• No results found

The roles of Canoe (Cno) in the regulation of morphological changes during Drosophila embryogenesis

N/A
N/A
Protected

Academic year: 2020

Share "The roles of Canoe (Cno) in the regulation of morphological changes during Drosophila embryogenesis"

Copied!
29
0
0

Loading.... (view fulltext now)

Full text

(1)

The roles of Canoe (Cno) in the regulation of morphological

changes during

Drosophila

embryogenesis

Kuo-Chen Jung

A thesis submitted to the faculty of the University of North Carolina at Chapel Hill in partial fulfillment of the requirements for the degree of Master of Science in the

Department of Biology

Chapel Hill 2012

Approved by: Mark Peifer Alan Fanning

(2)

ii

ABSTRACT

KUO-CHEN JUNG: The roles of Canoe (Cno) in the regulation of morphological changes during Drosophila embryogenesis

(Under the direction of Mark Peifer)

(3)

iii

ACKNOWLEDGEMENTS

I would like to express my appreciation to my advisor Mark Peifer. He guided

me into the fascinating world of science. I also want to show my gratitude to my lab

members: Ben, Colleen, Ed, John, Mira, Nathan, Steph, and Wangsun. Mark’s

mentorship and all the support from Peifer lab help me become more mature both in

research and as a person. My committee members, Alan Fanning and Bob Goldstein

have been very helpful throughout my time at UNC. I would also want to thank my

parents, Hsuan-Chu Jung and Yu-Mei Chou, as well as my friends Jui-Hua Hsieh,

Hung-Ching Hsia, Yini Su, Alice Weng, Ben Chung and Congying Wu for supporting

(4)

iv

TABLE OF CONTENTS

LIST OF FIGURES ...v

LIST OF ABBREVIATIONS ...vi

I: INTRODUCTION ...1

II: SUMMARY OF CONTRIBUTIONS TO PUBLISHED WORK Preface of Chio et. al., 2011 ...8

Preface of Sawyer et. al., 2011 ...9

Preface of Roberts et. al., 2012 ...10

III: CONCLUSION ...11

IV: FIGURES ………...…………14

(5)

v

LIST OF FIGURES

Fig.1 Adherens junction (AJ) complex………70 Fig.2 Domain structure of Afadin and Canoe. Afadin……….71 Fig.3 Sequence alignment of human Rap1A, Rap1B (H-RAP1A and H-RAP1-B),

and Drosophila Rap1 (D-RAP1)……….………….72 Fig.4 Dorsal closure………..73 Fig.5 Cross-section of a Drosophila embryo transiting from syncytial stages

(6)

vi

LIST OF ABBREVIATIONS

ABD Actin binding domain AJ Adherens Junction

ADIP Afadin DIL domain-interacting protein

AF-6 ALL1-fused gene from chromosome 6 protein AS Amnioserosa

AP Anterior-posterior aPKC Atypical kinase C Baz Bazooka

Cno Canoe

Dpp Decapentaplegic DIL Dilute domain Dlg Disc Large DC Dorsal closure DV Dorsal-ventral Ecad Ecadherin

FHA Forkhead associated GBE Germband extension JNK Jun N-terminal kinase LE Leading edge

PDZ PSD-95-Dlg-1-ZO-1 Puc Puckered

(7)

Chapter I

INTRODUCTION

Embryogenesis cannot be accomplished without proper morphological changes. Accomplishing accurate morphogenesis in organisms requires both the coordination between Adherens Junctions (AJ) and cytoskeleton network and also adhesion between cells to maintain tissue integrity. In the Peifer lab, we are interested in the machinery which controls the coordination between AJ and the cytoskeletal network.

The Adherens Junction

The AJ is composed of a core complex: Ecadherin (Ecad) and the catenins plus Nectin and Afadin (Fig.1). The relationship between the Ecad/catenin complex and the actin cytoskeleton has been well studied. The prevalent model was that the cytoplasmic domain of Ecad can interact with actin filaments via the β-catenin/α-catenin complex [1], but work from Weis’ and Nelson’s groups suggested a new view. They found that when α-catenin binds to

(8)

2

involved in E-cadherin-based intercellular AJ formation [4] and also can bind to the PDZ domain of the actin binding protein Afadin (Drosophila Canoe), through its C terminal region.

Afadin/ Canoe

Afadin is a multiple domain scaffolding protein. RA (Ras associating domain) domains within Afadin bind to both Ras [5] and Rap1 [6]. The forkhead associated (FHA) domain is a putative nuclear signaling domain, and the dilute domain (DIL) domain binds to Afadin DIL domain-interacting protein (ADIP) [7]. The PSD-95–Dlg-1–ZO-1 (PDZ) domain can bind to the transmembrane proteins Nectin [8], Eph receptor [9], SPA-1 [10], and Bcr [11]. The region adjacent to the PDZ domain is less conserved, and contains high percentage of proline. This region physically interacts with certain adaptors or actin regulators such as ponsin [12], profilin [13], α-catenin [14, 15], and the tight junction protein ZO-1 [16]. The C terminal actin binding domain (ABD) binds to F-actin [17] (Fig.2).

Afadin’s human homolog, ALL1-fused gene from chromosome 6 protein (AF-6), was

(9)

3

and also eliminate maternally loaded Cno, have a mesodermal cell invagination defect. The apical constriction of mesodermal cells initiated but was not completed, due to the separation of AJs and the actomyosin network. Thus we believe that Cno functions as an essential linker between AJs and the actomyosin network [24]. We also found that in cnoMZmutants, germband extension (GBE) is reduced, myosin detaches from the anterior and posterior cell boundaries, planar polarity of apical polarity proteins, Bazooka and aPKC, is dramatically enhanced in epithelial cells during GBE, and both Baz and aPKC are largely reduced on anterior and posterior cell borders [25].

Rap1

Genetic interactions suggest Cno may participate in several signaling pathways, including Jun N-terminal kinase (JNK) [16], Notch [26], and Wnt signaling [27]. One of the most studied upstream regulators is Rap1, a member of the Ras super family of small GTPases. Like Afadin, Drosophila Cno also binds to Rap1 via its N terminal RA domain [28]. Rap1 cycles between an active GTP-bound form and an inactive GDP-bound form to participate in signal transduction cascades. In humans, there are two Rap1 genes, Rap1A and Rap1B while in Drosophila there is only one Rap1 (Fig3). Rap1 was first identified by its ability to attenuate Ras-ERK signaling activity and thus antagonize Ras oncogenic transformation [29, 30]. Its activity is regulated by its upstream activators (such as PDZ-GEF, C3G, RapGRP, Dock4, and Epac) and inactivators (such as RapGAP, Spa1, and E6-TP1). Rap1 has multiple downstream effectors, including RapL, Riam, AF-6, Vav2, and Tiam, allowing Rap1 to be in the center of different signal pathways (reviewed in [31-33]).

(10)

4

cell adhesion junction maintenance [37]. Both Rap1 and Afadin are reported to inhibit E-cadherin endocytosis in MDCK cells [38]. Furthermore, in Drosophila imaginal disc cells, Ecad and Cno are no longer uniformly localized around the cell cortex in Rap1 mutant clones with Ecad, most likely being lost between newly divided daughter cells [37]. In addition, Rap1 mutants also have a range of other morphological defects, such as a reduced number of nurse cells in egg chambers, abnormal pole cell migration, ventral furrow invagination defect, and dorsal closure failure. [28, 39, 40].

Both Cno and Rap1 accumulate around the cell cortex during dorsal closure. Zygotic cno null mutants gastrulate normally, but die at the end of embryogenesis; most embryos have head involution defects and a minority have both head involution and dorsal closure defects [24, 28]. In cno, the expression of puckered (puc), a JNK target gene, is eliminated in leading edge cells [16]. This data indicates that Cno is a critical regulator of dorsal closure. Consistent with these findings, embryos expressing Rap1DN are reported to have severe defects in dorsal closure. When scientists introduced a dominant negative form of Rap1, Rap1N17, AS cell detached from LE cells during late dorsal closure. Cno lacking the N terminal RA domain failed to bind to Rap1 in yeast two hybrid system, but still localized to the junction sites and retained partial ability to rescue cno defects during dorsal closure. Dpp expression is reduced in cno mutants but is not affected in Rap1 mutants. This implies that Rap1 has both Cno dependent and independent functions, probably through downstream effectors other than Cno [6, 28].

Bazooka(PAR-3) and aPKC

(11)

5

complex. It differs from conventional PKCs by a N terminal PB1 domain, which is known to interact with PAR6. It also has a C terminal catalytic domain which is known to phosphorylate several proteins such as PAR3 [42]. In mammalian system, two aPKC proteins have been identified: aPKCλ/ι and aPKCζ [43]. They colocalize with the tight junction protein ZO-1 in MDCKII cells [44]. Introduction of a dominant form of aPKC in MDCK cells results in disrupted tight junction formation and mislocalized PAR6 and PAR3 [45]. In Drosophila, there is only one aPKC [46]. Studies in Drosophila revealed that aPKC is required for proper microtubule organization and symmetric AJ positioning during Drosophila early embryogenesis [47].

Drosophila Bazooka (Baz) is the ortholog of C. elegan PAR-3. It localizes to the anterior end of 1-cell C. elegan embryos and appears to play a central role in polarity establishment [48]. In mammalian cells, there are two PAR-3 proteins: PAR-3A and PAR-3B. Studies on PAR-3A showed that it also colocalizes with a tight junction marker ZO-1 and is required for PAR-6 and aPKC recruitment, and proper tight junction structure (reviewed in [41]). In Drosophila, Baz, PAR-6, and aPKC are enriched to the cortex of anterior and lateral sides of oocyte. However, within Drosophila primary epithelia, Baz is slightly below PAR-6 and aPKC in the Z axis (reviewed in [49]). It plays a role in directing apical assembly of AJs in Drosophila epithelia [50].

Drosophila embryogenesis

(12)

6

closure. For the past several years, I have focused on helping to understand the functions of Rap1/ Cno in the following three developmental events:

Initial cell polarity establishment

In Drosophila syncytial stages, before polarity established, the apical marker Baz, Ecad and the basal lateral marker Disc Large (Dlg) localize together at the cortex. After 13 nuclear divisions (Fig.4 A), cellularization occurs (Fig. 4B) and embryos start to build up their cell polarity (Fig.4 B to D). During this process, pseudocleavage furrows elongate. Baz, E-cad, and Dlg localize apically of the furrow. However, during cellularization, Baz and E-cad colocalize and accumulate in the apical region, while Dlg is along the entire furrow with slight enrichment at the apical end. It had been thought that AJs were the key landmark for cell polarity establishment. However, previous work from our lab demonstrated that Baz, one of the components of apical polarity complex, acts as an upstream regulator of AJ positioning. In arm or e-cadMZ mutants, Baz still localizes properly to its apical site, but in bazMZmutants, E-cad is no longer restrained in the apical region and shifts basally along the lateral membrane [50]. This indicates that Baz act upstream of AJs to determine their polarity.

Germband extension

(13)

7

germ-band movement is largely driven by cell intercalation. Cell intercalation requires proper planar polarity of AJs and apical polarity proteins (Baz and aPKC) which are enriched along DV borders, as well as actin and nonmuscle myosinII (myosinII) which enriched along AP boarders[25]. Disrupted planar polarity causes delay in this process.

Dorsal closure

(14)

8

Chapter II

SUMMARY OF CONTRIBUTIONS TO PUBLISHED WORK

Preface

This chapter includes three publications on which I am a co-author. Each is preceded by a preface describing my contributions to the work.

Choi et al, 2011

In this publication, we generated null alleles of the fly homolog of the mammalian tight junction protein ZO-1, Drosophila polychaetoid (pyd), to addresses the role of Pyd in

morphological changes. Our results show that Pyd is required for tracheal dorsal branch fusion, tracheal cell fate specification and proper morphogenesis during dorsal closure in Drosophila

embryogenesis. We further demonstrated that mutants lacking Pyd or its binding partner Afadin/ Canoe (Cno) share similar morphological defects, including: a) failure of segmental groove retraction b) a disrupted leading edge actin cable c) a reduced zippering d) mislocalization of the actin regulator Enabled (Ena).

My contribution in this work focused on characterizing cno phenotypes (Fig.6 E, G, L, Fig.8, and Fig. 9). I found that the localization of core AJ components D-Ecadherin (DEcad) and Armadillo (Arm), as well as Pyd, are not affected by Cno loss (Fig.8 F’, H’, and M, respectively).

(15)

9

Sawyer et al, 2011

This publication highlights the importance of Afadin /Canoe (Cno) in linking the contractile actomyosin network to AJs during morphological changes. We used cno maternal zygotic mutants to identify the role of Cno in Drosophila germband extension. In the absence of Cno, the apical actomyosin network detaches from AJs at AP cell borders within epithelial cells, and the planar polarity of the apical polarity proteins Bazooka (Baz)/Par3 as well as atypical protein kinase C (aPKC) is substantially altered. We further demonstrated that this effect mimics the disruption of actin polymerization.

(16)

10

Roberts et. al, 2012

This publication contributes four novel ideas to the mechanism by which β-catenin, Drosophila Armadillo (Arm), is regulated by using Drosophila as a model system. First, Roc1 is involved in SCF-βTrCP mediates Arm destruction in Drosophila cultured cells. Second, Arm levels have different sensitivity in response to inactivation of APC between Drosophila embryos and larvae. Third, there is no correlation between the roles in downregulating Arm protein and putative positive roles in Wnt signaling among different APC2 mutant proteins. Finally, N-terminal Armadillo repeat domain deleted APC could negatively-regulate Wnt signaling but was not able to downregulate Arm level.

(17)

Chapter III

CONCLUSION

One of the interests in our lab is to explore the mechanisms which regulate

morphogenetic movement during embryogenesis. Cno, an AJ-actin cytoskeleton linker protein, is essential for mesodermal cell apical constriction during gastrulation, as well as cell intercalation during GBE stages, and acts by linking AJ to contractile acto-myosin contractile ring [24, 25]. However, how Cno functions in other cell behaviors in different stages is largely unknown. Dorsal closure inlvoves in two types of cell that work together to seal the dorsal hole: First, AS cells cyclically constrict apically. Second, LE cells form actomyosin cables, send out actin protrusions, and elongate during the closure process. By generating zygotic cno mutants,we were able to reduce Cno protein levels to study Cno functions in regulating different cell

behaviors, such as cell elongation, assymetric actin cables, and polarized protrusion formation, in late Drosophila developmental stages. We found cno mutants have the following phenotypes: a) deep segmental grooves b) a discontinuous cable c) irregular leading edge cell diameter, and d) zippering defects. [57]

Potential players with which Cno works to contribute to morphogenetic movements

(18)

12

eliminating Pyd completely share the above mentioned phenotypes with Cno. Genetic interaction tests further demonstrate that pyd works together with Cno to help dorsal closure completion [1]. However, in cno, Pyd is still localized properly as seen in WT. Further characterization of Pyd protein levels in cno mutants could provide clues to answer whether Cno could help anchor Pyd in apical cortex region as WT [1].

How might these two proteins work together to help dorsal closure completion? One of the interesting players that may work downstream of Pyd and Cno is Enabled (Ena), an

anticapping protein. From previous studies we already know that Ena induces philopodia formation in mammalian cell culture [2]. In both cno and pyd mutants, Ena localization is

changed [1]. Genetic interaction test further demonstrates that Ena works both with Pyd and Cno to help dorsal closure process completion. However, cuticle morphology shows ena also has milder defect in epithelial integrity than cno does. Mutants in which Ena protein is eliminated completely only have deep segmental grooves and zippering defects but the actin cable is still present in the leading edge front. This probably could provide one of the reasons why ena has milder defect than cno does.

(19)

13

and Pyd localization in ed mutants, during dorsal closure stage embryos could help understand whether and how these proteins work together to regulate LE cable assembly. Which domain is essential for cable formation? Ectopic expression of Ed lacking its C terminus ( including the intracellular and PDZ binding domain) in stripes in both AS and LE cells results in abnormal retaining of Ed protein at the LE front. Similar results were obtained for extopic expression of both full length Ed and Ed lacking the PDZ binding domain. By generating mosaic clones in folicle cells, Laplante et. al. further demonstrated that the PDZ binding domain (reported to interact with Cno and Baz) is dispensable for actin cable formation whereas the intracellular domain is necessary [4]. Further experiments are needed to determine whether this implies the interaction between Ed and Cno is not important for Ed mediated cable formation, and if Cno is involved in cable formation independent from Ed.

Finally, the known Cno upstream regulator Rap1 is also expressed in the epithelial cell cortex, partially overlapping with Cno. Rap1 is present along cell membranes from apical to basal while Cno only appears in apical area. Cuticle data of Rap1 germline clones show ventral holes and this implies the morphology defect mainly happened during mesoderm invagination. Zygotic Rap1 mutants are not embryonic lethal, probably due to high maternal loading. These make Rap1DN becomes better tool to study Rap1 function in dorsal closure stages. Expressing Rap1DNin two stripes per segment of lateral ectoderm and the amnioserosa cells causes 93 % dorsal hole phenotype. As the dorsal closure process progressing, in some embryos, the AS cells detach from the LE cells, LE cells failed to elongate ,and the AS cells shrink [5]. Further

(20)

Chapter IV

FIGURES

(21)

15

Fig2.Domain structure of Afadin and Canoe. Rat Afadin, 1829aa; Drosophila Canoe, 2051aa; the DrosophilacnoR2

(22)

16

Fig3. Sequence alignment of human Rap1A, Rap1B (H-RAP1A and H-RAP1-B), and Drosophila Rap1 (D-RAP1)

The comparison of human Rap1 and

(23)

17

(24)

18

(25)

19

REFERENCE

1. Rimm, D.L., et al., Alpha 1(E)-catenin is an actin-binding and -bundling protein mediating the attachment of F-actin to the membrane adhesion complex. Proc Natl Acad Sci U S A, 1995.

92(19): p. 8813-7.

2. Yamada, S., et al., Deconstructing the cadherin-catenin-actin complex. Cell, 2005. 123(5): p. 889-901.

3. Takai, Y., et al., The immunoglobulin-like cell adhesion molecule nectin and its associated protein afadin. Annu Rev Cell Dev Biol, 2008. 24: p. 309-42.

4. Honda, T., et al., Antagonistic and agonistic effects of an extracellular fragment of nectin on formation of E-cadherin-based cell-cell adhesion. Genes Cells, 2003. 8(1): p. 51-63.

5. Kuriyama, M., et al., Identification of AF-6 and canoe as putative targets for Ras. J Biol Chem, 1996. 271(2): p. 607-10.

6. Linnemann, T., et al., Thermodynamic and kinetic characterization of the interaction between the Ras binding domain of AF6 and members of the Ras subfamily. J Biol Chem, 1999. 274(19): p. 13556-62.

7. Asada, M., et al., ADIP, a novel Afadin- and alpha-actinin-binding protein localized at cell-cell adherens junctions. J Biol Chem, 2003. 278(6): p. 4103-11.

8. Takahashi, K., et al., Nectin/PRR: an immunoglobulin-like cell adhesion molecule recruited to cadherin-based adherens junctions through interaction with Afadin, a PDZ domain-containing protein. J Cell Biol, 1999. 145(3): p. 539-49.

9. Buchert, M., et al., The junction-associated protein AF-6 interacts and clusters with specific Eph receptor tyrosine kinases at specialized sites of cell-cell contact in the brain. J Cell Biol, 1999.

144(2): p. 361-71.

10. Su, L., et al., AF-6 controls integrin-mediated cell adhesion by regulating Rap1 activation through the specific recruitment of Rap1GTP and SPA-1. J Biol Chem, 2003. 278(17): p. 15232-8.

11. Radziwill, G., et al., The Bcr kinase downregulates Ras signaling by phosphorylating AF-6 and binding to its PDZ domain. Mol Cell Biol, 2003. 23(13): p. 4663-72.

12. Mandai, K., et al., Ponsin/SH3P12: an l-afadin- and vinculin-binding protein localized at cell-cell and cell-matrix adherens junctions. J Cell Biol, 1999. 144(5): p. 1001-17.

(26)

20

14. Pokutta, S., et al., Biochemical and structural definition of the l-afadin- and actin-binding sites of alpha-catenin. J Biol Chem, 2002. 277(21): p. 18868-74.

15. Miyahara, M., et al., Interaction of nectin with afadin is necessary for its clustering at cell-cell contact sites but not for its cis dimerization or trans interaction. J Biol Chem, 2000. 275(1): p. 613-8.

16. Takahashi, K., et al., Direct binding between two PDZ domain proteins Canoe and ZO-1 and their roles in regulation of the jun N-terminal kinase pathway in Drosophila morphogenesis. Mech Dev, 1998. 78(1-2): p. 97-111.

17. Mandai, K., et al., Afadin: A novel actin filament-binding protein with one PDZ domain localized at cadherin-based cell-to-cell adherens junction. J Cell Biol, 1997. 139(2): p. 517-28.

18. Prasad, R., et al., Cloning of the ALL-1 fusion partner, the AF-6 gene, involved in acute myeloid leukemias with the t(6;11) chromosome translocation. Cancer Res, 1993. 53(23): p. 5624-8.

19. Miyata, M., et al., Localization of nectin-free afadin at the leading edge and its involvement in directional cell movement induced by platelet-derived growth factor. J Cell Sci, 2009. 122(Pt 23): p. 4319-29.

20. Lorger, M. and K. Moelling, Regulation of epithelial wound closure and intercellular adhesion by interaction of AF6 with actin cytoskeleton. J Cell Sci, 2006. 119(Pt 16): p. 3385-98.

21. Miyata, M., et al., Regulation by afadin of cyclical activation and inactivation of Rap1, Rac1, and RhoA small G proteins at leading edges of moving NIH3T3 cells. J Biol Chem, 2009. 284(36): p. 24595-609.

22. Ikeda, W., et al., Afadin: A key molecule essential for structural organization of cell-cell junctions of polarized epithelia during embryogenesis. J Cell Biol, 1999. 146(5): p. 1117-32.

23. Zhadanov, A.B., et al., Absence of the tight junctional protein AF-6 disrupts epithelial cell-cell junctions and cell polarity during mouse development. Curr Biol, 1999. 9(16): p. 880-8.

24. Sawyer, J.K., et al., The Drosophila afadin homologue Canoe regulates linkage of the actin cytoskeleton to adherens junctions during apical constriction. J Cell Biol, 2009. 186(1): p. 57-73.

25. Sawyer, J.K., et al., A contractile actomyosin network linked to adherens junctions by Canoe/afadin helps drive convergent extension. Mol Biol Cell.

26. Miyamoto, H., et al., canoe encodes a novel protein containing a GLGF/DHR motif and functions with Notch and scabrous in common developmental pathways in Drosophila. Genes Dev, 1995.

9(5): p. 612-25.

27. Carmena, A., S. Speicher, and M. Baylies, The PDZ protein Canoe/AF-6 links Ras-MAPK, Notch and Wingless/Wnt signaling pathways by directly interacting with Ras, Notch and Dishevelled.

(27)

21

28. Boettner, B., et al., The AF-6 homolog canoe acts as a Rap1 effector during dorsal closure of the Drosophila embryo. Genetics, 2003. 165(1): p. 159-69.

29. Cook, S.J., et al., RapV12 antagonizes Ras-dependent activation of ERK1 and ERK2 by LPA and EGF in Rat-1 fibroblasts. EMBO J, 1993. 12(9): p. 3475-85.

30. Kitayama, H., et al., A ras-related gene with transformation suppressor activity. Cell, 1989. 56(1): p. 77-84.

31. Boettner, B. and L. Van Aelst, Control of cell adhesion dynamics by Rap1 signaling. Curr Opin Cell Biol, 2009. 21(5): p. 684-93.

32. Hattori, M. and N. Minato, Rap1 GTPase: functions, regulation, and malignancy. J Biochem, 2003.

134(4): p. 479-84.

33. Kooistra, M.R., N. Dube, and J.L. Bos, Rap1: a key regulator in cell-cell junction formation. J Cell Sci, 2007. 120(Pt 1): p. 17-22.

34. Posern, G., et al., Activity of Rap1 is regulated by bombesin, cell adhesion, and cell density in NIH3T3 fibroblasts. J Biol Chem, 1998. 273(38): p. 24297-300.

35. Reedquist, K.A., et al., The small GTPase, Rap1, mediates CD31-induced integrin adhesion. J Cell Biol, 2000. 148(6): p. 1151-8.

36. Hogan, C., et al., Rap1 regulates the formation of E-cadherin-based cell-cell contacts. Mol Cell Biol, 2004. 24(15): p. 6690-700.

37. Knox, A.L. and N.H. Brown, Rap1 GTPase regulation of adherens junction positioning and cell adhesion. Science, 2002. 295(5558): p. 1285-8.

38. Hoshino, T., et al., Regulation of E-cadherin endocytosis by nectin through afadin, Rap1, and p120ctn. J Biol Chem, 2005. 280(25): p. 24095-103.

39. Asha, H., et al., The Rap1 GTPase functions as a regulator of morphogenesis in vivo. EMBO J, 1999. 18(3): p. 605-15.

40. Hariharan, I.K., R.W. Carthew, and G.M. Rubin, The Drosophila roughened mutation: activation of a rap homolog disrupts eye development and interferes with cell determination. Cell, 1991.

67(4): p. 717-22.

41. Assemat, E., et al., Polarity complex proteins. Biochim Biophys Acta, 2008. 1778(3): p. 614-30.

42. Nagai-Tamai, Y., et al., Regulated protein-protein interaction between aPKC and PAR-3 plays an essential role in the polarization of epithelial cells. Genes Cells, 2002. 7(11): p. 1161-71.

(28)

22

44. Izumi, Y., et al., An atypical PKC directly associates and colocalizes at the epithelial tight junction with ASIP, a mammalian homologue of Caenorhabditis elegans polarity protein PAR-3. J Cell Biol, 1998. 143(1): p. 95-106.

45. Suzuki, A., et al., Atypical protein kinase C is involved in the evolutionarily conserved par protein complex and plays a critical role in establishing epithelia-specific junctional structures. J Cell Biol, 2001. 152(6): p. 1183-96.

46. Wodarz, A., et al., Drosophila atypical protein kinase C associates with Bazooka and controls polarity of epithelia and neuroblasts. J Cell Biol, 2000. 150(6): p. 1361-74.

47. Harris, T.J. and M. Peifer, aPKC controls microtubule organization to balance adherens junction symmetry and planar polarity during development. Dev Cell, 2007. 12(5): p. 727-38.

48. Tabuse, Y., et al., Atypical protein kinase C cooperates with PAR-3 to establish embryonic polarity in Caenorhabditis elegans. Development, 1998. 125(18): p. 3607-14.

49. St Johnston, D. and J. Ahringer, Cell polarity in eggs and epithelia: parallels and diversity. Cell.

141(5): p. 757-74.

50. Harris, T.J. and M. Peifer, Adherens junction-dependent and -independent steps in the establishment of epithelial cell polarity in Drosophila. J Cell Biol, 2004. 167(1): p. 135-47.

51. Martin, A.C., M. Kaschube, and E.F. Wieschaus, Pulsed contractions of an actin-myosin network drive apical constriction. Nature, 2009. 457(7228): p. 495-9.

52. David, D.J., A. Tishkina, and T.J. Harris, The PAR complex regulates pulsed actomyosin contractions during amnioserosa apical constriction in Drosophila. Development. 137(10): p. 1645-55.

53. Toyama, Y., et al., Apoptotic force and tissue dynamics during Drosophila embryogenesis.

Science, 2008. 321(5896): p. 1683-6.

54. Millard, T.H. and P. Martin, Dynamic analysis of filopodial interactions during the zippering phase of Drosophila dorsal closure. Development, 2008. 135(4): p. 621-6.

55. Noselli, S. and F. Agnes, Roles of the JNK signaling pathway in Drosophila morphogenesis. Curr Opin Genet Dev, 1999. 9(4): p. 466-72.

56. Hou, X.S., E.S. Goldstein, and N. Perrimon, Drosophila Jun relays the Jun amino-terminal kinase signal transduction pathway to the Decapentaplegic signal transduction pathway in regulating epithelial cell sheet movement. Genes Dev, 1997. 11(13): p. 1728-37.

57. Choi, W., et al., The single Drosophila ZO-1 protein Polychaetoid regulates embryonic morphogenesis in coordination with Canoe/Afadin and Enabled. Mol Biol Cell.

(29)

23

59. Bear, J.E., et al., Antagonism between Ena/VASP proteins and actin filament capping regulates fibroblast motility. Cell, 2002. 109(4): p. 509-21.

60. Lin, H.P., et al., Cell adhesion molecule Echinoid associates with unconventional myosin VI/Jaguar motor to regulate cell morphology during dorsal closure in Drosophila. Dev Biol, 2007.

311(2): p. 423-33.

Figure

Fig. 4  Cross-section  of  a Drosophila embryo transit from syncytial stage to cellularization stage

References

Related documents

A study to assess the effectiveness of planned teaching program regarding the knowledge of psycho active substance abuse and its consequences on general health

Results: There are a few types of interventions such as the multisensory method, the phonological intervention, and the cognitive training method which can be used to

learning strategies as the main content, and collects a variety of teaching models and teaching methodsguided with foreign language teaching theory [2].To cultivate

In contrast with chemisorption, in which the electronic structure of bonding atoms or molecules is changed and covalent or ionic bonds are formed where as

In this paper, we provide a fault-tolerant replacement sequence for almost all the instructions of the Thumb- 2 instruction set and provide a formal verification for this

In four of the 24 participating centers, we scrutinized hospital inpatient data to identify all colorectal cancer patients who underwent surgery, in order to ascertain all

We also observed that both saliva and serum obestatin were clearly lower in the IDA patients compared with the control subjects; and after iron supplementation,

Overall, the data suggest that Se Pat activity is responsive to small changes in the levels of Ac-CoA and that oligomerization in response to Ac-CoA produces the cooperative