1
|
Matoo S, Graves MJ, Choi MS, Idris RAES, Acharya P, Thapa G, Nguyen T, Atallah SY, Tipirneni AK, Stevenson PJ, Crawley SW. The microvillar protocadherin CDHR5 associates with EBP50 to promote brush border assembly. Mol Biol Cell 2024; 35:ar36. [PMID: 38170579 PMCID: PMC10916864 DOI: 10.1091/mbc.e23-02-0065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 10/27/2023] [Accepted: 12/22/2023] [Indexed: 01/05/2024] Open
Abstract
Transporting epithelial cells of the gut and kidney interact with their luminal environment through a densely packed collection of apical microvilli known as a brush border (BB). Proper brush border assembly depends on the intermicrovillar adhesion complex (IMAC), a protocadherin-based adhesion complex found at the distal tips of microvilli that mediates adhesion between neighboring protrusions to promote their organized packing. Loss of the IMAC adhesion molecule Cadherin-related family member 5 (CDHR5) results in significant brush border defects, though the functional properties of this protocadherin have not been thoroughly explored. Here, we show that the cytoplasmic tail of CDHR5 contributes to its correct apical targeting and functional properties in an isoform-specific manner. Library screening identified the Ezrin-associated scaffolds EBP50 and E3KARP as cytoplasmic binding partners for CDHR5. Consistent with this, loss of EBP50 disrupted proper brush border assembly with cells exhibiting markedly reduced apical IMAC levels. Together, our results shed light on the apical targeting determinants of CDHR5 and further define the interactome of the IMAC involved in brush border assembly.
Collapse
Affiliation(s)
- Samaneh Matoo
- Department of Biological Sciences, University of Toledo, Toledo, Ohio 43606
| | - Maura J. Graves
- Department of Biological Sciences, University of Toledo, Toledo, Ohio 43606
| | - Myoung Soo Choi
- Department of Biological Sciences, University of Toledo, Toledo, Ohio 43606
| | | | - Prashun Acharya
- Department of Biological Sciences, University of Toledo, Toledo, Ohio 43606
| | - Garima Thapa
- Department of Biological Sciences, University of Toledo, Toledo, Ohio 43606
| | - Tram Nguyen
- Department of Biological Sciences, University of Toledo, Toledo, Ohio 43606
| | - Sarah Y. Atallah
- Department of Biological Sciences, University of Toledo, Toledo, Ohio 43606
| | - Ashna K. Tipirneni
- Department of Biological Sciences, University of Toledo, Toledo, Ohio 43606
| | | | - Scott W. Crawley
- Department of Biological Sciences, University of Toledo, Toledo, Ohio 43606
| |
Collapse
|
2
|
Kalashyan M, Raghunathan K, Oller H, Bayer MT, Jimenez L, Roland JT, Kolobova E, Hagen SJ, Goldsmith JD, Shub MD, Goldenring JR, Kaji I, Thiagarajah JR. Patient-derived enteroids provide a platform for the development of therapeutic approaches in microvillus inclusion disease. J Clin Invest 2023; 133:e169234. [PMID: 37643022 PMCID: PMC10575727 DOI: 10.1172/jci169234] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 08/22/2023] [Indexed: 08/31/2023] Open
Abstract
Microvillus inclusion disease (MVID), caused by loss-of-function mutations in the motor protein myosin Vb (MYO5B), is a severe infantile disease characterized by diarrhea, malabsorption, and acid/base instability, requiring intensive parenteral support for nutritional and fluid management. Human patient-derived enteroids represent a model for investigation of monogenic epithelial disorders but are a rare resource from MVID patients. We developed human enteroids with different loss-of function MYO5B variants and showed that they recapitulated the structural changes found in native MVID enterocytes. Multiplex immunofluorescence imaging of patient duodenal tissues revealed patient-specific changes in localization of brush border transporters. Functional analysis of electrolyte transport revealed profound loss of Na+/H+ exchange (NHE) activity in MVID patient enteroids with near-normal chloride secretion. The chloride channel-blocking antidiarrheal drug crofelemer dose-dependently inhibited agonist-mediated fluid secretion. MVID enteroids exhibited altered differentiation and maturation versus healthy enteroids. γ-Secretase inhibition with DAPT recovered apical brush border structure and functional Na+/H+ exchange activity in MVID enteroids. Transcriptomic analysis revealed potential pathways involved in the rescue of MVID cells including serum/glucocorticoid-regulated kinase 2 (SGK2) and NHE regulatory factor 3 (NHERF3). These results demonstrate the utility of patient-derived enteroids for developing therapeutic approaches to MVID.
Collapse
Affiliation(s)
- Meri Kalashyan
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Krishnan Raghunathan
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Haley Oller
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Marie-Theres Bayer
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Lissette Jimenez
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Congenital Enteropathy Program, Boston Children’s Hospital, Boston, Massachusetts, USA
- PediCODE Consortium, as detailed in Supplemental Acknowledgments
| | - Joseph T. Roland
- Section of Surgical Sciences and
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Elena Kolobova
- Section of Surgical Sciences and
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Susan J. Hagen
- Department of Surgery, Division of Surgical Sciences, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Jeffrey D. Goldsmith
- PediCODE Consortium, as detailed in Supplemental Acknowledgments
- Department of Pathology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Mitchell D. Shub
- Department of Child Health, University of Arizona College of Medicine–Phoenix, and Division of Gastroenterology, Phoenix Children’s Hospital, Phoenix, Arizona, USA
| | - James R. Goldenring
- PediCODE Consortium, as detailed in Supplemental Acknowledgments
- Section of Surgical Sciences and
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Nashville VA Medical Center, Nashville, Tennessee, USA
| | - Izumi Kaji
- PediCODE Consortium, as detailed in Supplemental Acknowledgments
- Section of Surgical Sciences and
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jay R. Thiagarajah
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Congenital Enteropathy Program, Boston Children’s Hospital, Boston, Massachusetts, USA
- PediCODE Consortium, as detailed in Supplemental Acknowledgments
- Harvard Digestive Disease Center, Boston, Massachusetts, USA
| |
Collapse
|
3
|
Nakagawa M, Matsumoto T, Yokoi A, Hashimura M, Oguri Y, Konno R, Ishibashi Y, Ito T, Ohhigata K, Harada Y, Fukagawa N, Kodera Y, Saegusa M. Interaction between membranous EBP50 and myosin 9 as a favorable prognostic factor in ovarian clear cell carcinoma. Mol Oncol 2023; 17:2168-2182. [PMID: 37539980 PMCID: PMC10552901 DOI: 10.1002/1878-0261.13503] [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: 04/18/2023] [Revised: 06/29/2023] [Accepted: 08/01/2023] [Indexed: 08/05/2023] Open
Abstract
Ezrin-radixin-moesin-binding phosphoprotein 50 (EBP50) is a scaffold protein that is required for epithelial polarity. Knockout (KO) of membranous EBP50 (Me-EBP50) in ovarian clear cell carcinoma (OCCC) cells induced an epithelial-mesenchymal transition (EMT)-like phenotype, along with decreased proliferation, accelerated migration capability, and induction of cancer stem cell (CSC)-like properties. Shotgun proteomics analysis of proteins that co-immunoprecipitated with EBP50 revealed that Me-EBP50 strongly interacts with myosin 9 (MYH9). Specific inhibition of MYH9 with blebbistatin phenocopied Me-EBP50 KO, and blebbistatin treatment potentiated the effects of Me-EBP50 KO. In OCCC cells from clinical samples, Me-EBP50 and MYH9 were co-localized at the apical plasma membrane. Patients with a combination of Me-EBP50-high and MYH9-high scores had the best prognosis for overall and progression-free survival. Our data suggest that Me-EBP50 has tumor-suppressive effects through the establishment and maintenance of epithelial polarization. By contrast, loss of Me-EBP50 expression induces EMT-like phenotypes, probably due to MYH9 dysfunction; this results in increased cell mobility and enhanced CSC-like properties, which in turn promote OCCC progression.
Collapse
Affiliation(s)
- Mayu Nakagawa
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
| | - Toshihide Matsumoto
- Department of PathologyKitasato University School of Allied Health ScienceSagamiharaJapan
| | - Ako Yokoi
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
| | - Miki Hashimura
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
| | - Yasuko Oguri
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
| | - Ryo Konno
- Center for Disease Proteomics, School of ScienceKitasato UniversitySagamiharaJapan
| | - Yu Ishibashi
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
| | - Takashi Ito
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
| | - Kensuke Ohhigata
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
| | - Yohei Harada
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
| | - Naomi Fukagawa
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
| | - Yoshio Kodera
- Center for Disease Proteomics, School of ScienceKitasato UniversitySagamiharaJapan
| | - Makoto Saegusa
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
| |
Collapse
|
4
|
Thüring EM, Hartmann C, Schwietzer YA, Ebnet K. TMIGD1: Emerging functions of a tumor supressor and adhesion receptor. Oncogene 2023:10.1038/s41388-023-02696-5. [PMID: 37087524 DOI: 10.1038/s41388-023-02696-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 04/11/2023] [Accepted: 04/17/2023] [Indexed: 04/24/2023]
Abstract
The development of multicellular organisms depends on cell adhesion molecules (CAMs) that connect cells to build tissues. The immunoglobulin superfamily (IgSF) constitutes one of the largest families of CAMs. Members of this family regulate such diverse processes like synapse formation, spermatogenesis, leukocyte-endothelial interactions, or epithelial cell-cell adhesion. Through their extracellular domains, they undergo homophilic and heterophilic interactions in cis and trans. Their cytoplasmic domains frequently bind scaffolding proteins to assemble signaling complexes. Transmembrane and immunoglobulin domain-containing protein 1 (TMIGD1) is a IgSF member with two Ig-like domains and a short cytoplasmic tail that contains a PDZ domain-binding motif. Recent observations indicate that TMIGD1 has pleiotropic functions in epithelial cells and has a critical role in suppressing malignant cell behavior. Here, we review the molecular characteristics of TMIGD1, its interaction with cytoplasmic scaffolding proteins, the regulation of its expression, and its downregulation in colorectal and renal cancers.
Collapse
Affiliation(s)
- Eva-Maria Thüring
- Institute-associated Research Group "Cell adhesion and cell polarity", Institute of Medical Biochemistry, ZMBE, University of Münster, Münster, Germany
| | - Christian Hartmann
- Institute-associated Research Group "Cell adhesion and cell polarity", Institute of Medical Biochemistry, ZMBE, University of Münster, Münster, Germany
| | - Ysabel A Schwietzer
- Institute-associated Research Group "Cell adhesion and cell polarity", Institute of Medical Biochemistry, ZMBE, University of Münster, Münster, Germany
| | - Klaus Ebnet
- Institute-associated Research Group "Cell adhesion and cell polarity", Institute of Medical Biochemistry, ZMBE, University of Münster, Münster, Germany.
| |
Collapse
|
5
|
Kalashyan M, Raghunathan K, Oller H, Theres MB, Jimenez L, Roland JT, Kolobova E, Hagen SJ, Goldsmith JD, Shub MD, Goldenring JR, Kaji I, Thiagarajah JR. Therapy Development for Microvillus Inclusion Disease using Patient-derived Enteroids. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.28.526036. [PMID: 36747680 PMCID: PMC9900906 DOI: 10.1101/2023.01.28.526036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Microvillus Inclusion Disease (MVID), caused by loss-of-function mutations in the motor protein Myosin Vb (MYO5B), is a severe infantile disease characterized by diarrhea, malabsorption, and acid-base instability, requiring intensive parenteral support for nutritional and fluid management. Human patient-derived enteroids represent a model for investigation of monogenic epithelial disorders but are a rare resource from MVID patients. We developed human enteroids with different loss-of function MYO5B variants and showed that they recapitulated the structural changes found in native MVID enterocytes. Multiplex Immunofluorescence imaging of patient duodenal tissues revealed patient-specific changes in localization of brush border transporters. Functional analysis of electrolyte transport revealed profound loss of Na + /H + exchange (NHE) activity in MVID patient enteroids with near-normal chloride secretion. The chloride channel-blocking anti-diarrheal drug, Crofelemer, dose-dependently inhibited agonist-mediated fluid secretion. MVID enteroids exhibited altered differentiation and maturation versus healthy enteroids. Inhibition of Notch signaling with the γ-secretase inhibitor, DAPT, recovered apical brush border structure and functional Na + /H + exchange activity in MVID enteroids. Transcriptomic analysis revealed potential pathways involved in the rescue of MVID cells including serum- and glucocorticoid-induced protein kinase 2 (SGK2), and NHE regulatory factor 3 (NHERF3). These results demonstrate the utility of patient-derived enteroids for developing therapeutic approaches to MVID. Conflict-of-interest statement The authors have declared that no conflict of interest exists.
Collapse
Affiliation(s)
- Meri Kalashyan
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children’s Hospital; Harvard Medical School, Boston, MA
| | - Krishnan Raghunathan
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children’s Hospital; Harvard Medical School, Boston, MA
| | - Haley Oller
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children’s Hospital; Harvard Medical School, Boston, MA
| | - Marie-Bayer Theres
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children’s Hospital; Harvard Medical School, Boston, MA
| | - Lissette Jimenez
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children’s Hospital; Harvard Medical School, Boston, MA
- Congenital Enteropathy Program, Boston Children’s Hospital, Boston, MA
- PediCoDE Consortium
| | - Joseph T. Roland
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Elena Kolobova
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Susan J Hagen
- Department of Surgery, Division of Surgical Sciences, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Jeffrey D. Goldsmith
- Department of Pathology, Boston Children’s Hospital; Harvard Medical School, Boston, MA
- PediCoDE Consortium
| | - Mitchell D. Shub
- Department of Child Health University of Arizona College of Medicine-Phoenix and Division of Gastroenterology, Phoenix Children’s
| | - James R. Goldenring
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee
- Nashville VA Medical Center, Nashville, TN
- PediCoDE Consortium
| | - Izumi Kaji
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee
- PediCoDE Consortium
| | - Jay R. Thiagarajah
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children’s Hospital; Harvard Medical School, Boston, MA
- Congenital Enteropathy Program, Boston Children’s Hospital, Boston, MA
- Harvard Digestive Disease Center, Boston MA
- PediCoDE Consortium
| |
Collapse
|
6
|
Hartmann C, Thüring EM, Greune L, Michels BE, Pajonczyk D, Leußink S, Brinkmann F, Glaesner-Ebnet M, Wardelmann E, Zobel T, Schmidt MA, Janssen KP, Gerke V, Ebnet K. Intestinal brush border formation requires a TMIGD1-based intermicrovillar adhesion complex. Sci Signal 2022; 15:eabm2449. [PMID: 36099341 DOI: 10.1126/scisignal.abm2449] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Intestinal epithelial cells absorb nutrients through the brush border, composed of dense arrays of highly ordered microvilli at their apical membranes. A protocadherin-based intermicrovillar adhesion complex localized at microvilli tips mediates microvilli packing and organization. Here, we identified a second adhesion complex localized at the proximal base region of microvilli. This complex contained the immunoglobulin superfamily member TMIGD1, which directly interacted with the microvillar scaffolding proteins EBP50 and E3KARP. Complex formation with EBP50 required the activation of EBP50 by the actin-binding protein ezrin and was enhanced by the dephosphorylation of Ser162 in the PDZ2 domain of EBP50 by the phosphatase PP1α. Binding of the EBP50-ezrin complex to TMIGD1 enhanced the dynamic turnover of EBP50 at microvilli. Enterocyte-specific inactivation of Tmigd1 in mice resulted in microvillar blebbing, loss of intermicrovillar adhesion, and perturbed brush border formation. Thus, we identified a second adhesion complex in microvilli and propose a mechanism that promotes microvillar formation and dynamics.
Collapse
Affiliation(s)
- Christian Hartmann
- Institute-associated Research Group "Cell adhesion and cell polarity", ZMBE, University of Münster, D-48149 Münster, Germany.,Institute of Medical Biochemistry, ZMBE, University of Münster, D-48149 Münster, Germany
| | - Eva-Maria Thüring
- Institute-associated Research Group "Cell adhesion and cell polarity", ZMBE, University of Münster, D-48149 Münster, Germany.,Institute of Medical Biochemistry, ZMBE, University of Münster, D-48149 Münster, Germany
| | - Lilo Greune
- Institute of Infectiology, ZMBE, University of Münster, D-48149 Münster, Germany
| | - Birgitta E Michels
- Institute-associated Research Group "Cell adhesion and cell polarity", ZMBE, University of Münster, D-48149 Münster, Germany.,Institute of Medical Biochemistry, ZMBE, University of Münster, D-48149 Münster, Germany
| | - Denise Pajonczyk
- Institute-associated Research Group "Cell adhesion and cell polarity", ZMBE, University of Münster, D-48149 Münster, Germany.,Institute of Medical Biochemistry, ZMBE, University of Münster, D-48149 Münster, Germany
| | - Sophia Leußink
- Institute-associated Research Group "Cell adhesion and cell polarity", ZMBE, University of Münster, D-48149 Münster, Germany.,Institute of Medical Biochemistry, ZMBE, University of Münster, D-48149 Münster, Germany
| | - Frauke Brinkmann
- Institute-associated Research Group "Cell adhesion and cell polarity", ZMBE, University of Münster, D-48149 Münster, Germany.,Institute of Medical Biochemistry, ZMBE, University of Münster, D-48149 Münster, Germany
| | - Mark Glaesner-Ebnet
- Institute-associated Research Group "Cell adhesion and cell polarity", ZMBE, University of Münster, D-48149 Münster, Germany.,Institute of Medical Biochemistry, ZMBE, University of Münster, D-48149 Münster, Germany
| | - Eva Wardelmann
- Gerhard-Domagk-Institute of Pathology, University Hospital Münster, D-48149 Münster, Germany
| | - Thomas Zobel
- Imaging Network Microscopy, University of Münster, D-48149 Münster, Germany
| | - M Alexander Schmidt
- Institute of Infectiology, ZMBE, University of Münster, D-48149 Münster, Germany
| | | | - Volker Gerke
- Institute of Medical Biochemistry, ZMBE, University of Münster, D-48149 Münster, Germany.,Cells-in-Motion Interfaculty Center (CiMIC), University of Münster, D-48419 Münster, Germany
| | - Klaus Ebnet
- Institute-associated Research Group "Cell adhesion and cell polarity", ZMBE, University of Münster, D-48149 Münster, Germany.,Institute of Medical Biochemistry, ZMBE, University of Münster, D-48149 Münster, Germany.,Cells-in-Motion Interfaculty Center (CiMIC), University of Münster, D-48419 Münster, Germany.,Interdisciplinary Center for Clinical Research (IZKF), University of Münster, D-48149 Münster, Germany
| |
Collapse
|
7
|
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
|
8
|
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
|
9
|
Xie J, Jusuf PR, Bui BV, Dudczig S, Sztal TE, Goodbourn PT. Altered Visual Function in a Larval Zebrafish Knockout of Neurodevelopmental Risk Gene pdzk1. Invest Ophthalmol Vis Sci 2021; 62:29. [PMID: 33749720 PMCID: PMC7991922 DOI: 10.1167/iovs.62.3.29] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose The human PDZK1 gene is located in a genomic susceptibility region for neurodevelopmental disorders. A genome-wide association study identified links between PDZK1 polymorphisms and altered visual contrast sensitivity, an endophenotype for schizophrenia and autism spectrum disorder. The PDZK1 protein is implicated in neurological functioning, interacting with synaptic molecules including postsynaptic density 95 (PSD-95), N-methyl-d-aspartate receptors (NMDARs), corticotropin-releasing factor receptor 1 (CRFR1), and serotonin 2A receptors. The purpose of the present study was to elucidate the role of PDZK1. Methods We generated pdzk1-knockout (pdzk1-KO) zebrafish using CRISPR/Cas-9 genome editing. Visual function of 7-day-old fish was assessed at behavioral and functional levels using the optomotor response and scotopic electroretinogram (ERG). We also quantified retinal morphology and densities of PSD-95, NMDAR1, CRFR1, and serotonin in the synaptic inner plexiform layer at 7 days, 4 weeks, and 8 weeks of age. Standard RT-PCR and nonsense-mediated decay interference treatment were also performed to assess genetic compensation in mutants. Results Relative to wild-type, pdzk1-KO larvae showed spatial frequency tuning functions with increased amplitude (likely due to abnormal gain control) and reduced ERG b-waves (suggestive of inner retinal dysfunction). No synaptic phenotypes, but possible morphological retinal phenotypes, were identified. We confirmed that the absence of major histological phenotypes was not attributable to genetic compensatory mechanisms. Conclusions Our findings point to a role for pdzk1 in zebrafish visual function, and our model system provides a platform for investigating other genes associated with abnormal visual behavior.
Collapse
Affiliation(s)
- Jiaheng Xie
- School of BioSciences, The University of Melbourne, Melbourne, Australia
| | - Patricia R Jusuf
- School of BioSciences, The University of Melbourne, Melbourne, Australia
| | - Bang V Bui
- Department of Optometry and Vision Sciences, The University of Melbourne, Melbourne, Australia
| | - Stefanie Dudczig
- School of BioSciences, The University of Melbourne, Melbourne, Australia
| | - Tamar E Sztal
- School of Biological Sciences, Monash University, Melbourne, Australia
| | - Patrick T Goodbourn
- Melbourne School of Psychological Sciences, The University of Melbourne, Melbourne, Australia
| |
Collapse
|
10
|
Song GJ, Gupta DP, Rahman MH, Park HT, Al Ghouleh I, Bisello A, Lee MG, Park JY, Park HH, Jun JH, Chung KW, Choi BO, Suk K. Loss-of-function of EBP50 is a new cause of hereditary peripheral neuropathy: EBP50 functions in peripheral nerve system. Glia 2020; 68:1794-1809. [PMID: 32077526 DOI: 10.1002/glia.23805] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 02/04/2020] [Accepted: 02/07/2020] [Indexed: 12/12/2022]
Abstract
Finding causative genetic mutations is important in the diagnosis and treatment of hereditary peripheral neuropathies. This study was conducted to find new genes involved in the pathophysiology of hereditary peripheral neuropathy. We identified a new mutation in the EBP50 gene, which is co-segregated with neuropathic phenotypes, including motor and sensory deficit in a family with Charcot-Marie-Tooth disease. EBP50 is known to be important for the formation of microvilli in epithelial cells, and the discovery of this gene mutation allowed us to study the function of EBP50 in the nervous system. EBP50 was strongly expressed in the nodal and paranodal regions of sciatic nerve fibers, where Schwann cell microvilli contact the axolemma, and at the growth tips of primary Schwann cells. In addition, EBP50 expression was decreased in mouse models of peripheral neuropathy. Knockout mice were used to study EBP50 function in the peripheral nervous system. Interestingly motor function deficit and abnormal histology of nerve fibers were observed in EBP50+/- heterozygous mice at 12 months of age, but not 3 months. in vitro studies using Schwann cells showed that NRG1-induced AKT activation and migration were significantly reduced in cells overexpressing the I325V mutant of EBP50 or cells with knocked-down EBP50 expression. In conclusion, we show for the first time that loss of function due to EBP50 gene deficiency or mutation can cause peripheral neuropathy.
Collapse
Affiliation(s)
- Gyun Jee Song
- Department of Medical Science, Institute for Bio-Medical Convergence, Catholic Kwandong University, International St. Mary's Hospital, Incheon, Republic of Korea
| | - Deepak Prasad Gupta
- Department of Pharmacology, Brain Science and Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Md Habibur Rahman
- Department of Pharmacology, Brain Science and Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Hwan Tae Park
- Department of Molecular Neuroscience, College of Medicine, Dong-A University, Busan, Republic of Korea
| | - Imad Al Ghouleh
- Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Alessandro Bisello
- Department of Pharmacology and Chemical Biology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Maan-Gee Lee
- Department of Pharmacology, Brain Science and Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Jae-Yong Park
- School of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul, Republic of Korea
| | - Hyun Ho Park
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
| | - Jin Hyun Jun
- Department of Senior Healthcare, BK21 Plus Program, Graduate School of Eulji University, Department of Biomedical Laboratory Science, College of Health Science, Eulji University, Seongnam, Republic of Korea
| | - Ki Wha Chung
- Department of Biological Sciences, Kongju National University, Gongju, Republic of Korea
| | - Byung-Ok Choi
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Kyoungho Suk
- Department of Pharmacology, Brain Science and Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| |
Collapse
|
11
|
Lenzen H, Qian J, Manns MP, Seidler U, Jörns A. Restoration of mucosal integrity and epithelial transport function by concomitant anti-TNFα treatment in chronic DSS-induced colitis. J Mol Med (Berl) 2018; 96:831-843. [DOI: 10.1007/s00109-018-1658-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 05/27/2018] [Accepted: 06/01/2018] [Indexed: 12/25/2022]
|
12
|
Apodaca G. Role of Polarity Proteins in the Generation and Organization of Apical Surface Protrusions. Cold Spring Harb Perspect Biol 2018; 10:cshperspect.a027813. [PMID: 28264821 DOI: 10.1101/cshperspect.a027813] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Protruding from the apical surfaces of epithelial cells are specialized structures, including cilia, microplicae, microvilli, and stereocilia. These contribute to epithelial function by cushioning the apical surface, by amplifying its surface area to facilitate nutrient absorption, and by promoting sensory transduction and barrier function. Despite these important roles, and the diseases that result when their formation is perturbed, there remain significant gaps in our understanding of the biogenesis of apical protrusions, or the pathways that promote their organization and orientation once at the apical surface. Here, I review some general aspects of these apical structures, and then discuss our current understanding of their formation and organization with respect to proteins that specify apicobasolateral polarity and planar cell polarity.
Collapse
Affiliation(s)
- Gerard Apodaca
- Department of Medicine Renal-Electrolyte Division and the Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261
| |
Collapse
|
13
|
Avula LR, Chen T, Kovbasnjuk O, Donowitz M. Both NHERF3 and NHERF2 are necessary for multiple aspects of acute regulation of NHE3 by elevated Ca 2+, cGMP, and lysophosphatidic acid. Am J Physiol Gastrointest Liver Physiol 2018; 314:G81-G90. [PMID: 28882822 PMCID: PMC5866371 DOI: 10.1152/ajpgi.00140.2017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The intestinal epithelial brush border Na+/H+ exchanger NHE3 accounts for a large component of intestinal Na absorption. NHE3 is regulated during digestion by signaling complexes on its COOH terminus that include the four multi-PDZ domain-containing NHERF family proteins. All bind to NHE3 and take part in different aspects of NHE3 regulation. Because the roles of each NHERF appear to vary on the basis of the cell model or intestinal segment studied and because of our recent finding that a NHERF3-NHERF2 heterodimer appears important for NHE3 regulation in Caco-2 cells, we examined the role of NHERF3 and NHERF2 in C57BL/6 mouse jejunum using homozygous NHERF2 and NHERF3 knockout mice. NHE3 activity was determined with two-photon microscopy and the dual-emission pH-sensitive dye SNARF-4F. The jejunal apical membrane of NHERF3-null mice appeared similar to wild-type (WT) mice in surface area, microvillus number, and height, which is similar to results previously reported for jejunum of NHERF2-null mice. NHE3 basal activity was not different from WT in either NHERF2- or NHERF3-null jejunum, while d-glucose-stimulated NHE3 activity was reduced in NHERF2, but similar to WT in NHERF3 KO. LPA stimulation and UTP (elevated Ca2+) and cGMP inhibition of NHE3 were markedly reduced in both NHERF2- and NHERF3-null jejunum. Forskolin inhibited NHE3 in NHERF3-null jejunum, but the extent of inhibition was reduced compared with WT. The forskolin inhibition of NHE3 in NHERF2-null mice was too inconsistent to determine whether there was an effect and whether it was altered compared with the WT response. These results demonstrate similar requirement for NHERF2 and NHERF3 in mouse jejunal NHE3 regulation by LPA, Ca2+, and cGMP. The explanation for the similarity is not known but is consistent with involvement of a brush-border NHERF3-NHERF2 heterodimer or sequential NHERF-dependent effects in these aspects of NHE3 regulation. NEW & NOTEWORTHY NHERF2 and NHERF3 are apical membrane multi-PDZ domain-containing proteins that are involved in regulation of intestinal NHE3. This study demonstrates that NHERF2 and NHERF3 have overlapping roles in NHE3 stimulation by LPA and inhibition by elevated Ca2+ and cGMP. These results are consistent with their role being as a NHERF3-NHERF2 heterodimer or via sequential NHERF-dependent signaling steps, and they begin to clarify a role for multiple NHERF proteins in NHE3 regulation.
Collapse
Affiliation(s)
- Leela Rani Avula
- 1Department of Medicine, the Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Tiane Chen
- 1Department of Medicine, the Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Olga Kovbasnjuk
- 1Department of Medicine, the Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Mark Donowitz
- 1Department of Medicine, the Johns Hopkins School of Medicine, Baltimore, Maryland,2Department of Physiology, the Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
14
|
Oh YS, Heo K, Kim EK, Jang JH, Bae SS, Park JB, Kim YH, Song M, Kim SR, Ryu SH, Kim IH, Suh PG. Dynamic relocalization of NHERF1 mediates chemotactic migration of ovarian cancer cells toward lysophosphatidic acid stimulation. Exp Mol Med 2017; 49:e351. [PMID: 28684865 PMCID: PMC5565956 DOI: 10.1038/emm.2017.88] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 12/28/2016] [Accepted: 01/09/2017] [Indexed: 01/05/2023] Open
Abstract
NHERF1/EBP50 (Na+/H+ exchanger regulating
factor 1; Ezrin-binding phosphoprotein of 50 kDa) organizes stable
protein complexes beneath the apical membrane of polar epithelial cells. By
contrast, in cancer cells without any fixed polarity, NHERF1 often localizes in
the cytoplasm. The regulation of cytoplasmic NHERF1 and its role in cancer
progression remain unclear. In this study, we found that, upon lysophosphatidic
acid (LPA) stimulation, cytoplasmic NHERF1 rapidly translocated to the plasma
membrane, and subsequently to cortical protrusion structures, of ovarian cancer
cells. This movement depended on direct binding of NHERF1 to C-terminally
phosphorylated ERM proteins (cpERMs). Moreover, NHERF1 depletion downregulated
cpERMs and further impaired cpERM-dependent remodeling of the cell cortex,
suggesting reciprocal regulation between these proteins. The LPA-induced protein
complex was highly enriched in migratory pseudopodia, whose formation was
impaired by overexpression of NHERF1 truncation mutants. Consistent with this,
NHERF1 depletion in various types of cancer cells abolished chemotactic cell
migration toward a LPA gradient. Taken together, our findings suggest that the
high dynamics of cytosolic NHERF1 provide cancer cells with a means of
controlling chemotactic migration. This capacity is likely to be essential for
ovarian cancer progression in tumor microenvironments containing LPA.
Collapse
Affiliation(s)
- Yong-Seok Oh
- Department of Brain-Cognitive Science, Daegu-Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea
| | - Kyun Heo
- Research Institute, National Cancer Center, Goyang, Republic of Korea
| | - Eung-Kyun Kim
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, Republic of Korea
| | - Jin-Hyeok Jang
- Department of Brain-Cognitive Science, Daegu-Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea
| | - Sun Sik Bae
- MRC for Ischemic Tissue Regeneration, Department of Pharmacology, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Jong Bae Park
- Research Institute, National Cancer Center, Goyang, Republic of Korea.,Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Republic of Korea
| | - Yun Hee Kim
- Research Institute, National Cancer Center, Goyang, Republic of Korea.,Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Republic of Korea
| | - Minseok Song
- Synaptic Circuit Plasticity Laboratory, Department of Structure and Function of Neural Network, Korea Brain Research Institute, Daegu, Republic of Korea
| | - Sang Ryong Kim
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Institute of Life Science and Biotechnology, Kyungpook National University, Daegu, Republic of Korea
| | - Sung Ho Ryu
- Division of Molecular and Life Science, Department of Life Science, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - In-Hoo Kim
- Research Institute, National Cancer Center, Goyang, Republic of Korea.,Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Republic of Korea
| | - Pann-Ghill Suh
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, Republic of Korea
| |
Collapse
|
15
|
Yoshida S, Fukutomi T, Kimura T, Sakurai H, Hatano R, Yamamoto H, Mukaisho KI, Hattori T, Sugihara H, Asano S. Comprehensive proteome analysis of brush border membrane fraction of ileum of ezrin knockdown mice. Biomed Res 2017; 37:127-39. [PMID: 27108882 DOI: 10.2220/biomedres.37.127] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Ezrin is an actin binding protein which cross-links membrane proteins with cytoskeleton directly or indirectly via PDZ domain-containing scaffold proteins. It is mainly expressed at the brush border membrane (BBM) of gastrointestinal tracts, and is involved in the construction of microvilli structure and the functional expression of membrane protein complexes at the cell surface. To precisely study the roles of ezrin on the expression of membrane proteins at the cell surface, here we prepared the BBM fractions of ileums from the wild-type and ezrin-knockdown (Vil2(kd/kd)) mice, analyzed them by mass spectrometry, and compared their proteomic patterns. Totally 313 proteins were identified in the BBM fractions. Several transport proteins, cytoskeleton-associated proteins, and trafficking proteins were up- or down-regulated in the BBM fraction of the ileum in the Vil2(kd/kd) mice. Among them, the expressions of i) Na(+)/H(+) exchanger regulatory factor 1 (a PDZ domain-containing scaffold protein), ii) sodium monocarboxylate transporter 1, which contains a PDZ domain-binding motif at their carboxy-terminal, and iii) chloride intracellular channel protein 5 were down-regulated at the BBM fraction of the ileum in the Vil2(kd/kd) mice, suggesting that ezrin is involved in their expression in the BBM.
Collapse
Affiliation(s)
- Saori Yoshida
- Department of Molecular Physiology, College of Pharmaceutical Sciences, Ritsumeikan University
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Henkel B, Bintig W, Bhat SS, Spehr M, Neuhaus EM. NHERF1 in Microvilli of Vomeronasal Sensory Neurons. Chem Senses 2016; 42:25-35. [PMID: 27655939 DOI: 10.1093/chemse/bjw094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
In most mammals, the vomeronasal system detects a variety of (semio)chemicals that mediate olfactory-driven social and sexual behaviors. Vomeronasal chemosensation depends on G protein-coupled receptors (V1R, V2R, and FPR-rs) that operate at remarkably low stimulus concentrations, thus, indicating a highly sensitive and efficient signaling pathway. We identified the PDZ domain-containing protein, Na+/H+ exchanger regulatory factor-1 (NHERF1), as putative molecular organizer of signal transduction in vomeronasal neurons. NHERF1 is a protein that contains 2 PDZ domains and a carboxy-terminal ezrin-binding domain. It localizes to microvilli of vomeronasal sensory neurons and interacts with V1Rs. Furthermore, NHERF1 and Gαi2 are closely colocalized. These findings open up new aspects of the functional organization and regulation of vomeronasal signal transduction by PDZ scaffolding proteins.
Collapse
Affiliation(s)
- Bastian Henkel
- Pharmacology and Toxicology, University Hospital Jena, Friedrich Schiller University Jena, Drackendorfer Straße 1, 07743 Jena, Germany.,Cluster of Excellence NeuroCure, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany and.,Present address: Institute of Anatomy, Medical Faculty, University Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany
| | - Willem Bintig
- Cluster of Excellence NeuroCure, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany and.,Present address: Institute of Biochemistry, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - S Sneha Bhat
- Pharmacology and Toxicology, University Hospital Jena, Friedrich Schiller University Jena, Drackendorfer Straße 1, 07743 Jena, Germany
| | - Marc Spehr
- Department of Chemosensation, Institute for Biology II, RWTH-Aachen University, Worringerweg 3, 52074 Aachen, Germany
| | - Eva M Neuhaus
- Pharmacology and Toxicology, University Hospital Jena, Friedrich Schiller University Jena, Drackendorfer Straße 1, 07743 Jena, Germany, .,Cluster of Excellence NeuroCure, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany and
| |
Collapse
|
17
|
Abstract
The brush border on the apical surface of enterocytes is a highly specialized structure well-adapted for efficient digestion and nutrient transport, whilst at the same time providing a protective barrier for the intestinal mucosa. The brush border is constituted of a densely ordered array of microvilli, protrusions of the plasma membrane, which are supported by actin-based microfilaments and interacting proteins and anchored in an apical network of actomyosin and intermediate filaments, the so-called terminal web. The highly dynamic, specialized apical domain is both an essential partner for the gut microbiota and an efficient signalling platform that enables adaptation to physiological stimuli from the external and internal milieu. Nevertheless, genetic alterations or various pathological stresses, such as infection, inflammation, and mechanical or nutritional alterations, can jeopardize this equilibrium and compromise intestinal functions. Long-time neglected, the intestinal brush-border shall be enlightening again as the central actor of the complex but essential intestinal homeostasis. Here, we review the processes and components involved in brush border organization and discuss pathological mechanisms that can induce brush border defects and their physiological consequences.
Collapse
|
18
|
Sauvanet C, Wayt J, Pelaseyed T, Bretscher A. Structure, Regulation, and Functional Diversity of Microvilli on the Apical Domain of Epithelial Cells. Annu Rev Cell Dev Biol 2015; 31:593-621. [DOI: 10.1146/annurev-cellbio-100814-125234] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Cécile Sauvanet
- Department of Molecular Biology and Genetics and Weill Institute for Molecular and Cell Biology, Cornell University, Ithaca, New York 14853;
| | - Jessica Wayt
- Department of Molecular Biology and Genetics and Weill Institute for Molecular and Cell Biology, Cornell University, Ithaca, New York 14853;
| | - Thaher Pelaseyed
- Department of Molecular Biology and Genetics and Weill Institute for Molecular and Cell Biology, Cornell University, Ithaca, New York 14853;
| | - Anthony Bretscher
- Department of Molecular Biology and Genetics and Weill Institute for Molecular and Cell Biology, Cornell University, Ithaca, New York 14853;
| |
Collapse
|
19
|
Sauvanet C, Garbett D, Bretscher A. The function and dynamics of the apical scaffolding protein E3KARP are regulated by cell-cycle phosphorylation. Mol Biol Cell 2015; 26:3615-27. [PMID: 26310448 PMCID: PMC4603932 DOI: 10.1091/mbc.e15-07-0498] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 08/18/2015] [Indexed: 11/11/2022] Open
Abstract
We examine the dynamics and function of the apical scaffolding protein E3KARP/NHERF2, which consists of two PDZ domains and a tail containing an ezrin-binding domain. The exchange rate of E3KARP is greatly enhanced during mitosis due to phosphorylation at Ser-303 in its tail region. Whereas E3KARP can substitute for the function of the closely related scaffolding protein EBP50/NHERF1 in the formation of interphase microvilli, E3KARP S303D cannot. Moreover, the S303D mutation enhances the in vivo dynamics of the E3KARP tail alone, whereas in vitro the interaction of E3KARP with active ezrin is unaffected by S303D, implicating another factor regulating dynamics in vivo. A-Raf is found to be required for S303 phosphorylation in mitotic cells. Regulation of the dynamics of EBP50 is known to be dependent on its tail region but modulated by PDZ domain occupancy, which is not the case for E3KARP. Of interest, in both cases, the mechanisms regulating dynamics involve the tails, which are the most diverged region of the paralogues and probably evolved independently after a gene duplication event that occurred early in vertebrate evolution.
Collapse
Affiliation(s)
- Cécile Sauvanet
- Department of Molecular Biology and Genetics, Weill Institute for Molecular and Cell Biology, Cornell University, Ithaca, NY 14853
| | - Damien Garbett
- Department of Molecular Biology and Genetics, Weill Institute for Molecular and Cell Biology, Cornell University, Ithaca, NY 14853
| | - Anthony Bretscher
- Department of Molecular Biology and Genetics, Weill Institute for Molecular and Cell Biology, Cornell University, Ithaca, NY 14853
| |
Collapse
|
20
|
Zhang XD, Xie JJ, Liao LD, Long L, Xie YM, Li EM, Xu LY. 12-O-Tetradecanoylphorbol-13-Acetate Induces Up-Regulated Transcription of Variant 1 but Not Variant 2 of VIL2 in Esophageal Squamous Cell Carcinoma Cells via ERK1/2/AP-1/Sp1 Signaling. PLoS One 2015; 10:e0124680. [PMID: 25915860 PMCID: PMC4411055 DOI: 10.1371/journal.pone.0124680] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 03/17/2015] [Indexed: 12/25/2022] Open
Abstract
The membrane-cytoskeleton link organizer ezrin may be the most "dramatic" tumor marker, being strongly over-expressed in nearly one-third of human malignancies. However, the molecular mechanisms of aberrant ezrin expression still need to be clarified. Ezrin, encoded by the VIL2 gene, has two transcript variants that differ in the transcriptional start site (TSS): V1 and V2. Both V1 and V2 encode the same protein. Here, we found that 12-O-tetradecanoylphorbol-13-acetate (TPA) induced over-expression of human VIL2 in esophageal squamous cell carcinoma (ESCC) cells. Furthermore, VIL2 V1 but not V2 was up-regulated after TPA stimulation in a time-dependent manner. AP-1 and Sp1 binding sites within the promoter region of VIL2 V1 acted not only as basal transcriptional elements but also as a composite TPA-responsive element (TRE) for the transcription of VIL2 V1. TPA stimulation enhanced c-Jun and Sp1 binding to the TRE via activation of the ERK1/2 pathway and increased protein levels of c-Jun, c-Fos, and Sp1, resulting in over-expression of VIL2 V1, whereas the MEK1/2 inhibitor U0126 blocked these events. Finally, we showed that TPA promoted the migration of ESCC cells whereas MEK1/2 inhibitor or ezrin silencing could partially inverse this alteration. Taken together, these results suggest that TPA is able to induce VIL2 V1 over-expression in ESCC cells by activating MEK/ERK1/2 signaling and increasing binding of Sp1 and c-Jun to the TRE of the VIL2 V1 promoter, and that VIL2 is an important TPA-induced effector.
Collapse
Affiliation(s)
- Xiao-Dan Zhang
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Medical College of Shantou University, Shantou 514041, Guangdong, P.R. China; Institute of Oncologic Pathology, Medical College of Shantou University, Shantou 514041, Guangdong, P.R. China
| | - Jian-Jun Xie
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Medical College of Shantou University, Shantou 514041, Guangdong, P.R. China; Department of Biochemistry and Molecular Biology, Medical College of Shantou University, Shantou 514041, Guangdong, P.R. China
| | - Lian-Di Liao
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Medical College of Shantou University, Shantou 514041, Guangdong, P.R. China; Institute of Oncologic Pathology, Medical College of Shantou University, Shantou 514041, Guangdong, P.R. China
| | - Lin Long
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Medical College of Shantou University, Shantou 514041, Guangdong, P.R. China; Department of Biochemistry and Molecular Biology, Medical College of Shantou University, Shantou 514041, Guangdong, P.R. China
| | - Yang-Min Xie
- Department of Experimental Animal Center, Medical College of Shantou University, Shantou 515041, P. R. China
| | - En-Min Li
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Medical College of Shantou University, Shantou 514041, Guangdong, P.R. China; Department of Biochemistry and Molecular Biology, Medical College of Shantou University, Shantou 514041, Guangdong, P.R. China
| | - Li-Yan Xu
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Medical College of Shantou University, Shantou 514041, Guangdong, P.R. China; Institute of Oncologic Pathology, Medical College of Shantou University, Shantou 514041, Guangdong, P.R. China
| |
Collapse
|
21
|
Klingner C, Cherian AV, Fels J, Diesinger PM, Aufschnaiter R, Maghelli N, Keil T, Beck G, Tolić-Nørrelykke IM, Bathe M, Wedlich-Soldner R. Isotropic actomyosin dynamics promote organization of the apical cell cortex in epithelial cells. ACTA ACUST UNITED AC 2015; 207:107-21. [PMID: 25313407 PMCID: PMC4195824 DOI: 10.1083/jcb.201402037] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Apical membrane organization of nonconfluent epithelial cells is driven by a dynamic network of actin and myosin II filaments. Although cortical actin plays an important role in cellular mechanics and morphogenesis, there is surprisingly little information on cortex organization at the apical surface of cells. In this paper, we characterize organization and dynamics of microvilli (MV) and a previously unappreciated actomyosin network at the apical surface of Madin–Darby canine kidney cells. In contrast to short and static MV in confluent cells, the apical surfaces of nonconfluent epithelial cells (ECs) form highly dynamic protrusions, which are often oriented along the plane of the membrane. These dynamic MV exhibit complex and spatially correlated reorganization, which is dependent on myosin II activity. Surprisingly, myosin II is organized into an extensive network of filaments spanning the entire apical membrane in nonconfluent ECs. Dynamic MV, myosin filaments, and their associated actin filaments form an interconnected, prestressed network. Interestingly, this network regulates lateral mobility of apical membrane probes such as integrins or epidermal growth factor receptors, suggesting that coordinated actomyosin dynamics contributes to apical cell membrane organization.
Collapse
Affiliation(s)
- Christoph Klingner
- Cellular Dynamics and Cell Patterning and Department of Molecular Structural Biology, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany Institute of Cell Dynamics and Imaging and Cells-In-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, 48149 Münster, Germany Institute of Cell Dynamics and Imaging and Cells-In-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, 48149 Münster, Germany
| | - Anoop V Cherian
- Cellular Dynamics and Cell Patterning and Department of Molecular Structural Biology, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Johannes Fels
- Institute of Cell Dynamics and Imaging and Cells-In-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, 48149 Münster, Germany Institute of Cell Dynamics and Imaging and Cells-In-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, 48149 Münster, Germany
| | - Philipp M Diesinger
- Laboratory for Computational Biology & Biophysics, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Roland Aufschnaiter
- Cellular Dynamics and Cell Patterning and Department of Molecular Structural Biology, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany Institute of Cell Dynamics and Imaging and Cells-In-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, 48149 Münster, Germany Institute of Cell Dynamics and Imaging and Cells-In-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, 48149 Münster, Germany
| | - Nicola Maghelli
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Thomas Keil
- Cellular Dynamics and Cell Patterning and Department of Molecular Structural Biology, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Gisela Beck
- Cellular Dynamics and Cell Patterning and Department of Molecular Structural Biology, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany Institute of Cell Dynamics and Imaging and Cells-In-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, 48149 Münster, Germany Institute of Cell Dynamics and Imaging and Cells-In-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, 48149 Münster, Germany
| | - Iva M Tolić-Nørrelykke
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany Division of Molecular Biology, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| | - Mark Bathe
- Laboratory for Computational Biology & Biophysics, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Roland Wedlich-Soldner
- Cellular Dynamics and Cell Patterning and Department of Molecular Structural Biology, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany Institute of Cell Dynamics and Imaging and Cells-In-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, 48149 Münster, Germany Institute of Cell Dynamics and Imaging and Cells-In-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, 48149 Münster, Germany
| |
Collapse
|
22
|
Yeruva S, Chodisetti G, Luo M, Chen M, Cinar A, Ludolph L, Lünnemann M, Goldstein J, Singh AK, Riederer B, Bachmann O, Bleich A, Gereke M, Bruder D, Hagen S, He P, Yun C, Seidler U. Evidence for a causal link between adaptor protein PDZK1 downregulation and Na⁺/H⁺ exchanger NHE3 dysfunction in human and murine colitis. Pflugers Arch 2014; 467:1795-807. [PMID: 25271043 PMCID: PMC4383727 DOI: 10.1007/s00424-014-1608-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2014] [Revised: 09/02/2014] [Accepted: 09/03/2014] [Indexed: 12/13/2022]
Abstract
A dysfunction of the Na(+)/H(+) exchanger isoform 3 (NHE3) significantly contributes to the reduced salt absorptive capacity of the inflamed intestine. We previously reported a strong decrease in the NHERF family member PDZK1 (NHERF3), which binds to NHE3 and regulates its function in a mouse model of colitis. The present study investigates whether a causal relationship exists between the decreased PDZK1 expression and the NHE3 dysfunction in human and murine intestinal inflammation. Biopsies from the colon of patients with ulcerative colitis, murine inflamed ileal and colonic mucosa, NHE3-transfected Caco-2BBe colonic cells with short hairpin RNA (shRNA) knockdown of PDZK1, and Pdzk1-gene-deleted mice were studied. PDZK1 mRNA and protein expression was strongly decreased in inflamed human and murine intestinal tissue as compared to inactive disease or control tissue, whereas that of NHE3 or NHERF1 was not. Inflamed human and murine intestinal tissues displayed correct brush border localization of NHE3 but reduced acid-activated NHE3 transport activity. A similar NHE3 transport defect was observed when PDZK1 protein content was decreased by shRNA knockdown in Caco-2BBe cells or when enterocyte PDZK1 protein content was decreased to similar levels as found in inflamed mucosa by heterozygote breeding of Pdzk1-gene-deleted and WT mice. We conclude that a decrease in PDZK1 expression, whether induced by inflammation, shRNA-mediated knockdown, or heterozygous breeding, is associated with a decreased NHE3 transport rate in human and murine enterocytes. We therefore hypothesize that inflammation-induced loss of PDZK1 expression may contribute to the NHE3 dysfunction observed in the inflamed intestine.
Collapse
Affiliation(s)
- Sunil Yeruva
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Dynamics of ezrin and EBP50 in regulating microvilli on the apical aspect of epithelial cells. Biochem Soc Trans 2014; 42:189-94. [PMID: 24450650 DOI: 10.1042/bst20130263] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Microvilli are found on the apical surface of epithelial cells. Recent studies on the microvillar proteins ezrin and EBP50 (ezrin/radixin/moesin-binding phosphoprotein of 50 kDa) have revealed both the dynamics and the regulation of microvillar components, and how a dynamic ezrin phosphocycle is necessary to confine microvilli to the apical membrane. In the present review, we first summarize the background to allow us to place these advances in context.
Collapse
|
24
|
Abstract
The cell cortex is a dynamic and heterogeneous structure that governs cell identity and behavior. The ERM proteins (ezrin, radixin and moesin) are major architects of the cell cortex, and they link plasma membrane phospholipids and proteins to the underlying cortical actin cytoskeleton. Recent studies in several model systems have uncovered surprisingly dynamic and complex molecular activities of the ERM proteins and have provided new mechanistic insight into how they build and maintain cortical domains. Among many well-established and essential functions of ERM proteins, this Cell Science at a Glance article and accompanying poster will focus on the role of ERMs in organizing the cell cortex during cell division and apical morphogenesis. These examples highlight an emerging appreciation that the ERM proteins both locally alter the mechanical properties of the cell cortex, and control the spatial distribution and activity of key membrane complexes, establishing the ERM proteins as a nexus for the physical and functional organization of the cell cortex and making it clear that they are much more than scaffolds. This article is part of a Minifocus on Establishing polarity.
Collapse
Affiliation(s)
- Andrea I McClatchey
- Massachusetts General Hospital Center for Cancer Research, Harvard Medical School Department of Pathology, 149 13th Street, Charlestown, MA 02129, USA
| |
Collapse
|
25
|
Yang J, Singh V, Chen TE, Sarker R, Xiong L, Cha B, Jin S, Li X, Tse CM, Zachos NC, Donowitz M. NHERF2/NHERF3 protein heterodimerization and macrocomplex formation are required for the inhibition of NHE3 activity by carbachol. J Biol Chem 2014; 289:20039-53. [PMID: 24867958 DOI: 10.1074/jbc.m114.562413] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
NHERF1, NHERF2, and NHERF3 belong to the NHERF (Na(+)/H(+) exchanger regulatory factor) family of PSD-95/Discs-large/ZO-1 (PDZ) scaffolding proteins. Individually, each NHERF protein has been shown to be involved in the regulation of multiple receptors or transporters including Na(+)/H(+) exchanger 3 (NHE3). Although NHERF dimerizations have been reported, results have been inconsistent, and the physiological function of NHERF dimerizations is still unknown. The current study semiquantitatively compared the interaction strength among all possible homodimerizations and heterodimerizations of these three NHERF proteins by pulldown and co-immunoprecipitation assays. Both methods showed that NHERF2 and NHERF3 heterodimerize as the strongest interaction among all NHERF dimerizations. In vivo NHERF2/NHERF3 heterodimerization was confirmed by FRET and FRAP (fluorescence recovery after photobleach). NHERF2/NHERF3 heterodimerization is mediated by PDZ domains of NHERF2 and the C-terminal PDZ domain recognition motif of NHERF3. The NHERF3-4A mutant is defective in heterodimerization with NHERF2 and does not support the inhibition of NHE3 by carbachol. This suggests a role for NHERF2/NHERF3 heterodimerization in the regulation of NHE3 activity. In addition, both PDZ domains of NHERF2 could be simultaneously occupied by NHERF3 and another ligand such as NHE3, α-actinin-4, and PKCα, promoting formation of NHE3 macrocomplexes. This study suggests that NHERF2/NHERF3 heterodimerization mediates the formation of NHE3 macrocomplexes, which are required for the inhibition of NHE3 activity by carbachol.
Collapse
Affiliation(s)
- Jianbo Yang
- From the Department of Medicine, Division of Gastroenterology and
| | - Varsha Singh
- From the Department of Medicine, Division of Gastroenterology and
| | - Tian-E Chen
- From the Department of Medicine, Division of Gastroenterology and
| | - Rafiquel Sarker
- From the Department of Medicine, Division of Gastroenterology and
| | - Lishou Xiong
- From the Department of Medicine, Division of Gastroenterology and
| | - Boyoung Cha
- From the Department of Medicine, Division of Gastroenterology and
| | - Shi Jin
- From the Department of Medicine, Division of Gastroenterology and
| | - Xuhang Li
- From the Department of Medicine, Division of Gastroenterology and
| | - C Ming Tse
- From the Department of Medicine, Division of Gastroenterology and
| | | | - Mark Donowitz
- From the Department of Medicine, Division of Gastroenterology and Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| |
Collapse
|
26
|
Braunger JA, Brückner BR, Nehls S, Pietuch A, Gerke V, Mey I, Janshoff A, Steinem C. Phosphatidylinositol 4,5-bisphosphate alters the number of attachment sites between ezrin and actin filaments: a colloidal probe study. J Biol Chem 2014; 289:9833-43. [PMID: 24500715 DOI: 10.1074/jbc.m113.530659] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Direct linkage between the plasma membrane and the actin cytoskeleton is controlled by the protein ezrin, a member of the ezrin-radixin-moesin protein family. To function as a membrane-cytoskeleton linker, ezrin needs to be activated in a process that involves binding of ezrin to phosphatidylinositol 4,5-bisphosphate (PIP2) and phosphorylation of a conserved threonine residue. Here, we used colloidal probe microscopy to quantitatively analyze the interaction between ezrin and F-actin as a function of these activating factors. We show that the measured individual unbinding forces between ezrin and F-actin are independent of the activating parameters, in the range of approximately 50 piconewtons. However, the cumulative adhesion energy greatly increases in the presence of PIP2 demonstrating that a larger number of bonds between ezrin and F-actin has formed. In contrast, the phosphorylation state, represented by phosphor-mimetic mutants of ezrin, only plays a minor role in the activation process. These results are in line with in vivo experiments demonstrating that an increase in PIP2 concentration recruits more ezrin to the apical plasma membrane of polarized cells and significantly increases the membrane tension serving as a measure of the adhesion sites between the plasma membrane and the F-actin network.
Collapse
Affiliation(s)
- Julia A Braunger
- From the Institute of Organic and Biomolecular Chemistry, University of Göttingen, Tammannstrasse 2, 37077 Göttingen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Dhekne HS, Hsiao NH, Roelofs P, Kumari M, Slim CL, Rings EHHM, van Ijzendoorn SCD. Myosin Vb and Rab11a regulate phosphorylation of ezrin in enterocytes. J Cell Sci 2014; 127:1007-17. [PMID: 24413175 DOI: 10.1242/jcs.137273] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Microvilli at the apical surface of enterocytes allow the efficient absorption of nutrients in the intestine. Ezrin activation by its phosphorylation at T567 is important for microvilli development, but how such ezrin phosphorylation is controlled is not well understood. We demonstrate that a subset of kinases that phosphorylate ezrin closely co-distributes with apical recycling endosome marker Rab11a in the subapical domain. Expression of dominant-negative Rab11a mutant or depletion of the Rab11a-binding motor protein myosin Vb prevents the subapical enrichment of Rab11a and these kinases and inhibits ezrin phosphorylation and microvilli development, without affecting the polarized distribution of ezrin itself. We observe a similar loss of the subapical enrichment of Rab11a and the kinases and reduced phosphorylation of ezrin in microvillus inclusion disease, which is associated with MYO5B mutations, intestinal microvilli atrophy and malabsorption. Thus, part of the machinery for ezrin activation depends on recycling endosomes controlled by myosin Vb and Rab11a which, we propose, might act as subapical signaling platforms that enterocytes use to regulate development of microvilli and maintain human intestinal function.
Collapse
Affiliation(s)
- Herschel S Dhekne
- Department of Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
28
|
Viswanatha R, Wayt J, Ohouo PY, Smolka MB, Bretscher A. Interactome analysis reveals ezrin can adopt multiple conformational states. J Biol Chem 2013; 288:35437-51. [PMID: 24151071 PMCID: PMC3853291 DOI: 10.1074/jbc.m113.505669] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Revised: 10/18/2013] [Indexed: 12/16/2022] Open
Abstract
Ezrin, a member of the ezrin-radixin-moesin family (ERM), is an essential regulator of the structure of microvilli on the apical aspect of epithelial cells. Ezrin provides a linkage between membrane-associated proteins and F-actin, oscillating between active/open and inactive/closed states, and is regulated in part by phosphorylation of a C-terminal threonine. In the open state, ezrin can bind a number of ligands, but in the closed state the ligand-binding sites are inaccessible. In vitro analysis has proposed that there may be a third hyperactivated form of ezrin. To gain a better understanding of ezrin, we conducted an unbiased proteomic analysis of ezrin-binding proteins in an epithelial cell line, Jeg-3. We refined our list of interactors by comparing the interactomes using quantitative mass spectrometry between wild-type ezrin, closed ezrin, open ezrin, and hyperactivated ezrin. The analysis reveals several novel interactors confirmed by their localization to microvilli, as well as a significant class of proteins that bind closed ezrin. Taken together, the data indicate that ezrin can exist in three different conformational states, and different ligands "perceive" ezrin conformational states differently.
Collapse
Affiliation(s)
- Raghuvir Viswanatha
- From the Department of Molecular Biology and Genetics and Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York 14853
| | - Jessica Wayt
- From the Department of Molecular Biology and Genetics and Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York 14853
| | - Patrice Y. Ohouo
- From the Department of Molecular Biology and Genetics and Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York 14853
| | - Marcus B. Smolka
- From the Department of Molecular Biology and Genetics and Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York 14853
| | - Anthony Bretscher
- From the Department of Molecular Biology and Genetics and Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York 14853
| |
Collapse
|
29
|
Antelmi E, Cardone RA, Greco MR, Rubino R, Di Sole F, Martino NA, Casavola V, Carcangiu M, Moro L, Reshkin SJ. ß1 integrin binding phosphorylates ezrin at T567 to activate a lipid raft signalsome driving invadopodia activity and invasion. PLoS One 2013; 8:e75113. [PMID: 24086451 PMCID: PMC3782503 DOI: 10.1371/journal.pone.0075113] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Accepted: 08/09/2013] [Indexed: 01/11/2023] Open
Abstract
Extracellular matrix (ECM) degradation is a critical process in tumor cell invasion and requires matrix degrading protrusions called invadopodia. The Na+/H+ exchanger (NHE1) has recently been shown to be fundamental in the regulation of invadopodia actin cytoskeleton dynamics and activity. However, the structural link between the invadopodia cytoskeleton and NHE1 is still unknown. A candidate could be ezrin, a linker between the NHE1 and the actin cytoskeleton known to play a pivotal role in invasion and metastasis. However, the mechanistic basis for its role remains unknown. Here, we demonstrate that ezrin phosphorylated at T567 is highly overexpressed in the membrane of human breast tumors and positively associated with invasive growth and HER2 overexpression. Further, in the metastatic cell line, MDA-MB-231, p-ezrin was almost exclusively expressed in invadopodia lipid rafts where it co-localized in a functional complex with NHE1, EGFR, ß1-integrin and phosphorylated-NHERF1. Manipulation by mutation of ezrins T567 phosphorylation state and/or PIP2 binding capacity or of NHE1s binding to ezrin or PIP2 demonstrated that p-ezrin expression and binding to PIP2 are required for invadopodia-mediated ECM degradation and invasion and identified NHE1 as the membrane protein that p-ezrin regulates to induce invadopodia formation and activity.
Collapse
Affiliation(s)
- Ester Antelmi
- Department of Bioscience, Biotechnology and Biopharmacologics, University of Bari, Bari, Italy
- Department of Pathology, Anatomic Pathology A Unit, Istituto Nazionale Tumori, Milan, Italy
| | - Rosa A. Cardone
- Department of Bioscience, Biotechnology and Biopharmacologics, University of Bari, Bari, Italy
| | - Maria R. Greco
- Department of Bioscience, Biotechnology and Biopharmacologics, University of Bari, Bari, Italy
| | - Rosa Rubino
- Department of Bioscience, Biotechnology and Biopharmacologics, University of Bari, Bari, Italy
| | - Francesca Di Sole
- Department of Medicine, University of Maryland School of Medicine and the Medical Service, Department of Veterans Affairs Medical Center, Baltimore, Maryland, United States of America
| | - Nicola A. Martino
- Department of Animal Production, Faculty of Biotechnological Sciences, University of Bari, Bari, Italy
| | - Valeria Casavola
- Department of Bioscience, Biotechnology and Biopharmacologics, University of Bari, Bari, Italy
| | - MariaLuisa Carcangiu
- Department of Pathology, Anatomic Pathology A Unit, Istituto Nazionale Tumori, Milan, Italy
| | - Loredana Moro
- Institute of Biomembranes and Bioenergetics (IBBE), CNR, Bari, Italy
| | - Stephan J. Reshkin
- Department of Bioscience, Biotechnology and Biopharmacologics, University of Bari, Bari, Italy
- * E-mail:
| |
Collapse
|
30
|
Garbett D, Sauvanet C, Viswanatha R, Bretscher A. The tails of apical scaffolding proteins EBP50 and E3KARP regulate their localization and dynamics. Mol Biol Cell 2013; 24:3381-92. [PMID: 23985317 PMCID: PMC3814156 DOI: 10.1091/mbc.e13-06-0330] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
ERM-binding protein of 50 kDa (EBP50) and NHE3 kinase A regulatory protein (E3KARP) are closely related but show dramatically different dynamics in microvilli. The high dynamics of EBP50 is determined by a region in its tail and is inhibited by its PDZ domains, but is activated upon PDZ ligand binding. Proteomic analysis of the effects of EBP50 dynamics identifies a novel PDZ binding partner, IRSp53. The closely related apical scaffolding proteins ERM-binding phosphoprotein of 50 kDa (EBP50) and NHE3 kinase A regulatory protein (E3KARP) both consist of two postsynaptic density 95/disks large/zona occludens-1 (PDZ) domains and a tail ending in an ezrin-binding domain. Scaffolding proteins are thought to provide stable linkages between components of multiprotein complexes, yet in several types of epithelial cells, EBP50, but not E3KARP, shows rapid exchange from microvilli compared with its binding partners. The difference in dynamics is determined by the proteins’ tail regions. Exchange rates of EBP50 and E3KARP correlated strongly with their abilities to precipitate ezrin in vivo. The EBP50 tail alone is highly dynamic, but in the context of the full-length protein, the dynamics is lost when the PDZ domains are unable to bind ligand. Proteomic analysis of the effects of EBP50 dynamics on binding-partner preferences identified a novel PDZ1 binding partner, the I-BAR protein insulin receptor substrate p53 (IRSp53). Additionally, the tails promote different microvillar localizations for EBP50 and E3KARP, which localized along the full length and to the base of microvilli, respectively. Thus the tails define the localization and dynamics of these scaffolding proteins, and the high dynamics of EBP50 is regulated by the occupancy of its PDZ domains.
Collapse
Affiliation(s)
- Damien Garbett
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853
| | | | | | | |
Collapse
|
31
|
Pelaseyed T, Gustafsson JK, Gustafsson IJ, Ermund A, Hansson GC. Carbachol-induced MUC17 endocytosis is concomitant with NHE3 internalization and CFTR membrane recruitment in enterocytes. Am J Physiol Cell Physiol 2013; 305:C457-67. [PMID: 23784542 DOI: 10.1152/ajpcell.00141.2013] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
We have reported that transmembrane mucin MUC17 binds PDZ protein PDZK1, which retains MUC17 apically in enterocytes. MUC17 and transmembrane mucins MUC3 and MUC12 are suggested to build the enterocyte apical glycocalyx. Carbachol (CCh) stimulation of the small intestine results in gel-forming mucin secretion from goblet cells, something that requires adjacent enterocytes to secrete chloride and bicarbonate for proper mucin formation. Surface labeling and confocal imaging demonstrated that apically expressed MUC17 in Caco-2 cells and Muc3(17) in murine enterocytes were endocytosed upon stimulation with CCh. Relocation of MUC17 in response to CCh was specific as MUC3 and MUC12 did not relocate following CCh stimulation. MUC17 colocalized with PDZK1 under basal conditions, while MUC17 relocated to the terminal web and into early endosomes after CCh stimulation. CCh stimulation concomitantly internalized the Na(+/)H(+) exchanger 3 (NHE3) and recruited cystic fibrosis transmembrane conductance regulator (CFTR) to the apical membranes, a process that was important for CFTR-mediated bicarbonate secretion necessary for proper gel-forming mucin unfolding. The reason for the specific internalization of MUC17 is not understood, but it could limit the diffusion barrier for ion secretion caused by the apical enterocyte glycocalyx or alternatively act to sample luminal bacteria. Our results reveal well-orchestrated mucus secretion and trafficking of ion channels and the MUC17 mucin.
Collapse
Affiliation(s)
- Thaher Pelaseyed
- Department of Medical Biochemistry, University of Gothenburg, Gothenburg, Sweden
| | | | | | | | | |
Collapse
|
32
|
Jiang L, Phang JM, Yu J, Harrop SJ, Sokolova AV, Duff AP, Wilk KE, Alkhamici H, Breit SN, Valenzuela SM, Brown LJ, Curmi PMG. CLIC proteins, ezrin, radixin, moesin and the coupling of membranes to the actin cytoskeleton: a smoking gun? BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2013; 1838:643-57. [PMID: 23732235 DOI: 10.1016/j.bbamem.2013.05.025] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Revised: 05/20/2013] [Accepted: 05/21/2013] [Indexed: 12/20/2022]
Abstract
The CLIC proteins are a highly conserved family of metazoan proteins with the unusual ability to adopt both soluble and integral membrane forms. The physiological functions of CLIC proteins may include enzymatic activity in the soluble form and anion channel activity in the integral membrane form. CLIC proteins are associated with the ERM proteins: ezrin, radixin and moesin. ERM proteins act as cross-linkers between membranes and the cortical actin cytoskeleton. Both CLIC and ERM proteins are controlled by Rho family small GTPases. CLIC proteins, ERM and Rho GTPases act in a concerted manner to control active membrane processes including the maintenance of microvillar structures, phagocytosis and vesicle trafficking. All of these processes involve the interaction of membranes with the underlying cortical actin cytoskeleton. The relationships between Rho GTPases, CLIC proteins, ERM proteins and the membrane:actin cytoskeleton interface are reviewed. Speculative models are proposed involving the formation of localised multi-protein complexes on the membrane surface that assemble via multiple weak interactions. This article is part of a Special Issue entitled: Reciprocal influences between cell cytoskeleton and membrane channels, receptors and transporters. Guest Editor: Jean Claude Hervé.
Collapse
Affiliation(s)
- Lele Jiang
- St Vincent's Centre for Applied Medical Research, St Vincent's Hospital, Sydney, NSW 2010, Australia
| | - Juanita M Phang
- School of Physics, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Jiang Yu
- School of Physics, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Stephen J Harrop
- School of Physics, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Anna V Sokolova
- Australian Nuclear Science and Technology Organisation, Lucas Heights, NSW, Australia
| | - Anthony P Duff
- Australian Nuclear Science and Technology Organisation, Lucas Heights, NSW, Australia
| | - Krystyna E Wilk
- School of Physics, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Heba Alkhamici
- School of Medical and Molecular Biosciences, The University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Samuel N Breit
- St Vincent's Centre for Applied Medical Research, St Vincent's Hospital, Sydney, NSW 2010, Australia
| | - Stella M Valenzuela
- School of Medical and Molecular Biosciences, The University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Louise J Brown
- Department of Chemistry and Biomolecular Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Paul M G Curmi
- St Vincent's Centre for Applied Medical Research, St Vincent's Hospital, Sydney, NSW 2010, Australia; School of Physics, The University of New South Wales, Sydney, NSW 2052, Australia.
| |
Collapse
|
33
|
Abstract
Mucins--large, highly glycosylated proteins--are important for the luminal protection of the gastrointestinal tract. Enterocytes have their apical surface covered by transmembrane mucins and goblet cells produce the secreted gel-forming mucins that form mucus. The small intestine has a single unattached mucus layer, which in cystic fibrosis becomes attached, accounting for the intestinal manifestations of this disease. The stomach and colon have two layers of mucus; the inner layer is attached and the outer layer is less dense and unattached. In the colon, the outer mucus layer is the habitat for commensal bacteria. The inner mucus layer is impervious to bacteria and is renewed every hour by surface goblet cells. The crypt goblet cells have the ability to restitute the mucus layer by secretion, for example after an ischaemic challenge. Proteases of certain parasites and some bacteria can cleave mucins and dissolve the mucus as part of their pathogenicity. The inner mucus layer can, however, also become penetrable to bacteria by several other mechanisms, including aberrations in the immune system. When bacteria reach the epithelial surface, the immune system is activated and inflammation is triggered. This mechanism might occur in some types of ulcerative colitis.
Collapse
|
34
|
Yang J, Singh V, Cha B, Chen TE, Sarker R, Murtazina R, Jin S, Zachos NC, Patterson GH, Tse CM, Kovbasnjuk O, Li X, Donowitz M. NHERF2 protein mobility rate is determined by a unique C-terminal domain that is also necessary for its regulation of NHE3 protein in OK cells. J Biol Chem 2013; 288:16960-16974. [PMID: 23612977 DOI: 10.1074/jbc.m113.470799] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Na(+)/H(+) exchanger regulatory factor (NHERF) proteins are a family of PSD-95/Discs-large/ZO-1 (PDZ)-scaffolding proteins, three of which (NHERFs 1-3) are localized to the brush border in kidney and intestinal epithelial cells. All NHERF proteins are involved in anchoring membrane proteins that contain PDZ recognition motifs to form multiprotein signaling complexes. In contrast to their predicted immobility, NHERF1, NHERF2, and NHERF3 were all shown by fluorescence recovery after photobleaching/confocal microscopy to be surprisingly mobile in the microvilli of the renal proximal tubule OK cell line. Their diffusion coefficients, although different among the three, were all of the same magnitude as that of the transmembrane proteins, suggesting they are all anchored in the microvilli but to different extents. NHERF3 moves faster than NHERF1, and NHERF2 moves the slowest. Several chimeras and mutants of NHERF1 and NHERF2 were made to determine which part of NHERF2 confers the slower mobility rate. Surprisingly, the slower mobility rate of NHERF2 was determined by a unique C-terminal domain, which includes a nonconserved region along with the ezrin, radixin, moesin (ERM) binding domain. Also, this C-terminal domain of NHERF2 determined its greater detergent insolubility and was necessary for the formation of larger multiprotein NHERF2 complexes. In addition, this NHERF2 domain was functionally significant in NHE3 regulation, being necessary for stimulation by lysophosphatidic acid of activity and increased mobility of NHE3, as well as necessary for inhibition of NHE3 activity by calcium ionophore 4-Br-A23187. Thus, multiple functions of NHERF2 require involvement of an additional domain in this protein.
Collapse
Affiliation(s)
- Jianbo Yang
- Department of Medicine, Division of Gastroenterology, Baltimore, Maryland 21205
| | - Varsha Singh
- Department of Medicine, Division of Gastroenterology, Baltimore, Maryland 21205
| | - Boyoung Cha
- Department of Medicine, Division of Gastroenterology, Baltimore, Maryland 21205
| | - Tian-E Chen
- Department of Medicine, Division of Gastroenterology, Baltimore, Maryland 21205
| | - Rafiquel Sarker
- Department of Medicine, Division of Gastroenterology, Baltimore, Maryland 21205
| | - Rakhilya Murtazina
- Department of Medicine, Division of Gastroenterology, Baltimore, Maryland 21205
| | - Shi Jin
- Department of Medicine, Division of Gastroenterology, Baltimore, Maryland 21205
| | - Nicholas C Zachos
- Department of Medicine, Division of Gastroenterology, Baltimore, Maryland 21205
| | - George H Patterson
- Biophotonics Section, National Institute of Biomedical Imaging and Bioengineering, NIH, Bethesda, Maryland 20892
| | - C Ming Tse
- Department of Medicine, Division of Gastroenterology, Baltimore, Maryland 21205
| | - Olga Kovbasnjuk
- Department of Medicine, Division of Gastroenterology, Baltimore, Maryland 21205
| | - Xuhang Li
- Department of Medicine, Division of Gastroenterology, Baltimore, Maryland 21205
| | - Mark Donowitz
- Department of Medicine, Division of Gastroenterology, Baltimore, Maryland 21205; Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205.
| |
Collapse
|
35
|
Ligand-induced dynamic changes in extended PDZ domains from NHERF1. J Mol Biol 2013; 425:2509-28. [PMID: 23583913 DOI: 10.1016/j.jmb.2013.04.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2012] [Revised: 03/31/2013] [Accepted: 04/03/2013] [Indexed: 01/09/2023]
Abstract
The multi-domain scaffolding protein NHERF1 modulates the assembly and intracellular trafficking of various transmembrane receptors and ion-transport proteins. The two PDZ (postsynaptic density 95/disk large/zonula occluden 1) domains of NHERF1 possess very different ligand-binding capabilities: PDZ1 recognizes a variety of membrane proteins with high affinity, while PDZ2 only binds limited number of target proteins. Here using NMR, we have determined the structural and dynamic mechanisms that differentiate the binding affinities of the two PDZ domains, for the type 1 PDZ-binding motif (QDTRL) in the carboxyl terminus of cystic fibrosis transmembrane regulator. Similar to PDZ2, we have identified a helix-loop-helix subdomain coupled to the canonical PDZ1 domain. The extended PDZ1 domain is highly flexible with correlated backbone motions on fast and slow timescales, while the extended PDZ2 domain is relatively rigid. The malleability of the extended PDZ1 structure facilitates the transmission of conformational changes at the ligand-binding site to the remote helix-loop-helix extension. By contrast, ligand binding has only modest effects on the conformation and dynamics of the extended PDZ2 domain. The study shows that ligand-induced structural and dynamic changes coupled with sequence variation at the putative PDZ binding site dictate ligand selectivity and binding affinity of the two PDZ domains of NHERF1.
Collapse
|
36
|
|
37
|
Li X, Xu WM, Yin TL, Zhao QH, Peng LY, Yang J. Temporal and spatial regulation of ezrin-radixin-moesin-binding phosphoprotein-50-kDa (EBP50) during embryo implantation in mouse uterus. Int J Mol Sci 2012. [PMID: 23208378 PMCID: PMC3546698 DOI: 10.3390/ijms131216418] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Embryo implantation is a crucial process for successful pregnancy. To date, the mechanism of embryo implantation remains unclear. Ezrin-radixin-moesin-binding protein-50-kDa (EBP50) is a scaffold protein, which has been shown to play an important role in cancer development. Embryo implantation and cancer follow a similar progression. Thus, in this article, we utilized immunohistochemical staining and western blot analyses to examine the spatiotemporal expression and regulation of EBP50 both in the mouse uterus during embryo implantation as well as in other related models. We found that EBP50 was detected in epithelial cells in all of the groups used in our study. During the peri-implantation period, EBP50 mainly localized in apical membranes. At the implantation site (IS) on day 5 (D5) of pregnancy, EBP50 was mainly expressed in the nuclei of stroma cells, whereas from day 6 to day 8 (D6–D8) of pregnancy, the expression of EBP50 was noted in the cytoplasm of decidual cells. The expression of EBP50 was not significantly different in the pseudopregnant uterus and decreased in the uteri subjected to activation of delayed implantation. Artificial decidualization also decreased EBP50 expression. Thus, the expression levels and location were affected by active blastocysts and decidualization during the window of implantation.
Collapse
Affiliation(s)
- Xing Li
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, China.
| | | | | | | | | | | |
Collapse
|
38
|
Viswanatha R, Ohouo PY, Smolka MB, Bretscher A. Local phosphocycling mediated by LOK/SLK restricts ezrin function to the apical aspect of epithelial cells. ACTA ACUST UNITED AC 2012; 199:969-84. [PMID: 23209304 PMCID: PMC3518218 DOI: 10.1083/jcb.201207047] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Local cycling of LOK/SLK-dependent phosphorylation of ezrin is required for its apical localization and for microvillus formation. In this paper, we describe how a dynamic regulatory process is necessary to restrict microvilli to the apical aspect of polarized epithelial cells. We found that local phosphocycling regulation of ezrin, a critical plasma membrane–cytoskeletal linker of microvilli, was required to restrict its function to the apical membrane. Proteomic approaches and ribonucleic acid interference knockdown identified lymphocyte-oriented kinase (LOK) and SLK as the relevant kinases. Using drug-resistant LOK and SLK variants showed that these kinases were sufficient to restrict ezrin function to the apical domain. Both kinases were enriched in microvilli and locally activated there. Unregulated kinase activity caused ezrin mislocalization toward the basolateral domain, whereas expression of the kinase regulatory regions of LOK or SLK resulted in local inhibition of ezrin phosphorylation by the endogenous kinases. Thus, the domain-specific presence of microvilli is a dynamic process requiring a localized kinase driving the phosphocycling of ezrin to continually bias its function to the apical membrane.
Collapse
Affiliation(s)
- Raghuvir Viswanatha
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA
| | | | | | | |
Collapse
|
39
|
Jayasundar JJ, Ju JH, He L, Liu D, Meilleur F, Zhao J, Callaway DJE, Bu Z. Open conformation of ezrin bound to phosphatidylinositol 4,5-bisphosphate and to F-actin revealed by neutron scattering. J Biol Chem 2012; 287:37119-33. [PMID: 22927432 PMCID: PMC3481312 DOI: 10.1074/jbc.m112.380972] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Revised: 08/22/2012] [Indexed: 11/06/2022] Open
Abstract
Ezrin is a member of the ezrin-radixin-moesin family (ERM) of adapter proteins that are localized at the interface between the cell membrane and the cortical actin cytoskeleton, and they regulate a variety of cellular functions. The structure representing a dormant and closed conformation of an ERM protein has previously been determined by x-ray crystallography. Here, using contrast variation small angle neutron scattering, we reveal the structural changes of the full-length ezrin upon binding to the signaling lipid phosphatidylinositol 4,5-bisphosphate (PIP(2)) and to F-actin. Ezrin binding to F-actin requires the simultaneous binding of ezrin to PIP(2). Once bound to F-actin, the opened ezrin forms more extensive contacts with F-actin than generally depicted, suggesting a possible role of ezrin in regulating the interfacial structure and dynamics between the cell membrane and the underlying actin cytoskeleton. In addition, using gel filtration, we find that the conformational opening of ezrin in response to PIP(2) binding is cooperative, but the cooperativity is disrupted by a phospho-mimic mutation S249D in the 4.1-ezrin/radixin/moesin (FERM) domain of ezrin. Using surface plasmon resonance, we show that the S249D mutation weakens the binding affinity and changes the kinetics of 4.1-ERM to PIP(2) binding. The study provides the first structural view of the activated ezrin bound to PIP(2) and to F-actin.
Collapse
Affiliation(s)
| | - Jeong Ho Ju
- From the Department of Chemistry, City College of New York, New York, New York 10031
| | - Lilin He
- the Center for Structural Molecular Biology and Chemical Sciences Division, Oak Ridge National Laboratory, Oak Ridge, Tennessee 37831
- the Neutron Sciences Directorate, Oak Ridge National Laboratory, Oak Ridge, Tennessee 37831
| | - Dazhi Liu
- the Neutron Sciences Directorate, Oak Ridge National Laboratory, Oak Ridge, Tennessee 37831
| | - Flora Meilleur
- the Neutron Sciences Directorate, Oak Ridge National Laboratory, Oak Ridge, Tennessee 37831
- the Department of Molecular and Structural Biochemistry, North Carolina State University, Raleigh, North Carolina 27695, and
| | - Jinkui Zhao
- the Neutron Sciences Directorate, Oak Ridge National Laboratory, Oak Ridge, Tennessee 37831
| | - David J. E. Callaway
- From the Department of Chemistry, City College of New York, New York, New York 10031
- the New York University School of Medicine, New York, New York 10016
| | - Zimei Bu
- From the Department of Chemistry, City College of New York, New York, New York 10031
| |
Collapse
|
40
|
Corcoran A, Cotter TG. FLT3-driven redox-modulation of Ezrin regulates leukaemic cell migration. Free Radic Res 2012; 47:20-34. [PMID: 23009217 DOI: 10.3109/10715762.2012.733385] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The concept of reactive oxygen species (ROS) being produced via the activation of specific oncogenes provides a basis for generating genomic instability and pro-survival signalling in tumour cells. The purpose of this study was to identify downstream targets of NADPH oxidase (Nox)-derived ROS signalling in acute myeloid leukaemia cells, by performing a proteomic analysis utilizing two-dimensional phosphotyrosine immunoblotting. The majority of the targets identified were cytoskeletal-associated proteins including Ezrin, a known regulator of the cytoskeleton, which was examined further. The study demonstrated that inhibition of Nox enzymes, using diphenyleneiodonium chloride in the acute myeloid leukaemia cell line MOLM-13, resulted in a decrease in Ezrin tyrosine phosphorylation and also triggered a shift in Ezrin sub-cellular localization as detected by immunofluorescence. The change in Ezrin localization coincided with altered cell morphology, observed using scanning electron microscopy and a decreased ability to migrate through a polycarbonate transwell membrane. Similar effects were observed upon inhibition of the oncogenic receptor tyrosine kinase FLT3 using the staurosporine derivate PKC412, implicating a role for FLT3 as an upstream regulator of Ezrin. Our results indicate that FLT3 drives production of ROS by Nox, which stimulates changes in Ezrin tyrosine phosphorylation and localization via redox regulation of Src. Furthermore, inhibition of FLT3 signalling leads to alterations in MOLM-13 cell morphology and has a significant influence on cell motility.
Collapse
Affiliation(s)
- Aoife Corcoran
- Tumour Biology Laboratory, Biochemistry Department, Biosciences Research Institute, University College Cork, Cork, Ireland
| | | |
Collapse
|
41
|
Garbett D, Bretscher A. PDZ interactions regulate rapid turnover of the scaffolding protein EBP50 in microvilli. ACTA ACUST UNITED AC 2012; 198:195-203. [PMID: 22801783 PMCID: PMC3410424 DOI: 10.1083/jcb.201204008] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Scaffolding proteins containing PDZ (postsynaptic density 95/discs large/zonula occludens-1) domains are believed to provide relatively stable linkages between components of macromolecular complexes and in some cases to bridge to the actin cytoskeleton. The microvillar scaffolding protein EBP50 (ERM-binding phosphoprotein of 50 kD), consisting of two PDZ domains and an ezrin-binding site, retains specific proteins in microvilli and is necessary for microvillar biogenesis. Our analysis of the dynamics of microvillar proteins in vivo indicated that ezrin and microvillar membrane proteins had dynamics consistent with actin treadmilling and microvillar lifetimes. However, EBP50 was highly dynamic, turning over within seconds. EBP50 turnover was reduced by mutations that inactivate its PDZ domains and was enhanced by protein kinase C phosphorylation. Using a novel in vitro photoactivation fluorescence assay, the EBP50-ezrin interaction was shown to have a slow off-rate that was dramatically enhanced in a PDZ-regulated manner by addition of cell extract to near in vivo levels. Thus, the linking of relatively stable microvillar components can be mediated by surprisingly dynamic EBP50, a finding that may have important ramifications for other scaffolding proteins.
Collapse
Affiliation(s)
- Damien Garbett
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA
| | | |
Collapse
|
42
|
Boratkó A, Gergely P, Csortos C. Cell cycle dependent association of EBP50 with protein phosphatase 2A in endothelial cells. PLoS One 2012; 7:e35595. [PMID: 22523604 PMCID: PMC3327649 DOI: 10.1371/journal.pone.0035595] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Accepted: 03/19/2012] [Indexed: 12/19/2022] Open
Abstract
Ezrin-radixin-moesin (ERM)-binding phosphoprotein 50 (EBP50) is a phosphorylatable PDZ domain-containing adaptor protein that is abundantly expressed in epithelium but was not yet studied in the endothelium. We report unusual nuclear localization of EBP50 in bovine pulmonary artery endothelial cells (BPAEC). Immunofluorescent staining and cellular fractionation demonstrated that EBP50 is present in the nuclear and perinuclear region in interphase cells. In the prophase of mitosis EBP50 redistributes to the cytoplasmic region in a phosphorylation dependent manner and during mitosis EBP50 co-localizes with protein phosphatase 2A (PP2A). Furthermore, in vitro wound healing of BPAEC expressing phospho-mimic mutant of EBP50 was accelerated indicating that EBP50 is involved in the regulation of the cell division. Cell cycle dependent specific interactions were detected between EBP50 and the subunits of PP2A (A, C, and Bα) with immunoprecipitation and pull-down experiments. The interaction of EBP50 with the Bα containing form of PP2A suggests that this holoenzyme of PP2A can be responsible for the dephosphorylation of EBP50 in cytokinesis. Moreover, the results underline the significance of EBP50 in cell division via reversible phosphorylation of the protein with cyclin dependent kinase and PP2A in normal cells.
Collapse
Affiliation(s)
- Anita Boratkó
- Department of Medical Chemistry, University of Debrecen Medical and Health Science Center, Debrecen, Hungary
| | - Pál Gergely
- Department of Medical Chemistry, University of Debrecen Medical and Health Science Center, Debrecen, Hungary
- Cell Biology and Signaling Research Group of the Hungarian Academy of Sciences, University of Debrecen Medical and Health Science Center, Debrecen, Hungary
| | - Csilla Csortos
- Department of Medical Chemistry, University of Debrecen Medical and Health Science Center, Debrecen, Hungary
- * E-mail:
| |
Collapse
|
43
|
Zwaenepoel I, Naba A, Da Cunha MML, Del Maestro L, Formstecher E, Louvard D, Arpin M. Ezrin regulates microvillus morphogenesis by promoting distinct activities of Eps8 proteins. Mol Biol Cell 2012; 23:1080-94. [PMID: 22262457 PMCID: PMC3302735 DOI: 10.1091/mbc.e11-07-0588] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The membrane cytoskeleton linker ezrin differentially regulates the activity of Eps8 and Eps8L1a in microvillar actin-F assembly. Eps8L1a displays F-actin capping activity, therefore controlling microvillus length, whereas, as previously shown, Eps8 displays bundling activity. The mechanisms that regulate actin filament polymerization resulting in the morphogenesis of the brush border microvilli in epithelial cells remain unknown. Eps8, the prototype of a family of proteins capable of capping and bundling actin filaments, has been shown to bundle the microvillar actin filaments. We report that Eps8L1a, a member of the Eps8 family and a novel ezrin-interacting partner, controls microvillus length through its capping activity. Depletion of Eps8L1a leads to the formation of long microvilli, whereas its overexpression has the opposite effect. We demonstrate that ezrin differentially modulates the actin-capping and -bundling activities of Eps8 and Eps8L1a during microvillus assembly. Coexpression of ezrin with Eps8 promotes the formation of membrane ruffles and tufts of microvilli, whereas expression of ezrin and Eps8L1a induces the clustering of actin-containing structures at the cell surface. These distinct morphological changes are neither observed when a mutant of ezrin defective in its binding to Eps8/Eps8L1a is coexpressed with Eps8 or Eps8L1a nor observed when ezrin is expressed with mutants of Eps8 or Eps8L1a defective in the actin-bundling or -capping activities, respectively. Our data show a synergistic effect of ezrin and Eps8 proteins in the assembly and organization of actin microvillar filaments.
Collapse
Affiliation(s)
- Ingrid Zwaenepoel
- Unité Mixte de Recherche 144, Centre National de la Recherche Scientifique/Institut Curie, 75248 Paris, France
| | | | | | | | | | | | | |
Collapse
|
44
|
Zhang J, Petit CM, King DS, Lee AL. Phosphorylation of a PDZ domain extension modulates binding affinity and interdomain interactions in postsynaptic density-95 (PSD-95) protein, a membrane-associated guanylate kinase (MAGUK). J Biol Chem 2011; 286:41776-41785. [PMID: 21965656 DOI: 10.1074/jbc.m111.272583] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Postsynaptic density-95 is a multidomain scaffolding protein that recruits glutamate receptors to postsynaptic sites and facilitates signal processing and connection to the cytoskeleton. It is the leading member of the membrane-associated guanylate kinase family of proteins, which are defined by the PSD-95/Discs large/ZO-1 (PDZ)-Src homology 3 (SH3)-guanylate kinase domain sequence. We used NMR to show that phosphorylation of conserved tyrosine 397, which occurs in vivo and is located in an atypical helical extension (α3), initiates a rapid equilibrium of docked and undocked conformations. Undocking reduced ligand binding affinity allosterically and weakened the interaction of PDZ3 with SH3 even though these domains are separated by a ~25-residue linker. Additional phosphorylation at two linker sites further disrupted the interaction, implicating α3 and the linker in tuning interdomain communication. These experiments revealed a novel mode of regulation by a detachable PDZ element and offer a first glimpse at the dynamic interaction of PDZ and SH3-guanylate kinase domains in membrane-associated guanylate kinases.
Collapse
Affiliation(s)
- Jun Zhang
- Department of Biochemistry and Biophysics, School of Medicine, University of North Carolina, Chapel Hill, North Carolina 27599
| | - Chad M Petit
- Department of Biochemistry and Biophysics, School of Medicine, University of North Carolina, Chapel Hill, North Carolina 27599
| | - David S King
- Howard Hughes Medical Institute Mass Spectrometry Laboratory and Department of Molecular and Cell Biology, University of California, Berkeley, California 94720
| | - Andrew L Lee
- Department of Biochemistry and Biophysics, School of Medicine, University of North Carolina, Chapel Hill, North Carolina 27599; Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599.
| |
Collapse
|
45
|
Ezrin-mediated apical integrity is required for intestinal homeostasis. Proc Natl Acad Sci U S A 2011; 108:11924-9. [PMID: 21730140 DOI: 10.1073/pnas.1103418108] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Individual cell types are defined by architecturally and functionally specialized cortical domains. The Ezrin, Radixin, and Moesin (ERM) proteins play a major role in organizing cortical domains by assembling membrane protein complexes and linking them to the cortical actin cytoskeleton. Many studies have focused on the individual roles of the ERM proteins in stabilizing the membrane-cytoskeleton interface, controlling the distribution and function of apical membrane complexes, regulating the small GTPase Rho, or establishing cell-cell junctions. We previously found that deletion of the mouse Ezrin gene yields severe defects in apical integrity throughout the developing intestinal epithelium, resulting in incomplete villus morphogenesis and neonatal death. However, the molecular function of Ezrin in building the apical surface of the intestinal epithelium was not clear. By deleting Ezrin in the adult mouse intestinal epithelium, we provide evidence that Ezrin performs multiple molecular functions that collaborate to build the functional apical surface of the intestinal epithelium in vivo. The loss of Ezrin-mediated apical integrity in the adult intestine yields severe morphological consequences during intestinal homeostasis, including defects in cell geometry, extrusion, junctional remodeling, and spindle orientation. Surprisingly, deletion of Ezrin either before or after villus morphogenesis yields villus fusion, revealing a previously unrecognized step in intestinal homeostasis. Our studies indicate that the function of Ezrin in building and maintaining the apical domain is essential not only for intestinal morphogenesis but also for homeostasis in the mature intestine.
Collapse
|
46
|
Lanzano L, Lei T, Okamura K, Giral H, Caldas Y, Masihzadeh O, Gratton E, Levi M, Blaine J. Differential modulation of the molecular dynamics of the type IIa and IIc sodium phosphate cotransporters by parathyroid hormone. Am J Physiol Cell Physiol 2011; 301:C850-61. [PMID: 21593452 DOI: 10.1152/ajpcell.00412.2010] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The kidney is a key regulator of phosphate homeostasis. There are two predominant renal sodium phosphate cotransporters, NaPi2a and NaPi2c. Both are regulated by parathyroid hormone (PTH), which decreases the abundance of the NaPi cotransporters in the apical membrane of renal proximal tubule cells. The time course of PTH-induced removal of the two cotransporters from the apical membrane, however, is markedly different for NaPi2a compared with NaPi2c. In animals and in cell culture, PTH treatment results in almost complete removal of NaPi2a from the brush border (BB) within 1 h whereas for NaPi2c this process in not complete until 4 to 8 h after PTH treatment. The reason for this is poorly understood. We have previously shown that the unconventional myosin motor myosin VI is required for PTH-induced removal of NaPi2a from the proximal tubule BB. Here we demonstrate that myosin VI is also necessary for PTH-induced removal of NaPi2c from the apical membrane. In addition, we show that, while at baseline the two cotransporters have similar diffusion coefficients within the membrane, after PTH addition the diffusion coefficient for NaPi2a initially exceeds that for NaPi2c. Thus NaPi2c appears to remain "tethered" in the apical membrane for longer periods of time after PTH treatment, accounting, at least in part, for the difference in response times to PTH of NaPi2a versus NaPi2c.
Collapse
Affiliation(s)
- Luca Lanzano
- Laboratory for Fluorescence Dynamics, University of California, Irvine, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Henning MS, Stiedl P, Barry DS, McMahon R, Morham SG, Walsh D, Naghavi MH. PDZD8 is a novel moesin-interacting cytoskeletal regulatory protein that suppresses infection by herpes simplex virus type 1. Virology 2011; 415:114-21. [PMID: 21549406 DOI: 10.1016/j.virol.2011.04.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2010] [Revised: 01/19/2011] [Accepted: 04/18/2011] [Indexed: 12/01/2022]
Abstract
The host cytoskeleton plays a central role in the life cycle of many viruses yet our knowledge of cytoskeletal regulators and their role in viral infection remains limited. Recently, moesin and ezrin, two members of the ERM (Ezrin/Radixin/Moesin) family of proteins that regulate actin and plasma membrane cross-linking and microtubule (MT) stability, have been shown to inhibit retroviral infection. To further understand how ERM proteins function and whether they also influence infection by other viruses, we identified PDZD8 as a novel moesin-interacting protein. PDZD8 is a poorly understood protein whose function is unknown. Exogenous expression of either moesin or PDZD8 reduced the levels of stable MTs, suggesting that these proteins functioned as part of a cytoskeletal regulatory complex. Additionally, exogenous expression or siRNA-mediated knockdown of either factor affected Herpes Simplex Virus type 1 (HSV-1) infection, identifying a cellular function for PDZD8 and novel antiviral properties for these two cytoskeletal regulatory proteins.
Collapse
Affiliation(s)
- Matthew S Henning
- Centre for Research in Infectious Diseases, School of Medicine and Medical Science, University College Dublin, Belfield, Dublin 4, Ireland
| | | | | | | | | | | | | |
Collapse
|
48
|
Farago B, Li J, Cornilescu G, Callaway DJE, Bu Z. Activation of nanoscale allosteric protein domain motion revealed by neutron spin echo spectroscopy. Biophys J 2011; 99:3473-82. [PMID: 21081097 DOI: 10.1016/j.bpj.2010.09.058] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2010] [Revised: 09/09/2010] [Accepted: 09/30/2010] [Indexed: 11/15/2022] Open
Abstract
NHERF1 is a multidomain scaffolding protein that assembles signaling complexes, and regulates the cell surface expression and endocytic recycling of a variety of membrane proteins. The ability of the two PDZ domains in NHERF1 to assemble protein complexes is allosterically modulated by the membrane-cytoskeleton linker protein ezrin, whose binding site is located as far as 110 Ångstroms away from the PDZ domains. Here, using neutron spin echo (NSE) spectroscopy, selective deuterium labeling, and theoretical analyses, we reveal the activation of interdomain motion in NHERF1 on nanometer length-scales and on submicrosecond timescales upon forming a complex with ezrin. We show that a much-simplified coarse-grained model suffices to describe interdomain motion of a multidomain protein or protein complex. We expect that future NSE experiments will benefit by exploiting our approach of selective deuteration to resolve the specific domain motions of interest from a plethora of global translational and rotational motions. Our results demonstrate that the dynamic propagation of allosteric signals to distal sites involves changes in long-range coupled domain motions on submicrosecond timescales, and that these coupled motions can be distinguished and characterized by NSE.
Collapse
|
49
|
Proteins move! Protein dynamics and long-range allostery in cell signaling. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2011; 83:163-221. [PMID: 21570668 DOI: 10.1016/b978-0-12-381262-9.00005-7] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
An emerging point of view in protein chemistry is that proteins are not the static objects that are displayed in textbooks but are instead dynamic actors. Protein dynamics plays a fundamental role in many diseases, and spans a large hierarchy of timescales, from picoseconds to milliseconds or even longer. Nanoscale protein domain motion on length scales comparable to protein dimensions is key to understanding how signals are relayed through multiple protein-protein interactions. A canonical example is how the scaffolding proteins NHERF1 and ezrin work in coordination to assemble crucial membrane complexes. As membrane-cytoskeleton scaffolding proteins, these provide excellent prototypes for understanding how regulatory signals are relayed through protein-protein interactions between the membrane and the cytoskeleton. Here, we review recent progress in understanding the structure and dynamics of the interaction. We describe recent novel applications of neutron spin echo spectroscopy to reveal the dynamic propagation of allosteric signals by nanoscale protein motion, and present a guide to the future study of dynamics and its application to the cure of disease.
Collapse
|
50
|
Lecce L, Lindsay LA, Murphy CR. Ezrin and EBP50 redistribute apically in rat uterine epithelial cells at the time of implantation and in response to cell contact. Cell Tissue Res 2010; 343:445-53. [PMID: 21120533 DOI: 10.1007/s00441-010-1088-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2010] [Accepted: 11/02/2010] [Indexed: 11/25/2022]
Abstract
Uterine epithelial cells (UECs) undergo extensive morphological remodelling in preparation for an implanting blastocyst. This remodelling involves changes in the actin cytoskeleton and surface structures including microvilli. Ezrin and ezrin-radixin-moesin-binding protein-50-kDa (EBP50) link actin filaments to intra-membranous adhesion molecules and are important molecules in polarised epithelia. The current study is the first to describe the colocalisation and molecular association of ezrin and EBP50 in rat UECs by using immunofluorescence microscopy and immunoprecipitation techniques. These proteins have also been localised in relation to uterine epithelial cytoskeletal rearrangement during early pregnancy in the rat and to the effect of apical surface contact between opposing epithelial cells, blastocyst contact and contact with a silicon filament. Immunofluorescence microscopy has revealed that ezrin and EBP50 respond to contact between opposing epithelial cells and increase apically on day 6 of pregnancy. This apical distribution is also observed in UECs in contact with a silicon filament. Ezrin and EBP50 are however absent within the implantation chamber itself, seemingly mimicking the events that take place in leucocyte-endothelium binding. Thus, ezrin and EBP50 occur apically in UECs at the time of implantation in the rat and in response to a substitute blastocyst (filament) suggesting a role for these proteins in the cytoskeletal rearrangements that facilitate uterine receptivity and blastocyst-epithelial adhesion. Their loss within the implantation chamber possibly allows the subsequent invasion of the embryo.
Collapse
Affiliation(s)
- Laura Lecce
- Cell and Reproductive Biology Laboratory, School of Medical Sciences, Anderson Stuart Building F13, Sydney, NSW, 2006, Australia.
| | | | | |
Collapse
|