Characterization of pin1 function in zebrafish development

166 223 0
Characterization of pin1 function in zebrafish development

Đang tải... (xem toàn văn)

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

Thông tin tài liệu

CHARACTERIZATION OF ZPIN1 FUNCTION IN ZEBRAFISH DEVELOPMENT ZHAO LIQUN NATIONAL UNIVERSITY OF SINGAPORE 2008 CHARACTERIZATION OF ZPIN1 FUNCTION IN ZEBRAFISH DEVELOPMENT ZHAO LIQUN B.Sc., Shandong University, China; M.Sc., Peking Union Medical College, China A THESIS SUBMITTED FOR THE DEGREE OF DOCTOR OF PHILOSOPHY DEPARTMENT OF BIOLOGICAL SCIENCES NATIONAL UNIVERSITY OF SINGAPORE 2008 Acknowledgements I greatly appreciate the help and support from my supervisor, and labmates during my graduate study in Department of Biological Sciences, National University of Singapore. Without them, I would not be able to accomplish my research work. First of all, I would like to express my sincere gratitude to my supervisor, Dr. Liou Yih-Cherng. He is such a knowledgeable, and wise person who is always guiding me and offering me wonderful advices whenever I have difficulties in my research. I am grateful for his consistent encouragement and support which help me finish my research project, and make my graduate career more enjoyable. I would like to thank Dr. Vladimir Korzh and Dr. Steven Fong. With generous help from Dr. Korzh, I was able to continue my research project in IMCB for later two years of my graduate career. There, I received systematic training on manipulating embryos, and taking excellent pictures from Dr. Steven. Furthermore, they introduced me to the wonderful world of neurobiology. Without their tremendous help both on techniques and on knowledge, I can not accomplish the latter part of this project. I want to express my sincere gratitude to my QE committee members: Dr. Wang Shu, Dr. Low Boon Chuan and Dr. Vladimir Korzh. They kindly give me helpful suggestions which are beneficial for my research work for my whole Ph.D period. I also want to thank Dr. Faroq, Dr. Liu Lihui, Dr. Zhu Shizen, Dr. Zhan Huiqing and Dr. Cathleen Teh for their valuable suggestions and enthusiastic help. I appreciate the help from my labmates: Dr. Wang Yu, Zhou Wei, Liu Jun, Xia Yun, Yang Qiaoyun, Tan Weiwei, Ye Fan and Lai Cherng-Yu. In my entire graduate career, they are very helpful in effective troubleshooting by discussing and solving problems. Lastly, my innermost gratitude goes to my family. Whenever I feel worried or depressed, it is their selfless love and encouragements that make me calm down and i continue my research work. I especially need to thank my husband, Li Hao. He is my spirit support through four years. Therefore, I would like to dedicate my thesis to my parents, my brother, and my husband. ii Table of contents Acknowledgements .i Table of contents iii List of Tables .ix List of Figures .x List of Abbreviations .xii Chapter Introduction 1.1 The function of Pin1 1.1.1 The relationship between Pin1 and protein phosphorylation 1.1.2 The characteristics of Pin1 1.1.3 Pin1 function in cell cycle and cancer 1.1.3.1 Pin1 function in M phase .5 1.1.3.2 Pin1 function in G1 and S phase 1.1.3.3 Pin1 function in oncogenesis .9 1.1.4 Pin1 function in apoptosis .10 1.1.5 Pin1 function in Alzheimer’s disease .11 1.1.6 Pin1 function in development .16 1.1.7 Pin1 function on protein stability 19 1.1.7.1 The ubiquitin-proteasome pathway 19 1.1.7.2 The role of Pin1 in protein stability .19 1.1.8 The summary of Pin1 function .23 1.2.1 Introduction of bHLH factors .25 1.2.2 Overview of NeuroD function 26 1.2.3 Phenotypes for NeuroD-null mice 27 1.2.4 Protein interactors of NeuroD .28 iii 1.2.5 Post-translational regulation of NeuroD .30 1.2.6 The upstream and downstream regulators of NeuroD 32 1.3 Advantages of zebrafish model 34 1.4 Introduction of zebrafish lateral line 35 1.4.1 Zebrafish lateral line developmental process 35 1.5 Objectives of this study 37 Chapter Materials and Methods 39 2.1 Molecular technology 39 2.1.1 Polymerase chain reaction (PCR) .39 2.1.2 PCR product purification 39 2.1.3 DNA ligation and transformation .40 2.1.4 DNA sequencing .41 2.1.5 Rapid amplification of cDNA ends (RACE) 41 2.1.6 Southern Blotting 42 2.1.6.1 Probe synthesis .42 2.1.6.2 Isolation of genomic DNA .42 2.1.6.3 Digestion of genomic DNA .43 2.1.6.4 Neutralization, transfer and fixation 43 2.1.6.5 Prehybridization and hybridization 44 2.1.6.6 Washing and blocking 44 2.1.6.7 Antibody incubation and detection 44 2.2 In vitro studies using cell lines .45 2.2.1 Cell lines and cell culture 45 2.2.2 Transfection and cell lysates collection 45 2.2.3 SDS-PAGE and Western blotting .46 iv 2.2.4 Site-directed mutagenesis .47 2.2.5 Co-immunoprecipitation .48 2.2.6 Expression and purification of recombinant GST-Pin1 49 2.2.7 GST pull-down assay 49 2.2.8 Stability and rescue assay .50 2.2.8.1 Protein concentration measurement .50 2.2.8.2 Stability assay 50 2.2.8.3 Rescue assay 51 2.2.9 Immunostaining 51 2.3 In vivo studies using zebrafish embryos 51 2.3.1 Maintenance and staging of zebrafish strains .51 2.3.2 In vitro transcription .52 2.3.3 Microinjection .52 2.3.4 In situ hybridization 53 2.3.4.1 Synthesis of labeled RNA probe 53 2.3.4.2 Embryos collection and fixation 54 2.3.4.3 Proteinase K treatment .55 2.3.4.4 Prehybridization .55 2.3.4.5 Hybridization .55 2.3.4.6 Post-hybridization washes .56 2.3.4.7 Antibody incubation .56 2.3.4.8 Color development .57 2.3.4.9 Mounting and photographing .57 2.3.5 Immunohistochemical staining on embryos .58 2.3.6 Acridine Orange staining 58 v 2.3.7 Total RNA extraction from zebrafish embryos 59 2.3.8 Reverse-transcriptase PCR (RT-PCR) 59 Chapter Results .61 3.1 Molecular analysis of zebrafish Pin1 .61 3.1.1 Characterization of zebrafish Pin1 61 3.1.2 Sequence alignment of Pin1 in various species 63 3.1.3 Expression pattern of zPin1 in zebrafish embryos 65 3.2.2 The interaction of zPin1 with NeuroD via pSer/Thr-Pro motif 73 3.3 zPin1 morpholino knockdown phenotypes 78 3.3.1 The knockdown efficiency of both zPin1 morpholinos 78 3.3.2 Developmental delay caused by zPin1 loss-of-function .80 3.3.3 Global apoptosis caused by zPin1 lost-of-function 84 3.3.4 M-phase arrest caused by zPin1 loss-of-function .85 3.3.5 Neuronal phenotypes caused by zPin1 lost-of-function .87 3.3.5.1 The defects on mature neurons 87 3.3.5.2 The defects for neuroD expression 89 3.3.5.3 Neuromasts hair cells defects .90 3.3.5.4 Neuromasts mantle cells defects 96 3.3.6 The specificity of neuromasts defects .96 3.4 Regulation of NeuroD stability by zPin1 .100 3.5 Rescue of NeuroD stability by Pin1 .101 3.6 The effects of zPin1 on insulin gene expression 105 Chapter Discussion and Conclusion 107 4.1 Molecular analysis of zebrafish Pin1 .107 4.2 The effects of zPin1 morpholinos 109 vi 4.3 Developmental delay caused by zPin1 knockdown .110 4.4 The interaction between zPin1 and NeuroD 112 4.5 Protein stabilizing effect of NeuroD by zPin1 regulation 114 4.6 The specificity of neuromasts defects 117 4.7 Transcriptional activity of NeuroD mediated by zPin1 .120 4.8 The expression of marker genes in zPin1 morphants 122 4.9 Pin1 as a novel regulator of bHLH family .125 4.10 Conclusion .126 References .128 Appendices 141 vii Summary The vertebrate pSer/pThr-Pro specific peptidyl-prolyl isomerase Pin1 has been shown to play important roles in cell cycle regulation, apoptosis, oncogenesis, and neuronal degeneration. However, its role in early neuronal development is not clear. With the use of zebrafish embryos, we examined zPin1’s effect on development. We showed that zebrafish Pin1 was expressed maternally and in a ubiquitous manner early in development, but by 48 hpf it was restricted to the brain and neuromasts. Coimmunoprecipitation assays (CoIP) in cell lines showed that zPin1 could interact with neuroD and ath1 but not ngn1. This binding was reduced when Ser/Thr phosphorylation sites were mutated. Antisense morpholino oligonucleotide (MO) knockdown of zPin1 led to delay in development. Accounting for the delay, neuroD expression was significantly diminished in the hindbrain of morphants by 48 hpf equivalent. Morphants in the background of Tol2/GFP enhancer trap lines with specific expressions in hair cells (ET4) and mantle cells (ET20) also displayed defects in neuromasts formation. It has been shown that specification of hair cells in neuromasts is neuroD dependent but ngn1 independent. Using siRNA Pin1 knockdown 293T cells and Pin1 knockout MEFs cells, we showed that neuroD protein was degraded more rapidly in the absence of Pin1. NeuroD stability was restored when Pin1 was over-expressed. This is the first study to demonstrate the functional regulation of a bHLH factor by cis-trans isomerization in neuronal specification. In view of the role of Pin1 in neurodegenative diseases, our results may have important pharmaco-therapeutic applications. viii References Metzner, M. et al. Functional replacement of the essential ESS1 in yeast by the plant parvulin DlPar13. J. Biol. Chem. 276(17): 13524-9 (2001). Miyachi, T. et al. Structure and regulation of the human NeuroD (BETA2/BHF1) gene. Brain Res. Mol. Brain Res. 69(2): 223-31 (1999). Miyata, T. et al. NeuroD is required for differentiation of the granule cells in the cerebellum and hippocampus. Genes Dev. 13(13): 1647-52 (1999). Moates, J. M. et al. BETA2 activates transcription from the upstream glucokinase gene promoter in islet beta-cells and gut endocrine cells. Diabetes 52(2): 403-8 (2003). Moens, C. B. and Prince, V. E. Constructing the hindbrain: insights from the zebrafish. Dev. Dyn. 224(1): 1-17 (2002). Moore, K. B. et al. Posttranslational mechanisms control the timing of bHLH function and regulate retinal cell fate. Neuron 34(2): 183-95 (2002). Nandi, D. et al. The ubiquitin-proteasome system. J. Biosci. 31(1): 137-55 (2006). Naya, F. J. et al. Diabetes, defective pancreatic morphogenesis, and abnormal enteroendocrine differentiation in BETA2/neuroD-deficient mice. Genes Dev. 11(18): 2323-34 (1997). Naya, F. J. et al. Tissue-specific regulation of the insulin gene by a novel basic helixloop-helix transcription factor. Genes Dev. 9(8): 1009-19 (1995). Nesbit, C. E. et al. MYC oncogenes and human neoplastic disease. Oncogene 18(19): 3004-16 (1999). Nigg, E. A. Mitotic kinases as regulators of cell division and its checkpoints. Nat. Rev. Mol. Cell Biol. 2(1): 21-32 (2001). Noll, M. Evolution and role of Pax genes. Curr. Opin. Genet. Dev. 3(4): 595-605 (1993). Norton, J. D. et al. Id helix-loop-helix proteins in cell growth and differentiation. Trends Cell Biol. 8(2): 58-65 (1998). Nunan, J. and Small, D. H. Proteolytic processing of the amyloid-beta protein precursor of Alzheimer's disease. Essays Biochem. 38: 37-49 (2002). Oates, A. C. et al. Too much interference: injection of double-stranded RNA has nonspecific effects in the zebrafish embryo. Dev. Biol. 224(1): 20-8 (2000). Oberst, A. et al. Regulation of the p73 protein stability and degradation. Biochem. Biophys. Res. Commun. 331(3): 707-12 (2005). Ogryzko, V. V et al. The transcriptional coactivators p300 and CBP are histone acetyltransferases. Cell 87(5): 953-9 (1996). 135 References Okamoto, K. and Sagata, N. Mechanism for inactivation of the mitotic inhibitory kinase Wee1 at M phase. Proc. Nat. Acad. Sci. U. S. A. 104(10): 3753-8 (2007). Osmani, A. H. et al. Parallel activation of the NIMA and p34cdc2 cell cycle-regulated protein kinases is required to initiate mitosis in A. nidulans. Cell 67(2): 283-91 (1991). Osmani, S. A. et al. Mitotic induction and maintenance by overexpression of a G2specific gene that encodes a potential protein kinase. Cell 53(2): 237-44 (1988). Pal, D. and Chakrabarti, P. Cis peptide bonds in proteins: residues involved, their conformations, interactions and locations. J. Mol. Biol. 294(1): 271-88 (1999). Parinov, S. et al. Tol2 transposon-mediated enhancer trap to identify developmentally regulated zebrafish genes in vivo. Dev. Dyn. 231(2): 449-59 (2004). Pastorino, L. et al. The prolyl isomerase Pin1 regulates amyloid precursor protein processing and amyloid-beta production. Nature 440(7083): 528-34 (2006). Pathan, N. et al. Microtubule-targeting drugs induce Bcl-2 phosphorylation and association with Pin1. Neoplasia 3(1): 70-9 (2001). Pawson, T. and Scott, J. D. Protein phosphorylation in signaling--50 years and counting. Trends Biochem. Sci. 30(6): 286-90 (2005). Petersen, H. V. et al. Glucose induced MAPK signalling influences NeuroD1mediated activation and nuclear localization. FEBS Lett. 528(1-3): 241-5 (2002). Pickart, C. M. Mechanisms underlying ubiquitination. Annu. Rev. Biochem. 70: 50333 (2001). Polakis, P. Wnt signaling and cancer. Genes Dev. 14(15): 1837-51 (2000). Pozzoli, O. et al. Xebf3 is a regulator of neuronal differentiation during primary neurogenesis in Xenopus. Dev. Biol. 233(2): 495-512 (2001). Preuss, U. and Mandelkow, E. M. Mitotic phosphorylation of tau protein in neuronal cell lines resembles phosphorylation in Alzheimer's disease. Eur. J. Cell Biol. 76(3): 176-84 (1998). Pu, R. T. and Osmani, S. A. Mitotic destruction of the cell cycle regulated NIMA protein kinase of Aspergillus nidulans is required for mitotic exit. EMBO J. 14(5): 995-1003 (1995). Putcha, G. V. et al. JNK-mediated BIM phosphorylation potentiates BAX-dependent apoptosis. Neuron 38(6): 899-914 (2003). Qiu, Y. et al. Acetylation of the BETA2 transcription factor by p300-associated factor is important in insulin gene expression. J. Biol. Chem. 279(11): 9796-802 (2004). 136 References Qiu, Y. et al. p300 mediates transcriptional stimulation by the basic helix-loop-helix activators of the insulin gene. Mol. Cell Biol. 18(5): 2957-64 (1998). Ramakrishnan, P. et al. Pin1 colocalization with phosphorylated tau in Alzheimer's disease and other tauopathies. Neurobiol. Dis. 14(2): 251-64 (2003). Ramelot, T. A. and Nicholson, L. K. Phosphorylation-induced structural changes in the amyloid precursor protein cytoplasmic tail detected by NMR. J Mol. Biol. 307(3): 871-84 (2001). Ranganathan, R. et al. Structural and functional analysis of the mitotic rotamase Pin1 suggests substrate recognition is phosphorylation dependent. Cell 89(6): 875-86 (1997). Ratineau, C. et al. Cyclin D1 represses the basic helix-loop-helix transcription factor, BETA2/NeuroD. J. Biol. Chem. 277(11): 8847-53 (2002). Reineke, E. L. et al. Degradation of the tumor suppressor PML by Pin1 contributes to the cancer phenotype of breast cancer MDA-MB-231 cells. Mol. Cell Biol. 28(3): 997-1006 (2008). Rippmann, J. F. et al. Phosphorylation-dependent proline isomerization catalyzed by Pin1 is essential for tumor cell survival and entry into mitosis. Cell Growth Differ. 11(7): 409-16 (2000). Roztocil, T. et al. NeuroM, a neural helix-loop-helix transcription factor, defines a new transition stage in neurogenesis. Development 124(17): 3263-72 (1997). Rubinstein, A. L. Zebrafish: from disease modeling to drug discovery. Curr. Opin. Drug Discov. Devel. 6(2): 218-23 (2003). Ryo, A. et al. A suppressive role of the prolyl isomerase Pin1 in cellular apoptosis mediated by the death-associated protein Daxx. J. Biol. Chem. 282(50): 36671-81 (2007). Ryo, A. et al. PIN1 is an E2F target gene essential for Neu/Ras-induced transformation of mammary epithelial cells. Mol. Cell Biol. 22(15): 5281-95 (2002). Ryo, A. et al. Pin1 regulates turnover and subcellular localization of beta-catenin by inhibiting its interaction with APC. Nat. Cell Biol. 3(9): 793-801 (2001). Ryo, A. et al. Regulation of NF-kappaB signaling by Pin1-dependent prolyl isomerization and ubiquitin-mediated proteolysis of p65/RelA. Mol. Cell 12(6): 141326 (2003). Saitoh, T. et al. Negative regulation of interferon-regulatory factor 3-dependent innate antiviral response by the prolyl isomerase Pin1. Nat. Immunol. 7(6): 598-605 (2006). Sampath, K. et al. Induction of the zebrafish ventral brain and floorplate requires cyclops/nodal signalling. Nature 395(6698): 185-9 (1998). 137 References Sarrazin, A. F. et al. Proneural gene requirement for hair cell differentiation in the zebrafish lateral line. Dev. Biol. 295(2): 534-45 (2006). Schwitzgebel, V. M. et al. Expression of neurogenin3 reveals an islet cell precursor population in the pancreas. Development 127(16): 3533-42 (2000). Sears, R. et al. Multiple Ras-dependent phosphorylation pathways regulate Myc protein stability. Genes Dev. 14(19): 2501-14 (2000). Seo, S. et al. Neurogenin and NeuroD direct transcriptional targets and their regulatory enhancers. EMBO J. 26(24): 5093-108 (2007). Seol, W. et al. An orphan nuclear hormone receptor that lacks a DNA binding domain and heterodimerizes with other receptors. Science 272(5266): 1336-9 (1996). Sharma, A. et al. The NeuroD1/BETA2 sequences essential for insulin gene transcription colocalize with those necessary for neurogenesis and p300/CREB binding protein binding. Mol. Cell Biol. 19(1): 704-13 (1999). Shen, M. et al. The essential mitotic peptidyl-prolyl isomerase Pin1 binds and regulates mitosis-specific phosphoproteins. Genes Dev. 12(5): 706-20 (1998). Song, A. et al. Phosphorylation of nuclear MyoD is required for its rapid degradation. Mol. Cell Biol. 18(9): 4994-9 (1998). Spillantini, M. G. and Goedert, M. Tau protein pathology in neurodegenerative diseases. Trends Neurosci. 21(10): 428-33 (1998). Stukenberg, P. T. and Kirschner, M. W. Pin1 acts catalytically to promote a conformational change in Cdc25. Mol. Cell 7(5): 1071-83 (2001). Suizu, F. et al. Pin1 regulates centrosome duplication, and its overexpression induces centrosome amplification, chromosome instability, and oncogenesis. Mol. Cell Biol. 26(4): 1463-79 (2006). Tessmar, K. et al. A screen for co-factors of Six3. Mech. Dev. 117(1-2): 103-13 (2002). Tintignac, L. A. et al. Cyclin E-cdk2 phosphorylation promotes late G1-phase degradation of MyoD in muscle cells. Exp. Cell Res. 259(1): 300-7 (2000). Turner, B. M. Decoding the nucleosome. Cell 75(1): 5-8 (1993). van Drogen, F. et al. Ubiquitylation of cyclin E requires the sequential function of SCF complexes containing distinct hCdc4 isoforms. Mol. Cell 23(1): 37-48 (2006). Vinals, F. et al. BMP-2 decreases Mash1 stability by increasing Id1 expression. EMBO J. 23(17): 3527-37 (2004). 138 References Vosper, J. M. et al. Regulation of neurogenin stability by ubiquitin-mediated proteolysis. Biochem. J. 407(2): 277-84 (2007). Weintraub, H. The MyoD family and myogenesis: redundancy, networks, and thresholds. Cell 75(7): 1241-4 (1993). Whitmarsh, A. J. and Davis, R. J. Transcription factor AP-1 regulation by mitogenactivated protein kinase signal transduction pathways. J. Mol. Med. 74(10): 589-607 (1996). Wintjens, R. et al. 1H NMR study on the binding of Pin1 Trp-Trp domain with phosphothreonine peptides. J. Biol. Chem. 276(27): 25150-6 (2001). Wu, X. et al. The Ess1 prolyl isomerase is linked to chromatin remodeling complexes and the general transcription machinery. EMBO J. 19(14): 3727-38 (2000). Wulf, G. et al. Phosphorylation-specific prolyl isomerization: is there an underlying theme? Nat. Cell Biol. 7(5): 435-41 (2005). Wulf, G. M. et al. Pin1 is overexpressed in breast cancer and cooperates with Ras signaling in increasing the transcriptional activity of c-Jun towards cyclin D1. EMBO J. 20(13): 3459-72 (2001). Xu, Y. X. and Manley, J. L. The prolyl isomerase Pin1 functions in mitotic chromosome condensation. Mol. Cell 26(2): 287-300 (2007). Yaffe, M. B. et al. Sequence-specific and phosphorylation-dependent proline isomerization: a potential mitotic regulatory mechanism. Science 278(5345): 1957-60 (1997). Yang, X. et al. Daxx, a novel Fas-binding protein that activates JNK and apoptosis. Cell 89(7): 1067-76 (1997). Yeh, E. et al. A signalling pathway controlling c-Myc degradation that impacts oncogenic transformation of human cells. Nat. Cell Biol. 6(4): 308-18 (2004). Yeh, E. S. et al. The loss of PIN1 deregulates cyclin E and sensitizes mouse embryo fibroblasts to genomic instability. J. Biol. Chem. 281(1): 241-51 (2006). Yeh, E. S. and Means, A. R. PIN1, the cell cycle and cancer. Nat. Rev. Cancer 7(5): 381-8 (2007). Yi, P. et al. Peptidyl-prolyl isomerase (Pin1) serves as a coactivator of steroid receptor by regulating the activity of phosphorylated steroid receptor coactivator (SRC-3/AIB1). Mol. Cell Biol. 25(21): 9687-99 (2005). Yu, L. et al. Regulation of Bruton tyrosine kinase by the peptidylprolyl isomerase Pin1. J. Biol. Chem. 281(26): 18201-7 (2006). 139 References Zacchi, P. et al. The prolyl isomerase Pin1 reveals a mechanism to control p53 functions after genotoxic insults. Nature 419(6909): 853-7 (2002). Zheng, H. et al. The prolyl isomerase Pin1 is a regulator of p53 in genotoxic response. Nature 419(6909): 849-53 (2002). Zhou, X. Z. et al. Pin1-dependent prolyl isomerization regulates dephosphorylation of Cdc25C and tau proteins. Mol. Cell 6(4): 873-83 (2000). Zhou, X. Z. et al. Phosphorylation-dependent prolyl isomerization: a novel signaling regulatory mechanism. Cell Mol. Life Sci. 56(9-10): 788-806 (1999). Zhu, J. X. et al. Identification and characterization of a novel and functional murine Pin1 isoform. Biochem. Biophys. Res. Commun. 359(3): 529-35 (2007). 140 Appendices Appendices Interaction between human Pin1 and zebrafish NeuroD It has been revealed that zebrafish Pin1 shares a high similarity (79%) with human Pin1. Moreover, we demonstrated that zPin1 could interact with NeuroD and this interaction was dependent on NeuroD phosphorylation. This result was the basis for all our thesis work. We indicated in the results and discussion part that rescue was unsuccessful on zebrafish embryos. Therefore, we had to perform rescue experiment on cell lines. To evaluate the feasibility of this assay, we used GST human Pin1 beads to pulldown cell lysates overexpressing zebrafish NeuroD. The result demonstrated that hPin1 could indeed associate with NeuroD (Fig. S.1). Therefore, it is plausible that in human HEK 293T cells, the absence of endogenous Pin1 can affect the stability of overexpressed NeuroD, which has been confirmed by our stability assay showing that loss of Pin1 can stabilize NeuroD. Furthermore, impaired stability of NeuroD can be restored by overexpressing zPin1 or hPin1. We could conclude that both hPin1 and zPin1 complement the reduced protein stability of NeuroD in 293T Pin1 knockdown cells. Rescue experiments were also demonstrated in MEFs cells. Indeed, in 1996, Lu et al. revealed that human Pin1 substituted the function of ESS1 in yeast. Our study demonstrated that zPin1 compensated the function mouse Pin1 and human Pin1 in mouse MEF cells and HEK 293T cells. These results provided a platform for studying general function of Pin1. 141 Appendices Detection of one zPin1 gene copy in zebrafish genome In Western blotting, we displayed that there seemed to be another isoform of zPin1 in muscle. However, with RACE experiment, we only retrieved one zPin1 sequence. To further confirm that there were no zPin1 isoforms in zebrafish, we performed Southern blotting experiment. We adopted several restriction enzymes (HindIII, EcoRI, XbaI, TaqI) to digest genome. Eventually, only one zPin1 gene copy was detected (Fig. S.2). A great number of problems occurred in this assay. Indeed, after being treated with enzymes, zebrafish genome should display a stripe of digested fragments (HindIII digested products, picture not shown). However, in our hands, one distinguished single non-cut band can be viewed on the gel after EcoRI and XbaI treatment, which means that they can not digest genome well. Therefore, one four-cutter enzyme (TaqI) was utilized and this enzyme can cut genome into small pieces. Finally two enzymes (HindIII and TaqI) worked and we detected only one zPin1 copy. However, one big problem was the dark background. Meanwhile, we did not place marker in this assay. Still, from the band position, we can identify that in TaqI digested genome, the band lies lower compared to that of HindIII digested genome, confirming the efficiency and specificity of Southern experiment. 142 S2 67 A S2 59 A S1 57 A WT WT Appendices neuroD GST-zPin1 GST pulldown GST only Input neuroD Figure S.1 Interaction between human Pin1 and zebrafish NeuroD. 293T cells were transfected with wild type NeuroD, S157A, S259A as well as S267A construct. Twenty four hours later, cell lysates were collected and subject to GST-hPin1 beads. After incubation for h at 4°C, beads were washed and analyzed by Western blotting. Anti-HA antibody was used to detect the presence of NeuroD. TaqI HindIII Figure S.2 Detection of gene copy numbers in zebrafish genome with Southern blotting. Zebrafish genomic DNA was extracted from adult fish. DNA was cut with HindIII and TaqI for hours for complete digestion. The digested DNA was probed with DIG-labeled Pin1 cDNA. One band was detected after Southern blotting. 143 Appendices Establishment of ET33-mi20 line Tol2 transopon-mediated enhancer trap (ET) technology has become a quite effective technology to establish mutant line in zebrafish, for its relatively easy insertion analysis, high mutation rate and efficiency (Kawakami et al., 1998; Kawakami et al., 1999; Koga et al., 2002). An ET construct contains the EGFP gene downstream of a promoter region from zebrafish keratin (krt8) gene (Gong et al., 2002). The Tol2 transposable element, which was first identified in medaka, contains a truncated transposase gene but still possesses the transposition activity. The ET construct was coinjected with Tol2 transposase mRNA for insertion of ET element into zebrafish genome (Kawakami et al., 2000). After screening, about 24 F1 were identified. One of them was called ET33, in which, a single Tol2 insertion in the 3’UTR of zic6 gene (Parinov et al., 2004). Because Tol2 insert could be made to jump to alternative positions simply by injecting transposase mRNA into ET lines, a few derivative lines were thus created in the hope that some of them will be insertional mutants. One of them, ET33-mi20, has Tol2 transposon cassette re-inserted into the first exon of neurogenin1 gene on ET33 background (Fig. S.3A). The line can be identified by the presence of abnormal posterior line ganglian at 32h (Fig. S.3B, C, D and E). Furthermore, in situ hybridization for ngn1 showed that in ET22-mi20, ngn1 transcripts were absent, confirming that ngn1 gen was indeed mutated (Fig. S.3F and G). This line was established by Igor Kondrychyn from Dr. Korzh’s lab in IMCB (unpublished paper). We used it in our experiment to demonstrate the specificity of our zPin1 morpholinos. 144 Appendices Figure S.3 Isolation of neurogenin mutant. ET33-mi20 line carries Tol2 insertion in the first exon of neurogenin gene. (A) Schematic representation of neurog1 structure. Transposon is inserted in the first exon coding 5’UTR. Direction of gfp transcription (green arrow) and sequence flanking the insertion site are shown. Start codon (ATG) is underlined, transposon 5’-end sequence is indicated in red and intron sequence (106 bp) is shown in small letters. Primers used for RT-PCR are shown as blue arrows. RT-PCR analysis of neurog1 transcript shows the absence of mRNA in mutant. Line 1: neurog1+/+, line 2: neurog1+/-, line 3: neurog1-/-. B-E: Confocal images of ET33-mi20 homozygous (B, D) and heterozygous (C, E) embryos. Arrow shows lateral line ganglion. F, G: Whole mount in situ hybridization with neurogenin RNA probe 145 Appendices Detection of early specification markers Before we focused the function of zPin1 on neuromasts hair cells formation, we characterized whether the specification of early cells fate was affected. We adopted gsc and ntl to stain embryos at hpf. The two markers were wellcharacterized and used extensively: gsc stains mesoderm shield area and ntl stains early axial mesoderm specifically (Schulte-Merker et al., 1994). Our result demonstrated that there was no difference for expression pattern of those two genes, which suggested that mesoderm development was not affected (Fig. S.4A, B, C and D). Another axial mesoderm marker shh, which was expressed in floor plate and notochord, was applied on 18 hpf embryos (Currie et al., 1996; Lawson et al., 2002). The staining for wild type and zPin1 morphant was similar, which further confirms that zPin1 did not impair early mesoderm specification. 146 Appendices Figure S.4 In situ hybridization of early specification markers. Wild type and zPin1 morphant were collected at hpf (A, B, C, D) or 18 hpf (E, F), respectively. The embryos collected at hpf were stained with gsc and ntl. Shh was used to stain the embryos at 18 hpf (E, F). (A, B) dorsal to the front. (C, D, E, F) dorsal to the right. 147 Appendices Endogenous NeuroD pulldown with GST-zPin1 beads We demonstrated that zPin1 can pulldown NeuroD. However, in all assays, we used cell lystaes overexpressing NeuroD. To confirm the relationship between zPin1 and NeuroD, we tried to pull down endogenous NeuroD wih zPin1. Firstly, several cell lines were tested for their endogenous NeuroD expression. Only SH-SY5Y cells express NeuroD, but only at a low level. With anti-NeuroD antibody we detected a single NeuroD protein at about 50 kDa. However, after pull down with GST-Pin1, there were several bands around this molecular weight. We initially thought these were phosphorylated NeuroD. These bands were excised and sent for Mass Spectrometer (MS) analysis. To our disappointment, none turned out to be NeuroD (Fig. S.5). The problem was that we can not identify which band was really the pulldown NeuroD band. It has been demonstrated previously that zPin1-NeuroD interaction was phosphorylation dependent. Therefore, it is highly possible that one of those bands that lied higher than endogenous NeuroD was indeed phosphorylated NeuroD. Due to time constraint and bad quality of NeuroD antibody, we did not continue this assay. In the future, it is likely that those bands could be confirmed by repeating the pulldown assay. 148 Appendices in1 T-z P GS To nl y GST pulldown GS SH ce -SY5 ll l ys Y a te s Input IB: neuroD Coomassie blue staining Figure S.5 GST-zPin1 pulldown of endogenous NeuroD. SH-SY5Y cell lysates were collected and assayed with NeuroD antibody. The cell lysates were then added into GST only beads and GST-zPin1 beads for pulldown assay. The tube was incubated at 4°C for hours. The beads were then washed and analyzed with Western blotting using NeuroD antibody. No band was detected after pulldown with GST beads, in contrast, several bands could be observed for GST-zPin1 pulldown at around 50 kDa. 149 Appendices References Currie, P. D. and Ingham, P. W. Induction of a specific muscle cell type by a hedgehog-like protein in zebrafish. Nature 382(6590): 452-5 (1996). Gong, Z. et al. Green fluorescent protein expression in germ-line transmitted transgenic zebrafish under a stratified epithelial promoter from keratin8. Dev. Dyn. 223(2): 204-15 (2002). Kawakami, K. et al. Excision of the tol2 transposable element of the medaka fish, Oryzias latipes, in zebrafish, Danio rerio. Gene 225(1-2): 17-22 (1998). Kawakami, K. and Shima, A. Identification of the Tol2 transposase of the medaka fish Oryzias latipes that catalyzes excision of a nonautonomous Tol2 element in zebrafish Danio rerio. Gene 240(1): 239-44 (1999). Kawakami, K. et al. Identification of a functional transposase of the Tol2 element, an Ac-like element from the Japanese medaka fish, and its transposition in the zebrafish germ lineage. Proc. Nat. Acad. Sci. U. S. A. 97(21): 11403-8 (2000). Koga, A. et al. Gene transfer and cloning of flanking chromosomal regions using the medaka fish Tol2 transposable element. Mar. Biotechnol. (NY) 4(1): 6-11 (2002). Lawson, N. D. et al. Sonic hedgehog and vascular endothelial growth factor act upstream of the Notch pathway during arterial endothelial differentiation. Dev. Cell 3(1): 127-36 (2002). Parinov, S. et al. Tol2 transposon-mediated enhancer trap to identify developmentally regulated zebrafish genes in vivo. Dev. Dyn. 231(2): 449-59 (2004). Schulte-Merker, S. et al. Expression of zebrafish goosecoid and no tail gene products in wild-type and mutant no tail embryos. Development 120(4): 843-52 (1994). 150 [...]... 67 Figure 3.4 In situ hybridization analysis of zpin1 expression at different stages 68 Figure 3.5 zPin1 interacts with NeuroD and Ath1 72 Figure 3.6 Identification of zPin1 potential Ser/Thr-Pro binding motifs on NeuroD 76 Figure 3.7 The reduction of zPin1 expression in zPin1 morphant embryos 79 Figure 3.8 Developmental delay in zPin1 morphant embryos 82 Figure 3.9 Neurogenin1 staining of the wild type... normal function of tau Therefore, Pin1 deregulation might act on multiple pathways to contribute to AD development (This figure was adapted from Lu et al., 2007) 15 Introduction 1.1.6 Pin1 function in development Although Pin1 s function has been studied extensively, little is known about function of Pin1 in development The characterized role of Pin1 in several model organisms has been presented in this... to become αAPP, Pin1 functions to convert cis conformation protein to trans one By doing so, Pin1 helps to avoid accumulation of too much cis APP protein as well as its toxic product (Pastorino et al., 2006) This kind of Pin1 function got further support from results that Aβ product was reduced in the case of Pin1 overexpression while increased Aβ secretion for depletion of Pin1 (Pastorino et al., 2006)... This study provides a novel finding regarding the role of Pin1 in activating the mitochondrial apoptosis machinery specifically in neuron cells An apoptosis model with Pin1 has been proposed (Becker et al., 2007) 1.1.5 Pin1 function in Alzheimer’s disease Expression level of Pin1 in neurons is higher than other tissues in mouse There is evidence to show that Pin1 expression is induced upon neuron differentiation... The binding of Pin1 with Emi1 stabilizes Emi1 and helps to 6 Introduction inhibit cyclin A and B proteins Therefore, cells can enter S and M phase smoothly (Bernis et al., 2007) Mitotic phosphorylated TopoIIα can also interact with Pin1 This binding localizes Pin1 on chromatin and promotes chromatin condensation Moreover, TopoIIα phosphorylation and binding ability to DNA are both increased due to Pin1. .. Model of cell cycle defects for PGCs 18 Figure 1.5 A spectrum of target activities by Pin1 isomerase 24 Figure 1.6 General interactions of bHLH proteins 26 Figure 1.7 Schematic of NeuroD protein 30 Figure 1.8 Composition of zebrafish lateral line 36 Figure 3.1 Full- length cDNA sequence of zebrafish Pin1 62 Figure 3.2 Amino acid sequence alignment of Pin1 in different species 64 Figure 3.3 zPin1 spatial... catch up zPin1 morphant embryos 83 Figure 3.10 Global apoptosis phenotype in zPin1 morphant embryos 84 Figure 3.11 Phospho-H3 staining of the wild type embryos and zPin1 morphant embryos 86 Figure 3.12 The neuronal defects in zPin1 morphant embryos 88 x Figure 3.13 Expression of marker genes in zPin1 morphant embryos 94 Figure 3.14 The impaired formation of posterior lateral line neuromasts in zPin1 morphant... 8 Introduction Detailed investigation shows that loss of Pin1 delays centrosome amplification; while overexpression promotes centrosome amplification (Suizu et al., 2006) These results indicate that Pin1 plays its role in S phase by affecting centrosome duplication without disturbing DNA synthesis 1.1.3.3 Pin1 function in oncogenesis Pin1 expression level is found to be increased in various kinds of. .. protein conformation, which may enhance dephosphorylation of Bcl-2, resulting in recovering to its non-phosphorylated state (Pathan et al., 2001; Basu et al., 2002) The apoptotic function of p53 is inhibited by one of the most conserved inhibitors iASPP iASPP binds to proline-rich domain of p53, thereby preventing binding of p53 to cell death-related promoters (Bergamaschi et al., 2006) Pin1 binds... untranslated region xiv Introduction Chapter 1 Introduction 1.1 The function of Pin1 1.1.1 The relationship between Pin1 and protein phosphorylation Protein phosphorylation is one of the most essential signaling modes for posttranslational modification The main effect of protein phosphorylation is to induce changes in protein conformation, which then further affects protein-protein interaction, subcellular . 1.1.3.3 Pin1 function in oncogenesis 9 1.1.4 Pin1 function in apoptosis 10 1.1.5 Pin1 function in Alzheimer’s disease 11 1.1.6 Pin1 function in development 16 1.1.7 Pin1 function on protein stability. between Pin1 and protein phosphorylation 1 1.1.2 The characteristics of Pin1 4 1.1.3 Pin1 function in cell cycle and cancer 5 1.1.3.1 Pin1 function in M phase 5 1.1.3.2 Pin1 function in G1. Molecular analysis of zebrafish Pin1 61 3.1.1 Characterization of zebrafish Pin1 61 3.1.2 Sequence alignment of Pin1 in various species 63 3.1.3 Expression pattern of zPin1 in zebrafish embryos

Ngày đăng: 12/09/2015, 09:55

Tài liệu cùng người dùng

  • Đang cập nhật ...

Tài liệu liên quan