Characterisation of the rho GTPase target dishevelled associated activator of morphogenesis 1 (DAAM1

140 184 0
Characterisation of the rho GTPase target dishevelled associated activator of morphogenesis 1 (DAAM1

Đang tải... (xem toàn văn)

Tài liệu hạn chế xem trước, để xem đầy đủ mời bạn chọn Tải xuống

Thông tin tài liệu

CHARACTERISATION OF THE RHO GTPASE TARGET DISHEVELLED ASSOCIATED ACTIVATOR OF MORPHOGENESIS (DAAM1) ANG SU FEN NATIONAL UNIVERSITY OF SINGAPORE 2008 CHARACTERISATION OF THE RHO GTPASE TARGET DISHEVELLED ASSOCIATED ACTIVATOR OF MORPHOGENESIS (DAAM1) ANG SU FEN (B.Sc. (Hons.), NUS) A THESIS SUBMITTED FOR THE DEGREE OF DOCTOR OF PHILOSOPHY NUS GRADUATE SCHOOL FOR INTEGRATIVE SCIENCES AND ENGINEERING NATIONAL UNIVERSITY OF SINGAPORE 2008 Acknowledgements Acknowledgements First and foremost, I would like to thank my supervisor, Dr Ed Manser, for his mentorship and guidance in these four years. Also, I would like to thank Dr Jackson Zhao, my co-supervisor, who has offered me tremendous help, be it in experimental techniques or data analysis. I am also grateful to Prof Louis Lim for granting me a position in GSK-IMCB lab to conduct my research, which finally led to the generation of this thesis. At the same time, I would like to extend my gratitude to members on my thesis advisory committee, Dr Victor Yu and Dr Hooi Shing Chuan, for their suggestions and guidance that helped me progress through different stages of my project. I am most fortunate to be part of the GSK-IMCB lab, a fun and interactive lab that one would never want to leave. I would like to thank all my fellow lab mates for their support and presence in the lab which makes it a lively place to be in. Special thanks go to Elsa, Yeow Fong and Jet Phey, who provided me with much assistance when I first joined the lab and in a way, inducted me into the lab. Thanks also go to Yohendran, Delina, Praju, Jeff Yong, Paochun and Soon Tuck, fellow students who join me in the pursuit of a doctorate. I would also like to thank members of the lab (past and present) who have made my four years of stay most memorable: Sinnisky, Evonne, Charis, Christy, Puneet, Irene, Dr Jeff Robens, Dr Chan Wing, Dr Dong Jing Ming, Dr Perry Chan, Dr Thomas Leung, Dr Ivan Tan, Mr Joel Lee, Ms Rossiter and Ms Rani. Apart from my lab members, I also want to thank my best friends, Chengying, Jiamin, Aifen, June and Huifen, whom I have known for more than ten years. They -i- Acknowledgements never fail to offer encouragement and advice in times of need, which helped me overcome difficulties encountered in different stages of my life. I would also like to express my thanks to members in my Honours lab and my ex-classmates. They include Mirtha, Baohua, Koh Shiuan, Lily, Hang Yee, Kah Weng and Shiqi. I am also glad to have Boon Kiat, someone who have always been my source of comfort and support, for company in this arduous stage of thesis writing, In addition, I would like to express my thanks to my family, both immediate and extended, for being so understanding and encouraging in this whole process. Their never-ending support and unconditional care give me the strength to persevere till the end. Finally, I would like to thank A*STAR for giving me this scholarship, without which I would not be able to come this far, and also IMCB for providing the necessary facilities for my research. I would also like to thank NGS for providing their best support to students. It would be impossible to mention everyone here, so before I end I would like to say a big thank you to all those who have helped me in one way or another and have contributed to making this thesis a success. Thank you very much! - ii - Table of Contents Table of Contents Acknowledgements .i Table of Contents . iii Abstract .ix List of Figures and Tables .xi Abbreviations . xiii Chapter 1. 1.1. Introduction Formins 1.1.1. Formin family of proteins 1.1.2. Formin domains .2 1.1.3. Lessons from Diaphanous 1.2. Rho GTPases 10 1.2.1. Rho GTPase family 10 1.2.2. Regulation of Rho GTPases .11 1.2.3. RhoA, Rac1 and Cdc42 13 1.2.4. Rho GTPases in cell migration 16 1.3. Dishevelled Associated Activator of Morphogenesis (DAAM1) .17 - iii - Table of Contents 1.4. Thesis objectives 18 Chapter 2. Materials and Methods 20 2.1. Cell culture .20 2.2. Antibodies 20 2.3. Reagents .21 2.4. Plasmids and oligonucleotides .21 2.4.1. List of DAAM1 primers .22 2.5. Site-directed mutagenesis 24 2.6. Sequence analyses and alignment 26 2.7. Transfection .26 2.8. siRNA Transfection .26 2.9. Drug treatment .27 2.10. Lipid raft purification .27 2.11. Immunoprecipitation 27 2.12. Western analysis 28 2.13. Wound healing assays 28 2.14. Microinjection 29 - iv - Table of Contents 2.15. Immunofluorescence 29 2.16. Live-cell imaging .30 Chapter 3. 3.1. Results .31 DAAM1 .31 3.1.1. DAAM1 domains .31 3.1.2. DAAM1 isoforms 31 3.1.3. DAAM1 versus DAAM2 .34 3.1.4. DAAM1 in different species 34 3.1.5. Analysis of DAAM1 FH3 domain .34 3.2. Generation and purification of DAAM1 antibody .38 3.3. Localisation of DAAM1 38 3.3.1. Design of DAAM1 truncation constructs 40 3.3.2. Localisation of endogenous DAAM1 40 3.3.3. DAAM1 localisation to the actin cytoskeleton 40 3.3.4. DAAM1 and the Golgi apparatus 44 3.3.5. Localisation of putatively active DAAM1: DAAM1 FH1-FH2-DAD 44 -v- Table of Contents 3.3.6. The FH1-FH2-DAD from different formins produce similar phenotypes .49 3.3.7. The activity of DAAM1 in vivo requires a C-terminal basic region 54 3.3.8. Lipid raft association of DAAM1 59 3.4. Effects of DAAM1 on the cytokeleton 63 3.4.1. Effect of DAAM1 on actin 63 3.4.2. Effect of DAAM1 on microtubules .63 3.4.3. Effect of DAAM1 on myosin II .63 3.5. Interaction of DAAM1 with Rho GTPases 64 3.5.1. Co-immunopreciptation assays and Western analysis .64 3.5.2. Immunofluorescence 67 3.5.3. Deriving GTPase binding defective DAAM1 67 3.5.4. Localisation of DAAM1 N-ter, (1-233) and (1-440) GBD mutants 70 3.6. siRNA knockdown of DAAM1 in cell lines 73 3.7. DAAM1 and Dishevelled (Dvl2) 73 3.8. A system to test DAAM1 involvement in cell migration 75 3.8.1. DAAM1 is essential for Golgi apparatus orientation in cell migration .79 - vi - Table of Contents Chapter 4. 4.1. Discussion 90 DAAM1 localisation and its significance 90 4.1.1. DAAM1 localisation to actin stress fibres .91 4.1.2. DAAM1 localisation to the Golgi 92 4.1.3. DAAM1: To Golgi or actin stress fibres? 92 4.1.4. DAAM1 localisation with a subset of myosin II fibres .93 4.2. Defining regions important for DAAM1 activity in vivo 94 4.2.1. DAAM1 FH1-FH2-DAD and actin polymerisation 94 4.2.2. DAAM1 FH1-FH2-DAD and the HeLa phase-dark structures .95 4.2.3. DAAM1 FH1-FH2-DAD and microtubule reorganisation and stabilisation 96 4.2.4. DAAM1 FH1-FH2-DAD and lipid raft association 97 4.2.5. Contribution of different domains to activation .97 4.2.6. DAAM1 localisation: COS versus HeLa 99 4.3. Rho GTPases and DAAM1 99 4.3.1. Interaction of DAAM1 with GTPases .99 4.3.2. Analysis of DAAM1 GBD mutants .100 - vii - Table of Contents 4.4. DAAM1 and Dishevelled (Dvl2) 101 4.4.1. DAAM1 and Dvl2 localisation 102 4.4.2. DAAM1, Rho GTPase and Dvl2 association 103 4.5. Role of DAAM1 in polarised migration 103 4.6. A summary of DAAM1 localisation 106 4.7. Conclusion .108 Bibliography 110 - viii - Chapter 4: Discussion 4.7. Conclusion DAAM1 shares common features with formins in terms of domain structure. The regulation of DAAM1, however, may differ from that of typical formins since the Cterminal 33 residues (10/33 basic) is required for generation of long, fine actin fibres in HeLa cells, a phenomenon observed in other active formins. In mDia1, deletion of its C-ter region up to the DAD is reported to generate an active phenotype [37]. It would be interesting to compare directly the effects of DAAM1 with those of other formins. My results with DAAM1 is the first suggestion that the region C-terminal to the DAD positively contributes to formin function, and indeed in the case of DAAM1, is essential for in vivo effects. It would be of interest to test this with respect to Wnt signalling. DAAM1 can bind to active versions of RhoA, Rac1 and Cdc42. It seems likely that localisation of DAAM1 actually determines the type of Rho GTPase it interacts with and the connection to lipid rafts suggests segregation of DAAM1 signalling at the plasma membrane as for RhoA and Rac1 [61, 97]. The significance of the DAAM1 FH1-FH2-DAD enriched phase-dark patches in HeLa cells and colocalisation of Glu-tubulin and cholera toxin B needs to be tempered by the observation that caveolin-1 does not co-segregate with these structures (Figure 3.14d). HeLa cells appear to express some components that generate these structures but there is no clue as yet to its nature; it is only known that this process is not DAAM1specific. In COS cells, I have shown that DAAM1 knockdown resulted in an impairment of Golgi/MTOC orientation in wound assays. This places DAAM1 in the - 108 - Chapter 4: Discussion cell polarity pathway together with various players like Cdc42, Par6, and PKC . Since DAAM1 has been shown to mediate actin polymerisation and microtubule stabilisation, it is likely that DAAM1 is required for Golgi orientation because of its ability to drive cytoskeletal remodelling. Targeting of DAAM1 to the Golgi is GTPase independent but requires the first 44 residues whose function is not revealed by homology search. Wound assays performed in the presence of dominant inhibitory Cdc42N17 show strong inhibition of Golgi orientation as established [65]. Therefore, I favour the notion that DAAM1 interacts with GTP bound Cdc42 at the Golgi region in spite of the weaker biochemical interaction with this GTPase. Existing data suggests that DAAM1 is a complex multi-domain protein subjected to multiple levels of regulation. Future studies of DAAM1 should be greatly aided by the observations I have made here regarding the targeting and structurefunction relationships of the protein. If crystal structures of larger fragments of DAAM1 become available, some of the regulatory issues might be resolved. The role of N- and C-terminal residues to its localisation requires screening for potential interactors (both lipids and proteins). On the other hand, the use of model systems such as Xenopus embryonic development to analyse DAAM1 would provide a firm physiological context. Developmental models should be used to study how the various regions of DAAM1 cooperate with Wnt and Dishevelled to mediate morphological changes at the tissue and organism level. - 109 - Bibliography Bibliography 1. Woychik, R.P., et al., 'Formins': proteins deduced from the alternative transcripts of the limb deformity gene. Nature, 1990. 346(6287): p. 850-3. 2. Higgs, H.N., Formin proteins: a domain-based approach. Trends Biochem Sci, 2005. 30(6): p. 342-53. 3. Wallar, B.J. and A.S. Alberts, The formins: active scaffolds that remodel the cytoskeleton. Trends Cell Biol, 2003. 13(8): p. 435-46. 4. Evangelista, M., S. Zigmond, and C. Boone, Formins: signaling effectors for assembly and polarization of actin filaments. J Cell Sci, 2003. 116(Pt 13): p. 2603-11. 5. Xu, Y., et al., Crystal structures of a Formin Homology-2 domain reveal a tethered dimer architecture. Cell, 2004. 116(5): p. 711-23. 6. Shimada, A., et al., The core FH2 domain of diaphanous-related formins is an elongated actin binding protein that inhibits polymerization. Mol Cell, 2004. 13(4): p. 511-22. 7. Otomo, T., et al., Structural basis of Rho GTPase-mediated activation of the formin mDia1. Mol Cell, 2005. 18(3): p. 273-81. 8. Rose, R., et al., Structural and mechanistic insights into the interaction between Rho and mammalian Dia. Nature, 2005. 435(7041): p. 513-8. 9. Lammers, M., et al., The regulation of mDia1 by autoinhibition and its release by Rho*GTP. Embo J, 2005. 24(23): p. 4176-87. 10. Nezami, A.G., F. Poy, and M.J. Eck, Structure of the autoinhibitory switch in formin mDia1. Structure, 2006. 14(2): p. 257-63. - 110 - Bibliography 11. Watanabe, N., et al., p140mDia, a mammalian homolog of Drosophila diaphanous, is a target protein for Rho small GTPase and is a ligand for profilin. Embo J, 1997. 16(11): p. 3044-56. 12. Li, F. and H.N. Higgs, The mouse Formin mDia1 is a potent actin nucleation factor regulated by autoinhibition. Curr Biol, 2003. 13(15): p. 1335-40. 13. Li, F. and H.N. Higgs, Dissecting requirements for auto-inhibition of actin nucleation by the formin, mDia1. J Biol Chem, 2005. 280(8): p. 6986-92. 14. Copeland, S.J., et al., The DID-DAD interaction is able to mediate heterodimerization between mDia1 and mDia2. J Biol Chem, 2007. 282(41): p. 30120-30. 15. Alberts, A.S., Identification of a carboxyl-terminal diaphanous-related formin homology protein autoregulatory domain. J Biol Chem, 2001. 276(4): p. 282430. 16. Wallar, B.J., et al., The basic region of the diaphanous-autoregulatory domain (DAD) is required for autoregulatory interactions with the diaphanous-related formin inhibitory domain. J Biol Chem, 2006. 281(7): p. 4300-7. 17. Petersen, J., et al., FH3, a domain found in formins, targets the fission yeast formin Fus1 to the projection tip during conjugation. J Cell Biol, 1998. 141(5): p. 1217-28. 18. Kato, T., et al., Localization of a mammalian homolog of diaphanous, mDia1, to the mitotic spindle in HeLa cells. J Cell Sci, 2001. 114(Pt 4): p. 775-84. 19. Tominaga, T., et al., Diaphanous-related formins bridge Rho GTPase and Src tyrosine kinase signaling. Mol Cell, 2000. 5(1): p. 13-25. - 111 - Bibliography 20. Fujiwara, T., et al., Rho small G-protein-dependent binding of mDia to an Src homology domain-containing IRSp53/BAIAP2. Biochem Biophys Res Commun, 2000. 271(3): p. 626-9. 21. Kovar, D.R. and T.D. Pollard, Insertional assembly of actin filament barbed ends in association with formins produces piconewton forces. Proc Natl Acad Sci U S A, 2004. 101(41): p. 14725-30. 22. Romero, S., et al., Formin is a processive motor that requires profilin to accelerate actin assembly and associated ATP hydrolysis. Cell, 2004. 119(3): p. 419-29. 23. Kovar, D.R., et al., The fission yeast cytokinesis formin Cdc12p is a barbed end actin filament capping protein gated by profilin. J Cell Biol, 2003. 161(5): p. 875-87. 24. Pring, M., et al., Mechanism of formin-induced nucleation of actin filaments. Biochemistry, 2003. 42(2): p. 486-96. 25. Castrillon, D.H. and S.A. Wasserman, Diaphanous is required for cytokinesis in Drosophila and shares domains of similarity with the products of the limb deformity gene. Development, 1994. 120(12): p. 3367-77. 26. Pruyne, D., et al., Role of formins in actin assembly: nucleation and barbed-end association. Science, 2002. 297(5581): p. 612-5. 27. Deeks, M.J., P.J. Hussey, and B. Davies, Formins: intermediates in signaltransduction cascades that affect cytoskeletal reorganization. Trends Plant Sci, 2002. 7(11): p. 492-8. 28. Pollard, T.D., Formins initiate new actin filaments. Nat Cell Biol, 2002. 4(8): p. E191. - 112 - Bibliography 29. Sagot, I., S.K. Klee, and D. Pellman, Yeast formins regulate cell polarity by controlling the assembly of actin cables. Nat Cell Biol, 2002. 4(1): p. 42-50. 30. Harris, E.S., F. Li, and H.N. Higgs, The mouse formin, FRLalpha, slows actin filament barbed end elongation, competes with capping protein, accelerates polymerization from monomers, and severs filaments. J Biol Chem, 2004. 279(19): p. 20076-87. 31. Zigmond, S.H., et al., Formin leaky cap allows elongation in the presence of tight capping proteins. Curr Biol, 2003. 13(20): p. 1820-3. 32. Moseley, J.B., et al., A conserved mechanism for Bni1- and mDia1-induced actin assembly and dual regulation of Bni1 by Bud6 and profilin. Mol Biol Cell, 2004. 15(2): p. 896-907. 33. Otomo, T., et al., Structural basis of actin filament nucleation and processive capping by a formin homology domain. Nature, 2005. 433(7025): p. 488-94. 34. Shemesh, T., et al., A novel mechanism of actin filament processive capping by formin: solution of the rotation paradox. J Cell Biol, 2005. 170(6): p. 889-93. 35. Pollard, T.D., Formins coming into focus. Dev Cell, 2004. 6(3): p. 312-4. 36. Krebs, A., et al., Characterization of functional domains of mDia1, a link between the small GTPase Rho and the actin cytoskeleton. J Cell Sci, 2001. 114(Pt 20): p. 3663-72. 37. Watanabe, N., et al., Cooperation between mDia1 and ROCK in Rho-induced actin reorganization. Nat Cell Biol, 1999. 1(3): p. 136-43. 38. Ishizaki, T., et al., Coordination of microtubules and the actin cytoskeleton by the Rho effector mDia1. Nat Cell Biol, 2001. 3(1): p. 8-14. - 113 - Bibliography 39. Palazzo, A.F., et al., mDia mediates Rho-regulated formation and orientation of stable microtubules. Nat Cell Biol, 2001. 3(8): p. 723-9. 40. Wen, Y., et al., EB1 and APC bind to mDia to stabilize microtubules downstream of Rho and promote cell migration. Nat Cell Biol, 2004. 6(9): p. 820-30. 41. Eng, C.H., T.M. Huckaba, and G.G. Gundersen, The formin mDia regulates GSK3beta through novel PKCs to promote microtubule stabilization but not MTOC reorientation in migrating fibroblasts. Mol Biol Cell, 2006. 17(12): p. 5004-16. 42. Copeland, J.W. and R. Treisman, The diaphanous-related formin mDia1 controls serum response factor activity through its effects on actin polymerization. Mol Biol Cell, 2002. 13(11): p. 4088-99. 43. Geneste, O., J.W. Copeland, and R. Treisman, LIM kinase and Diaphanous cooperate to regulate serum response factor and actin dynamics. J Cell Biol, 2002. 157(5): p. 831-8. 44. Sakata, D., et al., Impaired T lymphocyte trafficking in mice deficient in an actin-nucleating protein, mDia1. J Exp Med, 2007. 204(9): p. 2031-38. 45. Eisenmann, K.M., et al., T cell responses in mammalian diaphanous-related formin mDia1 knockout mice. J Biol Chem, 2007. 282(34): p. 25152-58 46. Colucci-Guyon, E., et al., A role for mammalian diaphanous-related formins in complement receptor (CR3)-mediated phagocytosis in macrophages. Curr Biol, 2005. 15(22): p. 2007-12. 47. Brandt, D.T., et al., Dia1 and IQGAP1 interact in cell migration and phagocytic cup formation. J Cell Biol, 2007. 178(2): p. 193-200. - 114 - Bibliography 48. Kitzing, T.M., et al., Positive feedback between Dia1, LARG, and RhoA regulates cell morphology and invasion. Genes Dev, 2007. 21(12): p. 1478-83. 49. Wallar, B.J., et al., RhoB and the mammalian Diaphanous-related formin mDia2 in endosome trafficking. Exp Cell Res, 2007. 313(3): p. 560-71. 50. Fernandez-Borja, M., et al., RhoB regulates endosome transport by promoting actin assembly on endosomal membranes through Dia1. J Cell Sci, 2005. 118(Pt 12): p. 2661-70. 51. Wennerberg, K. and C.J. Der, Rho-family GTPases: it's not only Rac and Rho (and I like it). J Cell Sci, 2004. 117(Pt 8): p. 1301-12. 52. Paduch, M., F. Jelen, and J. Otlewski, Structure of small G proteins and their regulators. Acta Biochim Pol, 2001. 48(4): p. 829-50. 53. Vetter, I.R. and A. Wittinghofer, The guanine nucleotide-binding switch in three dimensions. Science, 2001. 294(5545): p. 1299-304. 54. Feig, L.A., Tools of the trade: use of dominant-inhibitory mutants of Ras-family GTPases. Nat Cell Biol, 1999. 1(2): p. E25-7. 55. Leung, T., et al., The p160 RhoA-binding kinase ROK alpha is a member of a kinase family and is involved in the reorganization of the cytoskeleton. Mol Cell Biol, 1996. 16(10): p. 5313-27. 56. Ishizaki, T., et al., The small GTP-binding protein Rho binds to and activates a 160 kDa Ser/Thr protein kinase homologous to myotonic dystrophy kinase. Embo J, 1996. 15(8): p. 1885-93. 57. Kimura, K., et al., Regulation of myosin phosphatase by Rho and Rhoassociated kinase (Rho-kinase). Science, 1996. 273(5272): p. 245-8. - 115 - Bibliography 58. Arber, S., et al., Regulation of actin dynamics through phosphorylation of cofilin by LIM-kinase. Nature, 1998. 393(6687): p. 805-9. 59. Chong, L.D., et al., The small GTP-binding protein Rho regulates a phosphatidylinositol 4-phosphate 5-kinase in mammalian cells. Cell, 1994. 79(3): p. 507-13. 60. Ridley, A.J. and A. Hall, The small GTP-binding protein rho regulates the assembly of focal adhesions and actin stress fibers in response to growth factors. Cell, 1992. 70(3): p. 389-99. 61. Palazzo, A.F., et al., Localized stabilization of microtubules by integrin- and FAK-facilitated Rho signaling. Science, 2004. 303(5659): p. 836-9. 62. Ridley, A.J., et al., The small GTP-binding protein rac regulates growth factorinduced membrane ruffling. Cell, 1992. 70(3): p. 401-10. 63. Diekmann, D., et al., Interaction of Rac with p67phox and regulation of phagocytic NADPH oxidase activity. Science, 1994. 265(5171): p. 531-3. 64. Alison E. M. et al., CDC42 and CDC43, Two additional genes involved in budding and the establishment of cell polarity in the yeast Saccharomyces cerevisiae. J Cell Biol., 1990. 111(1): p. 131-42. 65. Nobes, C.D. and A. Hall, Rho, Rac, and Cdc42 GTPases regulate the assembly of multimolecular focal complexes associated with actin stress fibers, lamellipodia, and filopodia. Cell, 1995. 81(1): p. 53-62. 66. Kozma, R., et al., The Ras-related protein Cdc42Hs and bradykinin promote formation of peripheral actin microspikes and filopodia in Swiss 3T3 fibroblasts. Mol Cell Biol, 1995. 15(4): p. 1942-52. - 116 - Bibliography 67. Manser, E., et al., A brain serine/threonine protein kinase activated by Cdc42 and Rac1. Nature, 1994. 367(6458): p. 40-6. 68. Burbelo, P. D. et al., A conserved binding motif defines numerous candidate target proteins for both Cdc42 and Rac GTPases. J. Biol. Chem. 270(49): p. 29071–74. 69. Lei, M., et al., Structure of PAK1 in an autoinhibited conformation reveals a multistage activation switch. Cell, 2000. 102(3): p. 387-97. 70. Edwards, D.C., et al., Activation of LIM-kinase by Pak1 couples Rac/Cdc42 GTPase signalling to actin cytoskeletal dynamics. Nat Cell Biol, 1999. 1(5): p. 253-9. 71. Sanders, L.C., et al., Inhibition of myosin light chain kinase by p21-activated kinase. Science, 1999. 283(5410): p. 2083-5. 72. Wittmann, T., G.M. Bokoch, and C.M. Waterman-Storer, Regulation of microtubule destabilizing activity of Op18/stathmin downstream of Rac1. J Biol Chem, 2004. 279(7): p. 6196-203. 73. Manser, E., et al., Expression of constitutively active alpha-PAK reveals effects of the kinase on actin and focal complexes. Mol Cell Biol, 1997. 17(3): p. 112943. 74. Zhao, Z.S., et al., Coupling of PAK-interacting exchange factor PIX to GIT1 promotes focal complex disassembly. Mol Cell Biol, 2000. 20(17): p. 6354-63. 75. Machesky, L.M. and R.H. Insall, Scar1 and the related Wiskott-Aldrich syndrome protein, WASP, regulate the actin cytoskeleton through the Arp2/3 complex. Curr Biol, 1998. 8(25): p. 1347-56. - 117 - Bibliography 76. Yasuda, S., et al., Cdc42 and mDia3 regulate microtubule attachment to kinetochores. Nature, 2004. 428(6984): p. 767-71. 77. Fukata Y. et al., Phosphorylation of adducing by Rho-kinase plays a crucial role in cell motility. J. Cell Biol., 1999. 145: p.347-361. 78. Pertz O. et al., Spatiotemporal dynamics of RhoA activity in migrating cells. Nature, 2006. 440: p. 1069-1072. 79. Ridley, A.J., et al., Cell migration: integrating signals from front to back. Science, 2003. 302(5651): p. 1704-9. 80. Raftopoulou, M. and A. Hall, Cell migration: Rho GTPases lead the way. Dev Biol, 2004. 265(1): p. 23-32. 81. Habas, R., Y. Kato, and X. He, Wnt/Frizzled activation of Rho regulates vertebrate gastrulation and requires a novel Formin homology protein Daam1. Cell, 2001. 107(7): p. 843-54. 82. Sato, A., et al., Profilin is an effector for Daam1 in non-canonical Wnt signaling and is required for vertebrate gastrulation. Development, 2006. 133(21): p. 4219-31. 83. Matusek, T., et al., The Drosophila formin DAAM regulates the tracheal cuticle pattern through organizing the actin cytoskeleton. Development, 2006. 133(5): p. 957-66. 84. Aspenstrom, P., N. Richnau, and A.S. Johansson, The diaphanous-related formin DAAM1 collaborates with the Rho GTPases RhoA and Cdc42, CIP4 and Src in regulating cell morphogenesis and actin dynamics. Exp Cell Res, 2006. 312(12): p. 2180-94. - 118 - Bibliography 85. Liu, W. et al., Mechanism of activation of the Formin protein Daam1. PNAS, 2007. 105 (1): p. 210-215. 86. Lu, J., et al., Structure of the FH2 domain of Daam1: implications for formin regulation of actin assembly. J Mol Biol, 2007. 369(5): p. 1258-69. 87. Yamashita M. et al., Crystal structure of human DAAM1 formin homology domain. Genes Cells, 2007. 12(11): p. 1255-1265. 88. Foster, L.J., C.L. De Hoog, and M. Mann, Unbiased quantitative proteomics of lipid rafts reveals high specificity for signaling factors. Proc Natl Acad Sci U S A, 2003. 100(10): p. 5813-8. 89. Jaime Millán, M.C.d.M., Mohammed Qaidi, Alicia Batista, Fernando MartínBelmonte, and Miguel A. Alonso, Isolation of Lipid Raft-Associated Proteolipids. Membrane protein protocols: expression, purification, and characterization, ed. B.S. Selinsky. 2003, Totowa, N.J: Humana Press. 90. Li, F., et al., p21-activated kinase interacts with and phosphorylates histone H3 in breast cancer cells. EMBO Rep, 2002. 3(8): p. 767-73. 91. Zhao, Z.S., et al., The GIT-associated kinase PAK targets to the centrosome and regulates Aurora-A. Mol Cell, 2005. 20(2): p. 237-49. 92. Wallingford, J.B. and R. Habas, The developmental biology of Dishevelled: an enigmatic protein governing cell fate and cell polarity. Development, 2005. 132(20): p. 4421-36. 93. Smalley, M.J., et al., Interaction of axin and Dvl-2 proteins regulates Dvl-2stimulated TCF-dependent transcription. Embo J, 1999. 18(10): p. 2823-35. 94. Zhang, L., et al., Dapper antagonizes Wnt signaling by promoting dishevelled degradation. J Biol Chem, 2006. 281(13): p. 8607-12. - 119 - Bibliography 95. Gomes, E.R., S. Jani, and G.G. Gundersen, Nuclear movement regulated by Cdc42, MRCK, myosin, and actin flow establishes MTOC polarization in migrating cells. Cell, 2005. 121(3): p. 451-63. 96. Kolega, J., Cytoplasmic dynamics of myosin IIA and IIB: spatial 'sorting' of isoforms in locomoting cells. J Cell Sci, 1998. 111 (Pt 15): p. 2085-95. 97. del Pozo, M.A., et al., Integrins regulate Rac targeting by internalization of membrane domains. Science, 2004. 303(5659): p. 839-42. 98. Zhao, Z.S., et al., A conserved negative regulatory region in alphaPAK: inhibition of PAK kinases reveals their morphological roles downstream of Cdc42 and Rac1. Mol Cell Biol, 1998. 18(4): p. 2153-63. 99. Chong, C., et al., The mechanism of PAK activation. Autophosphorylation events in both regulatory and kinase domains control activity. J Biol Chem, 2001. 276(20): p. 17347-53. 100. Yvon, A.M., et al., Centrosome reorientation in wound-edge cells is cell type specific. Mol Biol Cell, 2002. 13(6): p. 1871-80. 101. Dunphy, W.G. and J.E. Rothman, Compartmental organization of the Golgi stack. Cell, 1985. 42(1): p. 13-21. 102. Chege, N.W. and S.R. Pfeffer, Compartmentation of the Golgi complex: brefeldin-A distinguishes trans-Golgi cisternae from the trans-Golgi network. J Cell Biol, 1990. 111(3): p. 893-9. 103. Totsukawa, G., et al., Distinct roles of ROCK (Rho-kinase) and MLCK in spatial regulation of MLC phosphorylation for assembly of stress fibers and focal adhesions in 3T3 fibroblasts. J Cell Biol, 2000. 150(4): p. 797-806. - 120 - Bibliography 104. Pellegrin, S. and H. Mellor, The Rho family GTPase Rif induces filopodia through mDia2. Curr Biol, 2005. 15(2): p. 129-33. 105. Schirenbeck, A., et al., Formins and VASPs may co-operate in the formation of filopodia. Biochem Soc Trans, 2005. 33(Pt 6): p. 1256-9. 106. Schirenbeck, A., et al., The Diaphanous-related formin dDia2 is required for the formation and maintenance of filopodia. Nat Cell Biol, 2005. 7(6): p. 61925. 107. Gundersen, G.G., M.H. Kalnoski, and J.C. Bulinski, Distinct populations of microtubules: tyrosinated and nontyrosinated alpha tubulin are distributed differently in vivo. Cell, 1984. 38(3): p. 779-89. 108. Gundersen, G.G., S. Khawaja, and J.C. Bulinski, Postpolymerization detyrosination of alpha-tubulin: a mechanism for subcellular differentiation of microtubules. J Cell Biol, 1987. 105(1): p. 251-64. 109. Bulinski, J.C. and G.G. Gundersen, Stabilization of post-translational modification of microtubules during cellular morphogenesis. Bioessays, 1991. 13(6): p. 285-93. 110. Bulinski, J.C., J.E. Richards, and G. Piperno, Posttranslational modifications of alpha tubulin: detyrosination and acetylation differentiate populations of interphase microtubules in cultured cells. J Cell Biol, 1988. 106(4): p. 1213-20. 111. Cook, T.A., T. Nagasaki, and G.G. Gundersen, Rho guanosine triphosphatase mediates the selective stabilization of microtubules induced by lysophosphatidic acid. J Cell Biol, 1998. 141(1): p. 175-85. 112. Goode, B.L. and M.J. Eck, Mechanism and function of formins in the control of actin assembly. Annu Rev Biochem, 2007. 76: p. 593-627. - 121 - Bibliography 113. Kupfer, A., D. Louvard, and S.J. Singer, Polarization of the Golgi apparatus and the microtubule-organizing center in cultured fibroblasts at the edge of an experimental wound. Proc Natl Acad Sci U S A, 1982. 79(8): p. 2603-7. 114. Sandoval, I.V., et al., Role of microtubules in the organization and localization of the Golgi apparatus. J Cell Biol, 1984. 99(1 Pt 2): p. 113s-118s. 115. Skoufias, D.A., T.L. Burgess, and L. Wilson, Spatial and temporal colocalization of the Golgi apparatus and microtubules rich in detyrosinated tubulin. J Cell Biol, 1990. 111(5 Pt 1): p. 1929-37. 116. Thyberg, J. and S. Moskalewski, Role of microtubules in the organization of the Golgi complex. Exp Cell Res, 1999. 246(2): p. 263-79. 117. Bershadsky, A.D. and A.H. Futerman, Disruption of the Golgi apparatus by brefeldin A blocks cell polarization and inhibits directed cell migration. Proc Natl Acad Sci U S A, 1994. 91(12): p. 5686-9. 118. Mellor, H., Cell motility: Golgi signalling shapes up to ship out. Curr Biol, 2004. 14(11): p. R434-5. 119. Watanabe, T., J. Noritake, and K. Kaibuchi, Regulation of microtubules in cell migration. Trends Cell Biol, 2005. 15(2): p. 76-83. 120. Etienne-Manneville, S. and A. Hall, Cdc42 regulates GSK-3beta and adenomatous polyposis coli to control cell polarity. Nature, 2003. 421(6924): p. 753-6. 121. Schlessinger, K., E.J. McManus, and A. Hall, Cdc42 and noncanonical Wnt signal transduction pathways cooperate to promote cell polarity. J Cell Biol, 2007. 178(3): p. 355-61. - 122 - Bibliography 122. Efimov, A., et al., Asymmetric CLASP-dependent nucleation of noncentrosomal microtubules at the trans-Golgi network. Dev Cell, 2007. 12(6): p. 917-30. 123. Yamana N. et al., The Rho-mDia1 pathway regulates cell polarity and focal adhesion turnover in migrating cells through mobilizing Apc and c-Src. Mol Cell Biol. 2006. 26(18): p. 6844-58. - 123 - [...]... Figure 1. 3 Crystal structure of mDia1 N ter and RhoC 5 Figure 1. 4 Dendrogram of Rho GTPase family 10 Figure 1. 5 Regulation of Rho GTPase activity 12 Figure 1. 6 Role of Rho GTPases in cell migration 16 Figure 2 .1 Site directed mutagenesis (two-step PCR) 25 Figure 3 .1 DAAM1 domain structure Figure 3.2 Alignment of DAAM1 isoforms 35 Figure 3.3 Alignment of DAAM1 and DAAM2 36 Figure 3.4 Alignment of FH3... Effects of DAAM1 10 0-350 on myosin II in COS 7 cells 66 Figure 3 .17 Interaction of DAAM1 with Rho GTPases (Co-IP) 68 Figure 3 .18 Interaction of DAAM1 with active Rho GTPases (IF) 69 Figure 3 .19 Generation of DAAM1 N-ter GBD mutants and determination of binding to the Rho GTPases 71 Figure 3.20 Localisation of DAAM1 GBD mutants 72 Figure 3. 21 siRNA knockdown of DAAM1 74 Figure 3.22 DAAM1 and Dishevelled. .. site - xv - Chapter 1: Introduction Chapter 1 Introduction 1. 1 Formins The focus of this thesis is on Dishevelled Associated Activator of Morphogenesis 1 (DAAM1) , a member of the formin family of proteins The name formin originated from the discovery of the limb deformity gene in mice This gene was found to be important for the formation of the limbs and kidneys, hence was given the name formin Mice... the biochemistry of DAAM1, the protein that is the focus of this thesis Habas et al identified DAAM1 as an interactor of the PDZ domain of mouse Dishevelled 2 (Dvl2) by yeast two hybrid [ 81] DAAM1 is placed downstream of Wnt signalling as DAAM1 is required for interaction of Dvl with RhoA upon Wnt stimulation The Dvl-RhoA-DAAM1 complex is proposed to be essential for activation of RhoA Use of DAAM1... endogenous Rho activation 1. 2.3 RhoA, Rac1 and Cdc42 RhoA, Rac1 and Cdc42 are the most well studied members of the Rho GTPase family They have been shown to be involved in various signalling pathways, many of which involve the cytoskeleton There are three members in the RhoA subfamily, RhoA, RhoB and RhoC RhoA has been shown to bind to downstream targets like Rhokinase (ROK/ROCK) [55, 56] and mDia1 [36]... In the case of the fission yeast, sequences within the FH3 of Fus1p targets it to the projection tip during conjugation [17 ] The C-terminal portion of the FH3 in mDia1 is required for its localisation to the mitotic spindle in HeLa cells [18 ] Given the absence of well defined residues and the lack of functional data, the existence of a FH3 “domain” is speculative -5- Chapter 1: Introduction The FH1... [11 ], many studies have been performed to investigate its structure and function Biochemical data from Watanabe et al supports the binding of endogenous mDia1 to activated RhoA and profilin [11 ] The interaction of mDia1 with profilin was independent of RhoA binding and was shown in later studies to be mediated by the FH1 domain [36] The FH1 and FH2 domains of mDia1 are both required for formation of. .. pathways generates the spatial and temporal delivery of components for movement of the cell [79, 80] Cdc42: directional sensing & polarity Rho: contractility Golgi Nucleus Rac: lamellipodia Rho: protrusions Trailing edge Leading edge Figure 1. 6 Role of Rho GTPases in cell migration - 16 - Chapter 1: Introduction 1. 3 Dishevelled Associated Activator of Morphogenesis 1 (DAAM1) Most studies of formins have... the lipid tails of the GTPases An activating signal is required to trigger the release of Rho GTPases from the GDIs and allow their translocation to the membrane for another round of activation Cycling between the “ON” and “OFF” states ensures that the Rho GTPases function as efficient switches in cellular signalling where fast activation and termination of the signal is required (Figure 1. 5) GTP Rho- GDP... [5] The structure of the GBD with either the DAD or RhoC was also resolved for mouse Diaphanous (mDia) [6 -10 ] These successes in protein structure resolution helped substantiate the -1- Chapter 1: Introduction proposed structural organisation of various domains and the mechanism of formin regulation and function Dia2 FMN1 DAAM1 DAAM2 Dia3 Dia1 FMN2 FRL2 Delphilin FRL3 FRL1 FHOD1 INF2 FHOD2 INF1 Figure . Rho GTPases 10 1. 2 .1. Rho GTPase family 10 1. 2.2. Regulation of Rho GTPases 11 1. 2.3. RhoA, Rac1 and Cdc42 13 1. 2.4. Rho GTPases in cell migration 16 1. 3. Dishevelled Associated Activator of. Chapter 3. Results 31 3 .1. DAAM1 31 3 .1. 1. DAAM1 domains 31 3 .1. 2. DAAM1 isoforms 31 3 .1. 3. DAAM1 versus DAAM2 34 3 .1. 4. DAAM1 in different species 34 3 .1. 5. Analysis of DAAM1 FH3 domain 34. Chapter 1: Introduction - 1 - Chapter 1. Introduction 1. 1. Formins The focus of this thesis is on Dishevelled Associated Activator of Morphogenesis 1 (DAAM1) , a member of the formin family of

Ngày đăng: 12/09/2015, 08:19

Từ khóa liên quan

Tài liệu cùng người dùng

  • Đang cập nhật ...

Tài liệu liên quan