1
|
English LA, Taylor RJ, Cameron CJ, Broker EA, Dent EW. F-BAR proteins CIP4 and FBP17 function in cortical neuron radial migration and process outgrowth. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.25.620310. [PMID: 39484544 PMCID: PMC11527352 DOI: 10.1101/2024.10.25.620310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Neurite initiation from newly born neurons is a critical step in neuronal differentiation and migration. Neuronal migration in the developing cortex is accompanied by dynamic extension and retraction of neurites as neurons progress through bipolar and multipolar states. However, there is a relative lack of understanding regarding how the dynamic extension and retraction of neurites is regulated during neuronal migration. In recent work we have shown that CIP4, a member of the F-BAR family of membrane bending proteins, inhibits cortical neurite formation in culture, while family member FBP17 induces premature neurite outgrowth. These results beg the question of how CIP4 and FBP17 function in radial neuron migration and differentiation in vivo , including the timing and manner of neurite extension and retraction. Indeed, the regulation of neurite outgrowth is essential for the transitions between bipolar and multipolar states during radial migration. To examine the effects of modulating expression of CIP4 and FBP17 in vivo , we used in utero electroporation, in combination with our published Double UP technique, to compare knockdown or overexpression cells with control cells within the same mouse tissue of either sex. We show that either knockdown or overexpression of CIP4 and FBP17 results in the marked disruption of radial neuron migration by modulating neuronal morphology and neurite outgrowth, consistent with our findings in culture. Our results demonstrate that the F-BAR proteins CIP4 and FBP17 are essential for proper radial migration in the developing cortex and thus play a key role in cortical development. SIGNIFICANCE STATEMENT During embryonic development, radial migration of newly born cortical neurons is a complex process that underlies the proper formation of the neocortex, the outermost layers of neurons in the brain. Disruptions in radial migration results in profound effects on cognitive function and can lead to devastating developmental disabilities. To better understand this critical process in brain development we examined two members of the F-BAR family of membrane bending proteins, CIP4 and FBP17, which are present in the developing brain. We demonstrate that intracellular concentrations of these proteins must be tightly regulated. Increasing or decreasing levels of either protein has profound effects on neuronal morphology and proper radial migration, suggesting they are key players in cortical development.
Collapse
|
2
|
Lemaigre C, Ceuppens A, Valades-Cruz CA, Ledoux B, Vanbeneden B, Hassan M, Zetterberg FR, Nilsson UJ, Johannes L, Wunder C, Renard HF, Morsomme P. N-BAR and F-BAR proteins-endophilin-A3 and PSTPIP1-control clathrin-independent endocytosis of L1CAM. Traffic 2023; 24:190-212. [PMID: 36843549 DOI: 10.1111/tra.12883] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/07/2023] [Accepted: 02/18/2023] [Indexed: 02/28/2023]
Abstract
Recent advances in the field demonstrate the high diversity and complexity of endocytic pathways. In the current study, we focus on the endocytosis of L1CAM. This glycoprotein plays a major role in the development of the nervous system, and is involved in cancer development and is associated with metastases and poor prognosis. Two L1CAM isoforms are subject to endocytosis: isoform 1, described as a clathrin-mediated cargo; isoform 2, whose endocytosis has never been studied. Deciphering the molecular machinery of isoform 2 internalisation should contribute to a better understanding of its pathophysiological role. First, we demonstrated in our cellular context that both isoforms of L1CAM are mainly a clathrin-independent cargo, which was not expected for isoform 1. Second, the mechanism of L1CAM endocytosis is specifically mediated by the N-BAR domain protein endophilin-A3. Third, we discovered PSTPIP1, an F-BAR domain protein, as a novel actor in this endocytic process. Finally, we identified galectins as endocytic partners and negative regulators of L1CAM endocytosis. In summary, the interplay of the BAR proteins endophilin-A3 and PSTPIP1, and galectins fine tune the clathrin-independent endocytosis of L1CAM.
Collapse
Affiliation(s)
- Camille Lemaigre
- UCLouvain, Louvain Institute of Biomolecular Science and Technology, Group of Molecular Physiology, Louvain-la-Neuve, Belgium
| | - Apolline Ceuppens
- UCLouvain, Louvain Institute of Biomolecular Science and Technology, Group of Molecular Physiology, Louvain-la-Neuve, Belgium
| | - Cesar Augusto Valades-Cruz
- Institut Curie, Université PSL, U1143 INSERM, UMR3666 CNRS, Cellular and Chemical Biology unit, Paris, France.,SERPICO Project Team, UMR144 CNRS Institut Curie, PSL Research University, Paris, France.,SERPICO Project Team, Inria Centre Rennes-Bretagne Atlantique, Campus Universitaire de Beaulieu, Rennes, France
| | - Benjamin Ledoux
- UCLouvain, Louvain Institute of Biomolecular Science and Technology, Group of Molecular Physiology, Louvain-la-Neuve, Belgium
| | - Bastien Vanbeneden
- UCLouvain, Louvain Institute of Biomolecular Science and Technology, Group of Molecular Physiology, Louvain-la-Neuve, Belgium
| | | | | | - Ulf J Nilsson
- Department of Chemistry, Lund University, Lund, Sweden
| | - Ludger Johannes
- Institut Curie, Université PSL, U1143 INSERM, UMR3666 CNRS, Cellular and Chemical Biology unit, Paris, France
| | - Christian Wunder
- Institut Curie, Université PSL, U1143 INSERM, UMR3666 CNRS, Cellular and Chemical Biology unit, Paris, France
| | - Henri-François Renard
- UNamur, NARILIS, Unité de recherche en biologie cellulaire animale (URBC), Namur, Belgium
| | - Pierre Morsomme
- UCLouvain, Louvain Institute of Biomolecular Science and Technology, Group of Molecular Physiology, Louvain-la-Neuve, Belgium
| |
Collapse
|
3
|
Lettau M, Janssen O. Intra- and Extracellular Effector Vesicles From Human T And NK Cells: Same-Same, but Different? Front Immunol 2022; 12:804895. [PMID: 35003134 PMCID: PMC8733945 DOI: 10.3389/fimmu.2021.804895] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 12/07/2021] [Indexed: 12/13/2022] Open
Abstract
Cytotoxic T lymphocytes (CTL) and Natural Killer (NK) cells utilize an overlapping effector arsenal for the elimination of target cells. It was initially proposed that all cytotoxic effector proteins are stored in lysosome-related effector vesicles (LREV) termed "secretory lysosomes" as a common storage compartment and are only released into the immunological synapse formed between the effector and target cell. The analysis of enriched LREV, however, revealed an uneven distribution of individual effectors in morphologically distinct vesicular entities. Two major populations of LREV were distinguished based on their protein content and signal requirements for degranulation. Light vesicles carrying FasL and 15 kDa granulysin are released in a PKC-dependent and Ca2+-independent manner, whereas dense granules containing perforin, granzymes and 9 kDa granulysin require Ca2+-signaling as a hallmark of classical degranulation. Notably, both types of LREV do not only contain the mentioned cytolytic effectors, but also store and transport diverse other immunomodulatory proteins including MHC class I and II, costimulatory and adhesion molecules, enzymes (i.e. CD26/DPP4) or cytokines. Interestingly, the recent analyses of CTL- or NK cell-derived extracellular vesicles (EV) revealed the presence of a related mixture of proteins in microvesicles or exosomes that in fact resemble fingerprints of the cells of origin. This overlapping protein profile indicates a direct relation of intra- and extracellular vesicles. Since EV potentially also interact with cells at distant sites (apart from the IS), they might act as additional effector vesicles or intercellular communicators in a more systemic fashion.
Collapse
Affiliation(s)
- Marcus Lettau
- Molecular Immunology, Institute of Immunology, University Hospital Schleswig-Holstein, Kiel, Germany.,Department of Internal Medicine II, Unit for Hematological Diagnostics, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Ottmar Janssen
- Molecular Immunology, Institute of Immunology, University Hospital Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
4
|
Liu Y, McDonald NA, Naegele SM, Gould KL, Wu JQ. The F-BAR Domain of Rga7 Relies on a Cooperative Mechanism of Membrane Binding with a Partner Protein during Fission Yeast Cytokinesis. Cell Rep 2020; 26:2540-2548.e4. [PMID: 30840879 PMCID: PMC6425953 DOI: 10.1016/j.celrep.2019.01.112] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 01/03/2019] [Accepted: 01/30/2019] [Indexed: 12/17/2022] Open
Abstract
F-BAR proteins bind the plasma membrane (PM) to scaffold and organize the actin cytoskeleton. To understand how F-BAR proteins achieve their PM association, we studied the localization of a Schizosaccharomyces pombe F-BAR protein Rga7, which requires the coiled-coil protein Rng10 for targeting to the division site during cytokinesis. We find that the Rga7 F-BAR domain directly binds a motif in Rng10 simultaneously with the PM, and that an adjacent Rng10 motif independently binds the PM. Together, these multivalent interactions significantly enhance Rga7 F-BAR avidity for membranes at physiological protein concentrations, ensuring the division site localization of Rga7. Moreover, the requirement for the F-BAR domain in Rga7 localization and function in cytokinesis is bypassed by tethering an Rga7 construct lacking its F-BAR to Rng10, indicating that at least some F-BAR domains are necessary but not sufficient for PM targeting and are stably localized to specific cortical positions through adaptor proteins. Liu et al. show that the Rga7 F-BAR domain binds an adaptor protein Rng10, which contains a second membrane-binding module, to enhance Rga7 membrane avidity and stabilize its membrane association. The authors reveal a mechanism by which F-BAR domains can achieve high-avidity binding with the plasma membrane.
Collapse
Affiliation(s)
- Yajun Liu
- Department of Molecular Genetics, The Ohio State University, Columbus, OH 43210, USA
| | - Nathan A McDonald
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37240, USA
| | - Shelby M Naegele
- Department of Molecular Genetics, The Ohio State University, Columbus, OH 43210, USA
| | - Kathleen L Gould
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37240, USA.
| | - Jian-Qiu Wu
- Department of Molecular Genetics, The Ohio State University, Columbus, OH 43210, USA; Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
5
|
Yeast as a Model to Understand Actin-Mediated Cellular Functions in Mammals-Illustrated with Four Actin Cytoskeleton Proteins. Cells 2020; 9:cells9030672. [PMID: 32164332 PMCID: PMC7140605 DOI: 10.3390/cells9030672] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 03/05/2020] [Accepted: 03/05/2020] [Indexed: 12/31/2022] Open
Abstract
The budding yeast Saccharomyces cerevisiae has an actin cytoskeleton that comprises a set of protein components analogous to those found in the actin cytoskeletons of higher eukaryotes. Furthermore, the actin cytoskeletons of S. cerevisiae and of higher eukaryotes have some similar physiological roles. The genetic tractability of budding yeast and the availability of a stable haploid cell type facilitates the application of molecular genetic approaches to assign functions to the various actin cytoskeleton components. This has provided information that is in general complementary to that provided by studies of the equivalent proteins of higher eukaryotes and hence has enabled a more complete view of the role of these proteins. Several human functional homologues of yeast actin effectors are implicated in diseases. A better understanding of the molecular mechanisms underpinning the functions of these proteins is critical to develop improved therapeutic strategies. In this article we chose as examples four evolutionarily conserved proteins that associate with the actin cytoskeleton: (1) yeast Hof1p/mammalian PSTPIP1, (2) yeast Rvs167p/mammalian BIN1, (3) yeast eEF1A/eEF1A1 and eEF1A2 and (4) yeast Yih1p/mammalian IMPACT. We compare the knowledge on the functions of these actin cytoskeleton-associated proteins that has arisen from studies of their homologues in yeast with information that has been obtained from in vivo studies using live animals or in vitro studies using cultured animal cell lines.
Collapse
|
6
|
Taylor KL, Taylor RJ, Richters KE, Huynh B, Carrington J, McDermott ME, Wilson RL, Dent EW. Opposing functions of F-BAR proteins in neuronal membrane protrusion, tubule formation, and neurite outgrowth. Life Sci Alliance 2019; 2:2/3/e201800288. [PMID: 31160379 PMCID: PMC6549137 DOI: 10.26508/lsa.201800288] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 05/15/2019] [Accepted: 05/17/2019] [Indexed: 01/08/2023] Open
Abstract
Neurite formation is a fundamental antecedent to axon and dendrite formation, but the mechanisms that underlie this important process are poorly characterized. Here, we demonstrate that two F-BAR proteins, CIP4 and FBP17, have opposing functions in early cortical neuron development. The F-BAR family of proteins play important roles in many cellular processes by regulating both membrane and actin dynamics. The CIP4 family of F-BAR proteins is widely recognized to function in endocytosis by elongating endocytosing vesicles. However, in primary cortical neurons, CIP4 concentrates at the tips of extending lamellipodia and filopodia and inhibits neurite outgrowth. Here, we report that the highly homologous CIP4 family member, FBP17, induces tubular structures in primary cortical neurons and results in precocious neurite formation. Through domain swapping and deletion experiments, we demonstrate that a novel polybasic region between the F-BAR and HR1 domains is required for membrane bending. Moreover, the presence of a poly-PxxP region in longer splice isoforms of CIP4 and FBP17 largely reverses the localization and function of these proteins. Thus, CIP4 and FBP17 function as an antagonistic pair to fine-tune membrane protrusion, endocytosis, and neurite formation during early neuronal development.
Collapse
Affiliation(s)
- Kendra L Taylor
- University of Wisconsin-Madison, Neuroscience Training Program, Madison, WI, USA
| | - Russell J Taylor
- University of Wisconsin-Madison, Neuroscience Training Program, Madison, WI, USA
| | - Karl E Richters
- University of Wisconsin-Madison, Department of Neuroscience, Madison, WI, USA
| | - Brandon Huynh
- University of Wisconsin-Madison, Department of Neuroscience, Madison, WI, USA
| | - Justin Carrington
- University of Wisconsin-Madison, Department of Neuroscience, Madison, WI, USA
| | - Maeve E McDermott
- University of Wisconsin-Madison, Department of Neuroscience, Madison, WI, USA
| | - Rebecca L Wilson
- University of Wisconsin-Madison, Department of Neuroscience, Madison, WI, USA
| | - Erik W Dent
- University of Wisconsin-Madison, Department of Neuroscience, Madison, WI, USA
| |
Collapse
|
7
|
Mierke CT. The matrix environmental and cell mechanical properties regulate cell migration and contribute to the invasive phenotype of cancer cells. REPORTS ON PROGRESS IN PHYSICS. PHYSICAL SOCIETY (GREAT BRITAIN) 2019; 82:064602. [PMID: 30947151 DOI: 10.1088/1361-6633/ab1628] [Citation(s) in RCA: 131] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
The minimal structural unit of a solid tumor is a single cell or a cellular compartment such as the nucleus. A closer look inside the cells reveals that there are functional compartments or even structural domains determining the overall properties of a cell such as the mechanical phenotype. The mechanical interaction of these living cells leads to the complex organization such as compartments, tissues and organs of organisms including mammals. In contrast to passive non-living materials, living cells actively respond to the mechanical perturbations occurring in their microenvironment during diseases such as fibrosis and cancer. The transformation of single cancer cells in highly aggressive and hence malignant cancer cells during malignant cancer progression encompasses the basement membrane crossing, the invasion of connective tissue, the stroma microenvironments and transbarrier migration, which all require the immediate interaction of the aggressive and invasive cancer cells with the surrounding extracellular matrix environment including normal embedded neighboring cells. All these steps of the metastatic pathway seem to involve mechanical interactions between cancer cells and their microenvironment. The pathology of cancer due to a broad heterogeneity of cancer types is still not fully understood. Hence it is necessary to reveal the signaling pathways such as mechanotransduction pathways that seem to be commonly involved in the development and establishment of the metastatic and mechanical phenotype in several carcinoma cells. We still do not know whether there exist distinct metastatic genes regulating the progression of tumors. These metastatic genes may then be activated either during the progression of cancer by themselves on their migration path or in earlier stages of oncogenesis through activated oncogenes or inactivated tumor suppressor genes, both of which promote the metastatic phenotype. In more detail, the adhesion of cancer cells to their surrounding stroma induces the generation of intracellular contraction forces that deform their microenvironments by alignment of fibers. The amplitude of these forces can adapt to the mechanical properties of the microenvironment. Moreover, the adhesion strength of cancer cells seems to determine whether a cancer cell is able to migrate through connective tissue or across barriers such as the basement membrane or endothelial cell linings of blood or lymph vessels in order to metastasize. In turn, exposure of adherent cancer cells to physical forces, such as shear flow in vessels or compression forces around tumors, reinforces cell adhesion, regulates cell contractility and restructures the ordering of the local stroma matrix that leads subsequently to secretion of crosslinking proteins or matrix degrading enzymes. Hence invasive cancer cells alter the mechanical properties of their microenvironment. From a mechanobiological point-of-view, the recognized physical signals are transduced into biochemical signaling events that guide cellular responses such as cancer progression after the malignant transition of cancer cells from an epithelial and non-motile phenotype to a mesenchymal and motile (invasive) phenotype providing cellular motility. This transition can also be described as the physical attempt to relate this cancer cell transitional behavior to a T1 phase transition such as the jamming to unjamming transition. During the invasion of cancer cells, cell adaptation occurs to mechanical alterations of the local stroma, such as enhanced stroma upon fibrosis, and therefore we need to uncover underlying mechano-coupling and mechano-regulating functional processes that reinforce the invasion of cancer cells. Moreover, these mechanisms may also be responsible for the awakening of dormant residual cancer cells within the microenvironment. Physicists were initially tempted to consider the steps of the cancer metastasis cascade as single events caused by a single mechanical alteration of the overall properties of the cancer cell. However, this general and simple view has been challenged by the finding that several mechanical properties of cancer cells and their microenvironment influence each other and continuously contribute to tumor growth and cancer progression. In addition, basement membrane crossing, cell invasion and transbarrier migration during cancer progression is explained in physical terms by applying physical principles on living cells regardless of their complexity and individual differences of cancer types. As a novel approach, the impact of the individual microenvironment surrounding cancer cells is also included. Moreover, new theories and models are still needed to understand why certain cancers are malignant and aggressive, while others stay still benign. However, due to the broad variety of cancer types, there may be various pathways solely suitable for specific cancer types and distinct steps in the process of cancer progression. In this review, physical concepts and hypotheses of cancer initiation and progression including cancer cell basement membrane crossing, invasion and transbarrier migration are presented and discussed from a biophysical point-of-view. In addition, the crosstalk between cancer cells and a chronically altered microenvironment, such as fibrosis, is discussed including the basic physical concepts of fibrosis and the cellular responses to mechanical stress caused by the mechanically altered microenvironment. Here, is highlighted how biophysical approaches, both experimentally and theoretically, have an impact on classical hallmarks of cancer and fibrosis and how they contribute to the understanding of the regulation of cancer and its progression by sensing and responding to the physical environmental properties through mechanotransduction processes. Finally, this review discusses various physical models of cell migration such as blebbing, nuclear piston, protrusive force and unjamming transition migration modes and how they contribute to cancer progression. Moreover, these cellular migration modes are influenced by microenvironmental perturbances such as fibrosis that can induce mechanical alterations in cancer cells, which in turn may impact the environment. Hence, the classical hallmarks of cancer need to be refined by including biomechanical properties of cells, cell clusters and tissues and their microenvironment to understand mechano-regulatory processes within cancer cells and the entire organism.
Collapse
|
8
|
Wang M, Nishihama R, Onishi M, Pringle JR. Role of the Hof1-Cyk3 interaction in cleavage-furrow ingression and primary-septum formation during yeast cytokinesis. Mol Biol Cell 2018; 29:597-609. [PMID: 29321253 PMCID: PMC6004579 DOI: 10.1091/mbc.e17-04-0227] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 12/26/2017] [Accepted: 01/03/2018] [Indexed: 11/11/2022] Open
Abstract
In Saccharomyces cerevisiae, it is well established that Hof1, Cyk3, and Inn1 contribute to septum formation and cytokinesis. Because hof1∆ and cyk3∆ single mutants have relatively mild defects but hof1∆ cyk3∆ double mutants are nearly dead, it has been hypothesized that these proteins contribute to parallel pathways. However, there is also evidence that they interact physically. In this study, we examined this interaction and its functional significance in detail. Our data indicate that the interaction 1) is mediated by a direct binding of the Hof1 SH3 domain to a proline-rich motif in Cyk3; 2) occurs specifically at the time of cytokinesis but is independent of the (hyper)phosphorylation of both proteins that occurs at about the same time; 3) is dispensable for the normal localization of both proteins; 4) is essential for normal primary-septum formation and a normal rate of cleavage-furrow ingression; and 5) becomes critical for growth when either Inn1 or the type II myosin Myo1 (a key component of the contractile actomyosin ring) is absent. The similarity in phenotype between cyk3∆ mutants and mutants specifically lacking the Hof1-Cyk3 interaction suggests that the interaction is particularly important for Cyk3 function, but it may be important for Hof1 function as well.
Collapse
Affiliation(s)
- Meng Wang
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305
| | - Ryuichi Nishihama
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305
| | - Masayuki Onishi
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305
| | - John R Pringle
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305
| |
Collapse
|
9
|
Aspenström P. BAR Domain Proteins Regulate Rho GTPase Signaling. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1111:33-53. [PMID: 30151649 DOI: 10.1007/5584_2018_259] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The Bin-Amphiphysin-Rvs (BAR) domain is a membrane lipid binding domain present in a wide variety of proteins, often proteins with a role in Rho-regulated signaling pathways. BAR domains do not only confer binding to lipid bilayers, they also possess a membrane sculpturing ability and thereby directly control the topology of biomembranes. BAR domain-containing proteins participate in a plethora of physiological processes but the common denominator is their capacity to link membrane dynamics to actin dynamics and thereby integrate processes such as endocytosis, exocytosis, vesicle trafficking, cell morphogenesis and cell migration. The Rho family of small GTPases constitutes an important bridging theme for many BAR domain-containing proteins. This review article will focus predominantly on the role of BAR proteins as regulators or effectors of Rho GTPases and it will only briefly discuss the structural and biophysical function of the BAR domains.
Collapse
Affiliation(s)
- Pontus Aspenström
- Department of Microbiology, and Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
10
|
Brüser L, Bogdan S. Adherens Junctions on the Move-Membrane Trafficking of E-Cadherin. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a029140. [PMID: 28096264 DOI: 10.1101/cshperspect.a029140] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cadherin-based adherens junctions are conserved structures that mediate epithelial cell-cell adhesion in invertebrates and vertebrates. Despite their pivotal function in epithelial integrity, adherens junctions show a remarkable plasticity that is a prerequisite for tissue architecture and morphogenesis. Epithelial cadherin (E-cadherin) is continuously turned over and undergoes cycles of endocytosis, sorting and recycling back to the plasma membrane. Mammalian cell culture and genetically tractable model systems such as Drosophila have revealed conserved, but also distinct, mechanisms in the regulation of E-cadherin membrane trafficking. Here, we discuss our current knowledge about molecules and mechanisms controlling endocytosis, sorting and recycling of E-cadherin during junctional remodeling.
Collapse
Affiliation(s)
- Lena Brüser
- Institut für Neurobiologie, Universität Münster, Badestraße 9, 48149 Münster, Germany
| | - Sven Bogdan
- Institut für Neurobiologie, Universität Münster, Badestraße 9, 48149 Münster, Germany.,Institut für Physiologie und Pathophysiologie, Abteilung Molekulare Zellphysiologie, Phillips-Universität Marburg, Emil-Mannkopff-Straße 2, 35037 Marburg, Germany
| |
Collapse
|
11
|
Meng DF, Xie P, Peng LX, Sun R, Luo DH, Chen QY, Lv X, Wang L, Chen MY, Mai HQ, Guo L, Guo X, Zheng LS, Cao L, Yang JP, Wang MY, Mei Y, Qiang YY, Zhang ZM, Yun JP, Huang BJ, Qian CN. CDC42-interacting protein 4 promotes metastasis of nasopharyngeal carcinoma by mediating invadopodia formation and activating EGFR signaling. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2017; 36:21. [PMID: 28129778 PMCID: PMC5273811 DOI: 10.1186/s13046-016-0483-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 12/23/2016] [Indexed: 01/08/2023]
Abstract
Background Nasopharyngeal carcinoma (NPC) is a common malignancy in Southern China and Southeast Asia. In this study, we investigated the functional and molecular mechanisms by which CDC42-interacting protein 4 (CIP4) influences NPC. Methods The expression levels of CIP4 were examined by Western blot, qRT-PCR or IHC. MTT assay was used to detect the proliferative rate of NPC cells. The invasive abilities were examined by matrigel and transwell assay. The metastatic abilities of NPC cells were revealed in BALB/c nude mice. Results We report that CIP4 is required for NPC cell motility and invasion. CIP4 promotes the activation of N-WASP that controls invadopodia formation and activates EGFR signaling, which induces downstream MMP2 (matrix metalloproteinase 2) upregulation. In addition, CIP4 could promote NPC metastasis by activating the EGFR pathway. In nude mouse models, distant metastasis was significantly inhibited in CIP4-silenced groups. High CIP4 expression is an independent adverse prognostic factor of overall survival (OS) and distant metastasis-free survival (DMFS). Conclusion We identify the critical role of CIP4 in metastasis of NPC which suggest that CIP4 may be a potential therapeutic target of NPC patients.
Collapse
Affiliation(s)
- Dong-Fang Meng
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Ping Xie
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Li-Xia Peng
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Rui Sun
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.,Department of Nasopharyngeal Carcinoma, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Dong-Hua Luo
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.,Department of Nasopharyngeal Carcinoma, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Qiu-Yan Chen
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.,Department of Nasopharyngeal Carcinoma, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Xing Lv
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.,Department of Nasopharyngeal Carcinoma, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Lin Wang
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.,Department of Nasopharyngeal Carcinoma, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Ming-Yuan Chen
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.,Department of Nasopharyngeal Carcinoma, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Hai-Qiang Mai
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.,Department of Nasopharyngeal Carcinoma, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Ling Guo
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.,Department of Nasopharyngeal Carcinoma, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Xiang Guo
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.,Department of Nasopharyngeal Carcinoma, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Li-Sheng Zheng
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Li Cao
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Jun-Ping Yang
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Meng-Yao Wang
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.,Radiotherapy Department, Affiliated Cancer Hospital of Guangzhou Medical University, Guangzhou, 510095, China
| | - Yan Mei
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Yuan-Yuan Qiang
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Zi-Meng Zhang
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Jing-Ping Yun
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.,Department of Pathology, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Bi-Jun Huang
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Chao-Nan Qian
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China. .,Department of Nasopharyngeal Carcinoma, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
| |
Collapse
|
12
|
Jiao Q, Wang L, Zhang Z, Wang Y, Yan H, Ma W, Jin W, Lu H, Liu Y. Dynamic expression of srGAP2 in cell nuclei and cytoplasm during the differentiation of rat neural stem cells in vitro. Mol Med Rep 2016; 14:4599-4605. [PMID: 27748913 PMCID: PMC5102019 DOI: 10.3892/mmr.2016.5795] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2015] [Accepted: 11/25/2016] [Indexed: 11/06/2022] Open
Abstract
Different SLIT-ROBO Rho GTPase-activating proteins (srGAPs) have different levels of expression and diverse functions during neural development. Although srGAP2 is expressed in developmental brain tissue, little is known about its influence on cellular development of the nervous system. In the current study, dynamic expression of endogenous srGAP2 during neural stem cell/progenitor cell (NSC/NPC) differentiation in vitro was investigated in order to elucidate the association between the dynamic expression of srGAP2 and neural development. srGAP2 was expressed in undifferentiated NSCs/NPCs, and differentiated neurons and astrocytes with distinct expression patterns. In conjunction with the differentiation of NSCs/NPCs in vitro, the number of srGAP2+ cells markedly reduced. The percentage of srGAP2+ cells in the population of nestin+ and β‑tubulin III+ cells was significantly downregulated while in the population of glial fibrillary acidic protein‑positive cells, almost all cells were srGAP2+. srGAP2 was predominantly expressed in the cell nucleus in all cell types. srGAP2 was also weakly expressed in the cytoplasm of nestin+ and β‑tubulin III+ cells at 3 and 7 days in vitro. However levels were gradually downregulated during the process of differentiation and almost disappeared in β‑tubulin III+ cells at 14 days. The results from the present study suggest that srGAP2 is involved in regulating NSC/NPC differentiation during neural development. The translocation of srGAP2 in the cytoplasm and cell nucleus in different cell types may function as a director in decisions regarding cell fate.
Collapse
Affiliation(s)
- Qian Jiao
- Institute of Neurobiology, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi 710061, P.R. China
| | - Li Wang
- Institute of Neurobiology, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi 710061, P.R. China
| | - Zhichao Zhang
- Institute of Neurobiology, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi 710061, P.R. China
| | - Yuanyuan Wang
- Institute of Neurobiology, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi 710061, P.R. China
| | - Hanqi Yan
- Institute of Neurobiology, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi 710061, P.R. China
| | - Wen Ma
- Institute of Neurobiology, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi 710061, P.R. China
| | - Weilin Jin
- School of Life Science and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, P.R. China
| | - Haixia Lu
- Institute of Neurobiology, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi 710061, P.R. China
| | - Yong Liu
- Institute of Neurobiology, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
13
|
Norton RL, Fredericks GJ, Huang Z, Fay JD, Hoffmann FW, Hoffmann PR. Selenoprotein K regulation of palmitoylation and calpain cleavage of ASAP2 is required for efficient FcγR-mediated phagocytosis. J Leukoc Biol 2016; 101:439-448. [PMID: 27601625 DOI: 10.1189/jlb.2a0316-156rr] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 08/07/2016] [Accepted: 08/10/2016] [Indexed: 12/17/2022] Open
Abstract
Effective activation of macrophages through phagocytic Fcγ receptors (FcγR) has been shown to require selenoprotein K (Selk). We set out to determine whether the FcγR-mediated uptake process itself also requires Selk and potential underlying mechanisms. Macrophages from Selk knockout (KO) mice were less efficient compared with wild-type (WT) controls in engulfing IgG-coated fluorescent beads. Using LC-MS/MS to screen for Selk-binding partners involved in FcγR-mediated phagocytosis, we identified Arf-GAP with SH3 domain, ANK repeat, and PH domain-containing protein 2 (ASAP2). Coimmunoprecipitation assays confirmed interactions between Selk and ASAP2. Selk was required for ASAP2 to be cleaved by calpain-2 within the Bin/Amphiphysin/Rvs (BAR) domain of ASAP2. BAR domains promote membrane association, which was consistent with our data showing that Selk deficiency led to retention of ASAP2 within the phagocytic cup. Because Selk was recently identified as a cofactor for the palmitoylation of certain proteins, we investigated whether ASAP2 was palmitoylated and whether this was related to its cleavage by calpain-2. Acyl/biotin exchange assays and MALDI-TOF analysis showed that cysteine-86 in ASAP2 was palmitoylated in WT, but to a much lesser extent in KO, mouse macrophages. Inhibitors of either palmitoylation or calpain-2 cleavage and rescue experiments with different versions of Selk demonstrated that Selk-dependent palmitoylation of ASAP2 leads to cleavage by calpain-2 within the BAR domain, which releases this protein from the maturing phagocytic cup. Overall, these findings identify ASAP2 as a new target of Selk-dependent palmitoylation and reveal a new mechanism regulating the efficiency of FcγR-mediated phagocytosis.
Collapse
Affiliation(s)
- Robert L Norton
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA; and
| | - Gregory J Fredericks
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA; and
| | - Zhi Huang
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, P. R. China
| | - Jeffrey D Fay
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA; and
| | - FuKun W Hoffmann
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA; and
| | - Peter R Hoffmann
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA; and
| |
Collapse
|
14
|
Finnigan GC, Duvalyan A, Liao EN, Sargsyan A, Thorner J. Detection of protein-protein interactions at the septin collar in Saccharomyces cerevisiae using a tripartite split-GFP system. Mol Biol Cell 2016; 27:2708-25. [PMID: 27385335 PMCID: PMC5007091 DOI: 10.1091/mbc.e16-05-0337] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 06/30/2016] [Indexed: 01/22/2023] Open
Abstract
A tripartite split-GFP system faithfully reports the order of the subunits in septin hetero-octamers (and thus can serve as a “molecular ruler”), conversely yields little or no false signal even with very highly expressed cytosolic proteins, and detects authentic interactions of other cellular proteins that are bona fide septin-binding proteins. Various methods can provide a readout of the physical interaction between two biomolecules. A recently described tripartite split-GFP system has the potential to report by direct visualization via a fluorescence signal the intimate association of minimally tagged proteins expressed at their endogenous level in their native cellular milieu and can capture transient or weak interactions. Here we document the utility of this tripartite split-GFP system to assess in living cells protein–protein interactions in a dynamic cytoskeletal structure—the septin collar at the yeast bud neck. We show, first, that for septin–septin interactions, this method yields a robust signal whose strength reflects the known spacing between the subunits in septin filaments and thus serves as a “molecular ruler.” Second, the method yields little or no spurious signal even with highly abundant cytosolic proteins readily accessible to the bud neck (including molecular chaperone Hsp82 and glycolytic enzyme Pgk1). Third, using two proteins (Bni5 and Hsl1) that have been shown by other means to bind directly to septins at the bud neck in vivo, we validate that the tripartite split-GFP method yields the same conclusions and further insights about specificity. Finally, we demonstrate the capacity of this approach to uncover additional new information by examining whether three other proteins reported to localize to the bud neck (Nis1, Bud4, and Hof1) are able to interact physically with any of the subunits in the septin collar and, if so, with which ones.
Collapse
Affiliation(s)
- Gregory C Finnigan
- Division of Biochemistry, Biophysics and Structural Biology, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720-3202
| | - Angela Duvalyan
- Division of Biochemistry, Biophysics and Structural Biology, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720-3202
| | - Elizabeth N Liao
- Division of Biochemistry, Biophysics and Structural Biology, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720-3202
| | - Aspram Sargsyan
- Division of Biochemistry, Biophysics and Structural Biology, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720-3202
| | - Jeremy Thorner
- Division of Biochemistry, Biophysics and Structural Biology, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720-3202
| |
Collapse
|
15
|
Winkle CC, Taylor KL, Dent EW, Gallo G, Greif KF, Gupton SL. Beyond the cytoskeleton: The emerging role of organelles and membrane remodeling in the regulation of axon collateral branches. Dev Neurobiol 2016; 76:1293-1307. [PMID: 27112549 DOI: 10.1002/dneu.22398] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 04/11/2016] [Accepted: 04/21/2016] [Indexed: 12/19/2022]
Abstract
The generation of axon collateral branches is a fundamental aspect of the development of the nervous system and the response of axons to injury. Although much has been discovered about the signaling pathways and cytoskeletal dynamics underlying branching, additional aspects of the cell biology of axon branching have received less attention. This review summarizes recent advances in our understanding of key factors involved in axon branching. This article focuses on how cytoskeletal mechanisms, intracellular organelles, such as mitochondria and the endoplasmic reticulum, and membrane remodeling (exocytosis and endocytosis) contribute to branch initiation and formation. Together this growing literature provides valuable insight as well as a platform for continued investigation into how multiple aspects of axonal cell biology are spatially and temporally orchestrated to give rise to axon branches. © 2016 Wiley Periodicals, Inc. Develop Neurobiol 76: 1293-1307, 2016.
Collapse
Affiliation(s)
- Cortney C Winkle
- Neurobiology Curriculum, University of North Carolina, Chapel Hill, North Carolina, 27599
| | - Kendra L Taylor
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, Wisconsin, 53705
| | - Erik W Dent
- Department of Neuroscience, University of Wisconsin-Madison, Madison, Wisconsin, 53705
| | - Gianluca Gallo
- Lewis Katz School of Medicine, Department of Anatomy and Cell Biology, Shriners Hospitals Pediatric Research Center, Temple University, Philadelphia, Pennsylvania, 19140
| | - Karen F Greif
- Department of Biology, Bryn Mawr College, Bryn Mawr, Pennsylvania, 19010
| | - Stephanie L Gupton
- Department of Cell Biology and Physiology, Neuroscience Center, University of North Carolina, Chapel Hill, North Carolina, 27599
| |
Collapse
|
16
|
Hu ZY, Liu YP, Xie LY, Wang XY, Yang F, Chen SY, Li ZG. AKAP-9 promotes colorectal cancer development by regulating Cdc42 interacting protein 4. Biochim Biophys Acta Mol Basis Dis 2016; 1862:1172-81. [PMID: 27039663 DOI: 10.1016/j.bbadis.2016.03.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Revised: 02/29/2016] [Accepted: 03/25/2016] [Indexed: 02/07/2023]
Abstract
Our previous studies have shown that PRKA kinase anchor protein 9 (AKAP-9) is involved in colorectal cancer (CRC) cell proliferation and migration in vitro. However, whether or not AKAP-9 is important for CRC development or metastasis in vivo remains unknown. In the present study, we found that AKAP-9 expression was significantly higher in human colorectal cancer tissues than the paired normal tissues. In fact, AKAP-9 level correlated with the CRC infiltrating depth and metastasis. Moreover, the higher AKAP-9 expression was associated with the lower survival rate in patients. In cultured CRC cells, knockdown of AKAP-9 inhibited cell proliferation, invasion, and migration. AKAP-9 deficiency also attenuated CRC tumor growth and metastasis in vivo. Mechanistically, AKAP-9 interacted with cdc42 interacting protein 4 (CIP4) and regulated its expression. CIP4 levels were interrelated to the AKAP-9 level in CRC cells. Functionally, AKAP-9 was essential for TGF-β1-induced epithelial-mesenchymal transition of CRC cells, and CIP4 played a critical role in mediating the function of AKAP-9. Importantly, CIP4 expression was significantly up-regulated in human CRC tissues. Taken together, our results demonstrated that AKAP-9 facilitates CRC development and metastasis via regulating CIP4-mediated epithelial-mesenchymal transition of CRC cells.
Collapse
Affiliation(s)
- Zhi-Yan Hu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Guangdong Provincial Key Laboratory of Molecular Tumour Pathology, Guangzhou 510515, China
| | - Yan-Ping Liu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Guangdong Provincial Key Laboratory of Molecular Tumour Pathology, Guangzhou 510515, China
| | - Lin-Ying Xie
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Guangdong Provincial Key Laboratory of Molecular Tumour Pathology, Guangzhou 510515, China
| | - Xiao-Yan Wang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Guangdong Provincial Key Laboratory of Molecular Tumour Pathology, Guangzhou 510515, China
| | - Fang Yang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Guangdong Provincial Key Laboratory of Molecular Tumour Pathology, Guangzhou 510515, China
| | - Shi-You Chen
- Department of Physiology & Pharmacology, University of Georgia, Athens, GA, United States.
| | - Zu-Guo Li
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Guangdong Provincial Key Laboratory of Molecular Tumour Pathology, Guangzhou 510515, China.
| |
Collapse
|
17
|
Zobel T, Brinkmann K, Koch N, Schneider K, Seemann E, Fleige A, Qualmann B, Kessels MM, Bogdan S. Cooperative functions of the two F-BAR proteins Cip4 and Nostrin in the regulation of E-cadherin in epithelial morphogenesis. J Cell Sci 2016; 128:499-515. [PMID: 25413347 DOI: 10.1242/jcs.155929] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
F-BAR proteins are prime candidates to regulate membrane curvature and dynamics during different developmental processes. Here, we analyzed nostrin, a so-far-unknown Drosophila melanogaster F-BAR protein related to Cip4. Genetic analyses revealed a strong synergism between nostrin and cip4 functions.Whereas single mutant flies are viable and fertile, combined loss of nostrin and cip4 results in reduced viability and fertility. Double mutant escaper flies show enhanced wing polarization defects and females exhibit strong egg chamber encapsulation defects. Live imaging analysis suggests that the observed phenotypes are caused by an impaired turnover of E-cadherin at the membrane. Simultaneous knockdown of Cip4 and Nostrin strongly increases the formation of tubular E-cadherin vesicles at adherens junctions. Cip4 and Nostrin localize at distinct membrane subdomains. Both proteins prefer similar membrane curvatures but seem to form distinct membrane coats and do not heterooligomerize. Our data suggest an important synergistic function of both F-BAR proteins in membrane dynamics. We propose a cooperative recruitment model, in which Cip4 initially promotes membrane invagination and early-actin-based endosomal motility, and Nostrin makes contacts with microtubules through the kinesin Khc-73 for trafficking of recycling endosomes.
Collapse
|
18
|
Liu S, Xiong X, Thomas SV, Xu Y, Cheng X, Zhao X, Yang X, Wang H. Analysis for Carom complex, signaling and function by database mining. Front Biosci (Landmark Ed) 2016; 21:856-72. [PMID: 26709809 DOI: 10.2741/4424] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Carom is a novel protein that regulates membrane curvature and transmits pathophysiological signaling. The tissue expression of Carom is unclear and its functional role and signaling are unknown. We employed a group of combined database mining strategies and established a working model of Carom signaling. We identified 26 Carom partners and established their expression profiles in human and mouse tissues. We classified three tiers of tissues for Carom/partner expression and found lymph node was the tier 1 tissue expressing Carom and most of its partners. Using GEO database, we discovered that four conditions (hypoxia, endometriosis, PPARgamma deletion and iPSC reprogramming) altered Carom/partner expression in endothelial cells. We identified 26 Carom partner signalings by Ingenuity pathway analysis. Ten of the 26 pathways and three genes (ITSN1, UBC and HSPA5) were reported to be regulated in the above four conditions. Paired induction of Carom/ITSN1 elevation was associated with pathological angiogenesis. Whereas, paired reduction of Carom/HSPA5 or UBC was associated with iPSC generation. These results provide an insight on identifying Carom complex model and predicting its functional implications.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Hong Wang
- Center for Metabolic Disease Research, Department of Pharmacology, Thrombosis Research Center,
| |
Collapse
|
19
|
Liu F, Guo H, Ou M, Hou X, Sun G, Gong W, Jing H, Tan Q, Xue W, Dai Y, Sui W. ARHGAP4 mutated in a Chinese intellectually challenged family. Gene 2015; 578:205-9. [PMID: 26707211 DOI: 10.1016/j.gene.2015.12.035] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Revised: 12/05/2015] [Accepted: 12/11/2015] [Indexed: 10/22/2022]
Abstract
OBJECTIVE Mental retardation is characterized by lower intelligence compared to the average intelligence of persons the same age. These patients have low adaptive capacity acquired by society. The genetic factors of causing MR include monogenic disease, chromosome structural aberration, and chromosome number aberration and so on. We explored the cause of a Chinese family suffering from mental retardation. METHODS We used karyotyping technology to determine the karyotype of the proband, and we used FISH to verify the result of the karyotyping. We used whole-exome sequencing to identify the disease-causing gene and used Sanger sequencing to verify the result of whole-exome sequencing to assess the family's gene expression. RESULTS The G-banding of the karyotype revealed that the patient's karyotype is 46, XY. FISH revealed that the patient does not have a trisomy syndrome. The karyotype of the proband is normal. Using whole-exome sequencing, we identified 108,767 variants in the exome gene of the patient, including 101,787 SNPs and 6980 InDels. Combining clinical information and bioinformatics analysis, including databases filtering and SIFT analysis, we found ARHGAP4 in X chromosome was candidate MR disease-causing gene. PCR and Sanger sequencing results were consistent with whole-exome sequencing. ARHGAP4 (T491M) mutation was present in the genome of the proband and his mother is a carrier, while his father, sister, and brother do not carry this mutation. CONCLUSION According to clinical information, whole-exome sequencing results and Sanger verification results, ARHGAP4 (T491M) mutation may be disease-causing gene of the MR patient. The relation between ARHGAP4 mutation and MR clinical characteristic is needed to be illuminated with participation of more MR patients.
Collapse
Affiliation(s)
- Fuhua Liu
- Nephrology Department of Guilin, 181 St Hospital, Guangxi Key Laboratory of Metabolic Diseases Research, 541002 Guilin, Guangxi, China; College of Life Science, Guangxi Normal University, 541004 Guilin, Guangxi, China
| | - Hui Guo
- Clinical Medical Research Center, the Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, 518020, Shenzhen, Guangdong, China
| | - Minglin Ou
- Nephrology Department of Guilin, 181 St Hospital, Guangxi Key Laboratory of Metabolic Diseases Research, 541002 Guilin, Guangxi, China
| | - Xianliang Hou
- Nephrology Department of Guilin, 181 St Hospital, Guangxi Key Laboratory of Metabolic Diseases Research, 541002 Guilin, Guangxi, China
| | - Guoping Sun
- Lab. Center, Shenzhen Pingshan People's Hospital, Shenzhen, Guangdong 518118, China
| | - Weiwei Gong
- Nephrology Department of Guilin, 181 St Hospital, Guangxi Key Laboratory of Metabolic Diseases Research, 541002 Guilin, Guangxi, China
| | - Huanyun Jing
- Nephrology Department of Guilin, 181 St Hospital, Guangxi Key Laboratory of Metabolic Diseases Research, 541002 Guilin, Guangxi, China
| | - Qiupei Tan
- Nephrology Department of Guilin, 181 St Hospital, Guangxi Key Laboratory of Metabolic Diseases Research, 541002 Guilin, Guangxi, China
| | - Wen Xue
- Nephrology Department of Guilin, 181 St Hospital, Guangxi Key Laboratory of Metabolic Diseases Research, 541002 Guilin, Guangxi, China
| | - Yong Dai
- Clinical Medical Research Center, the Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, 518020, Shenzhen, Guangdong, China.
| | - Weiguo Sui
- Nephrology Department of Guilin, 181 St Hospital, Guangxi Key Laboratory of Metabolic Diseases Research, 541002 Guilin, Guangxi, China.
| |
Collapse
|
20
|
Dupré L, Houmadi R, Tang C, Rey-Barroso J. T Lymphocyte Migration: An Action Movie Starring the Actin and Associated Actors. Front Immunol 2015; 6:586. [PMID: 26635800 PMCID: PMC4649030 DOI: 10.3389/fimmu.2015.00586] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 11/02/2015] [Indexed: 12/14/2022] Open
Abstract
The actin cytoskeleton is composed of a dynamic filament meshwork that builds the architecture of the cell to sustain its fundamental properties. This physical structure is characterized by a continuous remodeling, which allows cells to accomplish complex motility steps such as directed migration, crossing of biological barriers, and interaction with other cells. T lymphocytes excel in these motility steps to ensure their immune surveillance duties. In particular, actin cytoskeleton remodeling is a key to facilitate the journey of T lymphocytes through distinct tissue environments and to tune their stop and go behavior during the scanning of antigen-presenting cells. The molecular mechanisms controlling actin cytoskeleton remodeling during T lymphocyte motility have been only partially unraveled, since the function of many actin regulators has not yet been assessed in these cells. Our review aims to integrate the current knowledge into a comprehensive picture of how the actin cytoskeleton drives T lymphocyte migration. We will present the molecular actors that control actin cytoskeleton remodeling, as well as their role in the different T lymphocyte motile steps. We will also highlight which challenges remain to be addressed experimentally and which approaches appear promising to tackle them.
Collapse
Affiliation(s)
- Loïc Dupré
- INSERM, UMR 1043, Centre de Physiopathologie de Toulouse Purpan , Toulouse , France ; Université Toulouse III Paul-Sabatier , Toulouse , France ; CNRS, UMR 5282 , Toulouse , France
| | - Raïssa Houmadi
- INSERM, UMR 1043, Centre de Physiopathologie de Toulouse Purpan , Toulouse , France ; Université Toulouse III Paul-Sabatier , Toulouse , France ; CNRS, UMR 5282 , Toulouse , France
| | - Catherine Tang
- INSERM, UMR 1043, Centre de Physiopathologie de Toulouse Purpan , Toulouse , France ; Université Toulouse III Paul-Sabatier , Toulouse , France ; CNRS, UMR 5282 , Toulouse , France ; Master BIOTIN, Université Montpellier I , Montpellier , France
| | - Javier Rey-Barroso
- INSERM, UMR 1043, Centre de Physiopathologie de Toulouse Purpan , Toulouse , France ; Université Toulouse III Paul-Sabatier , Toulouse , France ; CNRS, UMR 5282 , Toulouse , France
| |
Collapse
|
21
|
Grega-Larson NE, Crawley SW, Erwin AL, Tyska MJ. Cordon bleu promotes the assembly of brush border microvilli. Mol Biol Cell 2015; 26:3803-15. [PMID: 26354418 PMCID: PMC4626065 DOI: 10.1091/mbc.e15-06-0443] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 09/02/2015] [Indexed: 01/24/2023] Open
Abstract
Microvilli are actin-based protrusions that amplify plasma membrane area and mediate interactions with the extracellular environment. We found that the multifunctional actin regulator cordon bleu promotes the growth of intestinal brush border microvilli. These results provide a new framework for investigating brush border biogenesis. Microvilli are actin-based protrusions found on the surface of diverse cell types, where they amplify membrane area and mediate interactions with the external environment. In the intestinal tract, these protrusions play central roles in nutrient absorption and host defense and are therefore essential for maintaining homeostasis. However, the mechanisms controlling microvillar assembly remain poorly understood. Here we report that the multifunctional actin regulator cordon bleu (COBL) promotes the growth of brush border (BB) microvilli. COBL localizes to the base of BB microvilli via a mechanism that requires its proline-rich N-terminus. Knockdown and overexpression studies show that COBL is needed for BB assembly and sufficient to induce microvillar growth using a mechanism that requires functional WH2 domains. We also find that COBL acts downstream of the F-BAR protein syndapin-2, which drives COBL targeting to the apical domain. These results provide insight into a mechanism that regulates microvillar growth during epithelial differentiation and have significant implications for understanding the maintenance of intestinal homeostasis.
Collapse
Affiliation(s)
- Nathan E Grega-Larson
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37240
| | - Scott W Crawley
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37240
| | - Amanda L Erwin
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37240
| | - Matthew J Tyska
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37240
| |
Collapse
|
22
|
Bezanilla M, Gladfelter AS, Kovar DR, Lee WL. Cytoskeletal dynamics: a view from the membrane. ACTA ACUST UNITED AC 2015; 209:329-37. [PMID: 25963816 PMCID: PMC4427793 DOI: 10.1083/jcb.201502062] [Citation(s) in RCA: 126] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Many aspects of cytoskeletal assembly and dynamics can be recapitulated in vitro; yet, how the cytoskeleton integrates signals in vivo across cellular membranes is far less understood. Recent work has demonstrated that the membrane alone, or through membrane-associated proteins, can effect dynamic changes to the cytoskeleton, thereby impacting cell physiology. Having identified mechanistic links between membranes and the actin, microtubule, and septin cytoskeletons, these studies highlight the membrane’s central role in coordinating these cytoskeletal systems to carry out essential processes, such as endocytosis, spindle positioning, and cellular compartmentalization.
Collapse
Affiliation(s)
- Magdalena Bezanilla
- Department of Biology, University of Massachusetts Amherst, Amherst, MA 01003
| | - Amy S Gladfelter
- Department of Biological Sciences, Dartmouth College, Hanover, NH 03755
| | - David R Kovar
- Department of Molecular Genetics and Cell Biology and Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL 60637 Department of Molecular Genetics and Cell Biology and Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL 60637
| | - Wei-Lih Lee
- Department of Biology, University of Massachusetts Amherst, Amherst, MA 01003
| |
Collapse
|
23
|
Abstract
BAR proteins comprise a heterogeneous group of multi-domain proteins with diverse biological functions. The common denominator is the Bin-Amphiphysin-Rvs (BAR) domain that not only confers targeting to lipid bilayers, but also provides scaffolding to mold lipid membranes into concave or convex surfaces. This function of BAR proteins is an important determinant in the dynamic reconstruction of membrane vesicles, as well as of the plasma membrane. Several BAR proteins function as linkers between cytoskeletal regulation and membrane dynamics. These links are provided by direct interactions between BAR proteins and actin-nucleation-promoting factors of the Wiskott-Aldrich syndrome protein family and the Diaphanous-related formins. The Rho GTPases are key factors for orchestration of this intricate interplay. This review describes how BAR proteins regulate the activity of Rho GTPases, as well as how Rho GTPases regulate the function of BAR proteins. This mutual collaboration is a central factor in the regulation of vital cellular processes, such as cell migration, cytokinesis, intracellular transport, endocytosis, and exocytosis.
Collapse
Affiliation(s)
- Pontus Aspenström
- a Department of Microbiology and Tumor and Cell Biology; Karolinska Institutet ; Stockholm , Sweden
| |
Collapse
|
24
|
Liu S, Xiong X, Zhao X, Yang X, Wang H. F-BAR family proteins, emerging regulators for cell membrane dynamic changes-from structure to human diseases. J Hematol Oncol 2015; 8:47. [PMID: 25956236 PMCID: PMC4437251 DOI: 10.1186/s13045-015-0144-2] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 04/27/2015] [Indexed: 02/08/2023] Open
Abstract
Eukaryotic cell membrane dynamics change in curvature during physiological and pathological processes. In the past ten years, a novel protein family, Fes/CIP4 homology-Bin/Amphiphysin/Rvs (F-BAR) domain proteins, has been identified to be the most important coordinators in membrane curvature regulation. The F-BAR domain family is a member of the Bin/Amphiphysin/Rvs (BAR) domain superfamily that is associated with dynamic changes in cell membrane. However, the molecular basis in membrane structure regulation and the biological functions of F-BAR protein are unclear. The pathophysiological role of F-BAR protein is unknown. This review summarizes the current understanding of structure and function in the BAR domain superfamily, classifies F-BAR family proteins into nine subfamilies based on domain structure, and characterizes F-BAR protein structure, domain interaction, and functional relevance. In general, F-BAR protein binds to cell membrane via F-BAR domain association with membrane phospholipids and initiates membrane curvature and scission via Src homology-3 (SH3) domain interaction with its partner proteins. This process causes membrane dynamic changes and leads to seven important cellular biological functions, which include endocytosis, phagocytosis, filopodium, lamellipodium, cytokinesis, adhesion, and podosome formation, via distinct signaling pathways determined by specific domain-binding partners. These cellular functions play important roles in many physiological and pathophysiological processes. We further summarize F-BAR protein expression and mutation changes observed in various diseases and developmental disorders. Considering the structure feature and functional implication of F-BAR proteins, we anticipate that F-BAR proteins modulate physiological and pathophysiological processes via transferring extracellular materials, regulating cell trafficking and mobility, presenting antigens, mediating extracellular matrix degradation, and transmitting signaling for cell proliferation.
Collapse
Affiliation(s)
- Suxuan Liu
- Department of Cardiology, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China. .,Center for Metabolic Disease Research, Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA, 19140, USA.
| | - Xinyu Xiong
- Center for Metabolic Disease Research, Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA, 19140, USA.
| | - Xianxian Zhao
- Department of Cardiology, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China.
| | - Xiaofeng Yang
- Center for Metabolic Disease Research, Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA, 19140, USA. .,Center for Cardiovascular Research, Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA, 19140, USA. .,Center for Thrombosis Research, Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA, 19140, USA.
| | - Hong Wang
- Department of Cardiology, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China. .,Center for Metabolic Disease Research, Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA, 19140, USA. .,Center for Cardiovascular Research, Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA, 19140, USA. .,Center for Thrombosis Research, Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA, 19140, USA.
| |
Collapse
|
25
|
Gerasimcik N, Dahlberg CIM, Baptista MAP, Massaad MJ, Geha RS, Westerberg LS, Severinson E. The Rho GTPase Cdc42 Is Essential for the Activation and Function of Mature B Cells. THE JOURNAL OF IMMUNOLOGY 2015; 194:4750-8. [PMID: 25870239 DOI: 10.4049/jimmunol.1401634] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Accepted: 03/16/2015] [Indexed: 12/15/2022]
Abstract
The Rho GTPase Cdc42 coordinates regulation of the actin and the microtubule cytoskeleton by binding and activating the Wiskott-Aldrich syndrome protein. We sought to define the role of intrinsic expression of Cdc42 by mature B cells in their activation and function. Mice with inducible deletion of Cdc42 in mature B cells formed smaller germinal centers and had a reduced Ab response, mostly of low affinity to T cell-dependent Ag, compared with wild-type (WT) controls. Spreading formation of long protrusions that contain F-actin, microtubules, and Cdc42-interacting protein 4, and assumption of a dendritic cell morphology in response to anti-CD40 plus IL-4 were impaired in Cdc42-deficient B cells compared with WT B cells. Cdc42-deficient B cells had an intact migratory response to chemokine in vitro, but their homing to the B cell follicles in the spleen in vivo was significantly impaired. Cdc42-deficient B cells induced a skewed cytokine response in CD4(+) T cells, compared with WT B cells. Our results demonstrate a critical role for Cdc42 in the motility of mature B cells, their cognate interaction with T cells, and their differentiation into Ab-producing cells.
Collapse
Affiliation(s)
- Natalija Gerasimcik
- Department of Molecular Biosciences, Wenner-Gren Institute, Stockholm University, SE-106 91 Stockholm, Sweden
| | - Carin I M Dahlberg
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Marisa A P Baptista
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Michel J Massaad
- Division of Immunology, Boston Children's Hospital, Boston, MA 02115; and Department of Pediatrics, Harvard Medical School, Boston, MA 02115
| | - Raif S Geha
- Division of Immunology, Boston Children's Hospital, Boston, MA 02115; and Department of Pediatrics, Harvard Medical School, Boston, MA 02115
| | - Lisa S Westerberg
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Eva Severinson
- Department of Molecular Biosciences, Wenner-Gren Institute, Stockholm University, SE-106 91 Stockholm, Sweden;
| |
Collapse
|
26
|
Kostan J, Salzer U, Orlova A, Törö I, Hodnik V, Senju Y, Zou J, Schreiner C, Steiner J, Meriläinen J, Nikki M, Virtanen I, Carugo O, Rappsilber J, Lappalainen P, Lehto VP, Anderluh G, Egelman EH, Djinović-Carugo K. Direct interaction of actin filaments with F-BAR protein pacsin2. EMBO Rep 2014; 15:1154-62. [PMID: 25216944 DOI: 10.15252/embr.201439267] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Two mechanisms have emerged as major regulators of membrane shape: BAR domain-containing proteins, which induce invaginations and protrusions, and nuclear promoting factors, which cause generation of branched actin filaments that exert mechanical forces on membranes. While a large body of information exists on interactions of BAR proteins with membranes and regulatory proteins of the cytoskeleton, little is known about connections between these two processes. Here, we show that the F-BAR domain protein pacsin2 is able to associate with actin filaments using the same concave surface employed to bind to membranes, while some other tested N-BAR and F-BAR proteins (endophilin, CIP4 and FCHO2) do not associate with actin. This finding reveals a new level of complexity in membrane remodeling processes.
Collapse
Affiliation(s)
- Julius Kostan
- Department of Structural and Computational Biology, Max F. Perutz Laboratories, University of Vienna, Vienna, Austria
| | - Ulrich Salzer
- Department of Structural and Computational Biology, Max F. Perutz Laboratories, University of Vienna, Vienna, Austria Department of Medical Biochemistry, Medical University of Vienna, Vienna, Austria
| | - Albina Orlova
- Department of Biochemistry and Molecular Genetics, University of Virginia Medical Center, Charlottesville, VA, USA
| | - Imre Törö
- Department of Structural and Computational Biology, Max F. Perutz Laboratories, University of Vienna, Vienna, Austria
| | - Vesna Hodnik
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Yosuke Senju
- Program in Cell and Molecular Biology, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Juan Zou
- Wellcome Trust Centre for Cell Biology, University of Edinburgh, Edinburgh, UK
| | - Claudia Schreiner
- Department of Structural and Computational Biology, Max F. Perutz Laboratories, University of Vienna, Vienna, Austria
| | - Julia Steiner
- Department of Structural and Computational Biology, Max F. Perutz Laboratories, University of Vienna, Vienna, Austria
| | - Jari Meriläinen
- Department of Pathology, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - Marko Nikki
- Department of Pathology, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - Ismo Virtanen
- Institute of Biomedicine/Anatomy, University of Helsinki, Helsinki, Finland
| | - Oliviero Carugo
- Department of Structural and Computational Biology, Max F. Perutz Laboratories, University of Vienna, Vienna, Austria Department of Chemistry, University of Pavia, Pavia, Italy
| | - Juri Rappsilber
- Department of Pathology, Haartman Institute, University of Helsinki, Helsinki, Finland Department of Biotechnology, Technological University of Berlin, Berlin, Germany
| | - Pekka Lappalainen
- Program in Cell and Molecular Biology, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Veli-Pekka Lehto
- Department of Pathology, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - Gregor Anderluh
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia National Institute of Chemistry, Ljubljana, Slovenia EN-FIST Centre of Excellence, Ljubljana, Slovena
| | - Edward H Egelman
- Department of Biochemistry and Molecular Genetics, University of Virginia Medical Center, Charlottesville, VA, USA
| | - Kristina Djinović-Carugo
- Department of Structural and Computational Biology, Max F. Perutz Laboratories, University of Vienna, Vienna, Austria Department of Biochemistry, Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
27
|
The CDC42-Interacting Protein 4 Controls Epithelial Cell Cohesion and Tumor Dissemination. Dev Cell 2014; 30:553-68. [DOI: 10.1016/j.devcel.2014.08.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Revised: 04/17/2014] [Accepted: 08/06/2014] [Indexed: 01/14/2023]
|
28
|
Abstract
Neutrophil granulocytes are key effector cells of the vertebrate immune system. They represent 50-70% of the leukocytes in the human blood and their loss by disease or drug side effect causes devastating bacterial infections. Their high turnover rate, their fine-tuned killing machinery, and their arsenal of toxic vesicles leave them particularly vulnerable to various genetic deficiencies. The aim of this review is to highlight those congenital immunodeficiencies which impede the dynamics of neutrophils, such as migration, cytoskeletal rearrangements, vesicular trafficking, and secretion.
Collapse
|
29
|
Cytoskeletal and signaling mechanisms of neurite formation. Cell Tissue Res 2014; 359:267-78. [PMID: 25080065 DOI: 10.1007/s00441-014-1955-0] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Accepted: 07/01/2014] [Indexed: 10/25/2022]
Abstract
The formation of a neurite, the basis for axons and dendrites, begins with the concerted accumulation and organization of actin and microtubules. Whereas much is known about the proteins that play a role in these processes, because they perform similar functions in axon branching and filopodia formation, much remains to be discovered concerning the interaction of these individual cytoskeletal regulators during neurite formation. Here, we review the literature regarding various models of filopodial formation and the way in which proteins that control actin organization and polymerization induce neurite formation. Although several different regulators of actin polymerization are involved in neurite initiation, redundancy occurs between these regulators, as the effects of the loss of a single regulator can be mitigated by the addition of neurite-promoting substrates and proteins. Similar to actin dynamics, both microtubule stabilizing and destabilizing proteins play a role in neurite initiation. Furthermore, interactions between the actin and microtubule cytoskeleton are required for neurite formation. Several lines of evidence indicate that the interactions between these two components of the cytoskeleton are needed for force generation and for the localization of microtubules at sites of nascent neurites. The general theme that emerges is the existence of several central regulatory pathways on which extracellular cues converge to control and organize both actin and microtubules to induce the formation of neurites.
Collapse
|
30
|
Martín-García R, Coll PM, Pérez P. F-BAR domain protein Rga7 collaborates with Cdc15 and Imp2 to ensure proper cytokinesis in fission yeast. J Cell Sci 2014; 127:4146-58. [PMID: 25052092 DOI: 10.1242/jcs.146233] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
F-BAR domain proteins act as linkers between the cell cortex and cytoskeleton, and are involved in membrane binding and bending. Rga7 is one of the seven F-BAR proteins present in the fission yeast Schizosaccharomyces pombe. In addition to the F-BAR domain in the N-terminal region, Rga7 possesses a Rho GTPase-activating protein (GAP) domain at its C-terminus. We show here that Rga7 is necessary to prevent fragmentation of the contracting ring and incorrect septum synthesis. Accordingly, cultures of cells lacking Rga7 contain a higher percentage of dividing cells and more frequent asymmetric or aberrant septa, which ultimately might cause cell death. The Rga7 F-BAR domain is necessary for the protein localization to the division site and to the cell tips, and also for the Rga7 roles in cytokinesis. In contrast, Rga7 GAP catalytic activity seems to be dispensable. Moreover, we demonstrate that Rga7 cooperates with the two F-BAR proteins Cdc15 and Imp2 to ensure proper cytokinesis. We have also detected association of Rga7 with Imp2, and its binding partners Fic1 and Pxl1. Taken together, our findings suggest that Rga7 forms part of a protein complex that coordinates the late stages of cytokinesis.
Collapse
Affiliation(s)
- Rebeca Martín-García
- Instituto de Biología Funcional y Genómica, Consejo Superior de Investigaciones Científicas/Universidad de Salamanca, 37007 Salamanca, Spain
| | - Pedro M Coll
- Instituto de Biología Funcional y Genómica, Consejo Superior de Investigaciones Científicas/Universidad de Salamanca, 37007 Salamanca, Spain
| | - Pilar Pérez
- Instituto de Biología Funcional y Genómica, Consejo Superior de Investigaciones Científicas/Universidad de Salamanca, 37007 Salamanca, Spain
| |
Collapse
|
31
|
Ebsen H, Lettau M, Kabelitz D, Janssen O. Identification of SH3 domain proteins interacting with the cytoplasmic tail of the a disintegrin and metalloprotease 10 (ADAM10). PLoS One 2014; 9:e102899. [PMID: 25036101 PMCID: PMC4103893 DOI: 10.1371/journal.pone.0102899] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Accepted: 06/25/2014] [Indexed: 11/19/2022] Open
Abstract
The a disintegrin and metalloproteases (ADAMs) play a pivotal role in the control of development, adhesion, migration, inflammation and cancer. Although numerous substrates of ADAM10 have been identified, the regulation of its surface expression and proteolytic activity is still poorly defined. One current hypothesis is that both processes are in part modulated by protein-protein interactions mediated by the intracellular portion of the protease. For related proteases, especially proline-rich regions serving as docking sites for Src homology domain 3 (SH3) domain-containing proteins proved to be important for mediating regulatory interactions. In order to identify ADAM10-binding SH3 domain proteins, we screened the All SH3 Domain Phager library comprising 305 human SH3 domains using a GST fusion protein with the intracellular region of human ADAM10 as a bait for selection. Of a total of 291 analyzed phage clones, we found 38 SH3 domains that were precipitated with the ADAM10-derived fusion protein but not with GST. We verified the binding to the cytosolic portion of ADAM10 for several candidates by co-immunoprecipitation and/or pull down analyses. Intriguingly, several of the identified proteins have been implicated in regulating surface appearance and/or proteolytic activity of related ADAMs. Thus, it seems likely that they also play a role in ADAM10 biology.
Collapse
Affiliation(s)
- Henriette Ebsen
- University of Kiel, Molecular Immunology, Institute for Immunology, University Hospital Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Marcus Lettau
- University of Kiel, Molecular Immunology, Institute for Immunology, University Hospital Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Dieter Kabelitz
- University of Kiel, Molecular Immunology, Institute for Immunology, University Hospital Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Ottmar Janssen
- University of Kiel, Molecular Immunology, Institute for Immunology, University Hospital Schleswig-Holstein Campus Kiel, Kiel, Germany
- * E-mail:
| |
Collapse
|
32
|
Muñoz S, Manjón E, Sánchez Y. The putative exchange factor Gef3p interacts with Rho3p GTPase and the septin ring during cytokinesis in fission yeast. J Biol Chem 2014; 289:21995-2007. [PMID: 24947517 DOI: 10.1074/jbc.m114.548792] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The small GTP-binding proteins of the Rho family and its regulatory proteins play a central role in cytokinetic actomyosin ring assembly and cytokinesis. Here we show that the fission yeast guanine nucleotide exchange factor Gef3p interacts with Rho3p at the division site. Gef3p contains a putative DH homology domain and a BAR/IMD-like domain. The protein localized to the division site late in mitosis, where it formed a ring that did not constrict with actomyosin ring (cytokinetic actomyosin ring) invagination; instead, it split into a double ring that resembled the septin ring. Gef3p co-localized with septins and Mid2p and required septins and Mid2p for its localization. Gef3p interacts physically with the GTP-bound form of Rho3p. Although Gef3p is not essential for cell separation, the simultaneous disruption of gef3(+) and Rho3p-interacting proteins, such as Sec8p, an exocyst component, Apm1p, a subunit of the clathrin adaptor complex or For3p, an actin-polymerizing protein, yielded cells with strong defects in septation and polarity respectively. Our results suggest that interactions between septins and Rho-GEFs provide a new targeting mechanism for GTPases in cytokinesis, in this case probably contributing to Rho3p function in vesicle tethering and vesicle trafficking in the later steps of cell separation.
Collapse
Affiliation(s)
- Sofía Muñoz
- From the Instituto de Biología Funcional y Genómica (IBFG), Consejo Superior de Investigaciones Científicas and Departamento de Microbiología y Genética, Universidad de Salamanca, C/ Zacarías González, s/n. 37007 Salamanca, Spain
| | - Elvira Manjón
- From the Instituto de Biología Funcional y Genómica (IBFG), Consejo Superior de Investigaciones Científicas and Departamento de Microbiología y Genética, Universidad de Salamanca, C/ Zacarías González, s/n. 37007 Salamanca, Spain
| | - Yolanda Sánchez
- From the Instituto de Biología Funcional y Genómica (IBFG), Consejo Superior de Investigaciones Científicas and Departamento de Microbiología y Genética, Universidad de Salamanca, C/ Zacarías González, s/n. 37007 Salamanca, Spain
| |
Collapse
|
33
|
Mishra M, Huang J, Balasubramanian MK. The yeast actin cytoskeleton. FEMS Microbiol Rev 2014; 38:213-27. [PMID: 24467403 DOI: 10.1111/1574-6976.12064] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 01/18/2014] [Accepted: 01/20/2014] [Indexed: 11/29/2022] Open
Abstract
The actin cytoskeleton is a complex network of dynamic polymers, which plays an important role in various fundamental cellular processes, including maintenance of cell shape, polarity, cell division, cell migration, endocytosis, vesicular trafficking, and mechanosensation. Precise spatiotemporal assembly and disassembly of actin structures is regulated by the coordinated activity of about 100 highly conserved accessory proteins, which nucleate, elongate, cross-link, and sever actin filaments. Both in vivo studies in a wide range of organisms from yeast to metazoans and in vitro studies of purified proteins have helped shape the current understanding of actin dynamics and function. Molecular genetics, genome-wide functional analysis, sophisticated real-time imaging, and ultrastructural studies in concert with biochemical analysis have made yeast an attractive model to understand the actin cytoskeleton, its molecular dynamics, and physiological function. Studies of the yeast actin cytoskeleton have contributed substantially in defining the universal mechanism regulating actin assembly and disassembly in eukaryotes. Here, we review some of the important insights generated by the study of actin cytoskeleton in two important yeast models the budding yeast Saccharomyces cerevisiae and the fission yeast Schizosaccharomyces pombe.
Collapse
Affiliation(s)
- Mithilesh Mishra
- Temasek Life Sciences Laboratory, National University of Singapore, Singapore
| | | | | |
Collapse
|
34
|
Differential protein–protein interactions of full length human FasL and FasL fragments generated by proteolysis. Exp Cell Res 2014; 320:290-301. [DOI: 10.1016/j.yexcr.2013.11.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 10/25/2013] [Accepted: 11/19/2013] [Indexed: 01/14/2023]
|
35
|
Lam Hui K, Kwak SI, Upadhyaya A. Adhesion-dependent modulation of actin dynamics in Jurkat T cells. Cytoskeleton (Hoboken) 2013; 71:119-35. [DOI: 10.1002/cm.21156] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2013] [Revised: 10/21/2013] [Accepted: 10/29/2013] [Indexed: 01/06/2023]
Affiliation(s)
- King Lam Hui
- Department of Physics; University of Maryland; College Park Maryland 20742
| | - Sae In Kwak
- Department of Chemistry and Biochemistry; University of Maryland; College Park Maryland 20742
| | - Arpita Upadhyaya
- Department of Physics; University of Maryland; College Park Maryland 20742
- Institute for Physical Science and Technology, University of Maryland; College Park Maryland 20742
| |
Collapse
|
36
|
Quan A, Robinson PJ. Syndapin--a membrane remodelling and endocytic F-BAR protein. FEBS J 2013; 280:5198-212. [PMID: 23668323 DOI: 10.1111/febs.12343] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Revised: 05/07/2013] [Accepted: 05/08/2013] [Indexed: 12/17/2022]
Abstract
Syndapin [also called PACSIN (protein kinase C and casein kinase II interacting protein)] is an Fes-CIP4 homology Bin-amphiphysin-Rvs161/167 (F-BAR) and Src-homology 3 domain-containing protein. Three genes give rise to three main isoforms in mammalian cells. They each function in different endocytic and vesicle trafficking pathways and provide critical links between the cytoskeletal network in different cellular processes, such as neuronal morphogenesis and cell migration. The membrane remodelling activity of syndapin via its F-BAR domain and its interaction partners, such as dynamin and neural Wiskott-Aldrich syndrome protein binding to its Src-homology 3 domain, are important with respect to its function. Its various partner proteins provide insights into its mechanism of action, as well as its differential roles in these cellular processes. Signalling pathways leading to the regulation of syndapin function by phosphorylation are now contributing to our understanding of the broader functions of this family of proteins.
Collapse
Affiliation(s)
- Annie Quan
- Cell Signalling Unit, Children's Medical Research Institute, The University of Sydney, New South Wales, Australia
| | | |
Collapse
|
37
|
Saengsawang W, Taylor KL, Lumbard DC, Mitok K, Price A, Pietila L, Gomez TM, Dent EW. CIP4 coordinates with phospholipids and actin-associated proteins to localize to the protruding edge and produce actin ribs and veils. J Cell Sci 2013; 126:2411-23. [PMID: 23572514 DOI: 10.1242/jcs.117473] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Cdc42-interacting protein 4 (CIP4), a member of the F-BAR family of proteins, plays important roles in a variety of cellular events by regulating both membrane and actin dynamics. In many cell types, CIP4 functions in vesicle formation, endocytosis and membrane tubulation. However, recent data indicate that CIP4 is also involved in protrusion in some cell types, including cancer cells (lamellipodia and invadopodia) and neurons (ribbed lamellipodia and veils). In neurons, CIP4 localizes specifically to extending protrusions and functions to limit neurite outgrowth early in development. The mechanism by which CIP4 localizes to the protruding edge membrane and induces lamellipodial/veil protrusion and actin rib formation is not known. Here, we show that CIP4 localization to the protruding edge of neurons is dependent on both the phospholipid content of the plasma membrane and the underlying organization of actin filaments. Inhibiting phosphatidylinositol (3,4,5)-trisphosphate (PIP3) production decreases CIP4 at the membrane. CIP4 localization to the protruding edge is also dependent on Rac1/WAVE1, rather than Cdc42/N-WASP. Capping actin filaments with low concentrations of cytochalasin D or by overexpressing capping protein dramatically decreases CIP4 at the protruding edge, whereas inactivating Arp2/3 drives CIP4 to the protruding edge. We also demonstrate that CIP4 dynamically colocalizes with Ena/VASP and DAAM1, two proteins known to induce unbranched actin filament arrays and play important roles in neuronal development. Together, this is the first study to show that the localization of an F-BAR protein depends on both actin filament architecture and phospholipids at the protruding edge of developing neurons.
Collapse
Affiliation(s)
- Witchuda Saengsawang
- University of Wisconsin-Madison, Department of Neuroscience, Madison, WI 53706, USA
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Kim JH, Lee YG, Yoo S, Oh J, Jeong D, Song WK, Yoo BC, Rhee MH, Park J, Cha SH, Hong S, Cho JY. Involvement of Src and the actin cytoskeleton in the antitumorigenic action of adenosine dialdehyde. Biochem Pharmacol 2013; 85:1042-56. [DOI: 10.1016/j.bcp.2013.01.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Revised: 12/28/2012] [Accepted: 01/18/2013] [Indexed: 01/06/2023]
|
39
|
Ma Y, Mi YJ, Dai YK, Fu HL, Cui DX, Jin WL. The inverse F-BAR domain protein srGAP2 acts through srGAP3 to modulate neuronal differentiation and neurite outgrowth of mouse neuroblastoma cells. PLoS One 2013; 8:e57865. [PMID: 23505444 PMCID: PMC3591447 DOI: 10.1371/journal.pone.0057865] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Accepted: 01/27/2013] [Indexed: 11/18/2022] Open
Abstract
The inverse F-BAR (IF-BAR) domain proteins srGAP1, srGAP2 and srGAP3 are implicated in neuronal development and may be linked to mental retardation, schizophrenia and seizure. A partially overlapping expression pattern and highly similar protein structures indicate a functional redundancy of srGAPs in neuronal development. Our previous study suggests that srGAP3 negatively regulates neuronal differentiation in a Rac1-dependent manner in mouse Neuro2a cells. Here we show that exogenously expressed srGAP1 and srGAP2 are sufficient to inhibit valporic acid (VPA)-induced neurite initiation and growth in the mouse Neuro2a cells. While ectopic- or over-expression of RhoGAP-defective mutants, srGAP1R542A and srGAP2R527A exert a visible inhibitory effect on neuronal differentiation. Unexpectedly, knockdown of endogenous srGAP2 fails to facilitate the neuronal differentiation induced by VPA, but promotes neurite outgrowth of differentiated cells. All three IF-BAR domains from srGAP1-3 can induce filopodia formation in Neuro2a, but the isolated IF-BAR domain from srGAP2, not from srGAP1 and srGAP3, can promote VPA-induced neurite initiation and neuronal differentiation. We identify biochemical and functional interactions of the three srGAPs family members. We propose that srGAP3-Rac1 signaling may be required for the effect of srGAP1 and srGAP2 on attenuating neuronal differentiation. Furthermore, inhibition of Slit-Robo interaction can phenocopy a loss-of-function of srGAP3, indicating that srGAP3 may be dedicated to the Slit-Robo pathway. Our results demonstrate the interplay between srGAP1, srGAP2 and srGAP3 regulates neuronal differentiation and neurite outgrowth. These findings may provide us new insights into the possible roles of srGAPs in neuronal development and a potential mechanism for neurodevelopmental diseases.
Collapse
Affiliation(s)
- Yue Ma
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, People's Republic of China
- Department of Bio-Nano-Science and Engineering, Institute of Micro-Nano Science and Technology, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Ya-Jing Mi
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, People's Republic of China
- Lab of Cell Biology & Translational Medicine, Xi'an Medical University, Xi'an, People's Republic of China
| | - Yun-Kai Dai
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Hua-Lin Fu
- Department of Bio-Nano-Science and Engineering, Institute of Micro-Nano Science and Technology, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Da-Xiang Cui
- Department of Bio-Nano-Science and Engineering, Institute of Micro-Nano Science and Technology, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Wei-Lin Jin
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, People's Republic of China
- Department of Bio-Nano-Science and Engineering, Institute of Micro-Nano Science and Technology, Shanghai Jiao Tong University, Shanghai, People's Republic of China
- * E-mail:
| |
Collapse
|
40
|
Meitinger F, Palani S, Hub B, Pereira G. Dual function of the NDR-kinase Dbf2 in the regulation of the F-BAR protein Hof1 during cytokinesis. Mol Biol Cell 2013; 24:1290-304. [PMID: 23447700 PMCID: PMC3639042 DOI: 10.1091/mbc.e12-08-0608] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
The conserved NDR-kinase Dbf2 plays a critical role in cytokinesis in budding yeast. Among its cytokinesis-related substrates is the F-BAR protein Hof1. Hof1 colocalizes at the cell division site with the septin complex and, as mitotic exit progresses, moves to the actomyosin ring (AMR). Neither the function of Hof1 at the septin complex nor the mechanism by which Hof1 supports AMR constriction is understood. Here we establish that Dbf2 has a dual function in Hof1 regulation. First, we show that the coiled-coil region, which is adjacent to the conserved F-BAR domain, is required for the binding of Hof1 to septins. The Dbf2-dependent phosphorylation of Hof1 at a single serine residue (serine 313) in this region diminishes the recruitment of Hof1 to septins both in vitro and in vivo. Genetic and functional analysis indicates that the binding of Hof1 to septins is important for septin rearrangement and integrity during cytokinesis. Furthermore, Dbf2 phosphorylation of Hof1 at serines 533 and 563 promotes AMR constriction most likely by inhibiting the SH3-domain-dependent interactions of Hof1. Thus our data show that Dbf2 coordinates septin and AMR functions during cytokinesis through the regulation/control of Hof1.
Collapse
Affiliation(s)
- Franz Meitinger
- Molecular Biology of Centrosomes and Cilia Unit, DKFZ-ZMBH Alliance, 69120 Heidelberg, Germany
| | | | | | | |
Collapse
|
41
|
Cadherins and their partners in the nematode worm Caenorhabditis elegans. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2013; 116:239-62. [PMID: 23481198 DOI: 10.1016/b978-0-12-394311-8.00011-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The extreme simplicity of Caenorhabditis elegans makes it an ideal system to study the basic principles of cadherin function at the level of single cells within the physiologically relevant context of a developing animal. The genetic tractability of C. elegans also means that components of cadherin complexes can be identified through genetic modifier screens, allowing a comprehensive in vivo characterization of the macromolecular assemblies involved in cadherin function during tissue formation and maintenance in C. elegans. This work shows that a single cadherin system, the classical cadherin-catenin complex, is essential for diverse morphogenetic events during embryogenesis through its interactions with a range of mostly conserved proteins that act to modulate its function. The role of other members of the cadherin family in C. elegans, including members of the Fat-like, Flamingo/CELSR and calsyntenin families is less well characterized, but they have clear roles in neuronal development and function.
Collapse
|
42
|
Huang L, Poke G, Gecz J, Gibson K. A novel contiguous gene deletion of AVPR2 and ARHGAP4 genes in male dizygotic twins with nephrogenic diabetes insipidus and intellectual disability. Am J Med Genet A 2012; 158A:2511-8. [PMID: 22965914 DOI: 10.1002/ajmg.a.35591] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2012] [Accepted: 06/28/2012] [Indexed: 11/08/2022]
Abstract
The clinical features of loss of ARHGAP4 function remain unclear despite several reports of different patterns of deletions inactivating different functional regions of the protein. The protein encoded by ARHGAP4 is thought to function as a Rho GTPase activating protein. Characterization of the genetic defect causing X-linked nephrogenic diabetes insipidus (NDI) and intellectual disability in two dizygotic twin brothers revealed a novel contiguous deletion of 17,905 bp encompassing the entire AVPR2 gene and extending into intron 7 of the ARHGAP4 gene. Examination of their mother showed that she was a carrier of this deletion. An attempt was made to distinguish the putative clinical signs of an ARHGAP4 deletion from the well-defined phenotype of X-linked NDI caused by an AVPR2 gene deletion. By reviewing all characterized deletions encompassing ARHGAP4, we reconsider the potential role of ARHGAP4 in cognition.
Collapse
Affiliation(s)
- Lingli Huang
- Institute of Reproductive and Stem Cell Engineering, Central South University, China
| | | | | | | |
Collapse
|
43
|
Loveless T, Hardin J. Cadherin complexity: recent insights into cadherin superfamily function in C. elegans. Curr Opin Cell Biol 2012; 24:695-701. [PMID: 22819515 DOI: 10.1016/j.ceb.2012.06.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Revised: 06/06/2012] [Accepted: 06/28/2012] [Indexed: 10/28/2022]
Abstract
Cadherin superfamily proteins mediate cell-cell adhesion during development. The C. elegans embryo is a powerful system for analyzing how cadherins function in highly stereotyped morphogenetic events. In the embryo, the classical cadherin HMR-1 acts along with the Rac pathway and SAX-7/L1CAM during gastrulation. As adherens junctions mature, PAR complex proteins differentially regulate cadherin complex localization, and SRGP-1/Slit/Robo GAP aids adhesion by promoting membrane bending. Once adherens junctions form, actin is linked to the cell surface via HMP-1/α-catenin, whose actin binding activity is regulated in novel ways. FMI-1/Flamingo and CDH-4/Fat-like regulate axonal morphology of both pioneer and follower neurons. C. elegans thus continues to be useful for uncovering precise functions for cadherin superfamily proteins and their associates in a simple metazoan.
Collapse
Affiliation(s)
- Timothy Loveless
- Cellular and Molecular Biology Program, University of Wisconsin-Madison, Madison, WI 53706, USA
| | | |
Collapse
|
44
|
Mishra M, Huang Y, Srivastava P, Srinivasan R, Sevugan M, Shlomovitz R, Gov N, Rao M, Balasubramanian M. Cylindrical cellular geometry ensures fidelity of division site placement in fission yeast. J Cell Sci 2012; 125:3850-7. [PMID: 22505610 DOI: 10.1242/jcs.103788] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Successful cytokinesis requires proper assembly of the contractile actomyosin ring, its stable positioning on the cell surface and proper constriction. Over the years, many of the key molecular components and regulators of the assembly and positioning of the actomyosin ring have been elucidated. Here we show that cell geometry and mechanics play a crucial role in the stable positioning and uniform constriction of the contractile ring. Contractile rings that assemble in locally spherical regions of cells are unstable and slip towards the poles. By contrast, actomyosin rings that assemble on locally cylindrical portions of the cell under the same conditions do not slip, but uniformly constrict the cell surface. The stability of the rings and the dynamics of ring slippage can be described by a simple mechanical model. Using fluorescence imaging, we verify some of the quantitative predictions of the model. Our study reveals an intimate interplay between geometry and actomyosin dynamics, which are likely to apply in a variety of cellular contexts.
Collapse
|
45
|
Oh E, Robinson I. Barfly: sculpting membranes at the Drosophila neuromuscular junction. Dev Neurobiol 2012; 72:33-56. [PMID: 21630471 DOI: 10.1002/dneu.20923] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The ability of a cell to change the shape of its membranes is intrinsic to many cellular functions. Proteins that can alter or recognize curved membrane structures and those that can act to recruit other proteins which stabilize the membrane curvature are likely to be essential in cell functions. The BAR (Bin, amphiphysin, RVS167 homology) domain is a protein domain that can either induce lipidic membranes to curve or can sense curved membranes. BAR domains are found in several proteins at neuronal synapses. We will review BAR domain structure and the role that BAR domain containing proteins play in regulating the morphology and function of the Drosophila neuromuscular junction. In flies the BAR domain containing proteins, endophilin and syndapin affect synaptic vesicle endocytosis, whereas CIP4, dRich, nervous wreck and syndapin affect synaptic morphology. We will review the growing evidence implicating mutations in BAR domain containing proteins being the cause of human pathologies.
Collapse
Affiliation(s)
- Eugene Oh
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | | |
Collapse
|
46
|
The BAR Domain Superfamily Proteins from Subcellular Structures to Human Diseases. MEMBRANES 2012; 2:91-117. [PMID: 24957964 PMCID: PMC4021885 DOI: 10.3390/membranes2010091] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2012] [Revised: 02/07/2012] [Accepted: 02/15/2012] [Indexed: 12/11/2022]
Abstract
Eukaryotic cells have complicated membrane systems. The outermost plasma membrane contains various substructures, such as invaginations and protrusions, which are involved in endocytosis and cell migration. Moreover, the intracellular membrane compartments, such as autophagosomes and endosomes, are essential for cellular viability. The Bin-Amphiphysin-Rvs167 (BAR) domain superfamily proteins are important players in membrane remodeling through their structurally determined membrane binding surfaces. A variety of BAR domain superfamily proteins exist, and each family member appears to be involved in the formation of certain subcellular structures or intracellular membrane compartments. Most of the BAR domain superfamily proteins contain SH3 domains, which bind to the membrane scission molecule, dynamin, as well as the actin regulatory WASP/WAVE proteins and several signal transduction molecules, providing possible links between the membrane and the cytoskeleton or other machineries. In this review, we summarize the current information about each BAR superfamily protein with an SH3 domain(s). The involvement of BAR domain superfamily proteins in various diseases is also discussed.
Collapse
|
47
|
The F-BAR protein CIP4 inhibits neurite formation by producing lamellipodial protrusions. Curr Biol 2012; 22:494-501. [PMID: 22361215 DOI: 10.1016/j.cub.2012.01.038] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2011] [Revised: 12/22/2011] [Accepted: 01/19/2012] [Indexed: 02/01/2023]
Abstract
Neurite formation is a seminal event in the early development of neurons. However, little is known about the mechanisms by which neurons form neurites. F-BAR proteins function in sensing and inducing membrane curvature. Cdc42-interacting protein 4 (CIP4), a member of the F-BAR family, regulates endocytosis in a variety of cell types. However, there is little data on how CIP4 functions in neurons. Here we show that CIP4 plays a novel role in neuronal development by inhibiting neurite formation. Remarkably, CIP4 exerts this effect not through endocytosis, but by producing lamellipodial protrusions. In primary cortical neurons CIP4 is concentrated specifically at the tips of extending lamellipodia and filopodia, instead of endosomes as in other cell types. Overexpression of CIP4 results in lamellipodial protrusions around the cell body, subsequently delaying neurite formation and enlarging growth cones. These effects depend on the F-BAR and SH3 domains of CIP4 and on its ability to multimerize. Conversely, cortical neurons from CIP4-null mice initiate neurites twice as fast as controls. This is the first study to demonstrate that an F-BAR protein functions differently in neuronal versus nonneuronal cells and induces lamellipodial protrusions instead of invaginations or filopodia-like structures.
Collapse
|
48
|
Palani S, Meitinger F, Boehm ME, Lehmann WD, Pereira G. Cdc14-dependent dephosphorylation of Inn1 contributes to Inn1-Cyk3 complex formation. J Cell Sci 2012; 125:3091-6. [DOI: 10.1242/jcs.106021] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
In Saccharomyces cerevisiae, the Cdc14 phosphatase plays a well-established role in reverting phosphorylation events on substrates of the mitotic cyclin-dependent kinase (M-Cdk1), thereby promoting mitotic exit and down-regulation of M-Cdk1 activity. Cdc14 localizes at the site of cell cleavage after M-Cdk1 inactivation, suggesting that Cdc14 may perform a critical, yet ill-defined, role during cytokinesis. Here, we identified Inn1, as a novel direct substrate of both M-Cdk1 and Cdc14. Cdc14 co-localizes with Inn1 at the cell division site and interacts with the C-terminal proline rich domain of Inn1 that mediates its binding to the SH3-domain containing proteins Hof1 and Cyk3. We show that phosphorylation of Inn1 by Cdk1 partially perturbs the interaction of Inn1 with Cyk3 thereby reducing the levels of Cyk3 at the cell division site. We propose that Cdc14 counteracts Cdk1 phosphorylation of Inn1 to facilitate Inn1-Cyk3 complex formation and so promote cytokinesis.
Collapse
|
49
|
Endris V, Haussmann L, Buss E, Bacon C, Bartsch D, Rappold G. SrGAP3 interacts with lamellipodin at the cell membrane and regulates Rac-dependent cellular protrusions. J Cell Sci 2011; 124:3941-55. [PMID: 22159416 DOI: 10.1242/jcs.077081] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
SrGAP3/MEGAP is a member of the Slit-Robo GAP (srGAP) family and is implicated in repulsive axon guidance and neuronal migration through Slit-Robo-mediated signal transduction. Here we describe an inhibitory role of srGAP3 on actin dynamics, specifically on lamellipodia formation. We show that the F-BAR domain localizes srGAP3 to the leading edge of cellular protrusions whereas the SH3 domain is important for focal adhesion targeting. We report on a novel srGAP3 interaction partner, lamellipodin, which localizes with srGAP3 at the leading edge. Live-cell analyses revealed that srGAP3 influences lamellipodin-evoked lamellipodial dynamics. Furthermore, we show that mouse embryonic fibroblasts derived from homozygous srGAP3-knockout embryos display an increased cell area and lamellipodia formation that can be blocked by shRNA-mediated knockdown of lamellipodin.
Collapse
Affiliation(s)
- Volker Endris
- Department of Human Molecular Genetics, University of Heidelberg, Germany
| | | | | | | | | | | |
Collapse
|
50
|
Baumgart T, Capraro BR, Zhu C, Das SL. Thermodynamics and mechanics of membrane curvature generation and sensing by proteins and lipids. Annu Rev Phys Chem 2011; 62:483-506. [PMID: 21219150 DOI: 10.1146/annurev.physchem.012809.103450] [Citation(s) in RCA: 272] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Research investigating lipid membrane curvature generation and sensing is a rapidly developing frontier in membrane physical chemistry and biophysics. The fast recent progress is based on the discovery of a plethora of proteins involved in coupling membrane shape to cellular membrane function, the design of new quantitative experimental techniques to study aspects of membrane curvature, and the development of analytical theories and simulation techniques that allow a mechanistic interpretation of quantitative measurements. The present review first provides an overview of important classes of membrane proteins for which function is coupled to membrane curvature. We then survey several mechanisms that are assumed to underlie membrane curvature sensing and generation. Finally, we discuss relatively simple thermodynamic/mechanical models that allow quantitative interpretation of experimental observations.
Collapse
Affiliation(s)
- Tobias Baumgart
- Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.
| | | | | | | |
Collapse
|