1
|
Silverman JB, Krystofiak EE, Caplan LR, Lau KS, Tyska MJ. Organization of a cytoskeletal superstructure in the apical domain of intestinal tuft cells. J Cell Biol 2024; 223:e202404070. [PMID: 39352498 PMCID: PMC11457492 DOI: 10.1083/jcb.202404070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 08/26/2024] [Accepted: 09/16/2024] [Indexed: 10/09/2024] Open
Abstract
Tuft cells are a rare epithelial cell type that play important roles in sensing and responding to luminal antigens. A defining morphological feature of this lineage is the actin-rich apical "tuft," which contains large fingerlike protrusions. However, details of the cytoskeletal ultrastructure underpinning the tuft, the molecules involved in building this structure, or how it supports tuft cell biology remain unclear. In the context of the small intestine, we found that tuft cell protrusions are supported by long-core bundles that consist of F-actin crosslinked in a parallel and polarized configuration; they also contain a tuft cell-specific complement of actin-binding proteins that exhibit regionalized localization along the bundle axis. Remarkably, in the sub-apical cytoplasm, the array of core actin bundles interdigitates and co-aligns with a highly ordered network of microtubules. The resulting cytoskeletal superstructure is well positioned to support subcellular transport and, in turn, the dynamic sensing functions of the tuft cell that are critical for intestinal homeostasis.
Collapse
Affiliation(s)
- Jennifer B. Silverman
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Evan E. Krystofiak
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
- Vanderbilt Cell Imaging Shared Resource, Vanderbilt University, Nashville, TN, USA
| | - Leah R. Caplan
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Ken S. Lau
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Matthew J. Tyska
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| |
Collapse
|
2
|
Yang Y, Xu B, Lu W. Phosphorylated ERM regulates meiotic maturation in mouse oocytes. Biochem Biophys Res Commun 2024; 734:150602. [PMID: 39243677 DOI: 10.1016/j.bbrc.2024.150602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 08/18/2024] [Accepted: 08/23/2024] [Indexed: 09/09/2024]
Abstract
The cytoskeleton of mammal oocytes provides structural support to the plasma membrane and contributes to critical cellular dynamic processes such as nuclear positioning, germinal vesicle breakdown, spindle orientation, chromosome segregation, polar body extrusion, and transmembrane signaling pathways. The ERM family (ezrin, radixin and moesin) well known as membrane-cytoskeletal crosslinkers play a crucial role in organizing plasma membrane domains through their capacity to interact with transmembrane proteins and the underlying cytoskeleton. Recent works mainly focused on the structural analysis of the ERM family members and their binding partners, together with multiple functions in cell mitosis, have significantly advanced our understanding of the importance of membrane-cytoskeletal interactions. In the present study, we documented that p-ERM was expressed and localized at cortical and nucleus during mouse oocyte meiosis. p-ERM and microfilaments were colocalized from GV to MII during mouse oocyte maturation. After being treated with cytochalasin B (CB), the F-actin was disassembled. Meanwhile, p-ERM exhibited a diffuse cytoplasmic distribution and no special staining was detected in either the oocyte membrane or condensed chromosomes. p-ERM depletion by trim-away caused the meiotic procedure arrest with a significantly lower polar body extrusion rate. Collectively, these data demonstrate that the subcellular distribution of p-ERM is correlated with microfilaments. Meanwhile, the p-ERM contributes to the first polar extrusion but does not regulate the microfilament assembly.
Collapse
Affiliation(s)
- Yifeng Yang
- Jilin Provincial International Joint Research Center of Animal Breeding & Reproduction Technology, Jilin Agricultural University, Changchun, 130118, China; Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education, Jilin Agricultural University, Jilin, Changchun, 130118, China; College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China; Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun, 130112, China
| | - Baozeng Xu
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun, 130112, China.
| | - Wenfa Lu
- Jilin Provincial International Joint Research Center of Animal Breeding & Reproduction Technology, Jilin Agricultural University, Changchun, 130118, China; Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education, Jilin Agricultural University, Jilin, Changchun, 130118, China; College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China.
| |
Collapse
|
3
|
Chong ZZ, Souayah N. Radixin: Roles in the Nervous System and Beyond. Biomedicines 2024; 12:2341. [PMID: 39457653 PMCID: PMC11504607 DOI: 10.3390/biomedicines12102341] [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: 09/16/2024] [Revised: 10/10/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Radixin is an ERM family protein that includes radixin, moesin, and ezrin. The importance of ERM family proteins has been attracting more attention, and studies on the roles of ERM in biological function and the pathogenesis of some diseases are accumulating. In particular, we have found that radixin is the most dramatically changed ERM protein in elevated glucose-treated Schwann cells. METHOD We systemically review the literature on ERM, radixin in focus, and update the roles of radixin in regulating cell morphology, interaction, and cell signaling pathways. The potential of radixin as a therapeutic target in neurodegenerative diseases and cancer was also discussed. RESULTS Radixin research has focused on its cell functions, activation, and pathogenic roles in some diseases. Radixin and other ERM proteins maintain cell shape, growth, and motility. In the nervous system, radixin has been shown to prevent neurodegeneration and axonal growth. The activation of radixin is through phosphorylation of its conserved threonine residues. Radixin functions in cell signaling pathways by binding to membrane proteins and relaying the cell signals into the cells. Deficiency of radixin has been involved in the pathogenic process of diseases in the central nervous system and diabetic peripheral nerve injury. Moreover, radixin also plays a role in cell growth and drug resistance in multiple cancers. The trials of therapeutic potential through radixin modulation have been accumulating. However, the exact mechanisms underlying the roles of radixin are far from clarification. CONCLUSIONS Radixin plays various roles in cells and is involved in developing neurodegenerative diseases and many types of cancers. Therefore, radixin may be considered a potential target for developing therapeutic strategies for its related diseases. Further elucidation of the function and the cell signaling pathways that are linked to radixin may open the avenue to finding novel therapeutic strategies for diseases in the nervous system and other body systems.
Collapse
Affiliation(s)
- Zhao Zhong Chong
- Department of Neurology, New Jersey Medical School, Rutgers University, 185 S. Orange Ave, Newark, NJ 07103, USA
| | - Nizar Souayah
- Department of Neurology, New Jersey Medical School, Rutgers University, 185 S. Orange Ave, Newark, NJ 07103, USA
- Department of Neurology, New Jersey Medical School, Rutgers University, 90 Bergen Street DOC 8100, Newark, NJ 07101, USA
| |
Collapse
|
4
|
Nhieu J, Wei CW, Ludwig M, Drake JM, Wei LN. CRABP1-complexes in exosome secretion. Cell Commun Signal 2024; 22:381. [PMID: 39075476 PMCID: PMC11285139 DOI: 10.1186/s12964-024-01749-w] [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: 05/10/2024] [Accepted: 07/13/2024] [Indexed: 07/31/2024] Open
Abstract
BACKGROUND Cellular retinoic acid binding protein 1 (CRABP1) mediates rapid, non-canonical activity of retinoic acid (RA) by forming signalosomes via protein-protein interactions. Two signalosomes have been identified previously: CRABP1-MAPK and CRABP1-CaMKII. Crabp1 knockout (CKO) mice exhibited altered exosome profiles, but the mechanism of CRABP1 action was unclear. This study aimed to screen for and identify novel CRABP1 signalosomes that could modulate exosome secretion by using a combinatorial approach involving biochemical, bioinformatic and molecular studies. METHODS Immunoprecipitation coupled with mass spectrometry (IP-MS) identified candidate CRABP1-interacting proteins which were subsequently analyzed using GO Term Enrichment, Functional Annotation Clustering; and Pathway Analysis. Gene expression analysis of CKO samples revealed altered expression of genes related to exosome biogenesis and secretion. The effect of CRABP1 on exosome secretion was then experimentally validated using CKO mice and a Crabp1 knockdown P19 cell line. RESULTS IP-MS identified CRABP1-interacting targets. Bioinformatic analyses revealed significant association with actin cytoskeletal dynamics, kinases, and exosome secretion. The effect of CRABP1 on exosome secretion was experimentally validated by comparing circulating exosome numbers of CKO and wild type (WT) mice, and secreted exosomes from WT and siCRABP1-P19 cells. Pathway analysis identified kinase signaling and Arp2/3 complex as the major pathways where CRABP1-signalosomes modulate exosome secretion, which was validated in the P19 system. CONCLUSION The combinatorial approach allowed efficient screening for and identification of novel CRABP1-signalosomes. The results uncovered a novel function of CRABP1 in modulating exosome secretion, and suggested that CRABP1 could play roles in modulating intercellular communication and signal propagation.
Collapse
Affiliation(s)
- Jennifer Nhieu
- Department of Pharmacology, University of Minnesota, 6-120 Jackson Hall, 321 Church St. SE, Minneapolis, MN, 55455, USA
| | - Chin-Wen Wei
- Department of Pharmacology, University of Minnesota, 6-120 Jackson Hall, 321 Church St. SE, Minneapolis, MN, 55455, USA
| | - Megan Ludwig
- Department of Pharmacology, University of Minnesota, 6-120 Jackson Hall, 321 Church St. SE, Minneapolis, MN, 55455, USA
| | - Justin M Drake
- Department of Pharmacology, University of Minnesota, 6-120 Jackson Hall, 321 Church St. SE, Minneapolis, MN, 55455, USA
| | - Li-Na Wei
- Department of Pharmacology, University of Minnesota, 6-120 Jackson Hall, 321 Church St. SE, Minneapolis, MN, 55455, USA.
| |
Collapse
|
5
|
Silverman JB, Krystofiak EE, Caplan LR, Lau KS, Tyska MJ. Intestinal tuft cells assemble a cytoskeletal superstructure composed of co-aligned actin bundles and microtubules. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.19.585757. [PMID: 38562898 PMCID: PMC10983963 DOI: 10.1101/2024.03.19.585757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Background & Aims All tissues consist of a distinct set of cell types, which collectively support organ function and homeostasis. Tuft cells are a rare epithelial cell type found in diverse epithelia, where they play important roles in sensing antigens and stimulating downstream immune responses. Exhibiting a unique polarized morphology, tuft cells are defined by an array of giant actin filament bundles that support ∼2 μm of apical membrane protrusion and extend over 7 μm towards the cell's perinuclear region. Despite their established roles in maintaining intestinal epithelial homeostasis, tuft cells remain understudied due to their rarity (e.g. ∼ 1% in the small intestinal epithelium). Details regarding the ultrastructural organization of the tuft cell cytoskeleton, the molecular components involved in building the array of giant actin bundles, and how these cytoskeletal structures support tuft cell biology remain unclear. Methods To begin to answer these questions, we used advanced light and electron microscopy to perform quantitative morphometry of the small intestinal tuft cell cytoskeleton. Results We found that tuft cell core bundles consist of actin filaments that are crosslinked in a parallel "barbed-end out" configuration. These polarized structures are also supported by a unique group of tuft cell enriched actin-binding proteins that are differentially localized along the giant core bundles. Furthermore, we found that tuft cell actin bundles are co-aligned with a highly ordered network of microtubules. Conclusions Tuft cells assemble a cytoskeletal superstructure that is well positioned to serve as a track for subcellular transport along the apical-basolateral axis and in turn, support the dynamic sensing functions that are critical for intestinal epithelial homeostasis. SYNOPSIS This research leveraged advanced light and electron microscopy to perform quantitative morphometry of the intestinal tuft cell cytoskeleton. Three-dimensional reconstructions of segmented image data revealed a co-aligned actin-microtubule superstructure that may play a fundamental role in tuft cell function.
Collapse
|
6
|
Lombardo AT, Mitchell CAR, Zaman R, McDermitt DJ, Bretscher A. ARHGAP18-ezrin functions as an autoregulatory module for RhoA in the assembly of distinct actin-based structures. eLife 2024; 13:e83526. [PMID: 38193818 PMCID: PMC10830128 DOI: 10.7554/elife.83526] [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: 09/17/2022] [Accepted: 01/08/2024] [Indexed: 01/10/2024] Open
Abstract
The location of different actin-based structures is largely regulated by Rho GTPases through specific effectors. We use the apical aspect of epithelial cells as a model system to investigate how RhoA is locally regulated to contribute to two distinct adjacent actin-based structures. Assembly of the non-muscle myosin-2 filaments in the terminal web is dependent on RhoA activity, and assembly of the microvilli also requires active RhoA for phosphorylation and activation of ezrin. We show that the RhoGAP, ARHGAP18, is localized by binding active microvillar ezrin, and this interaction enhances ARHGAP18's RhoGAP activity. We present a model where ezrin-ARHGAP18 acts as a negative autoregulatory module to locally reduce RhoA activity in microvilli. Consistent with this model, loss of ARHGAP18 results in disruption of the distinction between microvilli and the terminal web including aberrant assembly of myosin-2 filaments forming inside microvilli. Thus, ARHGAP18, through its recruitment and activation by ezrin, fine-tunes the local level of RhoA to allow for the appropriate distribution of actin-based structures between the microvilli and terminal web. As RhoGAPs vastly outnumber Rho GTPases, this may represent a general mechanism whereby individual Rho effectors drive specific actin-based structures.
Collapse
Affiliation(s)
- Andrew T Lombardo
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell UniversityIthacaUnited States
| | - Cameron AR Mitchell
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell UniversityIthacaUnited States
| | - Riasat Zaman
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell UniversityIthacaUnited States
| | - David J McDermitt
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell UniversityIthacaUnited States
| | - Anthony Bretscher
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell UniversityIthacaUnited States
| |
Collapse
|
7
|
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: 10] [Impact Index Per Article: 10.0] [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
|
8
|
Baqai U, Kurimchak AM, Trachtenberg IV, Purwin TJ, Haj JI, Han A, Luo K, Pachon NF, Jeon A, Chua V, Davies MA, Gutkind JS, Benovic JL, Duncan JS, Aplin AE. Kinome profiling identifies MARK3 and STK10 as potential therapeutic targets in uveal melanoma. J Biol Chem 2023; 299:105418. [PMID: 37923138 PMCID: PMC10716579 DOI: 10.1016/j.jbc.2023.105418] [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: 05/19/2023] [Revised: 10/05/2023] [Accepted: 10/26/2023] [Indexed: 11/07/2023] Open
Abstract
Most uveal melanoma cases harbor activating mutations in either GNAQ or GNA11. Despite activation of the mitogen-activated protein kinase (MAPK) signaling pathway downstream of Gαq/11, there are no effective targeted kinase therapies for metastatic uveal melanoma. The human genome encodes numerous understudied kinases, also called the "dark kinome". Identifying additional kinases regulated by Gαq/11 may uncover novel therapeutic targets for uveal melanoma. In this study, we treated GNAQ-mutant uveal melanoma cell lines with a Gαq/11 inhibitor, YM-254890, and conducted a kinase signaling proteomic screen using multiplexed-kinase inhibitors followed by mass spectrometry. We observed downregulated expression and/or activity of 22 kinases. A custom siRNA screen targeting these kinases demonstrated that knockdown of microtubule affinity regulating kinase 3 (MARK3) and serine/threonine kinase 10 (STK10) significantly reduced uveal melanoma cell growth and decreased expression of cell cycle proteins. Additionally, knockdown of MARK3 but not STK10 decreased ERK1/2 phosphorylation. Analysis of RNA-sequencing and proteomic data showed that Gαq signaling regulates STK10 expression and MARK3 activity. Our findings suggest an involvement of STK10 and MARK3 in the Gαq/11 oncogenic pathway and prompt further investigation into the specific roles and targeting potential of these kinases in uveal melanoma.
Collapse
Affiliation(s)
- Usman Baqai
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Alison M Kurimchak
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA
| | - Isabella V Trachtenberg
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Timothy J Purwin
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Jelan I Haj
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Anna Han
- Department of Food Science and Human Nutrition, Jeonbuk National University, Jeonju, Jeollabuk-do, Republic of Korea
| | - Kristine Luo
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Nikole Fandino Pachon
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Angela Jeon
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Vivian Chua
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Michael A Davies
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, The University of Texas, Houston, Texas, USA
| | - J Silvio Gutkind
- Moores Cancer Center, University of California San Diego, La Jolla, California, USA
| | - Jeffrey L Benovic
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA; Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - James S Duncan
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA
| | - Andrew E Aplin
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA; Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
9
|
Westphal D, Meinhardt M, Grützmann K, Schöne L, Steininger J, Neuhaus LT, Wiegel M, Schrimpf D, Aust DE, Schröck E, Baretton GB, Beissert S, Juratli TA, Schackert GG, Gravemeyer J, Becker JC, von Deimling A, Koelsche C, Klink B, Meier F, Schulz A, Muders MH, Seifert M. Identification of Epigenetically Regulated Genes Distinguishing Intracranial from Extracranial Melanoma Metastases. J Invest Dermatol 2023; 143:1233-1245.e17. [PMID: 36716920 DOI: 10.1016/j.jid.2023.01.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 12/13/2022] [Accepted: 01/09/2023] [Indexed: 01/29/2023]
Abstract
Despite remarkable advances in treating patients with metastatic melanoma, the management of melanoma brain metastases remains challenging. Recent evidence suggests that epigenetic reprogramming is an important mechanism for the adaptation of melanoma cells to the brain environment. In this study, the methylomes and transcriptomes of a cohort of matched melanoma metastases were evaluated by integrated omics data analysis. The identified 38 candidate genes displayed distinct promoter methylation and corresponding gene expression changes in intracranial compared with extracranial metastases. The 11 most promising genes were validated on protein level in both tumor and surrounding normal tissue using immunohistochemistry. In accordance with the underlying promoter methylation and gene expression changes, a significantly different protein expression was confirmed for STK10, PDXK, WDR24, CSSP1, NMB, RASL11B, phosphorylated PRKCZ, PRKCZ, and phosphorylated GRB10 in the intracranial metastases. The observed changes imply a distinct intracranial phenotype with increased protein kinase B phosphorylation and a higher frequency of proliferating cells. Knockdown of PRKCZ or GRB10 altered the expression of phosphorylated protein kinase B and decreased the viability of a brain-specific melanoma cell line. In summary, epigenetically regulated cancer-relevant alterations were identified that provide insights into the molecular mechanisms that discriminate brain metastases from other organ metastases, which could be exploited by targeting the affected signaling pathways.
Collapse
Affiliation(s)
- Dana Westphal
- Department of Dermatology, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany; National Center for Tumor Diseases (NCT/UCC), Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Medizinische Fakultät and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany.
| | - Matthias Meinhardt
- Institute of Pathology, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Konrad Grützmann
- National Center for Tumor Diseases (NCT/UCC), Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Medizinische Fakultät and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany; Core Unit for Molecular Tumor Diagnostics (CMTD), National Center for Tumor Diseases (NCT/UCC), Dresden, Germany; Institute for Medical Informatics and Biometry, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Lisa Schöne
- Department of Dermatology, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany; National Center for Tumor Diseases (NCT/UCC), Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Medizinische Fakultät and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany; Institute for Medical Informatics and Biometry, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Julian Steininger
- Department of Dermatology, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Lena T Neuhaus
- Institute of Pathology, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Miriam Wiegel
- Department of Dermatology, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Daniel Schrimpf
- Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany; Clinical Cooperation Unit Neuropathology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Daniela E Aust
- National Center for Tumor Diseases (NCT/UCC), Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Medizinische Fakultät and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany; Institute of Pathology, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany; Core Unit for Molecular Tumor Diagnostics (CMTD), National Center for Tumor Diseases (NCT/UCC), Dresden, Germany; German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany; BioBank Dresden (BBD), Tumor and Normal Tissue Bank (TNTB), National Center for Tumor Diseases (NCT/UCC), University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Evelin Schröck
- National Center for Tumor Diseases (NCT/UCC), Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Medizinische Fakultät and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany; Core Unit for Molecular Tumor Diagnostics (CMTD), National Center for Tumor Diseases (NCT/UCC), Dresden, Germany; German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany; Institute for Clinical Genetics, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Gustavo B Baretton
- National Center for Tumor Diseases (NCT/UCC), Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Medizinische Fakultät and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany; Institute of Pathology, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany; Core Unit for Molecular Tumor Diagnostics (CMTD), National Center for Tumor Diseases (NCT/UCC), Dresden, Germany; German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany; BioBank Dresden (BBD), Tumor and Normal Tissue Bank (TNTB), National Center for Tumor Diseases (NCT/UCC), University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Stefan Beissert
- Department of Dermatology, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany; National Center for Tumor Diseases (NCT/UCC), Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Medizinische Fakultät and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
| | - Tareq A Juratli
- Department of Neurosurgery, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Gabriele G Schackert
- Department of Neurosurgery, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Jan Gravemeyer
- Translational Skin Cancer Research, German Cancer Consortium (DKTK), Partner Site Essen, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jürgen C Becker
- Translational Skin Cancer Research, German Cancer Consortium (DKTK), Partner Site Essen, and German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Dermatology, University Hospital Essen, Essen, Germany
| | - Andreas von Deimling
- Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany; Clinical Cooperation Unit Neuropathology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christian Koelsche
- Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany; Clinical Cooperation Unit Neuropathology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of General Pathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Barbara Klink
- Core Unit for Molecular Tumor Diagnostics (CMTD), National Center for Tumor Diseases (NCT/UCC), Dresden, Germany; Institute for Clinical Genetics, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Friedegund Meier
- Department of Dermatology, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany; National Center for Tumor Diseases (NCT/UCC), Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Medizinische Fakultät and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany; Skin Cancer Center at the University Cancer Center and National Center for Tumor Diseases, Dresden, Germany
| | - Alexander Schulz
- Department of Dermatology, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany; National Center for Tumor Diseases (NCT/UCC), Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Medizinische Fakultät and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
| | - Michael H Muders
- Institute of Pathology, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Michael Seifert
- National Center for Tumor Diseases (NCT/UCC), Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Medizinische Fakultät and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany; Institute for Medical Informatics and Biometry, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
10
|
A biophysical perspective of the regulatory mechanisms of ezrin/radixin/moesin proteins. Biophys Rev 2022; 14:199-208. [PMID: 35340609 PMCID: PMC8921360 DOI: 10.1007/s12551-021-00928-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 12/14/2021] [Indexed: 02/08/2023] Open
Abstract
Many signal transductions resulting from ligand-receptor interactions occur at the cell surface. These signaling pathways play essential roles in cell polarization, membrane morphogenesis, and the modulation of membrane tension at the cell surface. However, due to the large number of membrane-binding proteins, including actin-membrane linkers, and transmembrane proteins present at the cell surface, the molecular mechanisms underlying the regulation at the cell surface are yet unclear. Here, we describe the molecular functions of one of the key players at the cell surface, ezrin/radixin/moesin (ERM) proteins from a biophysical point of view. We focus our discussion on biophysical properties of ERM proteins revealed by using biophysical tools in live cells and in vitro reconstitution systems. We first describe the structural properties of ERM proteins and then discuss the interactions of ERM proteins with PI(4,5)P2 and the actin cytoskeleton. These properties of ERM proteins revealed by using biophysical approaches have led to a better understanding of their physiological functions in cells and tissues. Supplementary Information The online version contains supplementary material available at 10.1007/s12551-021-00928-0.
Collapse
|
11
|
Avery L, Robertson TF, Wu CF, Roy NH, Chauvin SD, Perkey E, Vanderbeck A, Maillard I, Burkhardt JK. A Murine Model of X-Linked Moesin-Associated Immunodeficiency (X-MAID) Reveals Defects in T Cell Homeostasis and Migration. Front Immunol 2022; 12:726406. [PMID: 35069520 PMCID: PMC8770857 DOI: 10.3389/fimmu.2021.726406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 12/13/2021] [Indexed: 11/25/2022] Open
Abstract
X-linked moesin associated immunodeficiency (X-MAID) is a primary immunodeficiency disease in which patients suffer from profound lymphopenia leading to recurrent infections. The disease is caused by a single point mutation leading to a R171W amino acid change in the protein moesin (moesinR171W). Moesin is a member of the ERM family of proteins, which reversibly link the cortical actin cytoskeleton to the plasma membrane. Here, we describe a novel mouse model with global expression of moesinR171W that recapitulates multiple facets of patient disease, including severe lymphopenia. Further analysis reveals that these mice have diminished numbers of thymocytes and bone marrow precursors. X-MAID mice also exhibit systemic inflammation that is ameliorated by elimination of mature lymphocytes through breeding to a Rag1-deficient background. The few T cells in the periphery of X-MAID mice are highly activated and have mostly lost moesinR171W expression. In contrast, single-positive (SP) thymocytes do not appear activated and retain high expression levels of moesinR171W. Analysis of ex vivo CD4 SP thymocytes reveals defects in chemotactic responses and reduced migration on integrin ligands. While chemokine signaling appears intact, CD4 SP thymocytes from X-MAID mice are unable to polarize and rearrange cytoskeletal elements. This mouse model will be a valuable tool for teasing apart the complexity of the immunodeficiency caused by moesinR171W, and will provide new insights into how the actin cortex regulates lymphocyte function.
Collapse
Affiliation(s)
- Lyndsay Avery
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia Research Institute, Philadelphia, PA, United States
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Tanner F. Robertson
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia Research Institute, Philadelphia, PA, United States
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Christine F. Wu
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia Research Institute, Philadelphia, PA, United States
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Nathan H. Roy
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia Research Institute, Philadelphia, PA, United States
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Samuel D. Chauvin
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia Research Institute, Philadelphia, PA, United States
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Eric Perkey
- Graduate Program in Cellular and Molecular Biology and Medical Scientist Training Program, University of Michigan, Ann Arbor, MI, United States
| | - Ashley Vanderbeck
- Division of Hematology/Oncology, Department of Medicine and Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Ivan Maillard
- Division of Hematology/Oncology, Department of Medicine and Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Janis K. Burkhardt
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia Research Institute, Philadelphia, PA, United States
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
12
|
Serafim RAM, Sorrell FJ, Berger BT, Collins RJ, Vasconcelos SNS, Massirer KB, Knapp S, Bennett J, Fedorov O, Patel H, Zuercher WJ, Elkins JM. Discovery of a Potent Dual SLK/STK10 Inhibitor Based on a Maleimide Scaffold. J Med Chem 2021; 64:13259-13278. [PMID: 34463505 DOI: 10.1021/acs.jmedchem.0c01579] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
SLK (STE20-like kinase) and STK10 (serine/threonine kinase 10) are closely related kinases whose enzymatic activity is linked to the regulation of ezrin, radixin, and moesin function and to the regulation of lymphocyte migration and the cell cycle. We identified a series of 3-anilino-4-arylmaleimides as dual inhibitors of SLK and STK10 with good kinome-wide selectivity. Optimization of this series led to multiple SLK/STK10 inhibitors with nanomolar potency. Crystal structures of exemplar inhibitors bound to SLK and STK10 demonstrated the binding mode of the inhibitors and rationalized their selectivity. Cellular target engagement assays demonstrated the binding of the inhibitors to SLK and STK10 in cells. Further selectivity analyses, including analysis of activity of the reported inhibitors against off-targets in cells, identified compound 31 as the most potent and selective inhibitor of SLK and STK10 yet reported.
Collapse
Affiliation(s)
- Ricardo A M Serafim
- Center for Medicinal Chemistry (CQMED), Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas (UNICAMP), Campinas, SP 13083-875, Brazil.,Structural Genomics Consortium, University of Campinas (UNICAMP), Av. Dr. André Tosello, 550, Barão Geraldo, Campinas, SP 13083-886, Brazil
| | - Fiona J Sorrell
- Centre for Medicines Discovery, University of Oxford, Old Road Campus Research Building, Old Road Campus, Roosevelt Drive, Oxford OX3 7DQ, U.K
| | - Benedict-Tilman Berger
- Structural Genomics Consortium, Johann Wolfgang Goethe University, Buchmann Institute for Molecular Life Sciences, Max-von-Laue-Str. 15, D-60438 Frankfurt am Main, DE, Germany.,Institute for Pharmaceutical Chemistry, Johann Wolfgang Goethe University, Max-von-Laue-Str. 9, D-60438 Frankfurt am Main, DE, Germany
| | - Ross J Collins
- Medicines Discovery Institute, School of Biosciences, Cardiff University, Cardiff CF10 3AT, U.K
| | - Stanley N S Vasconcelos
- Center for Medicinal Chemistry (CQMED), Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas (UNICAMP), Campinas, SP 13083-875, Brazil.,Structural Genomics Consortium, University of Campinas (UNICAMP), Av. Dr. André Tosello, 550, Barão Geraldo, Campinas, SP 13083-886, Brazil
| | - Katlin B Massirer
- Center for Medicinal Chemistry (CQMED), Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas (UNICAMP), Campinas, SP 13083-875, Brazil.,Structural Genomics Consortium, University of Campinas (UNICAMP), Av. Dr. André Tosello, 550, Barão Geraldo, Campinas, SP 13083-886, Brazil
| | - Stefan Knapp
- Structural Genomics Consortium, Johann Wolfgang Goethe University, Buchmann Institute for Molecular Life Sciences, Max-von-Laue-Str. 15, D-60438 Frankfurt am Main, DE, Germany.,Institute for Pharmaceutical Chemistry, Johann Wolfgang Goethe University, Max-von-Laue-Str. 9, D-60438 Frankfurt am Main, DE, Germany
| | - James Bennett
- Centre for Medicines Discovery, University of Oxford, Old Road Campus Research Building, Old Road Campus, Roosevelt Drive, Oxford OX3 7DQ, U.K
| | - Oleg Fedorov
- Centre for Medicines Discovery, University of Oxford, Old Road Campus Research Building, Old Road Campus, Roosevelt Drive, Oxford OX3 7DQ, U.K
| | - Hitesh Patel
- Medicines Discovery Institute, School of Biosciences, Cardiff University, Cardiff CF10 3AT, U.K
| | - William J Zuercher
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Jonathan M Elkins
- Structural Genomics Consortium, University of Campinas (UNICAMP), Av. Dr. André Tosello, 550, Barão Geraldo, Campinas, SP 13083-886, Brazil.,Centre for Medicines Discovery, University of Oxford, Old Road Campus Research Building, Old Road Campus, Roosevelt Drive, Oxford OX3 7DQ, U.K
| |
Collapse
|
13
|
Zhang L, Lu SY, Guo R, Ma JX, Tang LY, Wang JJ, Shen CL, Lu LM, Liu J, Wang ZG, Zhang HX. STK10 knockout inhibits cell migration and promotes cell proliferation via modulating the activity of ERM and p38 MAPK in prostate cancer cells. Exp Ther Med 2021; 22:851. [PMID: 34149897 PMCID: PMC8210223 DOI: 10.3892/etm.2021.10283] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 05/25/2021] [Indexed: 12/24/2022] Open
Abstract
Prostate cancer (PCa) is one of the most common types of cancer and is a serious threat to men's health due to the high rate of incidence and metastasis. However, the exact underlying pathology of this malignant disease has yet to be fully elucidated. The ezrin-radixin-moesin (ERM) family of proteins are associated with the development and metastasis of various types of cancer. Serine threonine kinase 10 (STK10) is an ERM kinase that is involved in the activation of ERM proteins and serves essential roles in the aggregation and adhesion of lymphocytes. To evaluate the functional roles of STK10 in the pathogenesis of PCa, a STK10-knockout (KO) DU145 PCa cell line was generated using the CRISPR-Cas9 gene editing system, and the effects of STK10 deletion on tumor biological behaviors were further analyzed. The present data suggested that STK10 KO promoted PCa cell proliferation by inhibiting p38 MAPK activation and suppressed migration primarily via the inhibition of p38 MAPK signaling and ERM protein activation. To the best of our knowledge, this is the first study to provide evidence that STK10 plays important roles in the proliferation and migration of PCa cells, which will be useful for further investigation into the pathogenesis of this disease.
Collapse
Affiliation(s)
- Lu Zhang
- Research Center for Experimental Medicine, State Key Laboratory of Medical Genomics, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Shun-Yuan Lu
- Research Center for Experimental Medicine, State Key Laboratory of Medical Genomics, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Rui Guo
- Research Center for Experimental Medicine, State Key Laboratory of Medical Genomics, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Jin-Xia Ma
- Research Center for Experimental Medicine, State Key Laboratory of Medical Genomics, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Ling-Yun Tang
- Research Center for Experimental Medicine, State Key Laboratory of Medical Genomics, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Jin-Jin Wang
- Shanghai Model Organisms Center, Shanghai 201321, P.R. China
| | - Chun-Ling Shen
- Research Center for Experimental Medicine, State Key Laboratory of Medical Genomics, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Li-Ming Lu
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Jie Liu
- Shanghai Institute of Orthopaedics and Traumatology, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Zhu-Gang Wang
- Research Center for Experimental Medicine, State Key Laboratory of Medical Genomics, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Hong-Xin Zhang
- Research Center for Experimental Medicine, State Key Laboratory of Medical Genomics, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| |
Collapse
|
14
|
Abstract
Ras homology (RHO) GTPases are signalling proteins that have crucial roles in triggering multiple immune functions. Through their interactions with a broad range of effectors and kinases, they regulate cytoskeletal dynamics, cell polarity and the trafficking and proliferation of immune cells. The activity and localization of RHO GTPases are highly controlled by classical families of regulators that share consensus motifs. In this Review, we describe the recent discovery of atypical modulators and partners of RHO GTPases, which bring an additional layer of regulation and plasticity to the control of RHO GTPase activities in the immune system. Furthermore, the development of large-scale genetic screening has now enabled researchers to identify dysregulation of RHO GTPase signalling pathways as a cause of many immune system-related diseases. We discuss the mutations that have been identified in RHO GTPases and their signalling circuits in patients with rare diseases. The discoveries of new RHO GTPase partners and genetic mutations in RHO GTPase signalling hubs have uncovered unsuspected layers of crosstalk with other signalling pathways and may provide novel therapeutic opportunities for patients affected by complex immune or broader syndromes.
Collapse
|
15
|
Bhanja A, Rey-Suarez I, Song W, Upadhyaya A. Bidirectional feedback between BCR signaling and actin cytoskeletal dynamics. FEBS J 2021; 289:4430-4446. [PMID: 34124846 PMCID: PMC8669062 DOI: 10.1111/febs.16074] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 05/24/2021] [Accepted: 06/14/2021] [Indexed: 11/30/2022]
Abstract
When B cells are exposed to antigens, they use their B-cell receptors (BCRs) to transduce this external signal into internal signaling cascades and uptake antigen, which activate transcriptional programs. Signaling activation requires complex cytoskeletal remodeling initiated by BCR signaling. The actin cytoskeletal remodeling drives B-cell morphological changes, such as spreading, protrusion, contraction, and endocytosis of antigen by mechanical forces, which in turn affect BCR signaling. Therefore, the relationship between the actin cytoskeleton and BCR signaling is a two-way feedback loop. These morphological changes represent the indirect ways by which the actin cytoskeleton regulates BCR signaling. Recent studies using high spatiotemporal resolution microscopy techniques have revealed that actin also can directly influence BCR signaling. Cortical actin networks directly affect BCR mobility, not only during the resting stage by serving as diffusion barriers, but also at the activation stage by altering BCR diffusivity through enhanced actin flow velocities. Furthermore, the actin cytoskeleton, along with myosin, enables B cells to sense the physical properties of its environment and generate and transmit forces through the BCR. Consequently, the actin cytoskeleton modulates the signaling threshold of BCR to antigenic stimulation. This review discusses the latest research on the relationship between BCR signaling and actin remodeling, and the research techniques. Exploration of the role of actin in BCR signaling will expand fundamental understanding of the relationship between cell signaling and the cytoskeleton and the mechanisms underlying cytoskeleton-related immune disorders and cancer.
Collapse
Affiliation(s)
- Anshuman Bhanja
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, USA
| | - Ivan Rey-Suarez
- Institute for Physical Science and Technology, University of Maryland, College Park, MD, USA
| | - Wenxia Song
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, USA
| | - Arpita Upadhyaya
- Institute for Physical Science and Technology, University of Maryland, College Park, MD, USA.,Department Physics, University of Maryland, College Park, MD, USA
| |
Collapse
|
16
|
Garland B, Delisle S, Al-Zahrani KN, Pryce BR, Sabourin LA. The Ste20-like kinase - a Jack of all trades? J Cell Sci 2021; 134:261804. [PMID: 33961052 DOI: 10.1242/jcs.258269] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Over the past 20 years, the Ste20-like kinase (SLK; also known as STK2) has emerged as a central regulator of cytoskeletal dynamics. Reorganization of the cytoskeleton is necessary for a plethora of biological processes including apoptosis, proliferation, migration, tissue repair and signaling. Several studies have also uncovered a role for SLK in disease progression and cancer. Here, we review the recent findings in the SLK field and summarize the various roles of SLK in different animal models and discuss the biochemical mechanisms regulating SLK activity. Together, these studies have revealed multiple roles for SLK in coupling cytoskeletal dynamics to cell growth, in muscle repair and in negative-feedback loops critical for cancer progression. Furthermore, the ability of SLK to regulate some systems appears to be kinase activity independent, suggesting that it may be an important scaffold for signal transduction pathways. These various findings reveal highly complex functions and regulation patterns of SLK in development and disease, making it a potential therapeutic target.
Collapse
Affiliation(s)
- Brennan Garland
- Ottawa Hospital Research Institute, Cancer Therapeutics, Ottawa, Ontario, K1H8L1, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, K1H8L6, Canada
| | - Samuel Delisle
- Ottawa Hospital Research Institute, Cancer Therapeutics, Ottawa, Ontario, K1H8L1, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, K1H8L6, Canada
| | - Khalid N Al-Zahrani
- Center for Molecular and Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, M5G1X5, Canada
| | - Benjamin R Pryce
- Department of Pediatrics, Hollings Cancer Center, Medical University of South Carolina,Charleston, SC 29425, USA
| | - Luc A Sabourin
- Ottawa Hospital Research Institute, Cancer Therapeutics, Ottawa, Ontario, K1H8L1, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, K1H8L6, Canada
| |
Collapse
|
17
|
Zaman R, Lombardo A, Sauvanet C, Viswanatha R, Awad V, Bonomo LER, McDermitt D, Bretscher A. Effector-mediated ERM activation locally inhibits RhoA activity to shape the apical cell domain. J Cell Biol 2021; 220:211973. [PMID: 33836044 PMCID: PMC8185690 DOI: 10.1083/jcb.202007146] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 02/07/2021] [Accepted: 03/09/2021] [Indexed: 12/21/2022] Open
Abstract
Activated ezrin-radixin-moesin (ERM) proteins link the plasma membrane to the actin cytoskeleton to generate apical structures, including microvilli. Among many kinases implicated in ERM activation are the homologues LOK and SLK. CRISPR/Cas9 was used to knock out all ERM proteins or LOK/SLK in human cells. LOK/SLK knockout eliminates all ERM-activating phosphorylation. The apical domains of cells lacking LOK/SLK or ERMs are strikingly similar and selectively altered, with loss of microvilli and with junctional actin replaced by ectopic myosin-II–containing apical contractile structures. Constitutively active ezrin can reverse the phenotypes of either ERM or LOK/SLK knockouts, indicating that a central function of LOK/SLK is to activate ERMs. Both knockout lines have elevated active RhoA with concomitant enhanced myosin light chain phosphorylation, revealing that active ERMs are negative regulators of RhoA. As RhoA-GTP activates LOK/SLK to activate ERM proteins, the ability of active ERMs to negatively regulate RhoA-GTP represents a novel local feedback loop necessary for the proper apical morphology of epithelial cells.
Collapse
Affiliation(s)
- Riasat Zaman
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY
| | - Andrew Lombardo
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY
| | - Cécile Sauvanet
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY
| | - Raghuvir Viswanatha
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY
| | - Valerie Awad
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY
| | - Locke Ezra-Ros Bonomo
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY
| | - David McDermitt
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY
| | - Anthony Bretscher
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY
| |
Collapse
|
18
|
ERM Proteins at the Crossroad of Leukocyte Polarization, Migration and Intercellular Adhesion. Int J Mol Sci 2020; 21:ijms21041502. [PMID: 32098334 PMCID: PMC7073024 DOI: 10.3390/ijms21041502] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 02/18/2020] [Accepted: 02/19/2020] [Indexed: 12/12/2022] Open
Abstract
Ezrin, radixin and moesin proteins (ERMs) are plasma membrane (PM) organizers that link the actin cytoskeleton to the cytoplasmic tail of transmembrane proteins, many of which are adhesion receptors, in order to regulate the formation of F-actin-based structures (e.g., microspikes and microvilli). ERMs also effect transmission of signals from the PM into the cell, an action mainly exerted through the compartmentalized activation of the small Rho GTPases Rho, Rac and Cdc42. Ezrin and moesin are the ERMs more highly expressed in leukocytes, and although they do not always share functions, both are mainly regulated through phosphatidylinositol 4,5-bisphosphate (PIP2) binding to the N-terminal band 4.1 protein-ERM (FERM) domain and phosphorylation of a conserved Thr in the C-terminal ERM association domain (C-ERMAD), exerting their functions through a wide assortment of mechanisms. In this review we will discuss some of these mechanisms, focusing on how they regulate polarization and migration in leukocytes, and formation of actin-based cellular structures like the phagocytic cup-endosome and the immune synapse in macrophages/neutrophils and lymphocytes, respectively, which represent essential aspects of the effector immune response.
Collapse
|
19
|
de Toledo I, Grigolo TA, Bennett JM, Elkins JM, Pilli RA. Modular Synthesis of Di- and Trisubstituted Imidazoles from Ketones and Aldehydes: A Route to Kinase Inhibitors. J Org Chem 2019; 84:14187-14201. [PMID: 31460764 PMCID: PMC6829625 DOI: 10.1021/acs.joc.9b01844] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
![]()
A one-pot and modular approach to
the synthesis of 2,4(5)-disubstituted
imidazoles was developed based on ketone oxidation, employing catalytic
HBr and DMSO, followed by imidazole condensation with aldehydes. This
methodology afforded twenty-nine disubstituted NH-imidazoles (23%–85% yield). A three-step synthesis of 20
kinase inhibitors was achieved by employing this oxidation–condensation
protocol, followed by bromination and Suzuki coupling in the imidazole
ring to yield trisubstituted NH-imidazoles (23%–69%,
three steps). This approach was also employed in the synthesis of
known inhibitor GSK3037619A.
Collapse
Affiliation(s)
- Ian de Toledo
- Department of Organic Chemistry, Institute of Chemistry , University of Campinas, UNICAMP , Campinas , São Paulo 13083-970 , Brazil
| | - Thiago A Grigolo
- Department of Organic Chemistry, Institute of Chemistry , University of Campinas, UNICAMP , Campinas , São Paulo 13083-970 , Brazil
| | - James M Bennett
- Structural Genomics Consortium, Nuffield Department of Medicine , University of Oxford , Old Road Campus Research Building, Roosevelt Drive , Oxford OX3 7DQ , United Kingdom
| | - Jonathan M Elkins
- Structural Genomics Consortium, Nuffield Department of Medicine , University of Oxford , Old Road Campus Research Building, Roosevelt Drive , Oxford OX3 7DQ , United Kingdom.,Structural Genomics Consortium, Departamento de Genética e Evolução , Instituto de Biologia, UNICAMP , Campinas , São Paulo 13083-886 , Brazil
| | - Ronaldo A Pilli
- Department of Organic Chemistry, Institute of Chemistry , University of Campinas, UNICAMP , Campinas , São Paulo 13083-970 , Brazil
| |
Collapse
|
20
|
Derouiche A, Geiger KD. Perspectives for Ezrin and Radixin in Astrocytes: Kinases, Functions and Pathology. Int J Mol Sci 2019; 20:ijms20153776. [PMID: 31382374 PMCID: PMC6695708 DOI: 10.3390/ijms20153776] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 07/30/2019] [Accepted: 07/31/2019] [Indexed: 02/06/2023] Open
Abstract
Astrocytes are increasingly perceived as active partners in physiological brain function and behaviour. The structural correlations of the glia–synaptic interaction are the peripheral astrocyte processes (PAPs), where ezrin and radixin, the two astrocytic members of the ezrin-radixin-moesin (ERM) family of proteins are preferentially localised. While the molecular mechanisms of ERM (in)activation appear universal, at least in mammalian cells, and have been studied in great detail, the actual ezrin and radixin kinases, phosphatases and binding partners appear cell type specific and may be multiplexed within a cell. In astrocytes, ezrin is involved in process motility, which can be stimulated by the neurotransmitter glutamate, through activation of the glial metabotropic glutamate receptors (mGluRs) 3 or 5. However, it has remained open how this mGluR stimulus is transduced to ezrin activation. Knowing upstream signals of ezrin activation, ezrin kinase(s), and membrane-bound binding partners of ezrin in astrocytes might open new approaches to the glial role in brain function. Ezrin has also been implicated in invasive behaviour of astrocytomas, and glial activation. Here, we review data pertaining to potential molecular interaction partners of ezrin in astrocytes, with a focus on PKC and GRK2, and in gliomas and other diseases, to stimulate further research on their potential roles in glia-synaptic physiology and pathology.
Collapse
Affiliation(s)
- Amin Derouiche
- Institute of Anatomy II, Goethe-University Frankfurt, D-60590 Frankfurt am Main, Germany.
| | - Kathrin D Geiger
- Neuropathology, Institute for Pathology, Carl Gustav Carus University Hospital, TU Dresden, D-01307 Dresden, Germany
| |
Collapse
|
21
|
Ivetic A, Hoskins Green HL, Hart SJ. L-selectin: A Major Regulator of Leukocyte Adhesion, Migration and Signaling. Front Immunol 2019; 10:1068. [PMID: 31139190 PMCID: PMC6527602 DOI: 10.3389/fimmu.2019.01068] [Citation(s) in RCA: 270] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 04/26/2019] [Indexed: 12/12/2022] Open
Abstract
L-selectin (CD62L) is a type-I transmembrane glycoprotein and cell adhesion molecule that is expressed on most circulating leukocytes. Since its identification in 1983, L-selectin has been extensively characterized as a tethering/rolling receptor. There is now mounting evidence in the literature to suggest that L-selectin plays a role in regulating monocyte protrusion during transendothelial migration (TEM). The N-terminal calcium-dependent (C-type) lectin domain of L-selectin interacts with numerous glycans, including sialyl Lewis X (sLex) for tethering/rolling and proteoglycans for TEM. Although the signals downstream of L-selectin-dependent adhesion are poorly understood, they will invariably involve the short 17 amino acid cytoplasmic tail. In this review we will detail the expression of L-selectin in different immune cell subsets, and its influence on cell behavior. We will list some of the diverse glycans known to support L-selectin-dependent adhesion, within luminal and abluminal regions of the vessel wall. We will describe how each domain within L-selectin contributes to adhesion, migration and signal transduction. A significant focus on the L-selectin cytoplasmic tail and its proposed contribution to signaling via the ezrin-radixin-moesin (ERM) family of proteins will be outlined. Finally, we will discuss how ectodomain shedding of L-selectin during monocyte TEM is essential for the establishment of front-back cell polarity, bestowing emigrated cells the capacity to chemotax toward sites of damage.
Collapse
Affiliation(s)
- Aleksandar Ivetic
- King's College London, School of Cardiovascular Medicine and Sciences, BHF Center of Research Excellence, London, United Kingdom
| | - Hannah Louise Hoskins Green
- King's College London, School of Cardiovascular Medicine and Sciences, BHF Center of Research Excellence, London, United Kingdom
| | - Samuel James Hart
- King's College London, School of Cardiovascular Medicine and Sciences, BHF Center of Research Excellence, London, United Kingdom
| |
Collapse
|
22
|
Gold MR, Reth MG. Antigen Receptor Function in the Context of the Nanoscale Organization of the B Cell Membrane. Annu Rev Immunol 2019; 37:97-123. [DOI: 10.1146/annurev-immunol-042718-041704] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The B cell antigen receptor (BCR) plays a central role in the self/nonself selection of B lymphocytes and in their activation by cognate antigen during the clonal selection process. It was long thought that most cell surface receptors, including the BCR, were freely diffusing and randomly distributed. Since the advent of superresolution techniques, it has become clear that the plasma membrane is compartmentalized and highly organized at the nanometer scale. Hence, a complete understanding of the precise conformation and activation mechanism of the BCR must take into account the organization of the B cell plasma membrane. We review here the recent literature on the nanoscale organization of the lymphocyte membrane and discuss how this new information influences our view of the conformational changes that the BCR undergoes during activation.
Collapse
Affiliation(s)
- Michael R. Gold
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
- Life Sciences Institute, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Michael G. Reth
- BIOSS Centre for Biological Signaling Studies, University of Freiburg, 79104 Freiburg, Germany
- Department of Molecular Immunology, Institute of Biology III, Faculty of Biology, University of Freiburg, 79108 Freiburg, Germany
| |
Collapse
|
23
|
PTENα promotes neutrophil chemotaxis through regulation of cell deformability. Blood 2019; 133:2079-2089. [PMID: 30926592 DOI: 10.1182/blood-2019-01-899864] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 03/14/2019] [Indexed: 12/20/2022] Open
Abstract
Neutrophils are a major component of immune defense and are recruited through neutrophil chemotaxis in response to invading pathogens. However, the molecular mechanism that controls neutrophil chemotaxis remains unclear. Here, we report that PTENα, the first isoform identified in the PTEN family, regulates neutrophil deformability and promotes chemotaxis of neutrophils. A high level of PTENα is detected in neutrophils and lymphoreticular tissues. Homozygous deletion of PTENα impairs chemoattractant-induced migration of neutrophils. We show that PTENα physically interacts with cell membrane cross-linker moesin through its FERM domain and dephosphorylates moesin at Thr558, which disrupts the association of filamentous actin with the plasma membrane and subsequently induces morphologic changes in neutrophil pseudopodia. These results demonstrate that PTENα acts as a phosphatase of moesin and modulates neutrophil-mediated host immune defense. We propose that PTENα signaling is a potential target for the treatment of infections and immune diseases.
Collapse
|
24
|
Miller CJ, Lou HJ, Simpson C, van de Kooij B, Ha BH, Fisher OS, Pirman NL, Boggon TJ, Rinehart J, Yaffe MB, Linding R, Turk BE. Comprehensive profiling of the STE20 kinase family defines features essential for selective substrate targeting and signaling output. PLoS Biol 2019; 17:e2006540. [PMID: 30897078 PMCID: PMC6445471 DOI: 10.1371/journal.pbio.2006540] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 04/02/2019] [Accepted: 03/14/2019] [Indexed: 12/14/2022] Open
Abstract
Specificity within protein kinase signaling cascades is determined by direct and indirect interactions between kinases and their substrates. While the impact of localization and recruitment on kinase-substrate targeting can be readily assessed, evaluating the relative importance of direct phosphorylation site interactions remains challenging. In this study, we examine the STE20 family of protein serine-threonine kinases to investigate basic mechanisms of substrate targeting. We used peptide arrays to define the phosphorylation site specificity for the majority of STE20 kinases and categorized them into four distinct groups. Using structure-guided mutagenesis, we identified key specificity-determining residues within the kinase catalytic cleft, including an unappreciated role for the kinase β3-αC loop region in controlling specificity. Exchanging key residues between the STE20 kinases p21-activated kinase 4 (PAK4) and Mammalian sterile 20 kinase 4 (MST4) largely interconverted their phosphorylation site preferences. In cells, a reprogrammed PAK4 mutant, engineered to recognize MST substrates, failed to phosphorylate PAK4 substrates or to mediate remodeling of the actin cytoskeleton. In contrast, this mutant could rescue signaling through the Hippo pathway in cells lacking multiple MST kinases. These observations formally demonstrate the importance of catalytic site specificity for directing protein kinase signal transduction pathways. Our findings further suggest that phosphorylation site specificity is both necessary and sufficient to mediate distinct signaling outputs of STE20 kinases and imply broad applicability to other kinase signaling systems.
Collapse
Affiliation(s)
- Chad J. Miller
- Department of Pharmacology, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Hua Jane Lou
- Department of Pharmacology, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Craig Simpson
- Biotech Research and Innovation Center, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Bert van de Kooij
- Departments of Biological Engineering and Biology, MIT Center for Precision Cancer Medicine and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Byung Hak Ha
- Department of Pharmacology, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Oriana S. Fisher
- Department of Pharmacology, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Natasha L. Pirman
- Department of Cellular and Molecular Physiology and Systems Biology Institute, Yale University, New Haven, Connecticut, United States of America
| | - Titus J. Boggon
- Department of Pharmacology, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Jesse Rinehart
- Department of Cellular and Molecular Physiology and Systems Biology Institute, Yale University, New Haven, Connecticut, United States of America
| | - Michael B. Yaffe
- Departments of Biological Engineering and Biology, MIT Center for Precision Cancer Medicine and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Rune Linding
- Biotech Research and Innovation Center, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Benjamin E. Turk
- Department of Pharmacology, Yale School of Medicine, New Haven, Connecticut, United States of America
| |
Collapse
|
25
|
Li J, Yin W, Jing Y, Kang D, Yang L, Cheng J, Yu Z, Peng Z, Li X, Wen Y, Sun X, Ren B, Liu C. The Coordination Between B Cell Receptor Signaling and the Actin Cytoskeleton During B Cell Activation. Front Immunol 2019; 9:3096. [PMID: 30687315 PMCID: PMC6333714 DOI: 10.3389/fimmu.2018.03096] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Accepted: 12/13/2018] [Indexed: 01/27/2023] Open
Abstract
B-cell activation plays a crucial part in the immune system and is initiated via interaction between the B cell receptor (BCR) and specific antigens. In recent years with the help of modern imaging techniques, it was found that the cortical actin cytoskeleton changes dramatically during B-cell activation. In this review, we discuss how actin-cytoskeleton reorganization regulates BCR signaling in different stages of B-cell activation, specifically when stimulated by antigens, and also how this reorganization is mediated by BCR signaling molecules. Abnormal BCR signaling is associated with the progression of lymphoma and immunological diseases including autoimmune disorders, and recent studies have proved that impaired actin cytoskeleton can devastate the normal activation of B cells. Therefore, to figure out the coordination between the actin cytoskeleton and BCR signaling may reveal an underlying mechanism of B-cell activation, which has potential for new treatments for B-cell associated diseases.
Collapse
Affiliation(s)
- Jingwen Li
- Department of Microbiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Yin
- Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yukai Jing
- Department of Microbiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Danqing Kang
- Department of Microbiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lu Yang
- Department of Microbiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiali Cheng
- Department of Microbiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ze Yu
- Department of Microbiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zican Peng
- Department of Microbiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xingbo Li
- Department of Microbiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yue Wen
- Department of Microbiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xizi Sun
- Department of Microbiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Boxu Ren
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China
- Clinical Molecular Immunology Center, School of Medicine, Yangtze University, Jingzhou, China
| | - Chaohong Liu
- Department of Microbiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
26
|
Sphingosine 1-phosphate-mediated activation of ezrin-radixin-moesin proteins contributes to cytoskeletal remodeling and changes of membrane properties in epithelial otic vesicle progenitors. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1866:554-565. [PMID: 30611767 DOI: 10.1016/j.bbamcr.2018.12.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 11/24/2018] [Accepted: 12/18/2018] [Indexed: 12/20/2022]
Abstract
Hearing loss is among the most prevalent sensory impairments in humans. Cochlear implantable devices represent the current therapies for hearing loss but have various shortcomings. ERM (ezrin- radixin -moesin) are a family of adaptor proteins that link plasma membrane with actin cytoskeleton, playing a crucial role in cell morphology and in the formation of membrane protrusions. Recently, bioactive sphingolipids have emerged as regulators of ERM proteins. Sphingosine 1-phosphate (S1P) is a pleiotropic sphingolipid which regulates fundamental cellular functions such as proliferation, survival, migration as well as processes such as development and inflammation mainly via ligation to its specific receptors S1PR (S1P1-5). Experimental findings demonstrate a key role for S1P signaling axis in the maintenance of auditory function. Preservation of cellular junctions is a fundamental function both for S1P and ERM proteins, crucial for the maintenance of cochlear integrity. In the present work, S1P was found to activate ERM in a S1P2-dependent manner in murine auditory epithelial progenitors US/VOT-E36. S1P-induced ERM activation potently contributed to actin cytoskeletal remodeling and to the appearance of ionic currents and membrane passive properties changes typical of more differentiated cells. Moreover, PKC and Akt activation was found to mediate S1P-induced ERM phosphorylation. The obtained findings contribute to demonstrate the role of S1P signaling pathway in inner ear biology and to disclose potential innovative therapeutical approaches in the field of hearing loss prevention and treatment.
Collapse
|
27
|
Plasmodium falciparum Cyclic GMP-Dependent Protein Kinase Interacts with a Subunit of the Parasite Proteasome. Infect Immun 2018; 87:IAI.00523-18. [PMID: 30323024 DOI: 10.1128/iai.00523-18] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 09/29/2018] [Indexed: 12/20/2022] Open
Abstract
Malaria is caused by the protozoan parasite Plasmodium, which undergoes a complex life cycle in a human host and a mosquito vector. The parasite's cyclic GMP (cGMP)-dependent protein kinase (PKG) is essential at multiple steps of the life cycle. Phosphoproteomic studies in Plasmodium falciparum erythrocytic stages and Plasmodium berghei ookinetes have identified proteolysis as a major biological pathway dependent on PKG activity. To further understand PKG's mechanism of action, we screened a yeast two-hybrid library for P. falciparum proteins that interact with P. falciparum PKG (PfPKG) and tested peptide libraries to identify its phosphorylation site preferences. Our data suggest that PfPKG has a distinct phosphorylation site and that PfPKG directly phosphorylates parasite RPT1, one of six AAA+ ATPases present in the 19S regulatory particle of the proteasome. PfPKG and RPT1 interact in vitro, and the interacting fragment of RPT1 carries a PfPKG consensus phosphorylation site; a peptide carrying this consensus site competes with the RPT1 fragment for binding to PfPKG and is efficiently phosphorylated by PfPKG. These data suggest that PfPKG's phosphorylation of RPT1 could contribute to its regulation of parasite proteolysis. We demonstrate that proteolysis plays an important role in a biological process known to require Plasmodium PKG: invasion by sporozoites of hepatocytes. A small-molecule inhibitor of proteasomal activity blocks sporozoite invasion in an additive manner when combined with a Plasmodium PKG-specific inhibitor. Mining the previously described parasite PKG-dependent phosphoproteomes using the consensus phosphorylation motif identified additional proteins that are likely to be direct substrates of the enzyme.
Collapse
|
28
|
Pelaseyed T, Bretscher A. Regulation of actin-based apical structures on epithelial cells. J Cell Sci 2018; 131:131/20/jcs221853. [PMID: 30333133 DOI: 10.1242/jcs.221853] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Cells of transporting epithelia are characterized by the presence of abundant F-actin-based microvilli on their apical surfaces. Likewise, auditory hair cells have highly reproducible rows of apical stereocilia (giant microvilli) that convert mechanical sound into an electrical signal. Analysis of mutations in deaf patients has highlighted the critical components of tip links between stereocilia, and related structures that contribute to the organization of microvilli on epithelial cells have been found. Ezrin/radixin/moesin (ERM) proteins, which are activated by phosphorylation, provide a critical link between the plasma membrane and underlying actin cytoskeleton in surface structures. Here, we outline recent insights into how microvilli and stereocilia are built, and the roles of tip links. Furthermore, we highlight how ezrin is locally regulated by phosphorylation, and that this is necessary to maintain polarity. Localized phosphorylation is achieved through an intricate coincidence detection mechanism that requires the membrane lipid phosphatidylinositol 4,5-bisphosphate [PI(4,5)P2] and the apically localized ezrin kinase, lymphocyte-oriented kinase (LOK, also known as STK10) or Ste20-like kinase (SLK). We also discuss how ezrin-binding scaffolding proteins regulate microvilli and how, despite these significant advances, it remains to be discovered how the cell polarity program ultimately interfaces with these processes.
Collapse
Affiliation(s)
- Thaher Pelaseyed
- Institute of Biomedicine, Department of Medical Biochemistry and Cell Biology, University of Gothenburg, SE-405 30 Gothenburg, Sweden
| | - Anthony Bretscher
- Weill Institute for Cell and Molecular Biology, Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
29
|
Wang Z, Zhang Y, Pinkas DM, Fox AE, Luo J, Huang H, Cui S, Xiang Q, Xu T, Xun Q, Zhu D, Tu Z, Ren X, Brekken RA, Bullock AN, Liang G, Ding K, Lu X. Design, Synthesis, and Biological Evaluation of 3-(Imidazo[1,2- a]pyrazin-3-ylethynyl)-4-isopropyl- N-(3-((4-methylpiperazin-1-yl)methyl)-5-(trifluoromethyl)phenyl)benzamide as a Dual Inhibitor of Discoidin Domain Receptors 1 and 2. J Med Chem 2018; 61:7977-7990. [PMID: 30075624 PMCID: PMC6287892 DOI: 10.1021/acs.jmedchem.8b01045] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Discoidin-domain receptors 1 and 2 (DDR1 and DDR2) are new potential targets for anti-inflammatory-drug discovery. A series of heterocycloalkynylbenzimides were designed and optimized to coinhibit DDR1 and DDR2. One of the most promising compounds, 5n, tightly bound to DDR1 and DDR2 proteins with Kd values of 7.9 and 8.0 nM; potently inhibited the kinases with IC50 values of 9.4 and 20.4 nM, respectively; and was significantly less potent for a panel of 403 wild-type kinases at 1.0 μM. DDR1- and DDR2-kinase inhibition by 5n was validated by Western-blotting analysis in primary human lung fibroblasts. The compound also dose-dependently inhibited lipopolysaccharide (LPS)-induced interleukin 6 (IL-6) release in vitro and exhibited promising in vivo anti-inflammatory effects in an LPS-induced-acute-lung-injury (ALI) mouse model. Compound 5n may serve as a lead compound for new anti-inflammatory drug discovery.
Collapse
Affiliation(s)
- Zhen Wang
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy , Jinan University , 601 Huangpu Avenue West , Guangzhou 510632 , China.,Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences , 190 Kaiyuan Avenue , Guangzhou 510530 , China
| | | | - Daniel M Pinkas
- Structural Genomics Consortium , University of Oxford , Old Road Campus Research Building, Roosevelt Drive , Oxford OX3 7DQ , U.K
| | - Alice E Fox
- Structural Genomics Consortium , University of Oxford , Old Road Campus Research Building, Roosevelt Drive , Oxford OX3 7DQ , U.K
| | - Jinfeng Luo
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences , 190 Kaiyuan Avenue , Guangzhou 510530 , China
| | - Huocong Huang
- Nancy B. and Jake L. Hamon Centre for Therapeutic Oncology Research, Departments of Surgery and Pharmacology , University of Texas Southwestern Medical Center at Dallas , 5323 Harry Hines Boulevard , Dallas , Texas 75390 , United States
| | - Shengyang Cui
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences , 190 Kaiyuan Avenue , Guangzhou 510530 , China
| | - Qiuping Xiang
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences , 190 Kaiyuan Avenue , Guangzhou 510530 , China
| | | | - Qiuju Xun
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences , 190 Kaiyuan Avenue , Guangzhou 510530 , China
| | - Dongsheng Zhu
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences , 190 Kaiyuan Avenue , Guangzhou 510530 , China
| | - Zhengchao Tu
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences , 190 Kaiyuan Avenue , Guangzhou 510530 , China
| | - Xiaomei Ren
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy , Jinan University , 601 Huangpu Avenue West , Guangzhou 510632 , China
| | - Rolf A Brekken
- Nancy B. and Jake L. Hamon Centre for Therapeutic Oncology Research, Departments of Surgery and Pharmacology , University of Texas Southwestern Medical Center at Dallas , 5323 Harry Hines Boulevard , Dallas , Texas 75390 , United States
| | - Alex N Bullock
- Structural Genomics Consortium , University of Oxford , Old Road Campus Research Building, Roosevelt Drive , Oxford OX3 7DQ , U.K
| | | | - Ke Ding
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy , Jinan University , 601 Huangpu Avenue West , Guangzhou 510632 , China
| | - Xiaoyun Lu
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy , Jinan University , 601 Huangpu Avenue West , Guangzhou 510632 , China
| |
Collapse
|
30
|
LpMab-23-recognizing cancer-type podoplanin is a novel predictor for a poor prognosis of early stage tongue cancer. Oncotarget 2018; 9:21156-21165. [PMID: 29765527 PMCID: PMC5940393 DOI: 10.18632/oncotarget.24986] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 03/12/2018] [Indexed: 11/25/2022] Open
Abstract
Purpose We report that the reactivity of a novel monoclonal antibody LpMab-23 for human cancer-type podoplanin (PDPN) is a predictor for a poor prognosis of tongue cancer. Patients and Methods The association between LpMab-23-recognizing cancer-type PDPN expression and clinical/pathological features were analyzed on 60 patients with stage I and II tongue cancer treated with transoral resection of the primary tumor. Results In the mode of invasion, the LpMab-23-dull/negative cases were significantly larger in cases with low-grade malignancies and without late cervical lymph node metastasis, than in cases with high-grade malignancies and the metastasis. In the high-grade malignant cases, LpMab-23-positive cases were significantly larger than LpMab-23-dull/negative cases. The Kaplan–Meier curves of the five-year metastasis-free survival rate (MFS) were significantly lower in the LpMab-23 positive patients than in LpMab-23 dull/negative patients. The LpMab-23-dull/negative cases showed the highest MFS in all of the clinical/pathological features and particularly, the MFS of the LpMab-23 positive cases decreased to less than 60% in the first year. In the Cox proportional hazard regression models a comparison of the numbers of LpMab-23 dull/negative with positive cases showed the highest hazard ratio with statistical significance in all of the clinical/pathological features. Conclusions LpMab-23 positive cases may be considered to present a useful predictor of poor prognosis for early stage tongue cancer.
Collapse
|
31
|
Takenawa T, Kanai T, Kitamura T, Yoshimura Y, Sawa Y, Iida J. Expression and Dynamics of Podoplanin in Cultured Osteoblasts with Mechanostress and Mineralization Stimulus. Acta Histochem Cytochem 2018; 51:41-52. [PMID: 29622849 PMCID: PMC5880802 DOI: 10.1267/ahc.17031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 12/25/2017] [Indexed: 01/25/2023] Open
Abstract
This study investigates the significance of the expression and dynamics of podoplanin in mechanostress and mineralization in cultured murine osteoblasts. Podoplanin increased in osteoblasts subjected to straining in non-mineralization medium, suggesting that the mechanostress alone is a podoplanin induction factor. In osteoblasts subjected to vertical elongation straining in the mineralization medium, the mRNA amounts of podoplanin, osteopontin, and osteocalcin were significantly larger than those in cells not subjected to straining, suggesting that mechanostress is the cause of a synergistic effect in the expression of these proteins. In osteoblasts in the mineralization medium, significant increases in osteocalcin mRNA occurred earlier in cells subjected to straining than in the cells not subjected to straining, suggesting that the mechanostress is a critical factor to enhance the expression of osteocalcin. Western blot and ELISA analysis showed increased podoplanin production in osteoblasts with longer durations of straining. There was significantly less mineralization product in osteoblasts with antibodies for podoplanin, osteopontin, and osteocalcin. There was also less osteopontin and osteocalcin produced in osteoblasts with anti-podoplanin. These findings suggest that mechanostress induces the production of podoplanin in osteoblasts and that podoplanin may play a role in mineralization in cooperation with bone-associated proteins.
Collapse
Affiliation(s)
- Tomohiro Takenawa
- Department of Orthodontics, Faculty of Dental Medicine and Graduate School of Dental Medicine, Hokkaido University
| | - Takenori Kanai
- Department of Orthodontics, Faculty of Dental Medicine and Graduate School of Dental Medicine, Hokkaido University
| | - Tetsuya Kitamura
- Department of Oral Pathology and Biology, Faculty of Dental Medicine and Graduate School of Dental Medicine, Hokkaido University
| | - Yoshitaka Yoshimura
- Department of Molecular Cell Pharmacology, Faculty of Dental Medicine and Graduate School of Dental Medicine, Hokkaido University
| | - Yoshihiko Sawa
- Deparment of Oral Function & Anatomy, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences
| | - Junichiro Iida
- Department of Orthodontics, Faculty of Dental Medicine and Graduate School of Dental Medicine, Hokkaido University
| |
Collapse
|
32
|
Di Pietro C, Zhang PX, O'Rourke TK, Murray TS, Wang L, Britto CJ, Koff JL, Krause DS, Egan ME, Bruscia EM. Ezrin links CFTR to TLR4 signaling to orchestrate anti-bacterial immune response in macrophages. Sci Rep 2017; 7:10882. [PMID: 28883468 PMCID: PMC5589856 DOI: 10.1038/s41598-017-11012-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 08/17/2017] [Indexed: 12/16/2022] Open
Abstract
Macrophages (MΦs) with mutations in cystic fibrosis transmembrane conductance regulator (CFTR) have blunted induction of PI3K/AKT signaling in response to TLR4 activation, leading to hyperinflammation, a hallmark of cystic fibrosis (CF) disease. Here, we show that Ezrin links CFTR and TLR4 signaling, and is necessary for PI3K/AKT signaling induction in response to MΦ activation. Because PI3K/AKT signaling is critical for immune regulation, Ezrin-deficient MΦs are hyperinflammatory and have impaired Pseudomonas aeruginosa phagocytosis, phenocopying CF MΦs. Importantly, we show that activated CF MΦs have reduced protein levels and altered localization of the remaining Ezrin to filopodia that form during activation. In summary, we have described a direct link from CFTR to Ezrin to PI3K/AKT signaling that is disrupted in CF, and thus promotes hyper-inflammation and weakens phagocytosis.
Collapse
Affiliation(s)
- Caterina Di Pietro
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
| | - Ping-Xia Zhang
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
| | - Timothy K O'Rourke
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
- Quinnipiac University School of Medicine, Hamden, CT, USA
| | - Thomas S Murray
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, USA
- Quinnipiac University School of Medicine, Hamden, CT, USA
| | - Lin Wang
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Clemente J Britto
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Jonathan L Koff
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Diane S Krause
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, USA
- Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT, USA
| | - Marie E Egan
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
| | - Emanuela M Bruscia
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
33
|
Oh YS, Heo K, Kim EK, Jang JH, Bae SS, Park JB, Kim YH, Song M, Kim SR, Ryu SH, Kim IH, Suh PG. Dynamic relocalization of NHERF1 mediates chemotactic migration of ovarian cancer cells toward lysophosphatidic acid stimulation. Exp Mol Med 2017; 49:e351. [PMID: 28684865 PMCID: PMC5565956 DOI: 10.1038/emm.2017.88] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 12/28/2016] [Accepted: 01/09/2017] [Indexed: 01/05/2023] Open
Abstract
NHERF1/EBP50 (Na+/H+ exchanger regulating
factor 1; Ezrin-binding phosphoprotein of 50 kDa) organizes stable
protein complexes beneath the apical membrane of polar epithelial cells. By
contrast, in cancer cells without any fixed polarity, NHERF1 often localizes in
the cytoplasm. The regulation of cytoplasmic NHERF1 and its role in cancer
progression remain unclear. In this study, we found that, upon lysophosphatidic
acid (LPA) stimulation, cytoplasmic NHERF1 rapidly translocated to the plasma
membrane, and subsequently to cortical protrusion structures, of ovarian cancer
cells. This movement depended on direct binding of NHERF1 to C-terminally
phosphorylated ERM proteins (cpERMs). Moreover, NHERF1 depletion downregulated
cpERMs and further impaired cpERM-dependent remodeling of the cell cortex,
suggesting reciprocal regulation between these proteins. The LPA-induced protein
complex was highly enriched in migratory pseudopodia, whose formation was
impaired by overexpression of NHERF1 truncation mutants. Consistent with this,
NHERF1 depletion in various types of cancer cells abolished chemotactic cell
migration toward a LPA gradient. Taken together, our findings suggest that the
high dynamics of cytosolic NHERF1 provide cancer cells with a means of
controlling chemotactic migration. This capacity is likely to be essential for
ovarian cancer progression in tumor microenvironments containing LPA.
Collapse
Affiliation(s)
- Yong-Seok Oh
- Department of Brain-Cognitive Science, Daegu-Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea
| | - Kyun Heo
- Research Institute, National Cancer Center, Goyang, Republic of Korea
| | - Eung-Kyun Kim
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, Republic of Korea
| | - Jin-Hyeok Jang
- Department of Brain-Cognitive Science, Daegu-Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea
| | - Sun Sik Bae
- MRC for Ischemic Tissue Regeneration, Department of Pharmacology, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Jong Bae Park
- Research Institute, National Cancer Center, Goyang, Republic of Korea.,Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Republic of Korea
| | - Yun Hee Kim
- Research Institute, National Cancer Center, Goyang, Republic of Korea.,Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Republic of Korea
| | - Minseok Song
- Synaptic Circuit Plasticity Laboratory, Department of Structure and Function of Neural Network, Korea Brain Research Institute, Daegu, Republic of Korea
| | - Sang Ryong Kim
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Institute of Life Science and Biotechnology, Kyungpook National University, Daegu, Republic of Korea
| | - Sung Ho Ryu
- Division of Molecular and Life Science, Department of Life Science, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - In-Hoo Kim
- Research Institute, National Cancer Center, Goyang, Republic of Korea.,Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Republic of Korea
| | - Pann-Ghill Suh
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, Republic of Korea
| |
Collapse
|
34
|
Degryse B, Britto M, Shan CX, Wallace RG, Rochfort KD, Cummins PM, Meade G, Murphy RP. Moesin and merlin regulate urokinase receptor-dependent endothelial cell migration, adhesion and angiogenesis. Int J Biochem Cell Biol 2017; 88:14-22. [DOI: 10.1016/j.biocel.2017.04.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 04/02/2017] [Accepted: 04/28/2017] [Indexed: 10/19/2022]
|
35
|
Chen C, Nimlamool W, Miller CJ, Lou HJ, Turk BE. Rational Redesign of a Functional Protein Kinase-Substrate Interaction. ACS Chem Biol 2017; 12:1194-1198. [PMID: 28314095 PMCID: PMC5442603 DOI: 10.1021/acschembio.7b00089] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
![]()
Eukaryotic
protein kinases typically phosphorylate substrates in
the context of specific sequence motifs, contributing to specificity
essential for accurate signal transmission. Protein kinases recognize
their target sequences through complementary interactions within the
active site cleft. As a step toward the construction of orthogonal
kinase signaling systems, we have re-engineered the protein kinase
Pim1 to alter its phosphorylation consensus sequence. Residues in
the Pim1 catalytic domain interacting directly with a critical arginine
residue in the substrate were substituted to produce a kinase mutant
that instead accommodates a hydrophobic residue. We then introduced
a compensating mutation into a Pim1 substrate, the pro-apoptotic protein
BAD, to reconstitute phosphorylation both in vitro and in living cells. Coexpression of the redesigned kinase with
its substrate in cells protected them from apoptosis. Such orthogonal
kinase–substrate pairs provide tools to probe the functional
consequences of specific phosphorylation events in living cells and
to design synthetic signaling pathways.
Collapse
Affiliation(s)
- Catherine Chen
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut 06520, United States
| | - Wutigri Nimlamool
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut 06520, United States
| | - Chad J. Miller
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut 06520, United States
| | - Hua Jane Lou
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut 06520, United States
| | - Benjamin E. Turk
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut 06520, United States
| |
Collapse
|
36
|
Pelaseyed T, Viswanatha R, Sauvanet C, Filter JJ, Goldberg ML, Bretscher A. Ezrin activation by LOK phosphorylation involves a PIP 2-dependent wedge mechanism. eLife 2017; 6. [PMID: 28430576 PMCID: PMC5400502 DOI: 10.7554/elife.22759] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 03/27/2017] [Indexed: 12/15/2022] Open
Abstract
How cells specify morphologically distinct plasma membrane domains is poorly understood. Prior work has shown that restriction of microvilli to the apical aspect of epithelial cells requires the localized activation of the membrane-F-actin linking protein ezrin. Using an in vitro system, we now define a multi-step process whereby the kinase LOK specifically phosphorylates ezrin to activate it. Binding of PIP2 to ezrin induces a conformational change permitting the insertion of the LOK C-terminal domain to wedge apart the membrane and F-actin-binding domains of ezrin. The N-terminal LOK kinase domain can then access a site 40 residues distal from the consensus sequence that collectively direct phosphorylation of the appropriate threonine residue. We suggest that this elaborate mechanism ensures that ezrin is only phosphorylated at the plasma membrane, and with high specificity by the apically localized kinase LOK. DOI:http://dx.doi.org/10.7554/eLife.22759.001
Collapse
Affiliation(s)
- Thaher Pelaseyed
- Weill Institute for Molecular and Cell Biology, Cornell University, Ithaca, United States.,Department of Molecular Biology and Genetics, Cornell University, Ithaca, United States
| | - Raghuvir Viswanatha
- Weill Institute for Molecular and Cell Biology, Cornell University, Ithaca, United States.,Department of Molecular Biology and Genetics, Cornell University, Ithaca, United States.,Department of Genetics, Harvard Medical School, Boston, United States
| | - Cécile Sauvanet
- Weill Institute for Molecular and Cell Biology, Cornell University, Ithaca, United States.,Department of Molecular Biology and Genetics, Cornell University, Ithaca, United States
| | - Joshua J Filter
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, United States
| | - Michael L Goldberg
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, United States
| | - Anthony Bretscher
- Weill Institute for Molecular and Cell Biology, Cornell University, Ithaca, United States.,Department of Molecular Biology and Genetics, Cornell University, Ithaca, United States
| |
Collapse
|
37
|
Bastos P, Ferreira R, Manadas B, Moreira PI, Vitorino R. Insights into the human brain proteome: Disclosing the biological meaning of protein networks in cerebrospinal fluid. Crit Rev Clin Lab Sci 2017; 54:185-204. [PMID: 28393582 DOI: 10.1080/10408363.2017.1299682] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cerebrospinal fluid (CSF) is an excellent source of biological information regarding the nervous system, once it is in close contact and accurately reflects alterations in this system. Several studies have analyzed differential protein profiles of CSF samples between healthy and diseased human subjects. However, the pathophysiological mechanisms and how CSF proteins relate to diseases are still poorly known. By applying bioinformatics tools, we attempted to provide new insights on the biological and functional meaning of proteomics data envisioning the identification of putative disease biomarkers. Bioinformatics analysis of data retrieved from 99 mass spectrometry (MS)-based studies on CSF profiling highlighted 1985 differentially expressed proteins across 49 diseases. A large percentage of the modulated proteins originate from exosome vesicles, and the majority are involved in either neuronal cell growth, development, maturation, migration, or neurotransmitter-mediated cellular communication. Nevertheless, some diseases present a unique CSF proteome profile, which were critically analyzed in the present study. For instance, 48 proteins were found exclusively upregulated in the CSF of patients with Alzheimer's disease and are mainly involved in steroid esterification and protein activation cascade processes. A higher number of exclusively upregulated proteins were found in the CSF of patients with multiple sclerosis (76 proteins) and with bacterial meningitis (70 proteins). Whereas in multiple sclerosis, these proteins are mostly involved in the regulation of RNA metabolism and apoptosis, in bacterial meningitis the exclusively upregulated proteins participate in inflammation and antibacterial humoral response, reflecting disease pathogenesis. The exploration of the contribution of exclusively upregulated proteins to disease pathogenesis will certainly help to envision potential biomarkers in the CSF for the clinical management of nervous system diseases.
Collapse
Affiliation(s)
- Paulo Bastos
- a Department of Chemistry , University of Aveiro , Aveiro , Portugal.,b Department of Medical Sciences , Institute for Biomedicine - iBiMED, University of Aveiro , Aveiro , Portugal
| | - Rita Ferreira
- c QOPNA, Department of Chemistry , University of Aveiro , Aveiro , Portugal
| | - Bruno Manadas
- d CNC, Center for Neuroscience and Cell Biology, University of Coimbra , Coimbra , Portugal
| | - Paula I Moreira
- d CNC, Center for Neuroscience and Cell Biology, University of Coimbra , Coimbra , Portugal.,e Laboratory of Physiology, Faculty of Medicine , University of Coimbra , Coimbra , Portugal
| | - Rui Vitorino
- b Department of Medical Sciences , Institute for Biomedicine - iBiMED, University of Aveiro , Aveiro , Portugal.,f Departmento de Cirurgia e Fisiologia, Faculdade de Medicina , Unidade de Investigação Cardiovascular, Universidade do Porto , Porto , Portugal
| |
Collapse
|
38
|
Chen Y, Stookey J, Arsenault R, Scruten E, Griebel P, Napper S. Investigation of the physiological, behavioral, and biochemical responses of cattle to restraint stress. J Anim Sci 2017; 94:3240-3254. [PMID: 27695781 DOI: 10.2527/jas.2016-0549] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Stresses imposed on livestock have significant impact on their health and productivity as well as public perceptions of animal welfare. Understanding stress responses in livestock may help refine management procedures and facilitate selection of stress-tolerant animals. In this study, behavioral (chute entry order, chute behavior, and exit velocity), physiological (serum cortisol), and biochemical (kinome) responses were evaluated in cattle ( = 20) subjected to three 5-min restraint periods with weekly intervals. Correlations among stress responses were assessed across all animals as well as for subgroups ( = 4) representing animals consistently displaying a high and low extreme of serum cortisol responses. Across all animals, entry order ( = 0.006) and exit velocity ( = 0.023) were positively correlated with serum cortisol; however, these correlations were not consistently reproducible for the high and low serum cortisol responders. Kinome profiling of peripheral blood mononuclear cells revealed distinct signaling events between the high and low cortisol responders. In particular, kinome profiling revealed significant differences in carbohydrate metabolism and apoptosis that were independently validated. Furthermore, changes in serum glucose levels provided a reliable, inexpensive indicator of serum cortisol levels and often had greater predictive value than cortisol for stress-related behavioral responses. Serum cortisol levels displayed a pattern consistent with sensitization, whereas no habituation or sensitization was observed for serum glucose levels or behavioral responses. Collectively, this investigation provides insight into correlations among physiological, behavioral, and biochemical responses of cattle subjected to a brief restraint that may provide biomarkers for selection of stress-tolerant animals.
Collapse
|
39
|
Lee CF, Holownia A, Bennett JM, Elkins JM, St. Denis JD, Adachi S, Yudin AK. Oxalyl Boronates Enable Modular Synthesis of Bioactive Imidazoles. Angew Chem Int Ed Engl 2017. [DOI: 10.1002/ange.201611006] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- C. Frank Lee
- Davenport Research Laboratories; Department of Chemistry; University of Toronto; 80 St. George St. Toronto ON M5S 3H6 Canada
| | - Aleksandra Holownia
- Davenport Research Laboratories; Department of Chemistry; University of Toronto; 80 St. George St. Toronto ON M5S 3H6 Canada
| | - James M. Bennett
- Structural Genomics Consortium (SGC); Nuffield Department of Medicine; University of Oxford, Old Road Campus Research Building; Roosevelt Drive Oxford OX3 7DQ UK
| | - Jonathan M. Elkins
- Structural Genomics Consortium (SGC); Nuffield Department of Medicine; University of Oxford, Old Road Campus Research Building; Roosevelt Drive Oxford OX3 7DQ UK
| | - Jeffrey D. St. Denis
- Davenport Research Laboratories; Department of Chemistry; University of Toronto; 80 St. George St. Toronto ON M5S 3H6 Canada
| | - Shinya Adachi
- Davenport Research Laboratories; Department of Chemistry; University of Toronto; 80 St. George St. Toronto ON M5S 3H6 Canada
| | - Andrei K. Yudin
- Davenport Research Laboratories; Department of Chemistry; University of Toronto; 80 St. George St. Toronto ON M5S 3H6 Canada
| |
Collapse
|
40
|
Lee CF, Holownia A, Bennett JM, Elkins JM, St. Denis JD, Adachi S, Yudin AK. Oxalyl Boronates Enable Modular Synthesis of Bioactive Imidazoles. Angew Chem Int Ed Engl 2017; 56:6264-6267. [DOI: 10.1002/anie.201611006] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 01/16/2017] [Indexed: 01/15/2023]
Affiliation(s)
- C. Frank Lee
- Davenport Research Laboratories; Department of Chemistry; University of Toronto; 80 St. George St. Toronto ON M5S 3H6 Canada
| | - Aleksandra Holownia
- Davenport Research Laboratories; Department of Chemistry; University of Toronto; 80 St. George St. Toronto ON M5S 3H6 Canada
| | - James M. Bennett
- Structural Genomics Consortium (SGC); Nuffield Department of Medicine; University of Oxford, Old Road Campus Research Building; Roosevelt Drive Oxford OX3 7DQ UK
| | - Jonathan M. Elkins
- Structural Genomics Consortium (SGC); Nuffield Department of Medicine; University of Oxford, Old Road Campus Research Building; Roosevelt Drive Oxford OX3 7DQ UK
| | - Jeffrey D. St. Denis
- Davenport Research Laboratories; Department of Chemistry; University of Toronto; 80 St. George St. Toronto ON M5S 3H6 Canada
| | - Shinya Adachi
- Davenport Research Laboratories; Department of Chemistry; University of Toronto; 80 St. George St. Toronto ON M5S 3H6 Canada
| | - Andrei K. Yudin
- Davenport Research Laboratories; Department of Chemistry; University of Toronto; 80 St. George St. Toronto ON M5S 3H6 Canada
| |
Collapse
|
41
|
The protective role of MLCP-mediated ERM dephosphorylation in endotoxin-induced lung injury in vitro and in vivo. Sci Rep 2016; 6:39018. [PMID: 27976727 PMCID: PMC5157034 DOI: 10.1038/srep39018] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 11/17/2016] [Indexed: 12/13/2022] Open
Abstract
The goal of this study was to investigate the role of MLC phosphatase (MLCP) in a LPS model of acute lung injury (ALI). We demonstrate that ectopic expression of a constitutively-active (C/A) MLCP regulatory subunit (MYPT1) attenuates the ability of LPS to increase endothelial (EC) permeability. Down-regulation of MYPT1 exacerbates LPS-induced expression of ICAM1 suggesting an anti-inflammatory role of MLCP. To determine whether MLCP contributes to LPS-induced ALI in vivo, we utilized a nanoparticle DNA delivery method to specifically target lung EC. Expression of a C/A MYPT1 reduced LPS-induced lung inflammation and vascular permeability. Further, increased expression of the CS1β (MLCP catalytic subunit) also reduced LPS-induced lung inflammation, whereas the inactive CS1β mutant increased vascular leak. We next examined the role of the cytoskeletal targets of MLCP, the ERM proteins (Ezrin/Radixin/Moesin), in mediating barrier dysfunction. LPS-induced increase in EC permeability was accompanied by PKC-mediated increase in ERM phosphorylation, which was more prominent in CS1β-depleted cells. Depletion of Moesin and Ezrin, but not Radixin attenuated LPS-induced increases in permeability. Further, delivery of a Moesin phospho-null mutant into murine lung endothelium attenuated LPS-induced lung inflammation and vascular leak suggesting that MLCP opposes LPS-induced ALI by mediating the dephosphorylation of Moesin and Ezrin.
Collapse
|
42
|
Ji X, Liu Y, Hurd R, Wang J, Fitzmaurice B, Nishina PM, Chang B. Retinal Pigment Epithelium Atrophy 1 (rpea1): A New Mouse Model With Retinal Detachment Caused by a Disruption of Protein Kinase C, θ. Invest Ophthalmol Vis Sci 2016; 57:877-88. [PMID: 26978024 PMCID: PMC4794085 DOI: 10.1167/iovs.15-17495] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Retinal detachments (RDs), a separation of the light-sensitive tissue of the retina from its supporting layers in the posterior eye, isolate retinal cells from their normal supply of nourishment and can lead to their deterioration and death. We identified a new, spontaneous murine model of exudative retinal detachment, nm3342 (new mutant 3342, also referred to as rpea1: retinal pigment epithelium atrophy 1), which we characterize herein. Methods The chromosomal position for the recessive nm3342 mutation was determined by DNA pooling, and the causative mutation was discovered by comparison of whole exome sequences of mutant and wild-type controls. The effects of the mutation were examined in longitudinal studies by clinical evaluation, electroretinography (ERG), light microscopy, and marker and Western blot analyses. Results New mutant 3342, nm3342, also referred to as rpea1, causes an early-onset, complete RD on the ABJ/LeJ strain background, and central exudative RD and late-onset RPE atrophy on the C57BL/6J background. The ERG responses were normal at 2 months of age but deteriorate as mice age, concomitant with progressive pan-retinal photoreceptor loss. Genetic analysis localized rpea1 to mouse chromosome 2. By high-throughput sequencing of a whole exome capture library of an rpea1/rpea1 mutant and subsequent sequence analysis, a splice donor site mutation in the Prkcq (protein kinase C, θ) gene, was identified, leading to a skipping of exon 6, frame shift and premature termination. Homozygotes with a Prkcq-targeted null allele (Prkcqtm1Litt) have similar retinal phenotypes as homozygous rpea1 mice. We determined that the PKCθ protein is abundant in the lateral surfaces of RPE cells and colocalizes with both tight and adherens junction proteins. Phalloidin-stained RPE whole mounts showed abnormal RPE cell morphology with aberrant actin ring formation. Conclusions The homozygous Prkcqrpea1 and the null Prkcqtm1Litt mutants are reliable novel mouse models of RD and can also be used to study the effects of the disruption of PRKCQ (PKCθ) signaling in RPE cells.
Collapse
|
43
|
Tan L, Zhang Z, Gao D, Luo J, Tu ZC, Li Z, Peng L, Ren X, Ding K. 4-Oxo-1,4-dihydroquinoline-3-carboxamide Derivatives as New Axl Kinase Inhibitors. J Med Chem 2016; 59:6807-25. [PMID: 27379978 DOI: 10.1021/acs.jmedchem.6b00608] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Li Tan
- State
Key Laboratory of Respiratory Diseases, Guangzhou Institutes of Biomedicine
and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Guangzhou 510530, China
- University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing 100049, China
| | - Zhang Zhang
- State
Key Laboratory of Respiratory Diseases, Guangzhou Institutes of Biomedicine
and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Guangzhou 510530, China
- School
of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, China
| | - Donglin Gao
- State
Key Laboratory of Respiratory Diseases, Guangzhou Institutes of Biomedicine
and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Guangzhou 510530, China
- University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing 100049, China
| | - Jinfeng Luo
- State
Key Laboratory of Respiratory Diseases, Guangzhou Institutes of Biomedicine
and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Guangzhou 510530, China
| | - Zheng-Chao Tu
- State
Key Laboratory of Respiratory Diseases, Guangzhou Institutes of Biomedicine
and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Guangzhou 510530, China
| | - Zhengqiu Li
- School
of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, China
| | - Lijie Peng
- School
of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, China
| | - Xiaomei Ren
- State
Key Laboratory of Respiratory Diseases, Guangzhou Institutes of Biomedicine
and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Guangzhou 510530, China
- School
of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, China
| | - Ke Ding
- State
Key Laboratory of Respiratory Diseases, Guangzhou Institutes of Biomedicine
and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Guangzhou 510530, China
- School
of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, China
| |
Collapse
|
44
|
X-linked primary immunodeficiency associated with hemizygous mutations in the moesin (MSN) gene. J Allergy Clin Immunol 2016; 138:1681-1689.e8. [PMID: 27405666 DOI: 10.1016/j.jaci.2016.04.032] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 02/26/2016] [Accepted: 04/06/2016] [Indexed: 11/22/2022]
Abstract
BACKGROUND We investigated 7 male patients (from 5 different families) presenting with profound lymphopenia, hypogammaglobulinemia, fluctuating monocytopenia and neutropenia, a poor immune response to vaccine antigens, and increased susceptibility to bacterial and varicella zoster virus infections. OBJECTIVE We sought to characterize the genetic defect involved in a new form of X-linked immunodeficiency. METHODS We performed genetic analyses and an exhaustive phenotypic and functional characterization of the lymphocyte compartment. RESULTS We observed hemizygous mutations in the moesin (MSN) gene (located on the X chromosome and coding for MSN) in all 7 patients. Six of the latter had the same missense mutation, which led to an amino acid substitution (R171W) in the MSN four-point-one, ezrin, radixin, moesin domain. The seventh patient had a nonsense mutation leading to a premature stop codon mutation (R533X). The naive T-cell counts were particularly low for age, and most CD8+ T cells expressed the senescence marker CD57. This phenotype was associated with impaired T-cell proliferation, which was rescued by expression of wild-type MSN. MSN-deficient T cells also displayed poor chemokine receptor expression, increased adhesion molecule expression, and altered migration and adhesion capacities. CONCLUSION Our observations establish a causal link between an ezrin-radixin-moesin protein mutation and a primary immunodeficiency that could be referred to as X-linked moesin-associated immunodeficiency.
Collapse
|
45
|
Tsolis KC, Bagli E, Kanaki K, Zografou S, Carpentier S, Bei ES, Christoforidis S, Zervakis M, Murphy C, Fotsis T, Economou A. Proteome Changes during Transition from Human Embryonic to Vascular Progenitor Cells. J Proteome Res 2016; 15:1995-2007. [DOI: 10.1021/acs.jproteome.6b00180] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Konstantinos C. Tsolis
- Department
of Protein structure and Proteomics Facility, Institute of Molecular Biology and Biotechnology - FORTH, 70013 Iraklio, Crete, Greece
- Department
of Biology, University of Crete, 70013 Iraklio, Crete, Greece
| | - Eleni Bagli
- Division
of Biomedical Research, Institute of Molecular Biology and Biotechnology - FORTH, 45110 Ioaninna, Greece
| | - Katerina Kanaki
- Department
of Protein structure and Proteomics Facility, Institute of Molecular Biology and Biotechnology - FORTH, 70013 Iraklio, Crete, Greece
| | - Sofia Zografou
- Division
of Biomedical Research, Institute of Molecular Biology and Biotechnology - FORTH, 45110 Ioaninna, Greece
| | - Sebastien Carpentier
- SYBIOMA, KU Leuven facility for Systems Biology Based Mass Spectrometry, B-3000 Leuven Belgium
| | - Ekaterini S. Bei
- School
of Electronic and Computer Engineering, Technical University of Crete, 73100 Chania, Greece
| | - Savvas Christoforidis
- Division
of Biomedical Research, Institute of Molecular Biology and Biotechnology - FORTH, 45110 Ioaninna, Greece
- Laboratory
of Biological Chemistry, Medical School, University of Ioannina, 45110 Ioannina, Greece
| | - Michalis Zervakis
- School
of Electronic and Computer Engineering, Technical University of Crete, 73100 Chania, Greece
| | - Carol Murphy
- Division
of Biomedical Research, Institute of Molecular Biology and Biotechnology - FORTH, 45110 Ioaninna, Greece
- School
of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, U.K
| | - Theodore Fotsis
- Division
of Biomedical Research, Institute of Molecular Biology and Biotechnology - FORTH, 45110 Ioaninna, Greece
- Laboratory
of Biological Chemistry, Medical School, University of Ioannina, 45110 Ioannina, Greece
- School
of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, U.K
| | - Anastassios Economou
- Department
of Protein structure and Proteomics Facility, Institute of Molecular Biology and Biotechnology - FORTH, 70013 Iraklio, Crete, Greece
- Department
of Biology, University of Crete, 70013 Iraklio, Crete, Greece
- SYBIOMA, KU Leuven facility for Systems Biology Based Mass Spectrometry, B-3000 Leuven Belgium
| |
Collapse
|
46
|
Richter E, Ventz K, Harms M, Mostertz J, Hochgräfe F. Induction of Macrophage Function in Human THP-1 Cells Is Associated with Rewiring of MAPK Signaling and Activation of MAP3K7 (TAK1) Protein Kinase. Front Cell Dev Biol 2016; 4:21. [PMID: 27066479 PMCID: PMC4811913 DOI: 10.3389/fcell.2016.00021] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 03/09/2016] [Indexed: 12/12/2022] Open
Abstract
Macrophages represent the primary human host response to pathogen infection and link the immediate defense to the adaptive immune system. Mature tissue macrophages convert from circulating monocyte precursor cells by terminal differentiation in a process that is not fully understood. Here, we analyzed the protein kinases of the human monocytic cell line THP-1 before and after induction of macrophage differentiation by using kinomics and phosphoproteomics. When comparing the macrophage-like state with the monocytic precursor, 50% of the kinome was altered in expression and even 71% of covered kinase phosphorylation sites were affected. Kinome rearrangements are for example characterized by a shift of overrepresented cyclin-dependent kinases associated with cell cycle control in monocytes to calmodulin-dependent kinases and kinases involved in proinflammatory signaling. Eventually, we show that monocyte-to-macrophage differentiation is associated with major rewiring of mitogen-activated protein kinase signaling networks and demonstrate that protein kinase MAP3K7 (TAK1) acts as the key signaling hub in bacterial killing, chemokine production and differentiation. Our study proves the fundamental role of protein kinases and cellular signaling as major drivers of macrophage differentiation and function. The finding that MAP3K7 is central to macrophage function suggests MAP3K7 and its networking partners as promising targets in host-directed therapy for macrophage-associated disease.
Collapse
Affiliation(s)
- Erik Richter
- Junior Research Group Pathoproteomics, Competence Center Functional Genomics, University of Greifswald Greifswald, Germany
| | - Katharina Ventz
- Junior Research Group Pathoproteomics, Competence Center Functional Genomics, University of Greifswald Greifswald, Germany
| | - Manuela Harms
- Junior Research Group Pathoproteomics, Competence Center Functional Genomics, University of Greifswald Greifswald, Germany
| | - Jörg Mostertz
- Junior Research Group Pathoproteomics, Competence Center Functional Genomics, University of Greifswald Greifswald, Germany
| | - Falko Hochgräfe
- Junior Research Group Pathoproteomics, Competence Center Functional Genomics, University of Greifswald Greifswald, Germany
| |
Collapse
|
47
|
Abstract
In all eukaryotes, the plasma membrane is critically important as it maintains the architectural integrity of the cell. Proper anchorage and interaction between the plasma membrane and the cytoskeleton is critical for normal cellular processes. The ERM (ezrin-radixin-moesin) proteins are a class of highly homologous proteins involved in linking the plasma membrane to the cortical actin cytoskeleton. This review takes a succinct look at the biology of the ERM proteins including their structure and function. Current reports on their regulation that leads to activation and deactivation was examined before taking a look at the different interacting partners. Finally, emerging roles of each of the ERM family members in cancer was highlighted.
Collapse
Affiliation(s)
- Godwin A Ponuwei
- Cell migration laboratory, Molecular and Cellular Medicine Unit, Department of Biomedical Sciences, School of Biological Sciences, Hopkins Building, University of Reading, Whiteknights, Berkshire, UK. .,Molecular and Cellular Medicine unit, Department of Biomedical sciences, School of Life Sciences, Hopkins Building, Whiteknights Campus, University of Reading, Reading, Berkshire, UK.
| |
Collapse
|
48
|
Leroy C, Belkina NV, Long T, Deruy E, Dissous C, Shaw S, Tulasne D. Caspase Cleavages of the Lymphocyte-oriented Kinase Prevent Ezrin, Radixin, and Moesin Phosphorylation during Apoptosis. J Biol Chem 2016; 291:10148-61. [PMID: 26945071 DOI: 10.1074/jbc.m116.721365] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Indexed: 11/06/2022] Open
Abstract
The lymphocyte-oriented kinase (LOK), also called serine threonine kinase 10 (STK10), is synthesized mainly in lymphocytes. It is involved in lymphocyte migration and polarization and can phosphorylate ezrin, radixin, and moesin (the ERM proteins). In a T lymphocyte cell line and in purified human lymphocytes, we found LOK to be cleaved by caspases during apoptosis. The first cleavage occurs at aspartic residue 332, located between the kinase domain and the coiled-coil regulation domain. This cleavage generates an N-terminal fragment, p50 N-LOK, containing the kinase domain and a C-terminal fragment, which is further cleaved during apoptosis. Although these cleavages preserve the entire kinase domain, p50 N-LOK displays no kinase activity. In apoptotic lymphocytes, caspase cleavages of LOK are concomitant with a decrease in ERM phosphorylation. When non-apoptotic lymphocytes from mice with homozygous and heterozygous LOK knockout were compared, the latter showed a higher level of ERM phosphorylation, but when apoptosis was induced, LOK(-/-) and LOK(+/-) lymphocytes showed the same low level, confirming in vivo that LOK-induced ERM phosphorylation is prevented during lymphocyte apoptosis. Our results demonstrate that cleavage of LOK during apoptosis abolishes its kinase activity, causing a decrease in ERM phosphorylation, crucial to the role of the ERM proteins in linking the plasma membrane to actin filaments.
Collapse
Affiliation(s)
- Catherine Leroy
- From the University of Lille, CNRS, Institut Pasteur de Lille, Unité Mixte de Recherche (UMR) 8161, Mechanisms of Tumorigenesis and Target Therapies, 59000 Lille, France
| | | | - Thavy Long
- the University of Lille, CNRS, INSERM, Centre Hospitalier Régional Universitaire de Lille, Institut Pasteur de Lille, U1019-UMR 8204, Center for Infection and Immunity of Lille, 59000 Lille, France
| | - Emeric Deruy
- the University of Lille, CNRS, INSERM, Centre Hospitalier Régional Universitaire de Lille, Institut Pasteur de Lille, U1019-UMR 8204, Center for Infection and Immunity of Lille, 59000 Lille, France
| | - Colette Dissous
- the University of Lille, CNRS, INSERM, Centre Hospitalier Régional Universitaire de Lille, Institut Pasteur de Lille, U1019-UMR 8204, Center for Infection and Immunity of Lille, 59000 Lille, France
| | - Stephen Shaw
- Experimental Immunology Branch Branches, NCI/National Institutes of Health, Bethesda, Maryland 20892, and
| | - David Tulasne
- From the University of Lille, CNRS, Institut Pasteur de Lille, Unité Mixte de Recherche (UMR) 8161, Mechanisms of Tumorigenesis and Target Therapies, 59000 Lille, France
| |
Collapse
|
49
|
Locard-Paulet M, Lim L, Veluscek G, McMahon K, Sinclair J, van Weverwijk A, Worboys JD, Yuan Y, Isacke CM, Jørgensen C. Phosphoproteomic analysis of interacting tumor and endothelial cells identifies regulatory mechanisms of transendothelial migration. Sci Signal 2016; 9:ra15. [PMID: 26861043 PMCID: PMC6485367 DOI: 10.1126/scisignal.aac5820] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
The exit of metastasizing tumor cells from the vasculature, extravasation, is regulated by their dynamic interactions with the endothelial cells that line the internal surface of vessels. To elucidate signals controlling tumor cell adhesion to the endothelium and subsequent transendothelial migration, we performed phosphoproteomic analysis to map cell-specific changes in protein phosphorylation that were triggered by contact between metastatic MDA-MB-231 breast cancer cells and endothelial cells. From the 2669 unique phosphorylation sites identified, 77 and 43 were differentially phosphorylated in the tumor cells and endothelial cells, respectively. The receptor tyrosine kinase ephrin type A receptor 2 (EPHA2) exhibited decreased Tyr(772) phosphorylation in the cancer cells upon endothelial contact. Knockdown of EPHA2 increased adhesion of the breast cancer cells to human umbilical vein endothelial cells (HUVECs) and their transendothelial migration in coculture cell assays, as well as early-stage lung colonization in vivo. EPHA2-mediated inhibition of transendothelial migration of breast cancer cells depended on interaction with the ligand ephrinA1 on HUVECs and phosphorylation of EPHA2-Tyr(772). When EPHA2 phosphorylation dynamics were compared between cell lines of different metastatic ability, EPHA2-Tyr(772) was rapidly dephosphorylated after ephrinA1 stimulation specifically in cells targeting the lung. Knockdown of the phosphatase LMW-PTP reduced adhesion and transendothelial migration of the breast cancer cells. Overall, cell-specific phosphoproteomic analysis provides a bidirectional map of contact-initiated signaling between tumor and endothelial cells that can be further investigated to identify mechanisms controlling the transendothelial cell migration of cancer cells.
Collapse
Affiliation(s)
- Marie Locard-Paulet
- Division of Cancer Biology, The Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK. Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | - Lindsay Lim
- Division of Cancer Biology, The Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| | - Giulia Veluscek
- Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | - Kelly McMahon
- Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | - John Sinclair
- Division of Cancer Biology, The Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| | - Antoinette van Weverwijk
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
| | - Jonathan D Worboys
- Division of Cancer Biology, The Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK. Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | - Yinyin Yuan
- Centre for Evolution and Cancer and Centre for Molecular Pathology, Division of Molecular Pathology, The Institute of Cancer Research, 15 Cotswold Road, Sutton SM2 5NG, UK
| | - Clare M Isacke
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
| | - Claus Jørgensen
- Division of Cancer Biology, The Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK. Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester M20 4BX, UK.
| |
Collapse
|
50
|
Suda J, Rockey DC, Karvar S. Akt2-Dependent Phosphorylation of Radixin in Regulation of Mrp-2 Trafficking in WIF-B Cells. Dig Dis Sci 2016; 61:453-63. [PMID: 26500117 DOI: 10.1007/s10620-015-3905-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 09/24/2015] [Indexed: 12/09/2022]
Abstract
BACKGROUND The dominant ezrin/radixin/moesin protein in hepatocytes is radixin, which plays an important role in mediating the binding of F-actin to the plasma membrane after a conformational activation by phosphorylation at Thr564. AIM Here we have investigated the importance of Akt-mediated radixin Thr564 phosphorylation on Mrp-2 distribution and function in WIF-B cells. Mrp-2 is an adenosine triphosphate (ATP)-binding cassette transporter that plays an important role in detoxification and chemoprotection by transporting a wide range of compounds, especially conjugates of lipophilic substances with glutathione, organic anions, and drug metabolites such as glucuronides. METHODS Akt1 and Akt2 expression were manipulated using dominant active and negative constructs as well as Akt1 and Akt2 siRNA. Cellular distribution of radixin and Mrp-2 was visualized by fluorescence microscopy. A 5-chloromethylfluorescein diacetate, which is a substrate of the Mrp-2 and is actively transported in canalicular lumina, was used to measure Mrp-2 function. RESULTS Radixin phosphorylation was significantly increased in wild-type and dominant active Akt2 transfected cells. Furthermore, radixin and Mrp-2 were localized at the canalicular membrane, similar to control cells. In contrast, overexpression of dominant negative Akt2, siRNA knockdown of Akt2 and a specific Akt inhibitor prevented radixin phosphorylation and led to alteration of normal radixin and Mrp-2 localization; inhibition of Akt2, but not Akt1 function led to radixin localization to the cytoplasmic space. In addition, dominant negative and Akt2 knockdown led to a dramatically impaired hepatocyte secretory response, while wild-type and dominant active Akt2 transfected cells exhibited increased 5-chloromethylfluorescein diacetate excretion. In contrast to Akt2, Akt1 was not associated with radixin phosphorylation. CONCLUSIONS These studies, therefore, identify Akt2 as a critical kinase that regulates radixin phosphorylation and leads to Mrp-2 translocation and function.
Collapse
|