, After the incubation period, cells (1 × 10 7 cells) were sedimented, washed twice with PBS and suspended in 1 ml of PBS. Aliquots of the suspension (2 × 10 5 cells) were applied on microscopic slides and air-dried. After fixing the cells for 2 min in ice-cold methanol, the slides were air-dried for 20 min. Non-adherent cells were removed by gentle washing (0.1% Triton X-100 in PBS) followed by, Fluorescence microscopy. Promastigote cells were incubated for 72 h without RAD or with the IC 90 of RAD

, :25). After washing the slides thrice, Mowiol and coverslips were applied and the slides were left to dry for 24 h at 4 °C. Fluorescence microscopy was carried out on an EVOS FL Auto epifluorescence microscope, Triton-X 100 in PBS). Slides were then incubated for 1 h with monoclonal mouse anti-tubulin

;. Phosphoproteomics-;-pen/strep, . Gibco, and . Uk), Cultures were incubated at 34 °C/5% CO 2 for 72 h. Parasites were harvested by centrifugation at 2000 × g, 4 °C, and washed in 1) ice-cold wash buffer (21 mM HEPES pH 7.5, 137 mM NaCl, 5 mM KCl) and 2) ice-cold wash buffer with protease and phosphatase inhibitors (1 mM Na-orthovanadate, 0.1 µM okadaic acid, 10 mM NaF, 10 mM o-phenanthroline, EDTA-free protease inhibitors). For lysis, parasites were resuspended in a solution of 7 M urea, 2 M thiourea, 40 mM Tris, 1% n-octyl-?-D-glycopyranoside, 1 mM MgCl 2 , 1 mM o-phenanthroline, 300 U benzonase, 1 mM Na-pervanadate (Na-orthovanadate activated in 18% H 2 O 2 ), protease inhibitors (Roche EDTA-free protease inhibitor tablets), and phosphatase inhibitor cocktails (P2850 and P5726 from Sigma), and sonicated for 3 × 15 s on ice. Lysates were incubated at ?80 °C for 30 min prior to reduction (50 mM DTT) and alkylation with 50 mM iodoacetamide. Proteins were precipitated in an 8-fold excess of ice-cold acetone-ethanol, 1:1, v/v, by overnight incubation at ?20 °C. Protein precipitates were reconstituted in 6 M urea/2 M thiourea and diluted in 50 mM NH 4 HCO 3 for digestion with trypsin at a 1:75 enzyme-substrate ratio overnight. Digestion was quenched by addition of formic acid (FA) to a final concentration of 1%. Aliquots of 330 µg digested proteins were used for phosphopeptide enrichment by TiO 2 (Titansphere, 5 µm, Leishmania mexicana MNYC/BZ/62/M379 promastigotes were cultured in SDM-79 medium 93 supplemented with 10% heat-inactivated FCS

J. Alvar, Leishmaniasis worldwide and global estimates of its incidence, PloS one, vol.7, p.35671, 2012.

E. Barak, Differentiation of Leishmania donovani in host-free system: analysis of signal perception and response, Mol Biochem Parasitol, vol.141, pp.99-108, 2005.

S. Brandau, A. Dresel, and J. Clos, High constitutive levels of heat-shock proteins in human-pathogenic parasites of the genus Leishmania, Biochem J, vol.310, pp.225-232, 1995.

A. Hübel, S. Brandau, A. Dresel, and J. Clos, A member of the ClpB family of stress proteins is expressed during heat shock in Leishmania spp, Mol Biochem Parasitol, vol.70, pp.107-118, 1995.

S. Krobitsch, Leishmania donovani heat shock protein 100: characterization and function in amastigote stage differentiation, J. Biol. Chem, vol.273, pp.6488-6494, 1998.

,

A. Schlüter, Expression and Subcellular Localization of Cpn60 Protein Family Members in Leishmania donovani, Biochim. Biophys. Acta, vol.1491, pp.65-74, 2000.

F. B. Zamora-veyl, M. Kroemer, D. Zander, and J. Clos, Stage-specific expression of the mitochondrial co-chaperonin of Leishmania donovani, CPN10. Kinetoplastid Biol Dis, vol.4, p.3, 2005.

A. Hombach, G. Ommen, A. Macdonald, and J. Clos, A small heat shock protein is essential for thermotolerance and intracellular survival of Leishmania donovani, J Cell Sci, 2014.

J. M. Silverman, Leishmania exosomes modulate innate and adaptive immune responses through effects on monocytes and dendritic cells, J Immunol, vol.185, pp.5011-5022, 2010.

J. M. Silverman, An exosome-based secretion pathway is responsible for protein export from Leishmania and communication with macrophages, J Cell Sci, vol.123, pp.842-852, 2010.

S. L. Rutherford and C. S. Zuker, Protein folding and the regulation of signaling pathways, Cell, vol.79, pp.1129-1132, 1994.

T. Scheibel and J. Buchner, The Hsp90 complex-a super-chaperone machine as a novel drug target, Biochem Pharmacol, vol.56, pp.675-682, 1998.

S. L. Rutherford and S. Lindquist, Hsp90 as a capacitor for morphological evolution, Nature, vol.396, pp.336-342, 1998.

S. Lindquist and E. A. Craig, The heat-shock proteins, Annual review of genetics, vol.22, pp.631-677, 1988.

L. Whitesell, E. G. Mimnaugh, B. De-costa, C. E. Myers, and L. M. Neckers, Inhibition of heat shock protein HSP90-pp60v-src heteroprotein complex formation by benzoquinone ansamycins: essential role for stress proteins in oncogenic transformation, Proc Natl Acad Sci, vol.91, pp.8324-8328, 1994.

L. Whitesell and P. Cook, Stable and specific binding of heat shock protein 90 by geldanamycin disrupts glucocorticoid receptor function in intact cells, Mol Endocrinol, vol.10, pp.705-712, 1996.

A. Ali, S. Bharadwaj, R. O'carroll, and N. Ovsenek, HSP90 interacts with and regulates the activity of heat shock factor 1 in Xenopus oocytes, Mol Cell Biol, vol.18, pp.4949-4960, 1998.

H. R. Kim, C. H. Lee, Y. H. Choi, H. S. Kang, and H. D. Kim, Geldanamycin induces cell cycle arrest in K562 erythroleukemic cells, IUBMB Life, vol.48, pp.425-428, 1999.

M. Srethapakdi, F. Liu, R. Tavorath, and N. Rosen, Inhibition of Hsp90 function by ansamycins causes retinoblastoma gene productdependent G1 arrest, Cancer Res, vol.60, pp.3940-3946, 2000.

M. Wiesgigl and J. Clos, Heat Shock Protein 90 Homeostasis Controls Stage Differentiation inLeishmania donovani, Mol Biol Cell, vol.12, pp.3307-3316, 2001.

S. E. Graefe, M. Wiesgigl, I. Gaworski, A. Macdonald, and J. Clos, Inhibition of HSP90 in Trypanosoma cruzi Induces a Stress Response but No Stage Differentiation, Eukaryot Cell, vol.1, pp.936-943, 2002.

G. Banumathy, V. Singh, S. R. Pavithra, and U. Tatu, Heat shock protein 90 function is essential for Plasmodium falciparum growth in human erythrocytes, The Journal of biological chemistry, vol.278, pp.18336-18345, 2003.

T. W. Schulte, Antibiotic radicicol binds to the N-terminal domain of Hsp90 and shares important biologic activities with geldanamycin, Cell Stress Chaperones, vol.3, pp.100-108, 1998.

T. W. Schulte, Interaction of radicicol with members of the heat shock protein 90 family of molecular chaperones, Mol Endocrinol, vol.13, pp.1435-1448, 1999.

L. Whitesell and S. L. Lindquist, HSP90 and the chaperoning of cancer, Nature reviews, vol.5, pp.761-772, 2005.

M. Bente, Developmentally induced changes of the proteome in the protozoan parasite Leishmania donovani, Proteomics, vol.3, pp.1811-1829, 2003.

A. Hombach, G. Ommen, M. Chrobak, and J. Clos, The Hsp90-Sti1 Interaction is Critical forLeishmania donovaniProliferation in Both Life Cycle Stages, Cell Microbiol, vol.15, pp.585-600, 2013.

C. Prodromou, Regulation of Hsp90 ATPase activity by tetratricopeptide repeat (TPR)-domain co-chaperones, The EMBO journal, vol.18, pp.754-762, 1999.

J. L. Johnson and C. Brown, Plasticity of the Hsp90 chaperone machine in divergent eukaryotic organisms, Cell Stress Chaperones, vol.14, pp.83-94, 2009.

V. M. Longshaw, J. P. Chapple, M. S. Balda, M. E. Cheetham, and G. L. Blatch, Nuclear translocation of the Hsp70/Hsp90 organizing protein mSTI1 is regulated by cell cycle kinases, Journal of cell science, vol.117, pp.701-710, 2004.

M. Mollapour and L. Neckers, Post-translational modifications of Hsp90 and their contributions to chaperone regulation, Biochimica et biophysica acta, vol.1823, pp.648-655, 2012.

P. Muller, C-terminal phosphorylation of Hsp70 and Hsp90 regulates alternate binding to co-chaperones CHIP and HOP to determine cellular protein folding/degradation balances, Oncogene, vol.32, pp.3101-3110, 2013.

J. R. Webb, A. Campos-neto, Y. A. Skeiky, and S. G. Reed, Molecular characterization of the heat-inducible LmSTI1 protein of Leishmania major

, Mol Biochem Parasitol, vol.89, pp.179-193, 1997.

M. Morales, Phosphoproteome dynamics reveals heat shock protein complexes specific to theLeishmaniainfectious stage, Proc Natl Acad Sci, vol.107, pp.8381-8386, 2010.

G. Ommen, M. Chrobak, and J. Clos, The co-chaperone SGT ofLeishmania donovaniis essential for the parasite's viability, Cell Stress and Chaperones, vol.39, pp.541-546, 2010.

G. Ommen, S. Lorenz, and J. Clos, One-step generation of double-allele gene replacement mutants in Leishmania donovani, Int J Parasitol, vol.39, pp.541-546, 2009.

A. Hombach, G. Ommen, V. Sattler, and J. Clos, Leishmania donovani P23 protects parasites against HSP90 inhibitor-mediated growth arrest, Cell Stress Chaperones, vol.20, pp.673-685, 2015.

K. Bartsch, A. Hombach-barrigah, and J. Clos, Hsp90 inhibitors radicicol and geldanamycin have opposing effects on Leishmania Aha1-dependent proliferation, Cell Stress Chaperones, vol.22, pp.729-742, 2017.

M. A. Morales, Phosphoproteomic analysis of Leishmania donovanipro-and amastigote stages, Proteomics, vol.8, pp.350-363, 2008.

W. Yau, Cyclophilin 40-deficient Leishmania donovani fail to undergo stress-induced development of the infectious metacyclic stage, Cell. Microbiol, vol.93, pp.80-97, 2014.

A. D. Zuehlke, K. Beebe, L. Neckers, and T. Prince, Regulation and function of the human HSP90AA1 gene, Gene, vol.570, pp.8-16, 2015.

Y. G. Zhao, Hsp90 phosphorylation is linked to its chaperoning function. Assembly of the reovirus cell attachment protein, J Biol Chem, vol.276, pp.32822-32827, 2001.

M. Mollapour, S. Tsutsumi, Y. S. Kim, J. Trepel, and L. Neckers, Casein kinase 2 phosphorylation of Hsp90 threonine 22 modulates chaperone function and drug sensitivity, Oncotarget, vol.2, pp.407-417, 2011.

M. Mollapour, Threonine 22 phosphorylation attenuates Hsp90 interaction with cochaperones and affects its chaperone activity, Molecular cell, vol.41, pp.672-681, 2011.

W. Xu, Dynamic tyrosine phosphorylation modulates cycling of the HSP90-P50(CDC37)-AHA1 chaperone machine, Molecular cell, vol.47, pp.434-443, 2012.

K. M. Grant, Inhibitors of Leishmania mexicana CRK3 cyclin-dependent kinase: chemical library screen and antileishmanial activity, Antimicrob Agents Chemother, vol.48, pp.3033-3042, 2004.

M. Wiese, Leishmania MAP kinases-familiar proteins in an unusual context, Int J Parasitol, vol.37, pp.1053-1062, 2007.

E. Xingi, 6-Br-5methylindirubin-3?oxime (5-Me-6-BIO) targeting the leishmanial glycogen synthase kinase-3 (GSK-3) short form affects cell-cycle progression and induces apoptosis-like death: exploitation of GSK-3 for treating leishmaniasis, Int J Parasitol, vol.39, pp.1289-1303, 2009.
URL : https://hal.archives-ouvertes.fr/hal-00416035

G. F. Spath, S. Drini, and N. Rachidi, A touch of Zen: post-translational regulation of the Leishmania stress response, Cell Microbiol, vol.17, pp.632-638, 2015.
URL : https://hal.archives-ouvertes.fr/pasteur-01433407

S. M. Duncan, Conditional gene deletion with DiCre demonstrates an essential role for CRK3 in Leishmania mexicana cell cycle regulation, Mol Microbiol, vol.100, pp.931-944, 2016.

N. Rachidi, Pharmacological assessment defines Leishmania donovani casein kinase 1 as a drug target and reveals important functions in parasite viability and intracellular infection, Antimicrob Agents Chemother, vol.58, pp.2022-2035, 2014.
URL : https://hal.archives-ouvertes.fr/pasteur-01433412

M. Budini, Autophosphorylation of carboxy-terminal residues inhibits the activity of protein kinase CK1alpha, J Cell Biochem, vol.106, pp.399-408, 2009.

L. L. Vieira, N. Sacerdoti-sierra, and C. L. Jaffe, Effect of pH and temperature on protein kinase release by Leishmania donovani, Int J Parasitol, vol.32, pp.1085-1093, 2002.

J. M. Silverman, Proteomic analysis of the secretome of Leishmania donovani, Genome biology, vol.9, p.35, 2008.

J. Liu, Mammalian casein kinase 1alpha and its leishmanial ortholog regulate stability of IFNAR1 and type I interferon signaling, Mol Cell Biol, vol.29, pp.6401-6412, 2009.

M. Wiese, A mitogen-activated protein (MAP) kinase homologue of Leishmania mexicana is essential for parasite survival in the infected host, Embo J, vol.17, pp.2619-2628, 1998.

P. Kaur, M. Garg, A. Hombach-barrigah, J. Clos, and N. Goyal, MAPK1 of Leishmania donovani interacts and phosphorylates HSP70 and HSP90 subunits of foldosome complex, 2017.

P. Tsigankov, Regulation dynamics of Leishmania differentiation: deconvoluting signals and identifying phosphorylation trends, Mol Cell Proteomics, vol.13, pp.1787-1799, 2014.
URL : https://hal.archives-ouvertes.fr/pasteur-01433413

A. Zilka, S. Garlapati, E. Dahan, V. Yaolsky, and M. Shapira, Developmental regulation of HSP83 in Leishmania: transcript levels are controlled by the efficiency of 3? RNA processing and preferential translation is directed by a determinant in the 3?UTR, J Biol Chem, vol.11, p.11, 2001.

P. Hawle, The middle domain of Hsp90 acts as a discriminator between different types of client proteins, Mol Cell Biol, vol.26, pp.8385-8395, 2006.

E. Bifeld, Ribosome Profiling Reveals HSP90 Inhibitor Effects on Stage-specific Protein Synthesis in Leishmania donovani. mSystems accepted

D. L. Sacks and P. V. Perkins, Identification of an infective stage of Leishmania promastigotes, Science, vol.223, pp.1417-1419, 1984.

E. Bifeld, P. Tejera-nevado, J. Bartsch, J. Eick, and J. Clos, A versatile qPCR assay to quantify trypanosomatidic infections of host cells and tissues, Med Microbiol Immunol, vol.205, pp.449-458, 2016.

Y. L. Woods, The kinase DYRK phosphorylates protein-synthesis initiation factor eIF2Bepsilon at Ser539 and the microtubule-associated protein tau at Thr212: potential role for DYRK as a glycogen synthase kinase 3-priming kinase, Biochem J, vol.355, pp.609-615, 2001.

K. K. Ojo, Structure determination of glycogen synthase kinase-3 from Leishmania major and comparative inhibitor structure-activity relationships with Trypanosoma brucei GSK-3, Mol Biochem Parasitol, vol.176, pp.98-108, 2011.

U. Knippschild, The CK1 Family: Contribution to Cellular Stress Response and Its Role in Carcinogenesis, Front Oncol, vol.4, 2014.

O. Marin, A noncanonical sequence phosphorylated by casein kinase 1 in beta-catenin may play a role in casein kinase 1 targeting of important signaling proteins, Proc Natl Acad Sci, vol.100, pp.10193-10200, 2003.

F. Kawakami, K. Suzuki, and K. Ohtsuki, A novel consensus phosphorylation motif in sulfatide-and cholesterol-3-sulfate-binding protein substrates for CK1 in vitro, Biol Pharm Bull, vol.31, pp.193-200, 2008.

R. Amanchy, Identification of Novel Phosphorylation Motifs Through an Integrative Computational and Experimental Analysis of the Human Phosphoproteome, J Proteomics Bioinform, vol.4, pp.22-35, 2011.

M. Keller, R. L. Chan, L. H. Tessier, J. H. Weil, and P. Imbault, Post-transcriptional regulation by light of the biosynthesis of Euglena ribulose-1,5-bisphosphate carboxylase/oxygenase small subunit, Plant Mol Biol, vol.17, pp.73-82, 1991.

C. E. Clayton, Life without transcriptional control? From fly to man and back again, Embo J, vol.21, pp.1881-1888, 2002.

S. Martinez-calvillo, J. C. Vizuet-de-rueda, L. E. Florencio-martinez, R. G. Manning-cela, and E. E. Figueroa-angulo, Gene expression in trypanosomatid parasites, Journal of biomedicine & biotechnology, p.525241, 2010.

X. H. Liang, A. Haritan, S. Uliel, and S. Michaeli, trans and cis splicing in trypanosomatids: mechanism, factors, and regulation, Eukaryot Cell, vol.2, pp.830-840, 2003.

M. Mollapour, Asymmetric Hsp90 N domain SUMOylation recruits Aha1 and ATP-competitive inhibitors, Mol Cell, vol.53, pp.317-329, 2014.

Y. Xu, M. A. Singer, and S. Lindquist, Maturation of the tyrosine kinase c-src as a kinase and as a substrate depends on the molecular chaperone Hsp90, Proc Natl Acad Sci, vol.96, pp.109-114, 1999.

A. D. Zuehlke, An Hsp90 co-chaperone protein in yeast is functionally replaced by site-specific posttranslational modification in humans, Nat Commun, vol.8, 2017.

V. Legagneux, M. Morange, and O. Bensaude, Heat shock increases turnover of 90 kDa heat shock protein phosphate groups in HeLa cells, FEBS Lett, vol.291, pp.359-362, 1991.

M. Mollapour, S. Tsutsumi, and L. Neckers, Hsp90 phosphorylation, Wee1 and the cell cycle, Cell Cycle, vol.9, pp.2310-2316, 2010.

D. F. Nathan and S. Lindquist, Mutational analysis of Hsp90 function: interactions with a steroid receptor and a protein kinase, Mol Cell Biol, vol.15, pp.3917-3925, 1995.

M. K. Howard, G. Sayers, and M. A. Miles, Leishmania donovani metacyclic promastigotes: transformation in vitro, lectin agglutination, complement resistance, and infectivity, Exp Parasitol, vol.64, pp.147-156, 1987.

L. H. Pearl and C. Prodromou, Structure and mechanism of the Hsp90 molecular chaperone machinery, Annu Rev Biochem, vol.75, pp.271-294, 2006.

W. L. Yau, Phenotypic Characterization of a Leishmania donovani Cyclophilin 40 Null Mutant, J Eukaryot Microbiol, vol.63, pp.823-833, 2016.
URL : https://hal.archives-ouvertes.fr/pasteur-01451083

J. Li, K. Richter, and J. Buchner, Mixed Hsp90-cochaperone complexes are important for the progression of the reaction cycle, Nat Struct Mol Biol, vol.18, pp.61-66, 2011.

L. Sollelis, First efficient CRISPR-Cas9-mediated genome editing in Leishmania parasites, Cell Microbiol, vol.17, pp.1405-1412, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01992713

W. W. Zhang and G. Matlashewski, CRISPR-Cas9-Mediated Genome Editing inLeishmania donovani, MBio, vol.6, p.861, 2015.

P. Cohen, The regulation of protein function by multisite phosphorylation-a 25 year update, Trends Biochem Sci, vol.25, pp.596-601, 2000.

, Scientific RepoRts |, vol.9, p.5074, 2019.

T. Lahav, Multiple levels of gene regulation mediate differentiation of the intracellular pathogen Leishmania, FASEB journal: official publication of the Federation of American Societies for Experimental Biology, vol.25, pp.515-525, 2011.

D. Rosenzweig, Retooling Leishmania metabolism: from sand fly gut to human macrophage, FASEB J, vol.22, pp.590-602, 2008.

E. Bifeld, Leishmania: Methods and Protocols Methods in Molecular Biology, vol.12, 2019.

J. Sambrook and D. W. Russell, Molecular Cloning, 2001.

A. Schlüter, Expression and Subcellular Localization of Cpn60 Protein Family Members inLeishmania donovani, Biochim. Biophys. Acta, vol.1491, pp.65-74, 2000.

K. Choudhury, D. Zander, M. Kube, R. Reinhardt, and J. Clos, Identification of a Leishmania infantum gene mediating resistance to miltefosine and SbIII, Int J Parasitol, vol.38, pp.1411-1423, 2008.

R. Brun and . Schonenberger, Cultivation and in vitro cloning or procyclic culture forms of Trypanosoma brucei in a semi-defined medium, Acta Trop, vol.36, pp.289-292, 1979.

S. S. Jensen and M. R. Larsen, Evaluation of the impact of some experimental procedures on different phosphopeptide enrichment techniques, Rapid Commun Mass Spectrom, vol.21, pp.3635-3645, 2007.

J. Rappsilber, U. Ryder, A. I. Lamond, and M. Mann, Large-scale proteomic analysis of the human spliceosome, Genome Res, vol.12, pp.1231-1245, 2002.

P. Mortensen, MSQuant, an open source platform for mass spectrometry-based quantitative proteomics, Journal of proteome research, vol.9, pp.393-403, 2010.

S. A. Beausoleil, J. Villen, S. A. Gerber, J. Rush, and S. P. Gygi, A probability-based approach for high-throughput protein phosphorylation analysis and site localization, Nat Biotechnol, vol.24, pp.1285-1292, 2006.

M. Savitsky, Recording information on protein complexes in an information management system, J Struct Biol, vol.175, pp.224-229, 2011.

P. Poullet, S. Carpentier, and E. Barillot, myProMS, a web server for management and validation of mass spectrometry-based proteomic data, Proteomics, vol.7, pp.2553-2556, 2007.

B. Valot, O. Langella, E. Nano, and M. Zivy, MassChroQ: a versatile tool for mass spectrometry quantification, Proteomics, vol.11, pp.3572-3577, 2011.
URL : https://hal.archives-ouvertes.fr/hal-01481216

J. Clos and S. Brandau, pJC20 and pJC40-two high-copy-number vectors for T7 RNA polymerase-dependent expression of recombinant genes inEscherichia coli, Prot. Expression Purif, vol.5, pp.133-137, 1994.

A. Efstathiou, An inhibitor-driven study for enhancing the selectivity of indirubin derivatives towards leishmanial Glycogen Synthase Kinase-3 over leishmanial cdc2-related protein kinase 3, Parasites & vectors, vol.7, 2014.

U. K. Laemmli, Cleavage of structural proteins during the assembly of the head of bacteriophage T4, Nature, vol.227, pp.680-685, 1970.

H. B. Mann and D. R. Whitney, On a test of whether one of two random variables is stochastically larger than the other, Ann Math Statistics, vol.18, pp.50-60, 1947.