1
|
Huang YF, Wei YL, Wang SM, Yang PY, Hsieh PL, Yeh JC, Liao YW, Yu CC, Kuo WY. NCK1 antisense RNA 1 (NCK1-AS1) exerts pro-fibrosis property in oral mucosa through modulation of miR-137/NCK1 axis. J Dent Sci 2025; 20:632-638. [PMID: 39873098 PMCID: PMC11762667 DOI: 10.1016/j.jds.2024.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 11/01/2024] [Indexed: 01/30/2025] Open
Abstract
Background/Purpose Oral submucous fibrosis (OSF) is a premalignant condition of the oral cavity, and its pathogenesis remains largely unknown. A multitude of non-coding RNAs are aberrantly expressed in OSF, and their implication for the development of OSF is a matter meriting investigation. Materials and methods The functional role of long non-coding RNA NCK1-AS1 in myofibroblast activation of fibrotic buccal mucosal fibroblasts (fBMFs) derived from OSF tissues was assessed. Wound healing, collagen gel contraction and transwell migration assays have been employed to assess the myofibroblast activities. In addition, a luciferase-based reporter assay was used to illustrate the potential mechanism underlying the regulation of NCK1-AS1 in myofibroblast activation. Results Silencing of NCK1-AS1 markedly downregulated myofibroblast activation and the expression of fibrosis markers in fBMFs. Besides, we demonstrated that NCK1-AS1 directly interacted with microRNA-137 (miR-137) and was negatively correlated with it. Moreover, we found that NCK1 was a target of miR-137 and positively related to NCK1-AS1. Our results demonstrated that NCK1-AS1 may regulate myofibroblast activation by suppressing miR-137 and upregulating NCK1. Conclusion We showed that NCK1-AS1 acted as a sponge of miR-137 and titrated the suppressive effect of miR-137 on NCK1 to modulate myofibroblast activation in OSF condition.
Collapse
Affiliation(s)
- Yu-Feng Huang
- School of Dentistry, Chung Shan Medical University, Taichung, Taiwan
- Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Yu-Lei Wei
- School of Dentistry, Chung Shan Medical University, Taichung, Taiwan
- Institute of Oral Sciences, Chung Shan Medical University, Taichung, Taiwan
| | - Shih-Min Wang
- School of Dentistry, Chung Shan Medical University, Taichung, Taiwan
- Institute of Oral Sciences, Chung Shan Medical University, Taichung, Taiwan
| | - Po-Yu Yang
- School of Dentistry, Chung Shan Medical University, Taichung, Taiwan
- Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Pei-Ling Hsieh
- Department of Anatomy, School of Medicine, China Medical University, Taichung, Taiwan
| | - Jung-Chun Yeh
- School of Dentistry, Chung Shan Medical University, Taichung, Taiwan
- Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Yi-Wen Liao
- Institute of Oral Sciences, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Cheng-Chia Yu
- School of Dentistry, Chung Shan Medical University, Taichung, Taiwan
- Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan
- Institute of Oral Sciences, Chung Shan Medical University, Taichung, Taiwan
| | - Wan-Yin Kuo
- Department of Emergency Medicine, Chi Mei Medical Center, Tainan, Taiwan
- Department of Occupational Medicine, Chi Mei Medical Center, Tainan, Taiwan
| |
Collapse
|
2
|
Fülle JB, de Almeida RA, Lawless C, Stockdale L, Yanes B, Lane EB, Garrod DR, Ballestrem C. Proximity Mapping of Desmosomes Reveals a Striking Shift in Their Molecular Neighborhood Associated With Maturation. Mol Cell Proteomics 2024; 23:100735. [PMID: 38342409 PMCID: PMC10943070 DOI: 10.1016/j.mcpro.2024.100735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 01/29/2024] [Accepted: 02/08/2024] [Indexed: 02/13/2024] Open
Abstract
Desmosomes are multiprotein adhesion complexes that link intermediate filaments to the plasma membrane, ensuring the mechanical integrity of cells across tissues, but how they participate in the wider signaling network to exert their full function is unclear. To investigate this, we carried out protein proximity mapping using biotinylation (BioID). The combined interactomes of the essential desmosomal proteins desmocollin 2a, plakoglobin, and plakophilin 2a (Pkp2a) in Madin-Darby canine kidney epithelial cells were mapped and their differences and commonalities characterized as desmosome matured from Ca2+ dependence to the mature, Ca2+-independent, hyper-adhesive state, which predominates in tissues. Results suggest that individual desmosomal proteins have distinct roles in connecting to cellular signaling pathways and that these roles alter substantially when cells change their adhesion state. The data provide further support for a dualistic concept of desmosomes in which the properties of Pkp2a differ from those of the other, more stable proteins. This body of data provides an invaluable resource for the analysis of desmosome function.
Collapse
Affiliation(s)
- Judith B Fülle
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, UK
| | | | - Craig Lawless
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, UK
| | - Liam Stockdale
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, UK
| | - Bian Yanes
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, UK
| | - E Birgitte Lane
- Skin Research Institute of Singapore, Agency of Science Technology and Research (A∗STAR), Singapore, Singapore
| | - David R Garrod
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, UK.
| | - Christoph Ballestrem
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, UK.
| |
Collapse
|
3
|
Kumar M, Michael S, Alvarado-Valverde J, Zeke A, Lazar T, Glavina J, Nagy-Kanta E, Donagh J, Kalman Z, Pascarelli S, Palopoli N, Dobson L, Suarez C, Van Roey K, Krystkowiak I, Griffin J, Nagpal A, Bhardwaj R, Diella F, Mészáros B, Dean K, Davey N, Pancsa R, Chemes L, Gibson T. ELM-the Eukaryotic Linear Motif resource-2024 update. Nucleic Acids Res 2024; 52:D442-D455. [PMID: 37962385 PMCID: PMC10767929 DOI: 10.1093/nar/gkad1058] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/22/2023] [Accepted: 10/24/2023] [Indexed: 11/15/2023] Open
Abstract
Short Linear Motifs (SLiMs) are the smallest structural and functional components of modular eukaryotic proteins. They are also the most abundant, especially when considering post-translational modifications. As well as being found throughout the cell as part of regulatory processes, SLiMs are extensively mimicked by intracellular pathogens. At the heart of the Eukaryotic Linear Motif (ELM) Resource is a representative (not comprehensive) database. The ELM entries are created by a growing community of skilled annotators and provide an introduction to linear motif functionality for biomedical researchers. The 2024 ELM update includes 346 novel motif instances in areas ranging from innate immunity to both protein and RNA degradation systems. In total, 39 classes of newly annotated motifs have been added, and another 17 existing entries have been updated in the database. The 2024 ELM release now includes 356 motif classes incorporating 4283 individual motif instances manually curated from 4274 scientific publications and including >700 links to experimentally determined 3D structures. In a recent development, the InterPro protein module resource now also includes ELM data. ELM is available at: http://elm.eu.org.
Collapse
Affiliation(s)
- Manjeet Kumar
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg 69117, Germany
| | - Sushama Michael
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg 69117, Germany
| | - Jesús Alvarado-Valverde
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg 69117, Germany
- Collaboration for joint PhD degree between EMBL and Heidelberg University, Faculty of Biosciences, Germany
| | - András Zeke
- Institute of Enzymology, HUN-REN Research Centre for Natural Sciences, Budapest 1117, Hungary
| | - Tamas Lazar
- VIB-VUB Center for Structural Biology, Vlaams Instituut voor Biotechnologie, Pleinlaan 2, 1050 Brussels, Belgium
- Structural Biology Brussels, Department of Bioengineering, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium
| | - Juliana Glavina
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín (UNSAM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), CP 1650, Buenos Aires, Argentina
- Escuela de Bio y Nanotecnologías (EByN), Universidad Nacional de San Martín, Av. 25 de Mayo y Francia, CP1650 San Martín, Buenos Aires, Argentina
| | - Eszter Nagy-Kanta
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Práter u. 50/A, Budapest 1083, Hungary
| | - Juan Mac Donagh
- Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Bernal, Buenos Aires, Argentina
| | - Zsofia E Kalman
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg 69117, Germany
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Práter u. 50/A, Budapest 1083, Hungary
| | - Stefano Pascarelli
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Nicolas Palopoli
- Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Bernal, Buenos Aires, Argentina
| | - László Dobson
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg 69117, Germany
- Department of Bioinformatics, Semmelweis University, Tűzoltó u. 7, Budapest 1094, Hungary
| | - Carmen Florencia Suarez
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín (UNSAM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), CP 1650, Buenos Aires, Argentina
- Escuela de Bio y Nanotecnologías (EByN), Universidad Nacional de San Martín, Av. 25 de Mayo y Francia, CP1650 San Martín, Buenos Aires, Argentina
| | - Kim Van Roey
- Health Services Research, Sciensano, Brussels, Belgium
| | - Izabella Krystkowiak
- Institute of Cancer Research, Chester Beatty Laboratories, 237 Fulham Rd, Chelsea, London SW3 6JB, UK
| | - Juan Esteban Griffin
- Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Bernal, Buenos Aires, Argentina
| | - Anurag Nagpal
- Department of Biological Sciences, BITS Pilani, K. K. Birla Goa campus, Zuarinagar, Goa 403726, India
| | - Rajesh Bhardwaj
- Inselspital, University of Bern, Freiburgstrasse 15, CH-3010 Bern, Switzerland
| | - Francesca Diella
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg 69117, Germany
| | - Bálint Mészáros
- Department of Structural Biology and Center of Excellence for Data Driven Discovery, St Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Kellie Dean
- School of Biochemistry and Cell Biology, 3.91 Western Gateway Building, University College Cork, Cork, Ireland
| | - Norman E Davey
- Institute of Cancer Research, Chester Beatty Laboratories, 237 Fulham Rd, Chelsea, London SW3 6JB, UK
| | - Rita Pancsa
- Institute of Enzymology, HUN-REN Research Centre for Natural Sciences, Budapest 1117, Hungary
| | - Lucía B Chemes
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín (UNSAM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), CP 1650, Buenos Aires, Argentina
- Escuela de Bio y Nanotecnologías (EByN), Universidad Nacional de San Martín, Av. 25 de Mayo y Francia, CP1650 San Martín, Buenos Aires, Argentina
| | - Toby J Gibson
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg 69117, Germany
| |
Collapse
|
4
|
Golding AP, Ferrier B, New LA, Lu P, Martin CE, Shata E, Jones RA, Moorehead RA, Jones N. Distinct Requirements for Adaptor Proteins NCK1 and NCK2 in Mammary Gland Development. J Mammary Gland Biol Neoplasia 2023; 28:19. [PMID: 37479911 PMCID: PMC10361900 DOI: 10.1007/s10911-023-09541-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 06/20/2023] [Indexed: 07/23/2023] Open
Abstract
The adaptor proteins NCK1 and NCK2 are well-established signalling nodes that regulate diverse biological processes including cell proliferation and actin dynamics in many tissue types. Here we have investigated the distribution and function of Nck1 and Nck2 in the developing mouse mammary gland. Using publicly available single-cell RNA sequencing data, we uncovered distinct expression profiles between the two paralogs. Nck1 showed widespread expression in luminal, basal, stromal and endothelial cells, while Nck2 was restricted to luminal and basal cells, with prominent enrichment in hormone-sensing luminal subtypes. Next, using mice with global knockout of Nck1 or Nck2, we assessed mammary gland development during and after puberty (5, 8 and 12 weeks of age). Mice lacking Nck1 or Nck2 displayed significant defects in ductal outgrowth and branching at 5 weeks compared to controls, and the defects persisted in Nck2 knockout mice at 8 weeks before normalizing at 12 weeks. These defects were accompanied by an increase in epithelial cell proliferation at 5 weeks and a decrease at 8 weeks in both Nck1 and Nck2 knockout mice. We also profiled expression of several key genes associated with mammary gland development at these timepoints and detected temporal changes in transcript levels of hormone receptors as well as effectors of cell proliferation and migration in Nck1 and Nck2 knockout mice, in line with the distinct phenotypes observed at 5 and 8 weeks. Together these studies reveal a requirement for NCK proteins in mammary gland morphogenesis, and suggest that deregulation of Nck expression could drive breast cancer progression and metastasis.
Collapse
Affiliation(s)
- Adam P Golding
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada
| | - Benjamin Ferrier
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada
| | - Laura A New
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada
| | - Peihua Lu
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada
| | - Claire E Martin
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada
- Present address: Lunenfeld-Tanenbaum Research Institute, Toronto, ON, Canada
| | - Erka Shata
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada
| | - Robert A Jones
- Department of Biomedical Sciences, University of Guelph, Guelph, ON, Canada
| | - Roger A Moorehead
- Department of Biomedical Sciences, University of Guelph, Guelph, ON, Canada
| | - Nina Jones
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada.
| |
Collapse
|
5
|
Diab AM, Wigerius M, Quinn DP, Qi J, Shahin I, Paffile J, Krueger K, Karten B, Krueger SR, Fawcett JP. NCK1 Modulates Neuronal Actin Dynamics and Promotes Dendritic Spine, Synapse, and Memory Formation. J Neurosci 2023; 43:885-901. [PMID: 36535770 PMCID: PMC9908320 DOI: 10.1523/jneurosci.0495-21.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 12/05/2022] [Accepted: 12/10/2022] [Indexed: 12/23/2022] Open
Abstract
Memory formation and maintenance is a dynamic process involving the modulation of the actin cytoskeleton at synapses. Understanding the signaling pathways that contribute to actin modulation is important for our understanding of synapse formation and function, as well as learning and memory. Here, we focused on the importance of the actin regulator, noncatalytic region of tyrosine kinase adaptor protein 1 (NCK1), in hippocampal dependent behaviors and development. We report that male mice lacking NCK1 have impairments in both short-term and working memory, as well as spatial learning. Additionally, we report sex differences in memory impairment showing that female mice deficient in NCK1 fail at reversal learning in a spatial learning task. We find that NCK1 is expressed in postmitotic neurons but is dispensable for neuronal proliferation and migration in the developing hippocampus. Morphologically, NCK1 is not necessary for overall neuronal dendrite development. However, neurons lacking NCK1 have lower dendritic spine and synapse densities in vitro and in vivo EM analysis reveal increased postsynaptic density (PSD) thickness in the hippocampal CA1 region of NCK1-deficient mice. Mechanistically, we find the turnover of actin-filaments in dendritic spines is accelerated in neurons that lack NCK1. Together, these findings suggest that NCK1 contributes to hippocampal-dependent memory by stabilizing actin dynamics and dendritic spine formation.SIGNIFICANCE STATEMENT Understanding the molecular signaling pathways that contribute to memory formation, maintenance, and elimination will lead to a better understanding of the genetic influences on cognition and cognitive disorders and will direct future therapeutics. Here, we report that the noncatalytic region of tyrosine kinase adaptor protein 1 (NCK1) adaptor protein modulates actin-filament turnover in hippocampal dendritic spines. Mice lacking NCK1 show sex-dependent deficits in hippocampal memory formation tasks, have altered postsynaptic densities, and reduced synaptic density. Together, our work implicates NCK1 in the regulation of actin cytoskeleton dynamics and normal synapse development which is essential for memory formation.
Collapse
Affiliation(s)
- Antonios M Diab
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Michael Wigerius
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Dylan P Quinn
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Jiansong Qi
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Ibrahim Shahin
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Julia Paffile
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Kavita Krueger
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Barbara Karten
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Stefan R Krueger
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - James P Fawcett
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
- Department of Surgery, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| |
Collapse
|
6
|
Zhou L, Wang T, Zhang K, Zhang X, Jiang S. The development of small-molecule inhibitors targeting HPK1. Eur J Med Chem 2022; 244:114819. [DOI: 10.1016/j.ejmech.2022.114819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 09/29/2022] [Accepted: 09/30/2022] [Indexed: 11/25/2022]
|
7
|
Ilovich O, Dines M, Paul BK, Barkai E, Lamprecht R. Nck1 activity in lateral amygdala regulates long-term fear memory formation. Transl Psychiatry 2022; 12:475. [PMID: 36371406 PMCID: PMC9653413 DOI: 10.1038/s41398-022-02244-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 10/23/2022] [Accepted: 10/28/2022] [Indexed: 11/13/2022] Open
Abstract
Fear conditioning leads to long-term fear memory formation and is a model for studying fear-related psychopathological conditions such as phobias and post-traumatic stress disorder. Long-term fear memory formation is believed to involve alterations of synaptic efficacy mediated by changes in synaptic transmission and morphology in lateral amygdala (LA). Nck1 is a key neuronal adaptor protein involved in the regulation of the actin cytoskeleton and the neuronal processes believed to be involved in memory formation. However, the role of Nck1 in memory formation is not known. Here we explored the role of Nck1 in fear memory formation in lateral amygdala (LA). Reduction of Nck1 in excitatory neurons in LA enhanced long-term, but not short-term, auditory fear conditioning memory. Activation of Nck1, by using a photoactivatable Nck1 (PA-Nck1), during auditory fear conditioning in excitatory neurons in LA impaired long-term, but not short-term, fear memory. Activation of Nck1 immediately or a day after fear conditioning did not affect fear memory. The hippocampal-mediated contextual fear memory was not affected by the reduction or activation of Nck1 in LA. We show that Nck1 is localized to the presynapses in LA. Nck1 activation in LA excitatory neurons decreased the frequency of AMPA receptors-mediated miniature excitatory synaptic currents (mEPSCs). Nck1 activation did not affect GABA receptor-mediated inhibitory synaptic currents (mIPSCs). These results show that Nck1 activity in excitatory neurons in LA regulates glutamate release and sets the threshold for fear memory formation. Moreover, our research shows that Nck1 may serve as a target for pharmacological treatment of fear and anxiety disorders.
Collapse
Affiliation(s)
- Or Ilovich
- grid.18098.380000 0004 1937 0562Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Monica Dines
- grid.18098.380000 0004 1937 0562Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Blesson K. Paul
- grid.18098.380000 0004 1937 0562Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Edi Barkai
- grid.18098.380000 0004 1937 0562Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Raphael Lamprecht
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel.
| |
Collapse
|
8
|
Borroto A, Alarcón B, Navarro MN. Mutation of the Polyproline Sequence in CD3ε Evidences TCR Signaling Requirements for Differentiation and Function of Pro-Inflammatory Tγδ17 Cells. Front Immunol 2022; 13:799919. [PMID: 35432331 PMCID: PMC9008450 DOI: 10.3389/fimmu.2022.799919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 03/09/2022] [Indexed: 11/23/2022] Open
Abstract
Tγδ17 cells have emerged as a key population in the development of inflammatory and autoimmune conditions such as psoriasis. Thus, the therapeutic intervention of Tγδ17 cells can exert protective effects in this type of pathologies. Tγδ cells commit to IL-17 production during thymus development, and upon immune challenge, additional extrathymic signals induce the differentiation of uncommitted Tγδ cells into Tγδ17 effector cells. Despite the interest in Tγδ17 cells during the past 20 years, the role of TCR signaling in the generation and function of Tγδ17 cells has not been completely elucidated. While some studies point to the notion that Tγδ17 differentiation requires weak or no TCR signaling, other works suggest that Tγδ17 require the participation of specific kinases and adaptor molecules downstream of the TCR. Here we have examined the differentiation and pathogenic function of Tγδ17 cells in “knockin” mice bearing conservative mutations in the CD3ε polyproline rich sequence (KI-PRS) with attenuated TCR signaling due to lack of binding of the essential adaptor Nck. KI-PRS mice presented decreased frequency and numbers of Tγδ17 cells in adult thymus and lymph nodes. In the Imiquimod model of skin inflammation, KI-PRS presented attenuated skin inflammation parameters compared to wild-type littermates. Moreover, the generation, expansion and effector function Tγδ17 cells were impaired in KI-PRS mice upon Imiquimod challenge. Thus, we conclude that an intact CD3ε-PRS sequence is required for optimal differentiation and pathogenic function of Tγδ17 cells. These data open new opportunities for therapeutic targeting of specific TCR downstream effectors for treatment of Tγδ17-mediated diseases.
Collapse
Affiliation(s)
- Aldo Borroto
- Interactions with the Environment Program, Centro Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
| | - Balbino Alarcón
- Interactions with the Environment Program, Centro Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
| | - María N Navarro
- Interactions with the Environment Program, Centro Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
9
|
Dok-1 regulates mast cell degranulation negatively through inhibiting calcium-dependent F-actin disassembly. Clin Immunol 2022; 238:109008. [PMID: 35421591 DOI: 10.1016/j.clim.2022.109008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 04/06/2022] [Accepted: 04/07/2022] [Indexed: 11/24/2022]
Abstract
In food allergies, antigen-induced aggregation of FcεRI on mast cells initiates highly ordered and sequential signaling events. Dok-1(downstream of tyrosine kinase 1), undergoes intense tyrosine phosphorylation upon FcεRI stimulation, which negatively regulates Ras/Erk signaling and the subsequent cytokine release, but it remains unclear whether Dok-1 regulates Fc-mediated degranulation. In this study, we investigated the role of Dok-1 in FcεRI-mediated degranulation. Dok-1 overexpressing RBL-2H3 cells were established. Degranulation, immunoprecipitation, co-immunoprecipitation, immunoblotting and flow cytometry assay were performed to explore the effects of Dok-1 and its underlying mechanisms. We found that, following FcεRI activation, Dok-1 was recruited to the plasma membrane, leading to tyrosine phosphorylation. Phosphorylated Dok-1 inhibits FcεRI-operated calcium influx, and negatively regulated degranulation by inhibiting calcium-dependent disassembly of actin filaments. Our data revealed that Dok-1 is a negative regulator of FcεRI-mediated mast cell degranulation. These findings contribute to the identification of therapeutic targets for food allergies.
Collapse
|
10
|
Paensuwan P, Ngoenkam J, Wangteeraprasert A, Pongcharoen S. Essential function of adaptor protein Nck1 in platelet-derived growth factor receptor signaling in human lens epithelial cells. Sci Rep 2022; 12:1063. [PMID: 35058548 PMCID: PMC8776929 DOI: 10.1038/s41598-022-05183-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 01/06/2022] [Indexed: 11/24/2022] Open
Abstract
Binding of platelet-derived growth factor-BB (PDGF-BB) to its cognate receptor (PDGFR) promotes lens epithelial cell (LEC) proliferation and migration. After cataract surgery, these LEC behaviors have been proposed as an influential cause of posterior capsule opacification (PCO). Stimulated PDFGR undergoes dimerization and tyrosine phosphorylation providing docking sites for a SH2-domain-containing noncatalytic region of tyrosine kinase (Nck). Nck is an adaptor protein acting as a linker of the proximal and downstream signaling events. However, the functions of Nck1 protein in LEC have not been investigated so far. We reported here a crucial role of Nck1 protein in regulating PDGFR-mediated LEC activation using LEC with a silenced expression of Nck1 protein. The knockdown of Nck1 suppressed PDGF-BB-stimulated LEC proliferation and migration and disrupted the cell cycle progression especially G1/S transition. LEC lacking Nck1 protein failed to exhibit actin polymerization and membrane protrusions. The downregulation of Nck1 protein in LEC impaired PDGFR‐induced phosphorylation of intracellular signaling proteins, including Erk1/2, Akt, CREB and ATF1, which resulted in inhibition of LEC responses. Therefore, these data suggest that the loss of Nck1 expression may disturb LEC activation and Nck1 may potentially be a drug target to prevent PCO and lens-related disease.
Collapse
Affiliation(s)
- Pussadee Paensuwan
- Department of Optometry, Faculty of Allied Health Sciences, Naresuan University, Tapho District, Phitsanulok, 65000, Thailand.
| | - Jatuporn Ngoenkam
- Department of Microbiology and Parasitology, Faculty of Medical Science, Naresuan University, Tapho District, Phitsanulok, 65000, Thailand
| | - Apirath Wangteeraprasert
- Department of Medicine, Faculty of Medicine, Naresuan University, Tapho District, Phitsanulok, 65000, Thailand
| | - Sutatip Pongcharoen
- Department of Medicine, Faculty of Medicine, Naresuan University, Tapho District, Phitsanulok, 65000, Thailand.
| |
Collapse
|
11
|
Kotian N, Troike KM, Curran KN, Lathia JD, McDonald JA. A Drosophila RNAi screen reveals conserved glioblastoma-related adhesion genes that regulate collective cell migration. G3 GENES|GENOMES|GENETICS 2022; 12:6388037. [PMID: 34849760 PMCID: PMC8728034 DOI: 10.1093/g3journal/jkab356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 10/06/2021] [Indexed: 11/14/2022]
Abstract
Abstract
Migrating cell collectives are key to embryonic development but also contribute to invasion and metastasis of a variety of cancers. Cell collectives can invade deep into tissues, leading to tumor progression and resistance to therapies. Collective cell invasion is also observed in the lethal brain tumor glioblastoma (GBM), which infiltrates the surrounding brain parenchyma leading to tumor growth and poor patient outcomes. Drosophila border cells, which migrate as a small cell cluster in the developing ovary, are a well-studied and genetically accessible model used to identify general mechanisms that control collective cell migration within native tissue environments. Most cell collectives remain cohesive through a variety of cell–cell adhesion proteins during their migration through tissues and organs. In this study, we first identified cell adhesion, cell matrix, cell junction, and associated regulatory genes that are expressed in human brain tumors. We performed RNAi knockdown of the Drosophila orthologs in border cells to evaluate if migration and/or cohesion of the cluster was impaired. From this screen, we identified eight adhesion-related genes that disrupted border cell collective migration upon RNAi knockdown. Bioinformatics analyses further demonstrated that subsets of the orthologous genes were elevated in the margin and invasive edge of human GBM patient tumors. These data together show that conserved cell adhesion and adhesion regulatory proteins with potential roles in tumor invasion also modulate collective cell migration. This dual screening approach for adhesion genes linked to GBM and border cell migration thus may reveal conserved mechanisms that drive collective tumor cell invasion.
Collapse
Affiliation(s)
- Nirupama Kotian
- Division of Biology, Kansas State University, Manhattan, KS 66506, USA
| | - Katie M Troike
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Kristen N Curran
- Division of Biology, Kansas State University, Manhattan, KS 66506, USA
| | - Justin D Lathia
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | | |
Collapse
|
12
|
Shah K, Al-Haidari A, Sun J, Kazi JU. T cell receptor (TCR) signaling in health and disease. Signal Transduct Target Ther 2021; 6:412. [PMID: 34897277 PMCID: PMC8666445 DOI: 10.1038/s41392-021-00823-w] [Citation(s) in RCA: 202] [Impact Index Per Article: 50.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 11/02/2021] [Accepted: 11/02/2021] [Indexed: 12/18/2022] Open
Abstract
Interaction of the T cell receptor (TCR) with an MHC-antigenic peptide complex results in changes at the molecular and cellular levels in T cells. The outside environmental cues are translated into various signal transduction pathways within the cell, which mediate the activation of various genes with the help of specific transcription factors. These signaling networks propagate with the help of various effector enzymes, such as kinases, phosphatases, and phospholipases. Integration of these disparate signal transduction pathways is done with the help of adaptor proteins that are non-enzymatic in function and that serve as a scaffold for various protein-protein interactions. This process aids in connecting the proximal to distal signaling pathways, thereby contributing to the full activation of T cells. This review provides a comprehensive snapshot of the various molecules involved in regulating T cell receptor signaling, covering both enzymes and adaptors, and will discuss their role in human disease.
Collapse
Affiliation(s)
- Kinjal Shah
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
- Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Amr Al-Haidari
- Clinical Genetics and Pathology, Skåne University Hospital, Region Skåne, Lund, Sweden
- Clinical Sciences Department, Surgery Research Unit, Lund University, Malmö, Sweden
| | - Jianmin Sun
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
- Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, Lund, Sweden
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Science and Technology center, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Julhash U Kazi
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden.
- Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, Lund, Sweden.
| |
Collapse
|
13
|
Aboualizadeh F, Yao Z, Guan J, Drecun L, Pathmanathan S, Snider J, Umapathy G, Kotlyar M, Jurisica I, Palmer R, Stagljar I. Mapping the Phospho-dependent ALK Interactome to Identify Novel Components in ALK Signaling. J Mol Biol 2021; 433:167283. [PMID: 34606829 DOI: 10.1016/j.jmb.2021.167283] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/27/2021] [Accepted: 09/27/2021] [Indexed: 10/25/2022]
Abstract
Protein-protein interactions (PPIs) play essential roles in Anaplastic Lymphoma Kinase (ALK) signaling. Systematic characterization of ALK interactors helps elucidate novel ALK signaling mechanisms and may aid in the identification of novel therapeutics targeting related diseases. In this study, we used the Mammalian Membrane Two-Hybrid (MaMTH) system to map the phospho-dependent ALK interactome. By screening a library of 86 SH2 domain-containing full length proteins, 30 novel ALK interactors were identified. Many of their interactions are correlated to ALK phosphorylation activity: oncogenic ALK mutations potentiate the interactions and ALK inhibitors attenuate the interactions. Among the novel interactors, NCK2 was further verified in neuroblastoma cells using co-immunoprecipitation. Modulation of ALK activity by addition of inhibitors lead to concomitant changes in the tyrosine phosphorylation status of NCK2 in neuroblastoma cells, strongly supporting the functionality of the ALK/NCK2 interaction. Our study provides a resource list of potential novel ALK signaling components for further study.
Collapse
Affiliation(s)
| | - Zhong Yao
- Donnelly Centre, University of Toronto, Ontario, Canada
| | - Jikui Guan
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg SE-40530, Sweden
| | - Luka Drecun
- Donnelly Centre, University of Toronto, Ontario, Canada
| | | | - Jamie Snider
- Donnelly Centre, University of Toronto, Ontario, Canada
| | - Ganesh Umapathy
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg SE-40530, Sweden
| | - Max Kotlyar
- Krembil Research Institute, University Health Network, Ontario, Canada
| | - Igor Jurisica
- Krembil Research Institute, University Health Network, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Ontario, Canada; Department of Computer Science, University of Toronto, Ontario, Canada; Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovak Republic
| | - Ruth Palmer
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg SE-40530, Sweden
| | - Igor Stagljar
- Donnelly Centre, University of Toronto, Ontario, Canada; Department of Biochemistry, University of Toronto, Ontario, Canada; Department of Molecular Genetics, University of Toronto, Ontario, Canada; Mediterranean Institute for Life Sciences, Meštrovićevo Šetalište 45, Split, Croatia; School of Medicine, University of Split, Split, Croatia. https://twitter.com/stagljar
| |
Collapse
|
14
|
Abstract
The non-catalytic region of tyrosine kinase (Nck) family of adaptors, consisting of Nck1 and Nck2, contributes to selectivity and specificity in the flow of cellular information by recruiting components of signaling networks. Known to play key roles in cytoskeletal remodeling, Nck adaptors modulate host cell-pathogen interactions, immune cell receptor activation, cell adhesion and motility, and intercellular junctions in kidney podocytes. Genetic inactivation of both members of the Nck family results in embryonic lethality; however, viability of mice lacking either one of these adaptors suggests partial functional redundancy. In this Cell Science at a Glance and the accompanying poster, we highlight the molecular organization and functions of the Nck family, focusing on key interactions and pathways, regulation of cellular processes, development, homeostasis and pathogenesis, as well as emerging and non-redundant functions of Nck1 compared to those of Nck2. This article thus aims to provide a timely perspective on the biology of Nck adaptors and their potential as therapeutic targets.
Collapse
Affiliation(s)
- Briana C. Bywaters
- Department of Veterinary Pathobiology, Texas A&M University, College Station, TX 7783, USA
| | - Gonzalo M. Rivera
- Department of Veterinary Pathobiology, Texas A&M University, College Station, TX 7783, USA
| |
Collapse
|
15
|
Interaction Network Provides Clues on the Role of BCAR1 in Cellular Response to Changes in Gravity. COMPUTATION 2021. [DOI: 10.3390/computation9080081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
When culturing cells in space or under altered gravity conditions on Earth to investigate the impact of gravity, their adhesion and organoid formation capabilities change. In search of a target where the alteration of gravity force could have this impact, we investigated p130cas/BCAR1 and its interactions more thoroughly, particularly as its activity is sensitive to applied forces. This protein is well characterized regarding its role in growth stimulation and adhesion processes. To better understand BCAR1′s force-dependent scaffolding of other proteins, we studied its interactions with proteins we had detected by proteome analyses of MCF-7 breast cancer and FTC-133 thyroid cancer cells, which are both sensitive to exposure to microgravity and express BCAR1. Using linked open data resources and our experiments, we collected comprehensive information to establish a semantic knowledgebase and analyzed identified proteins belonging to signaling pathways and their networks. The results show that the force-dependent phosphorylation and scaffolding of BCAR1 influence the structure, function, and degradation of intracellular proteins as well as the growth, adhesion and apoptosis of cells similarly to exposure of whole cells to altered gravity. As BCAR1 evidently plays a significant role in cell responses to gravity changes, this study reveals a clear path to future research performing phosphorylation experiments on BCAR1.
Collapse
|
16
|
Jaufmann J, Franke FC, Sperlich A, Blumendeller C, Kloos I, Schneider B, Sasaki D, Janssen KP, Beer-Hammer S. The emerging and diverse roles of the SLy/SASH1-protein family in health and disease-Overview of three multifunctional proteins. FASEB J 2021; 35:e21470. [PMID: 33710696 DOI: 10.1096/fj.202002495r] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 01/22/2021] [Accepted: 02/08/2021] [Indexed: 12/12/2022]
Abstract
Intracellular adaptor proteins are indispensable for the transduction of receptor-derived signals, as they recruit and connect essential downstream effectors. The SLy/SASH1-adaptor family comprises three highly homologous proteins, all of them sharing conserved structural motifs. The initial characterization of the first member SLy1/SASH3 (SH3 protein expressed in lymphocytes 1) in 2001 was rapidly followed by identification of SLy2/HACS1 (hematopoietic adaptor containing SH3 and SAM domains 1) and SASH1/SLy3 (SAM and SH3 domain containing 1). Based on their pronounced sequence similarity, they were subsequently classified as one family of intracellular scaffold proteins. Despite their obvious homology, the three SLy/SASH1-members fundamentally differ with regard to their expression and function in intracellular signaling. On the contrary, growing evidence clearly demonstrates an important role of all three proteins in human health and disease. In this review, we systematically summarize what is known about the SLy/SASH1-adaptors in the field of molecular cell biology and immunology. To this end, we recapitulate current research about SLy1/SASH3, SLy2/HACS1, and SASH1/SLy3, with an emphasis on their similarities and differences.
Collapse
Affiliation(s)
- Jennifer Jaufmann
- Department of Pharmacology, Experimental Therapy and Toxicology, Institute of Experimental and Clinical Pharmacology and Pharmacogenomik and ICePhA, University of Tuebingen, Tuebingen, Germany
| | - Fabian Christoph Franke
- Department of Surgery, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Andreas Sperlich
- Department of Surgery, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Carolin Blumendeller
- Department of Pharmacology, Experimental Therapy and Toxicology, Institute of Experimental and Clinical Pharmacology and Pharmacogenomik and ICePhA, University of Tuebingen, Tuebingen, Germany
| | - Isabel Kloos
- Department of Pharmacology, Experimental Therapy and Toxicology, Institute of Experimental and Clinical Pharmacology and Pharmacogenomik and ICePhA, University of Tuebingen, Tuebingen, Germany
| | - Barbara Schneider
- Department of Pharmacology, Experimental Therapy and Toxicology, Institute of Experimental and Clinical Pharmacology and Pharmacogenomik and ICePhA, University of Tuebingen, Tuebingen, Germany
| | - Daisuke Sasaki
- Department of Surgery, Klinikum rechts der Isar, Technical University Munich, Munich, Germany.,Medical SC New Technology Strategy Office, General Research Institute, Nitto Boseki, Co., Ltd, Tokyo, Japan
| | - Klaus-Peter Janssen
- Department of Surgery, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Sandra Beer-Hammer
- Department of Pharmacology, Experimental Therapy and Toxicology, Institute of Experimental and Clinical Pharmacology and Pharmacogenomik and ICePhA, University of Tuebingen, Tuebingen, Germany
| |
Collapse
|
17
|
Lauko DI, Ohkawa T, Mares SE, Welch MD. Baculovirus actin-rearrangement-inducing factor ARIF-1 induces the formation of dynamic invadosome clusters. Mol Biol Cell 2021; 32:1433-1445. [PMID: 34133213 PMCID: PMC8351737 DOI: 10.1091/mbc.e20-11-0705] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The baculovirus Autographa californica multiple nucleopolyhedrovirus (AcMNPV), a pathogen of lepidopteran insects, has a striking dependence on the host cell actin cytoskeleton. During the delayed-early stage of infection, AcMNPV was shown to induce the accumulation of actin at the cortex of infected cells. However, the dynamics and molecular mechanism of cortical actin assembly remained unknown. Here, we show that AcMNPV induces dynamic cortical clusters of dot-like actin structures that mediate degradation of the underlying extracellular matrix and therefore function similarly to clusters of invadosomes in mammalian cells. Furthermore, we find that the AcMNPV protein actin-rearrangement-inducing factor-1 (ARIF-1), which was previously shown to be necessary and sufficient for cortical actin assembly and efficient viral infection in insect hosts, is both necessary and sufficient for invadosome formation. We mapped the sequences within the C-terminal cytoplasmic region of ARIF-1 that are required for invadosome formation and identified individual tyrosine and proline residues that are required for organizing these structures. Additionally, we found that ARIF-1 and the invadosome-associated proteins cortactin and the Arp2/3 complex localize to invadosomes and Arp2/3 complex is required for their formation. These ARIF-1-induced invadosomes may be important for the function of ARIF-1 in systemic virus spread.
Collapse
Affiliation(s)
- Domokos I Lauko
- Microbiology Graduate Group, University of California, Berkeley, Berkeley, CA 94720
| | - Taro Ohkawa
- Department of Molecular & Cell Biology, University of California, Berkeley, Berkeley, CA 94720
| | - Sergio E Mares
- Department of Molecular & Cell Biology, University of California, Berkeley, Berkeley, CA 94720
| | - Matthew D Welch
- Microbiology Graduate Group, University of California, Berkeley, Berkeley, CA 94720.,Department of Molecular & Cell Biology, University of California, Berkeley, Berkeley, CA 94720
| |
Collapse
|
18
|
Leyden F, Uthishtran S, Moorthi UK, York HM, Patil A, Gandhi H, Petrov EP, Bornschlögl T, Arumugam S. Rac1 activation can generate untemplated, lamellar membrane ruffles. BMC Biol 2021; 19:72. [PMID: 33849538 PMCID: PMC8042924 DOI: 10.1186/s12915-021-00997-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 02/26/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Membrane protrusions that occur on the dorsal surface of a cell are an excellent experimental system to study actin machinery at work in a living cell. Small GTPase Rac1 controls the membrane protrusions that form and encapsulate extracellular volumes to perform pinocytic or phagocytic functions. RESULTS Here, capitalizing on rapid volumetric imaging capabilities of lattice light-sheet microscopy (LLSM), we describe optogenetic approaches using photoactivable Rac1 (PA-Rac1) for controlled ruffle generation. We demonstrate that PA-Rac1 activation needs to be continuous, suggesting a threshold local concentration for sustained actin polymerization leading to ruffling. We show that Rac1 activation leads to actin assembly at the dorsal surface of the cell membrane that result in sheet-like protrusion formation without any requirement of a template. Further, this approach can be used to study the complex morpho-dynamics of the protrusions or to investigate specific proteins that may be enriched in the ruffles. Deactivating PA-Rac1 leads to complex contractile processes resulting in formation of macropinosomes. Using multicolour imaging in combination with these approaches, we find that Myo1e specifically is enriched in the ruffles. CONCLUSIONS Combining LLSM and optogenetics enables superior spatial and temporal control for studying such dynamic mechanisms. Demonstrated here, the techniques implemented provide insight into the complex nature of the molecular interplay involved in dynamic actin machinery, revealing that Rac1 activation can generate untemplated, lamellar protrusions.
Collapse
Affiliation(s)
- F Leyden
- Single Molecule Science, University of New South Wales, Sydney, Australia
| | - S Uthishtran
- Monash Biomedicine Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton/Melbourne, VIC, 3800, Australia
| | - U K Moorthi
- Monash Biomedicine Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton/Melbourne, VIC, 3800, Australia
- European Molecular Biological Laboratory Australia (EMBL Australia), Monash University, Clayton/Melbourne, VIC, 3800, Australia
| | - H M York
- Monash Biomedicine Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton/Melbourne, VIC, 3800, Australia
- European Molecular Biological Laboratory Australia (EMBL Australia), Monash University, Clayton/Melbourne, VIC, 3800, Australia
| | - A Patil
- Monash Biomedicine Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton/Melbourne, VIC, 3800, Australia
- European Molecular Biological Laboratory Australia (EMBL Australia), Monash University, Clayton/Melbourne, VIC, 3800, Australia
| | - H Gandhi
- Monash Biomedicine Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton/Melbourne, VIC, 3800, Australia
- European Molecular Biological Laboratory Australia (EMBL Australia), Monash University, Clayton/Melbourne, VIC, 3800, Australia
| | - E P Petrov
- Physikalisch-Technische Bundesanstalt, Abbestraße 2-12, 10587, Berlin, Germany
| | - T Bornschlögl
- L'Oréal Research & Innovation, 1 Avenue Eugène Schueller, 93601, Aulnay sous Bois, France
| | - S Arumugam
- Single Molecule Science, University of New South Wales, Sydney, Australia.
- Monash Biomedicine Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton/Melbourne, VIC, 3800, Australia.
- European Molecular Biological Laboratory Australia (EMBL Australia), Monash University, Clayton/Melbourne, VIC, 3800, Australia.
- ARC Centre of Excellence in Advanced Molecular Imaging, UNSW, Sydney, Australia.
| |
Collapse
|
19
|
Diab A, Qi J, Shahin I, Milligan C, Fawcett JP. NCK1 Regulates Amygdala Activity to Control Context-dependent Stress Responses and Anxiety in Male Mice. Neuroscience 2020; 448:107-125. [PMID: 32946951 DOI: 10.1016/j.neuroscience.2020.09.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 08/20/2020] [Accepted: 09/08/2020] [Indexed: 10/23/2022]
Abstract
Anxiety disorder (AD) is characterized by the development of maladaptive neuronal circuits and changes to the excitatory/inhibitory (E/I) balance of the central nervous system. Although AD is considered to be heritable, specific genetic markers remain elusive. Recent genome-wide association studies (GWAS) studies have identified non-catalytic region of tyrosine kinase adaptor protein 1 (NCK1), a gene that codes for an intracellular adaptor protein involved in actin dynamics, as an important gene in the regulation of mood. Using a murine model in which NCK1 is inactivated, we show that male, but not female, mice display increased levels of context-dependent anxiety-like behaviors along with an increase in circulating serum corticosterone relative to control. Treatment of male NCK1 mutant mice with a positive allosteric modulator of the GABAA receptor rescued the anxiety-like behaviors implicating NCK1 in regulating neuronal excitability. These defects are not attributable to apparent defects in gross brain structure or in axon guidance. However, when challenged in an approach-avoidance conflict paradigm, male NCK1-deficient mice have decreased neuronal activation in the prefrontal cortex (PFC), as well as decreased activation of inhibitory interneurons in the basolateral amygdala (BLA). Finally, NCK1 deficiency results in loss of dendritic spine density in principal neurons of the BLA. Taken together, these data implicate NCK1 in the control of E/I balance in BLA. Our work identifies a novel role for NCK1 in the regulation of sex-specific neuronal circuitry necessary for controlling anxiety-like behaviors. Further, our work points to this animal model as a useful preclinical tool for the study of novel anxiolytics and its significance towards understanding sex differences in anxiolytic function.
Collapse
Affiliation(s)
- Antonios Diab
- Department of Pharmacology, Dalhousie University, Canada
| | - Jiansong Qi
- Department of Pharmacology, Dalhousie University, Canada
| | - Ibrahim Shahin
- Department of Pharmacology, Dalhousie University, Canada
| | | | - James P Fawcett
- Department of Pharmacology, Dalhousie University, Canada; Department of Surgery, Dalhousie University, Canada.
| |
Collapse
|
20
|
Zhang X, Zhang B, Masoudi A, Wang X, Xue X, Li M, Xiao Q, Wang M, Liu J, Wang H. Comprehensive analysis of protein expression levels and phosphorylation levels in host skin in response to tick (Haemaphysalis longicornis) bite. J Proteomics 2020; 226:103898. [PMID: 32682108 DOI: 10.1016/j.jprot.2020.103898] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 07/01/2020] [Accepted: 07/08/2020] [Indexed: 11/15/2022]
Abstract
Ticks are parasitic arthropods that suck blood from the surface of most vertebrates. They can transmit a variety of pathogens. The blood sucking of ticks causes varying degrees of damage to the skin of the host. Proteins related to immune regulation, vascular repair, and wound healing in mammalian skin respond to tick bites by regulating their expression and post-translational modifications to protect the skin from injury. Phosphorylation of proteins, as the most common post-translational modification of proteins, plays an important role in the rapid regulation of cell signal transduction, gene expression and cell cycle. To systematically explore the molecular regulatory mechanisms employed by mammalian skin to resist tick bites, larval, nymphal, and adult Haemaphysalis longicornis were used to bite the skin tissues of healthy rabbits in the present study. The quantitative proteomic technology data-independent acquisition was then carried out to investigate in depth the changes in protein expression and phosphorylation in rabbit skin after tick bite. The results showed that among the 4034 proteins and 1795 phosphorylated proteins identified, a total of 202 proteins and 435 phosphorylation sites were changed after H. longicornis bite. In order to provide convenience for sucking blood, active substances in the saliva of H. longicornis injected into the rabbit's skin can cause the expression level of trichohyalin and peptidyl arginine deiminase 3 in the skin of the host downregulate, which can make the host hair loss and regeneration disorders. At the same time, the active substances in saliva of the H. longicornis led to the phosphorylation of microtubule actin cross-linking factor 1 in the host's skin and further inactivation, so as to delay the healing of the host wound. In response to tick bites, the host skin promotes coagulation through high expression of fibrinogen and fibronectin, and vascular repair through high expression of integrin linked kinase and tenascin C, as well as accelerated phosphorylation of the phosphorylated protein Nck adaptor protein 1, and wound healing through high expression of ezrin and integrin. The upregulation of proteins such as coronin, NADPH oxidase, calnexin, and calreticulin and phosphorylation level of IL-4R in the host skin after the H. longicornis bite indicated that the immune response was playing an important defensive role in response to tick bites. Meanwhile, we found that the upregulated two lectins, mannose receptor C-type 1 and DC-SIGN, may serve as molecular makers to identify and monitor whether the skin is bitten by ticks. SIGNIFICANCE: Haemaphysalis longicornis are parasitic arthropods that suck blood from the surface of most vertebrates. They can transmit a variety of pathogens and are harmful to humans and livestock. The present study is the first quantitative proteomic study on protein expression levels in the rabbit skin after infection by H. longicornis. It is also the first quantitative phosphoproteomic study in the host skin infected by ticks. In this study, we found that tick bites cause the host hair loss and regeneration disorders. For resisting tick bite, the host activates the immune response and initiates vascular repair and wound-healing systems. In addition, some phosphorylated proteins promote host immunity and vascular repair. These results can help us further understand the defence mechanism of the host against tick bites, provide a basis for the development of an anti-tick vaccine, the development of anti-tick drugs, and the diagnosis of tick-borne diseases.
Collapse
Affiliation(s)
- Xiaoli Zhang
- Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei 050024, PR China
| | - Baowen Zhang
- Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei 050024, PR China
| | - Abolfazl Masoudi
- Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei 050024, PR China
| | - Xiaoshuang Wang
- Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei 050024, PR China
| | - Xiaomin Xue
- Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei 050024, PR China
| | - Mengxue Li
- Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei 050024, PR China
| | - Qi Xiao
- Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei 050024, PR China
| | - Minjing Wang
- Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei 050024, PR China
| | - Jingze Liu
- Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei 050024, PR China.
| | - Hui Wang
- Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei 050024, PR China.
| |
Collapse
|
21
|
Wines-Samuelson M, Chowdhury S, Berk BC. Nck1 is a critical adaptor between proatherogenic blood flow, inflammation, and atherosclerosis. J Clin Invest 2020; 130:3968-3970. [PMID: 32657777 PMCID: PMC7410038 DOI: 10.1172/jci138536] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Atherosclerosis is an inflammatory condition of the arteries that has profound incidence and increasing prevalence. Although endothelial cells detect changes in blood flow, how endothelial activation contributes to atherogenic inflammation is not well understood. In this issue of the JCI, Alfaidi et al. used mouse models to explore flow-induced endothelial activation. The authors revealed a role for Nck1 and a specific activator of the innate immune response, the downstream interleukin receptor-associated kinase-1 (IRAK-1) in NF-κB-mediated inflammation and atherosclerosis susceptibility. These results link disturbed blood flow to NF-κB-mediated inflammation, which promotes atherosclerosis, and provide Nck1 as a potential target for the treatment of atherosclerosis.
Collapse
|
22
|
Tachie-Menson T, Gázquez-Gutiérrez A, Fulcher LJ, Macartney TJ, Wood NT, Varghese J, Gourlay R, Soares RF, Sapkota GP. Characterisation of the biochemical and cellular roles of native and pathogenic amelogenesis imperfecta mutants of FAM83H. Cell Signal 2020; 72:109632. [PMID: 32289446 PMCID: PMC7284315 DOI: 10.1016/j.cellsig.2020.109632] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 04/06/2020] [Accepted: 04/06/2020] [Indexed: 12/27/2022]
Abstract
The majority of mutations identified in patients with amelogenesis imperfecta have been mapped to FAM83H. As FAM83H expression is not limited to the enamel, how FAM83H contributes to amelogenesis is still largely unknown. We previously reported that members of the FAM83 family of proteins interact with and regulate the subcellular distribution of the promiscuous serine-threonine protein kinase CK1 family, through their shared N-terminal DUF1669 domains. FAM83H co-localises with CK1 isoforms to speckle-like structures in both the cytoplasm and nucleus. In this report, we show FAM83H, unlike other FAM83 proteins, interacts and colocalises with NCK1/2 tyrosine kinase adaptor proteins. This interaction is mediated by proline-rich motifs within the C-terminus of FAM83H, specifically interacting with the second and third SH3 domains of NCK1/2. Moreover, FAM83H pathogenic AI mutant proteins, which trigger C-terminal truncations of FAM83H, retain their interactions with CK1 isoforms but lose interaction with NCK1/2. These AI mutant FAM83H proteins acquire a nuclear localisation, and recruit CK1 isoforms to the nucleus where CK1 retains its kinase activity. As understanding the constituents of the FAM83H-localised speckles may hold the key to unravelling potential substrates of FAM83H-associated CK1 substrates, we employed a TurboID-based proximity labelling approach and uncovered several proteins including Iporin and BAG3 as potential constituents of the speckles.
Collapse
Affiliation(s)
- Theresa Tachie-Menson
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, United Kingdom
| | - Ana Gázquez-Gutiérrez
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, United Kingdom; University of Seville, Av. Sanchez Pizjuan, s/n, 41009, Seville, Spain
| | - Luke J Fulcher
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, United Kingdom
| | - Thomas J Macartney
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, United Kingdom
| | - Nicola T Wood
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, United Kingdom
| | - Joby Varghese
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, United Kingdom
| | - Robert Gourlay
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, United Kingdom
| | - Renata F Soares
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, United Kingdom
| | - Gopal P Sapkota
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, United Kingdom.
| |
Collapse
|
23
|
Nck1 promotes the progression of ovarian carcinoma by enhancing the PI3K/AKT/p70S6K signaling. Hum Cell 2020; 33:768-779. [PMID: 32166565 DOI: 10.1007/s13577-020-00344-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 03/02/2020] [Indexed: 02/05/2023]
Abstract
Non-catalytic region of tyrosine kinase adaptor protein 1 (Nck1) is crucial for the progression of cancers. However, little is known on the role of Nck1 in the progression of ovarian carcinoma (OC). Here, we show that Nck1 expression is up-regulated in 176 OC tissues, compared with non-carcinoma ovarian tissues, and the up-regulated Nck1 expression is associated with the aggressiveness of OC and shorter overall and disease-free survival in this population. Higher Nck1 expression was an independent risk factor for poor prognosis of OC. Furthermore, Nck1 silencing by short hairpin RNA (shRNA) technology significantly inhibited the proliferation, migration and invasion of OC cells in vitro and the growth and metastasis of implanted OC tumors in vivo. Human kinase phosphorylation array indicated that Nck1 silencing significantly reduced the relative levels of 11 kinase expression and phosphorylation in OC cells, particularly for decreased levels of p70S6 kinase (p70S6K) and protein kinase B (AKT) expression in SKOV3 cells. Actually, Nck1 silencing significantly decreased PI3K and AKT expression, and reduced AKT and p70S6K phosphorylation while Nck1 over-expression had opposite effects in OC cells. Therefore, our data indicate that Nck1 promotes the progression of OC by enhancing the PI3k/AKT/p70S6K signaling in OC. Our findings suggest that Nck1 expression may be valuable for evaluating the prognosis of OC and as a target for design of new therapies for OC.
Collapse
|
24
|
Wipa P, Paensuwan P, Ngoenkam J, Woessner NM, Minguet S, Schamel WW, Pongcharoen S. Actin polymerization regulates recruitment of Nck to CD3ε upon T-cell receptor triggering. Immunology 2019; 159:298-308. [PMID: 31674657 DOI: 10.1111/imm.13146] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 10/28/2019] [Accepted: 10/29/2019] [Indexed: 12/15/2022] Open
Abstract
Following T-cell antigen receptor (TCR) engagement, rearrangement of the actin cytoskeleton supports intracellular signal transduction and T-cell activation. The non-catalytic region of the tyrosine kinase (Nck) molecule is an adapter protein implicated in TCR-induced actin polymerization. Further, Nck is recruited to the CD3ε subunit of the TCR upon TCR triggering. Here we examine the role of actin polymerization in the recruitment of Nck to the TCR. To this end, Nck binding to CD3ε was quantified in Jurkat cells using the proximity ligation assay. We show that inhibition of actin polymerization using cytochalasin D delayed the recruitment of Nck1 to the TCR upon TCR triggering. Interestingly, CD3ε phosphorylation was also delayed. These findings suggest that actin polymerization promotes the recruitment of Nck to the TCR, enhancing downstream signaling, such as phosphorylation of CD3ε.
Collapse
Affiliation(s)
- Piyamaporn Wipa
- Department of Microbiology and Parasitology, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand
| | - Pussadee Paensuwan
- Department of Optometry, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, Thailand
| | - Jatuporn Ngoenkam
- Department of Microbiology and Parasitology, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand
| | - Nadine M Woessner
- Department of Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany.,Signaling Research Centers BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Susana Minguet
- Department of Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany.,Signaling Research Centers BIOSS and CIBSS, University of Freiburg, Freiburg, Germany.,Center for Chronic Immunodeficiency CCI, Medical Center Freiburg and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Wolfgang W Schamel
- Department of Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany.,Signaling Research Centers BIOSS and CIBSS, University of Freiburg, Freiburg, Germany.,Center for Chronic Immunodeficiency CCI, Medical Center Freiburg and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sutatip Pongcharoen
- Division of Immunology, Department of Medicine, Faculty of Medicine, Naresuan University, Phitsanulok, Thailand.,Center of Excellence in Medical Biotechnology, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand.,Research Center for Academic Excellence in Petroleum, Petrochemical, and Advanced Materials, Faculty of Science, Naresuan University, Phitsanulok, Thailand
| |
Collapse
|
25
|
Kim S, Kalappurakkal JM, Mayor S, Rosen MK. Phosphorylation of nephrin induces phase separated domains that move through actomyosin contraction. Mol Biol Cell 2019; 30:2996-3012. [PMID: 31599693 PMCID: PMC6857567 DOI: 10.1091/mbc.e18-12-0823] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 09/03/2019] [Accepted: 10/03/2019] [Indexed: 01/12/2023] Open
Abstract
The plasma membrane of eukaryotic cells is organized into lipid and protein microdomains, whose assembly mechanisms and functions are incompletely understood. We demonstrate that proteins in the nephrin/Nck/N-WASP actin-regulatory pathway cluster into micron-scale domains at the basal plasma membrane upon triggered phosphorylation of transmembrane protein nephrin. The domains are persistent but readily exchange components with their surroundings, and their formation is dependent on the number of Nck SH3 domains, suggesting they are phase separated polymers assembled through multivalent interactions among the three proteins. The domains form independent of the actin cytoskeleton, but acto-myosin contractility induces their rapid lateral movement. Nephrin phosphorylation induces larger clusters at the cell periphery, which are associated with extensive actin assembly and dense filopodia. Our studies illustrate how multivalent interactions between proteins at the plasma membrane can produce micron-scale organization of signaling molecules, and how the resulting clusters can both respond to and control the actin cytoskeleton.
Collapse
Affiliation(s)
- Soyeon Kim
- Department of Biophysics and Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390
- The HHMI/MBL Summer Institute, Marine Biological Laboratory, Woods Hole, MA 02543
| | - Joseph M. Kalappurakkal
- The HHMI/MBL Summer Institute, Marine Biological Laboratory, Woods Hole, MA 02543
- National Centre for Biological Sciences, Bangalore 560065, India
| | - Satyajit Mayor
- The HHMI/MBL Summer Institute, Marine Biological Laboratory, Woods Hole, MA 02543
- National Centre for Biological Sciences, Bangalore 560065, India
| | - Michael K. Rosen
- Department of Biophysics and Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390
- The HHMI/MBL Summer Institute, Marine Biological Laboratory, Woods Hole, MA 02543
| |
Collapse
|
26
|
Lillico DME, Pemberton JG, Niemand R, Stafford JL. Selective recruitment of Nck and Syk contribute to distinct leukocyte immune-type receptor-initiated target interactions. Cell Signal 2019; 66:109443. [PMID: 31626955 DOI: 10.1016/j.cellsig.2019.109443] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 10/14/2019] [Indexed: 01/18/2023]
Abstract
The ability of phagocytes to recognize, immobilize, and engulf extracellular targets are fundamental immune cell processes that allow for the destruction of a variety of microbial intruders. The phagocytic process depends onsignalling events that initiate dynamic changes in the plasma membrane architecture that are required to accommodate the internalization of large particulate targets. To better understand fundamental molecular mechanisms responsible for facilitating phagocytic receptor-mediated regulation of cytoskeletal networks, our research has focused on investigating representative immunoregulatory proteins from the channel catfish (Ictalurus punctatus) leukocyte immune-type receptor family (IpLITRs). Specifically, we have shown that a specific IpLITR-type can regulate the constitutive deployment of filopodial-like structures to actively capture and secure targets to the phagocyte surface, which is followed by F-actin mediated membrane dynamics that are associated with the formation of phagocytic cup-like structures that precede target engulfment. In the present study, we use confocal imaging to examine the recruitment of mediators of the F-actin cytoskeleton during IpLITR-mediated regulation of membrane dynamics. Our results provide novel details regarding the dynamic recruitment of the signaling effectors Nck and Syk during classical as well as atypical IpLITR-induced phagocytic processes.
Collapse
Affiliation(s)
- Dustin M E Lillico
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Joshua G Pemberton
- Section on Molecular Signal Transduction, Program for Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, United States
| | - Rikus Niemand
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - James L Stafford
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
27
|
Guan T, Huang K, Liu Y, Hou S, Hu C, Li Y, Zhang J, Zhao J, Zhang J, Wang R, Huang Y. Aristolochic acid inhibits Slit2-induced migration and tube formation via inactivation of Robo1/Robo2-NCK1/NCK2 signaling pathway in human umbilical vein endothelial cells. Toxicol Lett 2018; 300:51-58. [PMID: 30381256 DOI: 10.1016/j.toxlet.2018.10.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Revised: 10/15/2018] [Accepted: 10/22/2018] [Indexed: 11/28/2022]
Abstract
Robo1/Robo2-NCK1/NCK2 signaling pathway controls endothelial cell sprouting and migration induced by Slit2 or VEGF, but whether it is involved in peritubular capillary (PTC) rarefaction of Aristolochic acid nephropathy (AAN) is unclear. In the present study, we evaluated whether AA exerts antiangiogenic effects by targeting this signaling pathways in HUVECs. HUVECs or lentivirus-mediated NCK1-overexpressing HUVECs were stimulated with AA (1, 2 or 3 μg/ml) in the absence or presence of 6 nM Slit2. Our results showed that AAІ (1-3 μg/ml) dose-dependently inhibited the migration and tube formation of HUVECs. This inhibition was in parallel with down-regulated mRNA and protein expression of Slit2/Robo1/Robo2-NCK1/NCK2 signaling pathway. Importantly, overexpression of NCK1 rescued AAІ-impaired angiogenesis, as evidenced by the increase of cell migration and tube formation of HUVECs in response to Slit2. The down-regulation of NCK2 and decreased activation of Rac1 was also restored by overexpression of NCK1. Taken together, our findings show that AA inhibits Slit2-induced migration and tube formation via inactivation of Robo1/Robo2-NCK1/NCK2 signaling pathway in HUVECs, and NCK1 might be a potential agent for vascular remodeling in AAN and diseases associated with impaired angiogenesis.
Collapse
Affiliation(s)
- Tao Guan
- Department of Nephrology, Xinqiao Hospital, Army Medical University, The Third Military Medical University, Chongqing 400037, PR China
| | - Ke Huang
- Department of Dermatology, Rheumatic immunology, Xinqiao Hospital, Army Medical University, The Third Military Medical University, Chongqing, 400037, PR China
| | - Yuanyuan Liu
- Department of Nephrology, Xinqiao Hospital, Army Medical University, The Third Military Medical University, Chongqing 400037, PR China
| | - Shihui Hou
- Department of Nephrology, Xinqiao Hospital, Army Medical University, The Third Military Medical University, Chongqing 400037, PR China
| | - Chengfang Hu
- Department of Nephrology, Xinqiao Hospital, Army Medical University, The Third Military Medical University, Chongqing 400037, PR China
| | - Yi Li
- Department of Nephrology, Xinqiao Hospital, Army Medical University, The Third Military Medical University, Chongqing 400037, PR China
| | - Jingbo Zhang
- Department of Nephrology, Xinqiao Hospital, Army Medical University, The Third Military Medical University, Chongqing 400037, PR China
| | - Jinghong Zhao
- Department of Nephrology, Xinqiao Hospital, Army Medical University, The Third Military Medical University, Chongqing 400037, PR China
| | - Jun Zhang
- Department of Nephrology, Xinqiao Hospital, Army Medical University, The Third Military Medical University, Chongqing 400037, PR China
| | - Rupeng Wang
- Department of Dermatology, Rheumatic immunology, Xinqiao Hospital, Army Medical University, The Third Military Medical University, Chongqing, 400037, PR China
| | - Yunjian Huang
- Department of Nephrology, Xinqiao Hospital, Army Medical University, The Third Military Medical University, Chongqing 400037, PR China.
| |
Collapse
|
28
|
Zygmunt M, Hoinkis D, Hajto J, Piechota M, Skupień-Rabian B, Jankowska U, Kędracka-Krok S, Rodriguez Parkitna J, Korostyński M. Expression of alternatively spliced variants of the Dclk1 gene is regulated by psychotropic drugs. BMC Neurosci 2018; 19:55. [PMID: 30208879 PMCID: PMC6134793 DOI: 10.1186/s12868-018-0458-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 09/06/2018] [Indexed: 01/05/2023] Open
Abstract
Background The long-term effects of psychotropic drugs are associated with the reversal of disease-related alterations through the reorganization and normalization of neuronal connections. Molecular factors that trigger drug-induced brain plasticity remain only partly understood. Doublecortin-like kinase 1 (Dclk1) possesses microtubule-polymerizing activity during synaptic plasticity and neurogenesis. However, the Dclk1 gene shows a complex profile of transcriptional regulation, with two alternative promoters and exon splicing patterns that suggest the expression of multiple isoforms with different kinase activities. Results Here, we applied next-generation sequencing to analyze changes in the expression of Dclk1 gene isoforms in the brain in response to several psychoactive drugs with diverse pharmacological mechanisms of action. We used bioinformatics tools to define the range and levels of Dclk1 transcriptional regulation in the mouse nucleus accumbens and prefrontal cortex. We also sought to investigate the presence of DCLK1-derived peptides using mass spectrometry. We detected 15 transcripts expressed from the Dclk1 locus (FPKM > 1), including 2 drug-regulated variants (fold change > 2). Drugs that act on serotonin receptors (5-HT2A/C) regulate a subset of Dclk1 isoforms in a brain-region-specific manner. The strongest influence was observed for the mianserin-induced expression of an isoform with intron retention. The drug-activated expression of novel alternative Dclk1 isoforms was validated using qPCR. The drug-regulated isoform contains genetic variants of DCLK1 that have been previously associated with schizophrenia and hyperactivity disorder in humans. We identified a short peptide that might originate from the novel DCLK1 protein product. Moreover, protein domains encoded by the regulated variant indicate their potential involvement in the negative regulation of the canonical DCLK1 protein. Conclusions In summary, we identified novel isoforms of the neuroplasticity-related gene Dclk1 that are expressed in the brain in response to psychotropic drug treatments. Electronic supplementary material The online version of this article (10.1186/s12868-018-0458-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Magdalena Zygmunt
- Department of Molecular Neuropharmacology, Institute of Pharmacology of the Polish Academy of Sciences, Smetna 12, 31-343, Krakow, Poland
| | - Dżesika Hoinkis
- Department of Molecular Neuropharmacology, Institute of Pharmacology of the Polish Academy of Sciences, Smetna 12, 31-343, Krakow, Poland
| | - Jacek Hajto
- Department of Molecular Neuropharmacology, Institute of Pharmacology of the Polish Academy of Sciences, Smetna 12, 31-343, Krakow, Poland
| | - Marcin Piechota
- Department of Molecular Neuropharmacology, Institute of Pharmacology of the Polish Academy of Sciences, Smetna 12, 31-343, Krakow, Poland
| | - Bożena Skupień-Rabian
- Laboratory of Proteomics and Mass Spectrometry, Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
| | - Urszula Jankowska
- Laboratory of Proteomics and Mass Spectrometry, Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
| | - Sylwia Kędracka-Krok
- Department of Physical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Jan Rodriguez Parkitna
- Department of Molecular Neuropharmacology, Institute of Pharmacology of the Polish Academy of Sciences, Smetna 12, 31-343, Krakow, Poland
| | - Michał Korostyński
- Department of Molecular Neuropharmacology, Institute of Pharmacology of the Polish Academy of Sciences, Smetna 12, 31-343, Krakow, Poland.
| |
Collapse
|
29
|
Chang CJ, Chang MY, Lee YC, Chen KY, Hsu TI, Wu YH, Chuang JY, Kao TJ. Nck2 is essential for limb trajectory selection by spinal motor axons. Dev Dyn 2018; 247:1043-1056. [DOI: 10.1002/dvdy.24656] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 07/02/2018] [Accepted: 07/03/2018] [Indexed: 11/08/2022] Open
Affiliation(s)
- Chih-Ju Chang
- Department of Neurosurgery; Cathay General Hospital; Taipei Taiwan
- School of Medicine; Fu Jen Catholic University; New Taipei Taiwan
- Departemnt of Mechanical Engineering; National Central University; Taiwan
| | - Ming-Yuan Chang
- Division of Neurosurgery, Department of Surgery; Min-Sheng General Hospital; Taiwan
- Graduate Institute of Neural Regenerative Medicine, College of Medical Science and Technology; Taipei Medical University; Taipei Taiwan
| | - Yi-Chao Lee
- Graduate Institute of Neural Regenerative Medicine, College of Medical Science and Technology; Taipei Medical University; Taipei Taiwan
- Center for Neurotrauma and Neuroregeneration; Taipei Medical University; Taipei Taiwan
| | - Kai-Yun Chen
- Graduate Institute of Neural Regenerative Medicine, College of Medical Science and Technology; Taipei Medical University; Taipei Taiwan
- Center for Neurotrauma and Neuroregeneration; Taipei Medical University; Taipei Taiwan
| | - Tsung-I Hsu
- Center for Neurotrauma and Neuroregeneration; Taipei Medical University; Taipei Taiwan
| | - Yi-Hsin Wu
- Graduate Institute of Neural Regenerative Medicine, College of Medical Science and Technology; Taipei Medical University; Taipei Taiwan
- Center for Neurotrauma and Neuroregeneration; Taipei Medical University; Taipei Taiwan
| | - Jian-Ying Chuang
- Graduate Institute of Neural Regenerative Medicine, College of Medical Science and Technology; Taipei Medical University; Taipei Taiwan
- Center for Neurotrauma and Neuroregeneration; Taipei Medical University; Taipei Taiwan
| | - Tzu-Jen Kao
- Graduate Institute of Neural Regenerative Medicine, College of Medical Science and Technology; Taipei Medical University; Taipei Taiwan
- Center for Neurotrauma and Neuroregeneration; Taipei Medical University; Taipei Taiwan
| |
Collapse
|
30
|
Haider N, Dusseault J, Larose L. Nck1 Deficiency Impairs Adipogenesis by Activation of PDGFRα in Preadipocytes. iScience 2018; 6:22-37. [PMID: 30240612 PMCID: PMC6137712 DOI: 10.1016/j.isci.2018.07.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 05/22/2018] [Accepted: 07/13/2018] [Indexed: 02/06/2023] Open
Abstract
Obesity results from an excessive expansion of white adipose tissue (WAT), which is still poorly understood from an etiologic-mechanistic perspective. Here, we report that Nck1, a Src homology domain-containing adaptor, is upregulated during WAT expansion and in vitro adipogenesis. In agreement, Nck1 mRNA correlates positively with peroxisome proliferator-activated receptor (PPAR) γ and adiponectin mRNAs in the WAT of obese humans, whereas Nck1-deficient mice display smaller WAT depots with reduced number of adipocyte precursors and accumulation of extracellular matrix. Furthermore, silencing Nck1 in 3T3-L1 preadipocytes increases the proliferation and expression of genes encoding collagen, whereas it decreases the expression of adipogenic markers and impairs adipogenesis. Silencing Nck1 in 3T3-L1 preadipocytes also promotes the expression of platelet-derived growth factor (PDGF)-A and platelet-derived growth factor receptor (PDGFR) α activation and signaling. Preventing PDGFRα activation using imatinib, or through PDGF-A or PDGFRα deficiency, inhibits collagen expression in Nck1-deficient preadipocytes. Finally, imatinib rescues differentiation of Nck1-deficient preadipocytes. Altogether, our findings reveal that Nck1 modulates WAT development through PDGFRα-dependent remodeling of preadipocytes.
Collapse
Affiliation(s)
- Nida Haider
- Division of Experimental Medicine, Department of Medicine, McGill University and The Research Institute of McGill University Health Centre, Glen Site, Bloc E, Rm E02-7244, 1001 Decarie Boulevard, Montreal, QC H4A 3J1 Canada
| | - Julie Dusseault
- Division of Experimental Medicine, Department of Medicine, McGill University and The Research Institute of McGill University Health Centre, Glen Site, Bloc E, Rm E02-7244, 1001 Decarie Boulevard, Montreal, QC H4A 3J1 Canada
| | - Louise Larose
- Division of Experimental Medicine, Department of Medicine, McGill University and The Research Institute of McGill University Health Centre, Glen Site, Bloc E, Rm E02-7244, 1001 Decarie Boulevard, Montreal, QC H4A 3J1 Canada.
| |
Collapse
|
31
|
Jacquet K, Banerjee SL, Chartier FJM, Elowe S, Bisson N. Proteomic Analysis of NCK1/2 Adaptors Uncovers Paralog-specific Interactions That Reveal a New Role for NCK2 in Cell Abscission During Cytokinesis. Mol Cell Proteomics 2018; 17:1979-1990. [PMID: 30002203 DOI: 10.1074/mcp.ra118.000689] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Revised: 07/11/2018] [Indexed: 12/31/2022] Open
Abstract
Signals from cell surface receptors are often relayed via adaptor proteins. NCK1 and NCK2 are Src-Homology (SH) 2 and 3 domain adaptors that regulate processes requiring a remodeling of the actin cytoskeleton. Evidence from gene inactivation in mouse suggests that NCK1 and NCK2 are functionally redundant, although recent reports support the idea of unique functions for NCK1 and NCK2. We sought to examine this question further by delineating NCK1- and NCK2-specific signaling networks. We used both affinity purification-mass spectrometry and BioID proximity labeling to identify NCK1/2 signaling networks comprised of 98 proteins. Strikingly, we found 30 proteins restricted to NCK1 and 28 proteins specifically associated with NCK2, suggesting differences in their function. We report that Nck2 -/-, but not Nck1 -/- mouse embryo fibroblasts (MEFs) are multinucleated and display extended protrusions reminiscent of intercellular bridges, which correlate with an extended time spent in cytokinesis as well as a failure of a significant proportion of cells to complete abscission. Our data also show that the midbody of NCK2-deficient cells is not only increased in length, but also altered in composition, as judged by the mislocalization of AURKB, PLK1 and ECT2. Finally, we show that NCK2 function during cytokinesis requires its SH2 domain. Taken together, our data delineate the first high-confidence interactome for NCK1/2 adaptors and highlight several proteins specifically associated with either protein. Thus, contrary to what is generally accepted, we demonstrate that NCK1 and NCK2 are not completely redundant, and shed light on a previously uncharacterized function for the NCK2 adaptor protein in cell division.
Collapse
Affiliation(s)
- Kévin Jacquet
- From the ‡Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe Oncologie, Québec G1R 2J6, QC, Canada.,§Centre de recherche sur le cancer de l'Université Laval, Québec G1R 2J6, QC, Canada.,¶PROTEO-Quebec Network for Research on Protein Function, Engineering, and Applications, Québec G1V 0A6, QC, Canada
| | - Sara L Banerjee
- From the ‡Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe Oncologie, Québec G1R 2J6, QC, Canada.,§Centre de recherche sur le cancer de l'Université Laval, Québec G1R 2J6, QC, Canada.,¶PROTEO-Quebec Network for Research on Protein Function, Engineering, and Applications, Québec G1V 0A6, QC, Canada
| | - François J M Chartier
- From the ‡Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe Oncologie, Québec G1R 2J6, QC, Canada.,§Centre de recherche sur le cancer de l'Université Laval, Québec G1R 2J6, QC, Canada.,¶PROTEO-Quebec Network for Research on Protein Function, Engineering, and Applications, Québec G1V 0A6, QC, Canada
| | - Sabine Elowe
- §Centre de recherche sur le cancer de l'Université Laval, Québec G1R 2J6, QC, Canada.,¶PROTEO-Quebec Network for Research on Protein Function, Engineering, and Applications, Québec G1V 0A6, QC, Canada.,‖Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe Reproduction, santé de la mère et de l'enfant, Québec G1V 4G2, QC, Canada.,**Department of Pediatrics, Université Laval, Québec, QC, Canada
| | - Nicolas Bisson
- From the ‡Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe Oncologie, Québec G1R 2J6, QC, Canada; .,§Centre de recherche sur le cancer de l'Université Laval, Québec G1R 2J6, QC, Canada.,¶PROTEO-Quebec Network for Research on Protein Function, Engineering, and Applications, Québec G1V 0A6, QC, Canada.,‡‡Department of Molecular Biology, Medical Biochemistry and Pathology, Université Laval, Québec G1V 0A6, QC, Canada
| |
Collapse
|
32
|
Borroto A, Reyes-Garau D, Jiménez MA, Carrasco E, Moreno B, Martínez-Pasamar S, Cortés JR, Perona A, Abia D, Blanco S, Fuentes M, Arellano I, Lobo J, Heidarieh H, Rueda J, Esteve P, Cibrián D, Martinez-Riaño A, Mendoza P, Prieto C, Calleja E, Oeste CL, Orfao A, Fresno M, Sánchez-Madrid F, Alcamí A, Bovolenta P, Martín P, Villoslada P, Morreale A, Messeguer A, Alarcon B. First-in-class inhibitor of the T cell receptor for the treatment of autoimmune diseases. Sci Transl Med 2017; 8:370ra184. [PMID: 28003549 DOI: 10.1126/scitranslmed.aaf2140] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 08/25/2016] [Indexed: 12/14/2022]
Abstract
Modulating T cell activation is critical for treating autoimmune diseases but requires avoiding concomitant opportunistic infections. Antigen binding to the T cell receptor (TCR) triggers the recruitment of the cytosolic adaptor protein Nck to a proline-rich sequence in the cytoplasmic tail of the TCR's CD3ε subunit. Through virtual screening and using combinatorial chemistry, we have generated an orally available, low-molecular weight inhibitor of the TCR-Nck interaction that selectively inhibits TCR-triggered T cell activation with an IC50 (median inhibitory concentration) ~1 nM. By modulating TCR signaling, the inhibitor prevented the development of psoriasis and asthma and, furthermore, exerted a long-lasting therapeutic effect in a model of autoimmune encephalomyelitis. However, it did not prevent the generation of a protective memory response against a mouse pathogen, suggesting that the compound might not exert its effects through immunosuppression. These results suggest that inhibiting an immediate TCR signal has promise for treating a broad spectrum of human T cell-mediated autoimmune and inflammatory diseases.
Collapse
Affiliation(s)
- Aldo Borroto
- Centro de Biología Molecular Severo Ochoa, Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), Madrid, Spain
| | - Diana Reyes-Garau
- Centro de Biología Molecular Severo Ochoa, Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), Madrid, Spain
| | | | - Esther Carrasco
- Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Barcelona, Spain
| | - Beatriz Moreno
- Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS)-Hospital Clinic, Barcelona, Spain
| | - Sara Martínez-Pasamar
- Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS)-Hospital Clinic, Barcelona, Spain
| | - José R Cortés
- Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Almudena Perona
- Centro de Biología Molecular Severo Ochoa, Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), Madrid, Spain
| | - David Abia
- Centro de Biología Molecular Severo Ochoa, Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), Madrid, Spain
| | - Soledad Blanco
- Centro de Biología Molecular Severo Ochoa, Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), Madrid, Spain
| | - Manuel Fuentes
- Centro de Investigación del Cáncer, University of Salamanca-CSIC, Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
| | - Irene Arellano
- Centro de Biología Molecular Severo Ochoa, Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), Madrid, Spain
| | - Juan Lobo
- Centro de Biología Molecular Severo Ochoa, Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), Madrid, Spain
| | - Haleh Heidarieh
- Centro de Biología Molecular Severo Ochoa, Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), Madrid, Spain
| | - Javier Rueda
- Centro de Biología Molecular Severo Ochoa, Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), Madrid, Spain
| | - Pilar Esteve
- Centro de Biología Molecular Severo Ochoa, Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), Madrid, Spain
| | - Danay Cibrián
- Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Ana Martinez-Riaño
- Centro de Biología Molecular Severo Ochoa, Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), Madrid, Spain
| | - Pilar Mendoza
- Centro de Biología Molecular Severo Ochoa, Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), Madrid, Spain
| | - Cristina Prieto
- Centro de Biología Molecular Severo Ochoa, Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), Madrid, Spain
| | - Enrique Calleja
- Centro de Biología Molecular Severo Ochoa, Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), Madrid, Spain
| | - Clara L Oeste
- Centro de Biología Molecular Severo Ochoa, Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), Madrid, Spain
| | - Alberto Orfao
- Centro de Investigación del Cáncer, University of Salamanca-CSIC, Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
| | - Manuel Fresno
- Centro de Biología Molecular Severo Ochoa, Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), Madrid, Spain
| | | | - Antonio Alcamí
- Centro de Biología Molecular Severo Ochoa, Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), Madrid, Spain
| | - Paola Bovolenta
- Centro de Biología Molecular Severo Ochoa, Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), Madrid, Spain
| | - Pilar Martín
- Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Pablo Villoslada
- Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS)-Hospital Clinic, Barcelona, Spain
| | - Antonio Morreale
- Centro de Biología Molecular Severo Ochoa, Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), Madrid, Spain
| | - Angel Messeguer
- Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Barcelona, Spain
| | - Balbino Alarcon
- Centro de Biología Molecular Severo Ochoa, Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), Madrid, Spain.
| |
Collapse
|
33
|
Morris DC, Popp JL, Tang LK, Gibbs HC, Schmitt E, Chaki SP, Bywaters BC, Yeh AT, Porter WW, Burghardt RC, Barhoumi R, Rivera GM. Nck deficiency is associated with delayed breast carcinoma progression and reduced metastasis. Mol Biol Cell 2017; 28:3500-3516. [PMID: 28954862 PMCID: PMC5683761 DOI: 10.1091/mbc.e17-02-0106] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 09/15/2017] [Accepted: 09/20/2017] [Indexed: 12/16/2022] Open
Abstract
Nck promotes breast carcinoma progression and metastasis by directing the polarized interaction of carcinoma cells with collagen fibrils, decreasing actin turnover, and enhancing the localization and activity of MMP14 at the cell surface through modulation of the spatiotemporal activation of Cdc42 and RhoA. Although it is known that noncatalytic region of tyrosine kinase (Nck) regulates cell adhesion and migration by bridging tyrosine phosphorylation with cytoskeletal remodeling, the role of Nck in tumorigenesis and metastasis has remained undetermined. Here we report that Nck is required for the growth and vascularization of primary tumors and lung metastases in a breast cancer xenograft model as well as extravasation following injection of carcinoma cells into the tail vein. We provide evidence that Nck directs the polarization of cell–matrix interactions for efficient migration in three-dimensional microenvironments. We show that Nck advances breast carcinoma cell invasion by regulating actin dynamics at invadopodia and enhancing focalized extracellular matrix proteolysis by directing the delivery and accumulation of MMP14 at the cell surface. We find that Nck-dependent cytoskeletal changes are mechanistically linked to enhanced RhoA but restricted spatiotemporal activation of Cdc42. Using a combination of protein silencing and forced expression of wild-type/constitutively active variants, we provide evidence that Nck is an upstream regulator of RhoA-dependent, MMP14-mediated breast carcinoma cell invasion. By identifying Nck as an important driver of breast carcinoma progression and metastasis, these results lay the groundwork for future studies assessing the therapeutic potential of targeting Nck in aggressive cancers.
Collapse
Affiliation(s)
- David C Morris
- Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas 77843-4467
| | - Julia L Popp
- Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas 77843-4467
| | - Leung K Tang
- Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas 77843-4467
| | - Holly C Gibbs
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas 77843-4467
| | - Emily Schmitt
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas 77843-4467
| | - Sankar P Chaki
- Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas 77843-4467
| | - Briana C Bywaters
- Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas 77843-4467
| | - Alvin T Yeh
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas 77843-4467
| | - Weston W Porter
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas 77843-4467
| | - Robert C Burghardt
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas 77843-4467
| | - Rola Barhoumi
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas 77843-4467
| | - Gonzalo M Rivera
- Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas 77843-4467
| |
Collapse
|
34
|
IRE1α links Nck1 deficiency to attenuated PTP1B expression in HepG2 cells. Cell Signal 2017; 36:79-90. [DOI: 10.1016/j.cellsig.2017.04.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 04/11/2017] [Accepted: 04/23/2017] [Indexed: 12/23/2022]
|
35
|
Dubrac A, Genet G, Ola R, Zhang F, Pibouin-Fragner L, Han J, Zhang J, Thomas JL, Chedotal A, Schwartz MA, Eichmann A. Targeting NCK-Mediated Endothelial Cell Front-Rear Polarity Inhibits Neovascularization. Circulation 2015; 133:409-21. [PMID: 26659946 DOI: 10.1161/circulationaha.115.017537] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 12/04/2015] [Indexed: 12/22/2022]
Abstract
BACKGROUND Sprouting angiogenesis is a key process driving blood vessel growth in ischemic tissues and an important drug target in a number of diseases, including wet macular degeneration and wound healing. Endothelial cells forming the sprout must develop front-rear polarity to allow sprout extension. The adaptor proteins Nck1 and 2 are known regulators of cytoskeletal dynamics and polarity, but their function in angiogenesis is poorly understood. Here, we show that the Nck adaptors are required for endothelial cell front-rear polarity and migration downstream of the angiogenic growth factors VEGF-A and Slit2. METHODS AND RESULTS Mice carrying inducible, endothelial-specific Nck1/2 deletions fail to develop front-rear polarized vessel sprouts and exhibit severe angiogenesis defects in the postnatal retina and during embryonic development. Inactivation of NCK1 and 2 inhibits polarity by preventing Cdc42 and Pak2 activation by VEGF-A and Slit2. Mechanistically, NCK binding to ROBO1 is required for both Slit2- and VEGF-induced front-rear polarity. Selective inhibition of polarized endothelial cell migration by targeting Nck1/2 prevents hypersprouting induced by Notch or Bmp signaling inhibition, and pathological ocular neovascularization and wound healing, as well. CONCLUSIONS These data reveal a novel signal integration mechanism involving NCK1/2, ROBO1/2, and VEGFR2 that controls endothelial cell front-rear polarity during sprouting angiogenesis.
Collapse
Affiliation(s)
- Alexandre Dubrac
- From Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT (A.D., G.G., R.O., F.Z., J.H., J.Z., J.-L.T., A.E.); INSERM U1050, Collège de France, Center for Interdisciplinary Research in Biology (CIRB), Paris (L.P.-F., A.E.); Department of Neurology, Yale University School of Medicine, New Haven, CT (J.-L.T.); Institut du Cerveau et de la Moelle, Inserm, Université Pierre et Marie Curie, Paris, France (J.-L.T.); Sorbonne Universités, UPMC Universités Paris 06, INSERM, UMR-S968, CNRS, UMR-7210, Institut de la Vision, France (A.C.); Departments of Cell Biology and Biomedical Engineering, Yale University, New Haven, CT (M.A.S.); and Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT (A.E.)
| | - Gael Genet
- From Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT (A.D., G.G., R.O., F.Z., J.H., J.Z., J.-L.T., A.E.); INSERM U1050, Collège de France, Center for Interdisciplinary Research in Biology (CIRB), Paris (L.P.-F., A.E.); Department of Neurology, Yale University School of Medicine, New Haven, CT (J.-L.T.); Institut du Cerveau et de la Moelle, Inserm, Université Pierre et Marie Curie, Paris, France (J.-L.T.); Sorbonne Universités, UPMC Universités Paris 06, INSERM, UMR-S968, CNRS, UMR-7210, Institut de la Vision, France (A.C.); Departments of Cell Biology and Biomedical Engineering, Yale University, New Haven, CT (M.A.S.); and Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT (A.E.)
| | - Roxana Ola
- From Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT (A.D., G.G., R.O., F.Z., J.H., J.Z., J.-L.T., A.E.); INSERM U1050, Collège de France, Center for Interdisciplinary Research in Biology (CIRB), Paris (L.P.-F., A.E.); Department of Neurology, Yale University School of Medicine, New Haven, CT (J.-L.T.); Institut du Cerveau et de la Moelle, Inserm, Université Pierre et Marie Curie, Paris, France (J.-L.T.); Sorbonne Universités, UPMC Universités Paris 06, INSERM, UMR-S968, CNRS, UMR-7210, Institut de la Vision, France (A.C.); Departments of Cell Biology and Biomedical Engineering, Yale University, New Haven, CT (M.A.S.); and Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT (A.E.)
| | - Feng Zhang
- From Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT (A.D., G.G., R.O., F.Z., J.H., J.Z., J.-L.T., A.E.); INSERM U1050, Collège de France, Center for Interdisciplinary Research in Biology (CIRB), Paris (L.P.-F., A.E.); Department of Neurology, Yale University School of Medicine, New Haven, CT (J.-L.T.); Institut du Cerveau et de la Moelle, Inserm, Université Pierre et Marie Curie, Paris, France (J.-L.T.); Sorbonne Universités, UPMC Universités Paris 06, INSERM, UMR-S968, CNRS, UMR-7210, Institut de la Vision, France (A.C.); Departments of Cell Biology and Biomedical Engineering, Yale University, New Haven, CT (M.A.S.); and Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT (A.E.)
| | - Laurence Pibouin-Fragner
- From Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT (A.D., G.G., R.O., F.Z., J.H., J.Z., J.-L.T., A.E.); INSERM U1050, Collège de France, Center for Interdisciplinary Research in Biology (CIRB), Paris (L.P.-F., A.E.); Department of Neurology, Yale University School of Medicine, New Haven, CT (J.-L.T.); Institut du Cerveau et de la Moelle, Inserm, Université Pierre et Marie Curie, Paris, France (J.-L.T.); Sorbonne Universités, UPMC Universités Paris 06, INSERM, UMR-S968, CNRS, UMR-7210, Institut de la Vision, France (A.C.); Departments of Cell Biology and Biomedical Engineering, Yale University, New Haven, CT (M.A.S.); and Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT (A.E.)
| | - Jinah Han
- From Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT (A.D., G.G., R.O., F.Z., J.H., J.Z., J.-L.T., A.E.); INSERM U1050, Collège de France, Center for Interdisciplinary Research in Biology (CIRB), Paris (L.P.-F., A.E.); Department of Neurology, Yale University School of Medicine, New Haven, CT (J.-L.T.); Institut du Cerveau et de la Moelle, Inserm, Université Pierre et Marie Curie, Paris, France (J.-L.T.); Sorbonne Universités, UPMC Universités Paris 06, INSERM, UMR-S968, CNRS, UMR-7210, Institut de la Vision, France (A.C.); Departments of Cell Biology and Biomedical Engineering, Yale University, New Haven, CT (M.A.S.); and Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT (A.E.)
| | - Jiasheng Zhang
- From Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT (A.D., G.G., R.O., F.Z., J.H., J.Z., J.-L.T., A.E.); INSERM U1050, Collège de France, Center for Interdisciplinary Research in Biology (CIRB), Paris (L.P.-F., A.E.); Department of Neurology, Yale University School of Medicine, New Haven, CT (J.-L.T.); Institut du Cerveau et de la Moelle, Inserm, Université Pierre et Marie Curie, Paris, France (J.-L.T.); Sorbonne Universités, UPMC Universités Paris 06, INSERM, UMR-S968, CNRS, UMR-7210, Institut de la Vision, France (A.C.); Departments of Cell Biology and Biomedical Engineering, Yale University, New Haven, CT (M.A.S.); and Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT (A.E.)
| | - Jean-Léon Thomas
- From Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT (A.D., G.G., R.O., F.Z., J.H., J.Z., J.-L.T., A.E.); INSERM U1050, Collège de France, Center for Interdisciplinary Research in Biology (CIRB), Paris (L.P.-F., A.E.); Department of Neurology, Yale University School of Medicine, New Haven, CT (J.-L.T.); Institut du Cerveau et de la Moelle, Inserm, Université Pierre et Marie Curie, Paris, France (J.-L.T.); Sorbonne Universités, UPMC Universités Paris 06, INSERM, UMR-S968, CNRS, UMR-7210, Institut de la Vision, France (A.C.); Departments of Cell Biology and Biomedical Engineering, Yale University, New Haven, CT (M.A.S.); and Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT (A.E.)
| | - Alain Chedotal
- From Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT (A.D., G.G., R.O., F.Z., J.H., J.Z., J.-L.T., A.E.); INSERM U1050, Collège de France, Center for Interdisciplinary Research in Biology (CIRB), Paris (L.P.-F., A.E.); Department of Neurology, Yale University School of Medicine, New Haven, CT (J.-L.T.); Institut du Cerveau et de la Moelle, Inserm, Université Pierre et Marie Curie, Paris, France (J.-L.T.); Sorbonne Universités, UPMC Universités Paris 06, INSERM, UMR-S968, CNRS, UMR-7210, Institut de la Vision, France (A.C.); Departments of Cell Biology and Biomedical Engineering, Yale University, New Haven, CT (M.A.S.); and Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT (A.E.)
| | - Martin A Schwartz
- From Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT (A.D., G.G., R.O., F.Z., J.H., J.Z., J.-L.T., A.E.); INSERM U1050, Collège de France, Center for Interdisciplinary Research in Biology (CIRB), Paris (L.P.-F., A.E.); Department of Neurology, Yale University School of Medicine, New Haven, CT (J.-L.T.); Institut du Cerveau et de la Moelle, Inserm, Université Pierre et Marie Curie, Paris, France (J.-L.T.); Sorbonne Universités, UPMC Universités Paris 06, INSERM, UMR-S968, CNRS, UMR-7210, Institut de la Vision, France (A.C.); Departments of Cell Biology and Biomedical Engineering, Yale University, New Haven, CT (M.A.S.); and Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT (A.E.)
| | - Anne Eichmann
- From Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT (A.D., G.G., R.O., F.Z., J.H., J.Z., J.-L.T., A.E.); INSERM U1050, Collège de France, Center for Interdisciplinary Research in Biology (CIRB), Paris (L.P.-F., A.E.); Department of Neurology, Yale University School of Medicine, New Haven, CT (J.-L.T.); Institut du Cerveau et de la Moelle, Inserm, Université Pierre et Marie Curie, Paris, France (J.-L.T.); Sorbonne Universités, UPMC Universités Paris 06, INSERM, UMR-S968, CNRS, UMR-7210, Institut de la Vision, France (A.C.); Departments of Cell Biology and Biomedical Engineering, Yale University, New Haven, CT (M.A.S.); and Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT (A.E.).
| |
Collapse
|
36
|
Shp2 Associates with and Enhances Nephrin Tyrosine Phosphorylation and Is Necessary for Foot Process Spreading in Mouse Models of Podocyte Injury. Mol Cell Biol 2015; 36:596-614. [PMID: 26644409 DOI: 10.1128/mcb.00956-15] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 11/24/2015] [Indexed: 11/20/2022] Open
Abstract
In most forms of glomerular diseases, loss of size selectivity by the kidney filtration barrier is associated with changes in the morphology of podocytes. The kidney filtration barrier is comprised of the endothelial lining, the glomerular basement membrane, and the podocyte intercellular junction, or slit diaphragm. The cell adhesion proteins nephrin and neph1 localize to the slit diaphragm and transduce signals in a Src family kinase Fyn-mediated tyrosine phosphorylation-dependent manner. Studies in cell culture suggest nephrin phosphorylation-dependent signaling events are primarily involved in regulation of actin dynamics and lamellipodium formation. Nephrin phosphorylation is a proximal event that occurs both during development and following podocyte injury. We hypothesized that abrogation of nephrin phosphorylation following injury would prevent nephrin-dependent actin remodeling and foot process morphological changes. Utilizing a biased screening approach, we found nonreceptor Src homology 2 (sh2) domain-containing phosphatase Shp2 to be associated with phosphorylated nephrin. We observed an increase in nephrin tyrosine phosphorylation in the presence of Shp2 in cell culture studies. In the human glomerulopathies minimal-change nephrosis and membranous nephropathy, there is an increase in Shp2 phosphorylation, a marker of increased Shp2 activity. Mouse podocytes lacking Shp2 do not develop foot process spreading when subjected to podocyte injury in vivo using protamine sulfate or nephrotoxic serum (NTS). In the NTS model, we observed a lack of foot process spreading in mouse podocytes with Shp2 deleted and smaller amounts of proteinuria. Taken together, these results suggest that Shp2-dependent signaling events are necessary for changes in foot process structure and function following injury.
Collapse
|
37
|
Paensuwan P, Hartl FA, Yousefi OS, Ngoenkam J, Wipa P, Beck-Garcia E, Dopfer EP, Khamsri B, Sanguansermsri D, Minguet S, Schamel WW, Pongcharoen S. Nck Binds to the T Cell Antigen Receptor Using Its SH3.1 and SH2 Domains in a Cooperative Manner, Promoting TCR Functioning. THE JOURNAL OF IMMUNOLOGY 2015; 196:448-58. [PMID: 26590318 DOI: 10.4049/jimmunol.1500958] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 10/23/2015] [Indexed: 11/19/2022]
Abstract
Ligand binding to the TCR causes a conformational change at the CD3 subunits to expose the CD3ε cytoplasmic proline-rich sequence (PRS). It was suggested that the PRS is important for TCR signaling and T cell activation. It has been shown that the purified, recombinant SH3.1 domain of the adaptor molecule noncatalytic region of tyrosine kinase (Nck) can bind to the exposed PRS of CD3ε, but the molecular mechanism of how full-length Nck binds to the TCR in cells has not been investigated so far. Using the in situ proximity ligation assay and copurifications, we show that the binding of Nck to the TCR requires partial phosphorylation of CD3ε, as it is based on two cooperating interactions. First, the SH3.1(Nck) domain has to bind to the nonphosphorylated and exposed PRS, that is, the first ITAM tyrosine has to be in the unphosphorylated state. Second, the SH2(Nck) domain has to bind to the second ITAM tyrosine in the phosphorylated state. Likewise, mutations of the SH3.1 and SH2 domains in Nck1 resulted in the loss of Nck1 binding to the TCR. Furthermore, expression of an SH3.1-mutated Nck impaired TCR signaling and T cell activation. Our data suggest that the exact pattern of CD3ε phosphorylation is critical for TCR functioning.
Collapse
Affiliation(s)
- Pussadee Paensuwan
- Department of Microbiology and Parasitology, Faculty of Medical Science, Naresuan University, Phitsanulok 65000, Thailand
| | - Frederike A Hartl
- Department of Molecular Immunology, Faculty of Biology, BIOSS Centre for Biological Signaling Studies and Centre of Chronic Immunodeficiency, University of Freiburg, Freiburg 79108, Germany
| | - O Sascha Yousefi
- Department of Molecular Immunology, Faculty of Biology, BIOSS Centre for Biological Signaling Studies and Centre of Chronic Immunodeficiency, University of Freiburg, Freiburg 79108, Germany; Spemann Graduate School of Biology and Medicine, Albert Ludwigs University Freiburg, Freiburg 79104, Germany
| | - Jatuporn Ngoenkam
- Department of Microbiology and Parasitology, Faculty of Medical Science, Naresuan University, Phitsanulok 65000, Thailand
| | - Piyamaporn Wipa
- Department of Microbiology and Parasitology, Faculty of Medical Science, Naresuan University, Phitsanulok 65000, Thailand
| | - Esmeralda Beck-Garcia
- Department of Molecular Immunology, Faculty of Biology, BIOSS Centre for Biological Signaling Studies and Centre of Chronic Immunodeficiency, University of Freiburg, Freiburg 79108, Germany; International Max Planck Research School for Molecular and Cellular Biology, Freiburg 79108, Germany
| | - Elaine P Dopfer
- Department of Molecular Immunology, Faculty of Biology, BIOSS Centre for Biological Signaling Studies and Centre of Chronic Immunodeficiency, University of Freiburg, Freiburg 79108, Germany
| | - Boonruang Khamsri
- Department of Microbiology and Parasitology, Faculty of Medical Science, Naresuan University, Phitsanulok 65000, Thailand
| | - Donruedee Sanguansermsri
- Department of Microbiology and Parasitology, Faculty of Medical Science, Naresuan University, Phitsanulok 65000, Thailand
| | - Susana Minguet
- Department of Molecular Immunology, Faculty of Biology, BIOSS Centre for Biological Signaling Studies and Centre of Chronic Immunodeficiency, University of Freiburg, Freiburg 79108, Germany
| | - Wolfgang W Schamel
- Department of Molecular Immunology, Faculty of Biology, BIOSS Centre for Biological Signaling Studies and Centre of Chronic Immunodeficiency, University of Freiburg, Freiburg 79108, Germany;
| | - Sutatip Pongcharoen
- Centre of Excellence in Medical Biotechnology, Faculty of Medical Science, Naresuan University, Phitsanulok 65000, Thailand; Research Center for Academic Excellence in Petroleum, Petrochemical and Advanced Materials, Faculty of Science, Naresuan University, Phitsanulok 65000, Thailand; and Department of Medicine, Faculty of Medicine, Naresuan University, Phitsanulok 65000, Thailand
| |
Collapse
|
38
|
Conserved interdomain linker promotes phase separation of the multivalent adaptor protein Nck. Proc Natl Acad Sci U S A 2015; 112:E6426-35. [PMID: 26553976 DOI: 10.1073/pnas.1508778112] [Citation(s) in RCA: 131] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The organization of membranes, the cytosol, and the nucleus of eukaryotic cells can be controlled through phase separation of lipids, proteins, and nucleic acids. Collective interactions of multivalent molecules mediated by modular binding domains can induce gelation and phase separation in several cytosolic and membrane-associated systems. The adaptor protein Nck has three SRC-homology 3 (SH3) domains that bind multiple proline-rich segments in the actin regulatory protein neuronal Wiskott-Aldrich syndrome protein (N-WASP) and an SH2 domain that binds to multiple phosphotyrosine sites in the adhesion protein nephrin, leading to phase separation. Here, we show that the 50-residue linker between the first two SH3 domains of Nck enhances phase separation of Nck/N-WASP/nephrin assemblies. Two linear motifs within this element, as well as its overall positively charged character, are important for this effect. The linker increases the driving force for self-assembly of Nck, likely through weak interactions with the second SH3 domain, and this effect appears to promote phase separation. The linker sequence is highly conserved, suggesting that the sequence determinants of the driving forces for phase separation may be generally important to Nck functions. Our studies demonstrate that linker regions between modular domains can contribute to the driving forces for self-assembly and phase separation of multivalent proteins.
Collapse
|
39
|
Regulation of sarcoma cell migration, invasion and invadopodia formation by AFAP1L1 through a phosphotyrosine-dependent pathway. Oncogene 2015. [DOI: 10.1038/onc.2015.272] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
40
|
Hem CD, Sundvold-Gjerstad V, Granum S, Koll L, Abrahamsen G, Buday L, Spurkland A. T cell specific adaptor protein (TSAd) promotes interaction of Nck with Lck and SLP-76 in T cells. Cell Commun Signal 2015; 13:31. [PMID: 26163016 PMCID: PMC4499191 DOI: 10.1186/s12964-015-0109-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 07/03/2015] [Indexed: 11/12/2022] Open
Abstract
Background The Lck and Src binding adaptor protein TSAd (T cell specific adaptor) regulates actin polymerization in T cells and endothelial cells. The molecular details as to how TSAd regulates this process remain to be elucidated. Results To identify novel interaction partners for TSAd, we used a scoring matrix-assisted ligand algorithm (SMALI), and found that the Src homology 2 (SH2) domain of the actin regulator Non-catalytic region of tyrosine kinase adaptor protein (Nck) potentially binds to TSAd phosphorylated on Tyr280 (pTyr280) and pTyr305. These predictions were confirmed by peptide array analysis, showing direct binding of recombinant Nck SH2 to both pTyr280 and pTyr305 on TSAd. In addition, the SH3 domains of Nck interacted with the proline rich region (PRR) of TSAd. Pull-down and immunoprecipitation experiments further confirmed the Nck-TSAd interactions through Nck SH2 and SH3 domains. In line with this Nck and TSAd co-localized in Jurkat cells as assessed by confocal microscopy and imaging flow cytometry. Co-immunoprecipitation experiments in Jurkat TAg cells lacking TSAd revealed that TSAd promotes interaction of Nck with Lck and SLP-76, but not Vav1. TSAd expressing Jurkat cells contained more polymerized actin, an effect dependent on TSAd exon 7, which includes interactions sites for both Nck and Lck. Conclusions TSAd binds to and co-localizes with Nck. Expression of TSAd increases both Nck-Lck and Nck-SLP-76 interaction in T cells. Recruitment of Lck and SLP-76 to Nck by TSAd could be one mechanism by which TSAd promotes actin polymerization in activated T cells.
Collapse
Affiliation(s)
- Cecilie Dahl Hem
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, 0317, Norway.
| | - Vibeke Sundvold-Gjerstad
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, 0317, Norway.
| | - Stine Granum
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, 0317, Norway.
| | - Lise Koll
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, 0317, Norway.
| | - Greger Abrahamsen
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, 0317, Norway.
| | - Laszlo Buday
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, 1117, Hungary.
| | - Anne Spurkland
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, 0317, Norway. .,Institute of Basal Medical Sciences, University of Oslo, PB 1105, Blindern, Oslo, 0317, Norway.
| |
Collapse
|
41
|
Chaki SP, Barhoumi R, Rivera GM. Actin remodeling by Nck regulates endothelial lumen formation. Mol Biol Cell 2015; 26:3047-60. [PMID: 26157164 PMCID: PMC4551318 DOI: 10.1091/mbc.e15-06-0338] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 07/01/2015] [Indexed: 01/03/2023] Open
Abstract
Nck-dependent actin remodeling enables endothelial morphogenesis by promoting cell elongation and proper organization of VE-cadherin intercellular junctions. Nck determines spatiotemporal patterns of Cdc42/aPKC activation to regulate endothelial apical-basal polarity and lumen formation. Multiple angiogenic cues modulate phosphotyrosine signaling to promote vasculogenesis and angiogenesis. Despite its functional and clinical importance, how vascular cells integrate phosphotyrosine-dependent signaling to elicit cytoskeletal changes required for endothelial morphogenesis remains poorly understood. The family of Nck adaptors couples phosphotyrosine signals with actin dynamics and therefore is well positioned to orchestrate cellular processes required in vascular formation and remodeling. Culture of endothelial cells in three-dimensional collagen matrices in the presence of VEGF stimulation was combined with molecular genetics, optical imaging, and biochemistry to show that Nck-dependent actin remodeling promotes endothelial cell elongation and proper organization of VE-cadherin intercellular junctions. Major morphogenetic defects caused by abrogation of Nck signaling included loss of endothelial apical-basal polarity and impaired lumenization. Time-lapse imaging using a Förster resonance energy transfer biosensor, immunostaining with phospho-specific antibodies, and GST pull-down assays showed that Nck determines spatiotemporal patterns of Cdc42/aPKC activation during endothelial morphogenesis. Our results demonstrate that Nck acts as an important hub integrating angiogenic cues with cytoskeletal changes that enable endothelial apical-basal polarization and lumen formation. These findings point to Nck as an emergent target for effective antiangiogenic therapy.
Collapse
Affiliation(s)
- Sankar P Chaki
- Department of Veterinary Pathobiology, Texas A&M University, College Station, TX 77843-4467
| | - Rola Barhoumi
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843-4467
| | - Gonzalo M Rivera
- Department of Veterinary Pathobiology, Texas A&M University, College Station, TX 77843-4467
| |
Collapse
|
42
|
Lu KH, Keppler S, Leithäuser F, Mattfeldt T, Castello A, Kostezka U, Küblbeck G, Schmitt S, Klevenz A, Prokosch S, Pougialis G, Pawson T, Batista F, Tafuri A, Arnold B. Nck adaptor proteins modulate differentiation and effector function of T cells. J Leukoc Biol 2015; 98:301-11. [PMID: 25995205 DOI: 10.1189/jlb.1hi1114-565r] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Accepted: 04/02/2015] [Indexed: 11/24/2022] Open
Abstract
Understanding the molecular mechanisms regulating T cell reactivity is required for successful reprogramming of immune responses in medical conditions, characterized by dysfunctions of the immune system. Nck proteins are cytoplasmic adaptors mediating diverse cellular functions, including TCR signaling. By enhancing TCR signal strength, Nck proteins influence thymic selection and regulate the size and sensitivity of the peripheral T cell repertoire. Here, we investigated the contribution of Nck proteins to CD4(+) T cell differentiation and effector function using Nck.T(-/-) mice. Impaired GC formation and reduced Tfh were observed in Nck.T(-/-) mice after immunization with T cell-dependent antigens. Th2/Tfh-related cytokines, such as IL-4, IL-10, and IL-21, were decreased in Nck.T(-/-) mice T cells. Moreover, an increased susceptibility to cell death of Tfh cells in Nck.T(-/-) mice was associated with decreased levels of Akt phosphorylation. As a result of this dysregulation in Tfh cells of Nck.T(-/-) mice, we found impaired production and affinity maturation of antibodies against T cell-dependent antigens. Thus, Nck proteins not only participate in thymic selection and generation of the peripheral T cell repertoire but also are involved in the differentiation and effector functions of CD4(+) T cells.
Collapse
Affiliation(s)
- Kun-Hui Lu
- *Molecular Immunology, German Cancer Research Center, Heidelberg, Germany; Lymphocyte Interaction Laboratory, London Research Institute-Cancer Research UK, London, United Kingdom; Department of Pathology, Universitätsklinikum, Ulm, Germany; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada; Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Selina Keppler
- *Molecular Immunology, German Cancer Research Center, Heidelberg, Germany; Lymphocyte Interaction Laboratory, London Research Institute-Cancer Research UK, London, United Kingdom; Department of Pathology, Universitätsklinikum, Ulm, Germany; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada; Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Frank Leithäuser
- *Molecular Immunology, German Cancer Research Center, Heidelberg, Germany; Lymphocyte Interaction Laboratory, London Research Institute-Cancer Research UK, London, United Kingdom; Department of Pathology, Universitätsklinikum, Ulm, Germany; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada; Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Torsten Mattfeldt
- *Molecular Immunology, German Cancer Research Center, Heidelberg, Germany; Lymphocyte Interaction Laboratory, London Research Institute-Cancer Research UK, London, United Kingdom; Department of Pathology, Universitätsklinikum, Ulm, Germany; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada; Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Angelo Castello
- *Molecular Immunology, German Cancer Research Center, Heidelberg, Germany; Lymphocyte Interaction Laboratory, London Research Institute-Cancer Research UK, London, United Kingdom; Department of Pathology, Universitätsklinikum, Ulm, Germany; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada; Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Ulrike Kostezka
- *Molecular Immunology, German Cancer Research Center, Heidelberg, Germany; Lymphocyte Interaction Laboratory, London Research Institute-Cancer Research UK, London, United Kingdom; Department of Pathology, Universitätsklinikum, Ulm, Germany; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada; Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Günter Küblbeck
- *Molecular Immunology, German Cancer Research Center, Heidelberg, Germany; Lymphocyte Interaction Laboratory, London Research Institute-Cancer Research UK, London, United Kingdom; Department of Pathology, Universitätsklinikum, Ulm, Germany; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada; Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Sabine Schmitt
- *Molecular Immunology, German Cancer Research Center, Heidelberg, Germany; Lymphocyte Interaction Laboratory, London Research Institute-Cancer Research UK, London, United Kingdom; Department of Pathology, Universitätsklinikum, Ulm, Germany; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada; Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Alexandra Klevenz
- *Molecular Immunology, German Cancer Research Center, Heidelberg, Germany; Lymphocyte Interaction Laboratory, London Research Institute-Cancer Research UK, London, United Kingdom; Department of Pathology, Universitätsklinikum, Ulm, Germany; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada; Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Sandra Prokosch
- *Molecular Immunology, German Cancer Research Center, Heidelberg, Germany; Lymphocyte Interaction Laboratory, London Research Institute-Cancer Research UK, London, United Kingdom; Department of Pathology, Universitätsklinikum, Ulm, Germany; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada; Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Georg Pougialis
- *Molecular Immunology, German Cancer Research Center, Heidelberg, Germany; Lymphocyte Interaction Laboratory, London Research Institute-Cancer Research UK, London, United Kingdom; Department of Pathology, Universitätsklinikum, Ulm, Germany; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada; Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Tony Pawson
- *Molecular Immunology, German Cancer Research Center, Heidelberg, Germany; Lymphocyte Interaction Laboratory, London Research Institute-Cancer Research UK, London, United Kingdom; Department of Pathology, Universitätsklinikum, Ulm, Germany; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada; Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Facundo Batista
- *Molecular Immunology, German Cancer Research Center, Heidelberg, Germany; Lymphocyte Interaction Laboratory, London Research Institute-Cancer Research UK, London, United Kingdom; Department of Pathology, Universitätsklinikum, Ulm, Germany; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada; Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Anna Tafuri
- *Molecular Immunology, German Cancer Research Center, Heidelberg, Germany; Lymphocyte Interaction Laboratory, London Research Institute-Cancer Research UK, London, United Kingdom; Department of Pathology, Universitätsklinikum, Ulm, Germany; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada; Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Bernd Arnold
- *Molecular Immunology, German Cancer Research Center, Heidelberg, Germany; Lymphocyte Interaction Laboratory, London Research Institute-Cancer Research UK, London, United Kingdom; Department of Pathology, Universitätsklinikum, Ulm, Germany; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada; Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
43
|
Lillico DME, Zwozdesky MA, Pemberton JG, Deutscher JM, Jones LO, Chang JP, Stafford JL. Teleost leukocyte immune-type receptors activate distinct phagocytic modes for target acquisition and engulfment. J Leukoc Biol 2015; 98:235-48. [DOI: 10.1189/jlb.2a0215-039rr] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 04/15/2015] [Indexed: 12/22/2022] Open
|
44
|
Requisite role for Nck adaptors in cardiovascular development, endothelial-to-mesenchymal transition, and directed cell migration. Mol Cell Biol 2015; 35:1573-87. [PMID: 25691664 DOI: 10.1128/mcb.00072-15] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 02/07/2015] [Indexed: 01/06/2023] Open
Abstract
Development of the cardiovascular system is critically dependent on the ability of endothelial cells (ECs) to reorganize their intracellular actin architecture to facilitate migration, adhesion, and morphogenesis. Nck family cytoskeletal adaptors function as key mediators of actin dynamics in numerous cell types, though their role in EC biology remains largely unexplored. Here, we demonstrate an essential requirement for Nck within ECs. Mouse embryos lacking endothelial Nck1/2 expression develop extensive angiogenic defects that result in lethality at about embryonic day 10. Mutant embryos show immature vascular networks, with decreased vessel branching, aberrant perivascular cell recruitment, and reduced cardiac trabeculation. Strikingly, embryos deficient in endothelial Nck also fail to undergo the endothelial-to-mesenchymal transition (EnMT) required for cardiac valve morphogenesis, with loss of Nck disrupting expression of major EnMT markers, as well as suppressing mesenchymal outgrowth. Furthermore, we show that Nck-null ECs are unable to migrate downstream of vascular endothelial growth factor and angiopoietin-1, and they exhibit profound perturbations in cytoskeletal patterning, with disorganized cellular projections, impaired focal adhesion turnover, and disrupted actin-based signaling. Our collective findings thereby reveal a crucial role for Nck as a master regulator within the endothelium to control actin cytoskeleton organization, vascular network remodeling, and EnMT during cardiovascular development.
Collapse
|
45
|
Muscolini M, Camperio C, Porciello N, Caristi S, Capuano C, Viola A, Galandrini R, Tuosto L. Phosphatidylinositol 4–Phosphate 5–Kinase α and Vav1 Mutual Cooperation in CD28-Mediated Actin Remodeling and Signaling Functions. THE JOURNAL OF IMMUNOLOGY 2015; 194:1323-1333. [DOI: 10.4049/jimmunol.1401643] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Abstract
Phosphatidylinositol 4,5–biphosphate (PIP2) is a cell membrane phosphoinositide crucial for cell signaling and activation. Indeed, PIP2 is a pivotal source for second messenger generation and controlling the activity of several proteins regulating cytoskeleton reorganization. Despite its critical role in T cell activation, the molecular mechanisms regulating PIP2 turnover remain largely unknown. In human primary CD4+ T lymphocytes, we have recently demonstrated that CD28 costimulatory receptor is crucial for regulating PIP2 turnover by allowing the recruitment and activation of the lipid kinase phosphatidylinositol 4–phosphate 5–kinase (PIP5Kα). We also identified PIP5Kα as a key modulator of CD28 costimulatory signals leading to the efficient T cell activation. In this study, we extend these data by demonstrating that PIP5Kα recruitment and activation is essential for CD28-mediated cytoskeleton rearrangement necessary for organizing a complete signaling compartment leading to downstream signaling functions. We also identified Vav1 as the linker molecule that couples the C-terminal proline-rich motif of CD28 to the recruitment and activation of PIP5Kα, which in turn cooperates with Vav1 in regulating actin polymerization and CD28 signaling functions.
Collapse
Affiliation(s)
- Michela Muscolini
- *Department of Biology and Biotechnology “Charles Darwin,” Pasteur Institute–Cenci Bolognetti Foundation, Sapienza University, 00185 Rome, Italy
| | - Cristina Camperio
- *Department of Biology and Biotechnology “Charles Darwin,” Pasteur Institute–Cenci Bolognetti Foundation, Sapienza University, 00185 Rome, Italy
| | - Nicla Porciello
- *Department of Biology and Biotechnology “Charles Darwin,” Pasteur Institute–Cenci Bolognetti Foundation, Sapienza University, 00185 Rome, Italy
| | - Silvana Caristi
- *Department of Biology and Biotechnology “Charles Darwin,” Pasteur Institute–Cenci Bolognetti Foundation, Sapienza University, 00185 Rome, Italy
| | - Cristina Capuano
- †Department of Experimental Medicine, Sapienza University, 00185 Rome, Italy
| | - Antonella Viola
- ‡The Venetian Institute of Molecular Medicine, Padova 35129, Italy; and
- §Department of Biomedical Sciences, University of Padova, Padova 35121, Italy
| | | | - Loretta Tuosto
- *Department of Biology and Biotechnology “Charles Darwin,” Pasteur Institute–Cenci Bolognetti Foundation, Sapienza University, 00185 Rome, Italy
| |
Collapse
|
46
|
Baldanzi G, Graziani A. Physiological Signaling and Structure of the HGF Receptor MET. Biomedicines 2014; 3:1-31. [PMID: 28536396 PMCID: PMC5344233 DOI: 10.3390/biomedicines3010001] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 12/09/2014] [Indexed: 12/13/2022] Open
Abstract
The "hepatocyte growth factor" also known as "scatter factor", is a multifunctional cytokine with the peculiar ability of simultaneously triggering epithelial cell proliferation, movement and survival. The combination of those proprieties results in the induction of an epithelial to mesenchymal transition in target cells, fundamental for embryogenesis but also exploited by tumor cells during metastatization. The hepatocyte growth factor receptor, MET, is a proto-oncogene and a prototypical transmembrane tyrosine kinase receptor. Inhere we discuss the MET molecular structure and the hepatocyte growth factor driven physiological signaling which coordinates epithelial proliferation, motility and morphogenesis.
Collapse
Affiliation(s)
- Gianluca Baldanzi
- Department Translational Medicine, University Piemonte Orientale, via Solaroli 17, 28100 Novara, Italy.
| | - Andrea Graziani
- Department Translational Medicine, University Piemonte Orientale, via Solaroli 17, 28100 Novara, Italy.
- Università Vita-Salute San Raffaele, via Olgettina 58, 20132 Milano, Italy.
| |
Collapse
|
47
|
Lettau M, Kabelitz D, Janssen O. SDF1α-induced interaction of the adapter proteins Nck and HS1 facilitates actin polymerization and migration in T cells. Eur J Immunol 2014; 45:551-61. [PMID: 25359136 DOI: 10.1002/eji.201444473] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Revised: 10/07/2014] [Accepted: 10/24/2014] [Indexed: 12/22/2022]
Abstract
Noncatalytic region of tyrosine kinase (Nck) is an adapter protein that comprises one SH2 (Src homology) domain and three SH3 domains. Nck links receptors and receptor-associated tyrosine kinases or adapter proteins to proteins that regulate the actin cytoskeleton. Whereas the SH2 domain binds to phosphorylated receptors or associated phosphoproteins, individual interactions of the SH3 domains with proline-based recognition motifs result in the formation of larger protein complexes. In T cells, changes in cell polarity and morphology during T-cell activation and effector function require the T-cell receptor-mediated recruitment and activation of actin-regulatory proteins to initiate cytoskeletal reorganization at the immunological synapse. We previously identified the adapter protein HS1 as a putative Nck-interacting protein. We now demonstrate that the SH2 domain of Nck specifically interacts with HS1 upon phosphorylation of its tyrosine residue 378. We report that in human T cells, ligation of the chemokine receptor CXCR4 by stromal cell-derived factor 1α (SDF1α) induces a rapid and transient phosphorylation of tyrosine 378 of HS1 resulting in an increased association with Nck. Consequently, siRNA-mediated downregulation of HS1 and/or Nck impairs SDF1α-induced actin polymerization and T-cell migration.
Collapse
Affiliation(s)
- Marcus Lettau
- Institute of Immunology, University Hospital Schleswig-Holstein, Kiel, Germany
| | | | | |
Collapse
|
48
|
Pauker MH, Reicher B, Joseph N, Wortzel I, Jakubowicz S, Noy E, Perl O, Barda-Saad M. WASp family verprolin-homologous protein-2 (WAVE2) and Wiskott-Aldrich syndrome protein (WASp) engage in distinct downstream signaling interactions at the T cell antigen receptor site. J Biol Chem 2014; 289:34503-19. [PMID: 25342748 DOI: 10.1074/jbc.m114.591685] [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: 01/29/2023] Open
Abstract
T cell antigen receptor (TCR) engagement has been shown to activate pathways leading to actin cytoskeletal polymerization and reorganization, which are essential for lymphocyte activation and function. Several actin regulatory proteins were implicated in regulating the actin machinery, such as members of the Wiskott-Aldrich syndrome protein (WASp) family. These include WASp and the WASp family verprolin-homologous protein-2 (WAVE2). Although WASp and WAVE2 share several structural features, the precise regulatory mechanisms and potential redundancy between them have not been fully characterized. Specifically, unlike WASp, the dynamic molecular interactions that regulate WAVE2 recruitment to the cell membrane and specifically to the TCR signaling complex are largely unknown. Here, we identify the molecular mechanism that controls the recruitment of WAVE2 in comparison with WASp. Using fluorescence resonance energy transfer (FRET) and novel triple-color FRET (3FRET) technology, we demonstrate how WAVE2 signaling complexes are dynamically regulated during lymphocyte activation in vivo. We show that, similar to WASp, WAVE2 recruitment to the TCR site depends on protein-tyrosine kinase, ZAP-70, and the adaptors LAT, SLP-76, and Nck. However, in contrast to WASp, WAVE2 leaves this signaling complex and migrates peripherally together with vinculin to the membrane leading edge. Our experiments demonstrate that WASp and WAVE2 differ in their dynamics and their associated proteins. Thus, this study reveals the differential mechanisms regulating the function of these cytoskeletal proteins.
Collapse
Affiliation(s)
- Maor H Pauker
- From the Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 5290002, Israel
| | - Barak Reicher
- From the Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 5290002, Israel
| | - Noah Joseph
- From the Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 5290002, Israel
| | - Inbal Wortzel
- From the Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 5290002, Israel
| | - Shlomi Jakubowicz
- From the Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 5290002, Israel
| | - Elad Noy
- From the Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 5290002, Israel
| | - Orly Perl
- From the Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 5290002, Israel
| | - Mira Barda-Saad
- From the Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 5290002, Israel
| |
Collapse
|
49
|
Banjade S, Rosen MK. Phase transitions of multivalent proteins can promote clustering of membrane receptors. eLife 2014; 3. [PMID: 25321392 PMCID: PMC4238058 DOI: 10.7554/elife.04123] [Citation(s) in RCA: 416] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 10/16/2014] [Indexed: 12/14/2022] Open
Abstract
Clustering of proteins into micrometer-sized structures at membranes is observed in many signaling pathways. Most models of clustering are specific to particular systems, and relationships between physical properties of the clusters and their molecular components are not well understood. We report biochemical reconstitution on supported lipid bilayers of protein clusters containing the adhesion receptor Nephrin and its cytoplasmic partners, Nck and N-WASP. With Nephrin attached to the bilayer, multivalent interactions enable these proteins to polymerize on the membrane surface and undergo two-dimensional phase separation, producing micrometer-sized clusters. Dynamics and thermodynamics of the clusters are modulated by the valencies and affinities of the interacting species. In the presence of the Arp2/3 complex, the clusters assemble actin filaments, suggesting that clustering of regulatory factors could promote local actin assembly at membranes. Interactions between multivalent proteins could be a general mechanism for cytoplasmic adaptor proteins to organize membrane receptors into micrometer-scale signaling zones. DOI:http://dx.doi.org/10.7554/eLife.04123.001 The membrane that surrounds a cell is made up of a mixture of lipid molecules and proteins. Membrane proteins perform a wide range of roles, including transmitting signals into, and out of, cells and helping neighboring cells to stick together. To perform these tasks, these proteins commonly need to bind to other molecules—collectively known as ligands—that are found either inside or outside the cell. Membrane proteins are able to move around within the membrane, and in many systems, ligand binding causes the membrane proteins to cluster together. Although this clustering has been seen in many different systems, no general principles that describe how clustering occurs had been found. Now, Banjade and Rosen have constructed an artificial cell membrane to investigate the clustering of a membrane protein called Nephrin, which is essential for kidneys to function correctly. When it is activated, Nephrin interacts with protein ligands called Nck and N-WASP that are found inside cells and helps filaments of a protein called actin to form. These filaments perform a number of roles including enabling cells to adhere to each other and to move. In Banjade and Rosen's artificial system, when a critical concentration of ligands was exceeded, clusters of Nephrin, Nck and N-WASP suddenly formed. This suggests that the clusters form through a physical process known as ‘phase separation’. Banjade and Rosen found that this critical concentration depends on how strongly the proteins interact and the number of sites they possess to bind each other. Within the clusters, the three proteins formed large polymer chains. The clusters were mobile and, over time, small clusters coalesced into larger clusters. Even though the clusters persisted for hours, individual proteins did not stay in a given cluster for long and instead continuously exchanged back-and-forth between the cluster and its surroundings. When actin and another protein complex that interacts with N-WASP were added to the artificial membrane system, actin filaments began to form at the protein clusters. Banjade and Rosen suggest that such clusters act as ‘signaling zones’ that coordinate the construction of the actin filaments. Regions that are also found in many other signaling proteins mediate the interactions between Nephrin, Nck and N-WASP. Banjade and Rosen therefore suggest that phase separation and protein polymer formation could explain how many different types of membrane proteins form clusters. DOI:http://dx.doi.org/10.7554/eLife.04123.002
Collapse
Affiliation(s)
- Sudeep Banjade
- Department of Biophysics, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, United States
| | - Michael K Rosen
- Department of Biophysics, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, United States
| |
Collapse
|
50
|
Santos MS, Foss SM, Park CK, Voglmaier SM. Protein interactions of the vesicular glutamate transporter VGLUT1. PLoS One 2014; 9:e109824. [PMID: 25334008 PMCID: PMC4198130 DOI: 10.1371/journal.pone.0109824] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Accepted: 09/08/2014] [Indexed: 11/18/2022] Open
Abstract
Exocytotic release of glutamate depends upon loading of the neurotransmitter into synaptic vesicles by vesicular glutamate transporters, VGLUTs. The major isoforms, VGLUT1 and 2, exhibit a complementary pattern of expression in synapses of the adult rodent brain that correlates with the probability of release and potential for plasticity. Indeed, expression of different VGLUT protein isoforms confers different properties of release probability. Expression of VGLUT1 or 2 protein also determines the kinetics of synaptic vesicle recycling. To identify molecular determinants that may be related to reported differences in VGLUT trafficking and glutamate release properties, we investigated some of the intrinsic differences between the two isoforms. VGLUT1 and 2 exhibit a high degree of sequence homology, but differ in their N- and C-termini. While the C-termini of VGLUT1 and 2 share a dileucine-like trafficking motif and a proline-, glutamate-, serine-, and threonine-rich PEST domain, only VGLUT1 contains two polyproline domains and a phosphorylation consensus sequence in a region of acidic amino acids. The interaction of a VGLUT1 polyproline domain with the endocytic protein endophilin recruits VGLUT1 to a fast recycling pathway. To identify trans-acting cellular proteins that interact with the distinct motifs found in the C-terminus of VGLUT1, we performed a series of in vitro biochemical screening assays using the region encompassing the polyproline motifs, phosphorylation consensus sites, and PEST domain. We identify interactors that belong to several classes of proteins that modulate cellular function, including actin cytoskeletal adaptors, ubiquitin ligases, and tyrosine kinases. The nature of these interactions suggests novel avenues to investigate the modulation of synaptic vesicle protein recycling.
Collapse
Affiliation(s)
- Magda S. Santos
- Department of Psychiatry, University of California San Francisco, School of Medicine, San Francisco, California, United States of America
| | - Sarah M. Foss
- Department of Psychiatry, University of California San Francisco, School of Medicine, San Francisco, California, United States of America
- Graduate Program in Cell Biology, University of California San Francisco, School of Medicine, San Francisco, California, United States of America
| | - C. Kevin Park
- Department of Psychiatry, University of California San Francisco, School of Medicine, San Francisco, California, United States of America
| | - Susan M. Voglmaier
- Department of Psychiatry, University of California San Francisco, School of Medicine, San Francisco, California, United States of America
| |
Collapse
|