301
|
Baranwal S, Naydenov NG, Harris G, Dugina V, Morgan KG, Chaponnier C, Ivanov AI. Nonredundant roles of cytoplasmic β- and γ-actin isoforms in regulation of epithelial apical junctions. Mol Biol Cell 2012; 23:3542-53. [PMID: 22855531 PMCID: PMC3442403 DOI: 10.1091/mbc.e12-02-0162] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The functional effects of cytoplasmic actins on epithelial junctions are examined by using isoform-specific siRNAs and cell-permeable inhibitory peptides. Unique roles of cytoplasmic actin isoforms in regulating structure and remodeling of adherens and tight junctions are revealed. Association with the actin cytoskeleton is critical for normal architecture and dynamics of epithelial tight junctions (TJs) and adherens junctions (AJs). Epithelial cells express β-cytoplasmic (β-CYA) and γ-cytoplasmic (γ-CYA) actins, which have different cellular localization and functions. This study elucidates the roles of cytoplasmic actins in regulating structure and remodeling of AJs and TJs in model intestinal epithelia. Immunofluorescence labeling and latrunculin B treatment reveal affiliation of dynamic β-CYA filaments with newly assembled and mature AJs, whereas an apical γ-CYA pool is composed of stable perijunctional bundles and rapidly turning-over nonjunctional filaments. The functional effects of cytoplasmic actins on epithelial junctions are examined by using isoform-specific small interfering RNAs and cell-permeable inhibitory peptides. These experiments demonstrate unique roles of β-CYA and γ-CYA in regulating the steady-state integrity of AJs and TJs, respectively. Furthermore, β-CYA is selectively involved in establishment of apicobasal cell polarity. Both actin isoforms are essential for normal barrier function of epithelial monolayers, rapid AJ/TJ reassembly, and formation of three-dimensional cysts. Cytoplasmic actin isoforms play unique roles in regulating structure and permeability of epithelial junctions.
Collapse
Affiliation(s)
- Somesh Baranwal
- Department of Medicine, University of Rochester, Rochester, NY 14642, USA
| | | | | | | | | | | | | |
Collapse
|
302
|
Yu CG, Harris TJC. Interactions between the PDZ domains of Bazooka (Par-3) and phosphatidic acid: in vitro characterization and role in epithelial development. Mol Biol Cell 2012; 23:3743-53. [PMID: 22833561 PMCID: PMC3442420 DOI: 10.1091/mbc.e12-03-0196] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
The polarity regulator Bazooka (Par-3) is shown to directly bind the signaling lipid phosphatidic acid via its PDZ domains in vitro. In vivo, the interaction appears to support Bazooka functions important for Drosophila embryonic epithelial structure. Thus Bazooka has a closer connection to membrane lipids than previously recognized. Bazooka (Par-3) is a conserved polarity regulator that organizes molecular networks in a wide range of cell types. In epithelia, it functions as a plasma membrane landmark to organize the apical domain. Bazooka is a scaffold protein that interacts with proteins through its three PDZ (postsynaptic density 95, discs large, zonula occludens-1) domains and other regions. In addition, Bazooka has been shown to interact with phosphoinositides. Here we show that the Bazooka PDZ domains interact with the negatively charged phospholipid phosphatidic acid immobilized on solid substrates or in liposomes. The interaction requires multiple PDZ domains, and conserved patches of positively charged amino acid residues appear to mediate the interaction. Increasing or decreasing levels of diacylglycerol kinase or phospholipase D—enzymes that produce phosphatidic acid—reveal a role for phosphatidic acid in Bazooka embryonic epithelial activity but not its localization. Mutating residues implicated in phosphatidic acid binding revealed a possible role in Bazooka localization and function. These data implicate a closer connection between Bazooka and membrane lipids than previously recognized. Bazooka polarity landmarks may be conglomerates of proteins and plasma membrane lipids that modify each other's activities for an integrated effect on cell polarity.
Collapse
Affiliation(s)
- Cao Guo Yu
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | | |
Collapse
|
303
|
Harding MJ, Nechiporuk AV. Fgfr-Ras-MAPK signaling is required for apical constriction via apical positioning of Rho-associated kinase during mechanosensory organ formation. Development 2012; 139:3130-5. [PMID: 22833124 DOI: 10.1242/dev.082271] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Many morphogenetic movements during development require the formation of transient intermediates called rosettes. Within rosettes, cells are polarized with apical ends constricted towards the rosette center and nuclei basally displaced. Whereas the polarity and cytoskeletal machinery establishing these structures has been extensively studied, the extracellular cues and intracellular signaling cascades that promote their formation are not well understood. We examined how extracellular Fibroblast growth factor (Fgf) signals regulate rosette formation in the zebrafish posterior lateral line primordium (pLLp), a group of ∼100 cells that migrates along the trunk during embryonic development to form the lateral line mechanosensory system. During migration, the pLLp deposits rosettes from the trailing edge, while cells are polarized and incorporated into nascent rosettes in the leading region. Fgf signaling was previously shown to be crucial for rosette formation in the pLLp. We demonstrate that activation of Fgf receptor (Fgfr) induces intracellular Ras-MAPK, which is required for apical constriction and rosette formation in the pLLp. Inhibiting Fgfr-Ras-MAPK leads to loss of apically localized Rho-associated kinase (Rock) 2a, which results in failed actomyosin cytoskeleton activation. Using mosaic analyses, we show that a cell-autonomous Ras-MAPK signal is required for apical constriction and Rock2a localization. We propose a model whereby activated Fgfr signals through Ras-MAPK to induce apical localization of Rock2a in a cell-autonomous manner, activating the actomyosin network to promote apical constriction and rosette formation in the pLLp. This mechanism presents a novel cellular strategy for driving cell shape changes.
Collapse
Affiliation(s)
- Molly J Harding
- Department of Cell and Developmental Biology, Oregon Health & Science University, Portland, OR 97239, USA
| | | |
Collapse
|
304
|
Abstract
From the very early days of nuclear factor-κB (NF-κB) research, it was recognized that different protein kinase C (PKC) isoforms might be involved in the activation of NF-κB. Pharmacological tools and pseudosubstrate inhibitors suggested that these kinases play a role in this important inflammatory and survival pathway; however, it was the analysis of several genetic mouse knockout models that revealed the complexity and interrelations between the different components of the PB1 network in several cellular functions, including T-cell biology, bone homeostasis, inflammation associated with the metabolic syndrome, and cancer. These studies unveiled, for example, the critical role of PKCζ as a positive regulator of NF-κB through the regulation of RelA but also its inflammatory suppressor activities through the regulation of the interleukin-4 signaling cascade. This observation is of relevance in T cells, where p62, PKCζ, PKCλ/ι, and NBR1 establish a mesh of interactions that culminate in the regulation of T-cell effector responses through the modulation of T-cell polarity. Many questions remain to be answered, not just from the point of view of the implication for NF-κB activation but also with regard to the in vivo interplay between these pathways in pathophysiological processes like obesity and cancer.
Collapse
|
305
|
Cibrián Uhalte E, Kirchner M, Hellwig N, Allen JJ, Donat S, Shokat KM, Selbach M, Abdelilah-Seyfried S. In vivo conditions to identify Prkci phosphorylation targets using the analog-sensitive kinase method in zebrafish. PLoS One 2012; 7:e40000. [PMID: 22768194 PMCID: PMC3386912 DOI: 10.1371/journal.pone.0040000] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Accepted: 06/04/2012] [Indexed: 12/16/2022] Open
Abstract
Protein kinase C iota is required for various cell biological processes including epithelial tissue polarity and organ morphogenesis. To gain mechanistic insight into different roles of this kinase, it is essential to identify specific substrate proteins in their cellular context. The analog-sensitive kinase method provides a powerful tool for the identification of kinase substrates under in vivo conditions. However, it has remained a major challenge to establish screens based on this method in multicellular model organisms. Here, we report the methodology for in vivo conditions using the analog-sensitive kinase method in a genetically-tractable vertebrate model organism, the zebrafish. With this approach, kinase substrates can uniquely be labeled in the developing zebrafish embryo using bulky ATPγS analogs which results in the thiophosphorylation of substrates. The labeling of kinase substrates with a thiophosphoester epitope differs from phosphoesters that are generated by all other kinases and allows for an enrichment of thiophosphopeptides by immunoaffinity purification. This study provides the foundation for using the analog-sensitive kinase method in the context of complex vertebrate development, physiology, or disease.
Collapse
Affiliation(s)
| | | | - Nicole Hellwig
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Jasmina J. Allen
- Howard Hughes Medical Institute, University of California San Francisco, San Francisco, California, United States of America
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, California, United States of America
| | - Stefan Donat
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Kevan M. Shokat
- Howard Hughes Medical Institute, University of California San Francisco, San Francisco, California, United States of America
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, California, United States of America
| | | | | |
Collapse
|
306
|
The BEAF insulator regulates genes involved in cell polarity and neoplastic growth. Dev Biol 2012; 369:124-32. [PMID: 22743648 DOI: 10.1016/j.ydbio.2012.06.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2012] [Revised: 06/01/2012] [Accepted: 06/20/2012] [Indexed: 01/05/2023]
Abstract
Boundary Element Associated Factor-32 (BEAF-32) is an insulator protein predominantly found near gene promoters and thought to play a role in gene expression. We find that mutations in BEAF-32 are lethal, show loss of epithelial morphology in imaginal discs and cause neoplastic growth defects. To investigate the molecular mechanisms underlying this phenotype, we carried out a genome-wide analysis of BEAF-32 localization in wing imaginal disc cells. Mutation of BEAF-32 results in miss-regulation of 3850 genes by at least 1.5-fold, 794 of which are bound by this protein in wing imaginal cells. Up-regulated genes encode proteins involved in cell polarity, cell proliferation and cell differentiation. Among the down-regulated genes are those encoding components of the wingless pathway, which is required for cell differentiation. Miss-regulation of these genes explains the unregulated cell growth and neoplastic phenotypes observed in imaginal tissues of BEAF-32 mutants.
Collapse
|
307
|
Chang JT. Polarity and lymphocyte fate determination. Curr Opin Cell Biol 2012; 24:526-33. [PMID: 22658837 DOI: 10.1016/j.ceb.2012.05.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Accepted: 05/14/2012] [Indexed: 01/02/2023]
Abstract
Polarity within lymphocytes has been recognized to regulate a variety of processes, including migration, signaling, and the execution of effector function. It has been recently proposed, however, that this polarized behavior may also serve a different purpose in lymphocytes that have not yet encountered their foreign antigen-to coordinate asymmetric cell division. Asymmetric division is an evolutionarily conserved mechanism allowing a single cell to give rise to two distinct daughter cells from inception. In this review, recent findings in polarity and asymmetric division in lymphocytes are discussed.
Collapse
Affiliation(s)
- John T Chang
- Department of Medicine, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
308
|
Shafaq-Zadah M, Brocard L, Solari F, Michaux G. AP-1 is required for the maintenance of apico-basal polarity in the C. elegans intestine. Development 2012; 139:2061-70. [DOI: 10.1242/dev.076711] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Epithelial tubes perform functions that are essential for the survival of multicellular organisms. Understanding how their polarised features are maintained is therefore crucial. By analysing the function of the clathrin adaptor AP-1 in the C. elegans intestine, we found that AP-1 is required for epithelial polarity maintenance. Depletion of AP-1 subunits does not affect epithelial polarity establishment or the formation of the intestinal lumen. However, the loss of AP-1 affects the polarised distribution of both apical and basolateral transmembrane proteins. Moreover, it triggers de novo formation of ectopic apical lumens between intestinal cells along the lateral membranes later during embryogenesis. We also found that AP-1 is specifically required for the apical localisation of the small GTPase CDC-42 and the polarity determinant PAR-6. Our results demonstrate that AP-1 controls an apical trafficking pathway required for the maintenance of epithelial polarity in vivo in a tubular epithelium.
Collapse
Affiliation(s)
- Massiullah Shafaq-Zadah
- INSERM Avenir group, F-35043 Rennes, France
- CNRS, UMR6061, Institut de Génétique et Développement de Rennes, F-35043 Rennes, France
- Université de Rennes 1, UEB, IFR140, Faculté de Médecine, F-35043 Rennes, France
| | - Lysiane Brocard
- INSERM Avenir group, F-35043 Rennes, France
- CNRS, UMR6061, Institut de Génétique et Développement de Rennes, F-35043 Rennes, France
- Université de Rennes 1, UEB, IFR140, Faculté de Médecine, F-35043 Rennes, France
| | - Florence Solari
- CNRS UMR5534, Centre de Génétique et de Physiologie Moléculaires et Cellulaires, Université Claude Bernard Lyon 1 F-69622 Villeurbanne, France
| | - Grégoire Michaux
- INSERM Avenir group, F-35043 Rennes, France
- CNRS, UMR6061, Institut de Génétique et Développement de Rennes, F-35043 Rennes, France
- Université de Rennes 1, UEB, IFR140, Faculté de Médecine, F-35043 Rennes, France
| |
Collapse
|
309
|
The Dishevelled-associating protein Daple controls the non-canonical Wnt/Rac pathway and cell motility. Nat Commun 2012; 3:859. [PMID: 22643886 DOI: 10.1038/ncomms1861] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2012] [Accepted: 04/23/2012] [Indexed: 12/20/2022] Open
Abstract
Dishevelled is the common mediator of canonical and non-canonical Wnt signalling pathways, which are important for embryonic development, tissue maintenance and cancer progression. In the non-canonical Wnt signalling pathway, the Rho family of small GTPases acting downstream of Dishevelled has essential roles in cell migration. The mechanisms by which the non-canonical Wnt signalling pathway regulates Rac activation remain unknown. Here we show that Daple (Dishevelled-associating protein with a high frequency of leucine residues) regulates Wnt5a-mediated activation of Rac and formation of lamellipodia through interaction with Dishevelled. Daple increases the association of Dishevelled with an isoform of atypical protein kinase C, consequently promoting Rac activation. Accordingly, Daple deficiency impairs migration of fibroblasts and epithelial cells during wound healing in vivo. These findings indicate that Daple interacts with Dishevelled to direct the Dishevelled/protein kinase λ protein complex to activate Rac, which in turn mediates the non-canonical Wnt signalling pathway required for cell migration.
Collapse
|
310
|
Caenorhabditis elegans screen reveals role of PAR-5 in RAB-11-recycling endosome positioning and apicobasal cell polarity. Nat Cell Biol 2012; 14:666-76. [PMID: 22634595 DOI: 10.1038/ncb2508] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Accepted: 04/19/2012] [Indexed: 02/08/2023]
Abstract
Apically enriched Rab11-positive recycling endosomes (Rab11-REs) are important for establishing and maintaining epithelial polarity. Yet, little is known about the molecules controlling trafficking of Rab11-REs in an epithelium in vivo. Here, we report a genome-wide, image-based RNA interference screen for regulators of Rab11-RE positioning and transport of an apical membrane protein (PEPT-1) in C. elegans intestine. Among the 356 screen hits was the 14-3-3 and partitioning defective protein PAR-5, which we found to be specifically required for Rab11-RE positioning and apicobasal polarity maintenance. Depletion of PAR-5 induced abnormal clustering of Rab11-REs to ectopic sites at the basolateral cortex containing F-actin and other apical domain components. This phenotype required key regulators of F-actin dynamics and polarity, such as Rho GTPases (RHO-1 and the Rac1 orthologue CED-10) and apical PAR proteins. Our data suggest that PAR-5 acts as a regulatory hub for a polarity-maintaining network required for apicobasal asymmetry of F-actin and proper Rab11-RE positioning.
Collapse
|
311
|
Freeley M, O'Dowd F, Paul T, Kashanin D, Davies A, Kelleher D, Long A. L-plastin regulates polarization and migration in chemokine-stimulated human T lymphocytes. THE JOURNAL OF IMMUNOLOGY 2012; 188:6357-70. [PMID: 22581862 DOI: 10.4049/jimmunol.1103242] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Chemokines such as SDF-1α play a crucial role in orchestrating T lymphocyte polarity and migration via polymerization and reorganization of the F-actin cytoskeleton, but the role of actin-associated proteins in this process is not well characterized. In this study, we have investigated a role for L-plastin, a leukocyte-specific F-actin-bundling protein, in SDF-1α-stimulated human T lymphocyte polarization and migration. We found that L-plastin colocalized with F-actin at the leading edge of SDF-1α-stimulated T lymphocytes and was also phosphorylated at Ser(5), a site that when phosphorylated regulates the ability of L-plastin to bundle F-actin. L-plastin phosphorylation was sensitive to pharmacological inhibitors of protein kinase C (PKC), and several PKC isoforms colocalized with L-plastin at the leading edge of SDF-1α-stimulated lymphocytes. However, PKC ζ, an established regulator of cell polarity, was the only isoform that regulated L-plastin phosphorylation. Knockdown of L-plastin expression with small interfering RNAs demonstrated that this protein regulated the localization of F-actin at the leading edge of chemokine-stimulated cells and was also required for polarization, lamellipodia formation, and chemotaxis. Knockdown of L-plastin expression also impaired the Rac1 activation cycle and Akt phosphorylation in response to SDF-1α stimulation. Furthermore, L-plastin also regulated SDF-1α-mediated lymphocyte migration on the integrin ligand ICAM-1 by influencing velocity and persistence, but in a manner that was independent of LFA-1 integrin activation or adhesion. This study, therefore, demonstrates an important role for L-plastin and the signaling pathways that regulate its phosphorylation in response to chemokines and adds L-plastin to a growing list of proteins implicated in T lymphocyte polarity and migration.
Collapse
Affiliation(s)
- Michael Freeley
- Department of Clinical Medicine, Institute of Molecular Medicine, Trinity College Dublin, Dublin 2, Ireland.
| | | | | | | | | | | | | |
Collapse
|
312
|
Imbrie GA, Wu H, Seldin DC, Dominguez I. Asymmetric Localization of CK2α During Xenopus Oogenesis. ACTA ACUST UNITED AC 2012; Suppl 4:11328. [PMID: 25346867 PMCID: PMC4207361 DOI: 10.4172/2161-0436.s4-001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The establishment of the dorso-ventral axis is a fundamental process that occurs after fertilization. Dorsal axis specification in frogs starts immediately after fertilization, and depends upon activation of Wnt/β-catenin signaling. The protein kinase CK2α can modulate Wnt/β-catenin signaling and is necessary for dorsal axis specification in Xenopus laevis. Our previous experiments show that CK2α transcripts and protein are animally localized in embryos, overlapping the region where Wnt/β-catenin signaling is activated. Here we determined whether the animal localization of CK2α in the embryo is preceded by its localization in the oocyte. We found that CK2α transcripts were detected from stage I, their levels increased during oogenesis, and were animally localized as early as stage III. CK2α transcripts were translated during oogenesis and CK2α protein was localized to the animal hemisphere of stage VI oocytes. We cloned the CK2α 3’UTR and showed that the 2.8 kb CK2α transcript containing the 3’UTR was enriched during oogenesis. By injecting ectopic mRNAs, we demonstrated that both the coding and 3’UTR regions were necessary for proper CK2α transcript localization. This is the first report showing the involvement of coding and 3’UTR regions in animal transcript localization. Our findings demonstrate the pre-localization of CK2α transcript and thus, CK2α protein, in the oocyte. This may help restrict CK2α expression in preparation for dorsal axis specification.
Collapse
Affiliation(s)
- Gregory A Imbrie
- Hematology-Oncology Section, Department of Medicine, Boston University Medical School, 650 Albany Street, Boston, MA, USA
| | - Hao Wu
- Hematology-Oncology Section, Department of Medicine, Boston University Medical School, 650 Albany Street, Boston, MA, USA
| | - David C Seldin
- Hematology-Oncology Section, Department of Medicine, Boston University Medical School, 650 Albany Street, Boston, MA, USA
| | - Isabel Dominguez
- Hematology-Oncology Section, Department of Medicine, Boston University Medical School, 650 Albany Street, Boston, MA, USA
| |
Collapse
|
313
|
Ohara K, Enomoto A, Kato T, Hashimoto T, Isotani-Sakakibara M, Asai N, Ishida-Takagishi M, Weng L, Nakayama M, Watanabe T, Kato K, Kaibuchi K, Murakumo Y, Hirooka Y, Goto H, Takahashi M. Involvement of Girdin in the determination of cell polarity during cell migration. PLoS One 2012; 7:e36681. [PMID: 22574214 PMCID: PMC3344933 DOI: 10.1371/journal.pone.0036681] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2011] [Accepted: 04/05/2012] [Indexed: 02/05/2023] Open
Abstract
Cell migration is a critical cellular process that determines embryonic development and the progression of human diseases. Therefore, cell- or context-specific mechanisms by which multiple promigratory proteins differentially regulate cell migration must be analyzed in detail. Girdin (girders of actin filaments) (also termed GIV, Gα-interacting vesicle associated protein) is an actin-binding protein that regulates migration of various cells such as endothelial cells, smooth muscle cells, neuroblasts, and cancer cells. Here we show that Girdin regulates the establishment of cell polarity, the deregulation of which may result in the disruption of directional cell migration. We found that Girdin interacts with Par-3, a scaffolding protein that is a component of the Par protein complex that has an established role in determining cell polarity. RNA interference-mediated depletion of Girdin leads to impaired polarization of fibroblasts and mammary epithelial cells in a way similar to that observed in Par-3-depleted cells. Accordingly, the expression of Par-3 mutants unable to interact with Girdin abrogates cell polarization in fibroblasts. Further biochemical analysis suggests that Girdin is present in the Par protein complex that includes Par-3, Par-6, and atypical protein kinase C. Considering previous reports showing the role of Girdin in the directional migration of neuroblasts, network formation of endothelial cells, and cancer invasion, these data may provide a specific mechanism by which Girdin regulates cell movement in biological contexts that require directional cell movement.
Collapse
Affiliation(s)
- Kei Ohara
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Gastroenterology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Atsushi Enomoto
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Division of Molecular Pathology, Center for Neurological Disease and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
- * E-mail: (MT); (AE)
| | - Takuya Kato
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takahiko Hashimoto
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | - Naoya Asai
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Division of Molecular Pathology, Center for Neurological Disease and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Maki Ishida-Takagishi
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Liang Weng
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masanori Nakayama
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takashi Watanabe
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Katsuhiro Kato
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kozo Kaibuchi
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Core Research for Evolutionary Science and Technology (CREST), Japan Science and Technology Agency, Saitama, Japan
| | - Yoshiki Murakumo
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshiki Hirooka
- Department of Endoscopy, Nagoya University Hospital, Nagoya, Japan
| | - Hidemi Goto
- Department of Gastroenterology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Endoscopy, Nagoya University Hospital, Nagoya, Japan
| | - Masahide Takahashi
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Division of Molecular Pathology, Center for Neurological Disease and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
- * E-mail: (MT); (AE)
| |
Collapse
|
314
|
Graybill C, Wee B, Atwood SX, Prehoda KE. Partitioning-defective protein 6 (Par-6) activates atypical protein kinase C (aPKC) by pseudosubstrate displacement. J Biol Chem 2012; 287:21003-11. [PMID: 22544755 DOI: 10.1074/jbc.m112.360495] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Atypical protein kinase C (aPKC) controls cell polarity by modulating substrate cortical localization. Aberrant aPKC activity disrupts polarity, yet the mechanisms that control aPKC remain poorly understood. We used a reconstituted system with purified components and a cultured cell cortical displacement assay to investigate aPKC regulation. We find that aPKC is autoinhibited by two domains within its NH(2)-terminal regulatory half, a pseudosubstrate motif that occupies the kinase active site, and a C1 domain that assists in this process. The Par complex member Par-6, previously thought to inhibit aPKC, is a potent activator of aPKC in our assays. Par-6 and aPKC interact via PB1 domain heterodimerization, and this interaction activates aPKC by displacing the pseudosubstrate, although full activity requires the Par-6 CRIB-PDZ domains. We propose that, along with its previously described roles in controlling aPKC localization, Par-6 allosterically activates aPKC to allow for high spatial and temporal control of substrate phosphorylation and polarization.
Collapse
Affiliation(s)
- Chiharu Graybill
- Institute of Molecular Biology and Department of Chemistry, University of Oregon, Eugene, Oregon 97403, USA
| | | | | | | |
Collapse
|
315
|
Extensions of PSD-95/discs large/ZO-1 (PDZ) domains influence lipid binding and membrane targeting of syntenin-1. FEBS Lett 2012; 586:1445-51. [DOI: 10.1016/j.febslet.2012.04.024] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2012] [Revised: 04/10/2012] [Accepted: 04/12/2012] [Indexed: 11/21/2022]
|
316
|
Chatterjee S, Seifried L, Feigin ME, Gibbons DL, Scuoppo C, Lin W, Rizvi ZH, Lind E, Dissanayake D, Kurie J, Ohashi P, Muthuswamy SK. Dysregulation of cell polarity proteins synergize with oncogenes or the microenvironment to induce invasive behavior in epithelial cells. PLoS One 2012; 7:e34343. [PMID: 22529912 PMCID: PMC3329530 DOI: 10.1371/journal.pone.0034343] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2011] [Accepted: 02/26/2012] [Indexed: 11/18/2022] Open
Abstract
Changes in expression and localization of proteins that regulate cell and tissue polarity are frequently observed in carcinoma. However, the mechanisms by which changes in cell polarity proteins regulate carcinoma progression are not well understood. Here, we report that loss of polarity protein expression in epithelial cells primes them for cooperation with oncogenes or changes in tissue microenvironment to promote invasive behavior. Activation of ErbB2 in cells lacking the polarity regulators Scribble, Dlg1 or AF-6, induced invasive properties. This cooperation required the ability of ErbB2 to regulate the Par6/aPKC polarity complex. Inhibition of the ErbB2-Par6 pathway was sufficient to block ErbB2-induced invasion suggesting that two polarity hits may be needed for ErbB2 to promote invasion. Interestingly, in the absence of ErbB2 activation, either a combined loss of two polarity proteins, or exposure of cells lacking one polarity protein to cytokines IL-6 or TNFα induced invasive behavior in epithelial cells. We observed the invasive behavior only when cells were plated on a stiff matrix (Matrigel/Collagen-1) and not when plated on a soft matrix (Matrigel alone). Cells lacking two polarity proteins upregulated expression of EGFR and activated Akt. Inhibition of Akt activity blocked the invasive behavior identifying a mechanism by which loss of polarity promotes invasion of epithelial cells. Thus, we demonstrate that loss of polarity proteins confers phenotypic plasticity to epithelial cells such that they display normal behavior under normal culture conditions but display aggressive behavior in response to activation of oncogenes or exposure to cytokines.
Collapse
Affiliation(s)
- Samit Chatterjee
- Ontario Cancer Institute, Campbell Family Institute for Breast Cancer Research, University of Toronto, Toronto, Ontario, Canada
- Cold Spring Harbor Laboratory, Watson School of Biological Sciences, Cold Spring Harbor, New York, United States of America
| | - Laurie Seifried
- Ontario Cancer Institute, Campbell Family Institute for Breast Cancer Research, University of Toronto, Toronto, Ontario, Canada
| | - Michael E. Feigin
- Cold Spring Harbor Laboratory, Watson School of Biological Sciences, Cold Spring Harbor, New York, United States of America
| | - Don L. Gibbons
- Department of Thoracic/Head & Neck Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Claudio Scuoppo
- Cold Spring Harbor Laboratory, Watson School of Biological Sciences, Cold Spring Harbor, New York, United States of America
| | - Wei Lin
- Department of Thoracic/Head & Neck Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Zain H. Rizvi
- Department of Thoracic/Head & Neck Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Evan Lind
- Ontario Cancer Institute, Campbell Family Institute for Breast Cancer Research, University of Toronto, Toronto, Ontario, Canada
| | - Dilan Dissanayake
- Ontario Cancer Institute, Campbell Family Institute for Breast Cancer Research, University of Toronto, Toronto, Ontario, Canada
| | - Jonathan Kurie
- Department of Thoracic/Head & Neck Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Pam Ohashi
- Ontario Cancer Institute, Campbell Family Institute for Breast Cancer Research, University of Toronto, Toronto, Ontario, Canada
| | - Senthil K. Muthuswamy
- Ontario Cancer Institute, Campbell Family Institute for Breast Cancer Research, University of Toronto, Toronto, Ontario, Canada
- Cold Spring Harbor Laboratory, Watson School of Biological Sciences, Cold Spring Harbor, New York, United States of America
- * E-mail:
| |
Collapse
|
317
|
Seidl S, Braun UB, Leitges M. Functional comparison of protein domains within aPKCs involved in nucleocytoplasmic shuttling. Biol Open 2012; 1:436-45. [PMID: 23213435 PMCID: PMC3507206 DOI: 10.1242/bio.2012505] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The atypical protein kinases C (PKC) isoforms ι and ζ play crucial roles in regulation of signaling pathways related to proliferation, differentiation and cell survival. Over the years several interaction partners and phosphorylation targets have been identified. However, little is known about the regulation of atypical aPKC isoforms. To address this question, we performed a comparative analysis of atypical aPKCι/λ and ζ in MDCK cells. By using green fluorescence protein (GFP) fusion proteins containing the full-length or truncated proteins, we were able to recognize differences in subcellular localization and nucleocytoplasmic shuttling of both isoforms. We show, that an earlier described nuclear localization sequence (NLS), plays a role in the regulation of atypical aPKCζ but not in aPKCι, despite the fact that it is present in both isoforms. Leptomycin B treatment induces accumulation of GFP-fusion protein of both isoforms in the nucleus. Regardless, the loss of the NLS only decreases shuttling of aPKCζ, while aPKCι remains unaffected. In addition, we identified the hinge region as a potential regulator of localization of atypical PKCs. With a set of chimeric proteins we show that the hinge region of aPKCι mediates nuclear localization. In contrast, the hinge region of aPKCζ causes exclusion from the nucleus, indicating two different mechanisms leading to isoform specific regulation. Taken together, we show for the first time, that the atypical isoforms aPKCι and ζ underly different mechanisms regarding their regulation of subcellular localization and translocation into the nucleus in MDCK cells.
Collapse
Affiliation(s)
- Sebastian Seidl
- Biotechnology Centre of Oslo, University of Oslo , NO-0349, Oslo , Norway
| | | | | |
Collapse
|
318
|
Mashukova A, Forteza R, Wald FA, Salas PJ. PDK1 in apical signaling endosomes participates in the rescue of the polarity complex atypical PKC by intermediate filaments in intestinal epithelia. Mol Biol Cell 2012; 23:1664-74. [PMID: 22398726 PMCID: PMC3338434 DOI: 10.1091/mbc.e11-12-0988] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The polarity complex atypical PKC (aPKC) is rescued from degradation on intermediate filaments by Hsp70 chaperoning. The results indicate that PDK1 participates in the rescue mechanism and is localized to apical endosomes. Inhibition of dynamin-dependent endocytosis greatly decreases the steady-state levels of aPKC and Akt in their active conformation. Phosphorylation of the activation domain of protein kinase C (PKC) isoforms is essential to start a conformational change that results in an active catalytic domain. This activation is necessary not only for newly synthesized molecules, but also for kinase molecules that become dephosphorylated and need to be refolded and rephosphorylated. This “rescue” mechanism is responsible for the maintenance of the steady-state levels of atypical PKC (aPKC [PKCι/λ and ζ]) and is blocked in inflammation. Although there is consensus that phosphoinositide-dependent protein kinase 1 (PDK1) is the activating kinase for newly synthesized molecules, it is unclear what kinase performs that function during the rescue and where the rescue takes place. To identify the activating kinase during the rescue mechanism, we inhibited protein synthesis and analyzed the stability of the remaining aPKC pool. PDK1 knockdown and two different PDK1 inhibitors—BX-912 and a specific pseudosubstrate peptide—destabilized PKCι. PDK1 coimmunoprecipitated with PKCι in cells without protein synthesis, confirming that the interaction is direct. In addition, we showed that PDK1 aids the rescue of aPKC in in vitro rephosphorylation assays using immunodepletion and rescue with recombinant protein. Surprisingly, we found that in Caco-2 epithelial cells and intestinal crypt enterocytes PDK1 distributes to an apical membrane compartment comprising plasma membrane and apical endosomes, which, in turn, are in close contact with intermediate filaments. PDK1 comigrated with the Rab11 compartment and, to some extent, with the transferrin compartment in sucrose gradients. PDK1, pT555-aPKC, and pAkt were dependent on dynamin activity. These results highlight a novel signaling function of apical endosomes in polarized cells.
Collapse
Affiliation(s)
- Anastasia Mashukova
- Department of Cell Biology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | | | | | | |
Collapse
|
319
|
Iden S, Misselwitz S, Peddibhotla SSD, Tuncay H, Rehder D, Gerke V, Robenek H, Suzuki A, Ebnet K. aPKC phosphorylates JAM-A at Ser285 to promote cell contact maturation and tight junction formation. ACTA ACUST UNITED AC 2012; 196:623-39. [PMID: 22371556 PMCID: PMC3307692 DOI: 10.1083/jcb.201104143] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The PAR-3-atypical protein kinase C (aPKC)-PAR-6 complex has been implicated in the development of apicobasal polarity and the formation of tight junctions (TJs) in vertebrate epithelial cells. It is recruited by junctional adhesion molecule A (JAM-A) to primordial junctions where aPKC is activated by Rho family small guanosine triphosphatases. In this paper, we show that aPKC can interact directly with JAM-A in a PAR-3-independent manner. Upon recruitment to primordial junctions, aPKC phosphorylates JAM-A at S285 to promote the maturation of immature cell-cell contacts. In fully polarized cells, S285-phosphorylated JAM-A is localized exclusively at the TJs, and S285 phosphorylation of JAM-A is required for the development of a functional epithelial barrier. Protein phosphatase 2A dephosphorylates JAM-A at S285, suggesting that it antagonizes the activity of aPKC. Expression of nonphosphorylatable JAM-A/S285A interferes with single lumen specification during cyst development in three-dimensional culture. Our data suggest that aPKC phosphorylates JAM-A at S285 to regulate cell-cell contact maturation, TJ formation, and single lumen specification.
Collapse
Affiliation(s)
- Sandra Iden
- Institute-associated Research Group: Cell adhesion and cell polarity, University Münster, 48419 Münster, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
320
|
Aristizábal-Corrales D, Fontrodona L, Porta-de-la-Riva M, Guerra-Moreno A, Cerón J, Schwartz S. The 14-3-3 gene par-5 is required for germline development and DNA damage response in Caenorhabditis elegans. J Cell Sci 2012; 125:1716-26. [PMID: 22328524 DOI: 10.1242/jcs.094896] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
14-3-3 proteins have been extensively studied in organisms ranging from yeast to mammals and are associated with multiple roles, including fundamental processes such as the cell cycle, apoptosis and the stress response, to diseases such as cancer. In Caenorhabditis elegans, there are two 14-3-3 genes, ftt-2 and par-5. ftt-2 is expressed only in somatic lineages, whereas par-5 expression is detected in both soma and germline. During early embryonic development, par-5 is necessary to establish cell polarity. Although it is known that par-5 inactivation results in sterility, the role of this gene in germline development is poorly characterized. In the present study, we used a par-5 mutation and RNA interference to characterize par-5 functions in the germline. The lack of par-5 in germ cells caused cell cycle deregulation, the accumulation of endogenous DNA damage and genomic instability. Moreover, par-5 was required for checkpoint-induced cell cycle arrest in response to DNA-damaging agents. We propose a model in which PAR-5 regulates CDK-1 phosphorylation to prevent premature mitotic entry. This study opens a new path to investigate the mechanisms of 14-3-3 functions, which are not only essential for C. elegans development, but have also been shown to be altered in human diseases.
Collapse
Affiliation(s)
- David Aristizábal-Corrales
- Drug Delivery and Targeting, CIBBIM-Nanomedicine, Vall d'Hebron Research Institute, Universidad Autónoma de Barcelona, Barcelona, Spain
| | | | | | | | | | | |
Collapse
|
321
|
Tao H, Inoue KI, Kiyonari H, Bassuk AG, Axelrod JD, Sasaki H, Aizawa S, Ueno N. Nuclear localization of Prickle2 is required to establish cell polarity during early mouse embryogenesis. Dev Biol 2012; 364:138-48. [PMID: 22333836 DOI: 10.1016/j.ydbio.2012.01.025] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2011] [Revised: 01/29/2012] [Accepted: 01/30/2012] [Indexed: 12/27/2022]
Abstract
The establishment of trophectoderm (TE) manifests as the formation of epithelium, and is dependent on many structural and regulatory components that are commonly found and function in many epithelial tissues. However, the mechanism of TE formation is currently not well understood. Prickle1 (Pk1), a core component of the planar cell polarity (PCP) pathway, is essential for epiblast polarization before gastrulation, yet the roles of Pk family members in early mouse embryogenesis are obscure. Here we found that Pk2(-/-) embryos died at E3.0-3.5 without forming the blastocyst cavity and not maintained epithelial integrity of TE. These phenotypes were due to loss of the apical-basal (AB) polarity that underlies the asymmetric redistribution of microtubule networks and proper accumulation of AB polarity components on each membrane during compaction. In addition, we found GTP-bound active form of nuclear RhoA was decreased in Pk2(-/-) embryos during compaction. We further show that the first cell fate decision was disrupted in Pk2(-/-) embryos. Interestingly, Pk2 localized to the nucleus from the 2-cell to around the 16-cell stage despite its cytoplasmic function previously reported. Inhibiting farnesylation blocked Pk2's nuclear localization and disrupted AB cell polarity, suggesting that Pk2 farnesylation is essential for its nuclear localization and function. The cell polarity phenotype was efficiently rescued by nuclear but not cytoplasmic Pk2, demonstrating the nuclear localization of Pk2 is critical for its function.
Collapse
Affiliation(s)
- Hirotaka Tao
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Developmental Biology (CDB), Chuo-ku, Kobe, Japan
| | | | | | | | | | | | | | | |
Collapse
|
322
|
McKinley RFA, Yu CG, Harris TJC. Assembly of Bazooka polarity landmarks through a multifaceted membrane-association mechanism. J Cell Sci 2012; 125:1177-90. [PMID: 22303000 DOI: 10.1242/jcs.091884] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Epithelial cell polarity is essential for animal development. The scaffold protein Bazooka (Baz/PAR-3) forms apical polarity landmarks to organize epithelial cells. However, it is unclear how Baz is recruited to the plasma membrane and how this is coupled with downstream effects. Baz contains an oligomerization domain, three PDZ domains, and binding regions for the protein kinase aPKC and phosphoinositide lipids. With a structure-function approach, we dissected the roles of these domains in the localization and function of Baz in the Drosophila embryonic ectoderm. We found that a multifaceted membrane association mechanism localizes Baz to the apical circumference. Although none of the Baz protein domains are essential for cortical localization, we determined that each contributes to cortical anchorage in a specific manner. We propose that the redundancies involved might provide plasticity and robustness to Baz polarity landmarks. We also identified specific downstream effects, including the promotion of epithelial structure, a positive-feedback loop that recruits aPKC, PAR-6 and Crumbs, and a negative-feedback loop that regulates Baz.
Collapse
Affiliation(s)
- R F Andrew McKinley
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | | | | |
Collapse
|
323
|
Nakajima H, Tanoue T. Lulu2 regulates the circumferential actomyosin tensile system in epithelial cells through p114RhoGEF. ACTA ACUST UNITED AC 2012; 195:245-61. [PMID: 22006950 PMCID: PMC3198159 DOI: 10.1083/jcb.201104118] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The FERM domain–containing protein Lulu2 and p114RhoGEF function at epithelial cell–cell junctions to regulate the actomyosin belt that determines cell shape. Myosin II–driven mechanical forces control epithelial cell shape and morphogenesis. In particular, the circumferential actomyosin belt, which is located along apical cell–cell junctions, regulates many cellular processes. Despite its importance, the molecular mechanisms regulating the belt are not fully understood. In this paper, we characterize Lulu2, a FERM (4.1 protein, ezrin, radixin, moesin) domain–containing molecule homologous to Drosophila melanogaster Yurt, as an important regulator. In epithelial cells, Lulu2 is localized along apical cell–cell boundaries, and Lulu2 depletion by ribonucleic acid interference results in disorganization of the circumferential actomyosin belt. In its regulation of the belt, Lulu2 interacts with and activates p114RhoGEF, a Rho-specific guanine nucleotide exchanging factor (GEF), at apical cell–cell junctions. This interaction is negatively regulated via phosphorylation events in the FERM-adjacent domain of Lulu2 catalyzed by atypical protein kinase C. We further found that Patj, an apical cell polarity regulator, recruits p114RhoGEF to apical cell–cell boundaries via PDZ (PSD-95/Dlg/ZO-1) domain–mediated interaction. These findings therefore reveal a novel molecular system regulating the circumferential actomyosin belt in epithelial cells.
Collapse
Affiliation(s)
- Hiroyuki Nakajima
- Global Centers of Excellence Program for Integrative Membrane Biology, Graduate School of Medicine, Kobe University, Chuo-ku, Kobe 650-0017, Japan
| | | |
Collapse
|
324
|
Guilgur LG, Prudêncio P, Ferreira T, Pimenta-Marques AR, Martinho RG. Drosophila aPKC is required for mitotic spindle orientation during symmetric division of epithelial cells. Development 2012; 139:503-13. [DOI: 10.1242/dev.071027] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Epithelial cells mostly orient the spindle along the plane of the epithelium (planar orientation) for mitosis to produce two identical daughter cells. The correct orientation of the spindle relies on the interaction between cortical polarity components and astral microtubules. Recent studies in mammalian tissue culture cells suggest that the apically localised atypical protein kinase C (aPKC) is important for the planar orientation of the mitotic spindle in dividing epithelial cells. Yet, in chicken neuroepithelial cells, aPKC is not required in vivo for spindle orientation, and it has been proposed that the polarization cues vary between different epithelial cell types and/or developmental processes. In order to investigate whether Drosophila aPKC is required for spindle orientation during symmetric division of epithelial cells, we took advantage of a previously isolated temperature-sensitive allele of aPKC. We showed that Drosophila aPKC is required in vivo for spindle planar orientation and apical exclusion of Pins (Raps). This suggests that the cortical cues necessary for spindle orientation are not only conserved between Drosophila and mammalian cells, but are also similar to those required for spindle apicobasal orientation during asymmetric cell division.
Collapse
Affiliation(s)
- Leonardo G. Guilgur
- Instituto Gulbenkian de Ciência, Rua da Quinta Grande 6, Oeiras 2781-901, Portugal
| | - Pedro Prudêncio
- Instituto Gulbenkian de Ciência, Rua da Quinta Grande 6, Oeiras 2781-901, Portugal
| | - Tânia Ferreira
- Instituto Gulbenkian de Ciência, Rua da Quinta Grande 6, Oeiras 2781-901, Portugal
| | | | - Rui Gonçalo Martinho
- Instituto Gulbenkian de Ciência, Rua da Quinta Grande 6, Oeiras 2781-901, Portugal
- Regenerative Medicine Program, Departamento de Ciências Biomédicas e Medicina, and IBB-Institute for Biotechnology and Bioengineering, Centro de Biomedicina Molecular e Estrutural, Universidade do Algarve, Campus de Gambelas, 8005-139 Faro, Portugal
| |
Collapse
|
325
|
Chang KC, Wang C, Wang H. Balancing self-renewal and differentiation by asymmetric division: insights from brain tumor suppressors in Drosophila neural stem cells. Bioessays 2012; 34:301-10. [PMID: 22287225 DOI: 10.1002/bies.201100090] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Balancing self-renewal and differentiation of stem cells is an important issue in stem cell and cancer biology. Recently, the Drosophila neuroblast (NB), neural stem cell has emerged as an excellent model for stem cell self-renewal and tumorigenesis. It is of great interest to understand how defects in the asymmetric division of neural stem cells lead to tumor formation. Here, we review recent advances in asymmetric division and the self-renewal control of Drosophila NBs. We summarize molecular mechanisms of asymmetric cell division and discuss how the defects in asymmetric division lead to tumor formation. Gain-of-function or loss-of-function of various proteins in the asymmetric machinery can drive NB overgrowth and tumor formation. These proteins control either the asymmetric protein localization or mitotic spindle orientation of NBs. We also discuss other mechanisms of brain tumor suppression that are beyond the control of asymmetric division.
Collapse
Affiliation(s)
- Kai Chen Chang
- Neuroscience & Behavioral Disorder Program, Duke-NUS Graduate Medical School Singapore, Singapore, Singapore
| | | | | |
Collapse
|
326
|
Abstract
Mammalian preimplantation development is a process of dedifferentiation from the terminally differentiated eggs to the totipotent blastomeres at the cleavage stage, and then to the pluripotent cells at the blastocyst stage. Maternal factors that accumulate during oogenesis dominate early preimplantation development until the embryonic factors gain control after the activation of the embryonic genome. Recently, a handful of maternal factors that are encoded by the maternal-effect genes have been characterized in genetically modified mouse models. These factors are shown to participate in many aspects of preimplantation development, such as the degradation of maternal macromolecues, epigenetic modification, protein translation, cellular signaling transduction, and cell compaction. Even so, little is known about the interactions between different maternal factors. In this chapter, we have summarized the functions of known maternal factors and hopefully this will lead to a better understanding of the regulation of preimplantation embryogenesis by the maternal regulatory network.
Collapse
Affiliation(s)
- Wenjing Zheng
- Department of Cell and Molecular Biology, University of Gothenburg, Gothenburg, Sweden.
| | | |
Collapse
|
327
|
Matsumura S, Toyoshima F. ABL1 Joins the Cadre of Spindle Orientation Machinery. Cell Struct Funct 2012; 37:81-7. [DOI: 10.1247/csf.12011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Affiliation(s)
- Shigeru Matsumura
- Department of Cell Biology, Institute for Virus Research, Kyoto University
- Department of Cell Biology, Institute for Virus Research, Kyoto University
| | - Fumiko Toyoshima
- Department of Cell Biology, Institute for Virus Research, Kyoto University
- Department of Cell Biology, Institute for Virus Research, Kyoto University
| |
Collapse
|
328
|
Daley WP, Gervais EM, Centanni SW, Gulfo KM, Nelson DA, Larsen M. ROCK1-directed basement membrane positioning coordinates epithelial tissue polarity. Development 2012; 139:411-22. [PMID: 22186730 PMCID: PMC3243099 DOI: 10.1242/dev.075366] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/12/2011] [Indexed: 12/23/2022]
Abstract
The basement membrane is crucial for epithelial tissue organization and function. However, the mechanisms by which basement membrane is restricted to the basal periphery of epithelial tissues and the basement membrane-mediated signals that regulate coordinated tissue organization are not well defined. Here, we report that Rho kinase (ROCK) controls coordinated tissue organization by restricting basement membrane to the epithelial basal periphery in developing mouse submandibular salivary glands, and that ROCK inhibition results in accumulation of ectopic basement membrane throughout the epithelial compartment. ROCK-regulated restriction of PAR-1b (MARK2) localization in the outer basal epithelial cell layer is required for basement membrane positioning at the tissue periphery. PAR-1b is specifically required for basement membrane deposition, as inhibition of PAR-1b kinase activity prevents basement membrane deposition and disrupts overall tissue organization, and suppression of PAR-1b together with ROCK inhibition prevents interior accumulations of basement membrane. Conversely, ectopic overexpression of wild-type PAR-1b results in ectopic interior basement membrane deposition. Significantly, culture of salivary epithelial cells on exogenous basement membrane rescues epithelial organization in the presence of ROCK1 or PAR-1b inhibition, and this basement membrane-mediated rescue requires functional integrin β1 to maintain epithelial cell-cell adhesions. Taken together, these studies indicate that ROCK1/PAR-1b-dependent regulation of basement membrane placement is required for the coordination of tissue polarity and the elaboration of tissue structure in the developing submandibular salivary gland.
Collapse
Affiliation(s)
- William P. Daley
- Graduate program in Molecular, Cellular, Developmental, and Neural Biology, University at Albany, State University of New York, Albany, NY 12208, USA
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY 12208, USA
| | - Elise M. Gervais
- Graduate program in Molecular, Cellular, Developmental, and Neural Biology, University at Albany, State University of New York, Albany, NY 12208, USA
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY 12208, USA
| | - Samuel W. Centanni
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY 12208, USA
| | - Kathryn M. Gulfo
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY 12208, USA
| | - Deirdre A. Nelson
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY 12208, USA
| | - Melinda Larsen
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY 12208, USA
| |
Collapse
|
329
|
Mai Y, Higashi T, Terada K, Hatate C, Nepal P, Horiguchi M, Harada T, Miwa S, Horinouchi T. Nicotine- and Tar-free Cigarette Smoke Extract Induces Cell Injury via Intracellular Ca2+-Dependent Subtype-Specific Protein Kinase C Activation. J Pharmacol Sci 2012; 120:310-4. [DOI: 10.1254/jphs.12219sc] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|
330
|
Abstract
After years of extensive scientific discovery much has been learned about the networks that regulate epithelial homeostasis. Loss of expression or functional activity of cell adhesion and cell polarity proteins (including the PAR, crumbs (CRB) and scribble (SCRIB) complexes) is intricately related to advanced stages of tumour progression and invasiveness. But the key roles of these proteins in crosstalk with the Hippo and liver kinase B1 (LKB1)-AMPK pathways and in epithelial function and proliferation indicate that they may also be associated with the early stages of tumorigenesis. For example, deregulation of adhesion and polarity proteins can cause misoriented cell divisions and increased self-renewal of adult epithelial stem cells. In this Review, we highlight some advances in the understanding of how loss of epithelial cell polarity contributes to tumorigenesis.
Collapse
Affiliation(s)
- Fernando Martin-Belmonte
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Madrid 28049, Spain.
| | | |
Collapse
|
331
|
Yoshiura S, Ohta N, Matsuzaki F. Tre1 GPCR signaling orients stem cell divisions in the Drosophila central nervous system. Dev Cell 2011; 22:79-91. [PMID: 22178499 DOI: 10.1016/j.devcel.2011.10.027] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2011] [Revised: 09/14/2011] [Accepted: 10/26/2011] [Indexed: 11/20/2022]
Abstract
During development, directional cell division is a major mechanism for establishing the orientation of tissue growth. Drosophila neuroblasts undergo asymmetric divisions perpendicular to the overlying epithelium to produce descendant neurons on the opposite side, thereby orienting initial neural tissue growth. However, the mechanism remains elusive. We provide genetic evidence that extrinsic GPCR signaling determines the orientation of cortical polarity underlying asymmetric divisions of neuroblasts relative to the epithelium. The GPCR Tre1 activates the G protein oα subunit in neuroblasts by interacting with the epithelium to recruit Pins, which regulates spindle orientation. Because Pins associates with the Par-complex via Inscuteable, Tre1 consequently recruits the polarity complex to orthogonally orient the polarity axis to the epithelium. Given the universal role of the Par complex in cellular polarization, we propose that the GPCR-Pins system is a comprehensive mechanism controlling tissue polarity by orienting polarized stem cells and their divisions.
Collapse
Affiliation(s)
- Shigeki Yoshiura
- RIKEN Center for Developmental Biology, 2-2-3 Minatojima-Minamimachi, Chuo-ku, Kobe 650-0047, Japan
| | | | | |
Collapse
|
332
|
Hayashi K, Suzuki A, Ohno S. PAR-1/MARK: a kinase essential for maintaining the dynamic state of microtubules. Cell Struct Funct 2011; 37:21-5. [PMID: 22139392 DOI: 10.1247/csf.11038] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
The serine/threonine kinase, PAR-1, is an essential component of the evolutionary-conserved polarity-regulating system, PAR-aPKC system, which plays indispensable roles in establishing asymmetric protein distributions and cell polarity in various biological contexts (Suzuki, A. and Ohno, S. (2006). J. Cell Sci., 119: 979-987; Matenia, D. and Mandelkow, E.M. (2009). Trends Biochem. Sci., 34: 332-342). PAR-1 is also known as MARK, which phosphorylates classical microtubule-associated proteins (MAPs) and detaches MAPs from microtubules (Matenia, D. and Mandelkow, E.M. (2009). Trends Biochem. Sci., 34: 332-342). This MARK activity of PAR-1 suggests its role in microtubule (MT) dynamics, but surprisingly, only few studies have been carried out to address this issue. Here, we summarize our recent study on live imaging analysis of MT dynamics in PAR-1b-depleted cells, which clearly demonstrated the positive role of PAR-1b in maintaining MT dynamics (Hayashi, K., Suzuki, A., Hirai, S., Kurihara, Y., Hoogenraad, C.C., and Ohno, S. (2011). J. Neurosci., 31: 12094-12103). Importantly, our results further revealed the novel physiological function of PAR-1b in maintaining dendritic spine morphology in mature neurons.
Collapse
Affiliation(s)
- Kenji Hayashi
- Department of Molecular Biology, Yokohama City University Graduate School of Medical Science, Yokohama, Japan
| | | | | |
Collapse
|
333
|
Yadlapalli S, Yamashita YM. Spindle positioning in the stem cell niche. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2011; 1:215-30. [DOI: 10.1002/wdev.16] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
334
|
Nakamuta S, Funahashi Y, Namba T, Arimura N, Picciotto MR, Tokumitsu H, Soderling TR, Sakakibara A, Miyata T, Kamiguchi H, Kaibuchi K. Local Application of Neurotrophins Specifies Axons Through Inositol 1,4,5-Trisphosphate, Calcium, and Ca2+/Calmodulin-Dependent Protein Kinases. Sci Signal 2011; 4:ra76. [DOI: 10.1126/scisignal.2002011] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|
335
|
Yao S, Bee A, Brewer D, Dodson A, Beesley C, Ke Y, Ambroisine L, Fisher G, Møller H, Dickinson T, Gerard P, Lian LY, Risk J, Lane B, Smith P, Reuter V, Berney D, Gosden C, Scardino P, Cuzick J, Djamgoz MBA, Cooper C, Foster CS. PRKC-ζ Expression Promotes the Aggressive Phenotype of Human Prostate Cancer Cells and Is a Novel Target for Therapeutic Intervention. Genes Cancer 2011; 1:444-64. [PMID: 21779455 DOI: 10.1177/1947601910376079] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2010] [Revised: 05/25/2010] [Accepted: 05/25/2010] [Indexed: 01/26/2023] Open
Abstract
We show protein kinase C-zeta (PKC-ζ) to be a novel predictive biomarker for survival from prostate cancer (P < 0.001). We also confirm that transcription of the PRKC-ζ gene is crucial to the malignant phenotype of human prostate cancer. Following siRNA silencing of PRKC-ζ in PC3-M prostate cancer cells, stable transfectant cell line si-PRKC-ζ-PC3-M(T1-6) is phenotypically nonmalignant in vitro and in vivo. Genome-wide expression analysis identified 373 genes to be differentially expressed in the knockdown cells and 4 key gene networks to be significantly perturbed during phenotype modulation. Functional interconnection between some of the modulated genes is revealed, although these may be within different regulatory pathways, emphasizing the complexity of their mutual interdependence. Genes with altered expression following PRKC-ζ knockdown include HSPB1, RAD51, and ID1 that we have previously described to be critical in prostatic malignancy. Because expression of PRKC-ζ is functionally involved in promoting the malignant phenotype, we propose PKC-ζ as a novel and biologically relevant target for therapeutic intervention in prostate cancer.
Collapse
Affiliation(s)
- Sheng Yao
- Division of Cellular Pathology and Molecular Genetics, University of Liverpool, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
336
|
Yamahashi Y, Saito Y, Murata-Kamiya N, Hatakeyama M. Polarity-regulating kinase partitioning-defective 1b (PAR1b) phosphorylates guanine nucleotide exchange factor H1 (GEF-H1) to regulate RhoA-dependent actin cytoskeletal reorganization. J Biol Chem 2011; 286:44576-84. [PMID: 22072711 DOI: 10.1074/jbc.m111.267021] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Partitioning-defective 1b (PAR1b), also known as microtubule affinity-regulating kinase 2 (MARK2), is a member of evolutionally conserved PAR1/MARK serine/threonine kinase family, which plays a key role in the establishment and maintenance of cell polarity at least partly by phosphorylating microtubule-associated proteins (MAPs) that regulate microtubule stability. PAR1b has also been reported to influence actin cytoskeletal organization, raising the possibility that PAR1b functionally interacts with the Rho family of small GTPases, central regulators of the actin cytoskeletal system. Consistent with this notion, PAR1 was recently found to be physically associated with a RhoA-specific guanine nucleotide exchange factor H1 (GEF-H1). This observation suggests a functional link between PAR1b and GEF-H1. Here we show that PAR1b induces phosphorylation of GEF-H1 on serine 885 and serine 959. We also show that PAR1b-induced serine 885/serine 959 phosphorylation inhibits RhoA-specific GEF activity of GEF-H1. As a consequence, GEF-H1 phosphorylated on both of the serine residues loses the ability to stimulate RhoA and thereby fails to induce RhoA-dependent stress fiber formation. These findings indicate that PAR1b not only regulates microtubule stability through phosphorylation of MAPs but also influences actin stress fiber formation by inducing GEF-H1 phosphorylation. The dual function of PAR1b in the microtubule-based cytoskeletal system and the actin-based cytoskeletal system in the coordinated regulation of cell polarity, cell morphology, and cell movement.
Collapse
Affiliation(s)
- Yukie Yamahashi
- Division of Microbiology, Graduate School of Medicine, University of Tokyo, Tokyo 113-0033, Japan
| | | | | | | |
Collapse
|
337
|
Maintenance of dendritic spine morphology by partitioning-defective 1b through regulation of microtubule growth. J Neurosci 2011; 31:12094-103. [PMID: 21865452 DOI: 10.1523/jneurosci.0751-11.2011] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Dendritic spines are postsynaptic structures that receive excitatory synaptic input from presynaptic terminals. Actin and its regulatory proteins play a central role in morphogenesis of dendritic spines. In addition, recent studies have revealed that microtubules are indispensable for the maintenance of mature dendritic spine morphology by stochastically invading dendritic spines and regulating dendritic localization of p140Cap, which is required for actin reorganization. However, the regulatory mechanisms of microtubule dynamics remain poorly understood. Partitioning-defective 1b (PAR1b), a cell polarity-regulating serine/threonine protein kinase, is thought to regulate microtubule dynamics by inhibiting microtubule binding of microtubule-associated proteins. Results from the present study demonstrated that PAR1b participates in the maintenance of mature dendritic spine morphology in mouse hippocampal neurons. Immunofluorescent analysis revealed PAR1b localization in the dendrites, which was concentrated in dendritic spines of mature neurons. PAR1b knock-down cells exhibited decreased mushroom-like dendritic spines, as well as increased filopodia-like dendritic protrusions, with no effect on the number of protrusions. Live imaging of microtubule plus-end tracking proteins directly revealed decreases in distance and duration of microtubule growth following PAR1b knockdown in a neuroblastoma cell line and in dendrites of hippocampal neurons. In addition, reduced accumulation of GFP-p140Cap in dendritic protrusions was confirmed in PAR1b knock-down neurons. In conclusion, the present results suggested a novel function for PAR1b in the maintenance of mature dendritic spine morphology by regulating microtubule growth and the accumulation of p140Cap in dendritic spines.
Collapse
|
338
|
Beirowski B, Gustin J, Armour SM, Yamamoto H, Viader A, North BJ, Michán S, Baloh RH, Golden JP, Schmidt RE, Sinclair DA, Auwerx J, Milbrandt J. Sir-two-homolog 2 (Sirt2) modulates peripheral myelination through polarity protein Par-3/atypical protein kinase C (aPKC) signaling. Proc Natl Acad Sci U S A 2011; 108:E952-61. [PMID: 21949390 PMCID: PMC3203793 DOI: 10.1073/pnas.1104969108] [Citation(s) in RCA: 136] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The formation of myelin by Schwann cells (SCs) occurs via a series of orchestrated molecular events. We previously used global expression profiling to examine peripheral nerve myelination and identified the NAD(+)-dependent deacetylase Sir-two-homolog 2 (Sirt2) as a protein likely to be involved in myelination. Here, we show that Sirt2 expression in SCs is correlated with that of structural myelin components during both developmental myelination and remyelination after nerve injury. Transgenic mice lacking or overexpressing Sirt2 specifically in SCs show delays in myelin formation. In SCs, we found that Sirt2 deacetylates Par-3, a master regulator of cell polarity. The deacetylation of Par-3 by Sirt2 decreases the activity of the polarity complex signaling component aPKC, thereby regulating myelin formation. These results demonstrate that Sirt2 controls an essential polarity pathway in SCs during myelin assembly and provide insights into the association between intracellular metabolism and SC plasticity.
Collapse
Affiliation(s)
| | - Jason Gustin
- Sigma–Aldrich Biotechnology, St. Louis, MO 63103
| | - Sean M. Armour
- Department of Pathology, Harvard University School of Medicine, Cambridge, MA 02115
| | - Hiroyasu Yamamoto
- Laboratory for Integrative and Systems Physiology, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | | | - Brian J. North
- Department of Pathology, Harvard University School of Medicine, Cambridge, MA 02115
| | - Shaday Michán
- Instituto de Geriatria, Institutos Nacionales de Salud, Mexico D.F., 04510, Mexico
| | - Robert H. Baloh
- Neurology, and
- Hope Center for Neurological Diseases, St. Louis, MO 63110; and
| | - Judy P. Golden
- Department of Anesthesiology, Washington University Pain Center, St. Louis, MO 63110
| | - Robert E. Schmidt
- Pathology, Washington University School of Medicine, St. Louis, MO 63110
- Hope Center for Neurological Diseases, St. Louis, MO 63110; and
| | - David A. Sinclair
- Department of Pathology, Harvard University School of Medicine, Cambridge, MA 02115
| | - Johan Auwerx
- Laboratory for Integrative and Systems Physiology, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Jeffrey Milbrandt
- Departments of Genetics
- Hope Center for Neurological Diseases, St. Louis, MO 63110; and
| |
Collapse
|
339
|
Abstract
The MDCK cell line provides a tractable model for studying protein trafficking, polarity and junctions (tight, adherens, desmosome and gap) in epithelial cells. However, there are many different strains of MDCK cells available, including the parental line, MDCK I, MDCK II, MDCK.1, MDCK.2, superdome and supertube, making it difficult for new researchers to decide which strain to use. Furthermore, there is often inadequate reporting of strain types and where cells were obtained from in the literature. This review aims to provide new researchers with a guide to the different MDCK strains and a directory of where they can be obtained. We also hope to encourage experienced researchers to report the stain and origin of their MDCK cells.
Collapse
|
340
|
Dukes JD, Whitley P, Chalmers AD. The MDCK variety pack: choosing the right strain. BMC Cell Biol 2011. [PMID: 21982418 DOI: 10.1186/1471‐2121‐12‐43] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
The MDCK cell line provides a tractable model for studying protein trafficking, polarity and junctions (tight, adherens, desmosome and gap) in epithelial cells. However, there are many different strains of MDCK cells available, including the parental line, MDCK I, MDCK II, MDCK.1, MDCK.2, superdome and supertube, making it difficult for new researchers to decide which strain to use. Furthermore, there is often inadequate reporting of strain types and where cells were obtained from in the literature. This review aims to provide new researchers with a guide to the different MDCK strains and a directory of where they can be obtained. We also hope to encourage experienced researchers to report the stain and origin of their MDCK cells.
Collapse
|
341
|
Apico-basal polarity in polycystic kidney disease epithelia. Biochim Biophys Acta Mol Basis Dis 2011; 1812:1239-48. [DOI: 10.1016/j.bbadis.2011.05.008] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2011] [Revised: 05/19/2011] [Accepted: 05/24/2011] [Indexed: 12/29/2022]
|
342
|
Hsu T. NME genes in epithelial morphogenesis. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2011; 384:363-72. [PMID: 21336542 PMCID: PMC3337754 DOI: 10.1007/s00210-011-0607-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/28/2010] [Accepted: 01/27/2011] [Indexed: 01/29/2023]
Abstract
The NME family of genes encodes highly conserved multifunctional proteins that have been shown to participate in nucleic acid metabolism, energy homeostasis, cell signaling, and cancer progression. Some family members, particularly isoforms 1 and 2, have attracted extensive interests because of their potential anti-metastasis activity. Unfortunately, there have been few consensus mechanistic explanations for this critical function because of the numerous molecular functions ascribed to these proteins, including nucleoside diphosphate kinase, protein kinase, nuclease, transcription factor, growth factor, among others. In addition, different studies showed contradictory prognostic correlations between NME expression levels and tumor progression in clinical samples. Thus, analyses using pliable in vivo systems have become critical for unraveling at least some aspects of the complex functions of this family of genes. Recent works using the Drosophila genetic system have suggested a role for NME in regulating epithelial cell motility and morphogenesis, which has also been demonstrated in mammalian epithelial cell culture. This function is mediated by promoting internalization of growth factor receptors in motile epithelial cells, and the adherens junction components such as E-cadherin and β-catenin in epithelia that form the tissue linings. Interestingly, NME genes in epithelial cells appear to function in a defined range of expression levels. Either down-regulation or over-expression can perturb epithelial integrity, resulting in different aspects of epithelial abnormality. Such biphasic functions provide a plausible explanation for the documented anti-metastatic activity and the suspected oncogenic function. This review summarizes these recent findings and discusses their implications.
Collapse
Affiliation(s)
- Tien Hsu
- Department of Medicine, Boston University School of Medicine, 650 Albany St., Room 440, Boston, MA 02118, USA.
| |
Collapse
|
343
|
Complex cellular functions of the von Hippel-Lindau tumor suppressor gene: insights from model organisms. Oncogene 2011; 31:2247-57. [PMID: 21996733 DOI: 10.1038/onc.2011.442] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The von Hippel-Lindau tumor suppressor gene (VHL) has attracted intensive interest not only because its mutations predispose carriers to devastating tumors, but also because it is involved in oxygen sensing under physiological conditions. VHL loss-of-function mutations result in organ-specific tumors, such as hemangioblastoma of the central nervous system and renal cell carcinoma, both untreatable with conventional chemotherapies. The VHL protein is best known as an E3 ubiquitin ligase that targets hypoxia-inducible factor-α (HIF-α), but many diverse, non-canonical cellular functions have also been assigned to VHL, mainly based on studies in cell culture systems. As such, although the HIF-dependent role of VHL is critical, the full spectrum of pathophysiological functions of VHL is still unresolved. Such understanding requires careful cross-referencing with physiologically relevant experimental models. Studies in model systems, such as Caenorhabditis elegans, Drosophila, zebrafish and mouse have provided critical in vivo confirmation of the VHL-HIF pathway, and verification of potentially important cellular functions including microtubule stabilization and epithelial morphogenesis. More recently, animal models have also suggested systemic roles of VHL in hematopoiesis, metabolic homeostasis and inflammation. In this review, the studies performed in model organisms will be summarized and placed in context with existing clinical and in vitro data.
Collapse
|
344
|
Halbsgut N, Linnemannstöns K, Zimmermann LI, Wodarz A. Apical-basal polarity in Drosophila neuroblasts is independent of vesicular trafficking. Mol Biol Cell 2011; 22:4373-9. [PMID: 21937725 PMCID: PMC3216662 DOI: 10.1091/mbc.e11-03-0219] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Cell polarity in epithelia depends on the PAR proteins, which interact with the machinery for exocytic and endocytic vesicular trafficking. Polarity in Drosophila neural stem cells is independent of vesicular trafficking, although it depends on the PAR proteins, revealing different mechanisms of how polarity is controlled. The possession of apical–basal polarity is a common feature of epithelia and neural stem cells, so-called neuroblasts (NBs). In Drosophila, an evolutionarily conserved protein complex consisting of atypical protein kinase C and the scaffolding proteins Bazooka/PAR-3 and PAR-6 controls the polarity of both cell types. The components of this complex localize to the apical junctional region of epithelial cells and form an apical crescent in NBs. In epithelia, the PAR proteins interact with the cellular machinery for polarized exocytosis and endocytosis, both of which are essential for the establishment of plasma membrane polarity. In NBs, many cortical proteins show a strongly polarized subcellular localization, but there is little evidence for the existence of distinct apical and basolateral plasma membrane domains, raising the question of whether vesicular trafficking is required for polarization of NBs. We analyzed the polarity of NBs mutant for essential regulators of the main exocytic and endocytic pathways. Surprisingly, we found that none of these mutations affected NB polarity, demonstrating that NB cortical polarity is independent of plasma membrane polarity and that the PAR proteins function in a cell type–specific manner.
Collapse
Affiliation(s)
- Nils Halbsgut
- Stammzellbiologie, Abteilung Anatomie und Zellbiologie, Georg-August-Universität Göttingen, 37077 Göttingen, Germany
| | | | | | | |
Collapse
|
345
|
Dawes AT, Munro EM. PAR-3 oligomerization may provide an actin-independent mechanism to maintain distinct par protein domains in the early Caenorhabditis elegans embryo. Biophys J 2011; 101:1412-22. [PMID: 21943422 DOI: 10.1016/j.bpj.2011.07.030] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2011] [Revised: 06/11/2011] [Accepted: 07/06/2011] [Indexed: 02/06/2023] Open
Abstract
Par proteins establish discrete intracellular spatial domains to polarize many different cell types. In the single-cell embryo of the nematode worm Caenorhabditis elegans, the segregation of Par proteins is crucial for proper division and cell fate specification. Actomyosin-based cortical flows drive the initial formation of anterior and posterior Par domains, but cortical actin is not required for the maintenance of these domains. Here we develop a model of interactions between the Par proteins that includes both mutual inhibition and PAR-3 oligomerization. We show that this model gives rise to a bistable switch mechanism, allowing the Par proteins to occupy distinct anterior and posterior domains seen in the early C. elegans embryo, independent of dynamics or asymmetries in the actin cortex. The model predicts a sharp loss of cortical Par protein asymmetries during gradual depletion of the Par protein PAR-6, and we confirm this prediction experimentally. Together, these results suggest both mutual inhibition and PAR-3 oligomerization are sufficient to maintain distinct Par protein domains in the early C. elegans embryo.
Collapse
Affiliation(s)
- Adriana T Dawes
- Department of Mathematical and Statistical Statistical Sciences, University of Alberta, Edmonton, Alberta, Canada.
| | | |
Collapse
|
346
|
Abstract
The formation of axon/dendrite polarity is critical for the neuron to perform its signaling function in the brain. Recent advance in our understanding of cellular and molecular mechanisms underlying the development and maintenance of neuronal polarity has been greatly facilitated by the use of the culture system of dissociated hippocampal neurons. Among many polarization-related proteins, we here focus on the mammalian LKB1, the counterpart of the C. elegans Par-4, which is an upstream regulator among six Par (partitioning-defective) genes that act as master regulators of cell polarity in different cell types across evolutionary distant species. Recent studies have identified LKB1 and its downstream targets SAD/MARK kinases (mammalian homologs of Par-1) as key regulators of neuronal polarization and axon development in cultured neurons and in developing cortical neurons in vivo. We will review the properties of and interactions among proteins in this LKB1-SAD/MARK pathway, drawing upon information obtained from both neuronal and non-neuronal systems. Due to central role of the protein kinase A-dependent phosphorylation of LKB1 in the activation of this pathway, we will review recent findings on how cAMP and cGMP signaling may serve as antagonistic second messengers for axon/dendrite development, and how these cyclic nucleotides may mediate the action of extracellular polarizing factors by modulating the activity of the LKB1-SAD/MARK pathway.
Collapse
Affiliation(s)
- Maya Shelly
- Department of Neurobiology and Behavior, State University of New York, Stony Brook, NY 11794-5230, USA.
| | | |
Collapse
|
347
|
Coisne C, Engelhardt B. Tight junctions in brain barriers during central nervous system inflammation. Antioxid Redox Signal 2011; 15:1285-303. [PMID: 21338320 DOI: 10.1089/ars.2011.3929] [Citation(s) in RCA: 136] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Homeostasis within the central nervous system (CNS) is a prerequisite to elicit proper neuronal function. The CNS is tightly sealed from the changeable milieu of the blood stream by the blood-brain barrier (BBB) and the blood-cerebrospinal fluid (CSF) barrier (BCSFB). Whereas the BBB is established by specialized endothelial cells of CNS microvessels, the BCSFB is formed by the epithelial cells of the choroid plexus. Both constitute physical barriers by a complex network of tight junctions (TJs) between adjacent cells. During many CNS inflammatory disorders, such as multiple sclerosis, human immunodeficiency virus infection, or Alzheimer's disease, production of pro-inflammatory cytokines, matrix metalloproteases, and reactive oxygen species are responsible for alterations of CNS barriers. Barrier dysfunction can contribute to neurological disorders in a passive way by vascular leakage of blood-borne molecules into the CNS and in an active way by guiding the migration of inflammatory cells into the CNS. Both ways may directly be linked to alterations in molecular composition, function, and dynamics of the TJ proteins. This review summarizes current knowledge on the cellular and molecular aspects of the functional and dysfunctional TJ complexes at the BBB and the BCSFB, with a particular emphasis on CNS inflammation and the role of reactive oxygen species.
Collapse
Affiliation(s)
- Caroline Coisne
- Theodor Kocher Institute, University of Bern, Bern, Switzerland.
| | | |
Collapse
|
348
|
Chilov D, Sinjushina N, Rita H, Taketo MM, Mäkelä TP, Partanen J. Phosphorylated β-catenin localizes to centrosomes of neuronal progenitors and is required for cell polarity and neurogenesis in developing midbrain. Dev Biol 2011; 357:259-68. [DOI: 10.1016/j.ydbio.2011.06.029] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2010] [Revised: 05/16/2011] [Accepted: 06/21/2011] [Indexed: 10/18/2022]
|
349
|
Sato K, Watanabe T, Wang S, Kakeno M, Matsuzawa K, Matsui T, Yokoi K, Murase K, Sugiyama I, Ozawa M, Kaibuchi K. Numb controls E-cadherin endocytosis through p120 catenin with aPKC. Mol Biol Cell 2011; 22:3103-19. [PMID: 21775625 PMCID: PMC3164458 DOI: 10.1091/mbc.e11-03-0274] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Revised: 05/31/2011] [Accepted: 06/25/2011] [Indexed: 11/11/2022] Open
Abstract
Cadherin trafficking controls tissue morphogenesis and cell polarity. The endocytic adaptor Numb participates in apicobasal polarity by acting on intercellular adhesions in epithelial cells. However, it remains largely unknown how Numb controls cadherin-based adhesion. Here, we found that Numb directly interacted with p120 catenin (p120), which is known to interact with E-cadherin and prevent its internalization. Numb accumulated at intercellular adhesion sites and the apical membrane in epithelial cells. Depletion of Numb impaired E-cadherin internalization, whereas depletion of p120 accelerated internalization. Expression of the Numb-binding fragment of p120 inhibited E-cadherin internalization in a dominant-negative fashion, indicating that Numb interacts with the E-cadherin/p120 complex and promotes E-cadherin endocytosis. Impairment of Numb induced mislocalization of E-cadherin from the lateral membrane to the apical membrane. Atypical protein kinase C (aPKC), a member of the PAR complex, phosphorylated Numb and inhibited its association with p120 and α-adaptin. Depletion or inhibition of aPKC accelerated E-cadherin internalization. Wild-type Numb restored E-cadherin internalization in the Numb-depleted cells, whereas a phosphomimetic mutant or a mutant with defective α-adaptin-binding ability did not restore the internalization. Thus, we propose that aPKC phosphorylates Numb to prevent its binding to p120 and α-adaptin, thereby attenuating E-cadherin endocytosis to maintain apicobasal polarity.
Collapse
Affiliation(s)
- Kazuhide Sato
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Showa, Nagoya, Aichi 466-8550, Japan
| | - Takashi Watanabe
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Showa, Nagoya, Aichi 466-8550, Japan
- Institute for Advanced Research, Nagoya University, Furo, Chikusa, Nagoya, Aichi 464-8601, Japan
| | - Shujie Wang
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Showa, Nagoya, Aichi 466-8550, Japan
- Department of Anatomy, School of Medicine, Mie University, Tsu, Mie 514-8507, Japan
| | - Mai Kakeno
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Showa, Nagoya, Aichi 466-8550, Japan
| | - Kenji Matsuzawa
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Showa, Nagoya, Aichi 466-8550, Japan
| | - Toshinori Matsui
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Showa, Nagoya, Aichi 466-8550, Japan
| | - Keiko Yokoi
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Showa, Nagoya, Aichi 466-8550, Japan
| | - Kiyoko Murase
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Showa, Nagoya, Aichi 466-8550, Japan
| | - Ikuko Sugiyama
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Showa, Nagoya, Aichi 466-8550, Japan
| | - Masayuki Ozawa
- Department of Biochemistry and Molecular Biology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan
| | - Kozo Kaibuchi
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Showa, Nagoya, Aichi 466-8550, Japan
- CREST, Japan Science and Technology Agency, Kawaguchi, Saitama 332-0012, Japan
| |
Collapse
|
350
|
Abstract
Inositol phospholipids have been implicated in almost all aspects of cellular physiology including spatiotemporal regulation of cellular signaling, acquisition of cellular polarity, specification of membrane identity, cytoskeletal dynamics, and regulation of cellular adhesion, motility, and cytokinesis. In this review, we examine the critical role phosphoinositides play in these processes to execute the establishment and maintenance of cellular architecture. Epithelial tissues perform essential barrier and transport functions in almost all major organs. Key to their development and function is the establishment of epithelial cell polarity. We place a special emphasis on highlighting recent studies demonstrating phosphoinositide regulation of epithelial cell polarity and how individual cells use phosphoinositides to further organize into epithelial tissues.
Collapse
Affiliation(s)
- Annette Shewan
- Department of Anatomy, University of California, San Francisco, San Francisco, California 94143-2140, USA
| | | | | |
Collapse
|