Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.036
Filtrar
1.
Yi Chuan ; 46(3): 199-208, 2024 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-38632098

RESUMO

Polarity establishment is one of the key factors affecting early embryonic development. Polarity establishment begins with myosin phosphorylation in the 8-cell embryo, and phosphorylation activates actin leading to its initiation of contractility. Subsequently, actin undergoes reorganization to form an apical domain rich in microvilli on the non-contacting surface of each blastomere, and form the actomyosin ring that marks the maturation of the apical domain in conjunction with polar protein complexes and others. From the process of polarity establishment, it can be seen that the formation of the apical domain is influenced by actin-related proteins and polar protein complexes. Some zygote genome activation (ZGA) and lineage-specific genes also regulate polarity establishment. Polarity establishment underlies the first cell lineage differentiation during early embryonic development. It regulates lineage segregation and morphogenesis by affecting asymmetric cell division, asymmetric localization of lineage differentiation factors, and activity of the Hippo signaling pathway. In this review, we systematically summarize the mechanisms of early embryonic polarity establishment and its impact on lineage differentiation in mammals, and discuss the shortcomings of the currently available studies in terms of regulatory mechanisms and species, thereby providing clues and systematic perspectives for elucidating early embryonic polarity establishment.


Assuntos
Actinas , Actomiosina , Animais , Actomiosina/metabolismo , Citocinese , Diferenciação Celular , Linhagem da Célula , Polaridade Celular/fisiologia , Mamíferos/metabolismo
2.
J Cell Sci ; 137(5)2024 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-38465513

RESUMO

Drosophila neural stem cells, or neuroblasts, rapidly proliferate during embryonic and larval development to populate the central nervous system. Neuroblasts divide asymmetrically to create cellular diversity, with each division producing one sibling cell that retains the neuroblast fate and another that differentiates into glia or neurons. This asymmetric outcome is mediated by the transient polarization of numerous factors to the cell cortex during mitosis. The powerful genetics and outstanding imaging tractability of the neuroblast make it an excellent model system for studying the mechanisms of cell polarity. This Cell Science at a Glance article and the accompanying poster explore the phases of the neuroblast polarity cycle and the regulatory circuits that control them. We discuss the key features of the cycle - the targeted recruitment of proteins to specific regions of the plasma membrane and multiple phases of highly dynamic actomyosin-dependent cortical flows that pattern both protein distribution and membrane structure.


Assuntos
Proteínas de Drosophila , Células-Tronco Neurais , Animais , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Células-Tronco Neurais/metabolismo , Neurônios/metabolismo , Mitose , Proteínas de Ciclo Celular/metabolismo , Polaridade Celular/fisiologia
3.
Cells ; 13(3)2024 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-38334614

RESUMO

Planar cell polarity (PCP) proteins coordinate tissue morphogenesis by governing cell patterning and polarity. Asymmetrically localized on the plasma membrane of cells, transmembrane PCP proteins are trafficked by endocytosis, suggesting they may have intracellular functions that are dependent or independent of their extracellular role, but whether these functions extend to transcriptional control remains unknown. Here, we show the nuclear localization of transmembrane, PCP protein, VANGL2, in the HCC1569 breast cancer cell line, and in undifferentiated, but not differentiated, HC11 cells that serve as a model for mammary lactogenic differentiation. The loss of Vangl2 function results in upregulation of pathways related to STAT5 signaling. We identify DNA binding sites and a nuclear localization signal in VANGL2, and use CUT&RUN to demonstrate recruitment of VANGL2 to specific DNA binding motifs, including one in the Stat5a promoter. Knockdown (KD) of Vangl2 in HC11 cells and primary mammary organoids results in upregulation of Stat5a, Ccnd1 and Csn2, larger acini and organoids, and precocious differentiation; phenotypes are rescued by overexpression of Vangl2, but not Vangl2ΔNLS. Together, these results advance a paradigm whereby PCP proteins coordinate tissue morphogenesis by keeping transcriptional programs governing differentiation in check.


Assuntos
Polaridade Celular , Proteínas de Membrana , Polaridade Celular/fisiologia , Membrana Celular/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Transdução de Sinais , DNA/metabolismo
4.
J Cell Sci ; 137(5)2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38323986

RESUMO

Migratory cells - either individually or in cohesive groups - are critical for spatiotemporally regulated processes such as embryonic development and wound healing. Their dysregulation is the underlying cause of formidable health problems such as congenital abnormalities and metastatic cancers. Border cell behavior during Drosophila oogenesis provides an effective model to study temporally regulated, collective cell migration in vivo. Developmental timing in flies is primarily controlled by the steroid hormone ecdysone, which acts through a well-conserved, nuclear hormone receptor complex. Ecdysone signaling determines the timing of border cell migration, but the molecular mechanisms governing this remain obscure. We found that border cell clusters expressing a dominant-negative form of ecdysone receptor extended ineffective protrusions. Additionally, these clusters had aberrant spatial distributions of E-cadherin (E-cad), apical domain markers and activated myosin that did not overlap. Remediating their expression or activity individually in clusters mutant for ecdysone signaling did not restore proper migration. We propose that ecdysone signaling synchronizes the functional distribution of E-cadherin, atypical protein kinase C (aPKC), Discs large (Dlg1) and activated myosin post-transcriptionally to coordinate adhesion, polarity and contractility and temporally control collective cell migration.


Assuntos
Proteínas de Drosophila , Animais , Proteínas de Drosophila/metabolismo , Ecdisona/metabolismo , Drosophila/metabolismo , Caderinas/genética , Caderinas/metabolismo , Movimento Celular/fisiologia , Miosinas/metabolismo , Drosophila melanogaster/metabolismo , Polaridade Celular/fisiologia , Adesão Celular
5.
Curr Biol ; 34(3): 615-622.e4, 2024 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-38199065

RESUMO

Convergent extension (CE) requires the coordinated action of the planar cell polarity (PCP) proteins1,2 and the actin cytoskeleton,3,4,5,6 but this relationship remains incompletely understood. For example, PCP signaling orients actomyosin contractions, yet actomyosin is also required for the polarized localization of PCP proteins.7,8 Moreover, the actin-regulating Septins play key roles in actin organization9 and are implicated in PCP and CE in frogs, mice, and fish5,6,10,11,12 but execute only a subset of PCP-dependent cell behaviors. Septin loss recapitulates the severe tissue-level CE defects seen after core PCP disruption yet leaves overt cell polarity intact.5 Together, these results highlight the general fact that cell movement requires coordinated action by distinct but integrated actin populations, such as lamella and lamellipodia in migrating cells13 or medial and junctional actin populations in cells engaged in apical constriction.14,15 In the context of Xenopus mesoderm CE, three such actin populations are important, a superficial meshwork known as the "node-and-cable" system,4,16,17,18 a contractile network at deep cell-cell junctions,6,19 and mediolaterally oriented actin-rich protrusions, which are present both superficially and deeply.4,19,20,21 Here, we exploited the amenability of the uniquely "two-dimensional" node and cable system to probe the relationship between PCP proteins, Septins, and the polarization of this actin network. We find that the PCP proteins Vangl2 and Prickle2 and Septins co-localize at nodes, and that the node and cable system displays a cryptic, PCP- and Septin-dependent anteroposterior (AP) polarity in its organization and dynamics.


Assuntos
Actinas , Septinas , Camundongos , Animais , Septinas/metabolismo , Actinas/metabolismo , Actomiosina/metabolismo , Citoesqueleto de Actina/metabolismo , Movimento Celular/fisiologia , Polaridade Celular/fisiologia , Proteínas de Membrana/metabolismo , Proteínas com Domínio LIM/metabolismo
6.
J Cell Sci ; 137(5)2024 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-37818620

RESUMO

The membrane potential (MP) controls cell homeostasis by directing molecule transport and gene expression. How the MP is set upon epithelial differentiation is unknown. Given that tissue architecture also controls homeostasis, we investigated the relationship between basoapical polarity and resting MP in three-dimensional culture of the HMT-3522 breast cancer progression. A microelectrode technique to measure MP and input resistance reveals that the MP is raised by gap junction intercellular communication (GJIC), which directs tight-junction mediated apical polarity, and is decreased by the Na+/K+/2Cl- (NKCC, encoded by SLC12A1 and SLC12A2) co-transporter, active in multicellular structures displaying basal polarity. In the tumor counterpart, the MP is reduced. Cancer cells display diminished GJIC and do not respond to furosemide, implying loss of NKCC activity. Induced differentiation of cancer cells into basally polarized multicellular structures restores widespread GJIC and NKCC responses, but these structures display the lowest MP. The absence of apical polarity, necessary for cancer onset, in the non-neoplastic epithelium is also associated with the lowest MP under active Cl- transport. We propose that the loss of apical polarity in the breast epithelium destabilizes cellular homeostasis in part by lowering the MP.


Assuntos
Glândulas Mamárias Humanas , Humanos , Potenciais da Membrana , Epitélio/metabolismo , Mama , Comunicação Celular/fisiologia , Polaridade Celular/fisiologia , Células Epiteliais , Membro 2 da Família 12 de Carreador de Soluto/metabolismo
7.
J Cell Sci ; 137(5)2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-37888135

RESUMO

Polarised epithelial cell divisions represent a fundamental mechanism for tissue maintenance and morphogenesis. Morphological and mechanical changes in the plasma membrane influence the organisation and crosstalk of microtubules and actin at the cell cortex, thereby regulating the mitotic spindle machinery and chromosome segregation. Yet, the precise mechanisms linking plasma membrane remodelling to cell polarity and cortical cytoskeleton dynamics to ensure accurate execution of mitosis in mammalian epithelial cells remain poorly understood. Here, we manipulated the density of mammary epithelial cells in culture, which led to several mitotic defects. Perturbation of cell-cell adhesion formation impairs the dynamics of the plasma membrane, affecting the shape and size of mitotic cells and resulting in defects in mitotic progression and the generation of daughter cells with aberrant architecture. In these conditions, F- actin-astral microtubule crosstalk is impaired, leading to mitotic spindle misassembly and misorientation, which in turn contributes to chromosome mis-segregation. Mechanistically, we identify S100 Ca2+-binding protein A11 (S100A11) as a key membrane-associated regulator that forms a complex with E-cadherin (CDH1) and the leucine-glycine-asparagine repeat protein LGN (also known as GPSM2) to coordinate plasma membrane remodelling with E-cadherin-mediated cell adhesion and LGN-dependent mitotic spindle machinery. Thus, plasma membrane-mediated maintenance of mammalian epithelial cell identity is crucial for correct execution of polarised cell divisions, genome maintenance and safeguarding tissue integrity.


Assuntos
Actinas , Polaridade Celular , Animais , Adesão Celular , Actinas/metabolismo , Polaridade Celular/fisiologia , Mitose , Microtúbulos/metabolismo , Fuso Acromático/metabolismo , Membrana Celular/metabolismo , Caderinas/genética , Caderinas/metabolismo , Mamíferos/metabolismo
8.
EMBO Rep ; 24(12): e56997, 2023 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-37975164

RESUMO

Planar cell polarity (PCP) signaling polarizes epithelial cells within the plane of an epithelium. Core PCP signaling components adopt asymmetric subcellular localizations within cells to both polarize and coordinate polarity between cells. Achieving subcellular asymmetry requires additional effectors, including some mediating post-translational modifications of core components. Identification of such proteins is challenging due to pleiotropy. We used mass spectrometry-based proximity labeling proteomics to identify such regulators in the Drosophila wing. We identified the catalytic subunit of protein phosphatase1, Pp1-87B, and show that it regulates core protein polarization. Pp1-87B interacts with the core protein Van Gogh and at least one serine/threonine kinase, Dco/CKIε, that is known to regulate PCP. Pp1-87B modulates Van Gogh subcellular localization and directs its dephosphorylation in vivo. PNUTS, a Pp1 regulatory subunit, also modulates PCP. While the direct substrate(s) of Pp1-87B in control of PCP is not known, our data support the model that cycling between phosphorylated and unphosphorylated forms of one or more core PCP components may regulate acquisition of asymmetry. Finally, our screen serves as a resource for identifying additional regulators of PCP signaling.


Assuntos
Proteínas de Drosophila , Proteínas de Membrana , Animais , Polaridade Celular/fisiologia , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Proteínas de Membrana/metabolismo , Proteína Fosfatase 1/genética , Proteína Fosfatase 1/metabolismo , Processamento de Proteína Pós-Traducional , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais
9.
EMBO J ; 42(24): e113856, 2023 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-37953688

RESUMO

Apical-basal polarity is maintained by distinct protein complexes that reside in membrane junctions, and polarity loss in monolayered epithelial cells can lead to formation of multilayers, cell extrusion, and/or malignant overgrowth. Yet, how polarity loss cooperates with intrinsic signals to control directional invasion toward neighboring epithelial cells remains elusive. Using the Drosophila ovarian follicular epithelium as a model, we found that posterior follicle cells with loss of lethal giant larvae (lgl) or Discs large (Dlg) accumulate apically toward germline cells, whereas cells with loss of Bazooka (Baz) or atypical protein kinase C (aPKC) expand toward the basal side of wildtype neighbors. Further studies revealed that these distinct multilayering patterns in the follicular epithelium were determined by epidermal growth factor receptor (EGFR) signaling and its downstream target Pointed, a zinc-finger transcription factor. Additionally, we identified Rho kinase as a Pointed target that regulates formation of distinct multilayering patterns. These findings provide insight into how cell polarity genes and receptor tyrosine kinase signaling interact to govern epithelial cell organization and directional growth that contribute to epithelial tumor formation.


Assuntos
Polaridade Celular , Proteínas de Drosophila , Receptores ErbB , Animais , Polaridade Celular/fisiologia , Drosophila melanogaster , Proteínas de Drosophila/metabolismo , Células Epiteliais/metabolismo , Epitélio/metabolismo , Receptores ErbB/genética , Receptores ErbB/metabolismo
10.
PLoS Comput Biol ; 19(10): e1011523, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37782676

RESUMO

Many cells adjust the direction of polarized growth or migration in response to external directional cues. The yeast Saccharomyces cerevisiae orient their cell fronts (also called polarity sites) up pheromone gradients in the course of mating. However, the initial polarity site is often not oriented towards the eventual mating partner, and cells relocate the polarity site in an indecisive manner before developing a stable orientation. During this reorientation phase, the polarity site displays erratic assembly-disassembly behavior and moves around the cell cortex. The mechanisms underlying this dynamic behavior remain poorly understood. Particle-based simulations of the core polarity circuit revealed that molecular-level fluctuations are unlikely to overcome the strong positive feedback required for polarization and generate relocating polarity sites. Surprisingly, inclusion of a second pathway that promotes polarity site orientation generated relocating polarity sites with properties similar to those observed experimentally. This pathway forms a second positive feedback loop involving the recruitment of receptors to the cell membrane and couples polarity establishment to gradient sensing. This second positive feedback loop also allows cells to stabilize their polarity site once the site is aligned with the pheromone gradient.


Assuntos
Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae , Saccharomyces cerevisiae/metabolismo , Retroalimentação , Proteínas de Saccharomyces cerevisiae/metabolismo , Feromônios/metabolismo , Comunicação Celular , Polaridade Celular/fisiologia
11.
Curr Biol ; 33(20): 4312-4329.e6, 2023 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-37729910

RESUMO

During embryonic development, oriented cell divisions are important for patterned tissue growth and cell fate specification. Cell division orientation is controlled in part by asymmetrically localized polarity proteins, which establish functional domains of the cell membrane and interact with microtubule regulators to position the mitotic spindle. For example, in the 8-cell mouse embryo, apical polarity proteins form caps on the outside, contact-free surface of the embryo that position the mitotic spindle to execute asymmetric cell division. A similar radial or "inside-outside" polarity is established at an early stage in many other animal embryos, but in most cases, it remains unclear how inside-outside polarity is established and how it influences downstream cell behaviors. Here, we explore inside-outside polarity in C. elegans somatic blastomeres using spatiotemporally controlled protein degradation and live embryo imaging. We show that PAR polarity proteins, which form apical caps at the center of the contact-free membrane, localize dynamically during the cell cycle and contribute to spindle orientation and proper cell positioning. Surprisingly, isolated single blastomeres lacking cell contacts are able to break symmetry and form PAR-3/atypical protein kinase C (aPKC) caps. Polarity caps form independently of actomyosin flows and microtubules and can regulate spindle orientation in cooperation with the key polarity kinase aPKC. Together, our results reveal a role for apical polarity caps in regulating spindle orientation in symmetrically dividing cells and provide novel insights into how these structures are formed.


Assuntos
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Animais , Camundongos , Caenorhabditis elegans/fisiologia , Divisão Celular , Fuso Acromático/metabolismo , Ciclo Celular , Divisão Celular Assimétrica , Polaridade Celular/fisiologia , Proteínas de Caenorhabditis elegans/metabolismo
12.
Mol Biol Cell ; 34(12): ar116, 2023 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-37647145

RESUMO

The polarized distribution of membrane proteins into apical and basolateral domains provides the basis for specialized functions of epithelial tissues. The EGF receptor (EGFR) plays important roles in embryonic development, adult-epithelial tissue homeostasis, and growth and survival of many carcinomas. Typically targeted to basolateral domains, there is also considerable evidence of EGFR sorting plasticity but very limited knowledge regarding domain-specific EGFR substrates. Here we have investigated effects of selective EGFR mistargeting because of inactive-basolateral sorting signals on epithelial-cell homeostatic responses to growth-induced stress in MDCK cell models. Aberrant EGFR localization was associated with multilayer formation, anchorage-independent growth, and upregulated expression of the intermediate filament-protein vimentin characteristically seen in cells undergoing epithelial-to-mesenchymal transition. EGFRs were selectively retained following their internalization from apical membranes, and a signaling pathway involving the signaling adaptor Gab1 protein and extracellular signal-regulated kinase ERK5 had an essential role integrating multiple responses to growth-induced stress. Our studies highlight the potential importance of cellular machinery specifying EGFR polarity in epithelial pathologies associated with homeostatic imbalance.


Assuntos
Células Epiteliais , Receptores ErbB , Animais , Cães , Receptores ErbB/metabolismo , Células Epiteliais/metabolismo , Membrana Celular/metabolismo , Células Madin Darby de Rim Canino , Proteínas de Membrana/metabolismo , Homeostase , Polaridade Celular/fisiologia
13.
Nat Commun ; 14(1): 4056, 2023 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-37422455

RESUMO

During cell migration, front-rear polarity is spatiotemporally regulated; however, the underlying design of regulatory interactions varies. In rod-shaped Myxococcus xanthus cells, a spatial toggle switch dynamically regulates front-rear polarity. The polarity module establishes front-rear polarity by guaranteeing front pole-localization of the small GTPase MglA. Conversely, the Frz chemosensory system, by acting on the polarity module, causes polarity inversions. MglA localization depends on the RomR/RomX GEF and MglB/RomY GAP complexes that localize asymmetrically to the poles by unknown mechanisms. Here, we show that RomR and the MglB and MglC roadblock domain proteins generate a positive feedback by forming a RomR/MglC/MglB complex, thereby establishing the rear pole with high GAP activity that is non-permissive to MglA. MglA at the front engages in negative feedback that breaks the RomR/MglC/MglB positive feedback allosterically, thus ensuring low GAP activity at this pole. These findings unravel the design principles of a system for switchable front-rear polarity.


Assuntos
Proteínas Monoméricas de Ligação ao GTP , Myxococcus xanthus , Myxococcus xanthus/metabolismo , Proteínas de Bactérias/metabolismo , Movimento Celular/fisiologia , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Polaridade Celular/fisiologia
14.
Dev Cell ; 58(17): 1519-1533.e6, 2023 09 11.
Artigo em Inglês | MEDLINE | ID: mdl-37419117

RESUMO

Planar spindle orientation is critical for epithelial tissue organization and is generally instructed by the long cell-shape axis or cortical polarity domains. We introduced mouse intestinal organoids in order to study spindle orientation in a monolayered mammalian epithelium. Although spindles were planar, mitotic cells remained elongated along the apico-basal (A-B) axis, and polarity complexes were segregated to basal poles, so that spindles oriented in an unconventional manner, orthogonal to both polarity and geometric cues. Using high-resolution 3D imaging, simulations, and cell-shape and cytoskeleton manipulations, we show that planar divisions resulted from a length limitation in astral microtubules (MTs) which precludes them from interacting with basal polarity, and orient spindles from the local geometry of apical domains. Accordingly, lengthening MTs affected spindle planarity, cell positioning, and crypt arrangement. We conclude that MT length regulation may serve as a key mechanism for spindles to sense local cell shapes and tissue forces to preserve mammalian epithelial architecture.


Assuntos
Microtúbulos , Fuso Acromático , Animais , Camundongos , Fuso Acromático/fisiologia , Divisão Celular , Microtúbulos/fisiologia , Epitélio , Polaridade Celular/fisiologia , Mamíferos
15.
Curr Opin Plant Biol ; 74: 102383, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37285693

RESUMO

Land plants depend on oriented cell divisions that specify cell identities and tissue architecture. As such, the initiation and subsequent growth of plant organs require pathways that integrate diverse systemic signals to inform division orientation. Cell polarity is one solution to this challenge, allowing cells to generate internal asymmetry both spontaneously and in response to extrinsic cues. Here, we provide an update on our understanding of how plasma membrane-associated polarity domains control division orientation in plant cells. These cortical polar domains are flexible protein platforms whose positions, dynamics, and recruited effectors can be modulated by varied signals to control cellular behavior. Several recent reviews have explored the formation and maintenance of polar domains during plant development [1-4], so we focus here on substantial advances in our understanding of polarity-mediated division orientation from the last five years to provide a current snapshot of the field and highlight areas for future exploration.


Assuntos
Desenvolvimento Vegetal , Plantas , Divisão Celular , Polaridade Celular/fisiologia
16.
Development ; 150(14)2023 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-37390294

RESUMO

Caudal developmental defects, including caudal regression, caudal dysgenesis and sirenomelia, are devastating conditions affecting the skeletal, nervous, digestive, reproductive and excretory systems. Defects in mesodermal migration and blood supply to the caudal region have been identified as possible causes of caudal developmental defects, but neither satisfactorily explains the structural malformations in all three germ layers. Here, we describe caudal developmental defects in transmembrane protein 132a (Tmem132a) mutant mice, including skeletal, posterior neural tube closure, genitourinary tract and hindgut defects. We show that, in Tmem132a mutant embryos, visceral endoderm fails to be excluded from the medial region of early hindgut, leading directly to the loss or malformation of cloaca-derived genitourinary and gastrointestinal structures, and indirectly to the neural tube and kidney/ureter defects. We find that TMEM132A mediates intercellular interaction, and physically interacts with planar cell polarity (PCP) regulators CELSR1 and FZD6. Genetically, Tmem132a regulates neural tube closure synergistically with another PCP regulator Vangl2. In summary, we have identified Tmem132a as a new regulator of PCP, and hindgut malformation as the underlying cause of developmental defects in multiple caudal structures.


Assuntos
Defeitos do Tubo Neural , Camundongos , Animais , Defeitos do Tubo Neural/metabolismo , Tubo Neural/metabolismo , Neurulação , Camadas Germinativas/metabolismo , Polaridade Celular/fisiologia , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo
17.
Reproduction ; 166(2): 175-185, 2023 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-37318097

RESUMO

In brief: During the morula to blastocyst transformation, polarity establishment in outer cells is a prerequisite for trophectoderm lineage specification. This study reveals the roles of polarity proteins PATJ and MPDZ in trophectoderm lineage fate decision. Abstract: In mouse preimplantation embryos, cell polarity plays a crucial role in the first lineage specification. PATJ and its homolog MPDZ are the main members of CRB-PALS1-PATJ (CRUMBS-Protein associated with Lin7 1-Pals-associated tight junction protein) apical polarity complex. They act as adaptor proteins connecting CRB-PALS1 and tight junction proteins, making them essential for cell polarization and stabilization of apical junctions. However, their roles in regulating trophectoderm differentiation and blastocyst development remain unclear. In this study, PATJ and/or MPDZ were downregulated by the microinjection of specific RNA interference constructs into zygotes. Downregulation of PATJ alone did not severely affect early embryonic development and trophectoderm lineage differentiation although it slowed down the blastocyst formation. Depletion of PATJ and MPDZ did not affect compaction and morula development but impaired blastocyst formation. Furthermore, the expression of trophectoderm-specific transcription factors and trophoblast differentiation was compromised in the absence of PATJ/MPDZ. These abnormalities might result from the breakdown of apical domain in the outer cells of the embryo. The loss of PATJ/MPDZ caused the breakdown of CRB and PAR polarity complexes as well as deficiencies in tight junctions and actin filaments. These defects led to ectopic activation of Hippo signaling in the outer cells of developing embryos, ultimately suppressing Cdx2 expression and trophectoderm differentiation. Altogether, PATJ and MPDZ are essential for trophectoderm lineage differentiation and normal blastocyst morphogenesis via the regulation of the establishment of apical domain, formation of tight junctions, phosphorylation and localization of YAP, and expression of trophectoderm-specific transcription factors.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Fatores de Transcrição , Animais , Camundongos , Proteínas Adaptadoras de Transdução de Sinal/genética , Blastocisto , Diferenciação Celular , Linhagem da Célula , Polaridade Celular/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Junções Íntimas/metabolismo , Fatores de Transcrição/metabolismo
18.
Cell Rep ; 42(7): 112677, 2023 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-37352102

RESUMO

Polarity proteins regulate the proliferation and differentiation of neural progenitors to generate neurons during brain development through multiple signaling pathways. However, how cell polarity couples the signaling pathways remains unclear. Here, we show that coiled-coil domain-containing protein 85c (Ccdc85c) interacts with the polarity protein Par3 to regulate the proliferation of radial glial cells (RGCs) via phase separation coupled to percolation (PSCP). We find that the interaction with Ccdc85c relieves the intramolecular auto-inhibition of Par3, which leads to PSCP of Par3. Downregulation of Ccdc85c causes RGC differentiation. Importantly, the open conformation of Par3 facilitates the recruitment of the Notch regulator Numb to the Par3 condensates, which might prevent the attenuation of Notch activity to maintain RGC proliferation. Furthermore, ectopic activation of Notch signaling rescues RGC proliferation defects caused by the downregulation of Ccdc85c. These results suggest that Ccdc85c-mediated PSCP of Par3 regulates Notch signaling to control RGC proliferation during brain development.


Assuntos
Polaridade Celular , Transdução de Sinais , Polaridade Celular/fisiologia , Transdução de Sinais/fisiologia , Neurônios/metabolismo , Proliferação de Células , Receptores Notch/metabolismo
19.
Elife ; 122023 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-37144879

RESUMO

The vestibular maculae of the inner ear contain sensory receptor hair cells that detect linear acceleration and contribute to equilibrioception to coordinate posture and ambulatory movements. These hair cells are divided between two groups, separated by a line of polarity reversal (LPR), with oppositely oriented planar-polarized stereociliary bundles that detect motion in opposite directions. The transcription factor EMX2 is known to establish this planar polarized organization in mouse by regulating the distribution of the transmembrane receptor GPR156 at hair cell boundaries in one group of cells. However, the genes regulated by EMX2 in this context were previously not known. Using mouse as a model, we have identified the serine threonine kinase STK32A as a downstream effector negatively regulated by EMX2. Stk32a is expressed in hair cells on one side of the LPR in a pattern complementary to Emx2 expression in hair cells on the opposite side. Stk32a is necessary to align the intrinsic polarity of the bundle with the core planar cell polarity (PCP) proteins in EMX2-negative regions, and is sufficient to reorient bundles when ectopically expressed in neighboring EMX2-positive regions. We demonstrate that STK32A reinforces LPR formation by regulating the apical localization of GPR156. These observations support a model in which bundle orientation is determined through separate mechanisms in hair cells on opposite sides of the maculae, with EMX2-mediated repression of Stk32a determining the final position of the LPR.


Assuntos
Polaridade Celular , Vestíbulo do Labirinto , Animais , Camundongos , Polaridade Celular/fisiologia , Células Ciliadas Auditivas/metabolismo , Células Receptoras Sensoriais/metabolismo , Fatores de Transcrição/metabolismo , Vestíbulo do Labirinto/metabolismo
20.
PLoS One ; 18(5): e0285217, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37155619

RESUMO

Apical-basal cell polarity must be tightly controlled for epithelial cyst and tubule formation, and these are important functional units in various epithelial organs. Polarization is achieved through the coordination of several molecules that divide cells into an apical domain and a basolateral domain, which are separated from tight and adherens junctions. Cdc42 regulates cytoskeletal organization and the tight junction protein ZO-1 at the apical margin of epithelial cell junctions. MST kinases control organ size through the regulation of cell proliferation and cell polarity. For example, MST1 relays the Rap1 signal to induce cell polarity and adhesion of lymphocytes. Our previous study showed that MST3 was involved in E-cadherin regulation and migration in MCF7 cells. In vivo, MST3 knockout mice exhibited higher ENaC expression at the apical site of renal tubules, resulting in hypertension. However, it was not clear whether MST3 was involved in cell polarity. Here, control MDCK cells, HA-MST3 and HA-MST3 kinase-dead (HA-MST3-KD) overexpressing MDCK cells were cultured in collagen or Matrigel. We found that the cysts of HA-MST3 cells were fewer and smaller than those of control MDCK cells; ZO-1 was delayed to the apical site of cysts and in cell-cell contact in the Ca2+ switch assay. However, HA-MST3-KD cells exhibited multilumen cysts. Intensive F-actin stress fibers were observed in HA-MST3 cells with higher Cdc42 activity; in contrast, HA-MST3-KD cells had lower Cdc42 activity and weaker F-actin staining. In this study, we identified a new MST3 function in the establishment of cell polarity through Cdc42 regulation.


Assuntos
Cistos , Células Epiteliais , Animais , Camundongos , Actinas/metabolismo , Polaridade Celular/fisiologia , Cistos/metabolismo , Células Epiteliais/metabolismo , Junções Intercelulares/metabolismo , Transdução de Sinais , Junções Íntimas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...