and wood: a review, Nat. Prod. Commun, vol.10, pp.513-520, 2015.

D. J. Yelle, J. Ralph, F. Lu, and K. E. Hammel, Evidence for cleavage of lignin by a brown rot basidiomycete, Environ. Microbiol, vol.10, pp.1844-1849, 2008.

Y. Ra and M. Akhtar, Taxonomy of Industrially Important White-Rot Fungi, Environmentally friendly technologies for the pulp and paper industry, pp.259-272, 1998.

F. Yu, Q. Gu, Y. Yun, Y. Yin, J. Xu et al., The TOR signaling pathway regulates vegetative development and virulence in Fusarium graminearum, New Phytol, vol.203, pp.219-232, 2014.

C. Yun, H. Tamaki, R. Nakayama, K. Yamamoto, and H. Kumagai, Gpr1p, a Putative G-Protein Coupled Receptor, Regulates Glucose-Dependent Cellular cAMP Level in Yeast Saccharomyces cerevisiae, Biochem. Biophys. Res. Commun, vol.252, pp.29-33, 1998.

S. Zaman, S. I. Lippman, X. Zhao, and J. R. Broach, How Saccharomyces Responds to Nutrients, Annu. Rev. Genet, vol.42, pp.27-81, 2008.

S. Zeilinger, M. Schmoll, M. Pail, R. L. Mach, and C. P. Kubicek, Nucleosome transactions on the Hypocrea jecorina (Trichoderma reesei) cellulase promoter cbh2 associated with cellulase induction, Mol. Genet. Genomics MGG, vol.270, pp.46-55, 2003.

J. Zhang, S. Vaga, P. Chumnanpuen, R. Kumar, G. N. Vemuri et al., , 2011.

, Mapping the interaction of Snf1 with TORC1 in Saccharomyces cerevisiae, Mol. Syst. Biol, vol.7, p.545

L. Zhang, M. Huang, and E. Harsay, A Chemical Genetic Screen for Modulators of Exocytic Transport Identifies Inhibitors of a Transport Mechanism Linked to GTR2 Function, Eukaryot. Cell, vol.9, pp.116-126, 2010.

W. Zhang, Y. Kou, J. Xu, Y. Cao, G. Zhao et al., , 2013.

, Two Major Facilitator Superfamily Sugar Transporters from Trichoderma reesei and Their Roles in Induction of Cellulase Biosynthesis, J. Biol. Chem, vol.288, pp.32861-32872

X. Zheng, D. Fiorentino, J. Chen, G. R. Crabtree, and S. L. Schreiber, TOR kinase domains are required for two distinct functions, only one of which is inhibited by rapamycin, Cell, vol.82, pp.121-130, 1995.

E. A. Znameroski, X. Li, J. C. Tsai, J. M. Galazka, N. L. Glass et al., Evidence for Transceptor Function of Cellodextrin Transporters in Neurospora crassa, J. Biol. Chem, vol.289, pp.2610-2619, 2014.

S. A. Zurita-martinez and M. E. Cardenas, Tor and Cyclic AMP-Protein Kinase A: Two Parallel Pathways Regulating Expression of Genes Required for Cell Growth, Eukaryot. Cell, vol.4, pp.63-71, 2005.

S. Zuryn, S. L. Gras, K. Jamet, J. , and S. , A Strategy for Direct Mapping and Identification of Mutations by Whole-Genome Sequencing, Genetics, vol.186, pp.427-430, 2010.