1
|
Tuieng RJ, Cartmell SH, Kirwan CC, Eckersley A, Sherratt MJ. X-Ray Exposure Induces Structural Changes in Human Breast Proteins. Int J Mol Sci 2025; 26:5696. [PMID: 40565164 PMCID: PMC12193695 DOI: 10.3390/ijms26125696] [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: 04/30/2025] [Revised: 06/01/2025] [Accepted: 06/10/2025] [Indexed: 06/28/2025] Open
Abstract
During radiotherapy, X-rays can deliver significant doses of ionising radiation to both cancerous and healthy tissue, often leading to undesirable side effects that compromise patient outcomes. While the cellular effects of such therapeutic X-ray exposures are well studied, the impact on extracellular matrix (ECM) proteins remains poorly understood. This study characterises the response of ECM proteins, including the major tissue components collagen I and fibronectin (FN), to X-ray doses similar to those used in clinical practice (50 Gy, as employed in breast radiotherapy, and 100 Gy), using a combination of gel electrophoresis, biochemical assays, and mass spectrometry-based peptide location fingerprinting (PLF) analysis. In purified protein solutions, 50 Gy X-ray exposure led to the fragmentation of constituent collagen I α chains. Irradiation of purified plasma FN (pFN) induced localised changes in peptide yields (detected by liquid chromatography and tandem mass spectrometry (LC-MS/MS) and PLF) and enhanced its binding to collagen I. In complex environments, such as newly synthesised fibroblast-derived ECM and mature ex vivo breast tissue, X-ray exposure induced peptide yield changes in not only collagen I and FN but also key basement membrane proteins, including collagen IV, laminin, and perlecan. Intracellular proteins associated with gene expression (RPS3, MeCP2), the cytoskeleton (moesin, plectin), and the endoplasmic reticulum (calnexin) were also found to be impacted. These X-ray-induced structural changes may impair the ECM integrity and alter cell-ECM interactions, with potential implications for tissue stiffening, fibrosis, and impaired wound healing in irradiated tissues.
Collapse
Affiliation(s)
- Ren Jie Tuieng
- Division of Cell Matrix Biology & Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester M13 9PT, UK;
- Singapore Nuclear Research and Safety Institute, National University of Singapore, Singapore 118415, Singapore
| | - Sarah H. Cartmell
- Department of Materials, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, Manchester M13 9PL, UK;
- The Henry Royce Institute, Royce Hub Building, The University of Manchester, Manchester M13 9PL, UK
| | - Cliona C. Kirwan
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Oglesby Cancer Research Building, Manchester Cancer Research Centre, Wilmslow Road, Manchester M20 4BX, UK;
- The Nightingale Breast Cancer Unit, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester M23 9LT, UK
| | - Alexander Eckersley
- Division of Musculoskeletal & Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester M13 9PT, UK;
- Manchester Cell-Matrix Centre, Faculty of Biology, Medicine and Health, Manchester M13 9PT, UK
| | - Michael J. Sherratt
- Division of Cell Matrix Biology & Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester M13 9PT, UK;
- Manchester Cell-Matrix Centre, Faculty of Biology, Medicine and Health, Manchester M13 9PT, UK
| |
Collapse
|
2
|
Anselmi NK, Vanyo ST, Visser MB. Emerging oral Treponema membrane proteins disorder neutrophil phosphoinositide signaling via phosphatidylinositol-4-phosphate 5-kinase. FRONTIERS IN ORAL HEALTH 2025; 6:1568983. [PMID: 40248422 PMCID: PMC12003349 DOI: 10.3389/froh.2025.1568983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Accepted: 03/20/2025] [Indexed: 04/19/2025] Open
Abstract
Background Periodontitis (PD) is a group of inflammatory pathologies characterized by destruction of the tooth-supporting tissues. During PD, dysbiosis of the oral biofilm disrupts the host immune response and supports growth of pathogenic bacteria including the spirochetes Treponema denticola (Td), T. maltophilum (Tm), and T. lecithinolyticum (Tl). The outer membrane protein of Td, Msp, perturbs the function of neutrophils by modulating phosphoinositide (PIP) signaling. While Tm and Tl have similar outer membrane proteins, MspA and MspTL respectively, little is known of how these proteins affect neutrophil function. Methods This study examines putative mechanisms by which T. maltophilum MspA and T. lecithinolyticum MspTL inhibit neutrophil chemotaxis. Murine bone marrow neutrophils were treated with recombinant MspA or MspTL protein. Protein phosphorylation was assessed via immunoblot, phosphate release by malachite green assay, and PTEN and SHIP phosphatase activity through immunoprecipitation, enzymatic assays, and chemical inhibition. PIP quantification was assessed by immunofluorescence microscopy and Mass ELISAs, while small GTPase activity was measured with G-Protein Activation Assays. Neutrophil F-actin localization was determined through immunofluorescence. Results MspA and MspTL increase phosphate release in neutrophils, but unlike Msp, they do not affect PTEN or SHIP activity, despite modulating cellular levels of multiple PIP species [PI(3,4)P2, PI(4,5)P2, and PIP3]. Overall, MspA and MspTL differentially affected the metabolism of individual PIP species, but both increased PI(4,5)P2 levels in a PIP5K-dependent manner. Downstream effects of disrupted PIP signaling included inhibition of Akt and Rac1 activation and increased cortical F-actin localization. Conclusions Understanding distinct mechanistic relationships between novel Msp proteins and neutrophils provides important insight into how these understudied bacteria promote periodontitis progression.
Collapse
Affiliation(s)
| | | | - Michelle B. Visser
- Department of Oral Biology, The State University of New York at Buffalo, Buffalo, NY, United States
| |
Collapse
|
3
|
Cheung BCH, Chen X, Davis HJ, Nordmann CS, Toth J, Hodgson L, Segall JE, Shenoy VB, Wu M. Identification of CD44 as a key engager to hyaluronic acid-rich extracellular matrices for cell traction force generation and tumor invasion in 3D. Matrix Biol 2025; 135:1-11. [PMID: 39528207 PMCID: PMC11729355 DOI: 10.1016/j.matbio.2024.11.004] [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: 06/28/2024] [Revised: 11/07/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
Mechanical properties of the extracellular matrix (ECM) critically regulate a number of important cell functions including growth, differentiation and migration. Type I collagen and glycosaminoglycans (GAGs) are two primary components of ECMs that contribute to mammalian tissue mechanics, with the collagen fiber network sustaining tension, and GAGs withstanding compression. The architecture and stiffness of the collagen network are known to be important for cell-ECM mechanical interactions via cell surface adhesion receptor integrin. In contrast, studies of GAGs in modulating cell-ECM interactions are limited. Here, we present experimental studies on the roles of hyaluronic acid (HA) in single tumor cell traction force generation using a recently developed 3D cell traction force microscopy method. Our work reveals that CD44, a cell surface receptor to HA, is engaged in cell traction force generation in conjunction with β1-integrin. We find that HA significantly modifies the architecture and mechanics of the collagen fiber network, decreasing tumor cells' propensity to remodel the collagen network, attenuating traction force generation, transmission distance, and tumor invasion. Our findings point to a novel role for CD44 in traction force generation, which can be a potential therapeutic target for diseases involving HA rich ECMs such as breast cancer and glioblastoma.
Collapse
Affiliation(s)
- Brian C H Cheung
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, USA
| | - Xingyu Chen
- Center for Engineering MechanoBiology, University of Pennsylvania, Philadelphia, PA, USA; Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Hannah J Davis
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, USA; Department of Biological Sciences, Cornell University, Ithaca, NY, USA
| | - Cassidy S Nordmann
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, USA; Department of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Joshua Toth
- Center for Engineering MechanoBiology, University of Pennsylvania, Philadelphia, PA, USA; Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Louis Hodgson
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jeffrey E Segall
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Vivek B Shenoy
- Center for Engineering MechanoBiology, University of Pennsylvania, Philadelphia, PA, USA; Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Mingming Wu
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
4
|
Joachim J, Maselli D, Petsolari E, Aman J, Swiatlowska P, Killock D, Chaudhry H, Zarban AA, Sarker M, Fraser P, Cleary SJ, Amison R, Cuthbert I, Yang Y, Meier M, Fraternali F, Brain SD, Shah AM, Ivetic A. TNIK: A redox sensor in endothelial cell permeability. SCIENCE ADVANCES 2024; 10:eadk6583. [PMID: 39705357 DOI: 10.1126/sciadv.adk6583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 11/18/2024] [Indexed: 12/22/2024]
Abstract
Dysregulation of endothelial barrier integrity can lead to vascular leak and potentially fatal oedema. TNF-α controls endothelial permeability during inflammation and requires the actin organizing Ezrin-Radixin-Moesin (ERM) proteins. We identified TRAF2 and NCK-interacting kinase (TNIK) as a kinase directly phosphorylating and activating ERM, specifically at the plasma membrane of primary human endothelial cells. TNIK mediates TNF-α-dependent cellular stiffness and paracellular gap formation in vitro and is essential in driving inflammatory oedema formation in vivo. Unlike its homologs, TNIK activity is negatively and reversibly regulated by H2O2-mediated oxidation of C202 within the kinase domain. TNIK oxidation results in intermolecular disulfide bond formation and loss of kinase activity. Pharmacologic inhibition of endogenous reactive oxygen species production in endothelial cells elevated TNIK-dependent ERM phosphorylation, endothelial cell contraction, and cell rounding. Together, we highlight an interplay between TNIK, ERM phosphorylation, and redox signalling in regulating TNF-induced endothelial cell permeability.
Collapse
Affiliation(s)
- Justin Joachim
- School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, BHF Centre of Research Excellence, 125 Coldharbour Lane, King's College London, London SE5 9NU, UK
| | - Davide Maselli
- School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, BHF Centre of Research Excellence, 125 Coldharbour Lane, King's College London, London SE5 9NU, UK
| | - Emmanouela Petsolari
- Randall Centre for Cell and Molecular Biology, King's College London, London SE1 1UL, UK
| | - Jurjan Aman
- Department of Pulmonary Medicine, Amsterdam University Medical Center, location VUMC, Amsterdam, The Netherlands
| | - Pamela Swiatlowska
- Myocardial Function, National Heart and Lung Institute, Imperial College London, ICTEM, Hammersmith Hospital, London, UK
| | - David Killock
- School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, BHF Centre of Research Excellence, 125 Coldharbour Lane, King's College London, London SE5 9NU, UK
| | - Hiba Chaudhry
- School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, BHF Centre of Research Excellence, 125 Coldharbour Lane, King's College London, London SE5 9NU, UK
| | - Ali A Zarban
- School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, BHF Centre of Research Excellence, 125 Coldharbour Lane, King's College London, London SE5 9NU, UK
- Department of Pharmacological Sciences, Faculty of Pharmacy, Jazan University, Saudi Arabia
| | - Mosharraf Sarker
- Pharmacy and Biomolecular Sciences, Faculty of Science, Liverpool John Moores University, Liverpool, UK
| | - Paul Fraser
- School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, BHF Centre of Research Excellence, 125 Coldharbour Lane, King's College London, London SE5 9NU, UK
| | - Simon J Cleary
- Institute of Pharmaceutical Science, King's College London, Floor 5, Southwark Wing, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK
| | - Richard Amison
- School of Cancer and Pharmaceutical Sciences, Pulmonary Pharmacology Unit, King's College London, London, UK
| | - Isabelle Cuthbert
- School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, BHF Centre of Research Excellence, 125 Coldharbour Lane, King's College London, London SE5 9NU, UK
| | - Yue Yang
- School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, BHF Centre of Research Excellence, 125 Coldharbour Lane, King's College London, London SE5 9NU, UK
| | - Magda Meier
- School of Genetics and Genomic Medicine, University College London Institute of Child Health, London, UK
| | - Franca Fraternali
- Randall Centre for Cell and Molecular Biology, King's College London, London SE1 1UL, UK
- Division of Biosciences, Structural and Molecular Biology Department, University College London, Darwin (SMB) Building, Gower Street, London WC1E 6BT, UK
- Department of Structural and Molecular Biology, Division of Biosciences and Institute of Structural and Molecular Biology, University College London, London WC1E 6BT, UK Department of Biological Sciences, Birkbeck, University of London, London WC1E 7HX, United Kingdom
| | - Susan D Brain
- School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, BHF Centre of Research Excellence, 125 Coldharbour Lane, King's College London, London SE5 9NU, UK
| | - Ajay M Shah
- School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, BHF Centre of Research Excellence, 125 Coldharbour Lane, King's College London, London SE5 9NU, UK
| | - Aleksandar Ivetic
- School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, BHF Centre of Research Excellence, 125 Coldharbour Lane, King's College London, London SE5 9NU, UK
| |
Collapse
|
5
|
Kennedy C, Doyle R, Gough O, Mcevoy C, McAnallen S, Hughes M, Sheng X, Crifo B, Andrews D, Gaffney A, Rodriguez J, Kennedy S, Dillon E, Crean D, Zhang W, Yi Z, Nair V, Susztak K, Hirschhorn J, Florez J, Groop PH, Sandholm N, Kretzler M, McKay GJ, McKnight AJ, Maxwell AP, Matallanas D, Dorman A, Martin F, Conlon PJ, Sadlier DM, Brennan E, Godson C. A Novel Role for FERM Domain-Containing Protein 3 in CKD. KIDNEY360 2024; 5:1799-1812. [PMID: 39450948 PMCID: PMC11687992 DOI: 10.34067/kid.0000000602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 09/27/2024] [Indexed: 10/26/2024]
Abstract
Key Points We have identified a transcriptional signature of 93 genes associated with CKD severity and progression. Protein 4.1, ezrin, radixin, moesin domain-containing protein 3 gene expression is reduced in the context of more severe kidney disease and in individuals who go on to develop progressive disease. Protein 4.1, ezrin, radixin, moesin domain-containing protein 3 interacts with proteins of the cell cytoskeleton and cell-cell junctions in proximal tubule epithelial cells. Background Currently, there are limited methods to link disease severity and risk of disease progression in CKD. To better understand this potential relationship, we interrogated the renal transcriptomic profile of individuals with CKD with measures of CKD severity and identified protein 4.1, ezrin, radixin, moesin-domain containing protein 3 (FRMD3 ) as a candidate gene for follow-up study. Methods RNA-sequencing was used to profile the transcriptome of CKD biopsies from the North Dublin Renal BioBank, the results of which were correlated with clinical parameters. The potential function of FRMD3 was explored by interrogating the FRMD3 interactome and assessing the effect of lentiviral mediated FRMD3 knock down on human renal proximal tubule epithelial cells by assessing cell viability, metabolic activity, and structural markers. Results We identified a subset of 93 genes which are significantly correlated with eGFR and percentage tubulointerstitial fibrosis at time of biopsy and with CKD progression 5 years postbiopsy. These results were validated against transcriptomic data from an external cohort of 432 nephrectomy samples. One of the top-ranking genes from this subset, FRMD3, has previously been associated with the risk of developing diabetic kidney disease. Interrogating the interactome of FRMD3 in tubule epithelial cells revealed interactions with cytoskeletal components of cell-cell junctions. Knockdown of FRMD3 expression in tubule epithelial cells resulted in increased proapoptotic activity within the cells, as well as dysregulation of E-Cadherin. Conclusions We have identified a panel of kidney-specific transcripts correlated with severity and progression of kidney disease, and from this, we have identified a possible role for FRMD3 in tubule cell structure and health.
Collapse
Affiliation(s)
- Ciarán Kennedy
- UCD Diabetes Complications Research Centre, UCD School of Medicine, Conway Institute, University College Dublin, Dublin, Ireland
| | - Ross Doyle
- UCD Diabetes Complications Research Centre, UCD School of Medicine, Conway Institute, University College Dublin, Dublin, Ireland
- School of Medicine, Mater Misericordiae University Hospital, University College Dublin, Dublin, Ireland
| | - Oisin Gough
- UCD Diabetes Complications Research Centre, UCD School of Medicine, Conway Institute, University College Dublin, Dublin, Ireland
| | - Caitriona Mcevoy
- UCD Diabetes Complications Research Centre, UCD School of Medicine, Conway Institute, University College Dublin, Dublin, Ireland
- Tallaght University Hospital, Dublin; and Trinity Kidney Centre, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Susan McAnallen
- UCD Diabetes Complications Research Centre, UCD School of Medicine, Conway Institute, University College Dublin, Dublin, Ireland
| | - Maria Hughes
- UCD Diabetes Complications Research Centre, UCD School of Medicine, Conway Institute, University College Dublin, Dublin, Ireland
| | - Xin Sheng
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Liangzhu Laboratory, Zhejiang University, Hangzhou, China
- Department of Nephrology, The Children's Hospital, Zhejiang University School of Medicine and National Clinical Research Center for Child Health, Hangzhou, China
| | - Bianca Crifo
- School of Biomolecular and Biomedical Science, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Darrell Andrews
- UCD Diabetes Complications Research Centre, UCD School of Medicine, Conway Institute, University College Dublin, Dublin, Ireland
| | - Andrew Gaffney
- UCD Diabetes Complications Research Centre, UCD School of Medicine, Conway Institute, University College Dublin, Dublin, Ireland
| | - Javier Rodriguez
- Systems Biology Ireland, University College Dublin, Dublin, Ireland
| | - Susan Kennedy
- Systems Biology Ireland, University College Dublin, Dublin, Ireland
- TriviumVet, Waterford, Ireland
| | - Eugene Dillon
- UCD Conway Institute Core Technologies, University College Dublin, Dublin, Ireland
| | - Daniel Crean
- UCD Diabetes Complications Research Centre, UCD School of Medicine, Conway Institute, University College Dublin, Dublin, Ireland
- School of Veterinary Medicine, University College Dublin, Dublin, Ireland
| | - Weijia Zhang
- Icahn School of Medicine at Mount Sinai, New York, New York
| | - Zhengzi Yi
- Icahn School of Medicine at Mount Sinai, New York, New York
| | - Viji Nair
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Katalin Susztak
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Joel Hirschhorn
- Endocrine Division and Diabetes Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Jose Florez
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Per-Henrik Groop
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Niina Sandholm
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Matthias Kretzler
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Gareth J. McKay
- Centre for Public Health, Queens University of Belfast, Northern Ireland, United Kingdom
| | - Amy Jayne McKnight
- Centre for Public Health, Queens University of Belfast, Northern Ireland, United Kingdom
| | - Alexander P. Maxwell
- Centre for Public Health, Queens University of Belfast, Northern Ireland, United Kingdom
| | - David Matallanas
- Systems Biology Ireland, University College Dublin, Dublin, Ireland
| | - Anthony Dorman
- Department of Pathology, Beaumont Hospital, Dublin, Ireland
| | - Finian Martin
- UCD Diabetes Complications Research Centre, UCD School of Medicine, Conway Institute, University College Dublin, Dublin, Ireland
| | - Peter J. Conlon
- National Kidney Transplant Service, Department of Nephrology and Kidney Transplantation, Beaumont Hospital, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Denise M. Sadlier
- School of Medicine, Mater Misericordiae University Hospital, University College Dublin, Dublin, Ireland
| | - Eoin Brennan
- UCD Diabetes Complications Research Centre, UCD School of Medicine, Conway Institute, University College Dublin, Dublin, Ireland
| | - Catherine Godson
- UCD Diabetes Complications Research Centre, UCD School of Medicine, Conway Institute, University College Dublin, Dublin, Ireland
| |
Collapse
|
6
|
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
|
7
|
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
|
8
|
Eshaq AM, Flanagan TW, Hassan SY, Al Asheikh SA, Al-Amoudi WA, Santourlidis S, Hassan SL, Alamodi MO, Bendhack ML, Alamodi MO, Haikel Y, Megahed M, Hassan M. Non-Receptor Tyrosine Kinases: Their Structure and Mechanistic Role in Tumor Progression and Resistance. Cancers (Basel) 2024; 16:2754. [PMID: 39123481 PMCID: PMC11311543 DOI: 10.3390/cancers16152754] [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: 05/29/2024] [Revised: 06/30/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
Protein tyrosine kinases (PTKs) function as key molecules in the signaling pathways in addition to their impact as a therapeutic target for the treatment of many human diseases, including cancer. PTKs are characterized by their ability to phosphorylate serine, threonine, or tyrosine residues and can thereby rapidly and reversibly alter the function of their protein substrates in the form of significant changes in protein confirmation and affinity for their interaction with protein partners to drive cellular functions under normal and pathological conditions. PTKs are classified into two groups: one of which represents tyrosine kinases, while the other one includes the members of the serine/threonine kinases. The group of tyrosine kinases is subdivided into subgroups: one of them includes the member of receptor tyrosine kinases (RTKs), while the other subgroup includes the member of non-receptor tyrosine kinases (NRTKs). Both these kinase groups function as an "on" or "off" switch in many cellular functions. NRTKs are enzymes which are overexpressed and activated in many cancer types and regulate variable cellular functions in response to extracellular signaling-dependent mechanisms. NRTK-mediated different cellular functions are regulated by kinase-dependent and kinase-independent mechanisms either in the cytoplasm or in the nucleus. Thus, targeting NRTKs is of great interest to improve the treatment strategy of different tumor types. This review deals with the structure and mechanistic role of NRTKs in tumor progression and resistance and their importance as therapeutic targets in tumor therapy.
Collapse
Affiliation(s)
- Abdulaziz M. Eshaq
- Department of Epidemiology and Biostatistics, Milken Institute School of Public Health, George Washington University, Washington, DC 20052, USA;
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (S.A.A.A.); (W.A.A.-A.); (M.O.A.); (M.O.A.)
| | - Thomas W. Flanagan
- Department of Pharmacology and Experimental Therapeutics, LSU Health Sciences Center, New Orleans, LA 70112, USA;
| | - Sofie-Yasmin Hassan
- Department of Pharmacy, Faculty of Science, Heinrich-Heine University Duesseldorf, 40225 Duesseldorf, Germany;
| | - Sara A. Al Asheikh
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (S.A.A.A.); (W.A.A.-A.); (M.O.A.); (M.O.A.)
| | - Waleed A. Al-Amoudi
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (S.A.A.A.); (W.A.A.-A.); (M.O.A.); (M.O.A.)
| | - Simeon Santourlidis
- Institute of Cell Therapeutics and Diagnostics, University Medical Center of Duesseldorf, 40225 Duesseldorf, Germany;
| | - Sarah-Lilly Hassan
- Department of Chemistry, Faculty of Science, Heinrich-Heine University Duesseldorf, 40225 Duesseldorf, Germany;
| | - Maryam O. Alamodi
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (S.A.A.A.); (W.A.A.-A.); (M.O.A.); (M.O.A.)
| | - Marcelo L. Bendhack
- Department of Urology, Red Cross University Hospital, Positivo University, Rua Mauá 1111, Curitiba 80030-200, Brazil;
| | - Mohammed O. Alamodi
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (S.A.A.A.); (W.A.A.-A.); (M.O.A.); (M.O.A.)
| | - Youssef Haikel
- Institut National de la Santé et de la Recherche Médicale, University of Strasbourg, 67000 Strasbourg, France;
- Department of Operative Dentistry and Endodontics, Dental Faculty, 67000 Strasbourg, France
- Pôle de Médecine et Chirurgie Bucco-Dentaire, Hôpital Civil, Hôpitaux Universitaire de Strasbourg, 67000 Strasbourg, France
| | - Mossad Megahed
- Clinic of Dermatology, University Hospital of Aachen, 52074 Aachen, Germany;
| | - Mohamed Hassan
- Institut National de la Santé et de la Recherche Médicale, University of Strasbourg, 67000 Strasbourg, France;
- Department of Operative Dentistry and Endodontics, Dental Faculty, 67000 Strasbourg, France
- Research Laboratory of Surgery-Oncology, Department of Surgery, Tulane University School of Medicine, New Orleans, LA 70112, USA
| |
Collapse
|
9
|
Strandberg H, Hagströmer CJ, Werin B, Wendler M, Johanson U, Törnroth-Horsefield S. Structural Basis for the Interaction between the Ezrin FERM-Domain and Human Aquaporins. Int J Mol Sci 2024; 25:7672. [PMID: 39062914 PMCID: PMC11277499 DOI: 10.3390/ijms25147672] [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: 06/20/2024] [Revised: 07/08/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
The Ezrin/Radixin/Moesin (ERM) family of proteins act as cross-linkers between the plasma membrane and the actin cytoskeleton. This mechanism plays an essential role in processes related to membrane remodeling and organization, such as cell polarization, morphogenesis and adhesion, as well as in membrane protein trafficking and signaling pathways. For several human aquaporin (AQP) isoforms, an interaction between the ezrin band Four-point-one, Ezrin, Radixin, Moesin (FERM)-domain and the AQP C-terminus has been demonstrated, and this is believed to be important for AQP localization in the plasma membrane. Here, we investigate the structural basis for the interaction between ezrin and two human AQPs: AQP2 and AQP5. Using microscale thermophoresis, we show that full-length AQP2 and AQP5 as well as peptides corresponding to their C-termini interact with the ezrin FERM-domain with affinities in the low micromolar range. Modelling of the AQP2 and AQP5 FERM complexes using ColabFold reveals a common mode of binding in which the proximal and distal parts of the AQP C-termini bind simultaneously to distinct binding sites of FERM. While the interaction at each site closely resembles other FERM-complexes, the concurrent interaction with both sites has only been observed in the complex between moesin and its C-terminus which causes auto-inhibition. The proposed interaction between AQP2/AQP5 and FERM thus represents a novel binding mode for extrinsic ERM-interacting partners.
Collapse
Affiliation(s)
| | | | | | | | | | - Susanna Törnroth-Horsefield
- Department of Biochemistry and Structural Biology, Lund University, 221 00 Lund, Sweden; (H.S.); (C.J.H.); (B.W.); (M.W.); (U.J.)
| |
Collapse
|
10
|
Brown ND, Vomhof-DeKrey EE. Focal Adhesion Kinase and Colony Stimulating Factors: Intestinal Homeostasis and Innate Immunity Crosstalk. Cells 2024; 13:1178. [PMID: 39056760 PMCID: PMC11274384 DOI: 10.3390/cells13141178] [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: 04/30/2024] [Revised: 07/08/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Thousands struggle with acute and chronic intestinal injury due to various causes. Epithelial intestinal healing is dependent on phenotypic transitions to a mobile phenotype. Focal adhesion kinase (FAK) is a ubiquitous protein that is essential for cell mobility. This phenotype change is mediated by FAK activation and proves to be a promising target for pharmaceutical intervention. While FAK is crucial for intestinal healing, new evidence connects FAK with innate immunity and the importance it plays in macrophage/monocyte chemotaxis, as well as other intracellular signaling cascades. These cascades play a part in macrophage/monocyte polarization, maturation, and inflammation that is associated with intestinal injury. Colony stimulating factors (CSFs) such as macrophage colony stimulating factor (M-CSF/CSF-1) and granulocyte macrophage colony stimulating factor (GM-CSF/CSF-2) play a critical role in maintaining homeostasis within intestinal mucosa by crosstalk capabilities between macrophages and epithelial cells. The communication between these cells is imperative in orchestrating healing upon injury. Diving deeper into these connections may allow us a greater insight into the role that our immune system plays in healing, as well as a better comprehension of inflammatory diseases of the gut.
Collapse
Affiliation(s)
- Nicholas D. Brown
- Department of Pathology, School of Medicine and the Health Sciences, University of North Dakota, Grand Forks, ND 58203, USA;
| | - Emilie E. Vomhof-DeKrey
- Department of Pathology, School of Medicine and the Health Sciences, University of North Dakota, Grand Forks, ND 58203, USA;
- Department of Surgery, School of Medicine and the Health Sciences, University of North Dakota, Grand Forks, ND 58203, USA
- Department of Biomedical Sciences, School of Medicine and the Health Sciences, University of North Dakota, Grand Forks, ND 58203, USA
| |
Collapse
|
11
|
Lee HS, Ku B, Shin HC, Kim SJ. Structural analysis of the FERM domain of human protein tyrosine phosphatase non-receptor type 21. Acta Crystallogr F Struct Biol Commun 2024; 80:148-153. [PMID: 38940939 PMCID: PMC11229555 DOI: 10.1107/s2053230x24005260] [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: 02/27/2024] [Accepted: 06/04/2024] [Indexed: 06/29/2024] Open
Abstract
Protein tyrosine phosphatase non-receptor type 21 (PTPN21) is a cytosolic protein tyrosine phosphatase that regulates cell growth and invasion. Due to its oncogenic properties, PTPN21 has recently emerged as a potential therapeutic target for cancer. In this study, the three-dimensional structure of the PTPN21 FERM domain was determined at 2.1 Å resolution by X-ray crystallography. The crystal structure showed that this domain harbors canonical FERM folding and consists of three subdomains that are tightly packed via highly conserved intramolecular hydrophobic interactions. Consistent with this, the PTPN21 FERM domain shares high structural homology with several other FERM domains. Moreover, structural superimposition demonstrated two putative protein-binding sites of the PTPN21 FERM domain, which are presumed to be associated with interaction with its binding partner, kinesin family member 1C. Thus, these data suggest that the FERM domain of PTPN21 serves as a module that mediates protein-protein interaction, like other FERM domains.
Collapse
Affiliation(s)
- Hye Seon Lee
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| | - Bonsu Ku
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| | - Ho Cheol Shin
- Critical Disease Diagnostics Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| | - Seung Jun Kim
- Critical Disease Diagnostics Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| |
Collapse
|
12
|
Chen L, Qian Z, Zheng Y, Zhang J, Sun J, Zhou C, Xiao H. Structural analysis of PTPN21 reveals a dominant-negative effect of the FERM domain on its phosphatase activity. SCIENCE ADVANCES 2024; 10:eadi7404. [PMID: 38416831 PMCID: PMC10901363 DOI: 10.1126/sciadv.adi7404] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 01/24/2024] [Indexed: 03/01/2024]
Abstract
PTPN21 belongs to the four-point-one, ezrin, radixin, moesin (FERM) domain-containing protein tyrosine phosphatases (PTP) and plays important roles in cytoskeleton-associated cellular processes like cell adhesion, motility, and cargo transport. Because of the presence of a WPE loop instead of a WPD loop in the phosphatase domain, it is often considered to lack phosphatase activity. However, many of PTPN21's biological functions require its catalytic activity. To reconcile these findings, we have determined the structures of individual PTPN21 FERM, PTP domains, and a complex between FERM-PTP. Combined with biochemical analysis, we have found that PTPN21 PTP is weakly active and is autoinhibited by association with its FERM domain. Disruption of FERM-PTP interaction results in enhanced ERK activation. The oncogenic HPV18 E7 protein binds to PTP at the same location as PTPN21 FERM, indicating that it may act by displacing the FERM domain from PTP. Our results provide mechanistic insight into PTPN21 and benefit functional studies of PTPN21-mediated processes.
Collapse
Affiliation(s)
- Lu Chen
- Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China
- School of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Zijun Qian
- Department of Hematology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China
| | - Yuyuan Zheng
- School of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Jie Zhang
- Department of Hematology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China
| | - Jie Sun
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Chun Zhou
- Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China
- School of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Haowen Xiao
- Department of Hematology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China
| |
Collapse
|
13
|
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: 6] [Impact Index Per Article: 6.0] [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
|
14
|
Fatima F, Chourasiya NK, Mishra M, Kori S, Pathak S, Das R, Kashaw V, Iyer AK, Kashaw SK. Curcumin and its Derivatives Targeting Multiple Signaling Pathways to Elicit Anticancer Activity: A Comprehensive Perspective. Curr Med Chem 2024; 31:3668-3714. [PMID: 37221681 DOI: 10.2174/0929867330666230522144312] [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: 12/04/2022] [Revised: 03/23/2023] [Accepted: 04/07/2023] [Indexed: 05/25/2023]
Abstract
The uncontrolled growth and spread of aberrant cells characterize the group of disorders known as cancer. According to GLOBOCAN 2022 analysis of cancer patients in either developed countries or developing countries the main concern cancers are breast cancer, lung cancer, and liver cancer which may rise eventually. Natural substances with dietary origins have gained interest for their low toxicity, anti-inflammatory, and antioxidant effects. The evaluation of dietary natural products as chemopreventive and therapeutic agents, the identification, characterization, and synthesis of their active components, as well as the enhancement of their delivery and bioavailability, have all received significant attention. Thus, the treatment strategy for concerning cancers must be significantly evaluated and may include the use of phytochemicals in daily lifestyle. In the present perspective, we discussed one of the potent phytochemicals, that has been used over the past few decades known as curcumin as a panacea drug of the "Cure-all" therapy concept. In our review firstly we included exhausted data from in vivo and in vitro studies on breast cancer, lung cancer, and liver cancer which act through various cancer-targeting pathways at the molecular level. Now, the second is the active constituent of turmeric known as curcumin and its derivatives are enlisted with their targeted protein in the molecular docking studies, which help the researchers design and synthesize new curcumin derivatives with respective implicated molecular and cellular activity. However, curcumin and its substituted derivatives still need to be investigated with unknown targeting mechanism studies in depth.
Collapse
Affiliation(s)
- Firdous Fatima
- Integrated Drug Discovery Research Laboratory, Department of Pharmaceutical Sciences, Dr. Harisingh Gour University (A Central University), Sagar (MP), India
| | - Nikhil Kumar Chourasiya
- Integrated Drug Discovery Research Laboratory, Department of Pharmaceutical Sciences, Dr. Harisingh Gour University (A Central University), Sagar (MP), India
| | - Mitali Mishra
- Integrated Drug Discovery Research Laboratory, Department of Pharmaceutical Sciences, Dr. Harisingh Gour University (A Central University), Sagar (MP), India
| | - Shivam Kori
- Integrated Drug Discovery Research Laboratory, Department of Pharmaceutical Sciences, Dr. Harisingh Gour University (A Central University), Sagar (MP), India
| | - Sandhya Pathak
- Department of Chemistry, Dr. Harisingh Gour University (A Central University), Sagar (MP), India
| | - Ratnesh Das
- Department of Chemistry, Dr. Harisingh Gour University (A Central University), Sagar (MP), India
| | - Varsha Kashaw
- Sagar Institute of Pharmaceutical Sciences, Sagar (M.P.), India
| | - Arun K Iyer
- Use-inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Wayne State University, Detroit, Michigan, USA
- Molecular Imaging Program, Karmanos Cancer Institute, Detroit, Michigan, USA
| | - Sushil Kumar Kashaw
- Integrated Drug Discovery Research Laboratory, Department of Pharmaceutical Sciences, Dr. Harisingh Gour University (A Central University), Sagar (MP), India
| |
Collapse
|
15
|
Du Y, Bradshaw WJ, Leisner TM, Annor-Gyamfi JK, Qian K, Bashore FM, Sikdar A, Nwogbo FO, Ivanov AA, Frye SV, Gileadi O, Brennan PE, Levey AI, Axtman AD, Pearce KH, Fu H, Katis VL. Discovery of FERM domain protein-protein interaction inhibitors for MSN and CD44 as a potential therapeutic approach for Alzheimer's disease. J Biol Chem 2023; 299:105382. [PMID: 37866628 PMCID: PMC10692723 DOI: 10.1016/j.jbc.2023.105382] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 09/11/2023] [Accepted: 09/27/2023] [Indexed: 10/24/2023] Open
Abstract
Proteomic studies have identified moesin (MSN), a protein containing a four-point-one, ezrin, radixin, moesin (FERM) domain, and the receptor CD44 as hub proteins found within a coexpression module strongly linked to Alzheimer's disease (AD) traits and microglia. These proteins are more abundant in Alzheimer's patient brains, and their levels are positively correlated with cognitive decline, amyloid plaque deposition, and neurofibrillary tangle burden. The MSN FERM domain interacts with the phospholipid phosphatidylinositol 4,5-bisphosphate (PIP2) and the cytoplasmic tail of CD44. Inhibiting the MSN-CD44 interaction may help limit AD-associated neuronal damage. Here, we investigated the feasibility of developing inhibitors that target this protein-protein interaction. We have employed structural, mutational, and phage-display studies to examine how CD44 binds to the FERM domain of MSN. Interestingly, we have identified an allosteric site located close to the PIP2 binding pocket that influences CD44 binding. These findings suggest a mechanism in which PIP2 binding to the FERM domain stimulates CD44 binding through an allosteric effect, leading to the formation of a neighboring pocket capable of accommodating a receptor tail. Furthermore, high-throughput screening of a chemical library identified two compounds that disrupt the MSN-CD44 interaction. One compound series was further optimized for biochemical activity, specificity, and solubility. Our results suggest that the FERM domain holds potential as a drug development target. Small molecule preliminary leads generated from this study could serve as a foundation for additional medicinal chemistry efforts with the goal of controlling microglial activity in AD by modifying the MSN-CD44 interaction.
Collapse
Affiliation(s)
- Yuhong Du
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia, USA; Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - William J Bradshaw
- Alzheimer's Research UK Oxford Drug Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine Research Building, University of Oxford, Oxford, UK
| | - Tina M Leisner
- Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, Center for Integrative Chemical Biology and Drug Discovery, Chapel Hill, North Carolina, USA
| | - Joel K Annor-Gyamfi
- Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, Structural Genomics Consortium, Chapel Hill, North Carolina, USA
| | - Kun Qian
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia, USA; Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Frances M Bashore
- Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, Structural Genomics Consortium, Chapel Hill, North Carolina, USA
| | - Arunima Sikdar
- Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, Center for Integrative Chemical Biology and Drug Discovery, Chapel Hill, North Carolina, USA
| | - Felix O Nwogbo
- Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, Center for Integrative Chemical Biology and Drug Discovery, Chapel Hill, North Carolina, USA
| | - Andrey A Ivanov
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia, USA; Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Stephen V Frye
- Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, Center for Integrative Chemical Biology and Drug Discovery, Chapel Hill, North Carolina, USA
| | - Opher Gileadi
- Alzheimer's Research UK Oxford Drug Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine Research Building, University of Oxford, Oxford, UK
| | - Paul E Brennan
- Alzheimer's Research UK Oxford Drug Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine Research Building, University of Oxford, Oxford, UK
| | - Allan I Levey
- Department of Neurology, Emory Goizueta Alzheimer's Disease Research Center, Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Alison D Axtman
- Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, Structural Genomics Consortium, Chapel Hill, North Carolina, USA.
| | - Kenneth H Pearce
- Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, Center for Integrative Chemical Biology and Drug Discovery, Chapel Hill, North Carolina, USA.
| | - Haian Fu
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia, USA; Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, Georgia, USA.
| | - Vittorio L Katis
- Alzheimer's Research UK Oxford Drug Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine Research Building, University of Oxford, Oxford, UK.
| |
Collapse
|
16
|
de Jonckheere B, Kollotzek F, Münzer P, Göb V, Fischer M, Mott K, Coman C, Troppmair NN, Manke MC, Zdanyte M, Harm T, Sigle M, Kopczynski D, Bileck A, Gerner C, Hoffmann N, Heinzmann D, Assinger A, Gawaz M, Stegner D, Schulze H, Borst O, Ahrends R. Critical shifts in lipid metabolism promote megakaryocyte differentiation and proplatelet formation. NATURE CARDIOVASCULAR RESEARCH 2023; 2:835-852. [PMID: 38075556 PMCID: PMC7615361 DOI: 10.1038/s44161-023-00325-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 07/27/2023] [Indexed: 08/29/2024]
Abstract
During megakaryopoiesis, megakaryocytes (MK) undergo cellular morphological changes with strong modification of membrane composition and lipid signaling. Here we adopt a lipid-centric multiomics approach to create a quantitative map of the MK lipidome during maturation and proplatelet formation. Data reveal that MK differentiation is driven by an increased fatty acyl import and de novo lipid synthesis, resulting in an anionic membrane phenotype. Pharmacological perturbation of fatty acid import and phospholipid synthesis blocked membrane remodeling and directly reduced MK polyploidization and proplatelet formation resulting in thrombocytopenia. The anionic lipid shift during megakaryopoiesis was paralleled by lipid-dependent relocalization of the scaffold protein CKIP-1 and recruitment of the kinase CK2α to the plasma membrane, which seems to be essential for sufficient platelet biogenesis. Overall, this study provides a framework to understand how the MK lipidome is altered during maturation and the impact of MK membrane lipid remodeling on MK kinase signaling involved in thrombopoiesis.
Collapse
Affiliation(s)
- Bianca de Jonckheere
- Institute of Analytical Chemistry, University of Vienna, Austria
- Vienna Doctoral School in Chemistry, University of Vienna, Austria
| | - Ferdinand Kollotzek
- DFG Heisenberg Group Cardiovascular Thromboinflammation and Translational Thrombocardiology, University of Tübingen, Germany
- Department of Cardiology and Angiology, University of Tübingen, Germany
| | - Patrick Münzer
- DFG Heisenberg Group Cardiovascular Thromboinflammation and Translational Thrombocardiology, University of Tübingen, Germany
- Department of Cardiology and Angiology, University of Tübingen, Germany
| | - Vanessa Göb
- Institute for Experimental Biomedicine, University Hospital Würzburg, Germany
- Rudolf Virchow Center for Integrative and Translational Bioimaging, Würzburg, Germany
| | - Melina Fischer
- DFG Heisenberg Group Cardiovascular Thromboinflammation and Translational Thrombocardiology, University of Tübingen, Germany
- Department of Cardiology and Angiology, University of Tübingen, Germany
| | - Kristina Mott
- Institute for Experimental Biomedicine, University Hospital Würzburg, Germany
| | - Cristina Coman
- Institute of Analytical Chemistry, University of Vienna, Austria
| | - Nina Nicole Troppmair
- Institute of Analytical Chemistry, University of Vienna, Austria
- Vienna Doctoral School in Chemistry, University of Vienna, Austria
| | - Mailin-Christin Manke
- DFG Heisenberg Group Cardiovascular Thromboinflammation and Translational Thrombocardiology, University of Tübingen, Germany
- Department of Cardiology and Angiology, University of Tübingen, Germany
| | - Monika Zdanyte
- DFG Heisenberg Group Cardiovascular Thromboinflammation and Translational Thrombocardiology, University of Tübingen, Germany
- Department of Cardiology and Angiology, University of Tübingen, Germany
| | - Tobias Harm
- Department of Cardiology and Angiology, University of Tübingen, Germany
| | - Manuel Sigle
- Department of Cardiology and Angiology, University of Tübingen, Germany
| | | | - Andrea Bileck
- Institute of Analytical Chemistry, University of Vienna, Austria
- Joint Metabolome Facility, University of Vienna and Medical University of Vienna, Austria
| | - Christopher Gerner
- Institute of Analytical Chemistry, University of Vienna, Austria
- Joint Metabolome Facility, University of Vienna and Medical University of Vienna, Austria
| | - Nils Hoffmann
- Institute of Analytical Chemistry, University of Vienna, Austria
- Forschungszentrum Jülich GmbH, Institute for Bio- and Geosciences (IBG-5) Jülich, Germany
| | - David Heinzmann
- Department of Cardiology and Angiology, University of Tübingen, Germany
| | - Alice Assinger
- Institute of Physiology, Centre of Physiology and Pharmacology, Medical University of Vienna, Austria
| | - Meinrad Gawaz
- Department of Cardiology and Angiology, University of Tübingen, Germany
| | - David Stegner
- Institute for Experimental Biomedicine, University Hospital Würzburg, Germany
- Rudolf Virchow Center for Integrative and Translational Bioimaging, Würzburg, Germany
| | - Harald Schulze
- Institute for Experimental Biomedicine, University Hospital Würzburg, Germany
| | - Oliver Borst
- DFG Heisenberg Group Cardiovascular Thromboinflammation and Translational Thrombocardiology, University of Tübingen, Germany
- Department of Cardiology and Angiology, University of Tübingen, Germany
| | - Robert Ahrends
- Institute of Analytical Chemistry, University of Vienna, Austria
| |
Collapse
|
17
|
Buenaventura RGM, Merlino G, Yu Y. Ez-Metastasizing: The Crucial Roles of Ezrin in Metastasis. Cells 2023; 12:1620. [PMID: 37371090 DOI: 10.3390/cells12121620] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/08/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
Ezrin is the cytoskeletal organizer and functions in the modulation of membrane-cytoskeleton interaction, maintenance of cell shape and structure, and regulation of cell-cell adhesion and movement, as well as cell survival. Ezrin plays a critical role in regulating tumor metastasis through interaction with other binding proteins. Notably, Ezrin has been reported to interact with immune cells, allowing tumor cells to escape immune attack in metastasis. Here, we review the main functions of Ezrin, the mechanisms through which it acts, its role in tumor metastasis, and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Rand Gabriel M Buenaventura
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Glenn Merlino
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yanlin Yu
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
18
|
Lee KM, Seo EC, Lee JH, Kim HJ, Hwangbo C. The Multifunctional Protein Syntenin-1: Regulator of Exosome Biogenesis, Cellular Function, and Tumor Progression. Int J Mol Sci 2023; 24:ijms24119418. [PMID: 37298370 DOI: 10.3390/ijms24119418] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 05/20/2023] [Accepted: 05/22/2023] [Indexed: 06/12/2023] Open
Abstract
Syntenin acts as an adaptor and scaffold protein through its two PSD-95, Dlg, and ZO-1 (PDZ) domains, participating in multiple signaling pathways and modulating cellular physiology. It has been identified as an oncogene, promoting cancer development, metastasis, and angiogenesis in various carcinomas. Syntenin-1 is also associated with the production and release of exosomes, small extracellular vesicles that play a significant role in intercellular communication by containing bioactive molecules such as proteins, lipids, and nucleic acids. The trafficking of exosomes involves a complex interplay of various regulatory proteins, including syntenin-1, which interacts with its binding partners, syndecan and activated leukocyte cell adhesion molecule (ALIX). Exosomal transfer of microRNAs, a key cargo, can regulate the expression of various cancer-related genes, including syntenin-1. Targeting the mechanism involving the regulation of exosomes by syntenin-1 and microRNAs may provide a novel treatment strategy for cancer. This review highlights the current understanding of syntenin-1's role in regulating exosome trafficking and its associated cellular signaling pathways.
Collapse
Affiliation(s)
- Kwang-Min Lee
- Division of Life Science, College of Natural Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea
- Division of Applied Life Science (BK21 Four), Research Institute of Life Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Eun-Chan Seo
- Division of Life Science, College of Natural Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea
- Division of Applied Life Science (BK21 Four), Research Institute of Life Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Jeong-Hyung Lee
- Department of Biochemistry (BK21 Four), College of Natural Sciences, Kangwon National University, Chuncheon 24414, Republic of Korea
| | - Hyo-Jin Kim
- Division of Life Science, College of Natural Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea
- Division of Applied Life Science (BK21 Four), Research Institute of Life Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Cheol Hwangbo
- Division of Life Science, College of Natural Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea
- Division of Applied Life Science (BK21 Four), Research Institute of Life Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea
| |
Collapse
|
19
|
Ali M, Khramushin A, Yadav VK, Schueler-Furman O, Ivarsson Y. Elucidation of Short Linear Motif-Based Interactions of the FERM Domains of Ezrin, Radixin, Moesin, and Merlin. Biochemistry 2023. [PMID: 37224425 DOI: 10.1021/acs.biochem.3c00096] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
The ERM (ezrin, radixin, and moesin) family of proteins and the related protein merlin participate in scaffolding and signaling events at the cell cortex. The proteins share an N-terminal FERM [band four-point-one (4.1) ERM] domain composed of three subdomains (F1, F2, and F3) with binding sites for short linear peptide motifs. By screening the FERM domains of the ERMs and merlin against a phage library that displays peptides representing the intrinsically disordered regions of the human proteome, we identified a large number of novel ligands. We determined the affinities for the ERM and merlin FERM domains interacting with 18 peptides and validated interactions with full-length proteins through pull-down experiments. The majority of the peptides contained an apparent Yx[FILV] motif; others show alternative motifs. We defined distinct binding sites for two types of similar but distinct binding motifs (YxV and FYDF) using a combination of Rosetta FlexPepDock computational peptide docking protocols and mutational analysis. We provide a detailed molecular understanding of how the two types of peptides with distinct motifs bind to different sites on the moesin FERM phosphotyrosine binding-like subdomain and uncover interdependencies between the different types of ligands. The study expands the motif-based interactomes of the ERMs and merlin and suggests that the FERM domain acts as a switchable interaction hub.
Collapse
Affiliation(s)
- Muhammad Ali
- Department of Chemistry - BMC, Uppsala University, Husargatan 3, 751 23 Uppsala, Sweden
| | - Alisa Khramushin
- Department of Microbiology and Molecular Genetics, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| | - Vikash K Yadav
- Department of Chemistry - BMC, Uppsala University, Husargatan 3, 751 23 Uppsala, Sweden
| | - Ora Schueler-Furman
- Department of Microbiology and Molecular Genetics, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| | - Ylva Ivarsson
- Department of Chemistry - BMC, Uppsala University, Husargatan 3, 751 23 Uppsala, Sweden
| |
Collapse
|
20
|
Ghalavand MA, Asghari A, Farhadi M, Taghizadeh-Hesary F, Garshasbi M, Falah M. The genetic landscape and possible therapeutics of neurofibromatosis type 2. Cancer Cell Int 2023; 23:99. [PMID: 37217995 PMCID: PMC10204202 DOI: 10.1186/s12935-023-02940-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 05/07/2023] [Indexed: 05/24/2023] Open
Abstract
Neurofibromatosis type 2 (NF2) is a genetic condition marked by the development of multiple benign tumors in the nervous system. The most common tumors associated with NF2 are bilateral vestibular schwannoma, meningioma, and ependymoma. The clinical manifestations of NF2 depend on the site of involvement. Vestibular schwannoma can present with hearing loss, dizziness, and tinnitus, while spinal tumor leads to debilitating pain, muscle weakness, or paresthesias. Clinical diagnosis of NF2 is based on the Manchester criteria, which have been updated in the last decade. NF2 is caused by loss-of-function mutations in the NF2 gene on chromosome 22, leading the merlin protein to malfunction. Over half of NF2 patients have de novo mutations, and half of this group are mosaic. NF2 can be managed by surgery, stereotactic radiosurgery, monoclonal antibody bevacizumab, and close observation. However, the nature of multiple tumors and the necessity of multiple surgeries over the lifetime, inoperable tumors like meningiomatosis with infiltration of the sinus or in the area of the lower cranial nerves, the complications caused by the operation, the malignancies induced by radiotherapy, and inefficiency of cytotoxic chemotherapy due to the benign nature of NF-related tumors have led a march toward exploring targeted therapies. Recent advances in genetics and molecular biology have allowed identifying and targeting of underlying pathways in the pathogenesis of NF2. In this review, we explain the clinicopathological characteristics of NF2, its genetic and molecular background, and the current knowledge and challenges of implementing genetics to develop efficient therapies.
Collapse
Affiliation(s)
- Mohammad Amin Ghalavand
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Alimohamad Asghari
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Skull Base Research Center, The Five Senses Health Institute, Hazrat Rasoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Farhadi
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Farzad Taghizadeh-Hesary
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Radiation Oncology Department, Iran University of Medical Sciences, Tehran, Iran
| | - Masoud Garshasbi
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Masoumeh Falah
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
21
|
Du Y, Bradshaw WJ, Leisner TM, Annor-Gyamfi JK, Qian K, Bashore FM, Sikdar A, Nwogbo FO, Ivanov AA, Frye SV, Gileadi O, Brennan PE, Levey AI, Emory-Sage-SGC TREAT-AD Center, Axtman AD, Pearce KH, Fu H, Katis VL. Development of FERM domain protein-protein interaction inhibitors for MSN and CD44 as a potential therapeutic strategy for Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.22.541727. [PMID: 37292860 PMCID: PMC10245921 DOI: 10.1101/2023.05.22.541727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Recent genome-wide association studies have revealed genetic risk factors for Alzheimer's disease (AD) that are exclusively expressed in microglia within the brain. A proteomics approach identified moesin (MSN), a FERM (four-point-one ezrin radixin moesin) domain protein, and the receptor CD44 as hub proteins found within a co-expression module strongly linked to AD clinical and pathological traits as well as microglia. The FERM domain of MSN interacts with the phospholipid PIP2 and the cytoplasmic tails of receptors such as CD44. This study explored the feasibility of developing protein-protein interaction inhibitors that target the MSN-CD44 interaction. Structural and mutational analyses revealed that the FERM domain of MSN binds to CD44 by incorporating a beta strand within the F3 lobe. Phage-display studies identified an allosteric site located close to the PIP2 binding site in the FERM domain that affects CD44 binding within the F3 lobe. These findings support a model in which PIP2 binding to the FERM domain stimulates receptor tail binding through an allosteric mechanism that causes the F3 lobe to adopt an open conformation permissive for binding. High-throughput screening of a chemical library identified two compounds that disrupt the MSN-CD44 interaction, and one compound series was further optimized for biochemical activity, specificity, and solubility. The results suggest that the FERM domain holds potential as a drug development target. The small molecule preliminary leads generated from the study could serve as a foundation for additional medicinal chemistry effort with the goal of controlling microglial activity in AD by modifying the MSN-CD44 interaction.
Collapse
Affiliation(s)
- Yuhong Du
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA 30322, USA; Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - William J. Bradshaw
- Alzheimer’s Research UK Oxford Drug Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine Research Building, Old Road Campus, University of Oxford, Oxford, OX3 7FZ, UK
| | - Tina M. Leisner
- UNC Eshelman School of Pharmacy, Division of Chemical Biology and Medicinal Chemistry, Center for Integrative Chemical Biology and Drug Discovery, Chapel Hill, NC 27599, USA
| | - Joel K. Annor-Gyamfi
- UNC Eshelman School of Pharmacy, Division of Chemical Biology and Medicinal Chemistry, Structural Genomics Consortium, Chapel Hill, NC 27599, USA
| | - Kun Qian
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA 30322, USA; Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, GA 30322, USA
- Current address: Chemical Biology Consortium Sweden, Division of Chemical Biology and Genome Engineering, Department of Medical Biochemistry and Biophysics, Karolinska Institute, 171 65 Solna, Sweden
| | - Frances M. Bashore
- UNC Eshelman School of Pharmacy, Division of Chemical Biology and Medicinal Chemistry, Structural Genomics Consortium, Chapel Hill, NC 27599, USA
| | - Arunima Sikdar
- UNC Eshelman School of Pharmacy, Division of Chemical Biology and Medicinal Chemistry, Center for Integrative Chemical Biology and Drug Discovery, Chapel Hill, NC 27599, USA
| | - Felix O. Nwogbo
- UNC Eshelman School of Pharmacy, Division of Chemical Biology and Medicinal Chemistry, Center for Integrative Chemical Biology and Drug Discovery, Chapel Hill, NC 27599, USA
| | - Andrey A. Ivanov
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA 30322, USA; Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Stephen V. Frye
- UNC Eshelman School of Pharmacy, Division of Chemical Biology and Medicinal Chemistry, Center for Integrative Chemical Biology and Drug Discovery, Chapel Hill, NC 27599, USA
| | - Opher Gileadi
- Alzheimer’s Research UK Oxford Drug Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine Research Building, Old Road Campus, University of Oxford, Oxford, OX3 7FZ, UK
- Current address: Structural Genomics Consortium, Department of Medicine, Karolinska Hospital and Karolinska Institute, 171 76 Stockholm, Sweden
| | - Paul E. Brennan
- Alzheimer’s Research UK Oxford Drug Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine Research Building, Old Road Campus, University of Oxford, Oxford, OX3 7FZ, UK
| | - Allan I. Levey
- Department of Neurology, Emory Goizueta Alzheimer’s Disease Research Center, Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | - Alison D. Axtman
- UNC Eshelman School of Pharmacy, Division of Chemical Biology and Medicinal Chemistry, Structural Genomics Consortium, Chapel Hill, NC 27599, USA
| | - Kenneth H. Pearce
- UNC Eshelman School of Pharmacy, Division of Chemical Biology and Medicinal Chemistry, Center for Integrative Chemical Biology and Drug Discovery, Chapel Hill, NC 27599, USA
| | - Haian Fu
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA 30322, USA; Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Vittorio L. Katis
- Alzheimer’s Research UK Oxford Drug Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine Research Building, Old Road Campus, University of Oxford, Oxford, OX3 7FZ, UK
| |
Collapse
|
22
|
Ehret T, Heißenberg T, de Buhr S, Aponte-Santamaría C, Steinem C, Gräter F. FERM domains recruit ample PI(4,5)P 2s to form extensive protein-membrane attachments. Biophys J 2023; 122:1325-1333. [PMID: 36814382 PMCID: PMC10111351 DOI: 10.1016/j.bpj.2023.02.027] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 01/26/2023] [Accepted: 02/18/2023] [Indexed: 02/23/2023] Open
Abstract
The four-point-one ezrin-radixin-moesin homology (FERM) protein domain is a multifunctional protein-lipid binding site, constituting an integral part of numerous membrane-associated proteins. Its interaction with the lipid phosphatidylinositol-4,5-bisphosphate (PIP2), located at the inner leaflet of eukaryotic plasma membranes, is important for localization, anchorage, and activation of FERM-containing proteins. FERM-PIP2 complexes structurally determined so far exclusively feature a 1:1 binding stoichiometry of protein and lipid, with a few basic FERM residues neutralizing the -4 charge of the bound PIP2. Whether this picture from static crystal structures also applies to the dynamic interaction of FERM domains on PIP2 membranes is unknown. We here quantified the stoichiometry of FERM-PIP2 binding in a lipid bilayer using atomistic molecular dynamics simulations and experiments on solid supported membranes for the FERM domains of focal adhesion kinase and ezrin. In contrast to the structural data, we find much higher average stoichiometries of FERM-PIP2 binding, amounting to 1:3 or 1:4 ratios, respectively. In simulations, the full set of basic residues at the membrane interface, 7 and 15 residues for focal adhesion kinase and ezrin, respectively, engages in PIP2 interactions. In addition, Na ions enter the FERM-membrane binding interface, compensating negative PIP2 charges in case of high charge surpluses from bound PIP2. We propose the multivalent binding of FERM domains to PIP2 in lipid bilayers to significantly enhance the stability of FERM-membrane binding and to render the FERM-membrane linkage highly adjustable.
Collapse
Affiliation(s)
- Thomas Ehret
- Heidelberg Institute for Theoretical Studies (HITS), Heidelberg, Germany
| | - Tim Heißenberg
- Institute of Organic and Biomolecular Chemistry, University of Göttingen, Göttingen, Germany; Max Planck School Matter to Life, Heidelberg and Göttingen, Germany
| | - Svenja de Buhr
- Heidelberg Institute for Theoretical Studies (HITS), Heidelberg, Germany
| | | | - Claudia Steinem
- Institute of Organic and Biomolecular Chemistry, University of Göttingen, Göttingen, Germany; Max Planck School Matter to Life, Heidelberg and Göttingen, Germany.
| | - Frauke Gräter
- Heidelberg Institute for Theoretical Studies (HITS), Heidelberg, Germany; Max Planck School Matter to Life, Heidelberg and Göttingen, Germany; Interdisciplinary Center for Scientific Computing (IWR), Heidelberg University, Heidelberg, Germany.
| |
Collapse
|
23
|
Zhao S, Luo J, Hu J, Wang H, Zhao N, Cao M, Zhang C, Hu R, Liu L. Role of Ezrin in Asthma-Related Airway Inflammation and Remodeling. Mediators Inflamm 2022; 2022:6255012. [PMID: 36530558 PMCID: PMC9750775 DOI: 10.1155/2022/6255012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 11/22/2022] [Accepted: 11/22/2022] [Indexed: 08/13/2023] Open
Abstract
Ezrin is an actin binding protein connecting the cell membrane and the cytoskeleton, which is crucial to maintaining cell morphology, intercellular adhesion, and cytoskeleton remodeling. Asthma involves dysfunction of inflammatory cells, cytokines, and airway structural cells. Recent studies have shown that ezrin, whose function is affected by extensive phosphorylation and protein interactions, is closely associated with asthma, may be a therapeutic target for asthma treatment. In this review, we summarize studies on ezrin and discuss its role in asthma-related airway inflammation and remodeling.
Collapse
Affiliation(s)
- Shumei Zhao
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China
- Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Jiaqi Luo
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China
- Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Jun Hu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China
- Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Hesheng Wang
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China
- Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Ningwei Zhao
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China
- Shimadzu Biomedical Research Laboratory, Shanghai 200233, China
| | - Meng Cao
- Nanjing University of Chinese Medicine, Nanjing 210029, China
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine Nanjing University of Chinese Medicine, Nanjing 210028, China
| | - Cong Zhang
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China
- Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Rongkui Hu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China
- Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Lanying Liu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China
- Nanjing University of Chinese Medicine, Nanjing 210029, China
| |
Collapse
|
24
|
Xie T, Dong J, Zhou X, Tang D, Li D, Chen J, Chen Y, Xu H, Xue W, Liu D, Hong X, Tang F, Yin L, Dai Y. Proteomics analysis of lysine crotonylation and 2-hydroxyisobutyrylation reveals significant features of systemic lupus erythematosus. Clin Rheumatol 2022; 41:3851-3858. [PMID: 35941338 PMCID: PMC9652266 DOI: 10.1007/s10067-022-06254-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 05/14/2022] [Accepted: 06/15/2022] [Indexed: 11/28/2022]
Abstract
INTRODUCTION/OBJECTIVES To seek significant features of systemic lupus erythematosus (SLE) by utilizing bioinformatics analysis. METHOD Liquid chromatography-tandem mass spectrometry (LC-MS/MS) was used to quantify lysine crotonylation (Kcr) and lysine 2-hydroxyisobutyrylation (Khib) in peripheral blood mononuclear cells (PBMCs) of systemic lupus erythematosus (SLE) patients and normal controls. RESULTS Seventy-six differentially modified proteins (DMPs) dually modified by Kcr and Khib were identified between SLE patients and healthy people. GO enrichment analysis prompted significant enrichment of seventy-six DMPs in MHC class II protein complex binding and leukocyte migration. KEGG pathways were enriched in antigen processing and presentation pathway and leukocyte transendothelial migration pathway. Six DMPs (CLTC, HSPA1B, HSPA8, HSP90AB1, HSPD1, and PDIA3) were identified in antigen processing and presentation pathway, of which HSPA8 was the core protein. Significant changes of Kcr and Khib in HSPA8 may increase ATP hydrolysis and promote antigen binding to MHC II molecule. In leukocyte transendothelial migration pathway, 7 DMPs (ACTN1, ACTN4, EZR, MSN, RAC1, RHOA, and VCL) were identified. MSN was the protein with the most modification sites in this pathway. In amino terminal ferm region of MSN, Kcr and Khib expression change may lead to the adhesion between leukocytes and endothelial cells, which was an important step of leukocyte migration. CONCLUSION Kcr and Khib may promote the antigen presentation and jointly regulate the tissue damage mediated by leukocyte migration in SLE patients, which may play key roles in the pathogenesis of SLE probably. Key Points • Antigen processing and presentation and leukocyte transendothelial migration may play key roles in the pathogenesis of SLE.
Collapse
Affiliation(s)
- Ting Xie
- Institute of Nephrology and Blood Purification, the First Affiliated Hospital of Jinan University, Guangzhou, 510632, Guangdong, China
| | - Jingjing Dong
- Institute of Nephrology and Blood Purification, the First Affiliated Hospital of Jinan University, Guangzhou, 510632, Guangdong, China
- Clinical Medical Research Center, the Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, 518020, Guangdong, China
| | - Xianqing Zhou
- Department of Pathology, Guangxi Key Laboratory of Metabolic Diseases Research, No.924 Hospital of PLA Joint Logistic Support Force, Guilin, 541002, Guangxi, China
| | - Donge Tang
- Clinical Medical Research Center, the Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, 518020, Guangdong, China
| | - Dandan Li
- Institute of Nephrology and Blood Purification, the First Affiliated Hospital of Jinan University, Guangzhou, 510632, Guangdong, China
| | - Jiejing Chen
- Department of Pathology, Guangxi Key Laboratory of Metabolic Diseases Research, No.924 Hospital of PLA Joint Logistic Support Force, Guilin, 541002, Guangxi, China
| | - Yumei Chen
- Clinical Medical Research Center, the Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, 518020, Guangdong, China
| | - Huixuan Xu
- Clinical Medical Research Center, the Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, 518020, Guangdong, China
| | - Wen Xue
- Department of Pathology, Guangxi Key Laboratory of Metabolic Diseases Research, No.924 Hospital of PLA Joint Logistic Support Force, Guilin, 541002, Guangxi, China
| | - Dongzhou Liu
- Clinical Medical Research Center, the Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, 518020, Guangdong, China
| | - Xiaoping Hong
- Clinical Medical Research Center, the Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, 518020, Guangdong, China
| | - Fang Tang
- Department of Pathology, Guangxi Key Laboratory of Metabolic Diseases Research, No.924 Hospital of PLA Joint Logistic Support Force, Guilin, 541002, Guangxi, China.
| | - Lianghong Yin
- Institute of Nephrology and Blood Purification, the First Affiliated Hospital of Jinan University, Guangzhou, 510632, Guangdong, China.
| | - Yong Dai
- Clinical Medical Research Center, the Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, 518020, Guangdong, China.
| |
Collapse
|
25
|
Winkenbach LP, Parker DM, Williams RTP, Nishimura EO. The ERM-1 membrane-binding domain directs erm-1 mRNA localization to the plasma membrane in the C. elegans embryo. Development 2022; 149:279335. [PMID: 36314842 PMCID: PMC9793419 DOI: 10.1242/dev.200930] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 10/10/2022] [Indexed: 11/06/2022]
Abstract
mRNA localization and transport are integral in regulating gene expression. In Caenorhabditis elegans embryos, the maternally inherited mRNA erm-1 (Ezrin/Radixin/Moesin) becomes concentrated in anterior blastomeres. erm-1 mRNA localizes within those blastomeres to the plasma membrane where the essential ERM-1 protein, a membrane-actin linker, is also found. We demonstrate that the localization of erm-1 mRNA to the plasma membrane is translation dependent and requires its encoded N-terminal, membrane-binding (FERM) domain. By perturbing translation through multiple methods, we found that erm-1 mRNA localization at the plasma membrane persisted only if the nascent peptide remained in complex with the translating mRNA. Indeed, re-coding the erm-1 mRNA coding sequence while preserving the encoded amino acid sequence did not disrupt erm-1 mRNA localization, corroborating that the information directing mRNA localization resides within its membrane-binding protein domain. A single-molecule inexpensive fluorescence in situ hybridization screen of 17 genes encoding similar membrane-binding domains identified three plasma membrane-localized mRNAs in the early embryo. Ten additional transcripts showed potential membrane localization later in development. These findings point to a translation-dependent pathway for localization of mRNAs encoding membrane-associated proteins.
Collapse
Affiliation(s)
- Lindsay P. Winkenbach
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA
| | - Dylan M. Parker
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA,Department of Chemistry and Biochemistry, Howard Hughes Medical Institute, University of Colorado, Boulder, CO 80303, USA
| | - Robert T. P. Williams
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA
| | - Erin Osborne Nishimura
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA,Author for correspondence ()
| |
Collapse
|
26
|
Distel JS, Flores RMO, Bienvenu A, Aguilera MO, Bonazzi M, Berón W. Ezrin and CD44 participate in the internalization process of
Coxiella burnetii
into non‐phagocytic cells. Biol Cell 2022; 114:237-253. [DOI: 10.1111/boc.202100096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 07/11/2022] [Accepted: 07/15/2022] [Indexed: 11/30/2022]
Affiliation(s)
- Jesús S. Distel
- Instituto de Histología y Embriología Facultad de Ciencias Médicas Universidad Nacional de Cuyo ‐ CONICET Mendoza 5500 Argentina
| | - Rodolfo M. Ortiz Flores
- Instituto de Histología y Embriología Facultad de Ciencias Médicas Universidad Nacional de Cuyo ‐ CONICET Mendoza 5500 Argentina
- Cátedra de Microbiología Parasitología e Inmunología Facultad de Odontología Universidad Nacional de Cuyo Mendoza Argentina
| | - Arthur Bienvenu
- Institut de Recherche en Infectiologie de Montpellier (IRIM) UMR 9004 CNRS Université de Montpellier Montpellier France
| | - Milton O. Aguilera
- Instituto de Histología y Embriología Facultad de Ciencias Médicas Universidad Nacional de Cuyo ‐ CONICET Mendoza 5500 Argentina
- Cátedra de Microbiología Parasitología e Inmunología Facultad de Odontología Universidad Nacional de Cuyo Mendoza Argentina
| | - Matteo Bonazzi
- Institut de Recherche en Infectiologie de Montpellier (IRIM) UMR 9004 CNRS Université de Montpellier Montpellier France
| | - Walter Berón
- Instituto de Histología y Embriología Facultad de Ciencias Médicas Universidad Nacional de Cuyo ‐ CONICET Mendoza 5500 Argentina
| |
Collapse
|
27
|
Huang L, Zhou Y, Chen W, Lin P, Xie Y, He K, Zhang S, Wu Y, Li N. Correlations of FRMD7 gene mutations with ocular oscillations. Sci Rep 2022; 12:9914. [PMID: 35705619 PMCID: PMC9200781 DOI: 10.1038/s41598-022-14144-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 05/11/2022] [Indexed: 11/30/2022] Open
Abstract
Mutations in the FERM domain containing 7 (FRMD7) gene have been proven to be responsible for infantile nystagmus (IN). The purpose of this study is to investigate FRMD7 gene mutations in patients with IN, and to evaluate the nystagmus intensity among patients with and without FRMD7 mutations. The affected males were subdivided into three groups according to whether or not having FRMD7 mutations and the types of mutations. Fifty-two mutations were detected in FRMD7 in 56 pedigrees and 34 sporadic patients with IN, including 28 novel and 24 previous reported mutations. The novel identified mutations further expand the spectrum of FRMD7 mutations. The parameters of nystagmus intensity and the patients’ best corrected visual acuity were not statistically different among the patients with and without identified FRMD7 mutations, and also not different among patients with different mutant types. The FERM-C domain, whose amino acids are encoded by exons 7, 8 and 9, could be the harbor region for most mutations. Loss-of-function is suggested to be the common molecular mechanism for the X-linked infantile nystagmus.
Collapse
Affiliation(s)
- Lijuan Huang
- Department of Ophthalmology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China.,Department of Ophthalmology, Beijing Children's Hospital, Capital Medical University, No 56. Nan Li Shi Rd, Xicheng District, Beijing, 100045, China
| | - Yunyu Zhou
- Department of Ophthalmology, Beijing Children's Hospital, Capital Medical University, No 56. Nan Li Shi Rd, Xicheng District, Beijing, 100045, China
| | - Wencong Chen
- Department of Biostatistics, Vanderbilt University Medical Center, 2525 West End Avenue, Suite 1100, Nashville, TN, 37203, USA
| | - Ping Lin
- Department of Ophthalmology, Xi'an Children's Hospital, Xi'an, 710002, China
| | - Yan Xie
- Department of Ophthalmology, Beijing Children's Hospital, Capital Medical University, No 56. Nan Li Shi Rd, Xicheng District, Beijing, 100045, China
| | - Kaiwen He
- Department of Ophthalmology, Beijing Children's Hospital, Capital Medical University, No 56. Nan Li Shi Rd, Xicheng District, Beijing, 100045, China
| | - Shasha Zhang
- Department of Ophthalmology, Xi'an Children's Hospital, Xi'an, 710002, China
| | - Yuyu Wu
- Department of Ophthalmology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China.
| | - Ningdong Li
- Department of Ophthalmology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China. .,Department of Ophthalmology, Beijing Children's Hospital, Capital Medical University, No 56. Nan Li Shi Rd, Xicheng District, Beijing, 100045, China. .,Department of Ophthalmology, Xi'an Children's Hospital, Xi'an, 710002, China. .,Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, 100045, China. .,Department of Ophthalmology, Children's Hospital, Capital Institute of Pediatrics, Beijing, 100020, China.
| |
Collapse
|
28
|
Matching protein surface structural patches for high-resolution blind peptide docking. Proc Natl Acad Sci U S A 2022; 119:e2121153119. [PMID: 35482919 PMCID: PMC9170164 DOI: 10.1073/pnas.2121153119] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Modeling interactions between short peptides and their receptors is a challenging docking problem due to the peptide flexibility, resulting in a formidable sampling problem of peptide conformation in addition to its orientation. Alternatively, the peptide can be viewed as a piece that complements the receptor monomer structure. Here, we show that the peptide conformation can be determined based on the receptor backbone only and sampled using local structural motifs found in solved protein monomers and interfaces, independent of sequence similarity. This approach outperforms current peptide docking protocols and promotes new directions for peptide interface design. Peptide docking can be perceived as a subproblem of protein–protein docking. However, due to the short length and flexible nature of peptides, many do not adopt one defined conformation prior to binding. Therefore, to tackle a peptide docking problem, not only the relative orientation, but also the bound conformation of the peptide needs to be modeled. Traditional peptide-centered approaches use information about peptide sequences to generate representative conformer ensembles, which can then be rigid-body docked to the receptor. Alternatively, one may look at this problem from the viewpoint of the receptor, namely, that the protein surface defines the peptide-bound conformation. Here, we present PatchMAN (Patch-Motif AligNments), a global peptide-docking approach that uses structural motifs to map the receptor surface with backbone scaffolds extracted from protein structures. On a nonredundant set of protein–peptide complexes, starting from free receptor structures, PatchMAN successfully models and identifies near-native peptide–protein complexes in 58%/84% within 2.5 Å/5 Å interface backbone RMSD, with corresponding sampling in 81%/100% of the cases, outperforming other approaches. PatchMAN leverages the observation that structural units of peptides with their binding pocket can be found not only within interfaces, but also within monomers. We show that the bound peptide conformation is sampled based on the structural context of the receptor only, without taking into account any sequence information. Beyond peptide docking, this approach opens exciting new avenues to study principles of peptide–protein association, and to the design of new peptide binders. PatchMAN is available as a server at https://furmanlab.cs.huji.ac.il/patchman/.
Collapse
|
29
|
Song M, Suh P. O‐GlcNAcylation regulates lysophosphatidic acid‐induced cell migration by regulating ERM family proteins. FEBS Open Bio 2022; 12:1220-1229. [PMID: 35347892 PMCID: PMC9157403 DOI: 10.1002/2211-5463.13404] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 10/04/2021] [Accepted: 03/28/2022] [Indexed: 11/18/2022] Open
Abstract
O‐GlcNAcylation of intracellular proteins (O‐GlcNAc) is a post‐translational modification that often competes with phosphorylation in diverse cellular signaling pathways. Recent studies on human malignant tumors have demonstrated that O‐GlcNAc is implicated in cellular features relevant to metastasis. Here, we report that lysophosphatidic acid (LPA)‐induced ovarian cancer cell (OVCAR‐3) migration is regulated by O‐GlcNAc. We found that O‐GlcNAc modification of ERM family proteins, a membrane‐cytoskeletal crosslinker, was inversely correlated with its phosphorylation status. Moreover, the LPA‐induced formation of membrane protrusion structures, as well as the migration of OVCAR‐3 cells, was reduced by the accumulation of O‐GlcNAc. Collectively, these findings suggest that O‐GlcNAc is an essential signaling element controlling ERM family proteins involved in OVCAR‐3 cell migration.
Collapse
Affiliation(s)
- Minseok Song
- Department of Life Sciences Yeungnam University Gyeongsan Gyeongbuk 38541 South Korea
| | - Pann‐Ghill Suh
- Korea Basic Science Research Institute (KBRI) Daegu Republic of Korea
| |
Collapse
|
30
|
Sepers JJ, Ramalho JJ, Kroll JR, Schmidt R, Boxem M. ERM-1 Phosphorylation and NRFL-1 Redundantly Control Lumen Formation in the C. elegans Intestine. Front Cell Dev Biol 2022; 10:769862. [PMID: 35198555 PMCID: PMC8860247 DOI: 10.3389/fcell.2022.769862] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 01/04/2022] [Indexed: 11/13/2022] Open
Abstract
Reorganization of the plasma membrane and underlying actin cytoskeleton into specialized domains is essential for the functioning of most polarized cells in animals. Proteins of the ezrin-radixin-moesin (ERM) and Na+/H+ exchanger 3 regulating factor (NHERF) family are conserved regulators of cortical specialization. ERM proteins function as membrane-actin linkers and as molecular scaffolds that organize the distribution of proteins at the membrane. NHERF proteins are PDZ-domain containing adapters that can bind to ERM proteins and extend their scaffolding capability. Here, we investigate how ERM and NHERF proteins function in regulating intestinal lumen formation in the nematode Caenorhabditis elegans. C. elegans has single ERM and NHERF family proteins, termed ERM-1 and NRFL-1, and ERM-1 was previously shown to be critical for intestinal lumen formation. Using CRISPR/Cas9-generated nrfl-1 alleles we demonstrate that NRFL-1 localizes at the intestinal microvilli, and that this localization is depended on an interaction with ERM-1. However, nrfl-1 loss of function mutants are viable and do not show defects in intestinal development. Interestingly, combining nrfl-1 loss with erm-1 mutants that either block or mimic phosphorylation of a regulatory C-terminal threonine causes severe defects in intestinal lumen formation. These defects are not observed in the phosphorylation mutants alone, and resemble the effects of strong erm-1 loss of function. The loss of NRFL-1 did not affect the localization or activity of ERM-1. Together, these data indicate that ERM-1 and NRFL-1 function together in intestinal lumen formation in C. elegans. We postulate that the functioning of ERM-1 in this tissue involves actin-binding activities that are regulated by the C-terminal threonine residue and the organization of apical domain composition through NRFL-1.
Collapse
Affiliation(s)
- Jorian J Sepers
- Division of Developmental Biology, Department of Biology, Faculty of Science, Institute of Biodynamics and Biocomplexity, Utrecht University, Utrecht, Netherlands
| | - João J Ramalho
- Division of Developmental Biology, Department of Biology, Faculty of Science, Institute of Biodynamics and Biocomplexity, Utrecht University, Utrecht, Netherlands.,Laboratory of Biochemistry, Wageningen University and Research, Wageningen, Netherlands
| | - Jason R Kroll
- Division of Developmental Biology, Department of Biology, Faculty of Science, Institute of Biodynamics and Biocomplexity, Utrecht University, Utrecht, Netherlands
| | - Ruben Schmidt
- Division of Developmental Biology, Department of Biology, Faculty of Science, Institute of Biodynamics and Biocomplexity, Utrecht University, Utrecht, Netherlands
| | - Mike Boxem
- Division of Developmental Biology, Department of Biology, Faculty of Science, Institute of Biodynamics and Biocomplexity, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
31
|
Transient exposure of a buried phosphorylation site in an autoinhibited protein. Biophys J 2022; 121:91-101. [PMID: 34864046 PMCID: PMC8758417 DOI: 10.1016/j.bpj.2021.11.2890] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 10/25/2021] [Accepted: 11/29/2021] [Indexed: 01/07/2023] Open
Abstract
Autoinhibition is a mechanism used to regulate protein function, often by making functional sites inaccessible through the interaction with a cis-acting inhibitory domain. Such autoinhibitory domains often display a substantial degree of structural disorder when unbound, and only become structured in the inhibited state. These conformational dynamics make it difficult to study the structural origin of regulation, including effects of regulatory post-translational modifications. Here, we study the autoinhibition of the Dbl Homology domain in the protein Vav1 by the so-called acidic inhibitory domain. We use molecular simulations to study the process by which a mostly unstructured inhibitory domain folds upon binding and how transient exposure of a key buried tyrosine residue makes it accessible for phosphorylation. We show that the inhibitory domain, which forms a helix in the bound and inhibited stated, samples helical structures already before binding and that binding occurs via a molten-globule-like intermediate state. Together, our results shed light on key interactions that enable the inhibitory domain to sample a finely tuned equilibrium between an inhibited and a kinase-accessible state.
Collapse
|
32
|
Swamy H, Glading AJ. Contribution of protein-protein interactions to the endothelial barrier-stabilizing function of KRIT1. J Cell Sci 2021; 135:274104. [PMID: 34918736 PMCID: PMC8917353 DOI: 10.1242/jcs.258816] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 12/08/2021] [Indexed: 11/20/2022] Open
Abstract
Krev-interaction trapped 1 (KRIT1) is an endothelial scaffold protein that promotes adherens junction (AJ) stability. The precise mechanism by which KRIT1 promotes barrier stabilization is unclear. We tested the ability of a panel of KRIT1 constructs containing mutations that inhibit Rap1 binding, ICAP1 binding, disrupt KRIT1's protein tyrosine binding domain (PTB), or direct KRIT1 to the plasma membrane, either alone or in combination, to restore barrier function in KRIT1-deficient endothelial cells. We found that ablating the 192NPAY195 motif or disrupting the PTB domain was sufficient to restore AJ protein localization and barrier function to control levels, irrespective of the junctional localization of KRIT1 or Rap1 binding. The ability of our KRIT1 constructs to rescue AJ/barrier function in KRIT1 depleted endothelial cells correlated with decreased 1 integrin activity and maintenance of cortical actin fibers. Together, our findings indicate that Rap1 binding, ICAP1 binding, and junctional localization are not required for the ability of KRIT1 to stabilize endothelial contacts, and suggest that the ability of KRIT1 to limit integrin activity may be involved in barrier stabilization.
Collapse
Affiliation(s)
- Harsha Swamy
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY, USA
| | - Angela J Glading
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY, USA
| |
Collapse
|
33
|
Gómez-Henao W, Saavedra R, Chávez-Sánchez FR, Lascurain R, Zenteno E, Tenorio EP. Expression Dynamics of the O-Glycosylated Proteins Recognized by Amaranthus leucocarpus Lectin in T Lymphocytes and Its Relationship With Moesin as an Alternative Mechanism of Cell Activation. Front Immunol 2021; 12:788880. [PMID: 34917095 PMCID: PMC8669815 DOI: 10.3389/fimmu.2021.788880] [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: 10/03/2021] [Accepted: 11/11/2021] [Indexed: 11/13/2022] Open
Abstract
T lymphocyte activation begins with antigen/MHC recognition by the TCR/CD3 complex followed by a costimulatory signal provided by CD28. The search for novel costimulatory molecules has been extensive due to their potential use as immunotherapeutic targets. Although some molecules have been identified, they are unable to provide sustainable signaling to allow for proper T cell activation and proliferation. It has been shown that the Amaranthus leucocarpus lectin (ALL) can be used as an in vitro costimulator of CD4+ lymphocytes in the presence of anti-CD3 mAb; this lectin specifically recognizes O-glycans of the Galβ1-3GalNAc-O-Ser/Thr type, including a 70-kDa moesin-like protein that has been suggested as the costimulatory molecule. However, the identity of this molecule has not been confirmed and such costimulation has not been analyzed in CD8+ lymphocytes. We show herein that the expression kinetics of the glycoproteins recognized by ALL (gpALL) is different in CD4+ and CD8+ T cells, unlike moesin expression. Results from IP experiments demonstrate that the previously described 70-kDa moesin-like protein is an O-glycosylated form of moesin (O-moesin) and that in vitro stimulation with anti-CD3 and anti-moesin mAb induces expression of the activation molecules CD69 and CD25, proliferation and IL-2 production as efficiently as cells costimulated with ALL or anti-CD28. Overall, our results demonstrate that O-moesin is expressed in CD4+ and CD8+ T lymphocytes and that moesin provides a new costimulatory activation signal in both T cell subsets.
Collapse
Affiliation(s)
- Wilton Gómez-Henao
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Rafael Saavedra
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | | | - Ricardo Lascurain
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Edgar Zenteno
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Eda Patricia Tenorio
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
34
|
Hu X, Liu Y, Bing Z, Ye Q, Li C. High Moesin Expression Is a Predictor of Poor Prognosis of Breast Cancer: Evidence From a Systematic Review With Meta-Analysis. Front Oncol 2021; 11:650488. [PMID: 34900662 PMCID: PMC8660674 DOI: 10.3389/fonc.2021.650488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 11/01/2021] [Indexed: 11/13/2022] Open
Abstract
Owing to metastases and drug resistance, the prognosis of breast cancer is still dismal. Therefore, it is necessary to find new prognostic markers to improve the efficacy of breast cancer treatment. Literature shows a controversy between moesin (MSN) expression and prognosis in breast cancer. Here, we aimed to conduct a systematic review and meta-analysis to evaluate the prognostic relationship between MSN and breast cancer. Literature retrieval was conducted in the following databases: PubMed, Web of Science, Embase, and Cochrane. Two reviewers independently performed the screening of studies and data extraction. The Gene Expression Omnibus (GEO) database including both breast cancer gene expression and follow-up datasets was selected to verify literature results. The R software was employed for the meta-analysis. A total of 9 articles with 3,039 patients and 16 datasets with 2,916 patients were ultimately included. Results indicated that there was a significant relationship between MSN and lymph node metastases (P < 0.05), and high MSN expression was associated with poor outcome of breast cancer patients (HR = 1.99; 95% CI 1.73-2.24). In summary, there is available evidence to support that high MSN expression has valuable importance for the poor prognosis in breast cancer patients. SYSTEMATIC REVIEW REGISTRATION https://inplasy.com/inplasy-2020-8-0039/.
Collapse
Affiliation(s)
- Xiaoli Hu
- Department of Medical Physics, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Heavy Ion Radiation Medicine of Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Heavy Ion Radiation Medicine of Gansu Province, Lanzhou, China
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou, China
| | - Yang Liu
- Department of Medical Physics, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Heavy Ion Radiation Medicine of Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Heavy Ion Radiation Medicine of Gansu Province, Lanzhou, China
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou, China
| | - Zhitong Bing
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou, China
| | - Qian Ye
- Department of Medical Physics, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Heavy Ion Radiation Medicine of Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Heavy Ion Radiation Medicine of Gansu Province, Lanzhou, China
- School of Stomatology, Lanzhou University, Lanzhou, China
| | - Chengcheng Li
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| |
Collapse
|
35
|
Chen D, Yu W, Aitken L, Gunn-Moore F. Willin/FRMD6: A Multi-Functional Neuronal Protein Associated with Alzheimer's Disease. Cells 2021; 10:cells10113024. [PMID: 34831245 PMCID: PMC8616527 DOI: 10.3390/cells10113024] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/24/2021] [Accepted: 10/25/2021] [Indexed: 12/18/2022] Open
Abstract
The FERM domain-containing protein 6 (FRMD6), also known as Willin, is an upstream regulator of Hippo signaling that has recently been shown to modulate actin cytoskeleton dynamics and mechanical phenotype of neuronal cells through ERK signaling. Physiological functions of Willin/FRMD6 in the nervous system include neuronal differentiation, myelination, nerve injury repair, and vesicle exocytosis. The newly established neuronal role of Willin/FRMD6 is of particular interest given the mounting evidence suggesting a role for Willin/FRMD6 in Alzheimer's disease (AD), including a series of genome wide association studies that position Willin/FRMD6 as a novel AD risk gene. Here we describe recent findings regarding the role of Willin/FRMD6 in the nervous system and its actions in cellular perturbations related to the pathogenesis of AD.
Collapse
|
36
|
Inoue H, Takatsu H, Hamamoto A, Takayama M, Nakabuchi R, Muranaka Y, Yagi T, Nakayama K, Shin HW. The interaction of ATP11C-b with ezrin contributes to its polarized localization. J Cell Sci 2021; 134:272204. [PMID: 34528675 DOI: 10.1242/jcs.258523] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 09/09/2021] [Indexed: 02/04/2023] Open
Abstract
ATP11C, a member of the P4-ATPase family, translocates phosphatidylserine and phosphatidylethanolamine at the plasma membrane. We previously revealed that its C-terminal splice variant ATP11C-b exhibits polarized localization in motile cell lines, such as MDA-MB-231 and Ba/F3. In the present study, we found that the C-terminal cytoplasmic region of ATP11C-b interacts specifically with ezrin. Notably, the LLxY motif in the ATP11C-b C-terminal region is crucial for its interaction with ezrin as well as its polarized localization on the plasma membrane. A constitutively active, C-terminal phosphomimetic mutant of ezrin was colocalized with ATP11C-b in polarized motile cells. ATP11C-b was partially mislocalized in cells depleted of ezrin alone, and exhibited greater mislocalization in cells simultaneously depleted of the family members ezrin, radixin and moesin (ERM), suggesting that ERM proteins, particularly ezrin, contribute to the polarized localization of ATP11C-b. Furthermore, Atp11c knockout resulted in C-terminally phosphorylated ERM protein mislocalization, which was restored by exogenous expression of ATP11C-b but not ATP11C-a. These observations together indicate that the polarized localizations of ATP11C-b and the active form of ezrin to the plasma membrane are interdependently stabilized.
Collapse
Affiliation(s)
- Hiroki Inoue
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Hiroyuki Takatsu
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Asuka Hamamoto
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Masahiro Takayama
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Riki Nakabuchi
- Faculty of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Yumeka Muranaka
- Faculty of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Tsukasa Yagi
- Faculty of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Kazuhisa Nakayama
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Hye-Won Shin
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| |
Collapse
|
37
|
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: 10] [Impact Index Per Article: 2.5] [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
|
38
|
Padarti A, Abou-Fadel J, Zhang J. Resurgence of phosphotyrosine binding domains: Structural and functional properties essential for understanding disease pathogenesis. Biochim Biophys Acta Gen Subj 2021; 1865:129977. [PMID: 34391832 DOI: 10.1016/j.bbagen.2021.129977] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 07/30/2021] [Accepted: 07/30/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND Phosphotyrosine Binding (PTB) Domains, usually found on scaffold proteins, are pervasive in many cellular signaling pathways. These domains are the second-largest family of phosphotyrosine recognition domains and since their initial discovery, dozens of PTB domains have been structurally determined. SCOPE OF REVIEW Due to its signature sequence flexibility, PTB domains can bind to a large variety of ligands including phospholipids. PTB peptide binding is divided into classical binding (canonical NPXY motifs) and non-classical binding (all other motifs). The first atypical PTB domain was discovered in cerebral cavernous malformation 2 (CCM2) protein, while only one third in size of the typical PTB domain, it remains functionally equivalent. MAJOR CONCLUSIONS PTB domains are involved in numerous signaling processes including embryogenesis, neurogenesis, and angiogenesis, while dysfunction is linked to major disorders including diabetes, hypercholesterolemia, Alzheimer's disease, and strokes. PTB domains may also be essential in infectious processes, currently responsible for the global pandemic in which viral cellular entry is suspected to be mediated through PTB and NPXY interactions. GENERAL SIGNIFICANCE We summarize the structural and functional updates in the PTB domain over the last 20 years in hopes of resurging interest and further analyzing the importance of this versatile domain.
Collapse
Affiliation(s)
- Akhil Padarti
- Department of Molecular and Translational Medicine (MTM), Texas Tech University Health Science Center El Paso, 5001 El Paso Drive, El Paso, TX 79905, USA
| | - Johnathan Abou-Fadel
- Department of Molecular and Translational Medicine (MTM), Texas Tech University Health Science Center El Paso, 5001 El Paso Drive, El Paso, TX 79905, USA
| | - Jun Zhang
- Department of Molecular and Translational Medicine (MTM), Texas Tech University Health Science Center El Paso, 5001 El Paso Drive, El Paso, TX 79905, USA.
| |
Collapse
|
39
|
Primi MC, Rangarajan ES, Patil DN, Izard T. Conformational flexibility determines the Nf2/merlin tumor suppressor functions. Matrix Biol Plus 2021; 12:100074. [PMID: 34337379 PMCID: PMC8318988 DOI: 10.1016/j.mbplus.2021.100074] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 06/14/2021] [Accepted: 06/15/2021] [Indexed: 10/26/2022] Open
Abstract
The Neurofibromatosis type 2 gene encodes the Nf2/merlin tumor suppressor protein that is responsible for the regulation of cell proliferation. Once activated, Nf2/merlin modulates adhesive signaling pathways and thereby inhibits cell growth. Nf2/merlin controls oncogenic gene expression by modulating the Hippo pathway. By responding to several physical and biochemical stimuli, Hippo signaling determines contact inhibition of proliferation as well as organ size. The large tumor suppressor (LATS) serine/threonine-protein kinase is the key enzyme in the highly conserved kinase cascade that negatively regulates the activity and localization of the transcriptional coactivators Yes-associated protein (YAP) and its paralogue transcriptional coactivator with PDZ-binding motif (TAZ). Nf2/merlin belongs to the band 4.1, ezrin, radixin, moesin (FERM) gene family that links the actin cytoskeleton to adherens junctions, remodels adherens junctions during epithelial morphogenesis and maintains organized apical surfaces on the plasma cell membrane. Nf2/merlin and ERM proteins have a globular N-terminal cloverleaf head domain, the FERM domain, that binds to the plasma membrane, a central α-helical domain, and a tail domain that binds to its head domain. Here we present the high-resolution crystal structure of Nf2/merlin bound to LATS1 which shows that LATS1 binding to Nf2/merlin displaces the Nf2/merlin tail domain and causes an allosteric shift in the Nf2/merlin α-helix that extends from its FERM domain. This is consistent with the fact that full-length Nf2/merlin binds LATS1 ~10-fold weaker compared to LATS1 binding to the Nf2/merlin-PIP2 complex. Our data increase our understanding of Nf2/merlin biology by providing mechanistic insights into the Hippo pathway that are relevant to several diseases in particular oncogenic features that are associated with cancers.
Collapse
Affiliation(s)
- Marina C Primi
- Cell Adhesion Laboratory, Department of Integrative Structural and Computational Biology, The Scripps Research Institute, Jupiter 33458, FL, United States
| | - Erumbi S Rangarajan
- Cell Adhesion Laboratory, Department of Integrative Structural and Computational Biology, The Scripps Research Institute, Jupiter 33458, FL, United States
| | - Dipak N Patil
- Cell Adhesion Laboratory, Department of Integrative Structural and Computational Biology, The Scripps Research Institute, Jupiter 33458, FL, United States
| | - Tina Izard
- Cell Adhesion Laboratory, Department of Integrative Structural and Computational Biology, The Scripps Research Institute, Jupiter 33458, FL, United States
| |
Collapse
|
40
|
Carmi G, Gorohovski A, Frenkel-Morgenstern M. EvoProDom: Evolutionary modeling of protein families by assessing translocations of protein domains. FEBS Open Bio 2021; 11:2507-2524. [PMID: 34196123 PMCID: PMC8409312 DOI: 10.1002/2211-5463.13245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 06/22/2021] [Accepted: 06/30/2021] [Indexed: 11/29/2022] Open
Abstract
Here, we introduce a novel ‘evolution of protein domains’ (EvoProDom) model for describing the evolution of proteins based on the ‘mix and merge’ of protein domains. We assembled and integrated genomic and proteomic data comprising protein domain content and orthologous proteins from 109 organisms. In EvoProDom, we characterized evolutionary events, particularly, translocations, as reciprocal exchanges of protein domains between orthologous proteins in different organisms. We showed that protein domains that translocate with highly frequency are generated by transcripts enriched in trans‐splicing events, that is, the generation of novel transcripts from the fusion of two distinct genes. In EvoProDom, we describe a general method to collate orthologous protein annotation from KEGG, and protein domain content from protein sequences using tools such as KoFamKOAL and Pfam. To summarize, EvoProDom presents a novel model for protein evolution based on the ‘mix and merge’ of protein domains rather than DNA‐based evolution models. This confers the advantage of considering chromosomal alterations as drivers of protein evolutionary events.
Collapse
Affiliation(s)
- Gon Carmi
- Cancer Genomics and BioComputing of Complex Diseases Lab, The Azrieli Faculty of Medicine, Bar-Ilan University, 8 Henrietta Szold St, Safed, 13195, Israel
| | - Alessandro Gorohovski
- Cancer Genomics and BioComputing of Complex Diseases Lab, The Azrieli Faculty of Medicine, Bar-Ilan University, 8 Henrietta Szold St, Safed, 13195, Israel
| | - Milana Frenkel-Morgenstern
- Cancer Genomics and BioComputing of Complex Diseases Lab, The Azrieli Faculty of Medicine, Bar-Ilan University, 8 Henrietta Szold St, Safed, 13195, Israel
| |
Collapse
|
41
|
Bu W, Levitskaya Z, Tan SM, Gao YG. Emerging evidence for kindlin oligomerization and its role in regulating kindlin function. J Cell Sci 2021; 134:256567. [PMID: 33912917 DOI: 10.1242/jcs.256115] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Integrin-mediated cell-extracellular matrix (ECM) interactions play crucial roles in a broad range of physiological and pathological processes. Kindlins are important positive regulators of integrin activation. The FERM-domain-containing kindlin family comprises three members, kindlin-1, kindlin-2 and kindlin-3 (also known as FERMT1, FERMT2 and FERMT3), which share high sequence similarity (identity >50%), as well as domain organization, but exhibit diverse tissue-specific expression patterns and cellular functions. Given the significance of kindlins, analysis of their atomic structures has been an attractive field for decades. Recently, the structures of kindlin and its β-integrin-bound form have been obtained, which greatly advance our understanding of the molecular functions that involve kindlins. In particular, emerging evidence indicates that oligomerization of kindlins might affect their integrin binding and focal adhesion localization, positively or negatively. In this Review, we presented an update on the recent progress of obtaining kindlin structures, and discuss the implication for integrin activation based on kindlin oligomerization, as well as the possible regulation of this process.
Collapse
Affiliation(s)
- Wenting Bu
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore637551.,Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China518055
| | - Zarina Levitskaya
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore637551
| | - Suet-Mien Tan
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore637551
| | - Yong-Gui Gao
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore637551.,NTU Institute of Structural Biology, Nanyang Technological University, 59 Nanyang Drive, Singapore639798
| |
Collapse
|
42
|
Leguay K, Decelle B, He YY, Pagniez A, Hogue M, Kobayashi H, Le Gouill C, Bouvier M, Carréno S. Development of conformational BRET biosensors that monitor ezrin, radixin and moesin activation in real time. J Cell Sci 2021; 134:237806. [PMID: 33712451 DOI: 10.1242/jcs.255307] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 03/01/2021] [Indexed: 11/20/2022] Open
Abstract
Ezrin, radixin and moesin compose the family of ERM proteins. They link actin filaments and microtubules to the plasma membrane to control signaling and cell morphogenesis. Importantly, their activity promotes invasive properties of metastatic cells from different cancer origins. Therefore, a precise understanding of how these proteins are regulated is important for the understanding of the mechanism controlling cell shape, as well as providing new opportunities for the development of innovative cancer therapies. Here, we developed and characterized novel bioluminescence resonance energy transfer (BRET)-based conformational biosensors, compatible with high-throughput screening, that monitor individual ezrin, radixin or moesin activation in living cells. We showed that these biosensors faithfully monitor ERM activation and can be used to quantify the impact of small molecules, mutation of regulatory amino acids or depletion of upstream regulators on their activity. The use of these biosensors allowed us to characterize the activation process of ERMs that involves a pool of closed-inactive ERMs stably associated with the plasma membrane. Upon stimulation, we discovered that this pool serves as a cortical reserve that is rapidly activated before the recruitment of cytoplasmic ERMs.
Collapse
Affiliation(s)
- Kévin Leguay
- Cellular Mechanisms of Morphogenesis during Mitosis and Cell Motility lab, Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, P.O. Box 6128, Station Centre-Ville, Montréal, QC H3C 3J7, Canada
| | - Barbara Decelle
- Cellular Mechanisms of Morphogenesis during Mitosis and Cell Motility lab, Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, P.O. Box 6128, Station Centre-Ville, Montréal, QC H3C 3J7, Canada
| | - Yu Yan He
- Cellular Mechanisms of Morphogenesis during Mitosis and Cell Motility lab, Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, P.O. Box 6128, Station Centre-Ville, Montréal, QC H3C 3J7, Canada.,Molecular pharmacology lab, Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, P.O. Box 6128, Station Centre-Ville, Montréal, QC H3C 3J7, Canada
| | - Anthony Pagniez
- Cellular Mechanisms of Morphogenesis during Mitosis and Cell Motility lab, Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, P.O. Box 6128, Station Centre-Ville, Montréal, QC H3C 3J7, Canada
| | - Mireille Hogue
- Molecular pharmacology lab, Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, P.O. Box 6128, Station Centre-Ville, Montréal, QC H3C 3J7, Canada
| | - Hiroyuki Kobayashi
- Molecular pharmacology lab, Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, P.O. Box 6128, Station Centre-Ville, Montréal, QC H3C 3J7, Canada
| | - Christian Le Gouill
- Molecular pharmacology lab, Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, P.O. Box 6128, Station Centre-Ville, Montréal, QC H3C 3J7, Canada
| | - Michel Bouvier
- Molecular pharmacology lab, Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, P.O. Box 6128, Station Centre-Ville, Montréal, QC H3C 3J7, Canada.,Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, Québec H3C 3J7, Canada
| | - Sébastien Carréno
- Cellular Mechanisms of Morphogenesis during Mitosis and Cell Motility lab, Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, P.O. Box 6128, Station Centre-Ville, Montréal, QC H3C 3J7, Canada.,Department of Pathology and Cell Biology, Université de Montréal, Montréal, Québec H3C 3J7, Canada
| |
Collapse
|
43
|
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: 5.0] [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
|
44
|
Zhang F, Liu B, Gao Y, Long J, Zhou H. The crystal structure of the FERM and C-terminal domain complex of Drosophila Merlin. Biochem Biophys Res Commun 2021; 553:92-98. [PMID: 33765559 DOI: 10.1016/j.bbrc.2021.03.065] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 03/12/2021] [Indexed: 12/25/2022]
Abstract
NF2/Merlin is an upstream regulator of hippo pathway, and it has two states: an auto-inhibited "closed" state and an active "open" form. Previous studies showed that Drosophila Merlin adopts a more closed conformation. However, the molecular mechanism of conformational regulation remains poorly understood. Here, we first confirmed the strong interaction between FERM and the C-terminal domain (CTD) of Merlin, and then determined the crystal structure of the FERM/CTD complex, which reveals the structural basis of Merlin adopting a more closed conformation compared to its human cognate NF2. Interestingly, we found that the conserved lipid-binding site of Merlin might be masked by a linker. Confocal analyses confirmed that all putative lipid-binding site are very important for the membranal location of Merlin. More, we found that the phosphomimic Thr616Asp mutation weakens the interaction between FERM and CTD of Merlin. Collectively, the crystal structure of the FERM/CTD complex not only provides a mechanistic explanation of functionally dormant conformation of Merlin may also serve as a foundation for revealing the mechanism of conformational regulation of Merlin.
Collapse
Affiliation(s)
- Fayou Zhang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Science, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Beibei Liu
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Science, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Yaqi Gao
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Science, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Jiafu Long
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Science, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin, 300071, China.
| | - Hao Zhou
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Science, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin, 300071, China.
| |
Collapse
|
45
|
Spectrum of MYO7A Mutations in an Indigenous South African Population Further Elucidates the Nonsyndromic Autosomal Recessive Phenotype of DFNB2 to Include Both Homozygous and Compound Heterozygous Mutations. Genes (Basel) 2021; 12:genes12020274. [PMID: 33671976 PMCID: PMC7919343 DOI: 10.3390/genes12020274] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 01/20/2021] [Accepted: 01/21/2021] [Indexed: 11/17/2022] Open
Abstract
MYO7A gene encodes unconventional myosin VIIA, which, when mutated, causes a phenotypic spectrum ranging from recessive hearing loss DFNB2 to deaf-blindness, Usher Type 1B (USH1B). MYO7A mutations are reported in nine DFNB2 families to date, none from sub-Saharan Africa.In DNA, from a cohort of 94 individuals representing 92 families from the Limpopo province of South Africa, eight MYO7A variations were detected among 10 individuals. Family studies identified homozygous and compound heterozygous mutations in 17 individuals out of 32 available family members. Four mutations were novel, p.Gly329Asp, p.Arg373His, p.Tyr1780Ser, and p.Pro2126Leufs*5. Two variations, p.Ser617Pro and p.Thr381Met, previously listed as of uncertain significance (ClinVar), were confirmed to be pathogenic. The identified mutations are predicted to interfere with the conformational properties of myosin VIIA through interruption or abrogation of multiple interactions between the mutant and neighbouring residues. Specifically, p.Pro2126Leufs*5, is predicted to abolish the critical site for the interactions between the tail and the motor domain essential for the autoregulation, leaving a non-functional, unregulated protein that causes hearing loss. We have identified MYO7A as a possible key deafness gene among indigenous sub-Saharan Africans. The spectrum of MYO7A mutations in this South African population points to DFNB2 as a specific entity that may occur in a homozygous or in a compound heterozygous state.
Collapse
|
46
|
De Luca E, Perrelli A, Swamy H, Nitti M, Passalacqua M, Furfaro AL, Salzano AM, Scaloni A, Glading AJ, Retta SF. Protein kinase Cα regulates the nucleocytoplasmic shuttling of KRIT1. J Cell Sci 2021; 134:jcs250217. [PMID: 33443102 PMCID: PMC7875496 DOI: 10.1242/jcs.250217] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 12/15/2020] [Indexed: 12/16/2022] Open
Abstract
KRIT1 is a scaffolding protein that regulates multiple molecular mechanisms, including cell-cell and cell-matrix adhesion, and redox homeostasis and signaling. However, rather little is known about how KRIT1 is itself regulated. KRIT1 is found in both the cytoplasm and the nucleus, yet the upstream signaling proteins and mechanisms that regulate KRIT1 nucleocytoplasmic shuttling are not well understood. Here, we identify a key role for protein kinase C (PKC) in this process. In particular, we found that PKC activation promotes the redox-dependent cytoplasmic localization of KRIT1, whereas inhibition of PKC or treatment with the antioxidant N-acetylcysteine leads to KRIT1 nuclear accumulation. Moreover, we demonstrated that the N-terminal region of KRIT1 is crucial for the ability of PKC to regulate KRIT1 nucleocytoplasmic shuttling, and may be a target for PKC-dependent regulatory phosphorylation events. Finally, we found that silencing of PKCα, but not PKCδ, inhibits phorbol 12-myristate 13-acetate (PMA)-induced cytoplasmic enrichment of KRIT1, suggesting a major role for PKCα in regulating KRIT1 nucleocytoplasmic shuttling. Overall, our findings identify PKCα as a novel regulator of KRIT1 subcellular compartmentalization, thus shedding new light on the physiopathological functions of this protein.
Collapse
Affiliation(s)
- Elisa De Luca
- Department of Clinical and Biological Sciences, University of Torino, 10043 Orbassano, Torino, Italy
- CCM Italia Research Network, National Coordination Center at the Department of Clinical and Biological Sciences, University of Torino, 10043 Orbassano, Torino, Italy
- Center for Biomolecular Nanotechnologies, Istituto Italiano di Tecnologia, 73010 Arnesano, Lecce, Italy
| | - Andrea Perrelli
- Department of Clinical and Biological Sciences, University of Torino, 10043 Orbassano, Torino, Italy
- CCM Italia Research Network, National Coordination Center at the Department of Clinical and Biological Sciences, University of Torino, 10043 Orbassano, Torino, Italy
| | - Harsha Swamy
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY 14642, USA
| | - Mariapaola Nitti
- Department of Experimental Medicine, University of Genoa, 16132 Genova, Italy
| | - Mario Passalacqua
- Department of Experimental Medicine, University of Genoa, 16132 Genova, Italy
| | - Anna Lisa Furfaro
- Department of Experimental Medicine, University of Genoa, 16132 Genova, Italy
| | - Anna Maria Salzano
- Proteomics & Mass Spectrometry Laboratory, ISPAAM, National Research Council, 80147 Napoli, Italy
| | - Andrea Scaloni
- Proteomics & Mass Spectrometry Laboratory, ISPAAM, National Research Council, 80147 Napoli, Italy
| | - Angela J Glading
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY 14642, USA
| | - Saverio Francesco Retta
- Department of Clinical and Biological Sciences, University of Torino, 10043 Orbassano, Torino, Italy
- CCM Italia Research Network, National Coordination Center at the Department of Clinical and Biological Sciences, University of Torino, 10043 Orbassano, Torino, Italy
| |
Collapse
|
47
|
Liu YG, Chen Y, Wang X, Zhao P, Zhu Y, Qi Z. Ezrin is essential for the entry of Japanese encephalitis virus into the human brain microvascular endothelial cells. Emerg Microbes Infect 2021; 9:1330-1341. [PMID: 32538298 PMCID: PMC7473060 DOI: 10.1080/22221751.2020.1757388] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Japanese encephalitis virus (JEV) remains the predominant cause of viral encephalitis worldwide. It reaches the central nervous system upon crossing the blood-brain barrier through pathogenic mechanisms that are not completely understood. Here, using a high-throughput siRNA screening assay combined with verification experiments, we found that JEV enters the primary human brain microvascular endothelial cells (HBMEC) through a caveolae-mediated endocytic pathway. The role of ezrin, an essential host factor for JEV entry based on our screening, in caveolae-mediated JEV internalization was investigated. We observed that JEV internalization in HBMEC is largely dependent on ezrin-mediated actin cytoskeleton polymerization. Moreover, Src, a protein predicted by a STRING database search, was found to be required in JEV entry. By a variety of pharmacological inhibition and immunoprecipitation assays, we found that Src, ezrin, and caveolin-1 were sequentially activated and formed a complex during JEV infection. A combination of in vitro kinase assay and subcellular analysis demonstrated that ezrin is essential for Src-caveolin-1 interactions. In vivo, both Src and ezrin inhibitors protected ICR suckling mice against JEV-induced mortality and diminished mouse brain viral load. Therefore, JEV entry into HBMEC requires the activation of the Src-ezrin-caveolin-1 signalling axis, which provides potential targets for restricting JEV infection.
Collapse
Affiliation(s)
- Yan-Gang Liu
- Department of Microbiology, Shanghai Key Laboratory of Medical Biodefense, Naval Medical University (Second Military Medical University), Shanghai, People's Republic of China
| | - Yang Chen
- Department of Microbiology, Shanghai Key Laboratory of Medical Biodefense, Naval Medical University (Second Military Medical University), Shanghai, People's Republic of China.,College of Basic Medicine, Naval Medical University (Second Military Medical University Shanghai), Shanghai, People's Republic of China
| | - Xiaohang Wang
- Department of Microbiology, Shanghai Key Laboratory of Medical Biodefense, Naval Medical University (Second Military Medical University), Shanghai, People's Republic of China.,College of Basic Medicine, Naval Medical University (Second Military Medical University Shanghai), Shanghai, People's Republic of China
| | - Ping Zhao
- Department of Microbiology, Shanghai Key Laboratory of Medical Biodefense, Naval Medical University (Second Military Medical University), Shanghai, People's Republic of China
| | - Yongzhe Zhu
- Department of Microbiology, Shanghai Key Laboratory of Medical Biodefense, Naval Medical University (Second Military Medical University), Shanghai, People's Republic of China
| | - Zhongtian Qi
- Department of Microbiology, Shanghai Key Laboratory of Medical Biodefense, Naval Medical University (Second Military Medical University), Shanghai, People's Republic of China
| |
Collapse
|
48
|
Wang M, Jiang X, Yang Y, Chen H, Zhang C, Xu H, Qi B, Yao C, Xia H. Rhoj Is a Novel Target for Progression and Invasion of Glioblastoma by Impairing Cytoskeleton Dynamics. Neurotherapeutics 2020; 17:2028-2040. [PMID: 32822001 PMCID: PMC7851251 DOI: 10.1007/s13311-020-00910-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Rho GTPase family members were identified as critical regulators of cell morphology, actin cytoskeleton organization, cell movement, and cell cycle and also contributed to tumor progression, which have been implicated in various types of cancer metastasis and growth. Here, we firstly reported the dysregulation of Rhoj in glioblastoma multiforme (GBM) and aimed to investigate the role and mechanism of Rhoj in GBM. We analyzed the expression of 21 Rho GTPases family members and validated the expression of Rhoj in GBM by immunohistochemistry. We further investigated the role and mechanism of Rhoj in GBM both in vitro and in vivo. We observed that Rhoj is significantly overexpressed in GBM and associated with patients' survival. However, the role and underlying molecular mechanism of Rhoj in GBM are still unclear. We demonstrated that transcription factor c-Jun regulated the expression of Rhoj, and Rhoj interacted with moesin to promote GBM cell proliferation and migration by potentiating the activation of Rac1/PAK pathway and cytoskeletal dynamics. Rhoj may promote migration and invasion of GBM cells by regulating epithelial-mesenchymal transition (EMT)-like process. In conclusion, the Rhoj/Rac1/PAK signaling mediates invasion and progression of GBM and is a potential therapeutic target for GBM treatment. Rhoj may also be a promising biomarker for GBM diagnosis and prognosis.
Collapse
Affiliation(s)
- Mei Wang
- Department of Microbiology and Immunology, Medical School of Southeast University, Nanjing, 210009, China
- Department of Pathology, School of Basic Medical Sciences & Sir Run Run Hospital & State Key Laboratory of Reproductive Medicine & Key Laboratory of Antibody Technique of National Health Commission, Nanjing Medical University, Nanjing, 211166, China
| | - Xiaochun Jiang
- Department of Neurosurgery, Yijishan Hospital of Wannan Medical College, Wuhu, 241001, China
| | - Yongbo Yang
- Department of Neurosurgery, The Affiliated Drum Tower Hospital of Nanjing University, Nanjing, 210008, China
| | - Hongjin Chen
- Department of Pathology, School of Basic Medical Sciences & Sir Run Run Hospital & State Key Laboratory of Reproductive Medicine & Key Laboratory of Antibody Technique of National Health Commission, Nanjing Medical University, Nanjing, 211166, China
| | - Chengfei Zhang
- Department of Pathology, School of Basic Medical Sciences & Sir Run Run Hospital & State Key Laboratory of Reproductive Medicine & Key Laboratory of Antibody Technique of National Health Commission, Nanjing Medical University, Nanjing, 211166, China
| | - Haojun Xu
- Department of Pathology, School of Basic Medical Sciences & Sir Run Run Hospital & State Key Laboratory of Reproductive Medicine & Key Laboratory of Antibody Technique of National Health Commission, Nanjing Medical University, Nanjing, 211166, China
| | - Bin Qi
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, 130021, Jilin, China
| | - Chengyun Yao
- Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jangsu Institute of Cancer Research, Nanjing, 2100092, China.
| | - Hongping Xia
- Department of Microbiology and Immunology, Medical School of Southeast University, Nanjing, 210009, China.
- Department of Pathology, School of Basic Medical Sciences & Sir Run Run Hospital & State Key Laboratory of Reproductive Medicine & Key Laboratory of Antibody Technique of National Health Commission, Nanjing Medical University, Nanjing, 211166, China.
- Department of Neurosurgery, Yijishan Hospital of Wannan Medical College, Wuhu, 241001, China.
- Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jangsu Institute of Cancer Research, Nanjing, 2100092, China.
| |
Collapse
|
49
|
Salman A, Hutton SB, Newall T, Scott JA, Griffiths HL, Lee H, Gomez-Nicola D, Lotery AJ, Self JE. Characterization of the Frmd7 Knock-Out Mice Generated by the EUCOMM/COMP Repository as a Model for Idiopathic Infantile Nystagmus (IIN). Genes (Basel) 2020; 11:genes11101157. [PMID: 33007925 PMCID: PMC7601595 DOI: 10.3390/genes11101157] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 09/21/2020] [Accepted: 09/24/2020] [Indexed: 12/30/2022] Open
Abstract
In this study, we seek to exclude other pathophysiological mechanisms by which Frmd7 knock-down may cause Idiopathic Infantile Nystagmus (IIN) using the Frmd7.tm1a and Frmd7.tm1b murine models. We used a combination of genetic, histological and visual function techniques to characterize the role of Frmd7 gene in IIN using a novel murine model for the disease. We demonstrate that the Frmd7.tm1b allele represents a more robust model of Frmd7 knock-out at the mRNA level. The expression of Frmd7 was investigated using both antibody staining and X-gal staining confirming previous reports that Frmd7 expression in the retina is restricted to starburst amacrine cells and demonstrating that X-gal staining recapitulates the expression pattern in this model. Thus, it offers a useful tool for further expression studies. We also show that gross retinal morphology and electrophysiology are unchanged in these Frmd7 mutant models when compared with wild-type mice. High-speed eye-tracking recordings of Frmd7 mutant mice confirm a specific horizontal optokinetic reflex defect. In summary, our study confirms the likely role for Frmd7 in the optokinetic reflex in mice mediated by starburst amacrine cells. We show that the Frmd7.tm1b model provides a more robust knock-out than the Frmd7.tm1a model at the mRNA level, although the functional consequence is unchanged. Finally, we establish a robust eye-tracking technique in mice that can be used in a variety of future studies using this model and others. Although our data highlight a deficit in the optiokinetic reflex as a result of the starburst amacrine cells in the retina, this does not rule out the involvement of other cells, in the brain or the retina where Frmd7 is expressed, in the pathophysiology of IIN.
Collapse
MESH Headings
- Alleles
- Amacrine Cells/metabolism
- Animals
- Cytoskeletal Proteins/genetics
- Cytoskeletal Proteins/metabolism
- Disease Models, Animal
- Electroretinography
- Female
- Gene Expression
- Genetic Diseases, X-Linked/genetics
- Genetic Diseases, X-Linked/pathology
- Genetic Diseases, X-Linked/physiopathology
- Male
- Mice
- Mice, Knockout
- Mutation
- Nystagmus, Congenital/genetics
- Nystagmus, Congenital/pathology
- Nystagmus, Congenital/physiopathology
- Nystagmus, Optokinetic
- Retina/metabolism
- Retina/pathology
- Retina/physiopathology
- Tomography, Optical Coherence
Collapse
Affiliation(s)
- Ahmed Salman
- Clinical and Experimental Neurosciences, University of Southampton, Southampton SO16 6YD, UK; (T.N.); (J.A.S.); (H.L.G.); (H.L.); (A.J.L.); (J.E.S.)
- Correspondence:
| | - Samuel B. Hutton
- School of Psychology, University of Sussex, Brighton BN1 9QH, UK;
| | - Tutte Newall
- Clinical and Experimental Neurosciences, University of Southampton, Southampton SO16 6YD, UK; (T.N.); (J.A.S.); (H.L.G.); (H.L.); (A.J.L.); (J.E.S.)
| | - Jennifer A. Scott
- Clinical and Experimental Neurosciences, University of Southampton, Southampton SO16 6YD, UK; (T.N.); (J.A.S.); (H.L.G.); (H.L.); (A.J.L.); (J.E.S.)
| | - Helen L. Griffiths
- Clinical and Experimental Neurosciences, University of Southampton, Southampton SO16 6YD, UK; (T.N.); (J.A.S.); (H.L.G.); (H.L.); (A.J.L.); (J.E.S.)
| | - Helena Lee
- Clinical and Experimental Neurosciences, University of Southampton, Southampton SO16 6YD, UK; (T.N.); (J.A.S.); (H.L.G.); (H.L.); (A.J.L.); (J.E.S.)
| | - Diego Gomez-Nicola
- School of Biological Sciences, University of Southampton, Southampton SO171BJ, UK;
| | - Andrew J. Lotery
- Clinical and Experimental Neurosciences, University of Southampton, Southampton SO16 6YD, UK; (T.N.); (J.A.S.); (H.L.G.); (H.L.); (A.J.L.); (J.E.S.)
| | - Jay E. Self
- Clinical and Experimental Neurosciences, University of Southampton, Southampton SO16 6YD, UK; (T.N.); (J.A.S.); (H.L.G.); (H.L.); (A.J.L.); (J.E.S.)
| |
Collapse
|
50
|
Zhang Y, Lin Z, Fang Y, Wu J. Prediction of Catch-Slip Bond Transition of Kindlin2/β3 Integrin via Steered Molecular Dynamics Simulation. J Chem Inf Model 2020; 60:5132-5141. [PMID: 32877187 DOI: 10.1021/acs.jcim.0c00837] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Kindlin2 is believed to be crucial in integrin activation, which mediates the cell-extracellular matrix adhesion and signaling, but the mechanoregulation of the interaction between Kindlin2 and integrin remains unclear. Here, we performed the so-called "ramp-clamp" steered molecular dynamics simulation on the crystal structure of Kindlin2 bound with β3 integrin. The results showed that the complex had a better mechanical strength for its rupture force of about 200 pN under pulling with the velocity of 1 Å/ns, and was mechanostable for its conformational conservation under constant tensile force (≤60 pN). The catch-slip bond transition with a force threshold of 20 pN was demonstrated by the dissociation probability, the interaction energy, the interface H-bond number, and the force-induced allostery of the complex. This study might provide a novel insight into force-dependent Kindlin2/integrin-related signaling and its structural basis in cellular processes as well as a rational SMD-based computer strategy for predicting the structure-function relationship of the stretched complex.
Collapse
Affiliation(s)
- Yan Zhang
- Institute of Biomechanics, School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510006, China
| | - Zhanyi Lin
- Department of Cardiology, Institute of Geriatric Medicine, Guangdong Academy of Medical Sciences, Guangdong General Hospital, Guangzhou 510080, China
| | - Ying Fang
- Institute of Biomechanics, School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510006, China
| | - Jianhua Wu
- Institute of Biomechanics, School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510006, China
| |
Collapse
|