Asymmetric cell division in the regulation of neural stem cell self renewel in drosophila melanogaster

150 284 0
Asymmetric cell division in the regulation of neural stem cell self renewel in drosophila melanogaster

Đang tải... (xem toàn văn)

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

Thông tin tài liệu

ASYMMETRIC CELL DIVISION IN THE REGULATION OF NEURAL STEM CELL SELFRENEWAL IN DROSOPHILA MELANOGASTER CHANG KAI CHEN B.Sc (Hons), NUS A THESIS SUBMITTED FOR THE DEGREE OF DOCTOR OF PHILOSOPHY TEMASEK LIFE SCIENCES LABORATORY NATIONAL UNIVERSITY OF SINGAPORE 2009 ACKNOWLEDGEMENTS I would like to express my heartfelt thanks to my supervisor Prof William Chia for his guidance, patience and support during the course of my PhD study I would also like to extend my sincere thanks and gratitude to a/P Wang Hongyan who went beyond her call of duty to supervise and guide me in the last year of my PhD study Her insightful suggestions and critical comments have been instrumental in shaping this work to its present form I am extremely grateful to Dr Greg Somers for his invaluable help and guidance for the initial part of the Zif and PP2A work Many thanks to Dr Rita Sousa-Nunes for giving me the zif mutant to work on, to Dr Wang Cheng for being a great collaborator for the PP2A project, and to my Ph.D advisory committee, A/P David Ng, A/P Sudipto Roy and a/P Toshie Kai for their suggestions during the committee meetings My sincere gratitude goes to Liu Ming for being a friend, and for his very kind assistance with microinjection for the generation of some of the zif transgenic lines I would also like to thank Dr Gisela Garcia-Alvarez for her contribution to the kinase assays in this work In addition, I would like to acknowledge Jacqueline Chin, Wong Jian Xiang and Soon Swee Beng for their technical assistance I also thank all the past and present members of Bill Chia lab and Wang Hongyan lab as well as the TLL fly community who have generously shared reagents with me at various stages of this work A very big thank you goes to dear friends like Shvetha, Vera, Maddy, Xiaodong, Ai Khim, Dawn, Kenneth, Kris and Charissa for their support during the trying times of my study I also thank my Toh Yi Caregroup for being understanding and supportive while I was busy with the writing of my thesis My most sincere thanks and gratitude go to my dearest friend Chin Fern, Coach Jo-Ann, Pastor Benjamin and Pauline for being my constant source of encouragement, support and wise counsel at various critical points of my PhD study Most importantly, I would like to extend my deepest thanks to my everso-supportive family - Dad, Mom, and my two sisters, Kai-Tirng and Kai Chirng I thank them for loving me the way they - for always being there to listen, to help, and to provide the emotional support that I need, especially during the toughest times I especially thank beloved Kai-Tirng for her love, patience, incessant support and prayers Without my loving family, I will not be where I am today Finally, my most heartfelt thanks and gratitude go to Mark Chong for faithfully supporting me in more ways than I can ask for I am deeply touched and extremely grateful for his love, patience, thoughtfulness and unyielding encouragement I thank him for the laughter and the joy that he has brought into my life, and for being the most faithful prayer warrior I’ve ever had Above all else, I am forever grateful to Jesus Christ, my Lord and Savior No amount of words can fully express my gratitude for His faithfulness, His unfailing love and His abundant grace toward me Kai Chen Dec2009 i TABLE OF CONTENTS ACKNOWLEDGEMENTS i TABLE OF CONTENTS ii ABBREVIATIONS vi LIST OF FIGURES AND TABLES ix SUMMARY xii CHAPTER 1: INTRODUCTION 1.1 Drosophila as a model system 1.2 Stem cell in development 1.3 Stem cells in Drosophila neurogenesis 1.4 Asymmetric cell division in Drosophila neural stem cell 1.4.1 Setting up neuroblast polarity 1.4.2 Mitotic spindle orientation 11 1.4.3 Asymmetric localization and segregation of cell fate determinants 13 1.4.3.1 Adaptor proteins required for asymmetric localization of cell fate determinants 16 1.5 Stem cells and cancer – the cancer stem cell hypothesis 20 1.6 Link between failure in stem cell asymmetric division and tumor formation 21 1.7 Drosophila Stem Cell self-renewal and Tumor suppression 24 1.7.1 Tumor growth induced by altered stem cell division 26 1.7.2 Mitotic Spindle orientation and tumor suppression 28 1.7.3 Tumor growth induced by impaired terminal differentiation 29 1.8 Cell cycle genes regulate asymmetric division and act as tumor suppressors 30 1.9 Protein phosphatases, asymmetric division and tumor suppression 33 ii 1.10 Objectives 36 CHAPTER 2: MATERIALS AND METHODS 38 Molecular Biology 38 2.1 2.1.1 Recombinant DNA methods 38 2.1.2 Bacterial host strains and growth conditions 38 2.1.3 Cloning strategies 39 2.1.4 Transformation of E coli cells 40 2.1.4.1 Preparation of competent cells for heatshock transformation 40 2.1.4.2 Heat shock transformation of E coli 40 2.1.4.3 Preparation of competent cells for electroporation 41 2.1.4.4 Electroporation transformation of E coli 41 2.1.5 Plasmid DNA preparations 42 2.1.6 Isolation of total genomic DNA from adult flies 42 2.1.7 Reverse Transcription (RT)-PCR 43 2.1.7.1 Isolation of total RNA 43 2.1.7.2 First strand cDNA synthesis 43 2.1.7.3 PCR reaction after RT 44 2.1.8 2.2 Site-directed mutagenesis 45 Cell Culture 45 2.2.1 Production of double-stranded RNA (dsRNA) 45 2.2.2 Cell culture, dsRNA and drug treatment 47 2.3 Biochemistry 47 2.3.1 Frequently used buffers and solutions 47 2.3.2 PAGE and Western transfer of protein samples 48 2.3.3 Immunological detection of proteins and antibodies used 49 2.3.4 Generation of anti-Zif polyclonal antibody 49 2.3.5 Fusion protein expression 50 2.3.6 Two-dimensional PAGE 51 iii 2.3.7 Chromatin Immunoprecipitation (ChIP) Assay 51 2.3.8 Luciferase Assay 54 2.3.9 In vitro Kinase Assay 54 2.4 Immunohistochemistry and microscopy 55 2.4.1 Frequently used reagents and buffers 55 2.4.2 Antibodies 55 2.4.3 Fixing and staining of Drosophila larval brains 56 2.4.4 Neuroblast quantification and brain orientation 57 2.4.5 BrdU labeling 57 2.4.6 Spindle orientation quantification 57 2.5 Fly Genetics 58 2.5.1 Fly stocks and growth conditions used in this study 58 2.5.2 Generation of positively labeled neuroblast MARCM clones 59 CHAPTER 3: A NOVEL ZINC FINGER PROTEIN NEGATIVELY REGULATES aPKC EXPRESSION TO INHIBIT EXCESS SELF-RENEWAL OF DROSOPHILA NEURAL STEM CELLS 60 3.1 Introduction 60 3.2 Results 62 3.2.1 Identification of a novel zinc-finger protein with a role in regulating neuroblast asymmetry 62 3.2.2 Disruption of zif leads to excess neuroblasts in larval brain clones 64 3.2.3 Subcellular localization of endogenous Zif 68 3.2.4 Zif transgene fully rescues defects in zif mutant neuroblasts 70 3.2.5 Asymmetric localization of aPKC and Numb/Pon requires Zif function70 3.2.6 Zif represses aPKC transcription and downregulates aPKC protein expression 76 3.2.7 Zif directly represses aPKC transcription to inhibit excess neuroblast self-renewal 78 iv 3.2.8 aPKC phosphorylates Zif 82 3.2.9 Nuclear localization of Zif depends on its phosphorylation state 84 3.2.10 Non-phosphorylatable form of Zif inhibits excess neuroblast selfrenewal 3.3 89 Discussion 91 3.3.1 Role of Zif in neuroblast self-renewal and neuroblast asymmetry 91 3.3.2 Zif is the first identified transcription factor to regulate neuroblast self-renewal through direct transcriptional repression of aPKC 93 3.3.3 aPKC phosphorylates Zif to regulate its nuclear localization, thereby modulating activity of Zif as a transcriptional repressor of aPKC 94 3.3.4 Reciprocal repression between aPKC and Zif in neuroblast asymmetric division and neuroblast self-renewal 96 CHAPTER 4: PROTEIN PHOSPHATASE 2A REGULATES SELFRENEWAL OF DROSOPHILA NEURAL STEM CELLS 98 4.1 Introduction 98 4.2 Results 98 4.2.1 Microtubule Star (Mts) is a novel brain tumor suppressor in Drosophila 99 4.2.2 PP2A can inhibit excess self-renewal of neuroblasts 102 4.2.3 PP2A regulates asymmetric protein localizations as well as mitotic spindle orientation 107 4.2.4 PP2A and Polo enhance Numb phosphorylation and asymmetric localization 110 4.2.5 PP2A acts in Polo/Numb pathway to inhibit neuroblast overgrowth 117 Discussion 119 CHAPTER 5: CONCLUSION AND PERSPECTIVES 122 REFERENCES 128 v ABBREVIATIONS aa a/P A/P aPKC APS Ase ATP Aur-A Baz bp Brat BSA C elegans amino acid Assistant Professor Associate Professor Atypical protein kinase C ammonium persulphate Asense Adenosine 5’ Triphosphate Aurora-A Bazooka basepairs Brain tumor Bovine serum albumin Caenorhabditis elegans CaCl2 cDNA ChIP CIP CNN CNS CNS Cy3 Cyc DEPC DGRC Dlg DM DM DNA dNTP Dpn DSHB dsRNA DTT E coli ECL EDTA Calcium chloride complementary DNA chromatin immunoprecipitation calf intestinal phosphatase Centrosomin Central nervous system Central nervous system Cyanine conjugated cyclin Diethyl Pyrocarbonate Drosophila Genomics Resource Center Disc large dorsomedial dorsomedial Deoxyribonucleic acid deoxynucleotide triphosphate Deadpan Developmental Studies Hybridoma Bank double-stranded ribonucleic acid 1, 4-Dithio-DL-threitol Escherichia coli Enhanced Chemiluminescence Ethylenediaminetetraacetic acid Ethylene glycol-bis(2-aminoethylether)-N,N,N’,N’tetraacetic acid Embryonic Lethal Abnormal Vision ethlymethane sulfonate Falafel FLP recombinase recombination target grams guanine-nucleotide-dissociation inhibitor Green fluorescent protein Ganglion mother cell EGTA ELAV EMS Flfl FRT g GDI GFP GMC vi GST HCl hr IEF IgG Insc IPG IPTG Kb KCl Khc73 Kv L3 LB LiCl Loco M Glutathione-S-Transferase Hydrochloric acid hour Isoelectric focusing Immunoglobulin Inscuteable immobilized pH gradient Isopropyl-β-thiogalactopyranoside Kilobase potassium chloride kinesin heavy chain 73 Kilovolts third-instar larval Luria bertani Lithium chloride Locomotion defects Molar MgCl2 mins Mira ml mM MOPs MTOC Mts Mud NB Magnesium chloride minutes Miranda millilitre millimolar 4-Morpholinepropanesulphonic acid microtubule organizing center Microtubule star Mushroom body defective Neuroblast N-terminal OD PAGE PAN Par PBS PCM PCR PH3 Pins PON PP2A Pp4 Pros RNA RNAi RT RT-PCR S SDS secs Amino (NH2) terminal optical density Polyacrylamide gel electrophoresis Posterior Asense-Negative partitioning defective Phosphate Buffered Saline Pericentriolar material Polymerase Chain Reaction phospho-histone H3 Partner of Inscuteable Partner of Numb Protein phosphatase 2A Protein phosphatase Prospero Ribonucleic acid RNA interference room temperature real-time polymerase chain reaction Serine Sodium Dodecyl Sulphate seconds vii SOP stau TE TEMED Tris Tws UAS V Wdb β-Gal μg μl μM 2-D PAGE Sensory organ precursor staufen Tris EDTA N, N, N’, N’ tetramethylethylene diamine Tris (hydroxymethyl) aminomethane Twins Upstream Activator Sequence Volts Widerborst β-Galactosidase microgram microlitre micromolar two-dimensional polyacrylamide gel electrophoresis viii List of Figures and Table LIST OF FIGURES AND TABLES FIGURES Chapter PAGE Figure Life cycle of Drosophila melanogaster Figure Regulation of stem cell division Figure Asymmetric neural stem cell division in Drosophila embryo Figure Postembryonic neuroblast development Figure Key players in neuroblast asymmetric division Figure Overgrowth of mutant brain tissues implanted into adult 23 hosts Figure Models of the origin of Drosophila stem-cell derived 25 tumors Figure Proposed mechanism where activation of Aur-A leads 32 to the phosphorylation of Numb Protein phosphatase 2A is a heterotrimeric complex 34 Figure 10 Mosaic Analysis with a Repressible Cell Marker 61 Figure 11 Disruption of a novel gene causes ectopic Miranda 62 Figure Chapter persistence Figure 12 Schematic of zif locus and depiction of molecular 1L15 lesions in zif , zif 2L745 and zif 63 2L497 Figure 13 Zif inhibits excess neuroblast self-renewal 65 Figure 14 Zif acts to suppress neuroblast self-renewal and 66 promote neuronal differentiation Figure 15 Zif inhibits excess neuroblast self-renewal in both Ase- 67 positive non-DM and Ase-negative DM neuroblast lineages Figure 16 Polyclonal antibody against full-length Zif specifically 68 recognizes Zif Figure 17 Subcellular localization of Zif in third instar larval brain 69 recapitulated by an inducible Zif transgene ix Conclusion and Perspectives CHAPTER CONCLUSION AND PERSPECTIVES For many years, Drosophila neuroblasts have proved to be a remarkably fruitful model system for investigating stem cell symmetry breaking The study of stem cell asymmetric division in Drosophila embryonic neuroblasts has brought about a period of rapid progress in identifying many central players in asymmetric cell divisions In the recent years, Drosophila larval brain neuroblasts have also emerged as a novel model for the study of stem cell self-renewal and tumor suppression as a growing body of evidence strongly suggest that defects in asymmetric cell division can upset homeostasis of stem cell self-renewal causing neural stem cells to turn into tumor-initiating cells that recapitulate several hallmarks typical of mammalian tumors The advent of new methods for self-renewal assays such as MARCM (Lee and Luo 1999), made it possible to generate single mutant neuroblast clones that lack a particular gene in a heterozygous (phenotypically) wild-type background Such mosaic clonal analyses made it easier to ascertain the effects of specific gene mutation on neuroblast self-renewal by comparing stem cell numbers with and without the activity of candidate self-renewal regulators In fact, many of the widely conserved cellular polarity proteins (e.g lgl and dlg) known to regulate asymmetric cell division were discovered to act as tumor suppressors during neuroblast self-renewal through such new techniques Consistent with the link between asymmetric cell division and tumor formation, molecular genetic data from the study of Drosophila neural stem 122 Conclusion and Perspectives cell together with results from mammalian studies provided evidence to show that mechanisms regulating stem cell self-renewal and tumor suppression via asymmetric cell division are evolutionarily conserved These studies show that breakdown of asymmetry in dividing Drosophila neuroblast stem cells leads to symmetric, proliferative divisions and hence impaired differentiation (Rolls, Albertson et al 2003; Caussinus and Gonzalez 2005)l(Lee, Robinson et al 2006) In addition, impaired basal targeting and defective cell fate determination during asymmetric neuroblast division in Drosophila lead to the formation of malignant neoplasm due to excessive numbers of overproliferating mutant progenitor cells (Caussinus and Gonzalez 2005; Bello, Reichert et al 2006; Betschinger, Mechtler et al 2006; Lee, Wilkinson et al 2006) Interestingly, it has been observed that only a small subset of the cells in a human tumor when transplanted into immuno-compromised mice can reinitiate tumor formation Data from these recent studies also demonstrated that these so-called ‘cancer stem-cells’ cells may cause leukemia, and solid tumors of the breast and brain (Lapidot, Sirard et al 1994; Bonnet and Dick 1997; Al-Hajj, Wicha et al 2003; Singh, Clarke et al 2003; Al-Hajj and Clarke 2004) lending further support to the notion that failure of the otherwise tightly regulated self-renewing capacities of either stem cells or progenitor cells can result in neoplasm Though it is true that more advanced tumors usually lack polarity, it is unclear at present whether there is a direct causal link between loss of cell polarity and tumor initiation in humans Nonetheless, data from Drosophila clearly demonstrate that impaired asymmetric cell division, and in turn errors in the process of normal differentiation can be initiating events in the formation of malignant tumors 123 Conclusion and Perspectives Despite tremendous progress in the field, mechanistic understanding of how these regulators of asymmetric cell division exert their function in the self-renewal of neuroblasts remain elusive Results from my Ph.D work on the two novel players in asymmetric cell division of Drosophila neuroblasts certainly reveal new perspectives to our understanding of the underlying mechanisms that controls stem cell self-renewal, progenitor differentiation, and shed light on the link between tumor suppression and asymmetric cell division Chapter outlines a novel zinc-finger protein, Zif, which suppresses excess neuroblast self-renewal primarily through modulating aPKC expression and/or localization When Zif function is compromised, not only is neuroblast polarity disrupted, aPKC transcript and protein levels are also upregulated Since aPKC is a potent positive regulator that enhances neuroblast self-renewal, it is no surprise that zif mutant exhibit neuroblast overgrowth phenotype In fact, recent data implicate the PAR-aPKC complex in human carcinogenesis Gene amplification and elevated constitutive activity of PKC-ι, one of two human aPKC homologs, was detected in ovarian, lung and colon cancer, suggesting the PKC-ι may be an oncogene (Murray, Jamieson et al 2004; Eder, Sui et al 2005; Regala, Weems et al 2005; Regala, Weems et al 2005) Not unexpectedly, tumors with elevated levels of aPKC had lost epithelial polarity, which is consistent with the overexpression phenotype of a constitutively active form of aPKC in Drosophila epithelia (Eder, Sui et al 2005; Lee, Robinson et al 2006) Using ChIP and Luciferase assays, I also showed that Zif is able to negatively regulate aPKC expression through direct transcriptional repression To the best of our knowledge, this is the first time that a transcription factor is reported to inhibit self-renewing capacity of neuroblasts in the Drosophila larval brain by direct transcriptional regulation of aPKC 124 Conclusion and Perspectives levels It has been shown previously that neuroblasts express a temporal series of transcription factors during embryonic and larval stages (Isshiki, Pearson et al 2001; Maurange, Cheng et al 2008) During pupal stages neuroblasts shrink and eventually undergo symmetric division leading to terminal differentiation However, when certain larval transcription factors of the neuroblast clock are missing (e.g seven up), this does not happen, and neuroblasts continue to proliferate into adulthood It could be that defects in asymmetric cell division perturb the neuroblast clock, causing stem cells to become tumorous It will be interesting to find out if Zif or its targets, possibly genes that are involved in both asymmetric cell division and stem cell selfrenewal, are regulated in a temporal manner Given the importance of aPKC as a central component of the polarity machinery and its vital role as a positive regulator of neuroblast self-renewal, it is with pleasant surprise that I further discovered that aPKC is able to phosphorylate Zif directly More intriguingly, this phosphorylation of Zif by aPKC is able to inhibit nuclear localization of Zif As a transcriptional repressor, exclusion from the nucleus would effectively inactivate Zif, making it non-functional in the modulation of aPKC expression, and hence its role in the regulation of neuroblast self-renewal Hence, the proposed reciprocal interaction between aPKC and Zif in the regulation of neuroblast asymmetric division and neuroblast self-renewal adds a new mechanism to the complex framework of regulatory pathways that orchestrates robust aPKC polarity for its normal function It will be interesting to investigate whether this reciprocal interaction between aPKC and Zif is conserved in other systems (e.g epithelial cells, ovarian follicle cells and photoreceptor cells) where aPKC also plays a pivotal role in establishing polarity Taking a more global view, a genome wide microarray analysis combined with a biochemical search for common binding 125 Conclusion and Perspectives partners of Zif and aPKC may shed light on the regulatory mechanisms that modulate this reciprocal interaction Chapter elaborates on a novel role of the serine/threoninephosphatase PP2A in the control of neuroblast self-renewal Though mammalian PP2A has been shown to participate in malignant transformation by regulating multiple pathways (Li, Scuderi et al 2002; Westermarck and Hahn 2008) as a tumor suppressor, this is the first time that PP2A is demonstrated to be involved in the link between asymmetric cell division and the regulation of neural stem cell homeostasis To limit neuroblast self-renewal, PP2A is not just involved in the regulation of asymmetric protein localization but also plays a role in mitotic spindle orientation – both are key aspects of proper asymmetric cell division (outlined in Chapter Introduction) We also discovered that when PP2A function is compromised, aPKC protein levels increase and Numb hyperphosphorylation is dramatically reduced These PP2A-dependent defects in asymmetric division of mitotic neuroblast resemble that of polo loss-of-function mutants Polo is a kinase that phosphorylates and thereby activates the cell fate determinant Numb Prompted to investigate the possible link between Polo and PP2A, we first discover that in polo loss-of-function mutant, hyperphosphorylation of Numb is also dramatically reduced Consistent with our prediction, we subsequently discovered that indeed, in the absence of PP2A, both Polo protein and transcript levels are dramatically reduced These results suggest that PP2A regulates Numb activity by promoting Polo expression Based on these and previous findings, we proposed a novel pathway in which PP2A acts upstream of Polo and Numb to block excessive neuroblast self-renewal It will be interesting to identify potential substrates of PP2A (possibly novel transcription factors) that are able to promote polo expression and control 126 Conclusion and Perspectives neural stem cell self-renewal Given the structural diversity of PP2A, and its involvement in a myriad of cellular and developmental processes, it is of utmost interest to find out whether this function of PP2A in neural stem cell self-renewal is conserved in other stem cell systems 127 References REFERENCES Albertson, R., and C.Q Doe 2003 Dlg, Scrib and Lgl regulate neuroblast cell size and mitotic spindle asymmetry Nat Cell Biol 5:166-70 Androutsellis-Theotokis, A., R.R Leker, F Soldner, D.J Hoeppner, R Ravin, S.W Poser, M.A Rueger, S.K Bae, R Kittappa, and R.D McKay 2006 Notch signalling regulates stem cell numbers in vitro and in vivo Nature Atwood, S.X., C Chabu, R.R Penkert, C.Q Doe, and K.E Prehoda 2007 Cdc42 acts downstream of Bazooka to regulate neuroblast polarity through Par-6 aPKC J Cell Sci 120:3200-6 Atwood, S.X., and K.E Prehoda 2009 aPKC phosphorylates Miranda to polarize fate determinants during neuroblast asymmetric cell division Curr Biol 19:723-9 Barros, C.S., C.B Phelps, and A.H Brand 2003 Drosophila nonmuscle myosin II promotes the asymmetric segregation of cell fate determinants by cortical exclusion rather than active transport Dev Cell 5:829-40 Beaucher, M., E Hersperger, A Page-McCaw, and A Shearn 2007 Metastatic ability of Drosophila tumors depends on MMP activity Dev Biol 303:625-34 Bello, B., H Reichert, and F Hirth 2006 The brain tumor gene negatively regulates neural progenitor cell proliferation in the larval central brain of Drosophila Development 133:2639-48 Bello, B.C., N Izergina, E Caussinus, and H Reichert 2008 Amplification of neural stem cell proliferation by intermediate progenitor cells in Drosophila brain development Neural Develop 3:5 Berdnik, D., and J.A Knoblich 2002 Drosophila Aurora-A is required for centrosome maturation and actin-dependent asymmetric protein localization during mitosis Curr Biol 12:640-7 Berdnik, D., T Torok, M Gonzalez-Gaitan, and J.A Knoblich 2002 The endocytic protein alpha-Adaptin is required for numb-mediated asymmetric cell division in Drosophila Dev Cell 3:221-31 Betschinger, J., F Eisenhaber, and J.A Knoblich 2005 Phosphorylationinduced autoinhibition regulates the cytoskeletal protein Lethal (2) giant larvae Curr Biol 15:276-82 Betschinger, J., and J.A Knoblich 2004 Dare to be different: asymmetric cell division in Drosophila, C elegans and vertebrates Curr Biol 14:R674-85 Betschinger, J., K Mechtler, and J.A Knoblich 2003 The Par complex directs asymmetric cell division by phosphorylating the cytoskeletal protein Lgl Nature 422:326-30 128 References Betschinger, J., K Mechtler, and J.A Knoblich 2006 Asymmetric segregation of the tumor suppressor brat regulates self-renewal in Drosophila neural stem cells Cell 124:1241-53 Boone, J.Q., and C.Q Doe 2008 Identification of Drosophila type II neuroblast lineages containing transit amplifying ganglion mother cells Dev Neurobiol 68:1185-95 Bossing, T., G Udolph, C.Q Doe, and G.M Technau 1996 The embryonic central nervous system lineages of Drosophila melanogaster I Neuroblast lineages derived from the ventral half of the neuroectoderm Dev Biol 179:4164 Bowman, S.K., R.A Neumuller, M Novatchkova, Q Du, and J.A Knoblich 2006 The Drosophila NuMA Homolog Mud regulates spindle orientation in asymmetric cell division Dev Cell 10:731-42 Bowman, S.K., V Rolland, J Betschinger, K.A Kinsey, G Emery, and J.A Knoblich 2008 The tumor suppressors Brat and Numb regulate transitamplifying neuroblast lineages in Drosophila Dev Cell 14:535-46 Bray, S.J 2006 Notch signalling: a simple pathway becomes complex Nat Rev Mol Cell Biol 7:678-89 Broadus, J., and C.Q Doe 1997 Extrinsic cues, intrinsic cues and microfilaments regulate asymmetric protein localization in Drosophila neuroblasts Curr Biol 7:827-35 Buescher, M., S.L Yeo, G Udolph, M Zavortink, X Yang, G Tear, and W Chia 1998 Binary sibling neuronal cell fate decisions in the Drosophila embryonic central nervous system are nonstochastic and require inscuteablemediated asymmetry of ganglion mother cells Genes Dev 12:1858-70 Campos-Ortega, J A., Hartenstein, V (1997) "The embryonic development of Drosophila melanogaster." Springer, Berlin Heidelberg New York Carmena, M., M.G Riparbelli, G Minestrini, A.M Tavares, R Adams, G Callaini, and D.M Glover 1998 Drosophila polo kinase is required for cytokinesis J Cell Biol 143:659-71 Castellanos, E., P Dominguez, and C Gonzalez 2008 Centrosome dysfunction in Drosophila neural stem cells causes tumors that are not due to genome instability Curr Biol 18:1209-14 Caussinus, E., and C Gonzalez 2005 Induction of tumor growth by altered stem-cell asymmetric division in Drosophila melanogaster Nat Genet 37:1125-9 Chia, W., W.G Somers, and H Wang 2008 Drosophila neuroblast asymmetric divisions: cell cycle regulators, asymmetric protein localization, and tumorigenesis J Cell Biol 180:267-72 Chen, F., V Archambault, A Kar, P Lio, P.P D'Avino, R Sinka, K Lilley, E.D Laue, P Deak, L Capalbo, and D.M Glover 2007 Multiple protein phosphatases are required for mitosis in Drosophila Curr Biol 17:293-303 129 References Choksi, S.P., T.D Southall, T Bossing, K Edoff, E de Wit, B.E Fischer, B van Steensel, G Micklem, and A.H Brand 2006 Prospero acts as a binary switch between self-renewal and differentiation in Drosophila neural stem cells Dev Cell 11:775-89 Clarke, M.F., and M Fuller 2006 Stem cells and cancer: two faces of eve Cell 124:1111-5 Deak, P., M Donaldson, and D.M Glover 2003 Mutations in makos, a Drosophila gene encoding the Cdc27 subunit of the anaphase promoting complex, enhance centrosomal defects in polo and are suppressed by mutations in twins/aar, which encodes a regulatory subunit of PP2A J Cell Sci 116:4147-58 Dietzl, G., D Chen, F Schnorrer, K.C Su, Y Barinova, M Fellner, B Gasser, K Kinsey, S Oppel, S Scheiblauer, A Couto, V Marra, K Keleman, and B.J Dickson 2007 A genome-wide transgenic RNAi library for conditional gene inactivation in Drosophila Nature 448:151-6 Doxsey, S., D McCollum, and W Theurkauf 2005 Centrosomes in cellular regulation Annu Rev Cell Dev Biol 21:411-34 Eichhorn, P.J., M.P Creyghton, and R Bernards 2009 Protein phosphatase 2A regulatory subunits and cancer Biochim Biophys Acta 1795:1-15 Encalada, S.E., P.R Martin, J.B Phillips, R Lyczak, D.R Hamill, K.A Swan, and B Bowerman 2000 DNA replication defects delay cell division and disrupt cell polarity in early Caenorhabditis elegans embryos Dev Biol 228:225-38 Ferres-Marco, D., I Gutierrez-Garcia, D.M Vallejo, J Bolivar, F.J GutierrezAvino, and M Dominguez 2006 Epigenetic silencers and Notch collaborate to promote malignant tumours by Rb silencing Nature 439:430-6 Frank, D.J., B.A Edgar, and M.B Roth 2002 The Drosophila melanogaster gene brain tumor negatively regulates cell growth and ribosomal RNA synthesis Development 129:399-407 Fuerstenberg, S., C.Y Peng, P Alvarez-Ortiz, T Hor, and C.Q Doe 1998 Identification of Miranda protein domains regulating asymmetric cortical localization, cargo binding, and cortical release Mol Cell Neurosci 12:325-39 Fuller, M.T., and A.C Spradling 2007 Male and female Drosophila germline stem cells: two versions of immortality Science 316:402-4 Fuse, N., K Hisata, A.L Katzen, and F Matsuzaki 2003 Heterotrimeric G proteins regulate daughter cell size asymmetry in Drosophila neuroblast divisions Curr Biol 13:947-54 Gateff, E., Schneidermann, H.A 1967 Developmental studies of a new mutant of Drosophila melanogaster lethal malignant brain tumor l(2)gl4 Am Zool 7:760 130 References Glover, D.M., M.H Leibowitz, D.A McLean, and H Parry 1995 Mutations in aurora prevent centrosome separation leading to the formation of monopolar spindles Cell 81:95-105 Go, M.J., D.S Eastman, and S Artavanis-Tsakonas 1998 Cell proliferation control by Notch signaling in Drosophila development Development 125:2031-40 Goldstein, B., and I.G Macara 2007 The PAR proteins: fundamental players in animal cell polarization Dev Cell 13:609-22 Hannus, M., F Feiguin, C.P Heisenberg, and S Eaton 2002 Planar cell polarization requires Widerborst, a B' regulatory subunit of protein phosphatase 2A Development 129:3493-503 Ikeshima-Kataoka, H., J.B Skeath, Y Nabeshima, C.Q Doe, and F Matsuzaki 1997 Miranda directs Prospero to a daughter cell during Drosophila asymmetric divisions Nature 390:625-9 Ito, K., and Y Hotta 1992 Proliferation pattern of postembryonic neuroblasts in the brain of Drosophila melanogaster Dev Biol 149:134-48 Izumi, Y., N Ohta, K Hisata, T Raabe, and F Matsuzaki 2006 Drosophila Pins-binding protein Mud regulates spindle-polarity coupling and centrosome organization Nat Cell Biol 8:586-93 Janssens, V., J Goris, and C Van Hoof 2005 PP2A: the expected tumor suppressor Curr Opin Genet Dev 15:34-41 Januschke, J., and C Gonzalez 2008 Drosophila asymmetric division, polarity and cancer Oncogene 27:6994-7002 Junttila, M.R., P Puustinen, M Niemela, R Ahola, H Arnold, T Bottzauw, R Ala-aho, C Nielsen, J Ivaska, Y Taya, S.L Lu, S Lin, E.K Chan, X.J Wang, R Grenman, J Kast, T Kallunki, R Sears, V.M Kahari, and J Westermarck 2007 CIP2A inhibits PP2A in human malignancies Cell 130:51-62 Kaltschmidt, J.A., C.M Davidson, N.H Brown, and A.H Brand 2000 Rotation and asymmetry of the mitotic spindle direct asymmetric cell division in the developing central nervous system Nat Cell Biol 2:7-12 Kim, J.K., H.W Gabel, R.S Kamath, M Tewari, A Pasquinelli, J.F Rual, S Kennedy, M Dybbs, N Bertin, J.M Kaplan, M Vidal, and G Ruvkun 2005 Functional genomic analysis of RNA interference in C elegans Science 308:1164-7 Knoblich, J.A 2001 Asymmetric cell division during animal development Nat Rev Mol Cell Biol 2:11-20 Knoblich, J.A., L.Y Jan, and Y.N Jan 1995 Asymmetric segregation of Numb and Prospero during cell division Nature 377:624-7 131 References Krahn, M.P., D Egger-Adam, and A Wodarz 2009 PP2A antagonizes phosphorylation of Bazooka by PAR-1 to control apical-basal polarity in dividing embryonic neuroblasts Dev Cell 16:901-8 Kraut, R., W Chia, L.Y Jan, Y.N Jan, and J.A Knoblich 1996 Role of inscuteable in orienting asymmetric cell divisions in Drosophila Nature 383:50-5 Landy, A 1989 Dynamic, structural, and regulatory aspects of lambda sitespecific recombination Annu Rev Biochem 58:913-49 Le Borgne, R., A Bardin, and F Schweisguth 2005 The roles of receptor and ligand endocytosis in regulating Notch signaling Development 132:1751-62 Lee, C.Y., R.O Andersen, C Cabernard, L Manning, K.D Tran, M.J Lanskey, A Bashirullah, and C.Q Doe 2006a Drosophila Aurora-A kinase inhibits neuroblast self-renewal by regulating aPKC/Numb cortical polarity and spindle orientation Genes Dev 20:3464-74 Lee, C.Y., K.J Robinson, and C.Q Doe 2006b Lgl, Pins and aPKC regulate neuroblast self-renewal versus differentiation Nature 439:594-8 Lee, C.Y., B.D Wilkinson, S.E Siegrist, R.P Wharton, and C.Q Doe 2006c Brat Is a Miranda Cargo Protein that Promotes Neuronal Differentiation and Inhibits Neuroblast Self-Renewal Dev Cell 10:441-9 Lee, T., and L Luo 1999 Mosaic analysis with a repressible cell marker for studies of gene function in neuronal morphogenesis Neuron 22:451-61 Li, L., and H Vaessin 2000 Pan-neural Prospero terminates cell proliferation during Drosophila neurogenesis Genes Dev 14:147-51 Li, L., and T Xie 2005 Stem cell niche: structure and function Annu Rev Cell Dev Biol 21:605-31 Li, X., A Scuderi, A Letsou, and D.M Virshup 2002 B56-associated protein phosphatase 2A is required for survival and protects from apoptosis in Drosophila melanogaster Mol Cell Biol 22:3674-84 Llamazares, S., A Moreira, A Tavares, C Girdham, B.A Spruce, C Gonzalez, R.E Karess, D.M Glover, and C.E Sunkel 1991 polo encodes a protein kinase homolog required for mitosis in Drosophila Genes Dev 5:2153-65 Lu, B., M Rothenberg, L.Y Jan, and Y.N Jan 1998 Partner of Numb colocalizes with Numb during mitosis and directs Numb asymmetric localization in Drosophila neural and muscle progenitors Cell 95:225-35 Maurange, C., and A.P Gould 2005 Brainy but not too brainy: starting and stopping neuroblast divisions in Drosophila Trends Neurosci 28:30-6 Mayer-Jaekel, R.E., S Baumgartner, G Bilbe, H Ohkura, D.M Glover, and B.A Hemmings 1992 Molecular cloning and developmental expression of the catalytic and 65-kDa regulatory subunits of protein phosphatase 2A in Drosophila Mol Biol Cell 3:287-98 132 References Micchelli, C.A., and N Perrimon 2006 Evidence that stem cells reside in the adult Drosophila midgut epithelium Nature 439:475-9 Newton, A.C 2001 Protein kinase C: structural and spatial regulation by phosphorylation, cofactors, and macromolecular interactions Chem Rev 101:2353-64 Nipper, R.W., K.H Siller, N.R Smith, C.Q Doe, and K.E Prehoda 2007 Galphai generates multiple Pins activation states to link cortical polarity and spindle orientation in Drosophila neuroblasts Proc Natl Acad Sci U S A 104:14306-11 Ogawa, H., N Ohta, W Moon, and F Matsuzaki 2009 Protein phosphatase 2A negatively regulates aPKC signaling by modulating phosphorylation of Par-6 in Drosophila neuroblast asymmetric divisions J Cell Sci 122:3242-9 Ohlstein, B., and A Spradling 2006 The adult Drosophila posterior midgut is maintained by pluripotent stem cells Nature 439:470-4 Ohshiro, T., T Yagami, C Zhang, and F Matsuzaki 2000 Role of cortical tumour-suppressor proteins in asymmetric division of Drosophila neuroblast Nature 408:593-6 Peng, C.Y., L Manning, R Albertson, and C.Q Doe 2000 The tumoursuppressor genes lgl and dlg regulate basal protein targeting in Drosophila neuroblasts Nature 408:596-600 Peterson, F.C., R.R Penkert, B.F Volkman, and K.E Prehoda 2004 Cdc42 regulates the Par-6 PDZ domain through an allosteric CRIB-PDZ transition Mol Cell 13:665-76 Petritsch, C., G Tavosanis, C.W Turck, L.Y Jan, and Y.N Jan 2003 The Drosophila myosin VI Jaguar is required for basal protein targeting and correct spindle orientation in mitotic neuroblasts Dev Cell 4:273-81 Prehoda K E 2009 Polarization of Drosophila Neuroblasts During Asymmetric Division Cold Spring Harb.Perspect Biol 1: a001388 Rebollo, E., M Roldan, and C Gonzalez 2009 Spindle alignment is achieved without rotation after the first cell cycle in Drosophila embryonic neuroblasts Development 136:3393-7 Rebollo, E., P Sampaio, J Januschke, S Llamazares, H Varmark, and C Gonzalez 2007 Functionally unequal centrosomes drive spindle orientation in asymmetrically dividing Drosophila neural stem cells Dev Cell 12:467-74 Reya, T., S.J Morrison, M.F Clarke, and I.L Weissman 2001 Stem cells, cancer, and cancer stem cells Nature 414:105-11 Rhyu, M.S., L.Y Jan, and Y.N Jan 1994 Asymmetric distribution of numb protein during division of the sensory organ precursor cell confers distinct fates to daughter cells Cell 76:477-91 133 References Rolls, M.M., R Albertson, H.P Shih, C.Y Lee, and C.Q Doe 2003 Drosophila aPKC regulates cell polarity and cell proliferation in neuroblasts and epithelia J Cell Biol 163:1089-98 Rusan, N.M., K Akong, and M Peifer 2008 Putting the model to the test: are APC proteins essential for neuronal polarity, axon outgrowth, and axon targeting? J Cell Biol 183:203-12 Rusan, N.M., and M Peifer 2007 A role for a novel centrosome cycle in asymmetric cell division J Cell Biol 177:13-20 Sambrook, J., and Russell, D.W Molecular Cloning a laboratory manual (Cold spring Harbour Laboratory Press) Schaefer, M., M Petronczki, D Dorner, M Forte, and J.A Knoblich 2001 Heterotrimeric G proteins direct two modes of asymmetric cell division in the Drosophila nervous system Cell 107:183-94 Schaefer, M., A Shevchenko, and J.A Knoblich 2000 A protein complex containing Inscuteable and the Galpha-binding protein Pins orients asymmetric cell divisions in Drosophila Curr Biol 10:353-62 Schwamborn, J.C., E Berezikov, and J.A Knoblich 2009 The TRIM-NHL protein TRIM32 activates microRNAs and prevents self-renewal in mouse neural progenitors Cell 136:913-25 Schweisguth, F 2004 Regulation of notch signaling activity Curr Biol 14:R129-38 Shen, C.P., L.Y Jan, and Y.N Jan 1997 Miranda is required for the asymmetric localization of Prospero during mitosis in Drosophila Cell 90:449-58 Shiomi, K., M Takeichi, Y Nishida, Y Nishi, and T Uemura 1994 Alternative cell fate choice induced by low-level expression of a regulator of protein phosphatase 2A in the Drosophila peripheral nervous system Development 120:1591-9 Siegrist, S.E., and C.Q Doe 2005 Microtubule-induced Pins/Galphai cortical polarity in Drosophila neuroblasts Cell 123:1323-35 Siller, K.H., C Cabernard, and C.Q Doe 2006 The NuMA-related Mud protein binds Pins and regulates spindle orientation in Drosophila neuroblasts Nat Cell Biol 8:594-600 Skeath, J.B., and C.Q Doe 1998 Sanpodo and Notch act in opposition to Numb to distinguish sibling neuron fates in the Drosophila CNS Development 125:1857-65 Skeath, J.B., and S Thor 2003 Genetic control of Drosophila nerve cord development Curr Opin Neurobiol 13:8-15 Slack, C., P.M Overton, R.I Tuxworth, and W Chia 2007 Asymmetric localisation of Miranda and its cargo proteins during neuroblast division 134 References requires the 134:3781-7 anaphase-promoting complex/cyclosome Development Smith, C.A., K.M Lau, Z Rahmani, S.E Dho, G Brothers, Y.M She, D.M Berry, E Bonneil, P Thibault, F Schweisguth, R Le Borgne, and C.J McGlade 2007 aPKC-mediated phosphorylation regulates asymmetric membrane localization of the cell fate determinant Numb EMBO J 26:46880 Snaith, H.A., C.G Armstrong, Y Guo, K Kaiser, and P.T Cohen 1996 Deficiency of protein phosphatase 2A uncouples the nuclear and centrosome cycles and prevents attachment of microtubules to the kinetochore in Drosophila microtubule star (mts) embryos J Cell Sci 109 ( Pt 13):3001-12 Sousa-Nunes, R., W Chia, and W.G Somers 2009 Protein phosphatase mediates localization of the Miranda complex during Drosophila neuroblast asymmetric divisions Genes Dev 23:359-72 Spana, E.P., and C.Q Doe 1996 Numb antagonizes Notch signaling to specify sibling neuron cell fates Neuron 17:21-6 Suzuki, A., and S Ohno 2006 The PAR-aPKC system: lessons in polarity J Cell Sci 119:979-87 Uemura, T., S Shepherd, L Ackerman, L.Y Jan, and Y.N Jan 1989 numb, a gene required in determination of cell fate during sensory organ formation in Drosophila embryos Cell 58:349-60 Uemura, T., K Shiomi, S Togashi, and M Takeichi 1993 Mutation of twins encoding a regulator of protein phosphatase 2A leads to pattern duplication in Drosophila imaginal discs Genes Dev 7:429-40 Wang, C., K.C Chang, G Somers, D Virshup, B.T Ang, C Tang, F Yu, and H Wang 2009 Protein phosphatase 2A regulates self-renewal of Drosophila neural stem cells Development 136:2287-96 Wang, H., Y Ouyang, W.G Somers, W Chia, and B Lu 2007 Polo inhibits progenitor self-renewal and regulates Numb asymmetry by phosphorylating Pon Nature 449:96-100 Wang, H., G.W Somers, A Bashirullah, U Heberlein, F Yu, and W Chia 2006 Aurora-A acts as a tumor suppressor and regulates self-renewal of Drosophila neuroblasts Genes Dev 20:3453-63 Westermarck, J., and W.C Hahn 2008 Multiple pathways regulated by the tumor suppressor PP2A in transformation Trends Mol Med 14:152-60 Wirtz-Peitz, F., T Nishimura, and J.A Knoblich 2008 Linking cell cycle to asymmetric division: Aurora-A phosphorylates the Par complex to regulate Numb localization Cell 135:161-73 Xu, T., and G.M Rubin 1993 Analysis of genetic mosaics in developing and adult Drosophila tissues Development 117:1223-37 135 References Xue, Y., J Ren, X Gao, C Jin, L Wen, and X Yao 2008 GPS 2.0, a tool to predict kinase-specific phosphorylation sites in hierarchy Mol Cell Proteomics 7:1598-608 Yamanaka, T., Y Horikoshi, A Suzuki, Y Sugiyama, K Kitamura, R Maniwa, Y Nagai, A Yamashita, T Hirose, H Ishikawa, and S Ohno 2001 PAR-6 regulates aPKC activity in a novel way and mediates cell-cell contactinduced formation of the epithelial junctional complex Genes Cells 6:721-31 Yu, F., X Morin, Y Cai, X Yang, and W Chia 2000 Analysis of partner of inscuteable, a novel player of Drosophila asymmetric divisions, reveals two distinct steps in inscuteable apical localization Cell 100:399-409 Yu, F., H Wang, H Qian, R Kaushik, M Bownes, X Yang, and W Chia 2005 Locomotion defects, together with Pins, regulates heterotrimeric Gprotein signaling during Drosophila neuroblast asymmetric divisions Genes Dev 19:1341-53 136 ... development of multicellular organisms, it is also a means of keeping stem cell self- renewal and differentiation in balance During asymmetric division of neural stem cells in Drosophila melanogaster; ... through asymmetric division Each stem cell generates another stem cell and a sibling daughter cell destined to differentiate B Extrinsic versus intrinsic regulation of asymmetric division Extrinsic... determinants during neuroblast asymmetric cell division results in both the daughter cells adopting a stem cell- like identity upon division In the absence of these basal targeting adaptor proteins

Ngày đăng: 11/09/2015, 14:32

Tài liệu cùng người dùng

  • Đang cập nhật ...

Tài liệu liên quan