142
Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah Lynn Windler A dissertation submitted in partial satisfaction of the requirements for the degree of Doctor of Philosophy in Molecular and Cell Biology in the Graduate Division of the University of California, Berkeley Committee in charge: Professor David Bilder, Chair Professor Iswar Hariharan Professor David Drubin Professor Kathleen Ryan Fall 2010

Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

  • Upload
    others

  • View
    2

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

Regulation of cellular signaling pathways by endocytosis and protein degradation

by

Sarah Lynn Windler

A dissertation submitted in partial satisfaction of the

requirements for the degree of

Doctor of Philosophy

in

Molecular and Cell Biology

in the

Graduate Division

of the

University of California, Berkeley

Committee in charge:

Professor David Bilder, Chair Professor Iswar Hariharan Professor David Drubin Professor Kathleen Ryan

Fall 2010

Page 2: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

Regulation of cellular signaling pathways by endocytosis and protein degradation

© 2010

by

Sarah Lynn Windler

Page 3: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

1

Abstract

Regulation of cellular signaling pathways by endocytosis and protein degradation

by

Sarah Lynn Windler

Doctor of Philosophy in Molecular and Cell Biology

University of California, Berkeley

Professor David Bilder, Chair

The defined shapes and sizes of the diverse tissues that comprise multicellular organisms are intimately linked to their specialized functions. The plethora of known cellular behaviors and characteristics that contribute to tissue function and development comes in large part from any given cell’s ability to receive and respond to signals from its environment. These signals are often received via transmembrane protein receptors that interpret extracellular signals and transmit them inside the cell to effect cellular changes. Cellular signaling pathways influence cellular shape, proliferative capacity, cell fate, cell movement and many other traits that are crucial for proper cellular and tissue function both during development and over the lifetime of an animal. Therefore, understanding the mechanisms by which cells regulate these signals is a central question in biology. Drosophila is an ideal model organism in which to study the regulation of cellular signaling pathways. In addition to the ease of genetic manipulation, Drosophila contains epithelial tissues comparable to those in mammals, which have defined apico-basal polarity that is essential for tissue function. Cell polarity signaling pathways in epithelial cells can be studied by the isolation of neoplastic tumor suppressor genes (TSGs) from genetic screens. Drosophila neoplastic TSGs are genes whose functions are absolutely required for proper apico-basal polarity and the regulation of epithelial cell proliferation, though the underlying mechanisms of this regulation and the links between polarity and proliferation remain unknown. In addition to studying how these genes regulate cellular polarity, the identification of certain classes of neoplastic TSGs, such as the endocytic regulators, has given us tools to investigate the regulation of other developmentally important signaling pathways.

A number of cellular signaling pathways are regulated by endocytosis, which is commonly thought to play a role in the attenuation of signaling, but recently appreciated to provide a much more complex method of regulation that may in some contexts promote signaling as well. In general, the roles of both cargo internalization routes and subsequent trafficking down a degradative or recycling pathway in the regulation of cellular signaling pathways are not understood. In Chapter 2 I investigate the role of endocytosis on both trafficking and signaling regulation of the Notch/Delta pathway in Drosophila. I address the roles of specific components of the endocytic internalization machinery in the endocytosis of Notch and Delta, and define whether distinct paths of entry into the cell impact Notch signaling. In Chapter 3, I explore how endocytic trafficking regulates epithelial cell polarity and identify

Page 4: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

2

links between the endocytic and junctional scaffold classes of tumor suppressors. In particular, I investigate the possibilities that a common underlying mechanism of tumor suppression exists for these different classes.

Another mechanism through which cells regulate signaling pathways is the ubiquitin-proteasome system. In Drosophila, one well-known example of this is the negative regulation of Hedgehog and Wingless signaling pathways via the ubiquitylation of downstream effectors by the E3 ubiquitin ligase SCFSlmb. In a screen for new neoplastic TSGs, a component of this E3 ubiquitin ligase was recovered along with many endocytic regulators. In Chapter 4, I detail my unexpected discovery that slmb regulates the epithelial cell polarity signaling pathway. I further describe my progress toward finding a presumed polarity-regulating Slmb substrate via a genetic interaction screen.

In sum, my graduate work sheds new light on the ways in which basic cell biological mechanisms, including endocytosis and regulated proteolysis, regulate specific signaling pathways to control cell fate, proliferation, and polarity.

Page 5: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

i

Table of Contents

Abstract…………………………………………………………………………………………...1 Table of Contents…………………………………………………………………………………i List of Figures…………………………………………………………………………………....ii List of Tables…………………………………………………………………………………….iv Acknowledgments………………………………………………………………………………..v Chapter 1: Introduction…………………………………………………………………………1 Chapter 2: Endocytic regulation routes required for Delta/Notch signaling……………….11 Abstract…………………………………………………………………………………..12 Introduction………………………………………………………………………………13 Results……………………………………………………………………………………14 Discussion………………………………………………………………………………..18 Materials and Methods…………………………………………………………………...20 Figures……………………………………………………………………………………21 Chapter 3: Evidence for regulation of endocytic traffic by junctional scaffold neoplastic tumor suppressors……..………………………………………………………………………..35 Abstract…………………………………………………………………………………..36 Introduction………………………………………………………………………………37 Results……………………………………………………………………………………39 Discussion………………………………………………………………………………..43 Materials and Methods…………………………………………………………………...46 Figures……………………………………………………………………………………48 Chapter 4: Characterization of an E3 ubiquitin ligase component, supernumerary limbs, as a tumor suppressor and polarity regulator…………………………………………………...60 Abstract…………………………………………………………………………………..61 Introduction………………………………………………………………………………62 Results……………………………………………………………………………………63 Discussion………………………………………………………………………………..68 Materials and Methods…………………………………………………………………...71 Figures and Tables……………………………………………………………………….73 References……………………………………………………………………………………...123

Page 6: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

ii

List of Figures

Figure 2.1……New MENE Mutants Disrupt Internalization from the Cell Surface………21 Figure 2.2……Identification of Null Mutations in Cell Surface Endocytic Regulators.…...23

Figure 2.3……Cargo Trafficking Phenotypes are Distinct in Cells with Disrupted

ADE, CDE, or DDE…………………………………………………………….25 Figure 2.4……Notch traffics distinctly in mutants that disrupt the internalization step of

endocytosis……………………………………………………………………...27 Figure 2.5……Endocytic Requirements for Delta/Notch Signaling in Signal-Receiving and

Signal-Sending Cells……………………..…………………………………..…29 Figure 2.6……Delta localization in mutant GLCs……………………………………….…..31 Figure 2.7……Delta signal-sending is not impaired in GLCs mutant for Rab11

Effectors………………………………………………………………………...33 Figure 3.1……AP-2, chc, and shi are neoplastic TSGs……………….……………………...48 Figure 3.2……Polarity defects in endocytic TSG mutant cells……….……………………..50 Figure 3.3……Subcellular localization of normally polarized proteins in endocytic

mutant tissue……………………………………………………………………52 Figure 3.4……Junctional scaffold neoplastic TSG mutant discs display

AP-2-dependent endocytosis…………………………………………………...54 Figure 3.5……Defects in hTfR trafficking in dlg mutants are suggestive of a recycling

pathway defect in junctional-scaffold nTSG mutants………………………..56 Figure 3.6……Neoplastic TSGs do not control polarity and proliferation by

regulating Crb levels…………………………………………………………....58 Figure 4.1……Identification of slmb as a tumor suppressor………………………………...73 Figure 4.2……Apico-basal polarity is disrupted in slmb mutant tissue…………………….75 Figure 4.3……Slmb mutant clone survival ability depends upon allele…………………….77 Figure 4.4……Slmb mutant discs display ectopic JNK pathway signaling as seen in other

neoplastic tumor suppressor mutant discs……………………………………79 Figure 4.5……Slmb mutant discs display ectopic JAK/STAT signaling…………………....81

Page 7: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

iii

Figure 4.6……Discs mutant for other components of the SCF complex partially phenocopy slmb mutant disc neoplastic transformation…………….….........83

Figure 4.7……Co-overexpression of stabilized forms of Ci and Arm in the

developing wing disc is insufficient to cause neoplastic transformation…….85 Figure 4.8……Known substrate Arm accumulates in slmb mutant cells, but

candidate substrate aPKC does not…………………………………………...87 Figure 4.9……Slmb knockdown in the wing causes a visible phenotype that is

rescued by expression of wild type slmb………………………………………89 Figure 4.10….Slmb knockdown phenotype is enhanced by reduction of SCF

complex member function……………………………………………………...91 Figure 4.11….Increasing the efficiency of slmb knockdown in the wing disc

causes neoplastic transformation………………………………………………93 Figure 4.12….The slmb knockdown wing phenotype can be suppressed or enhanced…….95

Page 8: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

iv

List of Tables

Table 4.1……Results of Slmb Dominant Modifier Screen on Chromosome II…….………97 Table 4.2……Results of Slmb Dominant Modifier Screen on Chromosome III…………..102 Table 4.3……Deficiency mapping 3rd chromosome suppressor of slmb wing

knockdown phenotype narrows suppressor region down to 72C1-72D1….106 Table 4.4……Testing candidates in 72C1-72D1 cytological region fails to identify a

suppressor of the slmb knockdown wing phenotype………………………...108 Table 4.5……Deficiency mapping 3rd chromosome lethal enhancer of slmb wing

knockdown phenotype narrows enhancer region down to cytological band 95B1…………………………………………………………110

Table 4.6……Results of testing candidates uncovered by Df(2L)BSC354 for lethal

enhancement of the slmb knockdown wing phenotype……………………...112 Table 4.7……Results of testing candidates of interest that may play a role in the slmb

polarity pathway………………………………………………………………114 Table 4.8……Results of testing candidates uncovered by enhancer Df(1)Exel6227……...117 Table 4.9……High priority slmb substrate candidates from screens and bioinformatic

work……………………………………………………………………………119

Page 9: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

v

Acknowledgments I would first like to express my deepest appreciation for David’s mentorship during my time in his lab. Over ten years ago, pushing flies in a genetics class, I never would have dreamed that I would willingly choose to do my graduate work in a Drosophila genetics lab. Thanks to David’s patience in teaching and his passion for the research, I have grown to love fruit flies and have a vast appreciation now for the amazing genetic tools that are available to address fundamental questions in biology. David’s approach to answering scientific questions and his scientific curiosity have been both instructive and inspiring and have shaped me, for the better, as a scientist. His ability to teach and gently correct in a constructive manner have made me a better researcher and writer. Finally, David’s basic kindness and understanding during both good and difficult times have conveyed to me that he truly cares about his lab members as people, not just workers. Taken together, the afore-mentioned traits add up to make an ideal scientific mentor to and for whom I am extremely grateful. I would like to thank my committee members, Iswar Hariharan, David Drubin, and Kathleen Ryan, who have all helped me to progress as a researcher. They have contributed invaluable scientific advice and have given freely of their time to help me. It is hard to know where to start when thanking the members of the Bilder Lab for making my time here as a graduate student so much more than just work! I have been truly lucky to be surrounded by such a diverse group of fun and intelligent people. Thanks to former post-docs Thomas Vaccari and Sally Horne-Badovinac for their positive, helpful attitudes and sage scientific advice. Thanks to Han Lu and Jen Zeitler, fellow graduate students who paved the way and who continue to offer advice and help whenever I need it. Thanks to former technician Laurent Menut for keeping the lab running so smoothly that I never had to worry about anything but my research, and for providing a great fly-pushing soundtrack. Thanks to current post-docs Lucy O’Brien, Anne Classen, and Lara Skwarek for their encouragement and for sharing their wisdom on matters both personal and professional. Thanks to my fellow graduate students Saori Haigo, Brandon Bunker, and Alejandra Figueroa Clarevega for stimulating conversations both scientific and not-so-scientific, as well as their support. Thanks to my fellow graduate student and excellent baymate, Holly Morrison, for showing me how it’s done (though setting the bar really high), and for support, advice, and encouragement over the years. Thanks to technician Josh Schoenfeld for all of his hard work on our collaborative scientific endeavors and to Lupe Coy, without whom there would be no flies to study. Thanks, also, to those who have taught me how to be a better mentor, former rotation students, David Steakley and Brock Roberts, and REU summer student Meenakshi Prajapati. The Bilder Lab just wouldn’t be the same without our neighbors in the Hariharan Lab. Thanks to Iswar and the members of the Hariharan Lab for sharing their flies, reagents, and advice. Thanks especially to fellow graduate student and friend, Abigail Gerhold, for many impromptu gab sessions, both about science and life, and for always being there to lend an ear and some sensible advice. My interest in research first blossomed in Anthony Otsuka’s lab at Illinois State University. Without him and another outstanding teacher and mentor, Wade Nichols, I don’t know that I would have ended up doing basic science research. With their encouragement, I pursued a Masters degree also at ISU in the lab of Laura Vogel, who I thank for being an excellent role model and mentor. Thanks to Anjen Chenn who gave me my first job as a Research Associate in his lab at Northwestern (where I generated my first scientific publication)

Page 10: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

vi

and who encouraged me in my decision to go back to school for a PhD. Thanks to Greg Woodhead, a former member of the Chenn Lab, who planted the idea in my head of applying to UC Berkeley. Many thanks to Michael Hart, who dropped everything to move across the country with me so that I could pursue my dream. Thanks to Kristin Scott and David Drubin for their advice and instruction and welcoming me into their labs for my first-year rotations. Without my parents’ obvious respect for and appreciation of the natural world I don’t know that I would have ever pursued a career in biology. Many thanks to them for instilling in me a curiosity about and an appreciation for nature, as well as their support, both moral and financial, throughout my many years in school. Thanks to the rest of my family, including my grandmothers, who never fail to let me know how proud they are of me, my sister Jenny, who probably thinks it’s a little crazy that I’m still in school but supports me nonetheless and who I can always count on for some goofy fun, Sam, Jane, and Melanie for their support and for generally being amazing siblings, and to Anne, who helped to raise me and who I know I can always count on for hugs and a sympathetic ear. Finally, to my best friend and amazing partner, Jeremy, I owe tremendous thanks. Thank you for choosing to share the adventure of life with me. Thanks for your endless patience with stressful late nights as I tried to finish writing or preparing a presentation. Thank you for actually being interested in the work that I do. Thank you for the feeling of security that someone is always there for me, on my side. Finally, thank you for making the last few hectic years some of the happiest of my life.

Page 11: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

1

Chapter 1

Introduction

Page 12: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

2

Cells receive and regulate signals from their environment A cell’s ability to function relies upon its ability to integrate information from its surroundings

The cells that comprise multicellular organisms must be capable of communication with their environment in order to perform functions crucial to the development and survival of the organism. Many types of cues, such as global nutritional, hormonal, and diffusible secreted protein cues or cell-cell contact cues, must be received from the environment, communicated across the highly selective barrier of the plasma membrane, and transduced into the cell for an appropriate response to the information. For example, the ability of cells to move in a directed manner in concert relies upon both long-range diffusible signals and contacts between the migrating cells. A cell’s interpretation of its position with respect to surrounding cells in a tissue may allow it to keep dividing in a regulated manner or may constrain its ability to continue proliferating by causing it to differentiate. Occasionally, cells arise that can no longer interpret cues from their surroundings. A cell that is insensitive to its surroundings and the normal constraints dictated by its resident tissue may act in an aberrant manner, such as is seen in overproliferating and metastatic cancer cells. Therefore, with respect to development and disease, it is critical to understand the numerous ways in which cells receive and regulate incoming information. How do cells translate information from outside to inside? Cells have solved the problem of interpreting and acting on information from the environment in part by the use of sensors that span the plasma membrane, with both extracellular and intracellular domains. The classical view is that these transmembrane receptor proteins bind cues in the extracellular space, often resulting in conformational changes or dimerization, that then impact the activity of the intracellular domains as well. These changes perpetuate signaling downstream of the receptor and can impact transcriptional regulation, protein stability, changes in the lipid composition of membranes, or reorganization of the cytoskeleton, to name but a few potential outcomes of cellular signaling events. Though perhaps less dynamic than the classical view of receptor signaling pathways, a cell’s maintenance of its size, shape and function within a given tissue might also be considered a translation of information from outside to inside. In epithelial cells, transmembrane proteins on one cell interact with those of its neighbor to maintain tissue integrity. Cell-cell adhesion and other signaling, such as interactions between receptors and the extracellular matrix, are also tightly linked to proper cell polarity. How do cells solve the problem of signaling regulation? Given that the sensory system of any cell contains a multitude of different transmembrane proteins, along with the potential for activation of multiple signaling pathways at any given time, much of the process of signaling regulation is still a mystery. In any given cell signaling pathway, the receptor that spans the boundary of the cell is a crucial component of the pathway and therefore a major focus of regulation. In order for a cell to receive a signal, its receptor must first be made, so transcriptional and translational regulation leading to the availability of the receptor itself or other components of its signaling pathway are a first layer of regulation. The receptor must also be targeted to the right place, meaning that another layer of

Page 13: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

3

regulation is the polarized biosynthetic trafficking of a receptor to a position in the membrane where it is poised to bind its ligand. Some receptors may require co-factors to make them signaling-competent or to keep them ‘turned off’. Post-translational modifications may also regulate the receptor’s availability at the cell surface, making it more or less accessible to its ligand.

A number of exciting discoveries over the last several decades have implicated endocytosis as a key cellular process that plays important roles in the regulation of cellular signaling pathways. Endocytosis at its most basic level involves the internalization of molecules from outside of the cell, along with the cell membrane and its resident proteins. More than that, though, endocytic uptake can down-regulate and silence activated signaling receptors or, conversely, facilitate their activation at specialized internal compartments. Generally, endocytosis has been shown to play a role in important biological processes such as cellular polarization, synaptic transmission, cell migration, and immunity to name a few. In a number of these processes, the role endocytosis plays is that of cell signaling regulator, but the diverse mechanisms by which it may fulfill this role are just beginning to be uncovered. My thesis has used the genetic tools of Drosophila to tackle the question of how endocytosis regulates particular cell signaling pathways. In the following two chapters I will discuss first, the role of components of the endocytic pathway in the regulation of the trafficking and signaling of the cell-fate signaling receptor, Notch, and its ligand Delta, and second, the relationship between endocytosis and the pathways regulating epithelial cell polarity. Pathways of entry into the cell AP-2 adaptor, Clathrin, and Dynamin are required for receptor-mediated endocytosis

In order to investigate how endocytosis regulates signaling, it is important to understand how transmembrane proteins are generally internalized. The simplified and classical view of receptor-mediated endocytosis is as follows: Upon ligand-binding, transmembrane receptors undergo modifications or conformational changes that allow for recognition by endocytic adaptor proteins, such as the AP-2 complex. Clathrin triskelions are then recruited to the site of internalization via the adaptor complexes to form a clathrin-coated pit. In the final step of internalization, the large GTPase Dynamin is recruited to the clathrin-coated pits where it forms helical filaments around the invaginating membrane to drive the vesicles inwards before pinching them off to form clathrin-coated vesicles. In many types of cells, actin polymerization has also been shown to play a crucial role during the internalization of newly-formed vesicles (reviewed in [1]). Upon internalization, Clathrin is removed from the vesicle during an uncoating step that relies upon the activity of Auxilin and the ATPase heat-shock chaperone protein Hsc70 (reviewed in [2]). After uncoating, the vesicles can fuse with each other and the early endosomal compartment via the activity of several proteins that function in tethering and vesicle fusion. From these initial steps, internalized cargo can be trafficked in different ways. If a receptor is to be returned to the membrane upon dissociation from its ligand, it will be shunted to the recycling pathway. If the receptor is to be destroyed, it will pass from the early endosomal compartment to a late endosomal compartment where it is then sorted, via complexes that recognize its ubiquitin tag, into internal vesicles of the multivesicular body (MVB). The MVB then fuses with the lysosome and internalized cargo is degraded (reviewed in [3]).

Page 14: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

4

Dynamin plays a more universal role in endocytosis than perhaps any other protein, as demonstrated by the fact that it is absolutely required for Clathrin-mediated endocytosis (CME) and is also required for some forms of non-CME (discussed below and reviewed in [4]). Dynamin was initially described as a microtubule-interacting protein, and has since been proposed to play a role in numerous cellular processes, but is most well-known for its role in the “pinching off” of invaginating vesicles from the plasma membrane (reviewed in [5]). Electron microscopy (EM) work on rat nerve terminals, where a high rate of endocytic turnover occurs, demonstrated that in the presence of a non-hydrolyzable GTP analog, vesicles invaginating from the plasma membrane develop long necks that are not pinched off. In addition, electron-dense ring structures are associated with these necks, suggesting that the hydrolysis of GTP by polymerized Dynamin at sites of internalization plays an important role in vesicle scission [6].

Clathrin coats were observed for the first time when EM was used to image yolk protein accumulation in mosquito oocytes [7]. The unique vesicles that were observed were referred to as ‘bristle-coated’ and it was speculated that these vesicles went on to lose their coats and fuse to form the yolk granule, the oocyte’s specialized endosome. Clathrin was first partially purified from coated intracellular vesicles isolated from pig brains, where it was found to be the dominant protein species associated with the vesicles [8]. Clathrin is a heterodimer composed of a heavy chain and a light chain. Three such heterodimers bind together to form a clathrin triskelion which is the basic repeating unit that polymerizes to form an organized latticework on invaginating membranes which is now known as the clathrin coat (reviewed in [9]). In addition to its well-known role in endocytic internalization, clathrin also plays roles in Golgi transport and in sorting of proteins at the early endosome [10,11]. The basic composition of an endocytic clathrin-coated pit, from outermost to most intracellular, includes the plasma membrane and its associated transmembrane cargo, endocytic adaptor proteins and clathrin [12].

The AP-2 adaptor complex has been best studied in mammalian cells, where AP-2 binding motifs in the cytosolic tails of many transmembrane proteins have been elucidated. AP-2 is a heterotetrameric complex thought to be a core component of the machinery involved in the formation of clathrin-coated vesicles. By binding to both endocytic cargo and lipids that are enriched at sites of internalization, AP-2 recruits clathrin triskelions and stabilizes clathrin-coated pits. Biochemical purification of AP-2 has allowed for crystallization studies, as well as insights into what proteins AP-2 interacts with inside the cell. Many cargoes require AP-2 for their internalization, either by binding directly to an AP-2 subunit or indirectly through binding to clathrin-associated sorting proteins (CLASPs) which also bind to AP-2. Unlike Dynamin and clathrin, which have roles distinct from endocytosis, all available evidence suggests that AP-2 functions exclusively in an endocytic capacity.

Clathrin-independent mechanisms of endocytosis

In addition to CME, it has become apparent in recent years that there are a variety of

other mechanisms by which endocytic cargo may enter the cell (reviewed in [13]). A brief description of non-CME will be given here, as it also plays important roles in the regulation of signaling pathways. Understanding of the mechanisms governing non-CME is far from complete, as classifications can be made based upon the morphology of carrier vesicles, pathway-specific cargoes, proteins required for internalization, dependence upon cholesterol, destinations upon trafficking, and proposed function (reviewed in [14]). The most basic definition uniting these pathways is that Clathrin is not required. Perhaps the most well-studied

Page 15: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

5

non-CME pathway is that of Caveolin-mediated endocytosis. Caveolae were first observed by EM and morphologically appear as flask-like invaginations in the plasma membrane. Caveolin-mediated endocytosis occurs at a slower rate than CME, is dependent upon cholesterol and Caveolin, and requires the activity of Dynamin.

Clathrin-independent carriers (CLICs) are non-coated tubular plasma membrane extensions that are sensitive to cholesterol depletion and were first described with respect to the mechanism of cell entry of the cholera toxin binding subunit [15]. They were also shown to be enriched in glycosylphosphatidylinosital-anchored proteins (GPI-APs) and fluid phase markers and together with their target GPI-AP enriched early endosomal compartments (GEEC) form the CLIC/GEEC endocytic pathway. CLIC/GEEC endocytosis relies upon Arf1 and Cdc42, is Dynamin-independent, and may require actin for scission from the plasma membrane. Other non-CME pathways include those regulated by Arf6 and by RhoA (reviewed in [16]).

Clathrin-dependent endocytosis in Drosophila The major components of the clathrin-mediated endocytic pathway have single homologs in Drosophila, where genetic experiments have been done to better understand their functions in CME within the context of intact tissues. Elegant work in Drosophila allowed researchers to demonstrate a role for Shibire (later identified as the single fly homolog of Dynamin) in endocytosis using a temperature-sensitive mutation. Shifting flies to the restrictive temperature resulted in an immediate paralysis that was reversed upon a shift back to the permissive temperature. Neurotransmission requires very rapid endocytic recycling and EM of the paralyzed fly neurons revealed CME intermediate structures as well as a depletion of synaptic vesicles suggesting that shibire plays a role in endocytosis [17] . Mutations in fly Chc were originally characterized as cell lethal, as one might expect for a protein that is thought to be crucial for both intracellular trafficking as well as many endocytic events [18]. The first AP-2 mutant in Drosophila was isolated in 1997 in work that demonstrated AP-2’s requirement for Clathrin- and Dynamin-dependent endocytosis in the CNS of the fly[19]. Since that time, other studies in Drosophila have demonstrated the importance of AP-2 in endocytic events in a number of different cellular contexts [20-22]. These data, in addition to the finding that AP-2 is expressed in many tissue types during Drosophila development [23] are suggestive that AP-2 may play a similar crucial role in CME in Drosophila as it is thought to in mammalian cells, but whether some cargo can be internalized in an AP-2-independent manner is unknown. Clathrin-independent pathways in Drosophila

Our understanding of non-CME pathways is almost entirely informed by work done in

mammalian cells, and little is known about whether these pathways are important, or even exist, in Drosophila. The Caveolin-mediated pathway is not thought to exist in flies, based upon the lack of any Caveolin homologs in the Drosophila genome, no evidence of caveolae in EM studies of fly cells, and finally the fact that cholesterol, which is essential for caveolae formation, is not present in fly cell plasma membranes. However, extensive work in fly hemocytes has demonstrated the presence of non-CME pathways using the temperature sensitive allele of shibire [24]. In addition, the existence of a GEEC pathway in cultured Drosophila S2 cells has also been described [25]. The existence of multiple pathways of entry into the cell in Drosophila, coupled with the wealth of genetic and cell biological tools available to study these

Page 16: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

6

processes, makes it an ideal organism for investigating how varying mechanisms of internalization regulate cellular signaling pathways. Endocytic regulation of signaling pathways

Endocytic regulation of receptor tyrosine kinase signaling

The endocytic regulation of cell signaling pathways has classically been associated with the downregulation of activated receptors via their removal from the cell surface, thereby making them inaccessible to further ligand interaction and promoting their degradation in the lysosome. Helping to further strengthen the paradigm for endocytosis playing a role in down-regulation of receptor tyrosine kinase (RTK) signaling, it was demonstrated in 1990 that a non-internalizible form of the Epidermal Growth Factor Receptor (EGFR) could cause excess signaling and transformation of mammalian cells even in the presence of low levels of ligand [26]. It was postulated that endocytic mutations that prevented growth factor pathway signaling attenuation could lead to neoplastic transformation. The regulation of activated RTKs via endocytosis has since been studied extensively due to its now established role in carcinogenesis (reviewed in [27-30]). However, in recent years, many studies have demonstrated that endocytic trafficking can play an important role in termination or propagation of receptor signaling (reviewed in [31-34]).

Studies on EGFR have been instrumental in bringing to light just how intricate and complex a role endocytic trafficking can play in the regulation of receptor signaling. From the earliest studies that introduced the paradigm of RTK internalization facilitating the attenuation of signal[35], other studies have since demonstrated that RTKs can also signal from the endosome [36], and that where signaling occurs influences which downstream effectors are stimulated [37]. In addition, other work has demonstrated that ligand concentration influences the mechanism by which EGFR is internalized from the cell surface, and that the mechanism of internalization determines whether it will be degraded or recycled. The surprising finding was that CME is actually required for sustained EGFR signaling via recycling of the receptor, whereas clathrin-independent endocytosis of the receptor commits it to a degradative pathway [38,39]. Further complexity is revealed by a recent study in endosomal sorting mutants in which differential effects on RTK signaling were seen depending on signaling pathway and cell type [40].

Endocytic regulation of other developmentally-important signaling pathways

The findings on endocytic regulation of RTK signaling demonstrate the diversity that also

exists with respect to other signaling pathways. Ligand-bound TGFβ receptors have been found to continue signaling upon CME into endosomes, whereas internalization via non-CME leads to degradation [41]. In addition, these receptors are constitutively recycled when not occupied by ligand, and perhaps predictably considering their wide range of signaling outputs, are differentially regulated by endocytosis depending on cell type [42]. Similarly, both canonical and non-canonical Wnt signaling pathways have been shown to be regulated positively and negatively by endocytosis, and by both CME and non-CME (reviewed in [43].

Endocytic regulation of signaling pathways in Drosophila

Page 17: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

7

Drosophila has proven to be an invaluable system in which to study endocytic regulation of signaling pathways due to the availability of endocytic mutants as well as reliable in vivo readouts of the activation of well-characterized signaling pathways. The complexity of endocytic regulation of signaling seen in mammalian cells also extends to Drosophila . One recent study showed that Wg and its receptors, Arrow and Dvl, co-localize to and signal from the early endosome [44], whereas another showed that endocytic internalization of Wg and its receptors, Arrow and DFz2, results in inhibition of signaling, though whether this occurs during earlier trafficking steps or upon arrival to the lysosome depends upon developmental context [45]. Another signaling pathway regulated by endocytosis is the JAK/STAT pathway, which in Drosophila, can be activated by the binding of the ligand Unpaired 1 (Upd1) to the Domeless receptor. Internalization and trafficking of ligand-bound receptor toward the lysosomal pathway was shown to allow for its transient activation, thus keeping a tight temporal control over activation of the pathway [46].

Endocytic regulation of the Notch signaling pathway

The role of endocytic regulation in Notch signaling is a particularly exciting area of

research with a number of as yet unanswered questions. Though much of what is known has come from studies in Drosophila, the pathway and its endocytic regulation appear to be highly conserved in mammalian cells, as well. The Notch signaling pathway plays important roles throughout development in patterning and cell-fate specification, and its misregulation is often associated with disease states. The Notch receptor is a heterodimeric transmembrane protein that, upon binding to its ligand Delta or Serrate on an adjacent cell, undergoes a series of cleavage events that ultimately ends in the release of a small intracellular peptide (NICD) which translocates to the nucleus to regulate the transcription of Notch target genes. Notch and its ligands are subject to post-translational modifications, and their regulation is intimately tied to their appropriate trafficking, as well as that of other players in the pathway. In addition to polarized membrane targeting, endocytosis of both the receptor and ligand also plays a very important role in activation of the pathway (reviewed in [47]), though the details of why remain unclear. What is known is that blocks early in the endocytic pathway in either the signal-sending or signal-receiving cell lead to blocks in Notch signaling. In Drosophila epithelia, mutations that prevent biogenesis of early endosomes or that block earlier endocytic events lead to an increase in Notch levels and a block in Notch signaling whereas increased Notch trafficking or endocytic blocks later in the pathway lead to ectopic signaling [48,49]. It has been previously reported that internalization of Notch requires both Dynamin and Clathrin, but whether the AP-2 adaptor plays a role in internalization or signaling is unknown. I report my work addressing the AP-2-dependence of Notch and Delta internalization and signaling in Chapter 2. The Notch ligand Delta has been particularly well-studied with respect to its endocytic requirement in signal-sending. Some studies suggest that Delta must be internalized because it must transit through the recycling pathway, the so-called ‘recycling model’. There are a number of reasons why Delta might need to be recycled before being competent to send signal. It may undergo modification, clustering, or binding with co-factors that enhance its signaling capability. It may need to be relocalized to a different region of the plasma membrane where it would be more closely juxtaposed with Notch receptors on an adjacent cell membrane. Alternatively, the ‘pulling force’ model posits that the Notch ligand must be internalized in order to generate a pulling force on Notch. This pulling force causes a conformational change in Notch which

Page 18: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

8

exposes the ADAM cleavage site, allowing for shedding of the extracellular domain and subsequent signaling. Distinguishing between the recycling and pulling force models of Delta’s endocytic signaling requirement is of great interest in the field and I present my findings on this question in Chapter 2.

Endocytosis and cell polarity pathways Cell polarity as a signaling pathway

Polarity is an essential attribute of many cell types, including epithelial cells. Epithelial cells form monolayered tissues, with the apical surface typically facing a lumen, cellular junctions along the lateral surface serving as a barrier between apical and basal surfaces, and the basolateral surface contacting a basement membrane. Apicobasal polarity refers to the segregation of continuous tracts of plasma membrane into particular domains where both transmembrane and cortically-associated proteins are restricted. In epithelial cells, apicobasal polarity is required for proper cellular function and appropriate regulation of proliferation (reviewed in [50]). In order to polarize appropriately, cells must rely upon a polarity signaling pathway. A cell must be able to translate the positional cues from its environment, such as contact with the extracellular matrix via a transmembrane receptor, into actions inside the cell that distinguish apical from basal identity, such as the targeting of proteins to specific regions of the plasma membrane. The signaling pathways that generate and maintain cellular polarity are incompletely understood, but some key downstream players have been intensely studied. Several protein modules regulate apicobasal polarity in epithelial cells

While many important discoveries about epithelial cell polarity have been made using

mammalian cell culture, I will focus here on work that has elucidated the role of various apicobasal polarity complexes in the context of developing epithelial tissues in Drosophila, though the mechanisms and players are thought to be largely conserved between the two systems. Briefly, in Drosophila embryonic epithelia, the first known requirement for distinguishing apical from basolateral surfaces is the proper apical localization of Bazooka (Baz), perhaps by apical actin and apicobasal microtubules, which functions as a cue for the formation of adherens junctions. This starts a cascade through which other apical components, such as the transmembrane protein and apical determinant Crumbs and its partners Stardust and PatJ, as well as aPKC, Par6 and Cdc42, are localized just apically of Baz (reviewed in [51]). Along the basolateral cortex, just basal to the zonula adherens at the septate junction, the scaffolding proteins Scribble (Scrib) and Discs Large (Dlg) co-localize and functionally interact with each other along with Lethal Giant Larvae (Lgl) which localizes subcortically throughout the cell but is excluded from the apical surface by the activity of aPKC. This Scrib/Dlg/Lgl module also excludes apically-localized proteins from the basolateral surface, and loss of any member of the complex leads to loss of polarity and overproliferation (reviewed in [52]). The signaling events that drive apicobasal polarity, as well as the mechanisms that regulate them, are of great interest in the field. Endocytosis regulates epithelial cell polarity

Page 19: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

9

Unknown components of the epithelial polarity pathway in Drosophila have been

discovered by exploiting two observations. First, loss of function of the basolaterally-localized polarity complex members leads to overproliferation of epithelial tissues in the developing larvae; and second, loss of polarity and overproliferation in even one set of epithelial tissues, such as the eye-antennal imaginal discs, is sufficient to kill the animal before metamorphosis and eclosion. A screen was performed for Drosophila polarity regulators/neoplastic tumor suppressors using these criteria, and this Mutant Eye No Eclosion (MENE) screen uncovered a number of novel tumor suppressor genes [53]. Among these were components of the endocytic pathway. Some of these function at the level of early endosome fusion, such as Avl, Rab5, Vps45 and Rabenosyn [48,54], and others function later in the degradative pathway to sort proteins to the lysosome, such as Vps25, a member of the ESCRT-II complex [55]. While several phenotypes of the ESCRT mutants, such as non-cell autonomous overgrowth, have been explained by the trapping and activation of Notch and its associated downstream effects, the cause of the cell-autonomous phenotypes of apicobasal polarity misregulation in the endocytic mutants is still incompletely understood. Determining whether blocking other steps of endocytosis, including specific internalization pathways, causes polarity disruption and neoplastic transformation may provide clues as to how endocytosis regulates polarity. How are endocytosis and epithelial cell polarity linked? At the most basic level, a block in endocytosis prevents proteins from being taken into the cell and trafficked appropriately. While it is possible that cellular overproliferation in endocytic mutants may be caused by their inability to attenuate active signaling pathway receptors that stimulate proliferation, another mechanism must be invoked for the loss of cell polarity, as constitutively active receptors that drive proliferation do not cause defects in polarity. An attractive candidate mechanism for polarity disruption in endocytic mutants would be the accumulation and expanded localization of a polarity-regulating protein. In a study on the early endosomal protein and tumor suppressor avl, it was found that overexpression of the apical polarity determinant Crb, or even its intracellular domain, in epithelial tissue was sufficient to drive ‘apicalization’ and neoplastic overgrowth of the tissue [48]. Because Crb appeared to accumulate at the cell surface in avl mutant cells, an appealing hypothesis was that the loss of polarity and overproliferation of the endocytic mutants was due to an inability to appropriately regulate Crb levels at the plasma membrane. As a proxy for decreasing Crb-pathway apical polarity signaling, several groups have demonstrated the ability of a dominant negative form of aPKC to suppress polarity and/or proliferation defects in endocytic and junctional scaffold tumor suppressor mutant imaginal discs [48,56,57]. However, the role of Crb in the neoplastic phenotypes of endocytic tumor suppressor mutants has not been directly tested. I specifically address this outstanding question in the field in Chapter 3. Aside from the appealing hypothesis that endocytosis regulates polarity pathways, it is formally possible that proteins associated with apicobasal polarity complexes may have a role in regulating endocytosis in a polarized manner. Several groups have shown that loss of function of apically-associated polarity proteins can alter the endocytic itinerary of the junctional transmembrane protein, E-cadherin (E-cad), which is important in the maintenance of epithelial architecture [58-60]. In addition to this work, a screen in C. elegans also revealed a role for the

Page 20: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

10

Par complex and Cdc42 in endocytic regulation [61]. Chapter 3 of this thesis is largely devoted to further exploration of the links between epithelial cell polarity and endocytosis. Summary A more detailed understanding of how endocytosis regulates cell signaling pathways will greatly benefit many areas of research, both with respect to the development of multicellular organisms, as well as disease states that arise when these pathways become mis-regulated. In the following chapters I present the work that I have done to elucidate the mechanisms of this regulation. In Chapter 2, I detail the roles of both cell-surface endocytic regulators and downstream endocytic components in regulating both the trafficking and signaling capabilities of the receptor Notch and its ligand Delta during development. In Chapter 3, I describe the finding that cell-surface regulators of endocytosis act as tumor suppressors, and go on to determine whether the neoplastic phenotypes seen in both endocytic and junctional scaffold tumor suppressors are superficially related or whether there is a common underlying cause for the disruption of polarity and proliferation in both classes of mutants. Finally, in Chapter 4, I discuss the work that I have done to characterize another tumor suppressor gene, supernumerary limbs (slmb), that regulates cell signaling by yet another mechanism, the degradation of proteins via the proteasome. Overall, my graduate work makes significant contributions to our understanding of the endocytic regulation of cellular signaling pathways, as well as bringing to light a novel mechanism by which cellular polarity pathways may be regulated.

Page 21: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

11

The following Chapter was originally published as a Report in Current Biology 2010 Mar 23;20(6):538-43

Chapter 2

Endocytic internalization routes required for Delta/Notch signaling

Page 22: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

12

Abstract

The internalization of transmembrane receptors from the cell surface plays a central role in signal regulation. A large body of literature has shown that receptor internalization can occur through different routes [13]; however, because of the difficulty in selectively blocking these routes in vivo, their roles in signaling are poorly understood. Here we investigate this question using null mutations in Drosophila Dynamin, Clathrin, and AP-2 adaptor subunits to analyze internalization requirements for the Delta ligand and its receptor Notch. Bulk Notch is internalized via AP-2-dependent endocytosis, but signaling by Notch requires AP-2-independent Clathrin-dependent endocytosis, highlighting a distinction between Notch endocytic routes required for degradation versus signaling activation. Signaling by the Notch ligand Delta has been shown to require Dynamin, but whether this generates a pulling force of Delta on Notch or allows for Delta entry into a recycling pathway to gain signaling competence is widely debated [62,63]. Surprisingly, we show that signaling by Delta in germline cells can occur by Clathrin-independent endocytosis, when endosomal entry is blocked, and when activity of Rab11 or its effectors is reduced, suggesting that Delta need not pass through a recognized recycling pathway to achieve signaling competence. The absolute requirement for Dynamin-dependent endocytosis but not endosomal entry or Rab11 activity supports ‘pulling force’ rather than ‘recycling’ models for Delta activation.

Page 23: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

13

Introduction Endocytosis has emerged as a key process governing intercellular interactions such as

cell-cell signaling. The most important endocytic step in signal regulation is internalization from the cell surface, as this controls receptor access to extracellular stimuli and may also influence intercellular signal transduction. A large body of literature has defined several distinct routes by which receptors are internalized [13,64]. For many receptors, internalization is mediated by cargo-specific adaptors, such as the AP-2 complex, which recruit Clathrin to form coated pits that subsequently undergo Dynamin-dependent scission. This route can be called AP-2-dependent endocytosis (ADE); however, AP-2-independent Clathrin-dependent endocytosis (CDE), Dynamin-dependent endocytosis (DDE) that is Clathrin-independent (CIE), and Dynamin-independent endocytosis (DIE) also exist. Interestingly, recent work suggests that the specific internalization route of a signaling receptor can lead to distinct signaling outcomes [38,41]. Despite the importance of the question, studies of internalization routes in animal tissues have been hampered by the expectation that regulators such as Dynamin and Clathrin are required for cell viability. This has prevented thorough genetic analysis, and accordingly our understanding of how surface internalization regulates metazoan signaling and development is incomplete.

Page 24: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

14

Results MENE(2L)-A and MENE(3R)-D identify genes that regulate an early stage of endocytosis

Drosophila is a valuable system to study how development is influenced by endocytosis,

and identification of mutants that selectively inactivate endocytic internalization routes would provide a means to investigate how these routes impact cell-cell signaling. To identify such mutants, we examined complementation groups isolated in a screen for neoplastic tumor phenotypes [53] in eye imaginal discs consisting predominantly of homozygous mutant cells (hereafter referred to as mutant discs). We compared the subcellular localization of Notch to cortical actin. Notch undergoes constitutive endocytosis; when any step of the endocytic pathway is disrupted, Notch accumulates and marks the point of blockage. Discs from two complementation groups (MENE(2L)-A and MENE(3R)-D) (2.1B-C’), in addition to infrequent internal puncta, showed strong Notch staining in puncta associated with the cell cortex (Fig. 2.1G-H), distinct from the large internal puncta seen in ESCRT mutant cells (Fig. 2.1F [55]) and the diffuse sub-cortical staining seen in Rab5 mutant cells (Fig. 2.1E [48]). This unique pattern of cargo localization suggests that an early step of the endocytic pathway, potentially cell surface internalization, is disrupted.

MENE(2L)-A and MENE(3R)-D disrupt subunits of the AP-2 endocytic adaptor complex

Interestingly, mapping of MENE(2L)-A and MENE(3R)-D placed both complementation

groups in regions containing genes encoding components of the AP-2 complex. Genomic sequencing revealed nonsense mutations in AP-2α (FlyBase: α-Adaptin [19]) in four MENE(2L)-A alleles (Fig. 2.2A). AP-2α phenotypes strongly resemble those of MENE(3R)-DNN20, which alters the initiating methionine of AP-2µ (FlyBase: AP-50) (Fig. 2.2A). Discs homozygous for a transposon insertion in the coding region of a third subunit, AP-2σ, showed phenotypes that recapitulated those of AP-2α and AP-2µ (Fig. 2.2A-B), suggesting that this represents the AP-2 null phenotype.

Disrupting ADE, CDE, and DDE reveals distinct defects in cargo internalization

AP-2 controls the internalization of certain proteins from the cell surface (Fig. 2.2C), an

endocytic step seldom analyzed using null mutations in animals. Since ADE also requires Clathrin heavy chain (Chc) and Dynamin, we sought to examine cells lacking either of these proteins. We first sequenced Chc3 [18] and found a nonsense mutation within the third coding exon (Fig. 2.2D). We then sequenced shiFL54 [65] and identified a frameshift that induces a premature stop codon within the GTPase binding domain (Fig. 2.2D). Imaginal discs homozygous for these putative null mutations show neoplastic phenotypes akin to those of AP-2 subunits, consistent with a role for endocytic internalization in epithelial polarity and tumor suppression (Fig. 2.2E-F). The role of AP-2, Chc, and Shi in cell polarity will be described elsewhere.

To compare the internalization steps disrupted by each mutant, we carried out endocytic assays in living tissue using Notch as a cargo. We directly monitored the endocytic population by following a surface-labeled cohort of Notch [48]. In WT discs, Notch is internalized after 10

Page 25: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

15

minutes into endocytic puncta; after 5 hours almost no labeled Notch is detected as it has been degraded in lysosomes (Fig. 2.3A, 2.4). In shi discs, Notch is trapped in a diffuse pattern at or near the cell surface, and almost no colocalization with endosomal markers is seen (Fig. 2.3D and data not shown). In striking contrast to shi, most Notch in AP-2 discs assumes a punctate cortex-associated distribution (Fig. 2.3B); internal puncta can also occasionally be seen (Fig. 2.4). In Chc discs, similar to shi discs, Notch was trapped at the cell cortex, but an additional population of endocytosed Notch could be visualized in enlarged internal puncta (Fig. 2.3C). These puncta colocalize with the endosomal protein Hrs, indicating that some Notch internalization still occurs in Chc cells (Fig. 2.3E); the endosomal trapping may reflect a subsequent requirement for Clathrin in endosomal sorting [11]. The overlapping but distinct phenotypes of the three mutants suggest that although most Notch turnover requires ADE, other populations exist that can be internalized by Dynamin-dependent CIE.

An AP-2-independent population of Notch internalized by CDE is competent for signaling

The isolation of null Drosophila mutants that specifically block ADE, CDE and DDE

provided the opportunity to define the requirements of these routes in animal development. We focused on the Notch signaling pathway because a particularly rich literature exists concerning its endocytic regulation. We first studied the Notch receptor, which shows a clear dependence on ADE (Fig. 2.1G-H, 2.3B). Mutations that block post-internalization stages of endocytosis, including early endosomal entry and subsequent endosomal sorting, strongly perturb Notch activity, though in dramatically different ways [66,67]. This suggests that Notch signaling potential is altered during its endocytic itinerary, but whether particular cell surface internalization routes are required for Notch signaling is unknown.

Notch signal transduction can be effectively studied in the Drosophila ovary, where the signal-sending germline cells and signal-receiving follicle cells (FCs) are separable by clonal analysis. We have previously shown that Notch entry into the early endosome in FCs is required for transcription of its target hindsight (hnt; FlyBase: pebbled) at stage 6, when germline Delta expression activates Notch in surrounding FCs (Fig. 2.5A [49]). To investigate cell surface internalization routes required for Notch activation, we examined Notch signaling in FCs mutant for AP-2, Chc, and shi. Consistent with an endocytic requirement for Notch activation, we found that in stage 6 shi or Chc FCs, Hnt is not expressed (Fig. 2.5C-D). Unexpectedly, stage 6 AP-2 mutant FCs show normal Hnt expression, indicating that the Notch signal has been efficiently transduced (Fig. 2.5E). Together with the trafficking data, these results suggest that although internalization and degradation of a significant population of Notch requires ADE, efficient Notch signaling itself is AP-2-independent.

Interestingly, despite the fact that a population of Notch reaches Hrs-positive endosomes through CIE (Fig. 2.3E), reporter assays suggest that this pool is incapable of efficient signaling (Fig. 2.5C and data not shown). Moreover, unlike other genotypes in which Notch accumulates in an Hrs-positive compartment, such as ESCRT and lethal giant discs (lgd) mutants [66,67] and constitutively-active Rab5 expression [49], Chc mutants do not show ectopic Notch signaling (Fig. 2.5C); this suggests that sorting by flat Clathrin coats [11] may promote access to the endosomal compartment permissive for Notch cleavage. In contrast, despite the prominent cell surface trapping of Notch seen in AP-2 cells, these cells contain a population of Notch that is capable of WT signaling; interestingly, a small amount of endocytic Notch can be detected in mutant cells (Fig. 2.4F-F’). These findings reveal that although populations of Notch can be

Page 26: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

16

internalized through AP-2-independent CDE and/or CIE, the AP-2-independent population can support WT signaling levels, while signaling cannot proceed without Clathrin. Delta signaling requires DDE but neither Clathrin nor the canonical recycling pathway

We focused subsequent experiments on the Notch ligand Delta, which has the best-

documented endocytic requirement for signaling [62,63]. Numerous studies suggest that DDE is required for Notch signaling [68,69], and two models have been proposed to account for this requirement. The ‘pulling force’ model suggests that endocytosis of Delta bound to the Notch extracellular domain enables Notch S2 cleavage and subsequent productive signaling to occur. The ‘recycling’ model suggests that initially inactive Delta must be endocytosed and returned to the membrane before it is competent for Notch signaling, perhaps because traffic through the recycling pathway promotes processing, association with co-factors, or appropriate localization and/or clustering. However, evidence for these models is indirect. Examining Delta signaling activity in the complete absence of endocytic regulators would reveal routes of endocytosis required for Delta activation, and potentially distinguish between the models. We therefore utilized null mutant alleles of endocytic genes to make homozygous clones in the germline, the Delta signal-sending tissue during oogenesis, and assayed Hnt expression in the surrounding WT FCs at stage 6.

We first analyzed mutants for the cell surface endocytic regulators AP-2, Chc, and shi. FCs surrounding AP-2 mutant germline clones (GLCs) showed normal Hnt expression, demonstrating that Delta activation is AP-2-independent (Fig. 2.5F and 2.5H). Though egg chambers containing Chc mutant GLCs showed morphological defects, Hnt expression was observed in most (12/13) cases (Fig. 2.5I), indicating that Clathrin is not required in the Delta signal-sending cell. Finally, in shi mutant GLCs, Hnt was not induced in FCs (Fig. 2.5J). Avl+ endocytic puncta colocalized with Delta in AP-2 and Chc mutants but only rarely in shi mutant germlines (Fig. 2.6). These data demonstrate that internalization of Delta is required for Notch signaling during oogenesis, but that this internalization and appropriate signaling can proceed by Dynamin-dependent CIE.

The lack of requirement for Clathrin in the germline was surprising, since Delta activation in other tissues requires the Epsin homolog Liquid Facets (Lqf), a Clathrin-interacting endocytic adaptor thought to bind ubiquitylated Delta [70,71]. To test whether Delta germline activation might occur in an unconventional fashion, we generated lqf GLCs. These egg chambers showed no Hnt expression, similar to GLCs of Delta (Fig. 2.5G and 2.5K). Overall, the common requirement for Lqf in signal-sending cells demonstrates that the egg chamber is not unique with respect to regulation of Delta signaling, and indicates that Epsin can function independently of Clathrin.

Finally, we analyzed GLCs mutant for regulators that control post-internalization steps of endocytosis. Virtually all cargoes internalized by ADE, CDE, and CIE traffic through a Rab5-positive endosome, the compartment from which recognized recycling routes emanate [13,72]. Interestingly, in egg chambers containing Rab5 GLCs, Hnt expression occurs normally at the appropriate stage (Fig. 2.5L). Delta is found predominantly near the cortex of Rab5 GLCs, as well as in certain internal Avl-positive structures (Fig. 2.6G); ultrastructural analysis suggests that the latter represent accumulations of early endocytic vesicles (data not shown and [73]). This result demonstrates that Rab5 function and entry into early endosomes is not necessary for Delta signaling activity. We next attempted to analyze GLCs mutant for null alleles of Rab11, a

Page 27: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

17

canonical regulator of recycling, but were unable to recover stage 6 egg chambers, presumably due to Rab11’s role in biosynthesis and/or cytokinesis [74,75]. However, we could recover stage 6 egg chambers expressing dominant-negative Rab11 in the germline; these show normal Hnt expression in FCs, indicating that Delta activation is unaffected (Fig. 2.5M). We could also recover egg chambers with disrupted function of Rab11 effectors. GLCs of the sec15 allele 3, which prevents Delta signaling in sensory organ precursor (SOP) cells [76], display appropriate Hnt expression, indicating that Sec15 is not required for Delta activation in the germline as in SOP cells (Fig. 2.7B). Appropriate Hnt expression is further seen in egg chambers with disrupted function of DRip11 and MyoV, which both mediate Rab11-dependent traffic [72,77-79] (Fig. 2.7D-E). Finally, we analyzed egg chambers mutant for the sec5 allele E13, which blocks recycling of the Yolkless receptor within the germline as well as E-cadherin in imaginal tissues [80,81], and again found normal Hnt expression (Fig. 2.7C). Together, these data reveal that germline Delta must be internalized by Dynamin-dependent forces but need not transit through the conventional recycling pathway in order to activate Notch signaling.

Page 28: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

18

Discussion

In this work we have used null alleles to block ADE, CDE, and DDE in Drosophila and address the routes of cargo internalization required for Delta/Notch signaling. While Clathrin and Dynamin have previously been posited to be essential for cell viability [18], our work reveals that cells lacking these proteins not only survive but can show tumorous phenotypes. AP-2 serves a restricted subset of cargo, but Clathrin and especially Dynamin’s involvement is more broad; it is thought that internalization of most CDE and CIE cargo requires Dynamin activity [13]. The survival of mutant cells and, in some cases, their ability to internalize endocytic cargo, emphasizes the robustness of Clathrin- and Dynamin-independent endocytic routes [13,82-84]. Moreover, while Clathrin and Dynamin have additional non-endocytic functions, the similarities between Chc, shi, and AP-2 cells indicate that these shared phenotypes result from defects in endocytosis.

One cargo whose internalization strongly requires ADE is Notch, which accumulates in clusters at the surface of AP-2 cells. Despite this prominent phenotype, there is no associated Notch signaling defect, as seen in for instance shi cells. The contrast between this result and the emerging paradigm that endocytosis of Notch is required for its cleavage and activation [66,67] suggests that some Notch must be internalized through AP-2-independent routes, which we confirmed by detecting rare endocytic Notch puncta in AP-2 cells. The small amount of internalized Notch seen in AP-2 cells could reflect upregulation of alternative endocytic pathways. Alternatively, it could reflect the existence of a pool of Notch, perhaps that activated by ligand, that is normally internalized through AP-2- independent pathways and can therefore access endosomes to permit appropriate cleavage when ADE but not CDE is disrupted. Differing entry routes for activated and non-activated receptors have been described for EGFR and TGFβ [38,41] and differing subsets of early endosomes are accessed by ADE and AP-2-independent CDE [85]. Our results justify further studies to determine whether cell surface entry routes are yet another step by which endocytosis regulates Notch signaling.

While the role of cell surface internalization in Notch activity has been little studied, there is a well-established but incompletely understood role for internalization in activity of Delta. In addition to demonstrating that Delta can be internalized and activated through Dynamin-dependent CIE, our studies with mutant GLCs provide critical information to distinguish between the ‘pulling force’ and ‘recycling’ models. Shi and lqf are required in the germline for Notch signaling, confirming that regulation of Delta activity in this tissue parallels that in others. However, Delta internalized in Rab5 germlines can still activate Notch signaling as in WT germlines. While loss of germline shi function prevents endocytic pits from undergoing scission from the cell surface [86], Rab5 germline cells form many internalized endocytic vesicles, but not early endosomes [54,73] through which cargoes destined for canonical recycling pathways pass [72], suggesting that Delta internalized in Rab5 GLCs cannot access the recycling pathway. The requirement for recycling could not be further analyzed by utilizing null Rab11 mutations, but we found normal Delta signaling in germline cells expressing dominant-negative Rab11, with disrupted function of Rab11 effectors, and which are homozygous for a mutation that blocks recycling of other transmembrane proteins. In the absence of an assay that can determine whether recycling of Delta actually occurs in WT germline cells, we cannot formally exclude that these mutants provide an incomplete block to any such recycling that might occur. Moreover, our results do not exclude that recycling is used to promote Delta activity in other specific developmental contexts, such as Drosophila SOP cells

Page 29: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

19

where it may serve to enrich Delta at specific plasma membrane subdomains or promote asymmetric inheritance of Delta [75,76,87,88]. However, they indicate that a basic mode of Delta signaling, used in the simple inductive signaling of the developing Drosophila egg chamber, requires Dynamin-dependent internalization but not trafficking through known recycling pathways. These analyses, which utilize endogenous proteins in endocytic mutant tissue in vivo, therefore support the pulling force model of Delta signaling activity.

Page 30: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

20

Materials and Methods Fly Stocks and Genetics

AP-2α40-31 FRT40A and AP-2µNN20 FRT82B [53] were generated using EMS mutagenesis.

AP-2α40-31 terminates at amino acid 510 in the trunk region, prior to the hinge and appendage domains, which mediate important functional interactions with cargo and endocytic accessory proteins [89]. P{SUPor-P}AP-2σKG0245 [90], which contains an insertion at codon 70, was obtained from the Bloomington stock center and recombined onto FRT82B. Because tissues mutant for putative null alleles of each of the three subunits have similar phenotypes, representative experiments shown performed with the AP-2α40-31 allele are labeled AP-2. Chc3 [18,91] truncates the protein shortly after the β-propeller repeat domain and would render any protein that was made unable to interact with other Clathrin heavy or light chains [9]. Molecular lesions in AP-2α, AP-2µ, Chc, and shi alleles were identified by sequencing amplified genomic DNA isolated from heterozygous adults using polymerase chain reaction. Details and primer sequences are available upon request. Additional fly stocks used include Rab52FRT40A [92], shiFL54FRT19A [65], lqfL71FRT80 [93], Vps25A3FRT42 [55], Dlrev10eFRT82 [94], sec15-3FRT82 [76], dRip11KG02485FRT19A [77], sec5E13FRT40 [95], UASp-YFPRab11.S25N [96], mattubGal4Vp16, UAS-MyoV-motor and UAS-MyoV-CT [78].

Follicle cell or germline clones were generated using the following stocks: ubGFP FRT19A; e22c-GAL4 UAS-FLP, ubGFP FRT40A; GR1-GAL4 UAS-FLP, e22c-GAL4 UAS-FLP; ubGFP FRT82, hsFLP;ovoD FRT40A (for AP-2α GLCs), hsFLP;;ovoD FRT80 (for lqf GLCs), ubGFP FRT19A;hsFLP, hsFLP;ubGFP FRT40A, and hsFLP;;ubGFP FRT82B. w l(1)CL8.7 P[m-w+ arm-lacZ] FRT19A / FM7a; eyFLP was a kind gift of Brett Pellock [97] and was used in addition to Dp(1;Y)W73 to generate mutant eye discs for genes on the X chromosome. For generating mutant eye discs for genes on other chromosomes, we used eyFLP cl GMR-hid FRT40A, eyGAL4-UASFLP; cl FRT40A, eyFLP;;M(3) FRT80, and eyFLP cl GMR-hid FRT82.

Immunohistochemistry and Microscopy

Antibody staining was performed using standard conditions [98]. Notch trafficking was carried out as described previously [48].

The following primary antibodies were used: mouse anti-Mmp1, mouse anti-NECD, mouse anti-Hnt, mouse anti-Dl (all from Developmental Studies Hybridoma Bank, see references therein), rabbit anti-Avl [48] and guinea pig anti-Hrs [99]. To visualize the cell cortex, we co-stained with TRITC-phalloidin (Sigma) upon secondary antibody staining. Secondary antibodies were from Molecular Probes. Images shown are single confocal sections collected on a Leica TCS confocal microscope and assembled using Adobe Photoshop CS4. At least five samples were analyzed for each experiment.

Page 31: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

21

Figure 2.1: New MENE Mutants Disrupt Internalization from the Cell Surface WT (A, A’) or mutant MENE(2L)-A or MENE(3R)-D eye imaginal discs (B, B’ and C, C’) stained for F-actin (red) and Mmp1 (magenta), an indicator of neoplastic transformation. D-H: Eye imaginal discs isolated from wandering L3 larvae stained for Notch (green) and F-actin (red): Notch localizes to the apical plasma membrane and in endocytic vesicles in WT cells (D), in a diffuse subcortical pattern in Rab5 mutants (E), in large internal puncta in Vps25 mutants (F), and predominantly in puncta along the cell cortex in mutants from complementation groups MENE(2L)-A and MENE(3R)-D (G and H). In D-H arrows indicate Notch at the cortex and arrowheads indicate internal Notch. Scale bars: 100µm (A-C’) and 10µm (D-H).

Page 32: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

22

Page 33: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

23

Figure 2.2: Identification of Null Mutations in Cell Surface Endocytic Regulators A: Coding regions of AP-2 complex subunits with allele locations and details and cartoon of AP-2 adaptor complex (inset). AP-2α is disrupted in the MENE(2L)-A complementation group, while AP-2µ is disrupted in the MENE(3R)-D complementation group. B: AP-2σ mutant L3 eye imaginal discs stained for F-actin (red), Mmp1 (magenta), and Notch (green) (left, middle, and right panels, respectively). C: Schematic of AP-2-dependent endocytosis and cell surface endocytic regulators. D: Chc and shi coding regions with location of mutant alleles. Chc (E) and shi (F) mutant L3 eye imaginal discs stained for F-actin (red) and Mmp1(magenta). Scale bars: 100µm (B, left and middle panels, E-F ) and 10µm (B, right panel). Δ = deletion, fs = frameshift, * = stop codon.

Page 34: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

24

Page 35: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

25

Figure 2.3: Cargo Trafficking Phenotypes are Distinct in Cells with Disrupted ADE, CDE, or DDE Notch staining (green) 5 hrs post-chase from live trafficking experiment. Surface-labeled Notch, which is degraded at this time point in WT discs (A), is prominently trapped in surface puncta in AP-2α with a few internal puncta also visible (B), cortically and in internal puncta in Chc (C), and in a diffuse cortical location in shi (D) mutant eye discs. Internally-trapped Notch (green) in Chc mutants colocalizes with the endosomal protein Hrs (red) (E). Dashed lines in A-D represent cell cortex as determined by F-actin staining. In B-E arrows indicate Notch localized at or near the cell surface, arrowheads indicate internalized Notch. Scale bar: 10µm.

Page 36: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

26

Page 37: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

27

Figure 2.4: Notch traffics distinctly in mutants that disrupt the internalization step of endocytosis Anti-NECD (green) was used to detect a cohort of surface-accessible Notch and follow it over time relative to the cell surface marked by F-actin (red) in WT (A-A’=0min, E-E’=10min, and I-I’=300min), AP-2α (B-B’= 0min, F-F’= 10min, J-J’= 300min), Chc (C-C’= 0min, G-G’= 10min, K-K’= 300min), and shi (D-D’= 0min, H-H’= 10min, L-L’= 300min) mutant eye imaginal discs from L3 larvae. Scale bar = 10µm.

Page 38: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

28

Page 39: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

29

Figure 2.5: Endocytic Requirements for Delta/Notch Signaling in Signal-Receiving and Signal-Sending Cells A: Schematic of Notch and Delta expression and signaling in the developing Drosophila ovary, adapted from [49]; Notch in follicle cells is activated at Stage 6/7 in response to Delta signaling from the germline. Staining for the Notch signaling target Hindsight (Hnt) (blue) in WT (B), Chc (C), shi (D), and AP-2α (E) follicle cell clones (FCs). Hnt staining is seen in AP-2α, but not Chc or shi mutant follicle cells. Hnt staining (blue) in WT follicle cells surrounding WT (F), Dl (G), AP-2α (H), Chc (I), shi(J), lqf (K), Rab5(L), and Rab11DN-expressing(M) germline clones (GLCs). Hnt is present in follicle cells surrounding AP-2α, Chc, Rab5, and Rab11-expressing, but not shi or lqf germline clones. All mutant clones are outlined by dashed lines. Scale bars: 30µm (B-E) and 20µm (F-M).

Page 40: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

30

Page 41: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

31

Figure 2.6: Delta localization in mutant GLCs Delta (blue) staining in WT (A) and mutant (absence of GFP) GLCs (B-J). Delta is also shown in germ line cells expressing UAS-MyoV-motor (K) and UAS-Rab11DN-YFP (L). Avl (red) is also shown in A and C-G. Scale bar = 20µm.

Page 42: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

32

Page 43: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

33

Figure 2.7: Delta signal-sending is not impaired in GLCs mutant for Rab11 effectors Hnt (blue) expression in WT follicle cells surrounding WT (A), sec15 (B), sec5 (C), rip11(D), and UAS-MyoV-motor-expressing (E) GLCs. F-actin is shown in red. Scale bar = 20µm.

Page 44: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

34

Page 45: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

35

Chapter 3

Evidence for regulation of endocytic traffic by junctional scaffold neoplastic tumor suppressors

Page 46: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

36

Abstract

Endocytosis is an important regulator of intercellular interactions, including signaling and adhesion. Recent studies have demonstrated a requirement for endocytosis in epithelial polarity as well, but the specific endocytic entry routes and cargo involved are not known. We show here that apicobasal polarity in Drosophila epithelia requires the activity of the AP-2 endocytic adaptor complex, as well as Clathrin Heavy Chain and the Dynamin homolog Shibire. Cells depleted of any of these components display inappropriately distributed apical membrane domains, accompanied by failures of differentiation and overproliferation in imaginal discs. These mutant phenotypes closely resemble those of previously identified endocytic regulators, as well as those of junctional scaffold neoplastic tumor suppressor genes (TSGs) such as scribble. However, despite the similar phenotypes, scrib mutant tissue does not show decreased rates of endocytic uptake, but rather displays phenotypes consistent with endocytic recycling defects. The transmembrane regulator of apical polarity Crumbs is internalized by endocytosis, but Crumbs is not required for the polarity defects nor the neoplastic transformation of endocytic mutant or scrib mutant tissue. We conclude that Drosophila epithelial polarity requires AP-2-dependent endocytosis of key polarity regulators other than Crumbs.

Page 47: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

37

Introduction

The specialized functions of many cell types require cell polarity. One prominent polarized cell type is epithelia, in which certain proteins show distinct and non-overlapping distributions along free apical or contacting basolateral plasma membranes. The mechanisms that dictate these polarized protein distributions are well-conserved and broadly utilized, so elucidating the cellular pathways that control epithelial polarity is important to understanding, for instance, the polarity underlying the earliest divisions of animal embryos and the asymmetric divisions of stem cells. Nevertheless, our knowledge of how polarized proteins achieve their restricted distribution in a cell is far from complete.

Epithelial cell polarity must, at least in part, be regulated by membrane protein trafficking. Several multiprotein modules have been identified as key regulators of epithelial polarity in both vertebrate and invertebrate animal species [51] and are potential candidates for the regulation of protein trafficking, but the way in which they might carry out this function remains unknown. Functionally, these polarizing modules can be divided into two groups. One group consists of proteins that specify the apical membrane domain. These are the Par and Crumbs (Crb) modules, which in Drosophila consist of Bazooka (Baz)/Par-6/atypical protein kinase C (aPKC)/Cdc42-GTP and Crb/Stardust/PATJ respectively. The Par module is the earliest actor in polarizing the apical membrane; Baz and Par-6 serve as scaffolding proteins that direct aPKC kinase activity to appropriate targets in response to a Cdc42-mediated cue [100]. One likely target is the Crb module, in which the large transmembrane protein Crb can specify the apical membrane domain via a poorly characterized but aPKC-dependent pathway [101].

The second group of polarizing modules consists of proteins that specify the basolateral domain. Interestingly, a major activity of these proteins is to antagonize the activity of apical polarizing modules, thus restricting the apical domain [102,103]. The primary basolateral polarizing module is the Scribble (Scrib) module, consisting in Drosophila of Scrib/Discs-large (Dlg)/Lethal giant larvae (Lgl) [104]. All three proteins are ‘junctional scaffolds’ that contain multiple protein-protein interaction domains and are associated with the lateral plasma membrane and cell junctions. Lgl can bind myosin and syntaxin proteins, and is inactivated when phosphorylated by aPKC [105], but how it and other Scrib module proteins act with the cellular machinery that dictates the distribution of polarized membrane proteins is not known.

Interestingly, the Scrib module is required not only for epithelial polarity but also for growth control of certain epithelial organs [50]. In Drosophila imaginal discs, loss of any Scrib module protein causes dramatic overproliferation of disorganized cells, accompanied by a failure to differentiate and the acquisition of invasive behaviors. This group of phenotypes is termed ‘neoplastic’, and genes that when mutated result in these phenotypes are called ‘neoplastic tumor suppressor genes (TSGs)’. The link between control of epithelial polarity and suppression of neoplastic phenotypes in Drosophila is not known, but evidence suggests that the Scrib module may also play a role in preventing mammalian tumor progression [106]. This role has added to the interest in understanding the fundamental activity of neoplastic TSGs in polarity determination.

To gain insight into this activity, we have carried out a large-scale forward genetic screen for additional Drosophila genes that control both cell polarity and cell proliferation [53]. This screen has greatly expanded our knowledge of the number and types of genes that regulate both processes beyond the three originally identified Scrib module junctional scaffold neoplastic TSGs. Strikingly, a number of new neoplastic TSGs have been found to encode central

Page 48: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

38

regulators of endocytosis, the process by which membrane proteins are internalized into the cell. Specifically, null mutations in Rab5 or three other genes that control cargo entry into early endosomes [48,54], or null mutations in six genes encoding ESCRT components that sort endosomal cargo for degradation [34,55,107-109], cause loss of polarity and overproliferation of imaginal disc epithelia; mutations that disrupt subsequent stages of endocytic degradation do not. These mutations in canonical endocytic regulators have provided insight into the control of Notch signaling by endocytosis [66], but how endocytosis controls epithelial polarity remains mysterious. Although several models can be proposed [110], important questions remain unanswered, such as which of the several endocytic entry and trafficking routes [13] are involved in polarity determination, what are the specific endocytic cargoes involved, and whether there is a functional relationship between the junctional scaffold and endocytic classes of neoplastic TSGs. Here we use null mutations in endocytic regulators, including those that control internalization from the cell surface, to shed light on these issues.

Page 49: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

39

Results Mutations in cell surface endocytic regulators cause neoplastic transformation

We recently reported the isolation and characterization of molecularly null mutations in

Drosophila genes that regulate the internalization of transmembrane cargo from the cell surface [111]. Cells lacking either the alpha, sigma, or mu subunits of the AP-2 adaptor complex, the Dynamin ortholog Shibire (Shi), or the Clathrin heavy chain (Chc) show defective endocytosis of the transmembrane protein Notch, some of which becomes trapped at the plasma membrane in mutant cells. Since genes that regulate subsequent endocytic stages act as neoplastic TSGs in Drosophila, we assayed hallmarks of neoplastic transformation in tissues mutant for genes that regulate cargo internalization. We found that eye imaginal discs consisting predominantly of cells mutant for AP-2 components, shi or chc (hereafter referred to as mutant eye discs; see Experimental Procedures) are severely disorganized, with disrupted cell shapes, an absence of epithelial monolayering, and increased levels of filamentous actin; these discs are also larger than their WT counterparts (Fig. 3.1A-D). Mutant eye discs also showed a loss of neuronal differentiation (Fig. 3.1E-H) and upregulation of the matrix metalloprotease Mmp1 (Fig. 3.1I-L). Finally, when clones are made in another epithelium, the follicle cells surrounding developing egg chambers in the adult ovary, mutant cells again lose columnar or cuboidal shapes and display multilayering phenotypes, particularly at the anterior and posterior poles (Fig. 3.1M-P). These results indicate that AP-2 components, shibire, and chc act as neoplastic TSGs. Cytosolic polarity proteins display similar localization defects in endocytic and junctional scaffold tumor suppressor mutant cells

We next examined the polarization of the plasma membrane in tissue mutant for cell

surface endocytic regulators. The follicle cell epithelium was chosen because of the less severe defects in tissue architecture caused by mutant cells and because of the ease of recovering genetic mosaic tissues in the absence of cell competition. We initially assessed the polarization of cell cortex-associated proteins, using the apical marker aPKC and the basolateral marker Dlg. In AP-2 mutant follicle cells, aPKC displayed a fragmented distribution (Fig. 3.2F-F”) that in some regions was interspersed with Dlg in a complementary, mutually exclusive fashion; in other regions the two proteins colocalized in a manner never seen in WT tissue (Fig. 3.2E-E”). This phenotype reflects a severe perturbation of apicobasal polarity. A very similar distribution of aPKC and Dlg was also seen in follicle cells mutant for Rab5 (Fig. 3.2H-H”), as well as those mutant for the ‘junctional scaffold’ neoplastic TSG mutant lgl (Fig. 3.2G-G”). These results demonstrate that proper epithelial polarity requires AP-2-dependent endocytosis, and further suggest that polarization might be controlled in a similar manner by both the endocytic and junctional scaffold classes of neoplastic TSGs. Apical transmembrane proteins display distinct localization patterns in AP-2 versus early endocytic and junctional scaffold tumor suppressor mutant cells We then compared the localization of polarized transmembrane proteins in endocytic and junctional scaffold TSG mutant cells. We found that in AP-2 cells, the apically restricted transmembrane proteins Crb and Cad87A were seen in a fragmented distribution around the

Page 50: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

40

plasma membrane, rather than in the single continuous domain of WT cells (Fig. 3.2A-B” and data not shown). Crb was intermixed with and showed regions of overlap as well as complementary distribution with Neuroglian (Nrg), a transmembrane protein normally localized to the basolateral domain [112]. Exclusion and overlap between apical and basolateral transmembrane proteins was also seen in lgl follicle cells (Fig. 3.2C-C”). Strikingly, however, both Crb and Cad87A showed an additional hazy, apparently sub-cortical distribution in lgl follicle cells; this distribution was also seen in scrib follicle cells, but this distinction was not evident in imaginal discs(Fig. 3.2C-C” and data not shown). This localization was quite distinct from the tight plasma membrane association seen in AP-2 follicle cells (Fig. 3.2B-B”). A subcortical haze of Crb and Cad87A was also seen in Rab5 follicle cells, raising the questions of whether this haze may reflect trapping in endocytic structures near the plasma membrane (Fig. 3.2D-D” and data not shown), and whether Crb endocytosis is altered in scrib mutants. Crb is an AP-2-dependent endo-lysosomal cargo

To determine whether Crb is regulated via AP-2-dependent endocytosis, we performed analysis in imaginal discs mutant for endocytic regulators. We confirmed that cellular Crb levels are controlled by lysosomal degradation [107] by showing that Crb was trapped in endocytic structures in cells mutant for the ESCRT component and neoplastic TSG Vps25, in which cargo sorting into the multivesicular body is blocked (Fig. 3.3D-E), and in cells mutant for the PIKFYVE kinase Fab1[113], in which lysosomal degradation is blocked, but cellular polarity is unaffected (Fig. 3.3F). By contrast, the cortex-associated polarity-regulating proteins aPKC and Cdc42 are not trapped in endosomes of neoplastic ESCRT or Fab1 mutant cells, nor do they show grossly different distribution in AP-2 mutants than in scrib mutant cells (Fig. 3.3B’-C’ and data not shown). We next determined whether Crb is an AP-2 dependent cargo. Examination of Crb localization in AP-2 mutant tissue revealed a pattern of ‘trapping’ reminiscent of Notch (Fig. 3.6A-D’), which is internalized largely through AP-2-dependent endocytosis and is found at the cell cortex in AP-2 mutant cells [111]. These data suggest that Crb undergoes AP-2-dependent endocytosis. Junctional scaffold mutants do not alter AP-2 dependent endocytosis, but display other endocytic trafficking defects

The polarity phenotypes shared by endocytic and junctional scaffold TSG mutant cells, as

well as the evidence for endocytic defects in the latter, raised the possibility that junctional scaffolds control epithelial polarity by regulating AP-2-dependent endocytic traffic. We therefore carried out endocytic assays in living imaginal discs using the AP-2 cargo Notch [111]. However, we found that that AP-2-dependent endocytosis is intact in imaginal discs mutant for dlg, scrib, or lgl. These discs, in contrast to discs mutant for cell surface endocytic regulators, internalize Notch into endocytic puncta and transport it for lysosomal degradation after five hours, without evident trapping at the plasma membrane or other intermediate steps of the endocytic pathway (Fig. 3.4A-D’). Similarly, discs that overexpress an activated form of aPKC, which antagonizes junctional scaffold neoplastic TSGs and is sufficient to induce polarity defects and neoplastic transformation [114], internalize and degrade Notch (Fig. 3.4E-E’). To investigate the possibility that mutant cells exhibit a decreased rate of endocytosis rather than a complete block, we assessed endocytic rates by quantifying the number of cargo-containing

Page 51: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

41

endocytic puncta per cell after 20’ of internalization, a time point when cargo has not been degraded in lysosomes. Quantitation of Notch endocytic puncta again revealed no significant difference between WT and scrib mutant cells (Fig. 3.4G). Because Notch internalization, like neoplastic tumor suppression, requires AP-2 function (Fig. 3.4F-F’ and [111]), we conclude that the junctional scaffold neoplastic TSGs do not regulate polarity via general control of AP-2 dependent endocytosis.

While examining Notch endocytosis, we also examined the fluid-phase endocytic tracer Dextran, to explore whether endocytosis of other cargoes might be altered in junctional scaffold neoplastic TSG mutant cells. We found that Dextran is internalized in cells mutant for junctional scaffold neoplastic TSGs (Fig. 3.4K-L’). However, analysis of the 20’ time point indicated that scrib mutant cells show more Dextran-containing endosomes than WT tissue (Fig. 3.4H), suggesting that while fluid-phase endocytosis is present, its kinetics are altered. Some portion of Dextran taken up by both WT and scrib mutant cells follows the degradative pathway, as only a few Dextran-positive puncta are seen after 5 hours of trafficking in either case and many more are seen after 5 hours when the tissues are treated with the lysosomal inhibitor chloroquine (Fig. 3.4I-L’). However, evidence exists that at least some Dextran undergoes ‘regurgitation’ in Drosophila cells [25], as well, raising the possibility that the increase in Dextran-positive puncta in scrib mutant cells could represent a block in a recycling pathway.

To reconcile the evidence for endocytic trapping of Crb and Cad87A with the normal degradative traffic of Notch in junctional scaffold TSG cells, we considered whether trapping might happen along a distinct endocytic route that these cargoes follow. Crb is known to transit Rab11 recycling endosomes in embryos [115], but direct assays for Crb recycling are not available. To assess whether junctional scaffold TSG cells might be defective in the recycling pathway, we therefore turned to the human Transferrin receptor (hTfR) a canonical AP-2-dependent recycling cargo in vertebrate cells that has been shown to undergo recycling in Drosophila cells [25]. We first performed live trafficking of fluorescently-labeled human holo-transferrin on Drosophila wing discs expressing UAS-hTfR under the control of enGAL4. After a 10 minute pulse followed by a 3 hour chase, a number of Tfn-positive puncta were still observed in WT cells (Fig. 3.5A); this number was increased upon chloroquine treatment (Fig. 3.5B) suggesting that some hTfR and Tfn is normally degraded via the lysosome. Surprisingly, in the same assay using dlg discs expressing hTfR, very small numbers of Tfn positive puncta were observed in either the absence or presence of chloroquine (Fig. 3.5C-D). This result suggested either a defect in hTfR expression, inability of hTfR to traffic to the membrane, or a defect in the recycling of hTfR in dlg mutant cells. We then stained for hTfR directly on fixed tissue (Fig. 3.5E and G) and found very little hTfR at steady-state in dlg mutant discs as compared to WT (Fig. 3.5G compared to Fig. 3.5E). This did not reflect general defects in transgene expression nor in exocytic delivery, as transmembrane-tethered proteins showed normal expression when driven by enGAL4 in dlg discs (data not shown) and endogenous transmembrane proteins were accessible at the cell surface of junctional scaffold mutant cells (Fig. 3.4B-D). Surprisingly, chloroquine treatment revealed that hTfR is indeed expressed in dlg cells but a majority traffics to lysosomes whereas only a minority does so in WT cells (Fig. 3.5F, H). These results demonstrate that hTfR, a normally recycled cargo, is shunted toward a degradative pathway in dlg cells.

Defective hTfR trafficking in dlg cells suggested that loss of junctional scaffold TSGs may interfere with the recycling pathway. As mentioned above, live trafficking assays for Crb, a cargo known to pass through a Rab11 positive recycling compartment, are unavailable.

Page 52: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

42

However, we tested whether this endogenously expressed recycled cargo might accumulate in lysosomes in junctional scaffold neoplastic TSG mutant discs similar to hTfR. We cultured either WT or scrib discs in the presence or absence of chloroquine for 5 hours, followed by antibody staining for Crb. In WT discs, Crb is localized exclusively to the apical plasma membrane (upper left in Fig. 3.5I-I’ and lower right in Fig. 3.5J-J’) and very few Crb-positive puncta are seen, even in the presence of chloroquine, suggesting that little Crb is normally trafficked to the lysosome. In scrib mutant cells Crb localizes around the entire cell periphery (Fig. 3.5K-K’) due to the loss of apicobasal polarity and is only rarely observed in puncta. However, upon 5 hour treatment with chloroquine, many more Crb-positive puncta are seen (Fig. 3.5L-L’), reminiscent of the accumulation of hTfR in dlg mutant cells treated with chloroquine. These data lend further support to the hypothesis that junctional scaffold tumor suppressors play a role in endocytic recycling of particular cargoes.

Neoplastic transformation in endocytic and junctional scaffold mutants does not require Crb

The above data, indicating specific endocytic trafficking defects in junctional scaffold

mutant cells, raise the question of what cargo is mistrafficked to alter apicobasal polarity. In particular, the common phenotype seen in cells mutant for either junctional scaffold or endocytic class neoplastic TSGs raises the possibility that levels and/or activity of a specific apical polarity-regulating cargo might be normally controlled by endocytosis. Crb overexpression has been shown to be sufficient to specify apical character on plasma membranes [116]. In addition, we previously found that the endocytic neoplastic TSG mutants Rab5 and avl show elevated Crb expression, and that overexpression of Crb alone in imaginal discs is sufficient to induce neoplastic growth [48]; these results led to the suggestion that endocytic downregulation of Crb is required to control polarity and proliferation. Unexpectedly, despite the common apicobasal polarity defects seen in cells mutant for AP-2 subunits and later endocytic regulators and the evidence for AP-2-dependent Crb internalization, we found that Crb levels are not strongly elevated in AP-2 mutant imaginal discs, in contrast to discs mutant for either Rab5 [48] or the functionally related neoplastic TSG vps45 [54] (Fig. 3.6E-G). While it is not clear why Crb levels are more elevated in vps45 and Avl than in AP-2 mutant tissues, these data raise the possibility that neoplastic transformation of tissue mutant for AP-2 and other neoplastic TSGs may not be due to the failure to internalize Crb from the plasma membrane.

To test this rigorously, we generated recombinant chromosomes that carried null mutations in both crb and Vps45, and used them to generate mutant discs. Interestingly, removing Crb from Vps45 mutant discs did not suppress the Vps45 imaginal disc phenotype. Double mutant discs lose epithelial architecture, overgrow, and express Mmp1 levels comparable to those mutant for Vps45 alone, indicating that Crb is not in fact required for neoplastic transformation (Fig. 3.6K-L’). We carried out an analogous experiment using discs mutant for the junctional scaffold tumor suppressor scrib, which display mispolarized Crb localization, and again found that loss of Crb in this context did not alter the neoplastic phenotype (Fig. 3.6I-J’) [56]. These data rule out Crb as the sole polarity-controlling cargo misregulated in neoplastic tumors, and furthermore indicate that epithelial polarity and proliferation require carefully controlled levels of other, currently unidentified, cargoes whose internalization requires AP-2-dependent endocytosis and perhaps endocytic recycling.

Page 53: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

43

Discussion Epithelial polarity requires AP-2 dependent endocytosis

We report here that three proteins which regulate endocytic internalization from the

plasma membrane are required for global apicobasal polarity in Drosophila epithelia. The most prominent phenotype associated with Chc, shi, and AP-2 mutant cells is a dramatic neoplastic transformation associated with the aberrant localization of polarized membrane proteins. We and others have previously shown that disrupting two later stages of the endocytic pathway – entry into early endosomes and sorting into multivesicular bodies (MVBs) – causes loss of polarity [110]. However, the requirement for Adaptin subunits in polarity determination identified here is a particular advance. While the endosome entry and MVB sorting regulators serve broad classes of cargo internalized by many different endocytic entry routes [13], Adaptins control internalization of only a subset of cargo, often with defined consensus sequences that bind directly to Adaptin subunits [1]. Moreover, while Clathrin and Dynamin can play roles in biosynthetic trafficking that, when disrupted, can impact polarity in cultured mammalian cells [117], these defects are different from the mislocalized apical proteins and change in cell architecture seen when their fly homologs are mutated in vivo. While we have not investigated biosynthetic transport, the common phenotype that Chc and shibire mutant cells share with cells lacking AP-2, which is involved only in endocytosis, are consistent with the polarity defects resulting from disruption of an endocytic role. A parsimonious model is that the requirements that we document for AP-2, Shibire and Chc, as well as the endosomal entry and MVB sorting regulators, all reflect a single role for AP-2-dependent endocytosis in epithelial polarity. Endocytic defects in junctional scaffold polarity mutants

Amongst the diverse trafficking routes that cargoes may follow after endocytic

internalization, which is key for regulating apicobasal polarity? Comparison of phenotypes between endocytic and junctional scaffold mutants provides insight. Our analysis indicates that both classes share many common phenotypes (mislocalization of apical proteins, unchecked cell proliferation, ectopic activation of JNK signaling), while differences such as the type of apical transmembrane protein mislocalization seen in internalization mutants versus junctional scaffold mutants may provide clues about underlying mechanisms of polarity regulation. One simple mechanism linking polarity regulation by the two neoplastic TSG classes would be that junctional scaffold tumor suppressors positively regulate AP-2-dependent internalization. Junctional scaffold proteins antagonize the activity and localization of the Par module, which has been recently shown to affect endocytic rates in cells [58-61]. However, we found no evidence for altered internalization, endosomal entry, or lysosomal delivery of Notch in junctional scaffold TSG mutant cells. While it is possible that there are cargo-specific effects that are not captured by our assays, these data rule out the possibility that polarity defects in junctional scaffold TSG mutant cells are caused by general inactivation of AP-2 dependent endocytosis.

Interestingly, we found evidence to support the hypothesis that junctional scaffold TSGs regulate a cargo-specific, post-internalization endocytic sorting pathway. We observed altered trafficking of Dextran, the human TfR, and Crb in junctional scaffold mutant cells. Interestingly, these are all cargo that are internalized from the apical surface and may traverse a Rab11 recycling endosome [25,115]. At steady state, hTfR and Crb are found at the apical domain of

Page 54: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

44

the cell, but loss of junctional scaffold TSG function leads to not only mislocalization to basolateral surfaces in the case of Crb or reduced localization at the apical domain in the case of hTfR, but also elevated traffic to the lysosome. If junctional scaffold mutants block endocytic recycling, it might be speculated that normally recycled cargoes would instead be shunted down the alternative pathway of lysosomal degradation. Indeed, there is some evidence that defects in endocytic recycling cause an overall decrease in levels of normally recycled cargo [118]. Role of endocytosis in epithelial polarity

What is the outcome of defective trafficking? One potentially appealing model for the

complementary domains of apical and basolateral proteins in wild type cells is that mutually exclusive localization requires endocytosis. For example, apical membrane proteins, perhaps through the activity of an enzymatic effector such as aPKC, would stimulate the endocytic clearance of basolateral proteins found nearby. However, we found only limited evidence for this, as AP-2 deficient cells did not show frequent overlap of apical and basolateral proteins, and often showed exclusive and complementary localization. These data suggest that an endocytic mechanism alone is insufficient to account for the exclusion between apical and basolateral proteins, and that another feature is central to polarity determination.

A model more consistent with our findings is that apical polarity in Drosophila requires transcytosis. In this scenario, a key apical regulatory protein is transported following biosynthesis to the basolateral surface, and must be internalized to prevent mixing of membrane domains. It is internalized through AP-2-dependent endocytosis, and then sorted into a Rab11 recycling pathway via which it reaches the apical cell surface. This model posits that junctional scaffold tumor suppressors, perhaps by antagonizing aPKC activity, promote the sorting of this protein into an apical recycling pathway. In their absence, accumulation of normally apical transmembrane proteins, such as Crb, leads to their ‘spreading’ around the membrane and perhaps a subsequent increase in trafficking to the lysosome in an attempt to clear the excess protein. Some mammalian cells show a heavy reliance on transcytosis for apical protein localization [119], but it is not necessary to invoke such a phenomenon in imaginal epithelia to explain our data, as transcytosis of a key apical polarity regulator could produce this outcome. Similarly, it is possible that transcytosis serves as an ‘error-correction’ mechanism for a minor fraction of the cargo, rather than its major route of biosynthetic traffic. Given the categorical separation of apical and basolateral domains in WT cells, and the deleterious effects on both cells and organisms when this is lost, a mechanism must exist to ensure robustness of polarity.

It is increasingly evident that endocytic sorting and trafficking of certain cargo are more complex than current paradigms can explain. Moreover, there may be cargo-specific effects that are not captured by our assays. Strict tests of the hypothesis that junctional scaffold neoplastic TSGs act primarily through an endocytic mechanism must therefore await identification of a specific polarity-regulating cargo involved.

What polarity-regulating cargo requires endocytic regulation?

Our data together suggest that the AP-2-dependent internalization, and perhaps

trafficking via recycling endosome, of at least one protein is critical for proper apicobasal polarity and proliferation control in wild type epithelia. Crb was an attractive candidate for this cargo because of its sufficiency when overexpressed to induce neoplastic transformation, its AP-

Page 55: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

45

2-dependent endocytosis, and because it traverses a Rab11 endosome in some fly tissues. The double-mutant studies, however, in which elimination of Crb does not suppress neoplastic phenotypes of discs mutant for either an endocytic or a junctional scaffold neoplastic TSG, show that Crb cannot be the sole cargo responsible for proper polarity control. We examined the localization of additional candidates – aPKC and Cdc42, which are cytosolic polarity proteins that associate with the cell cortex – and found no evidence that their levels at the plasma membrane are controlled by AP-2-dependent endocytosis, though we cannot rule out differences in activity. Nevertheless, reducing activity of the key polarity regulator aPKC can partially suppress neoplastic growth in discs mutant for both the endocytic [48,57] and junctional scaffold [56,120] classes of neoplastic TSGs. We conclude therefore that an unknown protein that shares many of Crb’s properties – undergoing AP-2-dependent endocytosis, traversing the recycling pathway, and stimulating aPKC activity – serves as a critical regulator of imaginal disc polarity. The identification of this cargo is a current goal.

Page 56: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

46

Materials and Methods Fly Stocks and Genetics

WT control flies were isogenized w;FRT40, w;FRT42, or w;;FRT82. Due to the similar mutant phenotypes of AP-2 complex subunits, representative experiments carried out with AP-2α40-31 FRT40A [111] allele are labeled AP-2. Additional stocks used include shiFL54 FRT19A and chc3 FRT19A[111], lgl4FRT40A,bazXI106FRT9-2, fab121 FRT42[121], Vps25A3 FRT42[55], UAS-aPKCWTCAAX [122], scrib1, scrib2 FRT82B, dlg40.2[123], Vps45GG11 FRT82B[54], enGAL4, UAS-hTfR [124] , crb11A22 FRT82B [125], Pbac{PB}Vps45c10943 [90], and Rab5 FRT40A [48]. Mutant eye discs were generated with w l(1)CL8.7 P[m-w+ arm-lacZ] FRT19A/FM7a; eyFLP , eyFLP cl GMR-hid FRT40A and eyFLP cl GMR-hid FRT82[53,111]. eyFLP; ub-GFP FRT42D was used to generate mosaic eye disc clones. Follicle cell clones were generated using the following stocks: ub-GFP FRT9-2; e22c-GAL4 UAS-FLP, w; ub-EGFP FRT40A; GR1-GAL4 UAS-FLP, and w; e22c-GAL4 UAS-FLP; ub-EGFP FRT82. Descriptions of Drosophila stocks can be found on Flybase [126]. Immunohistochemistry and Microscopy

Ovaries and L3 larvae were dissected in PBS, fixed in 4% formaldehyde in PBS for 20 min at room temperature, and subsequently stained using standard procedures [127]. Primary antibodies used were rat anti-ElaV, mouse anti-Notch ECD, mouse anti-Dlg, mouse anti-Mmp1, mouse anti-Crb (all from DSHB), rat anti-Crb (Ulrich Tepass), mouse anti-Nrg (Michael Hortsch), rabbit anti-PKCζ (Santa Cruz Biotechnology), mouse anti-hTfR (BD Biosciences), and guinea pig anti-Scrib. TRITC-phalloidin (Sigma) was used to visualize the cell cortex. All secondary antibodies used were from Molecular Probes. Eye discs used to obtain cross-sections were mounted in 2-Hydroxyethylagarose (Sigma). All tissues were mounted in SlowFade® (Molecular Probes) and imaged on a Leica TCS confocal microscope. Images were assembled with Adobe Photoshop CS4. Endocytic trafficking and quantitation

Notch and Dextran trafficking experiments were carried out as described previously [48]. Discs were mounted on slides using either double-sided tape or coverslips as spacers to preserve the tissue shape. To quantify endocytic uptake of Notch or dextran, XY sections were imaged every 0.5µm through the Z plane of the disc. These stacks were imported and analyzed in ImageJ using the LOCI Bio-Formats Importer plugin and Image5D plugin. Individual cells were identified through these stacks using F-actin staining to delineate cortical actin and the number of endocytic puncta in the volume of a given cell was manually counted. Statistical analysis was performed using a Mann-Whitney test calculator (http://faculty.vassar.edu/lowry/utest.html). For assays involving the culturing of live discs, Shields and Sang M3 Insect media (Sigma) was used and discs were incubated at room temperature. Chloroquine (Sigma) was used at a final concentration of 200µM and for the 5 hour culturing studies, was replenished in fresh M3 media halfway through the time point.

Page 57: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

47

Live trafficking of transferrin (Tfn) uptake was performed in a manner similar to that of Dextran trafficking, with hTfR-expressing discs being incubated with fluorescently-labeled human holo-Tfn (Molecular Probes) at a final concentration of 10µg/ml.

Page 58: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

48

Figure 3.1: AP-2, chc, and shi are neoplastic TSGs WT (A, E, I, M), AP-2 (B, F, J, N), chc (C, G, K, O) and shi (D, H, L, P) mutant tissue stained to assess hallmarks of neoplastic transformation; phalloidin staining to reveal F-actin is red in all images. A-D: X-Z sections of eye imaginal discs (inset shows plane of section in WT discs) demonstrate that rather than the organized epithelial monolayer of WT discs, mutant discs are overgrown, disorganized and multilayered. Nuclei are shown in blue. E-H: Neuronal differentiation (anti-Elav, green) present in WT discs is lost in mutant discs. I-L: Mmp1 (cyan) is absent from WT discs but present in mutant discs. M-P: As compared to the cuboidal monolayer of WT follicle cells (GFP, green), mutant follicle cells clones (lacking GFP) are round and form multilayers.

Page 59: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

49

Page 60: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

50

Figure 3.2: Polarity defects in endocytic TSG mutant cells Follicle cells are stained for markers of apical (A-H) or basolateral (A’-H’) membranes; merge (apical in magenta; basolateral in green) is shown in A’’-H’’. Note that the apical follicle cell membrane faces the germline; the basolateral surface is in contact with a basement membrane (not visualized). In WT follicle cells (A), the apical transmembrane protein Crb is found in a single, continuous membrane subdomain that is distinct from that occupied by the basolateral transmembrane protein Nrg. In AP-2 follicle cells (B), both Crb and Nrg are intermixed throughout the plasma membrane; areas of overlap and complementary but discontinuous localization can both be seen. In lgl follicle cells (C), a similar pattern is seen; however an additional cortical haze of Crb but not Nrg is evident. In Rab5 follicle cells (D) an expansion of Crb at the cortex of the cells is observed. Segregation of the apical cortex-associated protein aPKC from the basolateral cortex-associated protein Dlg is seen in WT cells (E, note that Dlg is also found on germline cell membranes). In AP-2 (F) and lgl (G) follicle cells, intermixing with overlap and fragmented complementary domains are seen; note that no cortical haze of aPKC is evident in lgl cells or Rab5 follicle cells (H).

Page 61: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

51

Page 62: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

52

Figure 3.3: Subcellular localization of normally polarized proteins in endocytic mutant tissue Imaginal discs genetically mosaic for endocytic mutants (homozygous mutant clones outlined with dotted lines) were stained for proteins that normally show polarized localization. Single channels are shown on left, with merge on right. aPKC (A-C and red in A”-C’’) is found at the plasma membrane in WT cells (A) and remains there in vps25 (B) and fab1 (C) mutant cells. It does not colocalize with the endocytic cargo Notch (A’-C’ and blue in A”-C’’), which becomes trapped in aberrant endocytic structures in mutant cells. Crb (D-F and yellow in D’’-F’’) is found at the plasma membrane in WT cells (D) and becomes trapped in aberrant endocytic structures in vps25 (E) and fab1 (F) mutant cells. Scrib (D’-F’ and blue in D’’-F’’) is not seen in endocytic structures.

Page 63: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

53

Page 64: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

54

Figure 3.4: Junctional scaffold neoplastic TSG mutant discs display AP-2-dependent endocytosis Notch staining (green) from live trafficking experiments in wild type (A-A”), dlg (B-B”), scrib (C-C”), lgl (D-D”), and AP-2a (F-F”) L3 eye imaginal discs, and wing discs expressing aPKCDA (E-E”). At time point zero (A-F), Notch is localized to the cell surface. After 300 minutes (A-F’), Notch staining is no longer seen in any of the discs except for AP-2a. Quantitation of endocytosed Notch (G) and Dextran (H) at 20’ time point in WT (blue points) and scrib (red points), **: p=.0011. Dextran (red) visualized with F-actin (green) after 5 hours of live trafficking in wild type (I-J’) and scrib mutant discs (K-L’). When treated with the lysosomal inhibitor chloroquine, Dextran accumulates in large intracellular puncta (J-J’, L-L’).

Page 65: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

55

Page 66: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

56

Figure 3.5: Defects in hTfR trafficking in dlg mutants are suggestive of a recycling pathway defect in junctional scaffold neoplastic TSG mutants Live trafficking of labeled holo-transferrin (green) in hTfR-expressing wild type (A-B) or dlg mutant discs (C-D) after 3 hours in the absence (A, C) or presence (B, D) of lysosomal inhibitor chloroquine. Staining for hTfR (blue) at steady-state (E, G) or after 5 hours of culturing discs in the presence of chloroquine (F, H). Wild type cells expressing hTfR (E, F) show an increase in hTfR+ intracellular puncta upon chloroquine treatment, as do dlg mutant cells expressing hTfR (G, H). Crb staining (green) in wild type (I-J’) or scrib mutant cells (K-L’) that have been cultured for 5 hours in the absence (I-I’, K-K’) or presence (J-J’, L-L’) of chloroquine. In all panels the cell cortex is visualized by F-actin staining (red).

Page 67: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

57

Page 68: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

58

Figure 3.6: Neoplastic TSGs do not control polarity and proliferation by regulating Crb levels Crb expression (green) in wild type (A-B’) and AP-2 mutant cells (C-D’) that have been cultured for 5 hours in the absence (A-A’, C-C’) or presence (B-B’, D-D’) of the lysosomal inhibitor chloroquine. The strongly increased levels of Crb (green) relative to wild type L3 eye imaginal discs (E) that are found in Vps45 mutant discs (F) are not present in AP-2 mutant discs (G). F-actin (red) and Mmp1 (green) staining in wild type (H-H’), scrib(I-I’), scrib crb (J-J’), Vps45(K-K’), and Vps45 crb(L-L’) mutant discs. Note that removing Crb does not alter the neoplastic phenotype of Vps45 or scrib mutant discs.

Page 69: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

59

Page 70: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

60

Chapter 4

Characterization of an E3 ubiquitin ligase component, supernumerary limbs, as a tumor suppressor and polarity regulator

Page 71: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

61

Abstract

Proper apicobasal polarity of epithelial cells in Drosophila is dependent upon a number of processes. Here I show for the first time that ubiquitin-mediated degradation via the SCFSlmb E3 ubiquitin ligase complex is one of those processes by identifying slmb as a neoplastic tumor suppressor gene (TSG) in Drosophila. I describe assays comparing epithelial cell polarity disruptions and signaling pathway activation in both slmb and junctional scaffold neoplastic TSG mutant discs, and conclude that slmb mutant eye discs are comparable in many ways to discs mutant for previously described neoplastic TSGs. In addition, I show that accumulation of active forms of known Slmb substrates is not sufficient to cause polarity disruption, suggesting that Slmb-mediated degradation of an unknown protein or proteins is required for proper apicobasal polarity in epithelial cells. In addition to using immunofluorescence to look for the accumulation of candidate Slmb substrates in slmb mutant tissue, I report here the results of an unbiased dominant modifier screen that was employed to identify unknown components of the Slmb polarity pathway.

Page 72: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

62

Introduction Cells must respond to their environment in rapid and efficient ways to accommodate changing conditions. As reviewed in Chapter 1, a number of proteins and/or cellular processes that regulate epithelial cell polarity have now been identified. Basolaterally-localized junctional scaffold proteins, Scrib, Dlg, and Lgl all regulate epithelial cell polarity through an as yet unidentified mechanism (reviewed in [50]), though intracellular trafficking may be involved as detailed in Chapter 3. Endocytic trafficking at multiple stages, global transcriptional regulation, and mRNA localization have also been described to regulate epithelial cell polarity and tumor suppression [48,54,55,128,129].

In this chapter I address another such post-translational regulatory mechanism, the covalent attachment to target substrates of a small protein, ubiquitin, which leads to their proteasomal degradation/processing, changes in protein trafficking, or transcriptional changes [130,131]. The basic pathway of ubiquitylation involves several steps and three core components. An E1 ubiquitin-activating enzyme passes the ubiquitin molecule to the E2 ubiquitin-conjugation protein which then binds to an E3 ubiquitin ligase that catalyses transfer of the ubiquitin to the target protein. Often more than one ubiquitin tag is added, forming a ubiquitin chain that is then recognized by the 26S proteasome which degrades the tagged protein (reviewed in [131]). Alternatively, ubiquitin tags can be recognized by components of the endocytic or other intracellular trafficking pathways to alter the localization of a protein within the cell (reviewed in [130]). The entire process is often regulated by the phosphorylation state of the target protein; when it is phosphorylated upon key residues it can be bound by its E3 ubiquitin ligase [132]. Ubiquitylation has been shown to play roles in diverse cellular processes including cell cycle progression, the regulation of growth and patterning signaling pathways, cellular migration, apoptosis and cellular polarity (reviewed in [133]).

Here I add proteasomal degradation via a specific E3 ubiquitin ligase to the growing list of cellular processes that govern epithelial cell polarity. In a genetic screen to identify regulators of epithelial cell polarity and proliferation, the E3 ubiquitin ligase substrate binding protein, Slmb, was identified. Slmb was originally described in Drosophila as a negative regulator of Hedgehog and Wingless signaling [134]. This and subsequent work showed that slmb encodes an F-box/WD40 repeat protein that serves as a substrate specificity factor for the Skp-Cullin-F-box (SCF)Slmb E3 ubiquitin ligase. Slmb binds to phosphorylated serines, often in a conserved motif, DSGXXS, and has been shown to bind to and allow for ubiquitylation of Cubitus interruptus (Ci), Armadillo (Arm), Period (Per), and SAK kinase [134-138]. Loss of Slmb leads to phenotypes associated with stabilization of these active proteins and overactivation of their respective pathways. The mammalian homolog of Slmb, β-TRCP, has many known substrates and has been characterized as an oncogene but has not been shown to play a role in regulating cellular polarity (reviewed in [139]). However, in this Chapter, I detail the characterization of slmb as a Drosophila tumor suppressor and polarity regulator, as well as my progress towards identifying the Slmb substrate(s) that regulates epithelial cell polarity via a dominant modifier screen.

Page 73: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

63

Results Characterization of slmb mutant tissue The E3 Ubiquitin Ligase component Slmb is a novel tumor suppressor in Drosophila In order to identify new regulators of epithelial polarity, we exploited a link between apicobasal polarity and imaginal proliferation control to perform the Mutant Eye No Eclosion (MENE) screen for Drosophila neoplastic tumor suppressor genes (TSGs) (described in [53]). A number of components of the endocytic pathway were identified in the MENE screen; therefore I was intrigued by one complementation group that did not show obvious evidence of endocytic trafficking defects. Deficiency mapping followed by failure to complement P{PZ}slmb00295 suggested that the gene disrupted in this complementation group may be slmb, which codes for an F-box WD40-repeat protein that functions as a specificity factor for the Skp-Cullin-F-box (SCF)slmb E3 ubiquitin ligase complex. Sequencing identified lesions in the coding region of all six slmb alleles recovered from the screen (Fig. 4.1A). The lesions in slmb3A1and slmbUU11 result in nonsense mutations causing truncation of the protein after the F-box domain and before the first WD40 domain. The alleles slmbYY11, slmb3G1, and slmbLL16 are caused by missense mutations in the 4th, 5th, and 6th WD40 repeat domains, respectively, while slmbMM8 results from a nonsense mutation causing a truncation of the protein in the 6th WD40 repeat domain. Furthermore, the lethality of slmb8/slmb3A1 transheterozygous flies was rescued by expression of wild type Slmb. Together these data support the identification of slmb as a new neoplastic TSG. Slmb mutant tissues share similarities with, and display differences from previously described tumor suppressor mutants

Characterization of eye discs made up almost entirely of slmb mutant tissue reveals hallmarks of neoplastic transformation, such as disrupted cell polarity and tissue architecture and increased F-actin staining (Fig. 4.1B-I). The previously described and widely utilized alleles, slmb1 and slmb2, show either no or little neoplastic transformation, respectively (Fig. 4.1C-D). Sequencing revealed that these alleles are missense mutations in the WD40 domain(Fig. 4.1A); the data together suggest that these alleles are not null. An additional available allele, slmb8 deletes the 5’ UTR and first exon of slmb [140]. Since the alleles recovered from the MENE screen phenocopy slmb8, we suggest that these alleles are stronger than slmb1 and slmb2 and that at least slmbUU11 and slmb3A1 appear to be molecular nulls (Fig. 4.1A).

Identifying commonalities and differences between tissues mutant for different classes of tumor suppressors will provide crucial information for understanding the mechanisms underlying loss of polarity and neoplastic transformation in these tissues. One hallmark of neoplastic tumor suppressor mutant epithelial cells is a loss of apicobasal polarity. To determine whether slmb mutant tissue displays polarity defects akin to those seen in other neoplastic tissues, I compared the localization of polarity and junctional proteins in both slmb and lgl mutant discs. Like discs mutant for the junctional scaffold neoplastic TSG lgl, slmb discs show disrupted apicobasal polarity as demonstrated by mislocalization of the apical polarity determinant Crumbs (Crb) (Fig. 4.2A-C). However, the pattern of localization in the two types of mutants is distinct, with Crb appearing to be more punctate and less associated with the membrane in slmb mutants (Fig. 4.2C and C”). The adherens junction protein Ecad displays patterns similar to those of Crb (Fig.

Page 74: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

64

4.2D’-F’). By contrast, the basolateral proteins Scrib and Dlg show similar misdistribution in both mutants (Fig. 4.2A’-C’, D-F). Therefore, while apicobasal polarity is disrupted in slmb mutant tissue, it appears that the mechanism by which this occurs may be distinct from the mechanism in junctional scaffold tumor suppressor mutant tissue.

Another shared trait of cells mutant for many neoplastic TSGs is that they are outcompeted by neighboring wild type cells in mosaic imaginal discs [141]. To investigate this trait, I generated discs mosaic for slmb mutant tissue (see Materials and Methods) and found that mutant clones for several alleles of slmb are not outcompeted by wild type tissue in L3 larval discs (Fig. 4.3), again suggesting that the mechanisms operating in slmb tumor suppression are not identical to those of other neoplastic tumor suppressors.

Like loss of apicobasal polarity, epithelial cells mutant for neoplastic TSGs also misregulate various signaling pathways. Two signaling pathways that are consistently ectopically activated in neoplastic TSG mutant discs are the JNK and JAK/STAT signaling pathways [142-145]. Slmb mutant discs also display this phenotype as demonstrated by increased Mmp1 staining (Fig. 4.4) and increased expression of a STATGFP reporter (Fig. 4.5), respectively. Though the role that these pathways play in neoplastic transformation is not fully clear, their activation in null slmb mutant cells supports the classification of slmb as a neoplastic TSG. Slmb’s tumor suppressive role involves its function in the SCFslmb E3 ubiquitin ligase complex

In order to determine whether Slmb’s role in tumor suppression is related to its known function as a component of the SCF E3 ubiquitin ligase complex, I generated discs that were mutant for other components of the SCF complex and looked for evidence of neoplastic transformation (Fig. 4.6). All SCF mutant discs caused a MENE phenotype and displayed evidence of ectopic JNK signaling, recapitulating part of the neoplastic phenotype seen in slmb mutant discs (Fig. 4.6A-D, compare to Fig. 4.4C). Though skpA and Cul1 mutant discs display disruption of tissue architecture by F-actin staining (Fig.4.6B’-C’), this disruption did not completely phenocopy the disruption seen in slmb null mutant tissue (Fig. 4.1E-I and Fig. 4.4C’). Roc1a mutant discs, however, more closely phenocopy the neoplastic phenotype seen in slmb mutant discs (Fig. 4.6D-D’). These data are consistent with a model in which Slmb’s tumor suppressor function depends upon its activity as an SCF E3 ubiquitin ligase substrate specificity factor, and furthermore implicate regulated protein degradation as a mechanism for regulating epithelial cell polarity. Co-overexpression of stabilized forms of known Slmb substrates is insufficient to recapitulate neoplastic transformation The failure to target a particular protein or proteins for degradation likely underlies the neoplastic transformation seen in slmb mutant discs. Identifying the substrate or substrates whose accumulation may be responsible for the loss of polarity and overproliferation in slmb mutant tissue is crucial for understanding the mechanism by which slmb acts as a tumor suppressor. Before considering an unknown substrate, I tested the ability of stabilized forms of two known Slmb substrates, Arm and Ci, to cause neoplastic transformation in the wing disc since both Wg and Hh signaling pathways are overactivated in slmb mutant tissue and have been implicated in growth in epithelial tissues [134]. Though co-overexpression of stabilized forms of

Page 75: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

65

Arm and Ci in the wing disc pouch leads to overgrowth (Fig. 4.7C), individual cells appear to retain polarity and general tissue architecture is maintained. In addition, in contrast to a wing disc composed almost entirely of slmb mutant tissue, no ectopic JNK signaling is observed (Fig. 4.7B-C). These data demonstrate that stabilization of effector proteins and subsequent ectopic co-activation of the Wg and Hh signaling pathways are not sufficient to cause neoplastic transformation, suggesting that another Slmb substrate normally regulates at least polarity in epithelial cells. Using immunohistochemistry to validate potential polarity-regulating Slmb substrates In slmb mutants, the turnover of Slmb substrates is blocked and leads to an accumulation of the stabilized proteins in the cell. One approach for identifying Slmb substrates is to use antibody staining of potential candidate substrates to assess whether accumulation of a particular protein occurs or not in the slmb mutant tissue. In order to verify that I can visualize differences in levels or localization of Slmb substrates, I stained slmb mutant clones for a known substrate, Arm, and compared the expression to wild type tissue. Cells that have lost Slmb function accumulate high levels of Arm throughout as compared to their wild type counterparts where most Arm is localized to the cell cortex (Fig. 4.8B-B’ and [134]). In order to make sure that any phenotype I observed was specific to Slmb function and not a phenotype common to all neoplastic tissues, I also stained for Arm in scrib mutant clones. Arm staining in scrib mutant cells was comparable to that seen in wild type cells (Fig. 4.8A-A’), suggesting that the increase seen in slmb mutants was indeed due to the failure of Arm to be degraded in the absence of Slmb. One candidate Slmb substrate of significant interest is aPKC. aPKC is a cytosolic protein like other known Slmb substrates and overexpression of an activated form can cause neoplastic transformation and loss of polarity [114]. In addition, one of its isoforms contains a canonical Slmb-binding motif. Staining for aPKC, however, indicated that both its levels and localization were similar in slmb and scrib mutant cells (Fig. 4.8C-D’). While its localization differs from that in wild type cells, this is consistent with the loss of apicobasal polarity that is seen in neoplastic tumor suppressor mutants. Though aPKC cannot be formally rejected as a candidate substrate from this work, these data do not provide support for aPKC as a Slmb substrate. Identifying components of the slmb polarity pathway via a dominant modifier screen Slmb knockdown in the wing disc produces a robust and modifiable phenotype in the adult wing While many biochemical studies have been performed to identify ubiquitin ligase substrates, including those of the Slmb mammalian homolog, β-TRCP, the utilization of a complementary genetic approach may identify substrates or regulators of the pathway that are not suited to identification via biochemical methods. To identify the putative Slmb substrate that regulates epithelial cell polarity, as well as other players in the ‘slmb polarity pathway’, I employed a dominant modifier screen. Dominant modifier screens have been successfully carried out to identify components of various signaling pathways (reviewed in [146]. The premise of such screens is that reduction of the activity of a protein of interest to a critical threshold level will allow for the identification of other proteins that function within the same pathway or cellular process in a dominant fashion. In other words, the removal of only one copy

Page 76: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

66

of an interacting gene in the sensitized background is sufficient to dominantly modify a visible phenotype.

In my screen, reduction of Slmb function in the wing is achieved using RNAi knockdown by expressing a UASslmbIR transgene specifically in the developing dorsal wing pouch using the MS1096-Gal4 driver. The resulting phenotype - adult wings that are smaller than wild type wings, have defective patterning, and are often blackened or shriveled - is due to reduced Slmb function specifically as it is rescued by expression of UASslmb (Fig. 4.9) which has no phenotype when expressed on its own (data not shown). This phenotype is also modifiable: it is enhanced by reducing by half the activity of various components of the SCFSlmb complex (Fig. 4.10A-D), but not modified by removing a copy of Roc2 (Fig. 4.10E), which encodes for a protein associated with a distinct Cullin-based E3 ubiquitin ligase complex. While the cause of the adult wing phenotype is not completely understood, it is at one end of a spectrum of phenotypes that includes neoplastic growth: improving the efficiency of slmb knockdown by co-expression of UASDcr2 causes larval death and neoplastic transformation in the wing disc (Fig. 4.11). My expectations are that removing a copy of the gene that codes for the polarity-regulating Slmb substrate will cause a suppression of the slmb knockdown phenotype because there will be a decrease in the accumulation of this substrate. Other components of the slmb polarity pathway may have differing effects on the knockdown phenotype depending upon their role within the pathway. The slmb dominant modifier screen using Deficiencies on chromosomes II and III reveals enhancers and suppressors of the slmb knockdown phenotype

To take an unbiased approach to the slmb dominant modifier screen (described in Materials and Methods), I used a collection of Deficiencies from chromosomes II and III that together uncover approximately 80% of the Drosophila genome. The Deficiencies selected were from three isogenic collections in an attempt to minimize the effects of different genetic backgrounds. For chromosome II, nine Deficiencies enhanced the slmb knockdown wing phenotype, with two of them enhancing to lethality and no Deficiencies suppressing the phenotype (Table 4.1). For chromosome III, 12 enhancers and 3 suppressors were found (Table 4.2). Examples of a strong enhancer and strong suppressor can be seen in Fig. 4.12. In order to narrow down the candidate genes uncovered by the strongest suppressor, Df(3L)BSC443, I crossed the slmb knockdown flies to other Deficiencies that overlapped that region of the chromosome. This sub-Deficiency mapping revealed that loss of a gene product within the cytological limits 72C1-72D1 was responsible for the suppression of the slmb knockdown phenotype (Table 4.3). Candidates within that region were tested by the use of available mutant stocks or RNAi lines, but thus far no candidates have suppressed to the same extent as the suppressor Deficiencies (Table 4.4). While none of the tested candidates suppress to the same extent as the original suppressor Deficiency, reducing the activity of brahma or CG5830 (Scp1) results in mild suppression of the slmb knockdown phenotype and therefore may be of interest for further study. In order to identify one of the strongest enhancers, Deficiencies that partially overlap with lethal enhancer, Df(3R)BSC489, were tested for their ability to cause lethality when crossed to the slmb knockdown line (Table 4.5). The resulting interactions indicate that the reduction of a gene product (or products) within the cytological band 95B1 enhances the slmb knockdown phenotype in the wing to cause lethality in the larvae. Perhaps not surprisingly, two genes within

Page 77: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

67

this region, Rpn9 and Tat, encode for subunits of the proteasome and the reduction of these gene products possibly enhances the slmb phenotype to lethality. Though not yet tested, if removing copies of these two components of the proteasome could enhance the slmb knockdown phenotype to neoplastic transformation, this would support degradation or processing of a substrate rather than its trafficking, as the mechanism regulating cell polarity.

Another Deficiency that enhanced to lethality, Df(2L)Exel6011, had few overlapping Deficiencies, but the one that was tested, Df(2L)BSC693, also enhanced the slmb knockdown phenotype to lethality. This narrows down the candidate enhancers to the cytological region 25C10-25D5 and includes the following list of genes: Cyp28d1, CG7242, CG14034, TpnC250, tkv, tRNA:CR31971, tRNA:CR31914, Cyp4ac1, Cyp4ac2, Cyp4ac3, Bsg25D, Bub1, and vri. Based upon findings that are reported below, reduction of translational machinery activity enhances several wing knockdown phenotypes including that of slmb, suggesting that the lethal interactors in these Deficiencies may be the tRNAs.

Finally, the other Deficiency that enhanced to lethality was Df(2L)BSC354 which uncovers a small enough region that candidate genes were tested directly (Table 4.6). Of the candidates tested thus far, none has recapitulated the lethal phenotype. Identifying components of the slmb polarity pathway by a candidate modifier approach

In addition to this unbiased approach of using Deficiency kits to screen for slmb interactors, a candidate approach was also employed to try to identify other components of the slmb polarity pathway. Initially, a number of potential substrates, components of the SCF complex or other E3 ubiquitin ligase complexes, kinases involved in polarity regulation, other known polarity regulators, and other known tumor suppressors were tested for interaction with slmb (Table 4.7), either by obtaining alleles of, RNAi lines against, or Deficiencies that uncover the candidates. As seen in the screen, there was a skewing toward enhancement of the slmb phenotype rather than suppression, which could in part be due to the number of signaling pathways that are required for proper wing development.

The Deficiency that uncovers AmpKα significantly enhanced the slmb knockdown phenotype, but rather than AmpKα interacting with slmb, it appears that removal of a ribosomal subunit stubarista (sta) may be sufficient to generate lethality and even neoplastic growth (Table 4.8 and data not shown). Knockdown of sta in the wing is lethal even in the absence of slmbIR, but the fact that removing one copy of another ribosomal subunit (Table 4.7, see Rpl14) also significantly enhances the slmb knockdown phenotype suggests that this interaction may be an artifact of reducing the amount of slmb activity even further, via reduced translation of the protein. Another interactor was recovered from this candidate Deficiency, but surprisingly when Sp95 is knocked down, it causes partial suppression rather than enhancement of the slmb knockdown phenotype, suggesting that it might play a role in the Slmb polarity pathway. Further assessment of its role is ongoing.

Page 78: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

68

Discussion

In this work I have identified the E3 ubiquitin ligase substrate binding protein, Slmb, as a neoplastic tumor suppressor and regulator of epithelial cell polarity in Drosophila. In addition I have characterized slmb mutant discs with respect to their phenotypic similarities to and differences from junctional scaffold neoplastic TSG mutant discs. Finally I have made inroads into identifying and ruling out potential Slmb substrates that regulate epithelial cell polarity by employing a dominant modifier screen. It is becoming increasingly appreciated that the regulation of polarity proteins via ubiquitin-mediated degradation can play an important role in cellular polarity. Both TRIM2 and Smurf2 have been shown to play important roles in the degradation of key polarity proteins to generate proper neuronal polarity [147-150]. The E3 ubiquitin ligase Smurf1 associates with PKCζ in protrusions of cell culture fibroblasts where it regulates polarity by localized degradation of RhoA [151]. Other E3 ubiquitin ligase components have been shown to play roles in planar cell polarity both in mammalian and Xenopus cells [152,153] as well as A/P polarity in the C. elegans embryo [154]. Though the process of ubiquitylation and proteasomal degradation has been previously been described as a tumor suppressive mechanism in Drosophila imaginal discs with the SCF-Ago complex regulating levels of Myc and CyclinE to prevent tissue overgrowth [155], it has not been described as a regulator of epithelial cell polarity in Drosophila imaginal discs. However, loss of the VHL E3 ubiquitin ligase and mammalian tumor suppressor causes multilayering and polarity defects in the follicle cell epithelium in Drosophila, thought to be due to its role in stabilizing microtubules, as well as (indirectly) aPKC, though whether this relates to its E3 ubiquitin ligase function has yet to be thoroughly demonstrated [156]. Determination of the mechanism by which Slmb functions to regulate the apicobasal polarity pathway will contribute to our general understanding of both epithelial cell polarity and neoplastic tumor suppression in Drosophila.

Comparison of the phenotypes seen in junctional scaffold, endocytic and slmb neoplastic TSG mutant discs may give us valuable information about the way that polarity is disrupted in these cells. The shared phenotypes of discs mutant for the different classes of neoplastic TSGs, which include loss of apicobasal polarity, increased F-actin expression, multilayering of cells, and the upregulation of JNK and JAK/STAT signaling pathways, suggest that the way in which these tumor suppressors impinge upon polarity and proliferation is shared in many respects. In addition to the aforementioned shared phenotypes, endocytic trafficking of some cargo may be disrupted or proceed with altered kinetics in discs mutant for the different classes of neoplastic TSGs. I describe this phenomenon for the junctional scaffold neoplastic TSG mutant discs in Chapter 3, and preliminary data suggests that there may be a reduced rate of endocytosis in slmb mutant discs, but not an absolute block. These data suggest that common polarity and other signaling pathways may be disrupted in similar ways in the various classes of neoplastic TSG mutants.

On the other hand, differences between the different classes may also help to shed light on the pathways that normally function in polarity and proliferation regulation. One striking difference seen between the two classes of neoplastic TSG mutants is that transmembrane apical or sub-apical junctional proteins, Crb and Ecad respectively, show a significant decrease at the membrane in slmb mutant cells, while they appear to expand their domain and stay associated with the membrane in junctional scaffold mutants. However, the cytosolic apically-localized polarity regulator aPKC appears expanded and membrane-localized in mutant discs of both

Page 79: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

69

classes, as do the basolateral junctional proteins Dlg and Scrib. This suggests that Slmb-mediated degradation of a particular substrate may serve to maintain proper localization and retention of Crb and Ecad at the membrane and that once it accumulates in slmb mutant tissue, misregulation occurs. Though Crb function can be lost at this stage of eye disc development with little immediate phenotypic consequence, the loss of both Crb and Ecad from the membrane may be sufficient to allow for a mixing of normally spatially distinct apical and basolateral domains leading to similar downstream effects as are seen in other neoplastic TSG mutant discs. This type of polarity disruption might then impinge upon common signaling pathways which could explain why JNK and JAK/STAT signaling pathways are ectopically activated in both classes of neoplastic TSG mutants. One inconsistency with this model is that the overexpression of Crb is sufficient to cause polarity loss and neoplastic transformation; it would therefore be difficult to reconcile how its loss or reduction would recapitulate the same phenotype.

Another assay in which junctional scaffold neoplastic TSG mutant cells and slmb mutant cells differ is the ability of mutant clones to survive in mosaic tissue. While JNK-mediated apoptosis has been implicated in the loss of neoplastic TSG mutant cells that are surrounded by wild-type tissue [157], and JNK signaling is upregulated in slmb mutant cells (this work), it is possible that the other pathways being ectopically activated in slmb mutants, such as the Wg and Hh pathways, are sufficient to block this apoptosis. Further study into this phenomenon may reveal more about how transformed cells can be protected from apoptosis.

Using what I know about epithelial cell polarity, neoplastic tumor suppression, and Slmb’s cellular function, I can speculate as to the nature of the substrate that may be accumulating in slmb mutant epithelial cells that can lead to loss of polarity and overproliferation. One very appealing candidate substrate is aPKC for reasons mentioned previously, including its role as a master polarity regulator. However, I do not find evidence that it is accumulated in slmb mutant tissue using immunofluorescence. Because a number of other lines of reasoning strongly support aPKC as a candidate, however, collaborative efforts are underway to directly test this. Arguments can also be made for the investigation of other known polarity regulators as candidate Slmb substrates. One of these, Dlg, is an obvious candidate due to the fact that it is a known substrate for the mammalian homolog, β-TRCP [158]. Expansion of Dlg around the cell membrane is observed in both slmb and lgl mutant discs, though no apparent accumulation above and beyond that seen in lgl mutants is seen in slmb mutants as might be expected if it were a Slmb target. The expansion itself is a disruption in polarity, but it would be difficult to argue causation of the polarity and neoplastic phenotype by failure to degrade Dlg, since dlg is a classic junctional scaffold neoplastic TSG. However, it cannot be ruled out as there may be some way to explain these apparent discrepancies and ongoing work will focus on Dlg as a candidate substrate.

A number of candidate substrates, kinases, and other potential players in the Slmb epithelial cell polarity pathway were evaluated using a genetic interaction assay in the adult wing. In addition to these candidates, a number of candidates have been recovered from the unbiased dominant modifier screen. Furthermore, global protein stability profiling in mammalian cells [159], a biochemical screen in Xenopus, coupled with bioinformatic work [160,161], and numerous studies on β-TRCP (reviewed in [139]) have all generated lists of potential candidate substrates that I have further narrowed down to test in the slmb knockdown genetic interaction assay (Table 4.9). For all candidates that show significant modification of the phenotype, follow-up studies will involve the modification of a slmb hypomorphic allele neoplastic phenotype in the eye disc to verify that the modification is related to the Slmb polarity

Page 80: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

70

pathway. Collaborative efforts will also allow us to verify whether or not potential candidate substrates accumulate in slmb knockdown S2 cells.

While the dominant modifier screen was initially promising, the followup to identify interactors has proven to be difficult. In particular, substrate identification has remained elusive. One clear impediment is that Deficiencies uncover a number of genes, some of which may have opposing effects upon the phenotype being modified. For example, in the attempt to narrow down the enhancer from one Deficiency using RNAi stocks against the candidate genes in the region, I identified an RNAi stock that partially suppressed the slmb knockdown phenotype. Therefore, though a large percentage of the genome was screened, some interactors could have easily been masked. Perhaps a complementary screen could be performed using RNAi stocks or a pBac library so that interactions with single genes can be more easily interpreted, the tradeoff being, of course, less genomic coverage.

Several other modifier screens have been performed in the lab to identify components of polarity/tumor suppressor pathways, two relying upon adult wing phenotypes caused by RNAi, one against lgl and the other against the ESCRT component gene Vps25, and another based upon overexpression of an activated form of Crb in the pupal eye that causes an adult eye phenotype. Comparisons between the four screens may give us clues about whether there exist common players in each of these pathways. While Df(3R)ED5416 strongly enhanced lgl and Vps25 knockdown and Crbintra overexpression phenotypes [162], it showed only very mild interaction with the slmb knockdown phenotype suggesting that perhaps the Slmb polarity pathway is distinct in particular ways from a pathway common to lgl, Vps25, and Crb. It is interesting to note, however, that Df(3L)BSC443 and Df(3L)ED4606 both significantly suppressed the slmb and Vps25 knockdown phenotypes. While it was predicted that suppression of the Vps25 knockdown was due to the removal of a copy of mib (T. Vaccari), thereby reducing the ectopic Notch pathway signaling known to occur in Vps25 mutants, removing a copy of mib did not suppress the slmb knockdown phenotype and has not been tested in the Vps25 knockdown. Therefore, it remains to be seen whether a component common to both endocytic and Slmb polarity pathways is uncovered by this Deficiency. While certain Deficiencies that caused lethality or strong enhancement in the slmb knockdown screen (Df(3R)BSC492 and Df(3R)Exel6214) also enhanced lgl and Vps25 knockdown phenotypes, potentially more revealing is that two Deficiencies (Df(3R)BSC489 and Df(3R)BSC497) that enhanced to lethality in the slmb knockdown screen showed no effect or even mild suppression in the other screens. Though all known Slmb substrates to date are thought to be degraded or processed upon ubiquitylation, it is formally possible that Slmb-mediated ubiquitylation serves to stabilize a substrate, as observed recently for β-TRCP and Myc [163]. If this were true of the polarity-regulating substrate, knockdown of Slmb function would actually cause a decrease in the substrate, and removal of one copy of the substrate would lead to enhancement of the phenotype. Therefore, enhancers, in addition to the possibility that they act at other steps in the Slmb polarity pathway, could also be potential substrates. Ongoing work will involve testing candidates uncovered by Deficiencies that modify the slmb knockdown phenotype with an eye to the results of the other above-mentioned dominant modifier screens to inform the direction of the work. The outcome of this work will inform us as to the utility of these types of dominant modifier screens for future studies.

Page 81: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

71

Materials and Methods Fly Stocks and Genetics The slmb alleles were generated using EMS mutagenesis [53]. Molecular lesions in these alleles were identified by sequencing amplified genomic DNA isolated from heterozygous adults using polymerase chain reaction. Details and primer sequences are available upon request. Additional fly stocks used include: P{PZ}slmb00295 [90], slmb1 FRT82B and slmb2 FRT82B [134], slmb8 FRT82B [140], 10XSTAT92E-GFP [164], skpA1FRT19A [165], Roc1aG1FRT19A [166], Cul1EX FRT42D [167], UASciM1-4 [168], UASArmS10 [169], scrib2 FRT82B [98], UASSlmb [136], and lgl4 FRT40A [170]. UASslmbIR ( P{GD10682}v34274) was obtained from the Vienna Drosophila RNAi Center (VDRC) and other fly stocks were obtained from the researchers who generated them or the Bloomington Stock Center. RNAi stocks used for candidate testing in the dominant modifier screen were obtained either from VDRC or Fly Stocks of National Institutes of Genetics. Discs containing mostly homozygous mutant cells were generated as previously described [111]. Entirely mutant wing discs were generated using the following stock UbxFLP/FM7;;cl FRT82B/TM6B. slmbUU11mosaic clones in eye discs were generated using an eyFLP;;ubGFPFRT82B stock while scrib2 eye disc clones were generated using an act>stop>GAL4 UASGFP/CyO; eyFLP cl GMRhid FRT82B/TM6B stock. Immunohistochemistry and Microscopy Antibody staining was carried out using standard conditions [98]. The following primary antibodies were used: mouse anti-Crb, rat anti-DCAD2, mouse anti-Mmp1, mouse anti-Arm, mouse anti-Dlg (all from Developmental Studies Hybridoma Bank, see references therein), guinea pig anti-Scrib, and rabbit anti-PKCζ (Santa Cruz Biotechnology). To visualize F-actin I used TRITC-phalloidin (Sigma) and secondary antibodies were from Molecular Probes. Images are single sections taken with a Leica TCS confocal microscope and assembled in Adobe Photoshop CS4. Dominant Modifier Screen

The screen was performed with the intention of identifying modifiers of the following stock: MS1096-GAL4;;UASslmbIR, which has a slmb knockdown phenotype specifically in the adult wing (further description in Results section). The screen was performed using Deficiency kits for chromosomes II and III which were obtained from the Bloomington Fly Stock Center. Deficiency stocks on chromosome II were originally balanced over CyO, a balancer that causes curly wings. Since the screen relies upon a wing phenotype, all chromosome II Deficiency stocks were first outcrossed to L/CyO flies, and males containing both the Deficiency and the dominant eye marker L were selected for the screen. For each cross, an attempt was made to observe at least 10 female flies containing both the driver, the RNAi transgene, and the Deficiency. These wings were compared to the wings of female flies carrying the driver and the RNAi transgene (baseline control) and assigned a score based upon whether they showed, on average, suppression, no change, or enhancement as compared to the baseline phenotype. Flies with wings that were comparable to wild type wings would be assigned a score of -3, no change

Page 82: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

72

from the slmb knockdown wing phenotype were assigned a score of 0, and the most severe enhancement a score of +3. Several Deficiencies also enhanced to lethality. On average, in the entire screen there is a trend toward enhancement of the phenotype when the tester flies are crossed to any Deficiency stock (Tables 1 and 2), and a cutoff for potential Deficiencies of interest was made at a score of 2 or higher. In general, suppressors were less common and a cutoff for potential Deficiencies of interest was made at a score of -1.5 or below. Select female adult wings with enhancer and suppressor phenotypes were mounted in Euparal (Carolina Biologicals) and imaged using a Leica Z16 APO microscope and DFC300 FX camera. Images were assembled in Adobe Photoshop CS4.

Page 83: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

73

Figure 4.1: Identification of slmb as a tumor suppressor Schematic of slmb coding region along with molecular lesions identified in both previously described alleles and alleles recovered in the MENE screen. Slmb8 was previously described as a deletion of the coding region (A). F-actin stain of wild type eye disc (B) and eye discs composed mostly of slmb mutant tissue (C-I). Previously isolated alleles slmb1 and slmb2 do not cause complete neoplastic transformation, as is seen with discs mutant for previously described and molecularly null allele, slmb8. All slmb mutants isolated from the MENE screen cause complete neoplastic transformation of the mutant eye discs.

Page 84: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

74

Page 85: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

75

Figure 4.2: Apicobasal polarity is disrupted in slmb mutant tissue Crb (green in A-C”) and scrib (red in A-C”) localization in the cells of a wild type L3 eye imaginal disc (A-A’’). Crb is normally apically localized with scrib localizing to baso-lateral junctions. Wild type localization is disrupted in the junctional scaffold neoplastic tumor suppressor lgl4 mutant discs (B-B’’) and disruption of apicobasal polarity is also seen in slmbYY11 mutant discs (C-C’’). Dlg (green in D-F”) and E-cad (red in D-F”) localization in the cells of a wild type L3 eye imaginal disc (D-D’’). Dlg is normally localized baso-laterally at the septate junction with E-cad localizing to adherens junctions. Wild type localization is disrupted in the junctional scaffold neoplastic tumor suppressor lgl4 mutant discs (E-E’’) and disruption of junctions is also seen in slmbYY11 mutant discs (F-F’’).

Page 86: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

76

Page 87: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

77

Figure 4.3: Slmb mutant clone survival ability depends upon allele Clones of wild type cells (A) or clones of various slmb alleles (B-E) were generated in a wild type background in developing eye discs. GFP marked tissue (green) represents wild type cells. Cells mutant for slmb2, a hypomorphic allele, and slmb8, a molecularly null allele, both appear to outcompete their surrounding wild type cells, or at the very least are not outcompeted as many tumor suppressor mutant cells are. While slmb3A1 mutant cells also do not appear to be outcompeted at this stage of development, slmb3G1 clones are rarely recovered.

Page 88: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

78

Page 89: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

79

Figure 4.4: Slmb mutant discs display ectopic JNK pathway signaling as seen in other neoplastic tumor suppressor mutant discs Whereas wild type (A-A’) L3 eye discs do not express Mmp1 (green), slmb2 (B-B’), and slmb8 (C-C’) L3 eye discs show ectopic expression of Mmp1, a target of the JNK signaling pathway.

Page 90: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

80

Page 91: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

81

Figure 4.5: Slmb mutant discs display ectopic JAK/STAT signaling Wild type L3 wing disc with stereotypical JAK/STAT signaling pattern as visualized by a GFP reporter downstream of 10 STAT binding elements (green) (A) and an L3 wing disc composed almost entirely of slmbUU11 mutant tissue displaying an expansion of the JAK/STAT signaling domain and an overall increase in JAK/STAT signaling (B) as has been seen in other neoplastic tumor suppressor mutant discs.

Page 92: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

82

Page 93: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

83

Figure 4.6: Discs mutant for other components of the SCF complex partially phenocopy slmb mutant disc neoplastic transformation Mmp1 expression (green) and F-actin staining (red) in wild type eye disc (A-A’) and skpA, Cul1, and Roc1a mutant eye discs (B-D’). While some Mmp1 staining is seen in skpA and Cul1 mutant discs, the Roc1a mutant disc phenotype most closely resembles the slmb null mutant disc phenotype.

Page 94: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

84

Page 95: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

85

Figure 4.7: Co-overexpression of stabilized forms of Ci and Arm in the developing wing disc is insufficient to cause neoplastic transformation Mmp1 expression (green) and F-actin staining (red) in a wild type L3 wing disc (A), an L3 wing disc composed almost entirely of slmbUU11 mutant tissue (B), and a wing disc with co-expression of stabilized forms of Ci and Arm being driven in the dorsal wing pouch using the MS1096-Gal4 driver (C). Though overgrowth is apparent, no evidence of neoplastic transformation is observed.

Page 96: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

86

Page 97: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

87

Figure 4.8: Known substrate Arm accumulates in slmb mutant cells, but candidate substrate aPKC does not Arm expression (green in A-B’) in scrib2 (A-A’) and slmbUU11 (B-B’) mutant clones. While Arm localization in both wild type and scrib mutant cells is similar, Arm appears both in the cytosol and at the membrane of slmb mutant cells suggesting an accumulation of Arm that is not being degraded. aPKC localization (green in C-D’) in scrib2 (C-C’) and slmb8 (D-D’) mutant clones. Unlike Arm, a known substrate for slmb-mediated degradation, aPKC does not show an accumulation in slmb mutant cells. However, as in scrib mutants, where apicobasal polarity is also disrupted, aPKC does have disrupted localization as compared to wild type cells. The cell cortex is visualized by F-actin staining (red) and the mutant clones are outlined by white-dashed lines.

Page 98: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

88

Page 99: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

89

Figure 4.9: Slmb knockdown in the wing causes a visible phenotype that is rescued by expression of wild type slmb Example of a wild type adult wing (A). Expression of a slmb RNAi construct in the developing wing pouch leads to an adult wing phenotype (B) that is rescued by co-expression of slmb (C).

Page 100: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

90

Page 101: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

91

Figure 4.10: Slmb knockdown phenotype is enhanced by reduction of SCF complex member function A representative slmb knockdown adult wing (A). Reducing slmb function even further by crossing to slmbUU11 causes enhancement of the phenotype (B), as does reducing the function of SCF core complex members Cul1 or Roc1a (C and D). Reducing the function of a component of an unrelated Cullin-based E3 ligase complex, Roc2, does not modify the slmb knockdown phenotype (E).

Page 102: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

92

Page 103: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

93

Figure 4.11: Increasing the efficiency of slmb knockdown in the wing disc causes neoplastic transformation A wild type L3 wing disc with normal Mmp1 (green) and F-actin (red) staining (A), and a wing disc co-expressing slmbIR and Dcr2 showing increased Mmp1 staining and neoplastic transformation (B).

Page 104: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

94

Page 105: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

95

Figure 4.12: The slmb knockdown wing phenotype can be suppressed or enhanced An adult wing with reduced slmb function (A). An example of suppression almost to wild type wing morphology, therefore scoring a -3 on the scoring scale (B). An example of severe enhancement of the slmb knockdown wing phenotype, scoring +3 (C).

Page 106: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

96

Page 107: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

97

Table 4.1: Results of Slmb Dominant Modifier Screen on Chromosome II Chromosome II Deficiency Kits were obtained from the Bloomington Stock Center and males containing Df/CyO were first crossed to L/CyO or Gla/CTG virgins. From these crosses, males containing Df/L or Df/Gla were selected and then crossed to MS1096-Gal4/FM7;;UASslmbIR/TM6B virgins. Crossing iso82 males to MS1096-Gal4/FM7;;UASslmbIR/TM6B virgins generated MS1096-Gal4/+;;UASslmbIR/FRT82 F1s whose wing phenotype was used as a baseline control. Scores were given as follows: -3 (total suppression) to +3 (severe enhancement) with 0 being no interaction as compared to baseline controls. When enhancement to lethality occurred, “lethal” was indicated rather than a numerical score. The wings of adult females specifically were used for scoring, as males had a much more severe phenotype making enhancement particularly challenging to score reliably. Every effort was made to base a score upon the analysis of at least 10 females, however, there were some instances in which less than 10 females were scored. In these cases, at least 5 females were used to generate an interaction score.

Page 108: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

98

Deficiency Cytology Interaction score Df(2L)ED5878 21B1;21B3 0 Df(2L)BSC106 21B7;21C2 -1 Df(2L)ED94 21E2;21E3 0 Df(2L)ED108 21F1;22A1 -1 Df(2L)ED7762 22A6;22D3 0 Df(2L)Exel6007 22D1;22E1 1 Df(2L)ED136 22F4;23A3 0 Df(2L)ED4651 23B8;23F3 -1 Df(2L)ED247 24A2;24C3 0 Df(2L)Exel6009 24C3;24C8 1 Df(2L)BSC295 24D4;24F3 2.5 Df(2L)BSC51 25A1;25C3 0 Df(2L)Exel6011 25C8;25D5 lethal Df(2L)BSC169 25E5;25F3 -1 Df(2L)ED347 25F5;26B5 0 Df(2L)ED384 26B2;26D7 0 Df(2L)BSC354 26D7;26E3 lethal Df(2L)BSC188 26F1;27A2 1 Df(2L)ED441 27A1;27E1 -1 Df(2L)ED489 27E4;28B1 0 Df(2L)BSC191 28C1;28D3 2 Df(2L)Exel7034 28E1;28F1 0 Df(2L)ED578 28F1;29A3 0 Df(2L)ED629 29B4;29E4 0 Df(2L)ED678 29F5;30B12 0 Df(2L)ED690 30B3;30E4 1 Df(2L)BSC50 30F5;31B1 -1 Df(2L)ED748 31B1;32A5 0 Df(2L)BSC230 32A5;32C1 2 Df(2L)Exel6028 32D5;32E4 0 Df(2L)BSC237 32F2;33B5 1 Df(2L)ED761 33A2;33E5 0 Df(2L)ED776 33E4;34A3 0 Df(2L)BSC277 34A1;34B2 2 Df(2L)BSC252 34D1;34F1 0 Df(2L)ED793 34E4;35B4 0 Df(2L)ED3 35B2;35D1 0 Df(2L)ED1054 35B10;35D4 1 Df(2L)ED1102 35F12;36A10 0 Df(2L)BSC148 36C8;36E3 0

Page 109: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

99

Deficiency Cytology Interaction score Df(2L)BSC149 36F5;36F10 1 Df(2L)ED1203 36F7;37C5 1 Df(2L)ED1303 37E5;38C6 -1 Df(2L)ED1315 38B4;38F5 0 Df(2L)ED1378 38F1;39D2 0 Df(2L)ED1473 39B4;40A5 0 Df(2R)M41A10 h38R--h46 0.5 In(2R)bwVDe2LCyR h42--42A3 0 Df(2R)BSC630 41D3;41F11 1 Df(2R)ED1484 42A2;42A14 2.5 Df(2R)ED1612 42A13;42E6 0 Df(2R)ED1673 42E1;43D3 0 Df(2R)ED1725 43E4;44B5 -0.5 Df(2R)BSC267 44A4;44C4 2 Df(2R)ED1770 44D5;45B4 0.5 Df(2R)ED1791 44F7;45F1 1 Df(2R)BSC280 45C4;45F4 0.5 Df(2R)BSC132 45F6;46B4 0.5 Df(2R)BSC298 46B2;46C7 0 Df(2R)X1 46C;47A1 0.5 Df(2R)BSC303 46E1;46F3 0.5 Df(2R)BSC281 46F1;47A9 0 Df(2R)BSC595 47A3;47F1 0.5 Df(2R)ED2219 47D6;48B6 0 Df(2R)ED2247 48A3;48D5 0.5 Df(2R)BSC199 48C5;48E4 0 Df(2R)BSC699 48D7;48E6 0 Df(2R)BSC425 48F1;49A1 0.5 Df(2R)Exel6061 48F1;49A6 1 Df(2R)vg135 49A;49E1--2 0.5 Df(2R)BSC305 49A4;49A10 1 Df(2R)Exel7121 49B5;49B12 -0.5 Df(2R)BSC485 49B10;49E6 1 Df(2R)Exel6062 49E6;49F1 1.5 Df(2R)Exel8057 49F1;49F10 0 Df(2R)BSC273 49F4;50A13 0 Df(2R)BSC274 50A7;50B4 0 Df(2R)BSC383 50C6;50D2 0 Df(2R)Exel7130 50D4;50E4 1 Df(2R)Exel7131 50E4;50F6 1

Page 110: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

100

Deficiency Cytology Interaction score Df(2R)Exel6284 51B1;51C2 1 Df(2R)BSC429 51C2;51D1 0.5 Df(2R)ED2426 51E2;52B1 0 Df(2R)ED2436 51F11;52D11 0 Df(2R)ED2457 52D11;52E7 -0.5 Df(2R)Jp6 52E3--5;52F 0.5 Df(2R)Exel9060 52E11;52F1 0 Df(2R)Exel6063 52F6;53C4 0 Df(2R)BSC550 53C1;53C6 0 Df(2R)Exel6064 53C11;53D11 1 Df(2R)ED2747 53D11;53F8 0 Df(2R)BSC331 53D14;54A1 0 Df(2R)Exel6066 53F8;54B6 1.5 Df(2R)BSC161 54B2;54B17 0.5 Df(2R)BSC355 54B16;54C3 0 Df(2R)Exel7149 54C10;54D5 0 Df(2R)BSC347 54D2;54E9 0 Df(2R)14H10W-35 54E5--7;55B5--7 0.5 Df(2R)ED3610 54F1;55C8 -0.5 Df(2R)ED3683 55C2;56C4 0 Df(2R)BSC135 56C11;56D5 0 Df(2R)BSC22 56D7--E3;56F9--12 0.5 Df(2R)BSC782 56D8;56D14 0 Df(2R)ED3728 56D10;56E2 0 Df(2R)BSC594 56E1;56F9 1.5 Df(2R)Exel7162 56F11;56F16 1 Df(2R)BSC19 56F12--14;57A4 0 Df(2R)BSC701 56F15;57A9 0 Df(2R)BSC702 57A2;57B3 0 Df(2R)PK1 57C5;57F5--6 1.5 Df(2R)BSC821 57D10;57E6 0 Df(2R)BSC664 57D12;58A3 1 Df(2R)BSC597 58A2;58F1 0 Df(2R)Exel7173 58D4;58E5 0 Df(2R)BSC598 58F3;59A1 0 Df(2R)BSC787 58F4;59B1 0 Df(2R)BSC599 59B1;59B3 0 Df(2R)BSC784 59B4;59B6 0 Df(2R)BSC769 59B7;59D9 0 Df(2R)BSC356 60B8;60C4 0

Page 111: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

101

Deficiency Cytology Interaction score Df(2R)M60E 60E2--3;60E11--12 1 Df(2R)gsb 60E9--10;60F1--2 0.5 Df(2R)BSC608 60E11;60F2 0 Df(2R)ED50004 60F5;60F5 -0.5

Page 112: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

102

Table 4.2: Results of Slmb Dominant Modifier Screen on Chromosome III Chromosome III Deficiency Kits were obtained from the Bloomington Stock Center and males containing Df/Balancer were crossed to MS1096-Gal4/FM7;;UASslmbIR/TM6B virgins. Crossing iso82 males to MS1096-Gal4/FM7;;UASslmbIR/TM6B virgins generated MS1096-Gal4/+;;UASslmbIR/FRT82 F1s whose wing phenotype was used as a baseline control. Scores were given as follows: -3 (total suppression) to +3 (severe enhancement) with 0 being no interaction as compared to baseline controls. When enhancement to lethality occurred, “lethal” was indicated rather than a numerical score. The wings of adult females specifically were used for scoring, as males had a much more severe phenotype making enhancement particularly challenging to score reliably. Every effort was made to base a score upon the analysis of at least 10 females, however, there were some instances in which less than 10 females were scored. In these cases, at least 5 females were used to generate an interaction score.

Page 113: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

103

Deficiency Cytology Interaction Score Df(3L)Exel6083 61A6;61B2 0.5 Df(3L)ED201 61B1;61C1 -0.5 Df(3L)ED4177 61C1;61E2 0 Df(3L)ED207 61C9;62A6 0 Df(3L)BSC181 62A11;62B7 1 Df(3L)ED4287 62B4;62E5 0.5 Df(3L)Exel6091 62E8;62F5 0.5 Df(3L)Exel6092 62F5;63A3 0.5 Df(3L)Bsc386 63A3;63B8 1 Df(3L)ED208 63C1;63F5 0 Df(3L)BSC368 63F1;64A4 0 Df(3L)ED4341 63F6;64B9 0 Df(3L)ED210 64B9;64C13 0 Df(3L)BSC437 64E8;65A1 0 Df(3L)BSC411 65A2;65C1 0.5 Df(3L)BSC224 65D5;65E6 0.5 Df(3L)BSC117 65E9;65F5 1 Df(3L)Exel8104 65F7;66A4 0 Df(3L)BSC375 66A3;66A19 -0.5 Df(3L)ED4408 66A22;66C5 -0.5 Df(3L)BSC389 66C12;66D8 1.5 Df(3L)ED4421 66D12;67B3 -1 Df(3L)BSC391 67B7;67C5 1 Df(3L)BSC283 67C7;67D5 0 Df(3L)ED4457 67E2;68A7 -1 Df(3L)ED4470 68A6;68E1 0 Df(3L)ED4475 68C13;69B4 0 Df(3L)ED4483 69A5;69D3 -0.5 Df(3L)ED4486 69C4;69F6 -1.5 Df(3L)ED4502 70A3;70C10 0.5 Df(3L)ED4543 70C6;70F4 1 Df(3L)ED217 70F4;71E1 0 Df(3L)BSC443 72B1;72E4 -2.5 Df(3L)ED4606 72D4;73C4 -1.5 Df(3L)ED4685 73D5;74E2 1 Df(3L)ED4710 74D1;75B11 1 Df(3L)ED224 75B1;75C6 1 Df(3L)ED225 75C1;75D4 0 Df(3L)BSC416 75D2;75E4 1.5 Df(3L)ED4782 75F2;76A1 -0.5

Page 114: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

104

Deficiency Cytology Interaction Score Df(3L)BSC448 77C6;77E4 1.5 Df(3L)ED4978 78D5;79A2 0 Df(3L)BSC223 79A3;79B3 1 Df(3L)ED230 79C2;80A4 0 Df(3L)ED5017 80A4;80C2 1 Df(3R)ED5100 81F6;82E7 1 Df(3R)ED5147 82E7;83A1 0 Df(3R)Exel6144 83A6;83B6 1 Df(3R)BSC464 83B7;83E1 0 Df(3R)BSC193 83E5;83F4 2 Df(3R)BSC467 83F1;84B2 0 Df(3R)ED7665 84B4;84E11 1 Df(3R)ED5230 84E6;85A5 -1 Df(3R)ED5330 85A5;85D1 1 Df(3R)BSC507 85D6;85D15 2 Df(3R)ED5416 85D16;85E6 0.5 Df(3R)ED5454 85E5;85F12 1 Df(3R)ED5495 85F16;86C7 0 Df(3R)ED5518 86C7;86E13 1 Df(3R)ED5559 86E11;87B11 0 Df(3R)BSC486 87B10;87E9 2 Df(3R)ED5623 87E3;88A4 0 Df(3R)ED5644 88A4;88C9 0 Df(3R)ED5664 88D1;88E3 1 Df(3R)Exel6174 88F1;88F7 0 Df(3R)BSC515 88F6;89A8 2 Df(3R)Exel7328 89A12;89B6 0 Df(3R)ED10639 89B7;89B18 -0.5 Df(3R)Exel6270 89B18;89D8 2 Df(3R)ED5780 89E11;90C1 0 Df(3R)ED5785 90C2;90D1 -1 Df(3R)BSC510 90E2;90F4 2 Df(3R)ED2 91A5;91F1 -0.5 Df(3R)ED5938 91D4;92A11 0 Df(3R)BSC517 92C1;92F13 1.5 Df(3R)ED10820 93A4;93B12 0 Df(3R)ED10845 93B9;93D4 1 Df(3R)ED6076 93E10;94A1 0.5 Df(3R)ED6096 94B5;94E7 0 Df(3R)BSC489 94F3;95D1 lethal

Page 115: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

105

Deficiency Cytology Interaction Score Df(3R)BSC492 95E7;96B17 3 Df(3R)BSC461 96B15;96D1 2 Df(3R)BSC140 96F1;96F10 0.5 Df(3R)ED6232 96F10;97D2 0 Df(3R)BSC497 97E6;98B5 lethal Df(3R)BSC460 98B6;98D2 1.5 Df(3R)Exel6210 98E1;98F5 0.5 Df(3R)ED6316 99A5;99C1 0 Df(3R)Exel6213 99C5;99D1 0 Df(3R)Exel6214 99D5;99E2 2.5 Df(3R)BSC503 99E3;99F6 1.5 Df(3R)Exel7378 99F8;100A5 1 Df(3R)ED6346 100A5;100B1 0 Df(3R)Exel6218 100B5;100C1 1 Df(3R)ED6361 100C7;100E3 -0.5

Page 116: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

106

Table 4.3: Deficiency mapping 3rd chromosome suppressor of slmb wing knockdown phenotype narrows suppressor region down to 72C1-72D1 A series of Deficiencies overlapping with the strongest suppressor recovered from the screen, Df(3L)BSC443, were crossed to the slmb wing knockdown flies to narrow down the region containing the suppressor of the slmb knockdown phenotype.

Page 117: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

107

Deficiency Cytology Interaction Score Df(3L)BSC845 71D3;72A1 0 Df(3L)BSC833 71D4;71F1 0 Df(3L)BSC575 71F1;72C1 0 Df(3L)BSC774 71F1;72D10 -2 Df(3L)brm11 71F1--4;72D1--10 -1 Df(3L)th102 71F3--5;72D12 -3 Df(3L)BSC443 72B1;72E4 -2.5 Df(3L)st-f13, Ki^1 72C1;73A4 -2 Df(3L)st-g24, Ki^1 72D1--2;73A9--10 -0.5 Df(3L)Exel6127 72D1;72D9 0 Df(3L)th117 72D2;72D5 0 Df(3L)ED220 72D4;72F1 0 Df(3L)ED4606 72D4;73C4 -1 Df(3L)st-e4, Ki^1 72D5--10;73A5--8 -1 Df(3L)Exel6128 72D9;72D10 0 Df(3L)BSC560 72D9;72E1 0 Df(3L)BSC559 72D9;72E1 0 Df(3L)st4 72D10;73C1 0 Df(3L)st-b11, Ki^1 72D10--11;73D1-2 0 Df(3L)BSC579 72D12;72E4 0 Df(3L)BSC649 72E1;72E4 0 Df(3L)BSC555 72E2;73A10 0 Df(3L)st8P 72E4;73B4 -0.5

Page 118: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

108

Table 4.4: Testing candidates in 72C1-72D1 cytological region fails to identify a suppressor of the slmb knockdown wing phenotype Using either RNAi lines or existing alleles, a number of candidates from the cytological region narrowed down by Deficiency mapping were tested for their ability to suppress the slmb knockdown wing phenotype, but none recapitulated the same degree of suppression of the phenotype. N/A indicates that the RNAi line tested had an adult wing phenotype on its own when driven with MS1096-Gal4 and was therefore not scored with respect to modification.

Page 119: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

109

Candidate Stock # Allele or IR Interaction Scores elgi 32830 IR 0 elgi 32832 IR 0 CG17032 102655 IR 0 CG17032 12098 IR 0 CG5931 43962 IR 0 brm 37720 IR N/A brm^2 3619 allele -1.5 CG17026 49200 IR 0 CG17026 49199 IR 0.5 CG17029 49566 IR 0 CG17029 49565 IR 0 CG17028 100100 IR 0 CG17028 32819 IR 0 CG17027 50075 IR 0 CG17027 103270 IR -0.5 Arf72A^N6 4107 allele 0 Arf72A 17826 IR 0.5 DNApolDelta 41027 IR N/A DNApolDelta 41028 IR N/A Hip14 8487 IR 0 CG5830 101539 IR ? mib1^3 4112 allele 1.5 mib1 27525 IR N/A mib1 27526 IR N/A Notum^d00939 19154 allele 1 Notum 13076R-2 IR 0 Notum 13076R-1 IR -0.5 CG5895 5895R-2 IR 0 CG5895 5895R-3 IR 0 th^4 5053 allele 0.5 th 12284R-2 IR 0 Mbs^EP3727 17279 allele 0

Page 120: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

110

Table 4.5: Deficiency mapping 3rd chromosome lethal enhancer of slmb wing knockdown phenotype narrows enhancer region down to cytological band 95B1 A series of Deficiencies overlapping with a lethal enhancer recovered from the screen, Df(3R)BSC489, were crossed to the slmb wing knockdown flies to narrow down the region containing the lethal enhancer of the slmb knockdown phenotype.

Page 121: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

111

Deficiency Cytology Interaction Score Df(3R)BSC137 94F1--95A4 0 Df(3R)BSC138 95A1--95A4 0 Df(3R)Exel6195 95A4--95B1 0 Df(3R)Exel9013 95B1--95B5 lethal Df(3R)Exel9014 95B1--95D1 0

Page 122: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

112

Table 4.6: Results of testing candidates uncovered by Df(2L)BSC354 for lethal enhancement of the slmb knockdown wing phenotype Using either RNAi lines or existing alleles, a number of candidates from the region uncovered by the lethal enhancer of the phenotype, Df(2L)BSC354, were tested for their ability to enhance to lethality the slmb knockdown wing phenotype, but of the candidates tested thus far, none has recapitulated the lethal enhancement.

Page 123: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

113

Candidate Stock # Allele or IR Interaction Scores CG9531 14661 allele -0.5 (mmy) not tested CG9536 27969 allele 0 Sec61alpha 25361 allele 0 Sec61alpha 10560 allele 0 DLP 21312 allele 0 DLP 24081 allele -1 (CG9542) not tested epsilonCOP 15699 allele 0 epsilonCOP 17016 allele 0 (CG9548) not tested CG31368 17311 allele 1 CG31368 22487 allele 0.5 CG9547 22487 allele 0.5 (CG9550) not tested CG31637 25529 allele 0.5 eya 2285 allele -1 (osm-6) not tested

Page 124: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

114

Table 4.7: Results of testing candidates of interest that may play a role in the slmb polarity pathway RNAi lines, alleles, and Deficiencies that reduce the function of candidate genes of interest were tested for their ability to modify the slmb knockdown wing phenotype. N/A indicates that the RNAi on its own driven by MS1096-Gal4 had an adult wing phenotype and was therefore not scored for modification.

Page 125: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

115

Candidate (allele, Df, or IR) Stock # (if applicable) Interaction Scores

ab^1d 9586 0 AmpK - Df(1)Exel6227 7704 lethal (escapers: 3) aPKC^k06403 10622 2.5 UAS-aPKCDN -0.5 arm^1 3378 2 Avl^1 -0.5 cact^1 583 2 Cul1 - Df(3R)BSC55 8941 1 Cul2^02074 11176 -0.5 Cul3^C7 0 Cul3^gft2 23866 0 Cul3 - Df(2L)ED3 6963 1 Cul3IR 11861R-1 N/A Cul3IR 11861R-2 N/A Cul3IR 109415 N/A Cul3IR 16331 N/A Cul3IR 25875 2 Cul4 - Df(2R)BSC266 26500 2 Cul4^JJ11 0 Cul5 - Df(3R)BSC806 27378 0 lgl^4 9042 0.5 Lkb1 A 1 Lkb1 - Df(3R)Exel6169 7648 0 medea - Df(3R)ED6361 24143 2.5 Par1 - Df(2R)ED3728 9067 0 Per - Df(1)64c18 936 0 RhoGEF2^4.1 9122 0 Roc1a^G1 2.5 Roc2 - Df(2R)ED2247 8912 0 RpL14/M(3) 3 SAK - Df(3L)BSC418 24922 0.5 scrib^1 1.5 sgg^1 4095 2.5 shg^G317 0 shg^R69 0 slmb^UU11 3 UAS-slmbWT -3 snail^1 25127 0 snail^18 2311 -0.5

Page 126: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

116

Candidate (allele, Df, or IR) Stock # (if applicable) Interaction Scores sydIR 35345 0 sydIR 35346 0 sydIR 101459 0 syd[2H] 0 tid^2 2 yurt - Df(3R)ED5613 9089 2.5

Page 127: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

117

Table 4.8: Results of testing candidates uncovered by enhancer Df(1)Exel6227 RNAi lines and alleles of candidate enhancers were tested for their ability to lethally enhance the slmb knockdown wing phenotype. RNAi against sta caused lethality, but * indicates that this lethality also occurred when the RNAi was expressed on its own driven by MS1096-Gal4. N/A indicates that the RNAi on its own driven by MS1096-Gal4 had an adult wing phenotype and was therefore not scored for modification.

Page 128: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

118

Candidate Stock # Allele or IR Interaction Scores png 31500 IR 1 AmpKα 1827 IR 0 AmpKα 106200 IR N/A futsch^N94 8805 allele 0 futsch^K68 8794 allele 0.5 Gr2a 18531R-1 IR 0 Gr2a 18531R-2 IR -0.5 Gr2a^f01153 18415 allele 0 CG14785 43594 IR N/A CG14785 109723 IR N/A CG14786 14786R-4 IR 0 CG14787 14787R-2 IR 0.5 CG14787 14787R-3 IR 0 ns3 14788R-2 IR N/A Ofut2 41361 IR 1 CG14777 14777R-2 IR 0 CG14778 44970 IR 0 CG14778 44969 IR 0 pck^EA97 4644 allele 0.5 pck 50306 IR 3 sp95 30977 IR 0 sp95 102148 IR -1.5 Rab27^EY05588 16660 allele 0.5 mei-38^1 4881 allele 0 mei-38 51804 IR 0 mei-38 51805 IR 0.5 CG14782 14782R-1 IR 0.5 CG14782 14782R-2 IR 0.5 CG14782 12795 IR 1 sta 14792R-1 IR lethal* sta 14792R-1 IR lethal* Nmdar2 14794R-1 IR 0 Nmdar2 14794R-3 IR 0.5 CG14795 28754 IR 1.5 CG14795 106558 IR 0 CG32810 18225 IR 0.5 CG32810 18226 IR N/A CG32810 108816 IR 0

Page 129: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

119

Table 4.9: High priority slmb substrate candidates from screens and bioinformatic work Data from several screens, bioinformatic work, and reviews regarding potential slmb homolog substrates was culled to several key potential substrates in Drosophila based mostly upon whether a conserved degron was present.

Page 130: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

120

Gene name Dros. homolog Degron in humans potential consensus

site

GSK3β site?

Notes

beta-catenin armadillo (arm) DSGIHS fully conserved -

DSGIHS Y

up in slmb mutants, not sufficient to cause loss of polarity

discs large discs large (dlg)

DSGLPS almost conserved - DDGLPS

mislocalized in slmb mutants, but not significantly increased

Prickle homolog 3 espinas DSGCAS almost conserved -

(SDD)DSGCA Y

TRIM67 Trim9 DSGYGSYTPSLKS

not completely conserved, but some of the serines and the threonine are

Y

IκBα cactus DSGLDS not aligned, but contains a DSGFIS

Growth hormone receptor

sallimus (sls) not determined DSGEYS

Jip3 Sunday driver (syd)

ESGQSS/CSGSVMS not conserved, but contains a DSGPPS Y

Page 131: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

121

Gene name Dros. homolog Degron in humans potential consensus

site

GSK3β site?

Notes

LZTS2 - leucine zipper, putative tumor suppressor 2/FEZ1

CG15365 (dFezl? Or erm?)

DSGRNS/DSGRSSSS

not conserved, but contains DSGISE Y

Gephyrin cinnamon DSGVAST not conserved, contains DSDRNS Y

IκBβ ank2 DSGLGS not conserved, contains DSDGDSGD

PRL-R PTP10D (low homology)

DSGRGS not conserved, contains DSGGGS

Claspin CG32251 DSGQGS not conserved, contains DSIKNS

WNK2 CG7177 DSGQGST/ESGFFHES

not conserved, contains KSGHRS, LSGTLKS

Y

FAM135A CG32333 DSGVESEPSSFAT not conserved, contains ESGWVSSRRSS

Y

IFNR Domeless N/A DSSVS is the closest

Page 132: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

122

Gene name Dros. homolog Degron in humans potential consensus

site

GSK3β site?

Notes

Bora bora DSGYNT no, but there is a DSGFNETS at C-term

KIF1C unc104 DSGDDS/ nothing obvious Y

WWTR1 yki, SuDx DSGSHS/

yorkie: SHS conserved/DAGSQSS/ closely spaced serines, Su(dx): some closely spaced serines

Y

stained slmb mutants for Ex (as a readout of yki activation) but staining was ambiguous (poor Ab?)

Snail wor esg sna DSGKGS

no, but lots of closely-spaced serines

Y

stained for snail but antibody does not work well in discs

PLCH1 norpA LSGSVLS/NSGESS (another isoform contains ESGKKS)

not conserved, a few closely spaced serines

RAP GEF1 gef26 DSGRGSWTSCSSGS

not conserved, lots of closely-spaced serines

TBC15 CG11490 DSGFCSYLESQDS not conserved, lots of closely-spaced serines

Page 133: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

123

References 1. Traub, L.M. (2009). Tickets to ride: selecting cargo for clathrin-regulated internalization. Nat. Rev. Mol. Cell Biol. 9, 583-596. 2. Eisenberg, E., and Greene, L.E. (2007). Multiple roles of auxilin and hsc70 in clathrin-mediated endocytosis. Traffic 6, 640-646. 3. Jovic, M., Sharma, M., Rahajeng, J., and Caplan, S. (2010). The early endosome: a busy sorting station for proteins at the crossroads. Histol. Histopathol. 1, 99-112. 4. Kumari, S., Mg, S., and Mayor, S. (2010). Endocytosis unplugged: multiple ways to enter the cell. Cell Res. 3, 256-275. 5. Rappoport, J.Z. (2008). Focusing on clathrin-mediated endocytosis. Biochem. J. 3, 415-423. 6. Takei, K., McPherson, P.S., Schmid, S.L., and De Camilli, P. (1995). Tubular membrane invaginations coated by dynamin rings are induced by GTP-gamma S in nerve terminals. Nature 6518, 186-190. 7. ROTH, T.F., and PORTER, K.R. (1964). Yolk Protein Uptake in the Oocyte of the Mosquito Aedes Aegypti. L. J. Cell Biol. 313-332. 8. Pearse, B.M. (1975). Coated vesicles from pig brain: purification and biochemical characterization. J. Mol. Biol. 1, 93-98. 9. Young, A. (2007). Structural insights into the clathrin coat. Semin. Cell Dev. Biol. 4, 448-458. 10. McNiven, M.A., and Thompson, H.M. (2006). Vesicle formation at the plasma membrane and trans-Golgi network: the same but different. Science 5793, 1591-1594. 11. Raiborg, C., Wesche, J., Malerod, L., and Stenmark, H. (2006). Flat clathrin coats on endosomes mediate degradative protein sorting by scaffolding Hrs in dynamic microdomains. J. Cell. Sci. Pt 12, 2414-2424. 12. Ungewickell, E.J., and Hinrichsen, L. (2007). Endocytosis: clathrin-mediated membrane budding. Curr. Opin. Cell Biol. 4, 417-425. 13. Mayor, S., and Pagano, R.E. (2007). Pathways of clathrin-independent endocytosis. Nat. Rev. Mol. Cell Biol. 8, 603-612. 14. Hansen, C.G., and Nichols, B.J. (2009). Molecular mechanisms of clathrin-independent endocytosis. J. Cell. Sci. Pt 11, 1713-1721. 15. Kirkham, M., Fujita, A., Chadda, R., Nixon, S.J., Kurzchalia, T.V., Sharma, D.K., Pagano, R.E., Hancock, J.F., Mayor, S., and Parton, R.G. (2005). Ultrastructural identification of uncoated caveolin-independent early endocytic vehicles. J. Cell Biol. 3, 465-476. 16. Howes, M.T., Mayor, S., and Parton, R.G. (2010). Molecules, mechanisms, and cellular roles of clathrin-independent endocytosis. Curr. Opin. Cell Biol. 4, 519-527. 17. Kosaka, T., and Ikeda, K. (1983). Possible temperature-dependent blockage of synaptic vesicle recycling induced by a single gene mutation in Drosophila. J. Neurobiol. 3, 207-225. 18. Bazinet, C., Katzen, A.L., Morgan, M., Mahowald, A.P., and Lemmon, S.K. (1993). The Drosophila clathrin heavy chain gene: clathrin function is essential in a multicellular organism. Genetics 4, 1119-1134. 19. Gonzalez-Gaitan, M., and Jackle, H. (1997). Role of Drosophila alpha-adaptin in presynaptic vesicle recycling. Cell 6, 767-776. 20. Berdnik, D., Torok, T., Gonzalez-Gaitan, M., and Knoblich, J.A. (2002). The endocytic protein alpha-Adaptin is required for numb-mediated asymmetric cell division in Drosophila. Dev. Cell. 2, 221-231.

Page 134: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

124

21. Hutterer, A., and Knoblich, J.A. (2005). Numb and alpha-Adaptin regulate Sanpodo endocytosis to specify cell fate in Drosophila external sensory organs. EMBO Rep. 9, 836-842. 22. Korolchuk, V.I., Schutz, M.M., Gomez-Llorente, C., Rocha, J., Lansu, N.R., Collins, S.M., Wairkar, Y.P., Robinson, I.M., and O'Kane, C.J. (2007). Drosophila Vps35 function is necessary for normal endocytic trafficking and actin cytoskeleton organisation. J. Cell. Sci. Pt 24, 4367-4376. 23. Dornan, S., Jackson, A.P., and Gay, N.J. (1997). Alpha-adaptin, a marker for endocytosis, is expressed in complex patterns during Drosophila development. Mol. Biol. Cell 8, 1391-1403. 24. Guha, A., Sriram, V., Krishnan, K.S., and Mayor, S. (2003). Shibire mutations reveal distinct dynamin-independent and -dependent endocytic pathways in primary cultures of Drosophila hemocytes. J. Cell. Sci. Pt 16, 3373-3386. 25. Gupta, G.D., Swetha, M.G., Kumari, S., Lakshminarayan, R., Dey, G., and Mayor, S. (2009). Analysis of endocytic pathways in Drosophila cells reveals a conserved role for GBF1 in internalization via GEECs. PLoS One 8, e6768. 26. Wells, A., Welsh, J.B., Lazar, C.S., Wiley, H.S., Gill, G.N., and Rosenfeld, M.G. (1990). Ligand-induced transformation by a noninternalizing epidermal growth factor receptor. Science 4945, 962-964. 27. Abella, J.V., and Park, M. (2009). Breakdown of endocytosis in the oncogenic activation of receptor tyrosine kinases. Am. J. Physiol. Endocrinol. Metab. 5, E973-84. 28. Grandal, M.V., and Madshus, I.H. (2008). Epidermal growth factor receptor and cancer: control of oncogenic signalling by endocytosis. J. Cell. Mol. Med. 5A, 1527-1534. 29. Roepstorff, K., Grovdal, L., Grandal, M., Lerdrup, M., and van Deurs, B. (2008). Endocytic downregulation of ErbB receptors: mechanisms and relevance in cancer. Histochem. Cell Biol. 5, 563-578. 30. Bache, K.G., Slagsvold, T., and Stenmark, H. (2004). Defective downregulation of receptor tyrosine kinases in cancer. EMBO J. 14, 2707-2712. 31. Le Roy, C., and Wrana, J.L. (2005). Clathrin- and non-clathrin-mediated endocytic regulation of cell signalling. Nat. Rev. Mol. Cell Biol. 2, 112-126. 32. Kikuchi, A., and Yamamoto, H. (2007). Regulation of Wnt signalling by receptor-mediated endocytosis. J. Biochem. 4, 443-451. 33. Chen, Y.G. (2009). Endocytic regulation of TGF-beta signaling. Cell Res. 1, 58-70. 34. Vaccari, T., and Bilder, D. (2009). At the crossroads of polarity, proliferation and apoptosis: the use of Drosophila to unravel the multifaceted role of endocytosis in tumor suppression. Mol. Oncol. 4, 354-365. 35. Carpenter, G., and Cohen, S. (1976). 125I-labeled human epidermal growth factor. Binding, internalization, and degradation in human fibroblasts. J. Cell Biol. 1, 159-171. 36. Wang, Y., Pennock, S., Chen, X., and Wang, Z. (2002). Endosomal signaling of epidermal growth factor receptor stimulates signal transduction pathways leading to cell survival. Mol. Cell. Biol. 20, 7279-7290. 37. Goh, L.K., Huang, F., Kim, W., Gygi, S., and Sorkin, A. (2010). Multiple mechanisms collectively regulate clathrin-mediated endocytosis of the epidermal growth factor receptor. J. Cell Biol. 5, 871-883. 38. Sigismund, S., Woelk, T., Puri, C., Maspero, E., Tacchetti, C., Transidico, P., Di Fiore, P.P., and Polo, S. (2005). Clathrin-independent endocytosis of ubiquitinated cargos. Proc. Natl. Acad. Sci. U. S. A. 8, 2760-2765.

Page 135: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

125

39. Sigismund, S., Argenzio, E., Tosoni, D., Cavallaro, E., Polo, S., and Di Fiore, P.P. (2008). Clathrin-mediated internalization is essential for sustained EGFR signaling but dispensable for degradation. Dev. Cell. 2, 209-219. 40. Chanut-Delalande, H., Jung, A.C., Baer, M.M., Lin, L., Payre, F., and Affolter, M. (2010). The Hrs/Stam complex acts as a positive and negative regulator of RTK signaling during Drosophila development. PLoS One 4, e10245. 41. Di Guglielmo, G.M., Le Roy, C., Goodfellow, A.F., and Wrana, J.L. (2003). Distinct endocytic pathways regulate TGF-beta receptor signalling and turnover. Nat. Cell Biol. 5, 410-421. 42. Dore, J.J.,Jr, Yao, D., Edens, M., Garamszegi, N., Sholl, E.L., and Leof, E.B. (2001). Mechanisms of transforming growth factor-beta receptor endocytosis and intracellular sorting differ between fibroblasts and epithelial cells. Mol. Biol. Cell 3, 675-684. 43. Kikuchi, A., Yamamoto, H., and Sato, A. (2009). Selective activation mechanisms of Wnt signaling pathways. Trends Cell Biol. 3, 119-129. 44. Seto, E.S., and Bellen, H.J. (2006). Internalization is required for proper Wingless signaling in Drosophila melanogaster. J. Cell Biol. 1, 95-106. 45. Rives, A.F., Rochlin, K.M., Wehrli, M., Schwartz, S.L., and DiNardo, S. (2006). Endocytic trafficking of Wingless and its receptors, Arrow and DFrizzled-2, in the Drosophila wing. Dev. Biol. 1, 268-283. 46. Devergne, O., Ghiglione, C., and Noselli, S. (2007). The endocytic control of JAK/STAT signalling in Drosophila. J. Cell. Sci. Pt 19, 3457-3464. 47. Fortini, M.E. (2009). Notch signaling: the core pathway and its posttranslational regulation. Dev. Cell. 5, 633-647. 48. Lu, H., and Bilder, D. (2005). Endocytic control of epithelial polarity and proliferation in Drosophila. Nat. Cell Biol. 12, 1232-1239. 49. Vaccari, T., Lu, H., Kanwar, R., Fortini, M.E., and Bilder, D. (2008). Endosomal entry regulates Notch receptor activation in Drosophila melanogaster. J. Cell Biol. 4, 755-762. 50. Bilder, D. (2004). Epithelial polarity and proliferation control: links from the Drosophila neoplastic tumor suppressors. Genes Dev. 16, 1909-1925. 51. St Johnston, D., and Ahringer, J. (2010). Cell polarity in eggs and epithelia: parallels and diversity. Cell 5, 757-774. 52. Hariharan, I.K., and Bilder, D. (2006). Regulation of imaginal disc growth by tumor-suppressor genes in Drosophila. Annu. Rev. Genet. 335-361. 53. Menut, L., Vaccari, T., Dionne, H., Hill, J., Wu, G., and Bilder, D. (2007). A mosaic genetic screen for Drosophila neoplastic tumor suppressor genes based on defective pupation. Genetics 3, 1667-1677. 54. Morrison, H.A., Dionne, H., Rusten, T.E., Brech, A., Fisher, W.W., Pfeiffer, B.D., Celniker, S.E., Stenmark, H., and Bilder, D. (2008). Regulation of early endosomal entry by the Drosophila tumor suppressors Rabenosyn and Vps45. Mol. Biol. Cell 10, 4167-4176. 55. Vaccari, T., and Bilder, D. (2005). The Drosophila tumor suppressor vps25 prevents nonautonomous overproliferation by regulating notch trafficking. Dev. Cell. 5, 687-698. 56. Leong, G.R., Goulding, K.R., Amin, N., Richardson, H.E., and Brumby, A.M. (2009). Scribble mutants promote aPKC and JNK-dependent epithelial neoplasia independently of Crumbs. BMC Biol. 62.

Page 136: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

126

57. Gilbert, M.M., Robinson, B.S., and Moberg, K.H. (2009). Functional interactions between the erupted/tsg101 growth suppressor gene and the DaPKC and rbf1 genes in Drosophila imaginal disc tumors. PLoS One 9, e7039. 58. Georgiou, M., Marinari, E., Burden, J., and Baum, B. (2008). Cdc42, Par6, and aPKC regulate Arp2/3-mediated endocytosis to control local adherens junction stability. Curr. Biol. 21, 1631-1638. 59. Leibfried, A., Fricke, R., Morgan, M.J., Bogdan, S., and Bellaiche, Y. (2008). Drosophila Cip4 and WASp define a branch of the Cdc42-Par6-aPKC pathway regulating E-cadherin endocytosis. Curr. Biol. 21, 1639-1648. 60. Harris, K.P., and Tepass, U. (2008). Cdc42 and Par proteins stabilize dynamic adherens junctions in the Drosophila neuroectoderm through regulation of apical endocytosis. J. Cell Biol. 6, 1129-1143. 61. Balklava, Z., Pant, S., Fares, H., and Grant, B.D. (2007). Genome-wide analysis identifies a general requirement for polarity proteins in endocytic traffic. Nat. Cell Biol. 9, 1066-1073. 62. Fischer, J.A., Eun, S.H., and Doolan, B.T. (2006). Endocytosis, endosome trafficking, and the regulation of Drosophila development. Annu. Rev. Cell Dev. Biol. 181-206. 63. Nichols, J.T., Miyamoto, A., and Weinmaster, G. (2007). Notch signaling--constantly on the move. Traffic 8, 959-969. 64. Benmerah, A., and Lamaze, C. (2007). Clathrin-coated pits: vive la difference? Traffic 8, 970-982. 65. Yan, Y., Denef, N., and Schupbach, T. (2009). The vacuolar proton pump, V-ATPase, is required for notch signaling and endosomal trafficking in Drosophila. Dev. Cell. 3, 387-402. 66. Tien, A.C., Rajan, A., and Bellen, H.J. (2009). A Notch updated. J. Cell Biol. 5, 621-629. 67. Fortini, M.E., and Bilder, D. (2009). Endocytic regulation of Notch signaling. Curr Opin Genet Dev 68. Seugnet, L., Simpson, P., and Haenlin, M. (1997). Requirement for dynamin during Notch signaling in Drosophila neurogenesis. Dev. Biol. 2, 585-598. 69. Parks, A.L., Klueg, K.M., Stout, J.R., and Muskavitch, M.A. (2000). Ligand endocytosis drives receptor dissociation and activation in the Notch pathway. Development 7, 1373-1385. 70. Overstreet, E., Fitch, E., and Fischer, J.A. (2004). Fat facets and Liquid facets promote Delta endocytosis and Delta signaling in the signaling cells. Development 21, 5355-5366. 71. Wang, W., and Struhl, G. (2004). Drosophila Epsin mediates a select endocytic pathway that DSL ligands must enter to activate Notch. Development 21, 5367-5380. 72. Grant, B.D., and Donaldson, J.G. (2009). Pathways and mechanisms of endocytic recycling. Nat. Rev. Mol. Cell Biol. 9, 597-608. 73. Compagnon, J., Gervais, L., Roman, M.S., Chamot-Boeuf, S., and Guichet, A. (2009). Interplay between Rab5 and PtdIns(4,5)P2 controls early endocytosis in the Drosophila germline. J. Cell. Sci. Pt 1, 25-35. 74. Satoh, A.K., O'Tousa, J.E., Ozaki, K., and Ready, D.F. (2005). Rab11 mediates post-Golgi trafficking of rhodopsin to the photosensitive apical membrane of Drosophila photoreceptors. Development 7, 1487-1497. 75. Emery, G., Hutterer, A., Berdnik, D., Mayer, B., Wirtz-Peitz, F., Gaitan, M.G., and Knoblich, J.A. (2005). Asymmetric Rab 11 endosomes regulate delta recycling and specify cell fate in the Drosophila nervous system. Cell 5, 763-773.

Page 137: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

127

76. Jafar-Nejad, H., Andrews, H.K., Acar, M., Bayat, V., Wirtz-Peitz, F., Mehta, S.Q., Knoblich, J.A., and Bellen, H.J. (2005). Sec15, a component of the exocyst, promotes notch signaling during the asymmetric division of Drosophila sensory organ precursors. Dev. Cell. 3, 351-363. 77. Li, B.X., Satoh, A.K., and Ready, D.F. (2007). Myosin V, Rab11, and dRip11 direct apical secretion and cellular morphogenesis in developing Drosophila photoreceptors. J. Cell Biol. 4, 659-669. 78. Krauss, J., Lopez de Quinto, S., Nusslein-Volhard, C., and Ephrussi, A. (2009). Myosin-V regulates oskar mRNA localization in the Drosophila oocyte. Curr. Biol. 12, 1058-1063. 79. Shaye, D.D., Casanova, J., and Llimargas, M. (2008). Modulation of intracellular trafficking regulates cell intercalation in the Drosophila trachea. Nat. Cell Biol. 8, 964-970. 80. Sommer, B., Oprins, A., Rabouille, C., and Munro, S. (2005). The exocyst component Sec5 is present on endocytic vesicles in the oocyte of Drosophila melanogaster. J. Cell Biol. 6, 953-963. 81. Classen, A.K., Anderson, K.I., Marois, E., and Eaton, S. (2005). Hexagonal packing of Drosophila wing epithelial cells by the planar cell polarity pathway. Dev. Cell. 6, 805-817. 82. Sandvig, K., Torgersen, M.L., Raa, H.A., and van Deurs, B. (2008). Clathrin-independent endocytosis: from nonexisting to an extreme degree of complexity. Histochem. Cell Biol. 3, 267-276. 83. Motley, A., Bright, N.A., Seaman, M.N., and Robinson, M.S. (2003). Clathrin-mediated endocytosis in AP-2-depleted cells. J. Cell Biol. 5, 909-918. 84. Hinrichsen, L., Harborth, J., Andrees, L., Weber, K., and Ungewickell, E.J. (2003). Effect of clathrin heavy chain- and alpha-adaptin-specific small inhibitory RNAs on endocytic accessory proteins and receptor trafficking in HeLa cells. J. Biol. Chem. 46, 45160-45170. 85. Lakadamyali, M., Rust, M.J., and Zhuang, X. (2006). Ligands for clathrin-mediated endocytosis are differentially sorted into distinct populations of early endosomes. Cell 5, 997-1009. 86. Kessell, I., Holst, B.D., and Roth, T.F. (1989). Membranous intermediates in endocytosis are labile, as shown in a temperature-sensitive mutant. Proc. Natl. Acad. Sci. U. S. A. 13, 4968-4972. 87. Coumailleau, F., Furthauer, M., Knoblich, J.A., and Gonzalez-Gaitan, M. (2009). Directional Delta and Notch trafficking in Sara endosomes during asymmetric cell division. Nature 7241, 1051-1055. 88. Rajan, A., Tien, A.C., Haueter, C.M., Schulze, K.L., and Bellen, H.J. (2009). The Arp2/3 complex and WASp are required for apical trafficking of Delta into microvilli during cell fate specification of sensory organ precursors. Nat. Cell Biol. 7, 815-824. 89. Traub, L.M. (2003). Sorting it out: AP-2 and alternate clathrin adaptors in endocytic cargo selection. J. Cell Biol. 2, 203-208. 90. Bellen, H.J., Levis, R.W., Liao, G., He, Y., Carlson, J.W., Tsang, G., Evans-Holm, M., Hiesinger, P.R., Schulze, K.L., Rubin, G.M. et al. (2004). The BDGP gene disruption project: single transposon insertions associated with 40% of Drosophila genes. Genetics 2, 761-781. 91. Watts, R.J., Hoopfer, E.D., and Luo, L. (2003). Axon pruning during Drosophila metamorphosis: evidence for local degeneration and requirement of the ubiquitin-proteasome system. Neuron 6, 871-885. 92. Wucherpfennig, T., Wilsch-Brauninger, M., and Gonzalez-Gaitan, M. (2003). Role of Drosophila Rab5 during endosomal trafficking at the synapse and evoked neurotransmitter release. J. Cell Biol. 3, 609-624.

Page 138: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

128

93. Overstreet, E., Chen, X., Wendland, B., and Fischer, J.A. (2003). Either part of a Drosophila epsin protein, divided after the ENTH domain, functions in endocytosis of delta in the developing eye. Curr. Biol. 10, 854-860. 94. Baker, N.E., and Yu, S.Y. (1997). Proneural function of neurogenic genes in the developing Drosophila eye. Curr. Biol. 2, 122-132. 95. Murthy, M., and Schwarz, T.L. (2004). The exocyst component Sec5 is required for membrane traffic and polarity in the Drosophila ovary. Development 2, 377-388. 96. Zhang, J., Schulze, K.L., Hiesinger, P.R., Suyama, K., Wang, S., Fish, M., Acar, M., Hoskins, R.A., Bellen, H.J., and Scott, M.P. (2007). Thirty-one flavors of Drosophila rab proteins. Genetics 2, 1307-1322. 97. Pellock, B.J., Buff, E., White, K., and Hariharan, I.K. (2007). The Drosophila tumor suppressors Expanded and Merlin differentially regulate cell cycle exit, apoptosis, and Wingless signaling. Dev. Biol. 1, 102-115. 98. Bilder, D., and Perrimon, N. (2000). Localization of apical epithelial determinants by the basolateral PDZ protein Scribble. Nature 6770, 676-680. 99. Lloyd, T.E., Atkinson, R., Wu, M.N., Zhou, Y., Pennetta, G., and Bellen, H.J. (2002). Hrs regulates endosome membrane invagination and tyrosine kinase receptor signaling in Drosophila. Cell 2, 261-269. 100. Goldstein, B., and Macara, I.G. (2007). The PAR proteins: fundamental players in animal cell polarization. Dev. Cell. 5, 609-622. 101. Bulgakova, N.A., and Knust, E. (2009). The Crumbs complex: from epithelial-cell polarity to retinal degeneration. J. Cell. Sci. Pt 15, 2587-2596. 102. Tanentzapf, G., and Tepass, U. (2003). Interactions between the crumbs, lethal giant larvae and bazooka pathways in epithelial polarization. Nat. Cell Biol. 1, 46-52. 103. Bilder, D., Schober, M., and Perrimon, N. (2003). Integrated activity of PDZ protein complexes regulates epithelial polarity. Nat. Cell Biol. 1, 53-58. 104. Yamanaka, T., and Ohno, S. (2008). Role of Lgl/Dlg/Scribble in the regulation of epithelial junction, polarity and growth. Front. Biosci. 6693-6707. 105. Vasioukhin, V. (2006). Lethal giant puzzle of Lgl. Dev. Neurosci. 1-2, 13-24. 106. Humbert, P.O., Grzeschik, N.A., Brumby, A.M., Galea, R., Elsum, I., and Richardson, H.E. (2008). Control of tumourigenesis by the Scribble/Dlg/Lgl polarity module. Oncogene 55, 6888-6907. 107. Moberg, K.H., Schelble, S., Burdick, S.K., and Hariharan, I.K. (2005). Mutations in erupted, the Drosophila ortholog of mammalian tumor susceptibility gene 101, elicit non-cell-autonomous overgrowth. Dev. Cell. 5, 699-710. 108. Thompson, B.J., Mathieu, J., Sung, H.H., Loeser, E., Rorth, P., and Cohen, S.M. (2005). Tumor suppressor properties of the ESCRT-II complex component Vps25 in Drosophila. Dev. Cell. 5, 711-720. 109. Herz, H.M., Chen, Z., Scherr, H., Lackey, M., Bolduc, C., and Bergmann, A. (2006). Vps25 Mosaics Display Non-Autonomous Cell Survival and Overgrowth, and Autonomous Apoptosis. Development 10, 1871-1880. 110. Shivas, J.M., Morrison, H.A., Bilder, D., and Skop, A.R. (2010). Polarity and endocytosis: reciprocal regulation. Trends Cell Biol. 8, 445-452. 111. Windler, S.L., and Bilder, D. (2010). Endocytic internalization routes required for delta/notch signaling. Curr. Biol. 6, 538-543.

Page 139: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

129

112. Wei, J., Hortsch, M., and Goode, S. (2004). Neuroglian stabilizes epithelial structure during Drosophila oogenesis. Dev. Dyn. 4, 800-808. 113. Rusten, T.E., Vaccari, T., Lindmo, K., Rodahl, L.M., Nezis, I.P., Sem-Jacobsen, C., Wendler, F., Vincent, J.P., Brech, A., Bilder, D. et al. (2007). ESCRTs and Fab1 regulate distinct steps of autophagy. Curr. Biol. 20, 1817-1825. 114. Eder, A.M., Sui, X., Rosen, D.G., Nolden, L.K., Cheng, K.W., Lahad, J.P., Kango-Singh, M., Lu, K.H., Warneke, C.L., Atkinson, E.N. et al. (2005). Atypical PKCiota contributes to poor prognosis through loss of apical-basal polarity and cyclin E overexpression in ovarian cancer. Proc. Natl. Acad. Sci. U. S. A. 35, 12519-12524. 115. Blankenship, J.T., Fuller, M.T., and Zallen, J.A. (2007). The Drosophila homolog of the Exo84 exocyst subunit promotes apical epithelial identity. J. Cell. Sci. Pt 17, 3099-3110. 116. Wodarz, A., Hinz, U., Engelbert, M., and Knust, E. (1995). Expression of crumbs confers apical character on plasma membrane domains of ectodermal epithelia of Drosophila. Cell 1, 67-76. 117. Deborde, S., Perret, E., Gravotta, D., Deora, A., Salvarezza, S., Schreiner, R., and Rodriguez-Boulan, E. (2008). Clathrin is a key regulator of basolateral polarity. Nature 7188, 719-723. 118. Guichard, A., McGillivray, S.M., Cruz-Moreno, B., van Sorge, N.M., Nizet, V., and Bier, E. (2010). Anthrax toxins cooperatively inhibit endocytic recycling by the Rab11/Sec15 exocyst. Nature 7317, 854-858. 119. Rodriguez-Boulan, E., Kreitzer, G., and Musch, A. (2005). Organization of vesicular trafficking in epithelia. Nat. Rev. Mol. Cell Biol. 3, 233-247. 120. Rolls, M.M., Albertson, R., Shih, H.P., Lee, C.Y., and Doe, C.Q. (2003). Drosophila aPKC regulates cell polarity and cell proliferation in neuroblasts and epithelia. J. Cell Biol. 5, 1089-1098. 121. Rusten, T.E., Rodahl, L.M., Pattni, K., Englund, C., Samakovlis, C., Dove, S., Brech, A., and Stenmark, H. (2006). Fab1 phosphatidylinositol 3-phosphate 5-kinase controls trafficking but not silencing of endocytosed receptors. Mol. Biol. Cell 9, 3989-4001. 122. Sotillos, S., Diaz-Meco, M.T., Caminero, E., Moscat, J., and Campuzano, S. (2004). DaPKC-dependent phosphorylation of Crumbs is required for epithelial cell polarity in Drosophila. J. Cell Biol. 4, 549-557. 123. Mendoza-Topaz, C., Urra, F., Barria, R., Albornoz, V., Ugalde, D., Thomas, U., Gundelfinger, E.D., Delgado, R., Kukuljan, M., Sanxaridis, P.D. et al. (2008). DLGS97/SAP97 is developmentally upregulated and is required for complex adult behaviors and synapse morphology and function. J. Neurosci. 1, 304-314. 124. Strigini, M., and Cohen, S.M. (2000). Wingless gradient formation in the Drosophila wing. Curr. Biol. 6, 293-300. 125. Tepass, U., Theres, C., and Knust, E. (1990). crumbs encodes an EGF-like protein expressed on apical membranes of Drosophila epithelial cells and required for organization of epithelia. Cell 5, 787-799. 126. Tweedie, S., Ashburner, M., Falls, K., Leyland, P., McQuilton, P., Marygold, S., Millburn, G., Osumi-Sutherland, D., Schroeder, A., Seal, R. et al. (2009). FlyBase: enhancing Drosophila Gene Ontology annotations. Nucleic Acids Res. Database issue, D555-9. 127. Bilder, D., Li, M., and Perrimon, N. (2000). Cooperative regulation of cell polarity and growth by Drosophila tumor suppressors. Science 5476, 113-116.

Page 140: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

130

128. Classen, A.K., Bunker, B.D., Harvey, K.F., Vaccari, T., and Bilder, D. (2009). A tumor suppressor activity of Drosophila Polycomb genes mediated by JAK-STAT signaling. Nat. Genet. 10, 1150-1155. 129. Horne-Badovinac, S., and Bilder, D. (2008). Dynein regulates epithelial polarity and the apical localization of stardust A mRNA. PLoS Genet. 1, e8. 130. Mukhopadhyay, D., and Riezman, H. (2007). Proteasome-independent functions of ubiquitin in endocytosis and signaling. Science 5809, 201-205. 131. Nandi, D., Tahiliani, P., Kumar, A., and Chandu, D. (2006). The ubiquitin-proteasome system. J. Biosci. 1, 137-155. 132. Hunter, T. (2007). The age of crosstalk: phosphorylation, ubiquitination, and beyond. Mol. Cell 5, 730-738. 133. Ardley, H.C., and Robinson, P.A. (2005). E3 ubiquitin ligases. Essays Biochem. 15-30. 134. Jiang, J., and Struhl, G. (1998). Regulation of the Hedgehog and Wingless signalling pathways by the F-box/WD40-repeat protein Slimb. Nature 6666, 493-496. 135. Smelkinson, M.G., and Kalderon, D. (2006). Processing of the Drosophila hedgehog signaling effector Ci-155 to the repressor Ci-75 is mediated by direct binding to the SCF component Slimb. Curr. Biol. 1, 110-116. 136. Grima, B., Lamouroux, A., Chelot, E., Papin, C., Limbourg-Bouchon, B., and Rouyer, F. (2002). The F-box protein slimb controls the levels of clock proteins period and timeless. Nature 6912, 178-182. 137. Ko, H.W., Jiang, J., and Edery, I. (2002). Role for Slimb in the degradation of Drosophila Period protein phosphorylated by Doubletime. Nature 6916, 673-678. 138. Cunha-Ferreira, I., Rodrigues-Martins, A., Bento, I., Riparbelli, M., Zhang, W., Laue, E., Callaini, G., Glover, D.M., and Bettencourt-Dias, M. (2009). The SCF/Slimb ubiquitin ligase limits centrosome amplification through degradation of SAK/PLK4. Curr. Biol. 1, 43-49. 139. Frescas, D., and Pagano, M. (2008). Deregulated proteolysis by the F-box proteins SKP2 and beta-TrCP: tipping the scales of cancer. Nat. Rev. Cancer. 6, 438-449. 140. Miletich, I., and Limbourg-Bouchon, B. (2000). Drosophila null slimb clones transiently deregulate Hedgehog-independent transcription of wingless in all limb discs, and induce decapentaplegic transcription linked to imaginal disc regeneration. Mech. Dev. 1-2, 15-26. 141. Brumby, A.M., and Richardson, H.E. (2003). scribble mutants cooperate with oncogenic Ras or Notch to cause neoplastic overgrowth in Drosophila. EMBO J. 21, 5769-5779. 142. Pastor-Pareja, J.C., Wu, M., and Xu, T. (2008). An innate immune response of blood cells to tumors and tissue damage in Drosophila. Dis. Model. Mech. 2-3, 144-54; discussion 153. 143. Wu, M., Pastor-Pareja, J.C., and Xu, T. (2010). Interaction between Ras(V12) and scribbled clones induces tumour growth and invasion. Nature 7280, 545-548. 144. Igaki, T., Pagliarini, R.A., and Xu, T. (2006). Loss of cell polarity drives tumor growth and invasion through JNK activation in Drosophila. Curr. Biol. 11, 1139-1146. 145. Uhlirova, M., Jasper, H., and Bohmann, D. (2005). Non-cell-autonomous induction of tissue overgrowth by JNK/Ras cooperation in a Drosophila tumor model. Proc. Natl. Acad. Sci. U. S. A. 37, 13123-13128. 146. St Johnston, D. (2002). The art and design of genetic screens: Drosophila melanogaster. Nat. Rev. Genet. 3, 176-188. 147. Balastik, M., Ferraguti, F., Pires-da Silva, A., Lee, T.H., Alvarez-Bolado, G., Lu, K.P., and Gruss, P. (2008). Deficiency in ubiquitin ligase TRIM2 causes accumulation of neurofilament light chain and neurodegeneration. Proc. Natl. Acad. Sci. U. S. A. 33, 12016-12021.

Page 141: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

131

148. Khazaei, M.R., Bunk, E.C., Hillje, A.L., Jahn, H.M., Riegler, E.M., Knoblich, J.A., Young, P., and Schwamborn, J.C. (2010). The E3-ubiquitin ligase TRIM2 regulates neuronal polarization. J. Neurochem. 149. Schwamborn, J.C., Khazaei, M.R., and Puschel, A.W. (2007). The interaction of mPar3 with the ubiquitin ligase Smurf2 is required for the establishment of neuronal polarity. J. Biol. Chem. 48, 35259-35268. 150. Schwamborn, J.C., Muller, M., Becker, A.H., and Puschel, A.W. (2007). Ubiquitination of the GTPase Rap1B by the ubiquitin ligase Smurf2 is required for the establishment of neuronal polarity. EMBO J. 5, 1410-1422. 151. Wang, H.R., Zhang, Y., Ozdamar, B., Ogunjimi, A.A., Alexandrova, E., Thomsen, G.H., and Wrana, J.L. (2003). Regulation of cell polarity and protrusion formation by targeting RhoA for degradation. Science 5651, 1775-1779. 152. Narimatsu, M., Bose, R., Pye, M., Zhang, L., Miller, B., Ching, P., Sakuma, R., Luga, V., Roncari, L., Attisano, L. et al. (2009). Regulation of planar cell polarity by Smurf ubiquitin ligases. Cell 2, 295-307. 153. Ganner, A., Lienkamp, S., Schafer, T., Romaker, D., Wegierski, T., Park, T.J., Spreitzer, S., Simons, M., Gloy, J., Kim, E. et al. (2009). Regulation of ciliary polarity by the APC/C. Proc. Natl. Acad. Sci. U. S. A. 42, 17799-17804. 154. Pacquelet, A., Zanin, E., Ashiono, C., and Gotta, M. (2008). PAR-6 levels are regulated by NOS-3 in a CUL-2 dependent manner in Caenorhabditiselegans. Dev. Biol. 2, 267-272. 155. Moberg, K.H., Mukherjee, A., Veraksa, A., Artavanis-Tsakonas, S., and Hariharan, I.K. (2004). The Drosophila F box protein archipelago regulates dMyc protein levels in vivo. Curr. Biol. 11, 965-974. 156. Duchi, S., Fagnocchi, L., Cavaliere, V., Hsouna, A., Gargiulo, G., and Hsu, T. (2010). Drosophila VHL tumor-suppressor gene regulates epithelial morphogenesis by promoting microtubule and aPKC stability. Development 9, 1493-1503. 157. Igaki, T., Pastor-Pareja, J.C., Aonuma, H., Miura, M., and Xu, T. (2009). Intrinsic tumor suppression and epithelial maintenance by endocytic activation of Eiger/TNF signaling in Drosophila. Dev. Cell. 3, 458-465. 158. Mantovani, F., and Banks, L. (2003). Regulation of the discs large tumor suppressor by a phosphorylation-dependent interaction with the beta-TrCP ubiquitin ligase receptor. J. Biol. Chem. 43, 42477-42486. 159. Yen, H.C., and Elledge, S.J. (2008). Identification of SCF ubiquitin ligase substrates by global protein stability profiling. Science 5903, 923-929. 160. Kim, N.G., Xu, C., and Gumbiner, B.M. (2009). Identification of targets of the Wnt pathway destruction complex in addition to beta-catenin. Proc. Natl. Acad. Sci. U. S. A. 13, 5165-5170. 161. Xu, C., Kim, N.G., and Gumbiner, B.M. (2009). Regulation of protein stability by GSK3 mediated phosphorylation. Cell. Cycle 24, 4032-4039. 162. Morrison, H.A. (2010). Epithelial cell polarity and proliferation control in Drosophila melanogaster.183. 163. Popov, N., Schulein, C., Jaenicke, L.A., and Eilers, M. (2010). Ubiquitylation of the amino terminus of Myc by SCFbeta-TrCP antagonizes SCFFbw7-mediated turnover. Nat. Cell Biol. 10, 973-981.

Page 142: Regulation of cellular signaling pathways by endocytosis and … · 2018-10-10 · 1 Abstract Regulation of cellular signaling pathways by endocytosis and protein degradation by Sarah

132

164. Bach, E.A., Ekas, L.A., Ayala-Camargo, A., Flaherty, M.S., Lee, H., Perrimon, N., and Baeg, G.H. (2007). GFP reporters detect the activation of the Drosophila JAK/STAT pathway in vivo. Gene Expr. Patterns 3, 323-331. 165. Murphy, T.D. (2003). Drosophila skpA, a component of SCF ubiquitin ligases, regulates centrosome duplication independently of cyclin E accumulation. J. Cell. Sci. Pt 11, 2321-2332. 166. Noureddine, M.A., Donaldson, T.D., Thacker, S.A., and Duronio, R.J. (2002). Drosophila Roc1a encodes a RING-H2 protein with a unique function in processing the Hh signal transducer Ci by the SCF E3 ubiquitin ligase. Dev. Cell. 6, 757-770. 167. Ou, C.Y., Lin, Y.F., Chen, Y.J., and Chien, C.T. (2002). Distinct protein degradation mechanisms mediated by Cul1 and Cul3 controlling Ci stability in Drosophila eye development. Genes Dev. 18, 2403-2414. 168. Chen, Y., Cardinaux, J.R., Goodman, R.H., and Smolik, S.M. (1999). Mutants of cubitus interruptus that are independent of PKA regulation are independent of hedgehog signaling. Development 16, 3607-3616. 169. Pai, L.M., Orsulic, S., Bejsovec, A., and Peifer, M. (1997). Negative regulation of Armadillo, a Wingless effector in Drosophila. Development 11, 2255-2266. 170. Mechler, B.M., McGinnis, W., and Gehring, W.J. (1985). Molecular cloning of lethal(2)giant larvae, a recessive oncogene of Drosophila melanogaster. EMBO J. 6, 1551-1557.