Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Cell Commun Signal ; 22(1): 152, 2024 02 27.
Article in English | MEDLINE | ID: mdl-38414029

ABSTRACT

BACKGROUND: Germline mutations of E-cadherin contribute to hereditary diffuse gastric cancer (HDGC) and congenital malformations, such as oral facial clefts (OFC). However, the molecular mechanisms through which E-cadherin loss-of-function triggers distinct clinical outcomes remain unknown. We postulate that E-cadherin-mediated disorders result from abnormal interactions with the extracellular matrix and consequent aberrant intracellular signalling, affecting the coordination of cell migration. METHODS: Herein, we developed in vivo and in vitro models of E-cadherin mutants associated with either OFC or HDGC. Using a Drosophila approach, we addressed the impact of the different variants in cell morphology and migration ability. By combining gap closure migration assays and time-lapse microscopy, we further investigated the migration pattern of cells expressing OFC or HDGC variants. The adhesion profile of the variants was evaluated using high-throughput ECM arrays, whereas RNA sequencing technology was explored for identification of genes involved in aberrant cell motility. RESULTS: We have demonstrated that cells expressing OFC variants exhibit an excessive motility performance and irregular leading edges, which prevent the coordinated movement of the epithelial monolayer. Importantly, we found that OFC variants promote cell adhesion to a wider variety of extracellular matrices than HDGC variants, suggesting higher plasticity in response to different microenvironments. We unveiled a distinct transcriptomic profile in the OFC setting and pinpointed REG1A as a putative regulator of this outcome. Consistent with this, specific RNAi-mediated inhibition of REG1A shifted the migration pattern of OFC expressing cells, leading to slower wound closure with coordinated leading edges. CONCLUSIONS: We provide evidence that E-cadherin variants associated with OFC activate aberrant signalling pathways that support dynamic rearrangements of cells towards improved adaptability to the microenvironment. This proficiency results in abnormal tissue shaping and movement, possibly underlying the development of orofacial malformations.


Subject(s)
Adenocarcinoma , Stomach Neoplasms , Humans , Cadherins/genetics , Cadherins/metabolism , Cell Adhesion , Cell Movement , Germ-Line Mutation , Lithostathine/genetics , Stomach Neoplasms/metabolism , Tumor Microenvironment , Animals , Drosophila melanogaster
2.
Curr Biol ; 33(5): 858-874.e7, 2023 03 13.
Article in English | MEDLINE | ID: mdl-36917931

ABSTRACT

Cell proliferation is central to epithelial tissue development, repair, and homeostasis. During cell division, small RhoGTPases control both actomyosin dynamics and cell-cell junction remodeling to faithfully segregate the genome while maintaining tissue polarity and integrity. To decipher the mechanisms of RhoGTPase spatiotemporal regulation during epithelial cell division, we generated a transgenic fluorescently tagged library for the 48 Drosophila Rho guanine exchange factors (RhoGEFs) and GTPase-activating proteins (GAPs), and we systematically characterized their endogenous distributions by time-lapse microscopy. Therefore, we unveiled candidate regulators of the interplay between actomyosin and junctional dynamics during epithelial cell division. Building on these findings, we established that the conserved RhoGEF Cysts and RhoGEF4 play sequential and distinct roles to couple cytokinesis with de novo junction formation. During ring contraction, Cysts via Rho1 participates in the neighbor mechanosensing response, promoting daughter-daughter cell membrane juxtaposition in preparation to de novo junction formation. Subsequently and upon midbody formation, RhoGEF4 via Rac acts in the dividing cell to ensure the withdrawal of the neighboring cell membranes, thus controlling de novo junction length and cell-cell arrangements upon cytokinesis. Altogether, our findings delineate how the RhoGTPases Rho and Rac are locally and temporally activated during epithelial cytokinesis, highlighting the RhoGEF/GAP library as a key resource to understand the broad range of biological processes regulated by RhoGTPases.


Subject(s)
Actomyosin , Epithelial Cells , Animals , Rho Guanine Nucleotide Exchange Factors/genetics , Rho Guanine Nucleotide Exchange Factors/metabolism , Actomyosin/metabolism , Cell Division , Cytokinesis , Drosophila
3.
Dis Model Mech ; 16(2)2023 02 01.
Article in English | MEDLINE | ID: mdl-36808468

ABSTRACT

Alterations in the expression or function of cell adhesion molecules have been implicated in all steps of tumor progression. Among those, P-cadherin is highly enriched in basal-like breast carcinomas, playing a central role in cancer cell self-renewal, collective cell migration and invasion. To establish a clinically relevant platform for functional exploration of P-cadherin effectors in vivo, we generated a humanized P-cadherin Drosophila model. We report that actin nucleators, Mrtf and Srf, are main P-cadherin effectors in fly. We validated these findings in a human mammary epithelial cell line with conditional activation of the SRC oncogene. We show that, prior to promoting malignant phenotypes, SRC induces a transient increase in P-cadherin expression, which correlates with MRTF-A accumulation, its nuclear translocation and the upregulation of SRF target genes. Moreover, knocking down P-cadherin, or preventing F-actin polymerization, impairs SRF transcriptional activity. Furthermore, blocking MRTF-A nuclear translocation hampers proliferation, self-renewal and invasion. Thus, in addition to sustaining malignant phenotypes, P-cadherin can also play a major role in the early stages of breast carcinogenesis by promoting a transient boost of MRTF-A-SRF signaling through actin regulation.


Subject(s)
Actins , Trans-Activators , Humans , Actins/metabolism , Trans-Activators/metabolism , Signal Transduction , Cadherins , Epithelial Cells/metabolism , Serum Response Factor/genetics , Serum Response Factor/metabolism
4.
Proc Natl Acad Sci U S A ; 119(42): e2204701119, 2022 10 18.
Article in English | MEDLINE | ID: mdl-36215502

ABSTRACT

The synaptonemal complex (SC) is a proteinaceous scaffold that is assembled between paired homologous chromosomes during the onset of meiosis. Timely expression of SC coding genes is essential for SC assembly and successful meiosis. However, SC components have an intrinsic tendency to self-organize into abnormal repetitive structures, which are not assembled between the paired homologs and whose formation is potentially deleterious for meiosis and gametogenesis. This creates an interesting conundrum, where SC genes need to be robustly expressed during meiosis, but their expression must be carefully regulated to prevent the formation of anomalous SC structures. In this manuscript, we show that the Polycomb group protein Sfmbt, the Drosophila ortholog of human MBTD1 and L3MBTL2, is required to avoid excessive expression of SC genes during prophase I. Although SC assembly is normal after Sfmbt depletion, SC disassembly is abnormal with the formation of multiple synaptonemal complexes (polycomplexes) within the oocyte. Overexpression of the SC gene corona and depletion of other Polycomb group proteins are similarly associated with polycomplex formation during SC disassembly. These polycomplexes are highly dynamic and have a well-defined periodic structure. Further confirming the importance of Sfmbt, germ line depletion of this protein is associated with significant metaphase I defects and a reduction in female fertility. Since transcription of SC genes mostly occurs during early prophase I, our results suggest a role of Sfmbt and other Polycomb group proteins in downregulating the expression of these and other early prophase I genes during later stages of meiosis.


Subject(s)
Meiosis , Synaptonemal Complex , Chromosomal Proteins, Non-Histone/genetics , Chromosome Pairing , Female , Humans , Meiotic Prophase I , Polycomb-Group Proteins/genetics , Synaptonemal Complex/genetics
5.
Curr Biol ; 32(20): 4411-4427.e8, 2022 10 24.
Article in English | MEDLINE | ID: mdl-36113470

ABSTRACT

Apical-basal polarity is an essential epithelial trait controlled by the evolutionarily conserved PAR-aPKC polarity network. Dysregulation of polarity proteins disrupts tissue organization during development and in disease, but the underlying mechanisms are unclear due to the broad implications of polarity loss. Here, we uncover how Drosophila aPKC maintains epithelial architecture by directly observing tissue disorganization after fast optogenetic inactivation in living adult flies and ovaries cultured ex vivo. We show that fast aPKC perturbation in the proliferative follicular epithelium produces large epithelial gaps that result from increased apical constriction, rather than loss of apical-basal polarity. Accordingly, we can modulate the incidence of epithelial gaps by increasing and decreasing actomyosin-driven contractility. We traced the origin of these large epithelial gaps to tissue rupture next to dividing cells. Live imaging shows that aPKC perturbation induces apical constriction in non-mitotic cells within minutes, producing pulling forces that ultimately detach dividing and neighboring cells. We further demonstrate that epithelial rupture requires a global increase of apical constriction, as it is prevented by the presence of non-constricting cells. Conversely, a global induction of apical tension through light-induced recruitment of RhoGEF2 to the apical side is sufficient to produce tissue rupture. Hence, our work reveals that the roles of aPKC in polarity and actomyosin regulation are separable and provides the first in vivo evidence that excessive tissue stress can break the epithelial barrier during proliferation.


Subject(s)
Drosophila Proteins , Drosophila , Animals , Drosophila/metabolism , Actomyosin/metabolism , Drosophila Proteins/metabolism , Cell Polarity/physiology , Constriction , Protein Kinase C/genetics , Protein Kinase C/metabolism , Epithelium/metabolism , Epithelial Cells/metabolism , Drosophila melanogaster/metabolism
6.
BMJ Case Rep ; 15(9)2022 Sep 13.
Article in English | MEDLINE | ID: mdl-36100284

ABSTRACT

Systemic sclerosis is an autoimmune disease that can result in lung fibrosis, and is strongly associated with the presence of serum anti-topoisomerase-I autoantibodies. A young man with genetic muscular dystrophy caused by titin-cap/telethonin (TCAP) gene mutation, developed a severe restrictive lung disease due to a fibrosing interstitial pneumonia secondary to systemic sclerosis with positive anti-topoisomerase-I antibodies. Using amino acid sequence alignment and protein structure modelling, we found that mutant telethonin exposes an amino acid sequence with significant homology to an immunodominant site of topoisomerase-I. Abnormal telethonin results in a loss of integrity of the sarcomere structure, which might result in rhabdomyolysis and abnormal protein exposure to the immune system. Our preliminary analysis suggests a possible role for mutant sarcomere protein telethonin as an immunogenic target recognised by anti-topoisomerase-I antibodies, which could explain the development of systemic sclerosis in this particular patient.


Subject(s)
Autoimmune Diseases , Muscular Dystrophies , Scleroderma, Systemic , Antibodies, Antinuclear , Humans , Male , Muscles , Muscular Dystrophies/genetics , Scleroderma, Systemic/complications , Scleroderma, Systemic/genetics
7.
J Cell Biol ; 221(10)2022 10 03.
Article in English | MEDLINE | ID: mdl-36121422

ABSTRACT

How cells spatially organize their plasma membrane, cytoskeleton, and cytoplasm remains a central question for cell biologists. In this issue of JCB, Calvi et al. (2022. J. Cell Biol.https://doi.org/10.1083/jcb.202201048) identify PP1 phosphatases as key regulators of C. elegans anterior-posterior polarity, by counterbalancing aPKC-mediated phosphorylation of PAR-2.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Cell Polarity , Phosphoprotein Phosphatases , Protein Kinase C , Animals , Caenorhabditis elegans/cytology , Caenorhabditis elegans/enzymology , Caenorhabditis elegans Proteins/metabolism , Cell Membrane , Cytoplasm , Microtubules/metabolism , Phosphoprotein Phosphatases/metabolism , Phosphorylation , Protein Kinase C/metabolism
8.
Gastric Cancer ; 25(1): 124-137, 2022 01.
Article in English | MEDLINE | ID: mdl-34486077

ABSTRACT

BACKGROUND: Tumour progression relies on the ability of cancer cells to penetrate and invade neighbouring tissues. E-cadherin loss is associated with increased cell invasion in gastric carcinoma, and germline mutations of the E-cadherin gene are causative of hereditary diffuse gastric cancer. Although E-cadherin dysfunction impacts cell-cell adhesion, cell dissemination also requires an imbalance of adhesion to the extracellular matrix (ECM). METHODS: To identify ECM components and receptors relevant for adhesion of E-cadherin dysfunctional cells, we implemented a novel ECM microarray platform coupled with molecular interaction networks. The functional role of putative candidates was determined by combining micropattern traction microscopy, protein modulation and in vivo approaches, as well as transcriptomic data of 262 gastric carcinoma samples, retrieved from the cancer genome atlas (TCGA). RESULTS: Here, we show that E-cadherin mutations induce an abnormal interplay of cells with specific components of the ECM, which encompasses increased traction forces and Integrin ß1 activation. Integrin ß1 synergizes with E-cadherin dysfunction, promoting cell scattering and invasion. The significance of the E-cadherin-Integrin ß1 crosstalk was validated in Drosophila models and found to be consistent with evidence from human gastric carcinomas, where increased tumour grade and poor survival are associated with low E-cadherin and high Integrin ß1 levels. CONCLUSIONS: Integrin ß1 is a key mediator of invasion in carcinomas with E-cadherin impairment and should be regarded as a biomarker of poor prognosis in gastric cancer.


Subject(s)
Integrin beta1 , Stomach Neoplasms , Animals , Cadherins/genetics , Cadherins/metabolism , Cell Adhesion/physiology , Drosophila melanogaster , Extracellular Matrix/metabolism , Humans , Integrin beta1/genetics , Integrin beta1/metabolism , Neoplasm Invasiveness , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism
9.
Cancers (Basel) ; 13(17)2021 Aug 28.
Article in English | MEDLINE | ID: mdl-34503169

ABSTRACT

E-cadherin, encoded by CDH1, is an essential molecule for epithelial homeostasis, whose loss or aberrant expression results in disturbed cell-cell adhesion, increased cell invasion and metastasis. Carriers of CDH1 germline mutations have a high risk of developing diffuse gastric cancer and lobular breast cancer, associated with the cancer syndrome Hereditary Diffuse Gastric Cancer (HDGC). The ubiquitous availability of cancer panels has led to the identification of an increasing amount of "incidental" CDH1 genetic variants that pose a serious clinical challenge. This has sparked intensive research aiming at an accurate classification of the variants and consequent validation of their clinical relevance. The present study addressed the significance of a novel CDH1 variant, G212E, identified in an unusually large pedigree displaying strong aggregation of diffuse gastric cancer. We undertook a comprehensive pipeline encompassing family data, in silico predictions, in vitro assays and in vivo strategies, which validated the deleterious phenotype induced by this genetic alteration. In particular, we demonstrated that the G212E variant affects the stability and localization, as well as the adhesive and anti-invasive functions of E-cadherin, triggering epithelial disruption and disorganization. Our findings illustrate the clinical implication of a complementary approach for effective variant categorization and patient management.

10.
Nat Struct Mol Biol ; 28(3): 278-289, 2021 03.
Article in English | MEDLINE | ID: mdl-33633400

ABSTRACT

Trim-Away is a recently developed technology that exploits off-the-shelf antibodies and the RING E3 ligase and cytosolic antibody receptor TRIM21 to carry out rapid protein depletion. How TRIM21 is catalytically activated upon target engagement, either during its normal immune function or when repurposed for targeted protein degradation, is unknown. Here we show that a mechanism of target-induced clustering triggers intermolecular dimerization of the RING domain to switch on the ubiquitination activity of TRIM21 and induce virus neutralization or drive Trim-Away. We harness this mechanism for selective degradation of disease-causing huntingtin protein containing long polyglutamine tracts and expand the Trim-Away toolbox with highly active TRIM21-nanobody chimeras that can also be controlled optogenetically. This work provides a mechanism for cellular activation of TRIM RING ligases and has implications for targeted protein degradation technologies.


Subject(s)
Proteolysis , Ribonucleoproteins/metabolism , Ubiquitination , Animals , Biocatalysis , Cell Line , Drosophila melanogaster/cytology , Humans , Huntingtin Protein/chemistry , Huntingtin Protein/metabolism , Mice , Models, Molecular , Optogenetics , Peptides/metabolism , Protein Binding , Protein Multimerization , Ribonucleoproteins/chemistry , Ubiquitin-Protein Ligases/metabolism
11.
Development ; 147(15)2020 08 07.
Article in English | MEDLINE | ID: mdl-32665243

ABSTRACT

Apical-basal polarity underpins the formation of epithelial barriers that are crucial for metazoan physiology. Although apical-basal polarity is long known to require the basolateral determinants Lethal Giant Larvae (Lgl), Discs Large (Dlg) and Scribble (Scrib), mechanistic understanding of their function is limited. Lgl plays a role as an aPKC inhibitor, but it remains unclear whether Lgl also forms complexes with Dlg or Scrib. Using fluorescence recovery after photobleaching, we show that Lgl does not form immobile complexes at the lateral domain of Drosophila follicle cells. Optogenetic depletion of plasma membrane PIP2 or dlg mutants accelerate Lgl cortical dynamics. However, Dlg and Scrib are required only for Lgl localization and dynamic behavior in the presence of aPKC function. Furthermore, light-induced oligomerization of basolateral proteins indicates that Lgl is not part of the Scrib-Dlg complex in the follicular epithelium. Thus, Scrib and Dlg are necessary to repress aPKC activity in the lateral domain but do not provide cortical binding sites for Lgl. Our work therefore highlights that Lgl does not act in a complex but in parallel with Scrib-Dlg to antagonize apical determinants.


Subject(s)
Drosophila Proteins/metabolism , Membrane Proteins/metabolism , Multiprotein Complexes/metabolism , Ovarian Follicle/metabolism , Protein Kinase C/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Drosophila Proteins/genetics , Drosophila melanogaster , Female , Membrane Proteins/genetics , Multiprotein Complexes/genetics , Protein Binding , Protein Kinase C/genetics , Tumor Suppressor Proteins/genetics
12.
Elife ; 82019 08 19.
Article in English | MEDLINE | ID: mdl-31424385

ABSTRACT

According to the prevailing 'clock' model, chromosome decondensation and nuclear envelope reformation when cells exit mitosis are byproducts of Cdk1 inactivation at the metaphase-anaphase transition, controlled by the spindle assembly checkpoint. However, mitotic exit was recently shown to be a function of chromosome separation during anaphase, assisted by a midzone Aurora B phosphorylation gradient - the 'ruler' model. Here we found that Cdk1 remains active during anaphase due to ongoing APC/CCdc20- and APC/CCdh1-mediated degradation of B-type Cyclins in Drosophila and human cells. Failure to degrade B-type Cyclins during anaphase prevented mitotic exit in a Cdk1-dependent manner. Cyclin B1-Cdk1 localized at the spindle midzone in an Aurora B-dependent manner, with incompletely separated chromosomes showing the highest Cdk1 activity. Slowing down anaphase chromosome motion delayed Cyclin B1 degradation and mitotic exit in an Aurora B-dependent manner. Thus, a crosstalk between molecular 'rulers' and 'clocks' licenses mitotic exit only after proper chromosome separation.


Subject(s)
Anaphase , Aurora Kinase B/metabolism , CDC2 Protein Kinase/metabolism , Cyclin B1/metabolism , Drosophila Proteins/metabolism , Animals , Cell Line , Drosophila , Humans , Proteolysis , Spatio-Temporal Analysis
13.
Curr Opin Cell Biol ; 60: 75-83, 2019 10.
Article in English | MEDLINE | ID: mdl-31153057

ABSTRACT

Epithelial cell division is essential to shape developing tissues and for the homeostasis of adult organs. However, the mechanical and biochemical reorganization of dividing cells represents a major challenge to the integrity and architecture of the epithelial barrier. Here, we portray recent findings from a variety of model organisms that revealed how apical-basal polarity and intercellular adhesion are modulated during cell division to maintain a permeability barrier and to transmit epithelial organization to daughter cells. This demands not only that dividing and neighboring cells remodel their adhesion and contractility, but also cell cycle-dependent regulation of apical-basal polarity in the dividing cell. Additionally, mitotic structures, such as the midbody, provide spatial cues to enforce epithelial cell organization.


Subject(s)
Cell Division , Cell Polarity , Epithelial Cells/cytology , Intercellular Junctions/metabolism , Animals , Epithelial Cells/metabolism , Humans , Mitosis , Models, Biological
14.
Biomolecules ; 9(2)2019 02 12.
Article in English | MEDLINE | ID: mdl-30759894

ABSTRACT

Drosophila Schneider 2 (S2) cells are a simple and powerful system commonly used in cell biology because they are well suited for high resolution microscopy and RNAi-mediated depletion. However, understanding dynamic processes, such as cell division, also requires methodology to interfere with protein function with high spatiotemporal control. In this research study, we report the adaptation of an optogenetic tool to Drosophila S2 cells. Light-activated reversible inhibition by assembled trap (LARIAT) relies on the rapid light-dependent heterodimerization between cryptochrome 2 (CRY2) and cryptochrome-interacting bHLH 1 (CIB1) to form large protein clusters. An anti-green fluorescent protein (GFP) nanobody fused with CRY2 allows this method to quickly trap any GFP-tagged protein in these light-induced protein clusters. We evaluated clustering kinetics in response to light for different LARIAT modules, and showed the ability of GFP-LARIAT to inactivate the mitotic protein Mps1 and to disrupt the membrane localization of the polarity regulator Lethal Giant Larvae (Lgl). Moreover, we validated light-induced co-clustering assays to assess protein-protein interactions in S2 cells. In conclusion, GFP-based LARIAT is a versatile tool to answer different biological questions, since it enables probing of dynamic processes and protein-protein interactions with high spatiotemporal resolution in Drosophila S2 cells.


Subject(s)
Drosophila Proteins/metabolism , Light , Optogenetics , Animals , Cell Line , Drosophila , Drosophila Proteins/chemistry , Protein Binding
15.
Cell Rep ; 26(2): 293-301.e7, 2019 01 08.
Article in English | MEDLINE | ID: mdl-30625311

ABSTRACT

Apical-basal polarity is a common trait that underlies epithelial function. Although the asymmetric distribution of cortical polarity proteins works in a functioning equilibrium, it also retains plasticity to accommodate cell division, during which the basolateral determinant Lgl is released from the cortex. Here, we investigated how Lgl restores its cortical localization to maintain the integrity of dividing epithelia. We show that cytoplasmic Lgl is reloaded to the cortex at mitotic exit in Drosophila epithelia. Lgl cortical localization depends on protein phosphatase 1, which dephosphorylates Lgl on the serines phosphorylated by aPKC and Aurora A kinases through a mechanism that relies on the regulatory subunit Sds22 and a PP1-interacting RVxF motif of Lgl. This mechanism maintains epithelial polarity and is of particular importance at mitotic exit to couple Lgl cortical reloading with the polarization of the apical domain. Hence, PP1-mediated dephosphorylation of Lgl preserves the apical-basal organization of proliferative epithelia.


Subject(s)
Cell Polarity , Drosophila Proteins/metabolism , Protein Phosphatase 1/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Aurora Kinase A/metabolism , Binding Sites , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Drosophila melanogaster , Epithelial Cells/metabolism , Mitosis , Protein Binding , Protein Transport , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/genetics
16.
Int J Mol Sci ; 18(12)2017 Dec 12.
Article in English | MEDLINE | ID: mdl-29231860

ABSTRACT

The role of E-cadherin in Hereditary Diffuse Gastric Cancer (HDGC) is unequivocal. Germline alterations in its encoding gene (CDH1) are causative of HDGC and occur in about 40% of patients. Importantly, while in most cases CDH1 alterations result in the complete loss of E-cadherin associated with a well-established clinical impact, in about 20% of cases the mutations are of the missense type. The latter are of particular concern in terms of genetic counselling and clinical management, as the effect of the sequence variants in E-cadherin function is not predictable. If a deleterious variant is identified, prophylactic surgery could be recommended. Therefore, over the last few years, intensive research has focused on evaluating the functional consequences of CDH1 missense variants and in assessing E-cadherin pathogenicity. In that context, our group has contributed to better characterize CDH1 germline missense variants and is now considered a worldwide reference centre. In this review, we highlight the state of the art methodologies to categorize CDH1 variants, as neutral or deleterious. This information is subsequently integrated with clinical data for genetic counseling and management of CDH1 variant carriers.


Subject(s)
Cadherins/genetics , Genetic Predisposition to Disease/genetics , Mutation, Missense , Stomach Neoplasms/genetics , Antigens, CD , Cell Adhesion/genetics , Cell Movement/genetics , Genetic Counseling , Germ-Line Mutation , Heterozygote , Humans , Stomach Neoplasms/pathology
17.
Bioarchitecture ; 6(2): 29-38, 2016.
Article in English | MEDLINE | ID: mdl-26919260

ABSTRACT

Intracellular asymmetries, often termed cell polarity, determine how cells organize and divide to ultimately control cell fate and shape animal tissues. The tumor suppressor Lethal giant larvae (Lgl) functions at the core of the evolutionarily conserved cell polarity machinery that controls apico-basal polarization. This function relies on its restricted basolateral localization via phosphorylation by aPKC. Here, we summarize the spatial and temporal control of Lgl during the cell cycle, highlighting two ideas that emerged from our recent findings: 1) Aurora A directly phosphorylates Lgl during symmetric division to couple reorganization of epithelial polarity with the cell cycle; 2) Phosphorylation of Lgl within three conserved serines controls its localization and function in a site-specific manner. Considering the importance of phosphorylation to regulate the concentration of Lgl at the plasma membrane, we will further discuss how it may work as an on-off switch for the interaction with cortical binding partners, with implications on epithelial polarization and spindle orientation.


Subject(s)
Aurora Kinase A/metabolism , Cell Division , Drosophila Proteins/metabolism , Protein Kinase C/metabolism , Spindle Apparatus/physiology , Tumor Suppressor Proteins/metabolism , Animals
18.
Curr Biol ; 25(1): 53-60, 2015 Jan 05.
Article in English | MEDLINE | ID: mdl-25484294

ABSTRACT

Mitotic spindle orientation is essential to control cell-fate specification and epithelial architecture. The tumor suppressor Lgl localizes to the basolateral cortex of epithelial cells, where it acts together with Dlg and Scrib to organize apicobasal polarity. Dlg and Scrib also control planar spindle orientation, but how the organization of polarity complexes is adjusted to control symmetric division is largely unknown. Here, we show that the Dlg complex is remodeled during Drosophila follicular epithelium cell division, when Lgl is released to the cytoplasm. Lgl redistribution during epithelial mitosis is reminiscent of asymmetric cell division, where it is proposed that Aurora A promotes aPKC activation to control the localization of Lgl and cell-fate determinants. We show that Aurora A controls Lgl localization directly, triggering its cortical release at early prophase in both epithelial and S2 cells. This relies on double phosphorylation within the putative aPKC phosphorylation site, which is required and sufficient for Lgl cortical release during mitosis and can be achieved by a combination of aPKC and Aurora A activities. Cortical retention of Lgl disrupts planar spindle orientation, but only when Lgl mutants that can bind Dlg are expressed. Hence, our work reveals that Lgl mitotic cortical release is not specifically linked to the asymmetric segregation of fate determinants, and we propose that Aurora A activation breaks the Dlg/Lgl interaction to allow planar spindle orientation during symmetric division via the Pins (LGN)/Dlg pathway.


Subject(s)
Aurora Kinase A/metabolism , Cell Division , Drosophila Proteins/metabolism , Protein Kinase C/metabolism , Spindle Apparatus/physiology , Tumor Suppressor Proteins/metabolism , Animals , Cell Cycle Proteins , Cell Polarity , Drosophila , Epithelial Cells/physiology , Guanine Nucleotide Dissociation Inhibitors/metabolism
19.
Development ; 141(15): 2984-92, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25053432

ABSTRACT

The Drosophila anterior-posterior axis is specified when the posterior follicle cells signal to polarise the oocyte, leading to the anterior/lateral localisation of the Par-6/aPKC complex and the posterior recruitment of Par-1, which induces a microtubule reorganisation that localises bicoid and oskar mRNAs. Here we show that oocyte polarity requires Slmb, the substrate specificity subunit of the SCF E3 ubiquitin ligase that targets proteins for degradation. The Par-6/aPKC complex is ectopically localised to the posterior of slmb mutant oocytes, and Par-1 and oskar mRNA are mislocalised. Slmb appears to play a related role in epithelial follicle cells, as large slmb mutant clones disrupt epithelial organisation, whereas small clones show an expansion of the apical domain, with increased accumulation of apical polarity factors at the apical cortex. The levels of aPKC and Par-6 are significantly increased in slmb mutants, whereas Baz is slightly reduced. Thus, Slmb may induce the polarisation of the anterior-posterior axis of the oocyte by targeting the Par-6/aPKC complex for degradation at the oocyte posterior. Consistent with this, overexpression of the aPKC antagonist Lgl strongly rescues the polarity defects of slmb mutant germline clones. The role of Slmb in oocyte polarity raises an intriguing parallel with C. elegans axis formation, in which PAR-2 excludes the anterior PAR complex from the posterior cortex to induce polarity, but its function can be substituted by overexpressing Lgl.


Subject(s)
Cell Cycle Proteins/physiology , Drosophila Proteins/metabolism , Drosophila Proteins/physiology , Epithelial Cells/cytology , Oocytes/cytology , Protein Kinase C/metabolism , Ubiquitin-Protein Ligases/physiology , Alleles , Animals , Body Patterning , Cell Cycle Proteins/metabolism , Cell Polarity , Drosophila Proteins/antagonists & inhibitors , Drosophila melanogaster/embryology , Female , Gene Expression Regulation, Developmental , Genetic Complementation Test , Green Fluorescent Proteins/metabolism , Microtubules/metabolism , Mutation , Phenotype , Protein Kinase C/antagonists & inhibitors , Protein Structure, Tertiary , RNA, Messenger/metabolism , Signal Transduction , Spectrin/metabolism , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...