Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
EMBO J ; 42(6): e112863, 2023 03 15.
Article in English | MEDLINE | ID: mdl-36807601

ABSTRACT

The Hippo pathway was originally discovered to control tissue growth in Drosophila and includes the Hippo kinase (Hpo; MST1/2 in mammals), scaffold protein Salvador (Sav; SAV1 in mammals) and the Warts kinase (Wts; LATS1/2 in mammals). The Hpo kinase is activated by binding to Crumbs-Expanded (Crb-Ex) and/or Merlin-Kibra (Mer-Kib) proteins at the apical domain of epithelial cells. Here we show that activation of Hpo also involves the formation of supramolecular complexes with properties of a biomolecular condensate, including concentration dependence and sensitivity to starvation, macromolecular crowding, or 1,6-hexanediol treatment. Overexpressing Ex or Kib induces formation of micron-scale Hpo condensates in the cytoplasm, rather than at the apical membrane. Several Hippo pathway components contain unstructured low-complexity domains and purified Hpo-Sav complexes undergo phase separation in vitro. Formation of Hpo condensates is conserved in human cells. We propose that apical Hpo kinase activation occurs in phase separated "signalosomes" induced by clustering of upstream pathway components.


Subject(s)
Drosophila Proteins , Hippo Signaling Pathway , Animals , Humans , Signal Transduction/physiology , Tumor Suppressor Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila/metabolism , Neurofibromin 2/metabolism , Drosophila melanogaster/metabolism , Mammals , Protein Serine-Threonine Kinases/metabolism , Intracellular Signaling Peptides and Proteins/metabolism
2.
EMBO J ; 40(13): e105770, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33950519

ABSTRACT

Wnt signalling induces a gradient of stem/progenitor cell proliferation along the crypt-villus axis of the intestine, which becomes expanded during intestinal regeneration or tumour formation. The YAP transcriptional co-activator is known to be required for intestinal regeneration, but its mode of regulation remains controversial. Here we show that the YAP-TEAD transcription factor is a key downstream effector of Wnt signalling in the intestine. Loss of YAP activity by Yap/Taz conditional knockout results in sensitivity of crypt stem cells to apoptosis and reduced cell proliferation during regeneration. Gain of YAP activity by Lats1/2 conditional knockout is sufficient to drive a crypt hyperproliferation response. In particular, Wnt signalling acts transcriptionally to induce YAP and TEAD1/2/4 expression. YAP normally localises to the nucleus only in crypt base stem cells, but becomes nuclear in most intestinal epithelial cells during intestinal regeneration after irradiation, or during organoid growth, in a Src family kinase-dependent manner. YAP-driven crypt expansion during regeneration involves an elongation and flattening of the Wnt signalling gradient. Thus, Wnt and Src-YAP signals cooperate to drive intestinal regeneration.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Intestines/physiology , Regeneration/genetics , Regeneration/physiology , Transcription Factors/genetics , Wnt Signaling Pathway/genetics , src-Family Kinases/genetics , Animals , Apoptosis/genetics , Cell Cycle Proteins/genetics , Cell Proliferation/genetics , Epithelial Cells/physiology , Intestinal Mucosa/physiology , Mice , Mice, Inbred C57BL , Stem Cells/physiology , YAP-Signaling Proteins
3.
Nat Rev Mol Cell Biol ; 14(5): 307-14, 2013 05.
Article in English | MEDLINE | ID: mdl-23609509

ABSTRACT

Cell polarity and cell-cell junctions have pivotal roles in organizing cells into tissues and in mediating cell-cell communication. The transmembrane protein Crumbs has a well-established role in the maintenance of epithelial polarity, and it can also regulate signalling via the Notch and Hippo pathways to influence tissue growth. The functions of Crumbs in epithelial polarity and Hippo-mediated growth depend on its short intracellular domain. Recent evidence now points to a conserved and fundamental role for the extracellular domain of Crumbs in mediating homophilic Crumbs-Crumbs interactions at cell-cell junctions.


Subject(s)
Cell Polarity/physiology , Intercellular Junctions/physiology , Membrane Proteins/physiology , Nerve Tissue Proteins/physiology , Animals , Cell Communication/physiology , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelial Cells/physiology , Humans , Intercellular Junctions/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism
4.
Development ; 147(5)2020 03 02.
Article in English | MEDLINE | ID: mdl-32122911

ABSTRACT

Mutations in the Ultrabithorax (Ubx) gene cause homeotic transformation of the normally two-winged Drosophila into a four-winged mutant fly. Ubx encodes a HOX family transcription factor that specifies segment identity, including transformation of the second set of wings into rudimentary halteres. Ubx is known to control the expression of many genes that regulate tissue growth and patterning, but how it regulates tissue morphogenesis to reshape the wing into a haltere is still unclear. Here, we show that Ubx acts by repressing the expression of two genes in the haltere, Stubble and Notopleural, both of which encode transmembrane proteases that remodel the apical extracellular matrix to promote wing morphogenesis. In addition, Ubx induces expression of the Tissue inhibitor of metalloproteases in the haltere, which prevents the basal extracellular matrix remodelling necessary for wing morphogenesis. Our results provide a long-awaited explanation for how Ubx controls morphogenetic transformation.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/embryology , Homeodomain Proteins/genetics , Morphogenesis/genetics , Transcription Factors/genetics , Wings, Animal/embryology , Animals , CRISPR-Cas Systems , Drosophila melanogaster/genetics , Matrix Metalloproteinase Inhibitors/metabolism , Membrane Proteins/genetics , Serine Endopeptidases/genetics
5.
PLoS Biol ; 17(10): e3000509, 2019 10.
Article in English | MEDLINE | ID: mdl-31613895

ABSTRACT

The Hippo signalling pathway restricts cell proliferation in animal tissues by inhibiting Yes-associated protein (YAP or YAP1) and Transcriptional Activator with a PDZ domain (TAZ or WW-domain-containing transcriptional activator [WWTR1]), coactivators of the Scalloped (Sd or TEAD) DNA-binding transcription factor. Drosophila has a single YAP/TAZ homolog named Yorkie (Yki) that is regulated by Hippo pathway signalling in response to epithelial polarity and tissue mechanics during development. Here, we show that Yki translocates to the nucleus to drive Sd-mediated cell proliferation in the ovarian follicle cell epithelium in response to mechanical stretching caused by the growth of the germline. Importantly, mechanically induced Yki nuclear localisation also requires nutritionally induced insulin/insulin-like growth factor 1 (IGF-1) signalling (IIS) via phosphatidyl inositol-3-kinase (PI3K), phosphoinositide-dependent kinase 1 (PDK1 or PDPK1), and protein kinase B (Akt or PKB) in the follicular epithelium. We find similar results in the developing Drosophila wing, where Yki becomes nuclear in the mechanically stretched cells of the wing pouch during larval feeding, which induces IIS, but translocates to the cytoplasm upon cessation of feeding in the third instar stage. Inactivating Akt prevents nuclear Yki localisation in the wing disc, while ectopic activation of the insulin receptor, PI3K, or Akt/PKB is sufficient to maintain nuclear Yki in mechanically stimulated cells of the wing pouch even after feeding ceases. Finally, IIS also promotes YAP nuclear localisation in response to mechanical cues in mammalian skin epithelia. Thus, the Hippo pathway has a physiological function as an integrator of epithelial cell polarity, tissue mechanics, and nutritional cues to control cell proliferation and tissue growth in both Drosophila and mammals.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Epithelial Cells/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Nuclear Proteins/genetics , Phosphatidylinositol 3-Kinase/genetics , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Trans-Activators/genetics , 3-Phosphoinositide-Dependent Protein Kinases/genetics , 3-Phosphoinositide-Dependent Protein Kinases/metabolism , Animals , Biomechanical Phenomena , Cell Nucleus/metabolism , Cell Nucleus/ultrastructure , Cell Polarity , Cell Proliferation , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Epithelial Cells/cytology , Female , Gene Expression Regulation, Developmental , Humans , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Larva/cytology , Larva/genetics , Larva/growth & development , Larva/metabolism , Mechanotransduction, Cellular , Mice , Nuclear Proteins/metabolism , Ovarian Follicle/cytology , Ovarian Follicle/growth & development , Ovarian Follicle/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein Transport , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Trans-Activators/metabolism , Wings, Animal/cytology , Wings, Animal/growth & development , Wings, Animal/metabolism , YAP-Signaling Proteins
6.
EMBO Rep ; 21(4): e49700, 2020 04 03.
Article in English | MEDLINE | ID: mdl-32030856

ABSTRACT

Epithelial cells undergo cortical rounding at the onset of mitosis to enable spindle orientation in the plane of the epithelium. In cuboidal epithelia in culture, the adherens junction protein E-cadherin recruits Pins/LGN/GPSM2 and Mud/NuMA to orient the mitotic spindle. In the pseudostratified columnar epithelial cells of Drosophila, septate junctions recruit Mud/NuMA to orient the spindle, while Pins/LGN/GPSM2 is surprisingly dispensable. We show that these pseudostratified epithelial cells downregulate E-cadherin as they round up for mitosis. Preventing cortical rounding by inhibiting Rho-kinase-mediated actomyosin contractility blocks downregulation of E-cadherin during mitosis. Mitotic activation of Rho-kinase depends on the RhoGEF ECT2/Pebble and its binding partners RacGAP1/MgcRacGAP/CYK4/Tum and MKLP1/KIF23/ZEN4/Pav. Cell cycle control of these Rho activators is mediated by the Aurora A and B kinases, which act redundantly during mitotic rounding. Thus, in Drosophila pseudostratified epithelia, disruption of adherens junctions during mitosis necessitates planar spindle orientation by septate junctions to maintain epithelial integrity.


Subject(s)
Adherens Junctions , Spindle Apparatus , Animals , Drosophila/genetics , Epithelial Cells , Mitosis
8.
Bioessays ; 42(5): e1900162, 2020 05.
Article in English | MEDLINE | ID: mdl-32128850

ABSTRACT

The transcriptional co-activators YAP (or YAP1) and TAZ (or WWTR1) are frequently activated during the growth and progression of many solid tumors, including lung, colorectal, breast, pancreatic, and liver carcinomas as well as melanoma and glioma. YAP/TAZ bind to TEAD-family co-activators to drive cancer cell survival, proliferation, invasive migration, and metastasis. YAP/TAZ activation may also confer resistance to chemotherapy, radiotherapy, or immunotherapy. YAP-TEAD cooperates with the RAS-induced AP-1 (FOS/JUN) transcription factor to drive tumor growth and cooperates with MRTF-SRF to promote activation of cancer-associated fibroblasts, matrix stiffening, and metastasis. The key upstream repressor of YAP/TAZ activation is the Hippo (MST1/2-LATS1/2) pathway and the key upstream activators are mechanically induced Integrin-SRC and E-cadherin-AJUBA/TRIP6/LIMD1, growth factor induced PI3K-AKT, and inflammation-induced G-protein coupled receptor (GPCR) signals, all of which antagonize the Hippo pathway. In this review, strategies to target YAP/TAZ activity in cancer are discussed along with the prospects for synergy with established pillars of cancer therapy.


Subject(s)
Melanoma , Phosphatidylinositol 3-Kinases , Adaptor Proteins, Signal Transducing/genetics , Humans , Intracellular Signaling Peptides and Proteins , LIM Domain Proteins , Transcription Factors/metabolism
9.
J Cell Sci ; 132(8)2019 04 25.
Article in English | MEDLINE | ID: mdl-30872454

ABSTRACT

The elaboration of polarity is central to organismal development and to the maintenance of functional epithelia. Among the controls determining polarity are the PAR proteins, PAR6, aPKCι and PAR3, regulating both known and unknown effectors. Here, we identify FARP2 as a 'RIPR' motif-dependent partner and substrate of aPKCι that is required for efficient polarisation and junction formation. Binding is conferred by a FERM/FA domain-kinase domain interaction and detachment promoted by aPKCι-dependent phosphorylation. FARP2 is shown to promote GTP loading of Cdc42, which is consistent with it being involved in upstream regulation of the polarising PAR6-aPKCι complex. However, we show that aPKCι acts to promote the localised activity of FARP2 through phosphorylation. We conclude that this aPKCι-FARP2 complex formation acts as a positive feedback control to drive polarisation through aPKCι and other Cdc42 effectors.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Epithelial Cells/cytology , Guanine Nucleotide Exchange Factors/metabolism , Protein Kinase C/metabolism , Tight Junctions/metabolism , cdc42 GTP-Binding Protein/metabolism , Caco-2 Cells , Cell Polarity , Guanine Nucleotide Exchange Factors/genetics , HCT116 Cells , Humans , Phosphorylation
10.
Development ; 145(5)2018 03 08.
Article in English | MEDLINE | ID: mdl-29440303

ABSTRACT

Animal cells are thought to sense mechanical forces via the transcriptional co-activators YAP (or YAP1) and TAZ (or WWTR1), the sole Drosophila homolog of which is named Yorkie (Yki). In mammalian cells in culture, artificial mechanical forces induce nuclear translocation of YAP and TAZ. Here, we show that physiological mechanical strain can also drive nuclear localisation of Yki and activation of Yki target genes in the Drosophila follicular epithelium. Mechanical strain activates Yki by stretching the apical domain, reducing the concentration of apical Crumbs, Expanded, Kibra and Merlin, and reducing apical Hippo kinase dimerisation. Overexpressing Hippo kinase to induce ectopic activation in the cytoplasm is sufficient to prevent Yki nuclear localisation even in flattened follicle cells. Conversely, blocking Hippo signalling in warts clones causes Yki nuclear localisation even in columnar follicle cells. We find no evidence for involvement of other pathways, such as Src42A kinase, in regulation of Yki. Finally, our results in follicle cells appear generally applicable to other tissues, as nuclear translocation of Yki is also readily detectable in other flattened epithelial cells such as the peripodial epithelium of the wing imaginal disc, where it promotes cell flattening.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Stress, Mechanical , Wings, Animal/embryology , Animals , Animals, Genetically Modified , Body Patterning/genetics , Cell Nucleus/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Embryo, Nonmammalian , Gene Expression Regulation, Developmental , Imaginal Discs/embryology , Imaginal Discs/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Mechanotransduction, Cellular/physiology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Transport/genetics , Signal Transduction/genetics , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/metabolism , Wings, Animal/metabolism , YAP-Signaling Proteins
11.
J Cell Sci ; 131(22)2018 11 22.
Article in English | MEDLINE | ID: mdl-30404826

ABSTRACT

Human cells can sense mechanical stress acting upon integrin adhesions and respond by sending the YAP (also known as YAP1) and TAZ (also known as WWTR1) transcriptional co-activators to the nucleus to drive TEAD-dependent transcription of target genes. How integrin signaling activates YAP remains unclear. Here, we show that integrin-mediated mechanotransduction requires the Enigma and Enigma-like proteins (PDLIM7 and PDLIM5, respectively; denoted for the family of PDZ and LIM domain-containing proteins). YAP binds to PDLIM5 and PDLIM7 (hereafter PDLIM5/7) via its C-terminal PDZ-binding motif (PBM), which is essential for full nuclear localization and activity of YAP. Accordingly, silencing of PDLIM5/7 expression reduces YAP nuclear localization, tyrosine phosphorylation and transcriptional activity. The PDLIM5/7 proteins are recruited from the cytoplasm to integrin adhesions and F-actin stress fibers in response to force by binding directly to the key stress fiber component α-actinin. Thus, forces acting on integrins recruit Enigma family proteins to trigger YAP activation during mechanotransduction.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cytoskeletal Proteins/metabolism , LIM Domain Proteins/metabolism , Transcription Factors/metabolism , Animals , Caco-2 Cells , Fibroblasts/metabolism , HEK293 Cells , Humans , Integrins/metabolism , Mechanotransduction, Cellular , Mice , Signal Transduction , Trans-Activators , Transcriptional Coactivator with PDZ-Binding Motif Proteins , YAP-Signaling Proteins
12.
EMBO J ; 34(7): 940-54, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25712476

ABSTRACT

The Spectrin cytoskeleton is known to be polarised in epithelial cells, yet its role remains poorly understood. Here, we show that the Spectrin cytoskeleton controls Hippo signalling. In the developing Drosophila wing and eye, loss of apical Spectrins (alpha/beta-heavy dimers) produces tissue overgrowth and mis-regulation of Hippo target genes, similar to loss of Crumbs (Crb) or the FERM-domain protein Expanded (Ex). Apical beta-heavy Spectrin binds to Ex and co-localises with it at the apical membrane to antagonise Yki activity. Interestingly, in both the ovarian follicular epithelium and intestinal epithelium of Drosophila, apical Spectrins and Crb are dispensable for repression of Yki, while basolateral Spectrins (alpha/beta dimers) are essential. Finally, the Spectrin cytoskeleton is required to regulate the localisation of the Hippo pathway effector YAP in response to cell density human epithelial cells. Our findings identify both apical and basolateral Spectrins as regulators of Hippo signalling and suggest Spectrins as potential mechanosensors.


Subject(s)
Cytoskeleton/metabolism , Drosophila Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Mechanotransduction, Cellular/physiology , Ovarian Follicle/metabolism , Protein Serine-Threonine Kinases/metabolism , Spectrin/metabolism , Animals , Cell Line , Cytoskeleton/genetics , Drosophila Proteins/genetics , Drosophila melanogaster , Female , Humans , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Ovarian Follicle/cytology , Protein Serine-Threonine Kinases/genetics , Spectrin/genetics , Trans-Activators/genetics , Trans-Activators/metabolism , YAP-Signaling Proteins
13.
Development ; 143(10): 1674-87, 2016 05 15.
Article in English | MEDLINE | ID: mdl-26989177

ABSTRACT

The skin is a squamous epithelium that is continuously renewed by a population of basal layer stem/progenitor cells and can heal wounds. Here, we show that the transcription regulators YAP and TAZ localise to the nucleus in the basal layer of skin and are elevated upon wound healing. Skin-specific deletion of both YAP and TAZ in adult mice slows proliferation of basal layer cells, leads to hair loss and impairs regeneration after wounding. Contact with the basal extracellular matrix and consequent integrin-Src signalling is a key determinant of the nuclear localisation of YAP/TAZ in basal layer cells and in skin tumours. Contact with the basement membrane is lost in differentiating daughter cells, where YAP and TAZ become mostly cytoplasmic. In other types of squamous epithelia and squamous cell carcinomas, a similar control mechanism is present. By contrast, columnar epithelia differentiate an apical domain that recruits CRB3, Merlin (also known as NF2), KIBRA (also known as WWC1) and SAV1 to induce Hippo signalling and retain YAP/TAZ in the cytoplasm despite contact with the basal layer extracellular matrix. When columnar epithelial tumours lose their apical domain and become invasive, YAP/TAZ becomes nuclear and tumour growth becomes sensitive to the Src inhibitor Dasatinib.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Homeostasis , Integrins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Phosphoproteins/metabolism , Signal Transduction , Skin/metabolism , Animals , Cell Cycle Proteins , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Dasatinib/pharmacology , Epithelium/drug effects , Epithelium/metabolism , ErbB Receptors/metabolism , Gene Expression Regulation/drug effects , Homeostasis/drug effects , Humans , Keratinocytes/drug effects , Keratinocytes/metabolism , Mice , Neoplasms, Squamous Cell/pathology , Phosphatidylinositol 3-Kinases/metabolism , Protein Stability/drug effects , Protein Transport/drug effects , Signal Transduction/drug effects , Skin/drug effects , Skin/pathology , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/metabolism , Trans-Activators , Transcription Factors , Transcriptional Coactivator with PDZ-Binding Motif Proteins , Wound Healing/drug effects , YAP-Signaling Proteins , src-Family Kinases/metabolism
14.
Genes Dev ; 25(2): 131-6, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21245166

ABSTRACT

Tissues can grow in a particular direction by controlling the orientation of cell divisions. This phenomenon is evident in the developing Drosophila wing epithelium, where the tissue becomes elongated along the proximal-distal axis. We show that orientation of cell divisions in the wing requires planar polarization of an atypical myosin, Dachs. Our evidence suggests that Dachs constricts cell-cell junctions to alter the geometry of cell shapes at the apical surface, and that cell shape then determines the orientation of the mitotic spindle. Using a computational model of a growing epithelium, we show that polarized cell tension is sufficient to orient cell shapes, cell divisions, and tissue growth. Planar polarization of Dachs is ultimately oriented by long-range gradients emanating from compartment boundaries, and is therefore a mechanism linking these gradients with the control of tissue shape.


Subject(s)
Cell Polarity/physiology , Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Gene Expression Regulation, Developmental , Myosins/metabolism , Animals , Cell Division/genetics , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Spindle Apparatus/metabolism , Wings, Animal/cytology , Wings, Animal/embryology
15.
J Cell Sci ; 129(13): 2651-9, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27231092

ABSTRACT

In epithelial tissues, polarisation of microtubules and actin microvilli occurs along the apical-basal axis of each cell, yet how these cytoskeletal polarisation events are coordinated remains unclear. Here, we examine the hierarchy of events during cytoskeletal polarisation in Drosophila melanogaster epithelia. Core apical-basal polarity determinants polarise the spectrin cytoskeleton to recruit the microtubule-binding proteins Patronin (CAMSAP1, CAMSAP2 and CAMSAP3 in humans) and Shortstop [Shot; MACF1 and BPAG1 (also known as DST) in humans] to the apical membrane domain. Patronin and Shot then act to polarise microtubules along the apical-basal axis to enable apical transport of Rab11 endosomes by the Nuf-Dynein microtubule motor complex. Finally, Rab11 endosomes are transferred to the MyoV (also known as Didum in Drosophila) actin motor to deliver the key microvillar determinant Cadherin 99C to the apical membrane to organise the biogenesis of actin microvilli.


Subject(s)
Drosophila Proteins/genetics , Microfilament Proteins/genetics , Microtubule-Associated Proteins/genetics , Microvilli/metabolism , Myosin Type V/genetics , rab GTP-Binding Proteins/genetics , Actin Cytoskeleton/genetics , Actin Cytoskeleton/metabolism , Animals , Cadherins/metabolism , Cell Membrane/genetics , Cell Membrane/metabolism , Cell Polarity/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Epithelium/growth & development , Epithelium/metabolism , Humans , Microtubules/genetics , Microvilli/genetics , Myosin Type V/metabolism , Protein Transport/genetics , rab GTP-Binding Proteins/metabolism
16.
Bioessays ; 38(7): 644-53, 2016 07.
Article in English | MEDLINE | ID: mdl-27173018

ABSTRACT

The YAP/TAZ family of transcriptional co-activators drives cell proliferation in epithelial tissues and cancers. Yet, how YAP and TAZ are physiologically regulated remains unclear. Here we review recent reports that YAP and TAZ act primarily as sensors of epithelial cell polarity, being inhibited when cells differentiate an apical membrane domain, and being activated when cells contact the extracellular matrix via their basal membrane domain. Apical signalling occurs via the canonical Crumbs/CRB-Hippo/MST-Warts/LATS kinase cascade to phosphorylate and inhibit YAP/TAZ. Basal signalling occurs via Integrins and Src family kinases to phosphorylate and activate YAP/TAZ. Thus, YAP/TAZ is localised to the nucleus in basal stem/progenitor cells and cytoplasm in differentiated squamous cells or columnar cells. In addition, other signals such as mechanical forces, tissue damage and possibly receptor tyrosine kinases (RTKs) can influence MST-LATS or Src family kinase activity to modulate YAP/TAZ activity.


Subject(s)
Cell Polarity , Nuclear Proteins/physiology , Phosphotransferases (Alcohol Group Acceptor)/physiology , Signal Transduction , Stem Cells/metabolism , Transcription Factors/physiology , Animals , Biomechanical Phenomena , Cell Cycle Proteins , Drosophila Proteins/physiology , Epithelium/metabolism , Epithelium/physiology , Humans , RNA-Binding Proteins , Repressor Proteins , Stem Cells/physiology , Trans-Activators/physiology , YAP-Signaling Proteins
17.
EMBO J ; 32(21): 2790-803, 2013 Oct 30.
Article in English | MEDLINE | ID: mdl-24022370

ABSTRACT

Orientation of cell divisions is a key mechanism of tissue morphogenesis. In the growing Drosophila wing imaginal disc epithelium, most of the cell divisions in the central wing pouch are oriented along the proximal-distal (P-D) axis by the Dachsous-Fat-Dachs planar polarity pathway. However, cells at the periphery of the wing pouch instead tend to orient their divisions perpendicular to the P-D axis despite strong Dachs polarization. Here, we show that these circumferential divisions are oriented by circumferential mechanical forces that influence cell shapes and thus orient the mitotic spindle. We propose that this circumferential pattern of force is not generated locally by polarized constriction of individual epithelial cells. Instead, these forces emerge as a global tension pattern that appears to originate from differential rates of cell proliferation within the wing pouch. Accordingly, we show that localized overgrowth is sufficient to induce neighbouring cell stretching and reorientation of cell division. Our results suggest that patterned rates of cell proliferation can influence tissue mechanics and thus determine the orientation of cell divisions and tissue shape.


Subject(s)
Drosophila/cytology , Wings, Animal/cytology , Animals , Cell Division , Cell Proliferation , Drosophila/growth & development , Epithelial Cells/cytology , Models, Biological , Wings, Animal/growth & development
18.
Development ; 141(21): 4098-103, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25256343

ABSTRACT

The atypical cadherins Dachsous (Ds) and Fat (Ft) are required to control the size and shape of tissues and organs in animals. In Drosophila, a key effector of Ds and Ft is the atypical myosin Dachs, which becomes planar polarised along the proximal-distal axis in developing epithelia to regulate tissue size via the Hippo pathway and tissue shape via modulating tension at junctions. How Ds and Ft control Dachs polarisation remains unclear. Here, we identify a ubiquitin ligase, FbxL7, as a novel component of the Ds-Ft-Dachs system that is required to control the level and localisation of Dachs. Loss of FbxL7 results in accumulation of Dachs, similar to loss of Ft. Overexpression of FbxL7 causes downregulation of Dachs, similar to overexpression of the Ft intracellular domain. In addition to regulating Dachs, FbxL7 also influences Ds in a similar manner. GFP-tagged FbxL7 localises to the plasma membrane in a Ft-dependent manner and is planar polarised. We propose that Ft recruits FbxL7 to the proximal side of the cell to help restrict Ds and Dachs to the distal side of the cell.


Subject(s)
Cadherins/metabolism , Cell Adhesion Molecules/metabolism , Cell Polarity/physiology , Drosophila Proteins/metabolism , Myosins/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Cadherins/genetics , Cell Adhesion Molecules/genetics , Cell Polarity/genetics , Drosophila , Drosophila Proteins/genetics , Myosins/genetics , Ubiquitin-Protein Ligases/genetics
19.
Semin Cell Dev Biol ; 28: 70-7, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24583474

ABSTRACT

Human colorectal cancers arise as benign adenomas, tumours that retain their epithelial character, and then progress to malignant adenocarcinomas and carcinomas in which the epithelium becomes disrupted. Carcinomas often exhibit transcriptional downregulation of E-cadherin and other epithelial genes in an epithelial-to-mesenchymal transition (EMT), a mechanism first discovered in Drosophila to be mediated by the transcription factors Twist and Snail. In contrast, adenocarcinomas retain expression of E-cadherin and disruption of the epithelium occurs through formation of progressively smaller epithelial cysts with apical Crumbs/CRB3, Stardust/PALS1, and Bazooka/PAR3 localised to the inner lumen. Results from Drosophila show that morphologically similar cysts form upon induction of clonal heterogeneity in Wnt, Smad, or Ras signalling levels, which causes extrusion of epithelial cells at clonal boundaries. Thus, intratumour heterogeneity might also promote formation of adenocarcinomas in humans. Finally, epithelial cysts can collectively migrate, as in the case of Drosophila border cells, a potential model system for the invasive migration of adenocarcinoma cells.


Subject(s)
Cadherins/metabolism , Colorectal Neoplasms/metabolism , Epithelium/metabolism , Signal Transduction/physiology , Transcription Factors/metabolism , Animals , Cell Movement/physiology , Drosophila , Epithelium/pathology , Humans
20.
Development ; 140(1): 13-21, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23222437

ABSTRACT

Determinants of cell polarity orient the behaviour of many cell types during development. Pioneering genetic screens in yeast, worms and flies have identified key polarity determinants that are evolutionarily conserved across the animal kingdom. Recent work in these three model organisms has combined computer modelling with experimental analysis to reveal the molecular mechanisms that drive the polarisation of determinants. Two key principles have emerged: the first is the requirement for a positive-feedback loop to drive self-recruitment of determinants to the plasma membrane; the second is the requirement for mutual antagonism between determinants that localise to opposite ends of the cell.


Subject(s)
Cell Polarity/physiology , Diptera/cytology , Helminths/cytology , Models, Anatomic , Models, Biological , Saccharomyces cerevisiae/cytology , Animals , Caenorhabditis elegans/cytology , Caenorhabditis elegans/physiology , Computer Simulation , Diptera/physiology , Epithelium/physiology , Helminths/physiology , Humans , Saccharomyces cerevisiae/physiology , Saccharomycetales/cytology , Saccharomycetales/physiology , Zygote/cytology , Zygote/physiology
SELECTION OF CITATIONS
SEARCH DETAIL