1
|
Strandberg H, Hagströmer CJ, Werin B, Wendler M, Johanson U, Törnroth-Horsefield S. Structural Basis for the Interaction between the Ezrin FERM-Domain and Human Aquaporins. Int J Mol Sci 2024; 25:7672. [PMID: 39062914 PMCID: PMC11277499 DOI: 10.3390/ijms25147672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 07/08/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
The Ezrin/Radixin/Moesin (ERM) family of proteins act as cross-linkers between the plasma membrane and the actin cytoskeleton. This mechanism plays an essential role in processes related to membrane remodeling and organization, such as cell polarization, morphogenesis and adhesion, as well as in membrane protein trafficking and signaling pathways. For several human aquaporin (AQP) isoforms, an interaction between the ezrin band Four-point-one, Ezrin, Radixin, Moesin (FERM)-domain and the AQP C-terminus has been demonstrated, and this is believed to be important for AQP localization in the plasma membrane. Here, we investigate the structural basis for the interaction between ezrin and two human AQPs: AQP2 and AQP5. Using microscale thermophoresis, we show that full-length AQP2 and AQP5 as well as peptides corresponding to their C-termini interact with the ezrin FERM-domain with affinities in the low micromolar range. Modelling of the AQP2 and AQP5 FERM complexes using ColabFold reveals a common mode of binding in which the proximal and distal parts of the AQP C-termini bind simultaneously to distinct binding sites of FERM. While the interaction at each site closely resembles other FERM-complexes, the concurrent interaction with both sites has only been observed in the complex between moesin and its C-terminus which causes auto-inhibition. The proposed interaction between AQP2/AQP5 and FERM thus represents a novel binding mode for extrinsic ERM-interacting partners.
Collapse
Affiliation(s)
| | | | | | | | | | - Susanna Törnroth-Horsefield
- Department of Biochemistry and Structural Biology, Lund University, 221 00 Lund, Sweden; (H.S.); (C.J.H.); (B.W.); (M.W.); (U.J.)
| |
Collapse
|
2
|
Wang Y, Ma B, Jian Y, Wu ST, Wong A, Wong J, Bonder EM, Zheng X. Deficiency of Pdcd10 causes urothelium hypertrophy and vesicle trafficking defects in ureter. FEBS J 2024; 291:1008-1026. [PMID: 38037455 DOI: 10.1111/febs.17022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 11/02/2023] [Accepted: 11/29/2023] [Indexed: 12/02/2023]
Abstract
The scaffolding protein programmed cell death protein 10 (Pdcd10) has been demonstrated to play a critical role in renal epithelial cell homeostasis and function by maintaining appropriate water reabsorption in collecting ducts. Both ureter and kidney collecting duct systems are derived from the ureter bud during development. Here, we report that cadherin-16 (Cdh16)-cre drives gene recombination with high specificity in the ureter, but not the bladder, urothelium. The consequences of Pdcd10 deletion on the stratified ureter urothelium were investigated using an integrated approach including messenger RNA (mRNA) expression analysis, immunocytochemistry, and high-resolution confocal and electron microscopy. Loss of Pdcd10 in the ureter urothelium resulted in increased expression of uroplakins (Upks) and keratins (Krts), as well as hypertrophy of the ureter urothelium with an associated increase in the number of proliferation marker protein Ki-67 (Ki67)-expressing cells specifically within the basal urothelium layer. Ultrastructural analysis documented significant modification of the intracellular membrane system, including intracellular vesicle genesis and transport along the basal- to umbrella-cell-layer axis. Additionally, Pdcd10 loss resulted in swelling of Golgi compartments, disruption of mitochondrial cristae structure, and increased lysosomal fusion. Lack of Pdcd10 also resulted in decreased fusiform vesicle formation in umbrella cells, increased secretion of exosome vesicles, and alteration in microvillar structure on apical membranes. Our findings indicate that Pdcd10 expression and its influence on homeostasis is associated with modulation of endomembrane trafficking and organelle biogenesis in the ureter urothelium.
Collapse
Affiliation(s)
- Yixuan Wang
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, and Center for Cardiovascular Diseases, Tianjin Medical University, China
| | - Baotao Ma
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, and Center for Cardiovascular Diseases, Tianjin Medical University, China
| | - Youli Jian
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Shi-Ting Wu
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, and Center for Cardiovascular Diseases, Tianjin Medical University, China
| | - Alex Wong
- Epigenetics and RNA Biology Program Centenary Institute and Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Justin Wong
- Epigenetics and RNA Biology Program Centenary Institute and Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Edward M Bonder
- Department of Biological Sciences, Rutgers, The State University of New Jersey, Newark, NJ, USA
| | - Xiangjian Zheng
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, and Center for Cardiovascular Diseases, Tianjin Medical University, China
| |
Collapse
|
3
|
Schacke S, Kirkpatrick J, Stocksdale A, Bauer R, Hagel C, Riecken LB, Morrison H. Ezrin deficiency triggers glial fibrillary acidic protein upregulation and a distinct reactive astrocyte phenotype. Glia 2022; 70:2309-2329. [PMID: 35929192 DOI: 10.1002/glia.24253] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 07/12/2022] [Accepted: 07/15/2022] [Indexed: 01/02/2023]
Abstract
Astrocytes are increasingly being recognized as contributors to physiological brain function and behavior. Astrocytes engage in glia-synaptic interactions through peripheral astrocyte processes, thus modulating synaptic signaling, for example, by handling glutamate removal from the synaptic cleft and (re)provision to axonal terminals. Peripheral astrocyte processes are ultrafine membrane protrusions rich in the membrane-to-actin cytoskeleton linker Ezrin, an essential component of in vitro filopodia formation and in vivo peripheral astrocyte process motility. Consequently, it has been postulated that Ezrin significantly contributes to neurodevelopment as well as astrocyte functions within the adult brain. However, while Ezrin has been studied in vitro within cultured primary astrocytes, in vivo studies on the role of Ezrin in astrocytes remain to be conducted and consequences of its depletion to be studied. Here, we investigated consequences of Ezrin deletion in the mouse brain starting from early neuronal specification. While Ezrin knockout did not impact prenatal cerebral cortex development, behavioral phenotyping depicted reduced exploratory behavior. Starting with postnatal appearance of glia cells, Ezrin was verified to remain predominantly expressed in astrocytes. Proteome analysis of Ezrin deficient astrocytes revealed alterations in glutamate and ion homeostasis, metabolism and cell morphology - important processes for synaptic signal transmission. Notably, Ezrin deletion in astrocytes provoked (GFAP) glial fibrillary acidic protein upregulation - a marker of astrocyte activation and reactive astrogliosis. However, this spontaneous, reactive astrogliosis exhibited proteome changes distinct from ischemic-induced reactive astrogliosis. Moreover, in experimental ischemic stroke, Ezrin knockout mice displayed reduced infarct volume, indicating a protective effect of the Ezrin deletion-induced changes and astrogliosis.
Collapse
Affiliation(s)
- Stephan Schacke
- Leibniz Institute on Aging, Fritz Lipmann Institute, Jena, Germany
| | | | - Amy Stocksdale
- Leibniz Institute on Aging, Fritz Lipmann Institute, Jena, Germany
| | - Reinhard Bauer
- Institute of Molecular Cell Biology, CMB, Jena University Hospital, Jena, Germany
| | - Christian Hagel
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Helen Morrison
- Leibniz Institute on Aging, Fritz Lipmann Institute, Jena, Germany.,Faculty of Biological Sciences, Friedrich-Schiller University, Jena, Germany
| |
Collapse
|
4
|
Barik GK, Sahay O, Paul D, Santra MK. Ezrin gone rogue in cancer progression and metastasis: An enticing therapeutic target. Biochim Biophys Acta Rev Cancer 2022; 1877:188753. [PMID: 35752404 DOI: 10.1016/j.bbcan.2022.188753] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 06/16/2022] [Accepted: 06/18/2022] [Indexed: 12/12/2022]
Abstract
Cancer metastasis is the primary cause of morbidity and mortality in cancer as it remains the most complicated, devastating, and enigmatic aspect of cancer. Several decades of extensive research have identified several key players closely associated with metastasis. Among these players, cytoskeletal linker Ezrin (the founding member of the ERM (Ezrin-Radixin-Moesin) family) was identified as a critical promoter of metastasis in pediatric cancers in the early 21st century. Ezrin was discovered 40 years ago as a aminor component of intestinal epithelial microvillus core protein, which is enriched in actin-containing cell surface structures. It controls gastric acid secretion and plays diverse physiological roles including maintaining cell polarity, regulating cell adhesion, cell motility and morphogenesis. Extensive research for more than two decades evinces that Ezrin is frequently dysregulated in several human cancers. Overexpression, altered subcellular localization and/or aberrant activation of Ezrin are closely associated with higher metastatic incidence and patient mortality, thereby justifying Ezrin as a valuable prognostic biomarker in cancer. Ezrin plays multifaceted role in multiple aspects of cancer, with its significant contribution in the complex metastatic cascade, through reorganizing the cytoskeleton and deregulating various cellular signaling pathways. Current preclinical studies using genetic and/or pharmacological approaches reveal that inactivation of Ezrin results in significant inhibition of Ezrin-mediated tumor growth and metastasis as well as increase in the sensitivity of cancer cells to various chemotherapeutic drugs. In this review, we discuss the recent advances illuminating the molecular mechanisms responsible for Ezrin dysregulation in cancer and its pleiotropic role in cancer progression and metastasis. We also highlight its potential as a prognostic biomarker and therapeutic target in various cancers. More importantly, we put forward some potential questions, which we strongly believe, will stimulate both basic and translational research to better understand Ezrin-mediated malignancy, ultimately leading to the development of Ezrin-targeted cancer therapy for the betterment of human life.
Collapse
Affiliation(s)
- Ganesh Kumar Barik
- Cancer Biology Division, National Centre for Cell Science, Ganeshkhind Road, Pune, Maharashtra 411007, India; Department of Biotechnology, Savitribai Phule Pune University, Ganeshkhind Road, Pune, Maharashtra 411007, India
| | - Osheen Sahay
- Cancer Biology Division, National Centre for Cell Science, Ganeshkhind Road, Pune, Maharashtra 411007, India; Department of Biotechnology, Savitribai Phule Pune University, Ganeshkhind Road, Pune, Maharashtra 411007, India
| | - Debasish Paul
- Laboratory of Cancer Biology and Genetics, Centre for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Manas Kumar Santra
- Cancer Biology Division, National Centre for Cell Science, Ganeshkhind Road, Pune, Maharashtra 411007, India.
| |
Collapse
|
5
|
Ezrin Regulates Ca 2+ Ionophore-Induced Plasma Membrane Translocation of Aquaporin-5. Int J Mol Sci 2021; 22:ijms222413505. [PMID: 34948308 PMCID: PMC8705411 DOI: 10.3390/ijms222413505] [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: 11/10/2021] [Revised: 12/03/2021] [Accepted: 12/14/2021] [Indexed: 01/16/2023] Open
Abstract
Aquaporin-5 (AQP5) is selectively expressed in the apical membrane of exocrine glands, such as salivary, sweat, and submucosal airway glands, and plays important roles in maintaining their secretory functions. Because AQP5 is not regulated by gating, localization on the plasma membrane is important for its water-permeable function. Ezrin is an ezrin-radixin-moesin family protein that serves as a crosslinker between the plasma membrane and actin cytoskeleton network. It plays important roles in translocation of various membrane proteins to mediate vesicle trafficking to the plasma membrane. In this study, we examined the effects of ezrin inhibition on membrane trafficking of AQP5. Ezrin inhibition selectively suppressed an ionomycin-induced increase in AQP5 translocation to the plasma membrane of mouse lung epithelial cells (MLE-12) without affecting the steady-state level of plasma membrane AQP5. Taken together, our data suggest that AQP5 translocates to the plasma membrane through at least two pathways and that ezrin is selectively involved in a stimulation-dependent pathway.
Collapse
|
6
|
Song X, Wang W, Wang H, Yuan X, Yang F, Zhao L, Mullen M, Du S, Zohbi N, Muthusamy S, Cao Y, Jiang J, Xia P, He P, Ding M, Emmett N, Ma M, Wu Q, Green HN, Ding X, Wang D, Wang F, Liu X. Acetylation of ezrin regulates membrane-cytoskeleton interaction underlying CCL18-elicited cell migration. J Mol Cell Biol 2021; 12:424-437. [PMID: 31638145 PMCID: PMC7333480 DOI: 10.1093/jmcb/mjz099] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 06/29/2019] [Accepted: 08/13/2019] [Indexed: 12/13/2022] Open
Abstract
Ezrin, a membrane–cytoskeleton linker protein, plays an essential role in cell polarity establishment, cell migration, and division. Recent studies show that ezrin phosphorylation regulates breast cancer metastasis by promoting cancer cell survivor and promotes intrahepatic metastasis via cell migration. However, it was less characterized whether there are additional post-translational modifications and/or post-translational crosstalks on ezrin underlying context-dependent breast cancer cell migration and invasion. Here we show that ezrin is acetylated by p300/CBP-associated factor (PCAF) in breast cancer cells in response to CCL18 stimulation. Ezrin physically interacts with PCAF and is a cognate substrate of PCAF. The acetylation site of ezrin was mapped by mass spectrometric analyses, and dynamic acetylation of ezrin is essential for CCL18-induced breast cancer cell migration and invasion. Mechanistically, the acetylation reduced the lipid-binding activity of ezrin to ensure a robust and dynamic cycling between the plasma membrane and cytosol in response to CCL18 stimulation. Biochemical analyses show that ezrin acetylation prevents the phosphorylation of Thr567. Using atomic force microscopic measurements, our study revealed that acetylation of ezrin induced its unfolding into a dominant structure, which prevents ezrin phosphorylation at Thr567. Thus, these results present a previously undefined mechanism by which CCL18-elicited crosstalks between the acetylation and phosphorylation on ezrin control breast cancer cell migration and invasion. This suggests that targeting PCAF signaling could be a potential therapeutic strategy for combating hyperactive ezrin-driven cancer progression.
Collapse
Affiliation(s)
- Xiaoyu Song
- School of Traditional Medicine, Beijing University of Chinese Medicine, Beijing, China.,MOE Key Laboratory for Membraneless Organelles & Cellular Dynamics, Hefei National Center for Physical Sciences at the Microscale, Hefei, China.,Morehouse School of Medicine, Keck Center for Organoids Plasticity, Atlanta, GA, USA
| | - Wanjuan Wang
- School of Traditional Medicine, Beijing University of Chinese Medicine, Beijing, China.,MOE Key Laboratory for Membraneless Organelles & Cellular Dynamics, Hefei National Center for Physical Sciences at the Microscale, Hefei, China
| | - Haowei Wang
- MOE Key Laboratory for Membraneless Organelles & Cellular Dynamics, Hefei National Center for Physical Sciences at the Microscale, Hefei, China.,Optics and Optical Engineering, University of Science and Technology of China, Hefei, China
| | - Xiao Yuan
- MOE Key Laboratory for Membraneless Organelles & Cellular Dynamics, Hefei National Center for Physical Sciences at the Microscale, Hefei, China
| | - Fengrui Yang
- MOE Key Laboratory for Membraneless Organelles & Cellular Dynamics, Hefei National Center for Physical Sciences at the Microscale, Hefei, China.,Morehouse School of Medicine, Keck Center for Organoids Plasticity, Atlanta, GA, USA
| | - Lingli Zhao
- MOE Key Laboratory for Membraneless Organelles & Cellular Dynamics, Hefei National Center for Physical Sciences at the Microscale, Hefei, China.,Morehouse School of Medicine, Keck Center for Organoids Plasticity, Atlanta, GA, USA
| | - McKay Mullen
- MOE Key Laboratory for Membraneless Organelles & Cellular Dynamics, Hefei National Center for Physical Sciences at the Microscale, Hefei, China.,Morehouse School of Medicine, Keck Center for Organoids Plasticity, Atlanta, GA, USA
| | - Shihao Du
- School of Traditional Medicine, Beijing University of Chinese Medicine, Beijing, China.,MOE Key Laboratory for Membraneless Organelles & Cellular Dynamics, Hefei National Center for Physical Sciences at the Microscale, Hefei, China
| | - Najdat Zohbi
- MOE Key Laboratory for Membraneless Organelles & Cellular Dynamics, Hefei National Center for Physical Sciences at the Microscale, Hefei, China.,Morehouse School of Medicine, Keck Center for Organoids Plasticity, Atlanta, GA, USA
| | - Saravanakumar Muthusamy
- MOE Key Laboratory for Membraneless Organelles & Cellular Dynamics, Hefei National Center for Physical Sciences at the Microscale, Hefei, China.,Morehouse School of Medicine, Keck Center for Organoids Plasticity, Atlanta, GA, USA
| | - Yalei Cao
- School of Traditional Medicine, Beijing University of Chinese Medicine, Beijing, China.,MOE Key Laboratory for Membraneless Organelles & Cellular Dynamics, Hefei National Center for Physical Sciences at the Microscale, Hefei, China
| | - Jiying Jiang
- MOE Key Laboratory for Membraneless Organelles & Cellular Dynamics, Hefei National Center for Physical Sciences at the Microscale, Hefei, China
| | - Peng Xia
- MOE Key Laboratory for Membraneless Organelles & Cellular Dynamics, Hefei National Center for Physical Sciences at the Microscale, Hefei, China
| | - Ping He
- MOE Key Laboratory for Membraneless Organelles & Cellular Dynamics, Hefei National Center for Physical Sciences at the Microscale, Hefei, China
| | - Mingrui Ding
- MOE Key Laboratory for Membraneless Organelles & Cellular Dynamics, Hefei National Center for Physical Sciences at the Microscale, Hefei, China.,Morehouse School of Medicine, Keck Center for Organoids Plasticity, Atlanta, GA, USA
| | - Nerimah Emmett
- Morehouse School of Medicine, Keck Center for Organoids Plasticity, Atlanta, GA, USA
| | - Mingming Ma
- MOE Key Laboratory for Membraneless Organelles & Cellular Dynamics, Hefei National Center for Physical Sciences at the Microscale, Hefei, China
| | - Quan Wu
- MOE Key Laboratory for Membraneless Organelles & Cellular Dynamics, Hefei National Center for Physical Sciences at the Microscale, Hefei, China
| | - Hadiyah-Nicole Green
- School of Traditional Medicine, Beijing University of Chinese Medicine, Beijing, China.,Morehouse School of Medicine, Keck Center for Organoids Plasticity, Atlanta, GA, USA
| | - Xia Ding
- School of Traditional Medicine, Beijing University of Chinese Medicine, Beijing, China.,MOE Key Laboratory for Membraneless Organelles & Cellular Dynamics, Hefei National Center for Physical Sciences at the Microscale, Hefei, China.,Morehouse School of Medicine, Keck Center for Organoids Plasticity, Atlanta, GA, USA
| | - Dongmei Wang
- MOE Key Laboratory for Membraneless Organelles & Cellular Dynamics, Hefei National Center for Physical Sciences at the Microscale, Hefei, China
| | - Fengsong Wang
- MOE Key Laboratory for Membraneless Organelles & Cellular Dynamics, Hefei National Center for Physical Sciences at the Microscale, Hefei, China.,School of Life Science, Anhui Medical University, Hefei, China
| | - Xing Liu
- School of Traditional Medicine, Beijing University of Chinese Medicine, Beijing, China.,MOE Key Laboratory for Membraneless Organelles & Cellular Dynamics, Hefei National Center for Physical Sciences at the Microscale, Hefei, China.,Morehouse School of Medicine, Keck Center for Organoids Plasticity, Atlanta, GA, USA
| |
Collapse
|
7
|
Wölffling S, Daddi AA, Imai-Matsushima A, Fritsche K, Goosmann C, Traulsen J, Lisle R, Schmid M, Reines-Benassar MDM, Pfannkuch L, Brinkmann V, Bornschein J, Malfertheiner P, Ordemann J, Link A, Meyer TF, Boccellato F. EGF and BMPs Govern Differentiation and Patterning in Human Gastric Glands. Gastroenterology 2021; 161:623-636.e16. [PMID: 33957136 DOI: 10.1053/j.gastro.2021.04.062] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 04/22/2021] [Accepted: 04/24/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS The homeostasis of the gastrointestinal epithelium relies on cell regeneration and differentiation into distinct lineages organized inside glands and crypts. Regeneration depends on Wnt/β-catenin pathway activation, but to understand homeostasis and its dysregulation in disease, we need to identify the signaling microenvironment governing cell differentiation. By using gastric glands as a model, we have identified the signals inducing differentiation of surface mucus-, zymogen-, and gastric acid-producing cells. METHODS We generated mucosoid cultures from the human stomach and exposed them to different growth factors to obtain cells with features of differentiated foveolar, chief, and parietal cells. We localized the source of the growth factors in the tissue of origin. RESULTS We show that epidermal growth factor is the major fate determinant distinguishing the surface and inner part of human gastric glands. In combination with bone morphogenetic factor/Noggin signals, epidermal growth factor controls the differentiation of foveolar cells vs parietal or chief cells. We also show that epidermal growth factor is likely to underlie alteration of the gastric mucosa in the precancerous condition atrophic gastritis. CONCLUSIONS Use of our recently established mucosoid cultures in combination with analysis of the tissue of origin provided a robust strategy to understand differentiation and patterning of human tissue and allowed us to draw a new, detailed map of the signaling microenvironment in the human gastric glands.
Collapse
Affiliation(s)
- Sarah Wölffling
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Alice Anna Daddi
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, United Kingdom
| | - Aki Imai-Matsushima
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany; Preemptive Medicine and Lifestyle-Related Diseases Research Center, Kyoto University Hospital, Kyoto, Japan
| | - Kristin Fritsche
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Christian Goosmann
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Jan Traulsen
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, United Kingdom
| | - Richard Lisle
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, United Kingdom
| | - Monika Schmid
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | | | - Lennart Pfannkuch
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Volker Brinkmann
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Jan Bornschein
- Translational Gastroenterology Unit, John Radcliffe Hospital, Oxford University Hospitals, Oxford, United Kingdom; Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Hospital, Magdeburg, Germany
| | - Peter Malfertheiner
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Hospital, Magdeburg, Germany
| | - Jürgen Ordemann
- Department of Bariatric and Metabolic Surgery, Helios Klinikum, Berlin, Germany; Center for Bariatric and Metabolic Surgery, Vivantes Klinikum Spandau, Berlin, Germany
| | - Alexander Link
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Hospital, Magdeburg, Germany
| | - Thomas F Meyer
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany; Laboratory of Infection Oncology, Institute of Clinical Molecular Biology, Christian Albrechts University of Kiel and University Hospital Schleswig-Holstein, Kiel, Germany.
| | - Francesco Boccellato
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany; Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, United Kingdom.
| |
Collapse
|
8
|
Wang S, Lin CW, Carleton AE, Cortez CL, Johnson C, Taniguchi LE, Sekulovski N, Townshend RF, Basrur V, Nesvizhskii AI, Zou P, Fu J, Gumucio DL, Duncan MC, Taniguchi K. Spatially resolved cell polarity proteomics of a human epiblast model. SCIENCE ADVANCES 2021; 7:7/17/eabd8407. [PMID: 33893097 PMCID: PMC8064645 DOI: 10.1126/sciadv.abd8407] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 03/05/2021] [Indexed: 05/08/2023]
Abstract
Critical early steps in human embryonic development include polarization of the inner cell mass, followed by formation of an expanded lumen that will become the epiblast cavity. Recently described three-dimensional (3D) human pluripotent stem cell-derived cyst (hPSC-cyst) structures can replicate these processes. To gain mechanistic insights into the poorly understood machinery involved in epiblast cavity formation, we interrogated the proteomes of apical and basolateral membrane territories in 3D human hPSC-cysts. APEX2-based proximity bioinylation, followed by quantitative mass spectrometry, revealed a variety of proteins without previous annotation to specific membrane subdomains. Functional experiments validated the requirement for several apically enriched proteins in cyst morphogenesis. In particular, we found a key role for the AP-1 clathrin adaptor complex in expanding the apical membrane domains during lumen establishment. These findings highlight the robust power of this proximity labeling approach for discovering novel regulators of epithelial morphogenesis in 3D stem cell-based models.
Collapse
Affiliation(s)
- Sicong Wang
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Chien-Wei Lin
- Division of Biostatistics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Amber E Carleton
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Chari L Cortez
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Craig Johnson
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Linnea E Taniguchi
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Nikola Sekulovski
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Ryan F Townshend
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Venkatesha Basrur
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Alexey I Nesvizhskii
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Peng Zou
- College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Jianping Fu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA.
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Deborah L Gumucio
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Mara C Duncan
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| | - Kenichiro Taniguchi
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
9
|
Huta Y, Nitzan Y, Breitbart H. Ezrin protects bovine spermatozoa from spontaneous acrosome reaction. Theriogenology 2020; 151:119-127. [PMID: 32334120 DOI: 10.1016/j.theriogenology.2020.04.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 04/12/2020] [Accepted: 04/13/2020] [Indexed: 11/18/2022]
Abstract
To interact and penetrate the egg, the spermatozoon must undergo a maturation step called the acrosome reaction (AR) in close proximity to the egg. This process can take place only after a series of biochemical changes to the sperm occur in the female reproductive tract, collectively called capacitation. Spermatozoa can undergo spontaneous-acrosome reaction (sAR) before reaching the vicinity of the egg, preventing successful fertilization. Several mechanisms were shown to protect spermatozoa from undergoing sAR. Here we describe the involvement of the actin cross-linker, Ezrin in the mechanism that protects spermatozoa from sAR. Inhibition of Ezrin stimulates sAR and inhibits actin polymerization. Ezrin is highly phosphorylated/activated during the first hour of the capacitation process, and its phosphorylation rate is subsequently decreased. Ezrin phosphorylation depends on protein kinase A (PKA) and calmodulin kinase II (CaMKII) activities, and to some extent on phosphatidyl-inositol-4-kinase (PI4K) activity. Inhibition of these three kinases stimulates sAR, in which the effect of PI4K inhibition, but not PKA or CaMKII inhibition, can be reversed by increasing p-Ezrin using a phosphatase inhibitor. All together, we showed that three kinases mediate Ezrin activation during spermatozoa capacitation, leading to actin polymerization in a mechanism that prevents sAR.
Collapse
Affiliation(s)
- Y Huta
- The Mina &Everard Faculty of Life Sciences,Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Y Nitzan
- Department of Clinical Laboratory Science, Zefat Academic College, Zefat, 1320611, Israel
| | - H Breitbart
- The Mina &Everard Faculty of Life Sciences,Bar-Ilan University, Ramat-Gan, 5290002, Israel.
| |
Collapse
|
10
|
Yao X, Smolka AJ. Gastric Parietal Cell Physiology and Helicobacter pylori-Induced Disease. Gastroenterology 2019; 156:2158-2173. [PMID: 30831083 PMCID: PMC6715393 DOI: 10.1053/j.gastro.2019.02.036] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 02/12/2019] [Accepted: 02/14/2019] [Indexed: 12/13/2022]
Abstract
Acidification of the gastric lumen poses a barrier to transit of potentially pathogenic bacteria and enables activation of pepsin to complement nutrient proteolysis initiated by salivary proteases. Histamine-induced activation of the PKA signaling pathway in gastric corpus parietal cells causes insertion of proton pumps into their apical plasma membranes. Parietal cell secretion and homeostasis are regulated by signaling pathways that control cytoskeletal changes required for apical membrane remodeling and organelle and proton pump activities. Helicobacter pylori colonization of human gastric mucosa affects gastric epithelial cell plasticity and homeostasis, promoting epithelial progression to neoplasia. By intervening in proton pump expression, H pylori regulates the abundance and diversity of microbiota that populate the intestinal lumen. We review stimulation-secretion coupling and renewal mechanisms in parietal cells and the mechanisms by which H pylori toxins and effectors alter cell secretory pathways (constitutive and regulated) and organelles to establish and maintain their inter- and intracellular niches. Studies of bacterial toxins and their effector proteins have provided insights into parietal cell physiology and the mechanisms by which pathogens gain control of cell activities, increasing our understanding of gastrointestinal physiology, microbial infectious disease, and immunology.
Collapse
Affiliation(s)
- Xuebiao Yao
- MOE Key Laboratory of Cellular Dynamics, CAS Center for Excellence in Molecular Cell Science, University of Science and Technology of China, Hefei, China; Keck Center for Cellular Dynamics and Organoids Plasticity, Morehouse School of Medicine, Atlanta, Georgia.
| | - Adam J. Smolka
- Gastroenterology and Hepatology Division, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
11
|
Abstract
Comprehensive theory explaining the relationship between estrogen (E2) and ezrin in metastasis of thyroid cancer remains non-elicited. In vitro results revealed that E2 could stimulate the expression and phosphorylation of ezrin in a time and dose dependent manner. Our data clearly showed that E2 enhanced the migration and invasion of cells, which was reversed by the transfection of cells with ezrin specific siRNA. Further, we observed that Phosphoinositide 3-kinase (PI3K) ROCK-2 are among the kinases responsible for E2 induced phosphorylation of ezrin. Clinical validation of ezrin/phospho-ezrin revealed that phospho-ezrin was intensely expressed in follicular thyroid carcinoma (FTC) and follicular variant of papillary thyroid carcinoma (FVPTC), while it was completely absent in follicular adenoma (FA) lesions in which the differentiation of the follicular neoplasms remains subtle. When histology of different carcinomas is correlated with benign FA with respect to phospho-ezrin, we observed that the marker was highly significant (p = 0.0001). 100% sensitivity, specificity and diagnostic accuracy of the above marker in the histological association of FTC, FVPTC with FA, enables us to suggest phospho-ezrin as a diagnostic marker to differentiate the follicular neoplasms. These data are the first to suggest the dynamic regulation of ezrin phosphorylation during metastasis in FTC.
Collapse
|
12
|
Tsai FC, Bertin A, Bousquet H, Manzi J, Senju Y, Tsai MC, Picas L, Miserey-Lenkei S, Lappalainen P, Lemichez E, Coudrier E, Bassereau P. Ezrin enrichment on curved membranes requires a specific conformation or interaction with a curvature-sensitive partner. eLife 2018; 7:37262. [PMID: 30234483 PMCID: PMC6167055 DOI: 10.7554/elife.37262] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 09/14/2018] [Indexed: 01/12/2023] Open
Abstract
One challenge in cell biology is to decipher the biophysical mechanisms governing protein enrichment on curved membranes and the resulting membrane deformation. The ERM protein ezrin is abundant and associated with cellular membranes that are flat, positively or negatively curved. Using in vitro and cell biology approaches, we assess mechanisms of ezrin’s enrichment on curved membranes. We evidence that wild-type ezrin (ezrinWT) and its phosphomimetic mutant T567D (ezrinTD) do not deform membranes but self-assemble anti-parallelly, zipping adjacent membranes. EzrinTD’s specific conformation reduces intermolecular interactions, allows binding to actin filaments, which reduces membrane tethering, and promotes ezrin binding to positively-curved membranes. While neither ezrinTD nor ezrinWT senses negative curvature alone, we demonstrate that interacting with curvature-sensing I-BAR-domain proteins facilitates ezrin enrichment in negatively-curved membrane protrusions. Overall, our work demonstrates that ezrin can tether membranes, or be targeted to curved membranes, depending on conformations and interactions with actin and curvature-sensing binding partners.
Collapse
Affiliation(s)
- Feng-Ching Tsai
- Laboratoire Physico Chimie Curie, Institut Curie, PSL Research University, CNRS UMR168, Paris, France.,Sorbonne Université, Paris, France
| | - Aurelie Bertin
- Laboratoire Physico Chimie Curie, Institut Curie, PSL Research University, CNRS UMR168, Paris, France.,Sorbonne Université, Paris, France
| | - Hugo Bousquet
- Sorbonne Université, Paris, France.,Compartimentation et dynamique cellulaire, Institut Curie, PSL Research University, CNRS UMR144, Paris, France
| | - John Manzi
- Laboratoire Physico Chimie Curie, Institut Curie, PSL Research University, CNRS UMR168, Paris, France.,Sorbonne Université, Paris, France
| | - Yosuke Senju
- Program in Cell and Molecular Biology, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Meng-Chen Tsai
- Université Côte d'Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France.,Département de Microbiologie, Unité des Toxines Bactériennes, Université Paris Descartes, Institut Pasteur, Paris, France
| | - Laura Picas
- Institut de Recherche en Infectiologie de Montpellier (IRIM), CNRS UMR 9004, Montpellier, France
| | - Stephanie Miserey-Lenkei
- Sorbonne Université, Paris, France.,Compartimentation et dynamique cellulaire, Institut Curie, PSL Research University, CNRS UMR144, Paris, France
| | - Pekka Lappalainen
- Program in Cell and Molecular Biology, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Emmanuel Lemichez
- Département de Microbiologie, Unité des Toxines Bactériennes, Université Paris Descartes, Institut Pasteur, Paris, France
| | - Evelyne Coudrier
- Sorbonne Université, Paris, France.,Compartimentation et dynamique cellulaire, Institut Curie, PSL Research University, CNRS UMR144, Paris, France
| | - Patricia Bassereau
- Laboratoire Physico Chimie Curie, Institut Curie, PSL Research University, CNRS UMR168, Paris, France.,Sorbonne Université, Paris, France
| |
Collapse
|
13
|
Okamoto CT. Regulation of Transporters and Channels by Membrane-Trafficking Complexes in Epithelial Cells. Cold Spring Harb Perspect Biol 2017; 9:a027839. [PMID: 28246186 PMCID: PMC5666629 DOI: 10.1101/cshperspect.a027839] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The vectorial secretion and absorption of fluid and solutes by epithelial cells is dependent on the polarized expression of membrane solute transporters and channels at the apical and basolateral membranes. The establishment and maintenance of this polarized expression of transporters and channels are affected by divers protein-trafficking complexes. Moreover, regulation of the magnitude of transport is often under control of physiological stimuli, again through the interaction of transporters and channels with protein-trafficking complexes. This review highlights the value in utilizing transporters and channels as cargo to characterize core trafficking machinery by which epithelial cells establish and maintain their polarized expression, and how this machinery regulates fluid and solute transport in response to physiological stimuli.
Collapse
Affiliation(s)
- Curtis T Okamoto
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California 90089-9121
| |
Collapse
|
14
|
Mytilinaiou M, Nikitovic D, Berdiaki A, Papoutsidakis A, Papachristou DJ, Tsatsakis A, Tzanakakis GN. IGF-I regulates HT1080 fibrosarcoma cell migration through a syndecan-2/Erk/ezrin signaling axis. Exp Cell Res 2017; 361:9-18. [PMID: 28962916 DOI: 10.1016/j.yexcr.2017.09.035] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 09/15/2017] [Accepted: 09/25/2017] [Indexed: 11/17/2022]
Abstract
Fibrosarcoma is a tumor of mesenchymal origin, originating from fibroblasts. IGF-I is an anabolic growth factor which exhibits significant involvement in cancer progression. In this study, we investigated the possible participation of syndecan-2 (SDC-2), a cell membrane heparan sulfate (HS) proteoglycan on IGF-I dependent fibrosarcoma cell motility. Our results demonstrate that SDC-2-deficient HT1080 cells exhibit attenuated IGF-I-dependent chemotactic migration (p < 0.001). SDC-2 was found to co-localize to IGF-I receptor (IGF-IR) in a manner dependent on IGF-I activity (P ≤ 0.01). In parallel, the downregulation of SDC-2 significantly inhibited both basal and due to IGF-I action ERK1/2 activation, (p < 0.001). The phosphorylation levels of ezrin (Thr567), which is suggested to act as a signaling bridge between the cellular membrane receptors and actin cytoskeleton, were strongly enhanced by IGF-I at both 1h and 24h (p < 0.05; p < 0.01). The formation of an immunoprecipitative complex revealed an association between SDC2 and ezrin which was enhanced through IGF-I action (p < 0.05). Immunoflourescence demonstrated a co-localization of IGF-IR, SDC2 and ezrin upregulated by IGF-I action. IGF-I enhanced actin polymerization and ezrin/actin specific localization to cell membranes. Finally, treatment with IGF-I strongly increased SDC2 expression at both the mRNA and protein level (p < 0.001). Therefore, we propose a novel SDC2-dependent mechanism, where SDC2 is co-localized with IGF-IR and enhances its' IGFI-dependent downstream signaling. SDC2 mediates directly IGFI-induced ERK1/2 activation, it recruits ezrin, contributes to actin polymerization and ezrin/actin specific localization to cell membranes, ultimately facilitating the progression of IGFI-dependent fibrosarcoma cell migration.
Collapse
Affiliation(s)
- Maria Mytilinaiou
- Laboratory of Anatomy-Histology-Embryology, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Dragana Nikitovic
- Laboratory of Anatomy-Histology-Embryology, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Aikaterini Berdiaki
- Laboratory of Anatomy-Histology-Embryology, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Antonis Papoutsidakis
- Laboratory of Anatomy-Histology-Embryology, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | | | - Aristidis Tsatsakis
- Laboratory of Anatomy-Histology-Embryology, Unit of Bone and Soft Tissue Studies, School of Medicine, University of Patras, Patras, Greece
| | - George N Tzanakakis
- Laboratory of Anatomy-Histology-Embryology, School of Medicine, University of Crete, 71003 Heraklion, Greece.
| |
Collapse
|
15
|
Li W, Jin WW, Tsuji K, Chen Y, Nomura N, Su L, Yui N, Arthur J, Cotecchia S, Paunescu TG, Brown D, Lu HAJ. Ezrin directly interacts with AQP2 and promotes its endocytosis. J Cell Sci 2017; 130:2914-2925. [PMID: 28754689 DOI: 10.1242/jcs.204842] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 07/21/2017] [Indexed: 11/20/2022] Open
Abstract
The water channel aquaporin-2 (AQP2) is a major regulator of water homeostasis in response to vasopressin (VP). Dynamic trafficking of AQP2 relies on its close interaction with trafficking machinery proteins and the actin cytoskeleton. Here, we report the identification of ezrin, an actin-binding protein from the ezrin/radixin/moesin (ERM) family as an AQP2-interacting protein. Ezrin was first detected in a co-immunoprecipitation (co-IP) complex using an anti-AQP2 antibody in a proteomic analysis. Immunofluorescence staining revealed the co-expression of ezrin and AQP2 in collecting duct principal cells, and VP treatment caused redistribution of both proteins to the apical membrane. The ezrin-AQP2 interaction was confirmed by co-IP experiments with an anti-ezrin antibody, and by pulldown assays using purified full-length and FERM domain-containing recombinant ezrin. By using purified recombinant proteins, we showed that ezrin directly interacts with AQP2 C-terminus through its N-terminal FERM domain. Knocking down ezrin expression with shRNA resulted in increased membrane accumulation of AQP2 and reduced AQP2 endocytosis. Therefore, through direct interaction with AQP2, ezrin facilitates AQP2 endocytosis, thus linking the dynamic actin cytoskeleton network with AQP2 trafficking.
Collapse
Affiliation(s)
- Wei Li
- Center for Systems Biology, Program in Membrane Biology and Division of Nephrology, Massachusetts General Hospital, and Harvard Medical School, Boston, MA 02114, USA
| | - William W Jin
- Washington University in St. Louis, College of Arts and Sciences, St Louis, MO 63130, USA
| | - Kenji Tsuji
- Center for Systems Biology, Program in Membrane Biology and Division of Nephrology, Massachusetts General Hospital, and Harvard Medical School, Boston, MA 02114, USA
| | - Ying Chen
- Center for Systems Biology, Program in Membrane Biology and Division of Nephrology, Massachusetts General Hospital, and Harvard Medical School, Boston, MA 02114, USA
| | - Naohiro Nomura
- Center for Systems Biology, Program in Membrane Biology and Division of Nephrology, Massachusetts General Hospital, and Harvard Medical School, Boston, MA 02114, USA
| | - Limin Su
- Center for Systems Biology, Program in Membrane Biology and Division of Nephrology, Massachusetts General Hospital, and Harvard Medical School, Boston, MA 02114, USA.,Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Naofumi Yui
- Center for Systems Biology, Program in Membrane Biology and Division of Nephrology, Massachusetts General Hospital, and Harvard Medical School, Boston, MA 02114, USA
| | - Julian Arthur
- Center for Systems Biology, Program in Membrane Biology and Division of Nephrology, Massachusetts General Hospital, and Harvard Medical School, Boston, MA 02114, USA
| | - Susanna Cotecchia
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne 1005, Switzerland
| | - Teodor G Paunescu
- Center for Systems Biology, Program in Membrane Biology and Division of Nephrology, Massachusetts General Hospital, and Harvard Medical School, Boston, MA 02114, USA
| | - Dennis Brown
- Center for Systems Biology, Program in Membrane Biology and Division of Nephrology, Massachusetts General Hospital, and Harvard Medical School, Boston, MA 02114, USA
| | - Hua A J Lu
- Center for Systems Biology, Program in Membrane Biology and Division of Nephrology, Massachusetts General Hospital, and Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
16
|
Li LY, Xie YH, Xie YM, Liao LD, Xu XE, Zhang Q, Zeng FM, Tao LH, Xie WM, Xie JJ, Xu LY, Li EM. Ezrin Ser66 phosphorylation regulates invasion and metastasis of esophageal squamous cell carcinoma cells by mediating filopodia formation. Int J Biochem Cell Biol 2017; 88:162-171. [PMID: 28504189 DOI: 10.1016/j.biocel.2017.05.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2016] [Revised: 04/18/2017] [Accepted: 05/09/2017] [Indexed: 02/05/2023]
Abstract
BACKGROUND Ezrin, links the plasma membrane to the actin cytoskeleton, and plays an important role in the development and progression of human esophageal squamous cell carcinoma (ESCC). However, the roles of ezrin S66 phosphorylation in tumorigenesis of ESCC remain unclear. METHODS Distribution of ezrin in membrane and cytosol fractions was examined by analysis of detergent-soluble/-insoluble fractions and cytosol/membrane fractionation. Both immunofluorescence and live imaging were used to explore the role of ezrin S66 phosphorylation in the behavior of ezrin and actin in cell filopodia. Cell proliferation, migration and invasion of ESCC cells were investigated by proliferation and migration assays, respectively. Tumorigenesis, local invasion and metastasis were assessed in a nude mouse model of regional lymph node metastasis. RESULTS Ezrin S66 phosphorylation enhanced the recruitment of ezrin to the membrane in ESCC cells. Additionally, non-phosphorylatable ezrin (S66A) significantly prevented filopodia formation, as well as caused a reduction in the number, length and lifetime of filopodia. Moreover, functional experiments revealed that expression of non-phosphorylatable ezrin (S66A) markedly suppressed migration and invasion but not proliferation of ESCC cells in vitro, and attenuated local invasion and regional lymph node metastasis, but not primary tumor growth of ESCC cells in vivo. CONCLUSION Ezrin S66 phosphorylation enhances filopodia formation, contributing to the regulation of invasion and metastasis of esophageal squamous cell carcinoma cells.
Collapse
Affiliation(s)
- Li-Yan Li
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, Guangdong, PR China; Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, Guangdong, PR China
| | - Ying-Hua Xie
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, Guangdong, PR China; Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, Guangdong, PR China
| | - Yang-Min Xie
- Experimental Animal Center, Shantou University Medical College, Shantou, Guangdong, PR China
| | - Lian-Di Liao
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, Guangdong, PR China; Institute of Oncologic Pathology, Shantou University Medical College, Shantou, Guangdong, PR China
| | - Xiu-E Xu
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, Guangdong, PR China; Institute of Oncologic Pathology, Shantou University Medical College, Shantou, Guangdong, PR China
| | - Qiang Zhang
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, Guangdong, PR China; Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, Guangdong, PR China
| | - Fa-Min Zeng
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, Guangdong, PR China; Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, Guangdong, PR China
| | - Li-Hua Tao
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, Guangdong, PR China; Institute of Oncologic Pathology, Shantou University Medical College, Shantou, Guangdong, PR China
| | - Wen-Ming Xie
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, Guangdong, PR China; Institute of Oncologic Pathology, Shantou University Medical College, Shantou, Guangdong, PR China
| | - Jian-Jun Xie
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, Guangdong, PR China; Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, Guangdong, PR China
| | - Li-Yan Xu
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, Guangdong, PR China; Institute of Oncologic Pathology, Shantou University Medical College, Shantou, Guangdong, PR China.
| | - En-Min Li
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, Guangdong, PR China; Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, Guangdong, PR China.
| |
Collapse
|
17
|
Shabardina V, Kramer C, Gerdes B, Braunger J, Cordes A, Schäfer J, Mey I, Grill D, Gerke V, Steinem C. Mode of Ezrin-Membrane Interaction as a Function of PIP2 Binding and Pseudophosphorylation. Biophys J 2017; 110:2710-2719. [PMID: 27332129 DOI: 10.1016/j.bpj.2016.05.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 04/14/2016] [Accepted: 05/05/2016] [Indexed: 12/28/2022] Open
Abstract
Ezrin, a protein of the ezrin, radixin, moesin (ERM) family, provides a regulated linkage between the plasma membrane and the cytoskeleton. The hallmark of this linkage is the activation of ezrin by phosphatidylinositol-4,5-bisphosphate (PIP2) binding and a threonine phosphorylation at position 567. To analyze the influence of these activating factors on the organization of ezrin on lipid membranes and the proposed concomitant oligomer-monomer transition, we made use of supported lipid bilayers in conjunction with atomic force microscopy and fluorescence microscopy. Bilayers doped with either PIP2 as the natural receptor lipid of ezrin or a Ni-nitrilotriacetic acid-equipped lipid to bind the proteins via their His6-tags to the lipid membrane were used to bind two different ezrin variants: ezrin wild-type and ezrin T567D mimicking the phosphorylated state. Using a combination of reflectometric interference spectroscopy, atomic force microscopy, and Förster resonance energy transfer experiments, we show that only the ezrin T567D mutant, upon binding to PIP2-containing bilayers, undergoes a remarkable conformational change, which we attribute to an opening of the conformation resulting in monomeric protein on the lipid bilayer.
Collapse
Affiliation(s)
- Victoria Shabardina
- Institute of Medical Biochemistry and Center for Molecular Biology of Inflammation, University of Münster, Münster, Germany
| | - Corinna Kramer
- Institute of Organic and Biomolecular Chemistry, University of Göttingen, Göttingen, Germany
| | - Benjamin Gerdes
- Institute of Organic and Biomolecular Chemistry, University of Göttingen, Göttingen, Germany
| | - Julia Braunger
- Institute of Organic and Biomolecular Chemistry, University of Göttingen, Göttingen, Germany
| | - Andrea Cordes
- Institute of Organic and Biomolecular Chemistry, University of Göttingen, Göttingen, Germany
| | - Jonas Schäfer
- Institute of Organic and Biomolecular Chemistry, University of Göttingen, Göttingen, Germany
| | - Ingo Mey
- Institute of Organic and Biomolecular Chemistry, University of Göttingen, Göttingen, Germany
| | - David Grill
- Institute of Medical Biochemistry and Center for Molecular Biology of Inflammation, University of Münster, Münster, Germany
| | - Volker Gerke
- Institute of Medical Biochemistry and Center for Molecular Biology of Inflammation, University of Münster, Münster, Germany.
| | - Claudia Steinem
- Institute of Organic and Biomolecular Chemistry, University of Göttingen, Göttingen, Germany; Göttingen Center for Molecular Biosciences, Göttingen, Germany.
| |
Collapse
|
18
|
Wang Y, Wang S, Bao Y, Li T, Chang X, Yang G, Meng X. Multipathway Integrated Adjustment Mechanism of Glycyrrhiza Triterpenes Curing Gastric Ulcer in Rats. Pharmacogn Mag 2017; 13:209-215. [PMID: 28539709 PMCID: PMC5421414 DOI: 10.4103/0973-1296.204550] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Revised: 03/02/2016] [Indexed: 12/17/2022] Open
Abstract
Background: Gastric ulcer is a common chronic disease in human digestive system, which is difficult to cure, easy to relapse, and endangers human health seriously. Compared with western medicine, traditional Chinese medicine has a unique advantage in improving the general situation, stablizing medical condition, and with little side effects. Glycyrrhiza known as “king of all the medicine”, has a range of pharmacological activities and is commonly used in a variety of proprietary Chinese medicines and formulations. Objective: On the basis of explicit antiulcer effect of Glycyrrhiza triterpenes, the molecular mechanisms of its therapeutic effect on acetic acid induced gastric ulcer in rats were explored. Materials and Methods: Acetic acid induced gastric ulcer model in rats was established to evaluate the curing effect of G. triterpenes and all of the rats were randomised into six groups: Control group, model group, omeprazole group (0.8 mg/mL), triterpenes high dose group (378.0 mg/mL), triterpenes middle dose group (126.0 mg/mL), and triterpenes low dose group (42.0 mg/mL). All rats in groups were orally administered the active group solution 1.5 mL once daily (model and control groups with saline) for 7 days. HPLC-TOF-MS analysis method was performed to obtain the plasma metabolites spectrums of control group, model group, triterpenes high, middle and low dose groups. Results: A total of 11 differential endogenous metabolites related to the therapeutic effect of G. triterpenes were identified, including tryptophan, phingosine-1-phosphate, pantothenic acid, and so on, among which tryptophan and phingosine-1-phosphate are related with the calcium signaling pathway and arachidonic acid (AA) metabolism. At the same time, in order to verify the above results, quantitative real time polymerase chain reaction were performed to evaluate the expression of H+-K+-ATPase alpha mRNA and phospholipase a 2 mRNA in relational signaling pathways. Combined with statistical analysis of plasma metabolic spectrum and gene expression in tissue, it is suggested that G. triterpenes has antiulcer effect on gastric ulcer in rats. Conclusion: G. triterpenes has a certain regulating effect on the metabolism of tryptophan, AA, sphingosine, and other endogenous metabolites, and we speculated that the antiulcer potential of G. triterpenes can be primarily attributed to its inhibiting gastric acid secretion, reducing the release of inflammatory mediators, and protecting gastric mucosa effects to prevent the further development of gastric ulcer. SUMMARY G. triterpenes can obviously relieve the symptoms of gastric ulcer, especially the low dose group. G. triterpenes can effectively regulate the amount of small molecule metablism in gastric ulcer rats in vivo, including tryptophan, phingosine-1-phosphate, etc. G. triterpenes resisting gastric ulcer is probably by regulating arachidonic acid metabolism, sphingosine metabolism, etc. Down-regulation of H+-K+-ATPase alpha subunit mRNA and up-regulation of PLA2 mRNA in gastric tissue of dose group validated the possible mechanisms of G. triterpenes for the treatment of gastric ulcer
Abbreviations used: HP: Helicobacter pylori, ECL: enterochromaffinlike, TCM: Traditional Chinese medicine; HPLC: High Performance Liquid Chromatography, HPLC/MS: High Performance Liquid chromatography Mass Spectrometry, HPLC-TOF-MS: High Performance Liquid Chromatography and Tof Mass Spectrometry, SD: Sprague Dawley, PCDL: Personal Compound Database and Library, MPP: Mass Profiler Professiona; PCA: principal component analysis, RT-PCR: real time polymerase chain reaction, PGE 2: Prostaglandin E2, COX1: cyclooxygenase 1 S1P: Sphingosine-1-phosphate, AA: Arachidonic acid, 5-HT: 5- hydroxytryptamine.
Collapse
Affiliation(s)
- Ying Wang
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, People's Republic of China
| | - Shuai Wang
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, People's Republic of China.,Component Medicine Engineering Research Center of Liaoning Province, Dalian, People's Republic of China.,Liaoning Province Modern Chinese Medicine Research Engineering Laboratory, Dalian, People's Republic of China
| | - Yongrui Bao
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, People's Republic of China.,Component Medicine Engineering Research Center of Liaoning Province, Dalian, People's Republic of China.,Liaoning Province Modern Chinese Medicine Research Engineering Laboratory, Dalian, People's Republic of China
| | - Tianjiao Li
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, People's Republic of China.,Component Medicine Engineering Research Center of Liaoning Province, Dalian, People's Republic of China.,Liaoning Province Modern Chinese Medicine Research Engineering Laboratory, Dalian, People's Republic of China
| | - Xin Chang
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, People's Republic of China
| | - Guanlin Yang
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, People's Republic of China
| | - Xiansheng Meng
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, People's Republic of China.,Component Medicine Engineering Research Center of Liaoning Province, Dalian, People's Republic of China.,Liaoning Province Modern Chinese Medicine Research Engineering Laboratory, Dalian, People's Republic of China
| |
Collapse
|
19
|
Qian J, Li Y, Yao H, Tian H, Wang H, Ai L, Xie Y, Bao Y, Liang L, Hu Y, Zhang Y, Wang J, Li C, Tang J, Chen Y, Xu J, Fang JY. ASAP3 regulates microvilli structure in parietal cells and presents intervention target for gastric acidity. Signal Transduct Target Ther 2017; 2:17003. [PMID: 29263912 PMCID: PMC5661632 DOI: 10.1038/sigtrans.2017.3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 01/20/2017] [Accepted: 01/22/2017] [Indexed: 01/28/2023] Open
Abstract
Gastric acidity-associated disorders such as peptic ulcer and reflux diseases are widespread, and the reported resistance and side effects of currently used medicines suggest an urgent requirement for alternative therapeutic approaches. Here we demonstrate a critical role of ASAP3 in regulating the microvilli structure of parietal cells in vivo, and reveal the feasibility of controlling gastric acidity by targeting ASAP3. Conditional knockout of ASAP3 in mice caused elongation and stacking of microvilli in parietal cells, and substantially decreased gastric acid secretion. These were associated with active assembly of F-actin caused by a higher level of GTP-bound Arf6 GTPase. Consistently, a small molecular compound QS11 inhibited ASAP3 function and significantly reduced gastric acidity in vivo. Of note, the expression of ASAP3 was positively correlated with gastric acid secretion in 90 human cases, and high expression of ASAP3 was associated with reflux disease and peptic ulcer. These results reveal for the first time that ASAP3 regulates the microvilli structures in parietal cells. Our data also suggest ASAP3 as a feasible and drugable therapeutic target for gastric acidity-associated diseases.
Collapse
Affiliation(s)
- Jin Qian
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Yueyuan Li
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Han Yao
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Haiying Tian
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Huanbin Wang
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Luoyan Ai
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Yuanhong Xie
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Yujie Bao
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Lunxi Liang
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Ye Hu
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Yao Zhang
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Jilin Wang
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Chushu Li
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Jiayin Tang
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Yingxuan Chen
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Jie Xu
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Jing-Yuan Fang
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, China
| |
Collapse
|
20
|
Jiang H, Wang W, Zhang Y, Yao WW, Jiang J, Qin B, Yao WY, Liu F, Wu H, Ward TL, Chen CW, Liu L, Ding X, Liu X, Yao X. Cell Polarity Kinase MST4 Cooperates with cAMP-dependent Kinase to Orchestrate Histamine-stimulated Acid Secretion in Gastric Parietal Cells. J Biol Chem 2015; 290:28272-28285. [PMID: 26405038 DOI: 10.1074/jbc.m115.668855] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Indexed: 01/13/2023] Open
Abstract
The digestive function of the stomach depends on acidification of the gastric lumen. Acid secretion into the lumen is triggered by activation of the PKA cascade, which ultimately results in the insertion of gastric H,K-ATPases into the apical plasma membranes of parietal cells. A coupling protein is ezrin, whose phosphorylation at Ser-66 by PKA is required for parietal cell activation. However, little is known regarding the molecular mechanism(s) by which this signaling pathway operates in gastric acid secretion. Here we show that PKA cooperates with MST4 to orchestrate histamine-elicited acid secretion by phosphorylating ezrin at Ser-66 and Thr-567. Histamine stimulation activates PKA, which phosphorylates MST4 at Thr-178 and then promotes MST4 kinase activity. Interestingly, activated MST4 then phosphorylates ezrin prephosphorylated by PKA. Importantly, MST4 is important for acid secretion in parietal cells because either suppression of MST4 or overexpression of non-phosphorylatable MST4 prevents the apical membrane reorganization and proton pump translocation elicited by histamine stimulation. In addition, overexpressing MST4 phosphorylation-deficient ezrin results in an inhibition of gastric acid secretion. Taken together, these results define a novel molecular mechanism linking the PKA-MST4-ezrin signaling cascade to polarized epithelial secretion in gastric parietal cells.
Collapse
Affiliation(s)
- Hao Jiang
- BUCM-USTC Joint Program in Cellular Dynamics and Anhui Key Laboratory for Cellular Dynamics, University of Science and Technology of China, Hefei 230027, China
| | - Wenwen Wang
- BUCM-USTC Joint Program in Cellular Dynamics and Anhui Key Laboratory for Cellular Dynamics, University of Science and Technology of China, Hefei 230027, China,; Molecular Imaging Center, Atlanta Clinical and Translational Science Institute, Atlanta, Georgia 30310
| | - Yin Zhang
- BUCM-USTC Joint Program in Cellular Dynamics and Anhui Key Laboratory for Cellular Dynamics, University of Science and Technology of China, Hefei 230027, China,; Beijing University of Chinese Medicine, Beijing 100029, China
| | - William W Yao
- Molecular Imaging Center, Atlanta Clinical and Translational Science Institute, Atlanta, Georgia 30310
| | - Jiying Jiang
- BUCM-USTC Joint Program in Cellular Dynamics and Anhui Key Laboratory for Cellular Dynamics, University of Science and Technology of China, Hefei 230027, China
| | - Bo Qin
- BUCM-USTC Joint Program in Cellular Dynamics and Anhui Key Laboratory for Cellular Dynamics, University of Science and Technology of China, Hefei 230027, China,; Molecular Imaging Center, Atlanta Clinical and Translational Science Institute, Atlanta, Georgia 30310
| | - Wendy Y Yao
- Molecular Imaging Center, Atlanta Clinical and Translational Science Institute, Atlanta, Georgia 30310
| | - Fusheng Liu
- BUCM-USTC Joint Program in Cellular Dynamics and Anhui Key Laboratory for Cellular Dynamics, University of Science and Technology of China, Hefei 230027, China,; Beijing University of Chinese Medicine, Beijing 100029, China; Airforce General Hospital, Beijing 100036, China
| | - Huihui Wu
- BUCM-USTC Joint Program in Cellular Dynamics and Anhui Key Laboratory for Cellular Dynamics, University of Science and Technology of China, Hefei 230027, China,; Molecular Imaging Center, Atlanta Clinical and Translational Science Institute, Atlanta, Georgia 30310
| | - Tarsha L Ward
- Molecular Imaging Center, Atlanta Clinical and Translational Science Institute, Atlanta, Georgia 30310
| | - Chun Wei Chen
- BUCM-USTC Joint Program in Cellular Dynamics and Anhui Key Laboratory for Cellular Dynamics, University of Science and Technology of China, Hefei 230027, China
| | - Lifang Liu
- Airforce General Hospital, Beijing 100036, China
| | - Xia Ding
- BUCM-USTC Joint Program in Cellular Dynamics and Anhui Key Laboratory for Cellular Dynamics, University of Science and Technology of China, Hefei 230027, China,; Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Xing Liu
- BUCM-USTC Joint Program in Cellular Dynamics and Anhui Key Laboratory for Cellular Dynamics, University of Science and Technology of China, Hefei 230027, China,; Molecular Imaging Center, Atlanta Clinical and Translational Science Institute, Atlanta, Georgia 30310.
| | - Xuebiao Yao
- BUCM-USTC Joint Program in Cellular Dynamics and Anhui Key Laboratory for Cellular Dynamics, University of Science and Technology of China, Hefei 230027, China,.
| |
Collapse
|
21
|
Henry C, Saadaoui B, Bouvier F, Cebo C. Phosphoproteomics of the goat milk fat globule membrane: New insights into lipid droplet secretion from the mammary epithelial cell. Proteomics 2015; 15:2307-17. [PMID: 25737190 DOI: 10.1002/pmic.201400245] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 01/26/2015] [Accepted: 02/26/2015] [Indexed: 12/24/2022]
Abstract
Mechanisms of milk lipid secretion are highly controversial. Analyzing the fine protein composition of the "milk fat globule membrane" (MFGM), the triple-layered membrane surrounding milk lipid droplets (LDs) can provide mechanistic clues to better understand LD biosynthesis and secretion pathways in mammary epithelial cells (MECs). We therefore combined a high-sensitive Q-Exactive LC-MS/MS analysis of MFGM-derived peptides to the use of an in-house database intended to improve protein identification in the goat species. Using this approach, we performed the identification of 442 functional groups of proteins in the MFGM from goat milk. To get a more dynamic view of intracellular mechanisms driving LD dynamics in the MECs, we decided to investigate for the first time whether MFGM proteins were phosphorylated. MFGM proteins were sequentially digested by lysine-C and trypsin proteases and the resulting peptides were fractionated by a strong cation exchange chromatography. Titanium beads were used to enrich phosphopeptides from strong cation exchange chromatography eluted fractions. This approach lets us pinpoint 271 sites of phosphorylation on 124 unique goat MFGM proteins. Enriched GO terms associated with phosphorylated MFGM proteins were protein transport and actin cytoskeleton organization. Gained data are discussed with regard to lipid secretory mechanisms in the MECs. All MS data have been deposited in the ProteomeXchange with identifier PXD001039 (http://proteomecentral.proteomexchange.org/dataset/PXD001039).
Collapse
Affiliation(s)
- Céline Henry
- INRA, UMR1319, MICALIS, Plateforme PAPSSO (Plateforme d'Analyse Protéomique Paris Sud Ouest), Jouy-en-Josas, France
| | - Besma Saadaoui
- Faculté des Sciences de Gabès, Université de Gabès, cité Erriadh Zrig, Tunisia
| | | | - Christelle Cebo
- INRA, UMR1313 Unité Génétique Animale et Biologie Intégrative, Equipe Génomique Fonctionnelle et Physiologie de la Glande Mammaire (GFP-GM), Jouy-en-Josas, France
| |
Collapse
|
22
|
Yu H, Zhou J, Takahashi H, Yao W, Suzuki Y, Yuan X, Yoshimura SH, Zhang Y, Liu Y, Emmett N, Bond V, Wang D, Ding X, Takeyasu K, Yao X. Spatial control of proton pump H,K-ATPase docking at the apical membrane by phosphorylation-coupled ezrin-syntaxin 3 interaction. J Biol Chem 2014; 289:33333-42. [PMID: 25301939 PMCID: PMC4246090 DOI: 10.1074/jbc.m114.581280] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 10/08/2014] [Indexed: 11/06/2022] Open
Abstract
The digestive function of the stomach depends on acidification of the gastric lumen. Acid secretion into the lumen is triggered by activation of a cAMP-dependent protein kinase (PKA) cascade, which ultimately results in the insertion of gastric H,K-ATPases into the apical plasma membranes of parietal cells. A coupling protein is ezrin whose phosphorylation at Ser-66 by PKA is required for parietal cell activation. However, little is known regarding the molecular mechanism(s) by which ezrin operates in gastric acid secretion. Here we show that phosphorylation of Ser-66 induces a conformational change of ezrin that enables its association with syntaxin 3 (Stx3) and provides a spatial cue for H,K-ATPase trafficking. This conformation-dependent association is specific for Stx3, and the binding interface is mapped to the N-terminal region. Biochemical analyses show that inhibition of ezrin phosphorylation at Ser-66 prevents ezrin-Stx3 association and insertion of H,K-ATPase into the apical plasma membrane of parietal cells. Using atomic force microscopic analyses, our study revealed that phosphorylation of Ser-66 induces unfolding of ezrin molecule to allow Stx3 binding to its N terminus. Given the essential role of Stx3 in polarized secretion, our study presents the first evidence in which phosphorylation-induced conformational rearrangement of the ezrin molecule provides a spatial cue for polarized membrane trafficking in epithelial cells.
Collapse
Affiliation(s)
- Huijuan Yu
- From the Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, University of Science and Technology of China School of Life Science, Hefei, China 230027
| | - Jiajia Zhou
- From the Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, University of Science and Technology of China School of Life Science, Hefei, China 230027, Morehouse School of Medicine, Atlanta, Georgia 30310, and
| | - Hirohide Takahashi
- Laboratory of Plasma Membrane, Graduate School of Biostudies, Kyoto University, Kyoto 606-8502, Japan
| | - William Yao
- Morehouse School of Medicine, Atlanta, Georgia 30310, and
| | - Yuki Suzuki
- Laboratory of Plasma Membrane, Graduate School of Biostudies, Kyoto University, Kyoto 606-8502, Japan
| | - Xiao Yuan
- From the Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, University of Science and Technology of China School of Life Science, Hefei, China 230027
| | - Shige H Yoshimura
- Laboratory of Plasma Membrane, Graduate School of Biostudies, Kyoto University, Kyoto 606-8502, Japan
| | - Yin Zhang
- From the Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, University of Science and Technology of China School of Life Science, Hefei, China 230027, Graduate School, Beijing University of Chinese Medicine, Beijing 100086, China
| | - Ya Liu
- From the Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, University of Science and Technology of China School of Life Science, Hefei, China 230027
| | | | - Vincent Bond
- Morehouse School of Medicine, Atlanta, Georgia 30310, and
| | - Dongmei Wang
- From the Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, University of Science and Technology of China School of Life Science, Hefei, China 230027
| | - Xia Ding
- Graduate School, Beijing University of Chinese Medicine, Beijing 100086, China
| | - Kunio Takeyasu
- Laboratory of Plasma Membrane, Graduate School of Biostudies, Kyoto University, Kyoto 606-8502, Japan,
| | - Xuebiao Yao
- From the Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, University of Science and Technology of China School of Life Science, Hefei, China 230027,
| |
Collapse
|
23
|
Nakano T, Sekine S, Ito K, Horie T. Ezrin regulates the expression of Mrp2/Abcc2 and Mdr1/Abcb1 along the rat small intestinal tract. Am J Physiol Gastrointest Liver Physiol 2013; 305:G807-17. [PMID: 24091598 DOI: 10.1152/ajpgi.00187.2013] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Multidrug resistance-associated protein 2 (MRP2)/ATP-binding cassette protein C2 (ABCC2) and multidrug resistance protein 1 (MDR1)/ABCB1 are well-known efflux transporters located on the brush border membrane of the small intestinal epithelia, where they limit the absorption of a broad range of substrates. The expression patterns of MRP2/ABCC2 and MDR1/ABCB1 along the small intestinal tract are tightly regulated. Several reports have demonstrated the participation of ERM (ezrin/radixin/moesin) proteins in the posttranslational modulation of MRP2/ABCC2 and MDR1/ABCB1, especially with regard to their membrane localization. The present study focused on the in vivo expression profiles of MRP2/ABCC2, MDR1/ABCB1, ezrin, and phosphorylated ezrin to further elucidate the relationship between the efflux transporters and the ERM proteins. The current results showed good correlation between the phosphorylation status of ezrin and Mrp2/Abcc2 expression along the gastrointestinal tract of rats and between the expression profiles of both ezrin and Mdr1/Abcb1 in the small intestine. We also demonstrated the involvement of conventional protein kinase C isoforms in the regulation of ezrin phosphorylation. Furthermore, experiments conducted with wild-type (WT) ezrin and a T567A (Ala substituted Thr) dephosphorylated mutant showed a decrease in membrane surface-localized and total expressed MRP2/ABCC2 in T567A-expressing vs. WT ezrin-expressing Caco-2 cells. In contrast, T567A- and WT-expressing cells both showed an increase in membrane surface-localized and total expressed MDR1/ABCB1. These findings suggest that the phosphorylation status and the expression profile of ezrin differentially direct MRP2/ABCC2 and MDR1/ABCB1 expression, respectively, along the small intestinal tract.
Collapse
Affiliation(s)
- Takafumi Nakano
- Faculty of Pharmaceutical Sciences, Teikyo Heisei University, 4-21-2 Nakano, Nakano-ku, Tokyo 164-8530, Japan.
| | | | | | | |
Collapse
|
24
|
Adada M, Canals D, Hannun YA, Obeid LM. Sphingolipid regulation of ezrin, radixin, and moesin proteins family: implications for cell dynamics. Biochim Biophys Acta Mol Cell Biol Lipids 2013; 1841:727-37. [PMID: 23850862 DOI: 10.1016/j.bbalip.2013.07.002] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Revised: 06/30/2013] [Accepted: 07/02/2013] [Indexed: 12/13/2022]
Abstract
A key but poorly studied domain of sphingolipid functions encompasses endocytosis, exocytosis, cellular trafficking, and cell movement. Recently, the ezrin, radixin and moesin (ERM) family of proteins emerged as novel potent targets regulated by sphingolipids. ERMs are structural proteins linking the actin cytoskeleton to the plasma membrane, also forming a scaffold for signaling pathways that are used for cell proliferation, migration and invasion, and cell division. Opposing functions of the bioactive sphingolipid ceramide and sphingosine-1-phosphate (S1P), contribute to ERM regulation. S1P robustly activates whereas ceramide potently deactivates ERM via phosphorylation/dephosphorylation, respectively. This recent dimension of cytoskeletal regulation by sphingolipids opens up new avenues to target cell dynamics, and provides further understanding of some of the unexplained biological effects mediated by sphingolipids. In addition, these studies are providing novel inroads into defining basic mechanisms of regulation and action of bioactive sphingolipids. This review describes the current understanding of sphingolipid regulation of the cytoskeleton, it also describes the biologies in which ERM proteins have been involved, and finally how these two large fields have started to converge. This article is part of a Special Issue entitled New Frontiers in Sphingolipid Biology.
Collapse
Affiliation(s)
- Mohamad Adada
- The Department of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Daniel Canals
- The Department of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Yusuf A Hannun
- The Department of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Lina M Obeid
- The Department of Medicine, Stony Brook University, Stony Brook, NY 11794, USA; The Northport VA Medical Center, Northport, NY 11768, USA.
| |
Collapse
|
25
|
Solaymani-Mohammadi S, Singer SM. Regulation of intestinal epithelial cell cytoskeletal remodeling by cellular immunity following gut infection. Mucosal Immunol 2013; 6:369-78. [PMID: 22910215 PMCID: PMC4094376 DOI: 10.1038/mi.2012.80] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Gut infections often lead to epithelial cell damage followed by a healing response. We examined changes in the epithelial cell cytoskeleton and the involvement of host adaptive immunity in these events using an in vivo model of parasitic infection. We found that both ezrin and villin, key components of the actin cytoskeleton comprising the brush border (BB) of intestinal epithelial cells (IECs), underwent significant post-translational changes following gut infection and during the recovery phase of gut infection. Intriguingly, using mice lacking either CD4(+) or CD8(+) T-cell responses, we demonstrated that the mechanisms by which ezrin and villin are regulated in response to infection are different. Both ezrin and villin undergo proteolysis during the recovery phase of infection. Cleavage of ezrin requires CD4(+) but not CD8(+) T cells, whereas cleavage of villin requires both CD4(+) and CD8(+) T-cell responses. Both proteins were also regulated by phosphorylation; reduced levels of phosphorylated ezrin and increased levels of villin phosphorylation were observed at the peak of infection and correlated with reduced BB enzyme activity. Finally, we show that infection also leads to enhanced proliferation of IECs in this model. Cytoskeletal remodeling in IECs can have critical roles in the immunopathology and healing responses observed during many infectious and non-infectious intestinal conditions. These data indicate that cellular immune responses can be significant drivers of these processes.
Collapse
Affiliation(s)
- Shahram Solaymani-Mohammadi
- Department of Biology and Center for Infectious Disease, Georgetown University, Washington, DC, 20057, USA,Correspondence should be addressed to Shahram Solaymani-Mohammadi or Steven M. Singer, Mailing address: Shahram Solaymani-Mohammadi, University of California, San Diego, 9500 Gilman Drive, Mail Code 0623D, La Jolla CA 92093-0623 USA; Phone (858)534-4625; Fax: 858-534-5691; ; Steven M. Singer, Georgetown University, 406 Reiss Building, 37th and O Streets, NW, Washington, DC 20057 USA; Phone (202)-687-9884; Fax: (202)-687-5662;
| | - Steven M. Singer
- Department of Biology and Center for Infectious Disease, Georgetown University, Washington, DC, 20057, USA,Correspondence should be addressed to Shahram Solaymani-Mohammadi or Steven M. Singer, Mailing address: Shahram Solaymani-Mohammadi, University of California, San Diego, 9500 Gilman Drive, Mail Code 0623D, La Jolla CA 92093-0623 USA; Phone (858)534-4625; Fax: 858-534-5691; ; Steven M. Singer, Georgetown University, 406 Reiss Building, 37th and O Streets, NW, Washington, DC 20057 USA; Phone (202)-687-9884; Fax: (202)-687-5662;
| |
Collapse
|
26
|
Irie-Maezono R, Tsuyama S. Immunohistochemical Analysis of the Acid Secretion Potency in Gastric Parietal Cells. Cell 2013. [DOI: 10.4236/cellbio.2013.24020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
27
|
Bosk S, Braunger JA, Gerke V, Steinem C. Activation of F-actin binding capacity of ezrin: synergism of PIP₂ interaction and phosphorylation. Biophys J 2011; 100:1708-17. [PMID: 21463584 DOI: 10.1016/j.bpj.2011.02.039] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2010] [Revised: 02/18/2011] [Accepted: 02/23/2011] [Indexed: 11/27/2022] Open
Abstract
Ezrin is a membrane-cytoskeleton linker protein that can bind F-actin in its active conformation. Several means of regulation of ezrin's activity have been described including phosphorylation of Thr-567 and binding of L-α-phosphatidylinositol-4,5-bisphosphate (PIP(2)). However, the relative contributions of these events toward activation of the protein and their potential interdependence are not known. We developed an assay based on solid-supported membranes, to which different ezrin mutants (ezrin T567A (inactive mutant), wild-type, and T567D (active pseudophosphorylated mutant)) were bound, that enabled us to analyze the influence of phosphorylation and PIP(2) binding on ezrin's activation state in vitro. The lipid bilayers employed contained either DOGS-NTA-Ni to bind the proteins via an N-terminal His-tag, or PIP(2), to which ezrin binds via specific binding sites located in the N-terminal region of the protein. Quantitative analysis of the binding behavior of all three proteins to the two different receptor lipids revealed that all three bind with high affinity and specificity to the two receptor lipids. Fluorescence microscopy on ezrin-decorated solid-supported membranes showed that, dependent on the mode of binding and the phosphorylation state, ezrin is capable of binding actin filaments. A clear synergism between phosphorylation and the receptor lipid PIP(2) was observed, suggesting a conformational switch from the dormant to the active, F-actin binding state by recognition of PIP(2), which is enhanced by the phosphorylation.
Collapse
Affiliation(s)
- Sabine Bosk
- Institute of Organic and Biomolecular Chemistry, University of Göttingen, Göttingen, Germany
| | | | | | | |
Collapse
|
28
|
Chen Y, Wang D, Guo Z, Zhao J, Wu B, Deng H, Zhou T, Xiang H, Gao F, Yu X, Liao J, Ward T, Xia P, Emenari C, Ding X, Thompson W, Ma K, Zhu J, Aikhionbare F, Dou K, Cheng SY, Yao X. Rho kinase phosphorylation promotes ezrin-mediated metastasis in hepatocellular carcinoma. Cancer Res 2011; 71:1721-9. [PMID: 21363921 DOI: 10.1158/0008-5472.can-09-4683] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
During progression of hepatocellular carcinoma, multiple genetic and epigenetic alterations act to posttranslationally modulate the function of proteins that promote cancer invasion and metastasis. To define such abnormalities that contribute to liver cancer metastasis, we carried out a proteomic comparison of primary hepatocellular carcinoma and samples of intravascular thrombi from the same patient. Mass spectrometric analyses of the liver cancer samples revealed a series of acidic phospho-isotypes associated with the intravascular thrombi samples. In particular, we found that Thr567 hyperphosphorylation of the cytoskeletal protein ezrin was tightly correlated to an invasive phenotype of clinical hepatocellular carcinomas and to poor outcomes in tumor xenograft assays. Using phospho-mimicking mutants, we showed that ezrin phosphorylation at Thr567 promoted in vitro invasion by hepatocarcinoma cells. Phospho-mimicking mutant ezrinT567D, but not the nonphosphorylatable mutant ezrinT567A, stimulated formation of membrane ruffles, suggesting that Thr567 phosphorylation promotes cytoskeletal-membrane remodeling. Importantly, inhibition of Rho kinase, either by Y27632 or RNA interference, resulted in inhibition of Thr567 phosphorylation and a blockade to cell invasion, implicating Rho kinase-ezrin signaling in hepatocellular carcinoma cell invasion. Our findings suggest a strategy to reduce liver tumor metastasis by blocking Rho kinase-mediated phosphorylation of ezrin.
Collapse
Affiliation(s)
- Yong Chen
- Department of Hepatobiliary Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shanxi, P.R. China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Song P, Groos S, Riederer B, Feng Z, Krabbenhöft A, Manns MP, Smolka A, Hagen SJ, Neusch C, Seidler U. Kir4.1 channel expression is essential for parietal cell control of acid secretion. J Biol Chem 2011; 286:14120-8. [PMID: 21367857 DOI: 10.1074/jbc.m110.151191] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Kir4.1 channels were found to colocalize with the H(+)/K(+)-ATPase throughout the parietal cell (PC) acid secretory cycle. This study was undertaken to explore their functional role. Acid secretory rates, electrophysiological parameters, PC ultrastructure, and gene and protein expression were determined in gastric mucosae of 7-8-day-old Kir4.1-deficient mice and WT littermates. Kir4.1(-/-) mucosa secreted significantly more acid and initiated secretion significantly faster than WT mucosa. No change in PC number but a relative up-regulation of H(+)/K(+)-ATPase gene and protein expression (but not of other PC ion transporters) was observed. Electron microscopy revealed fully fused canalicular membranes and a lack of tubulovesicles in resting state Kir4.1(-/-) PCs, suggesting that Kir4.1 ablation may also interfere with tubulovesicle endocytosis. The role of this inward rectifier in the PC apical membrane may therefore be to balance between K(+) loss via KCNQ1/KCNE2 and K(+) reabsorption by the slow turnover of the H(+)/K(+)-ATPase, with consequences for K(+) reabsorption, inhibition of acid secretion, and membrane recycling. Our results demonstrate that Kir4.1 channels are involved in the control of acid secretion and suggest that they may also affect secretory membrane recycling.
Collapse
Affiliation(s)
- Penghong Song
- Department of Gastroenterology, Hepatology, and Endocrinology, Hannover Medical School, D-30625 Hannover, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Suda J, Zhu L, Karvar S. Phosphorylation of radixin regulates cell polarity and Mrp-2 distribution in hepatocytes. Am J Physiol Cell Physiol 2011; 300:C416-24. [DOI: 10.1152/ajpcell.00467.2010] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Radixin, the dominant ezrin-radixin-moesin (ERM) protein in hepatocytes, has two important binding domains: an NH2-terminal region that binds to plasma membrane and a COOH-terminal region that binds to F-actin after a conformational activation by phosphorylation at Thr564. The present studies were undertaken to investigate the cellular changes in expression of radixin in WIF-B cells and to assess radixin distribution and its influence on cell polarity. We used a recombinant adenoviral expression system encoding radixin wild-type and Thr564 mutants fused to cyan fluorescent protein (CFP), as well as conventional immunostaining procedures. Functional analyses were characterized quantitatively. Similar to endogenous radixin, adenovirus-infected radixin-CFP-wild type and nonphosphorylatable radixin-CFP-T564A were found to be expressed heavily in the compartment of canalicular membrane vacuoles, typically colocalizing with multidrug resistance-associated protein 2 (Mrp-2). Expression of radixin-CFP-T564D, which mimics constant phosphorylation, was quite different, being rarely associated with canalicular membranes. The WIF-B cells were devoid of a secretory response, T567D radixin became predominantly redistributed to the basolateral membrane, usually in the form of dense, long spikes and fingerlike projections, and the altered cell polarity involved changes in apical membrane markers. Differences in polar distribution of radixin suggest a role for the linker protein in promoting formation and plasticity of membrane surface projections and also suggest that radixin might be an organizer and regulator of Mrp-2 and cell polarity in hepatocytes.
Collapse
Affiliation(s)
- Jo Suda
- Division of Gastrointestinal and Liver Diseases, Keck School of Medicine, University of Southern California, Los Angeles, California; and
| | - Lixin Zhu
- Digestive Diseases and Nutrition Center, University at Buffalo, State University of New York, Buffalo, New York
| | - Serhan Karvar
- Division of Gastrointestinal and Liver Diseases, Keck School of Medicine, University of Southern California, Los Angeles, California; and
| |
Collapse
|
31
|
Chirivino D, Del Maestro L, Formstecher E, Hupé P, Raposo G, Louvard D, Arpin M. The ERM proteins interact with the HOPS complex to regulate the maturation of endosomes. Mol Biol Cell 2010; 22:375-85. [PMID: 21148287 PMCID: PMC3031467 DOI: 10.1091/mbc.e10-09-0796] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
In the degradative pathway, the progression of cargos through endosomal compartments involves a series of fusion and maturation events. The HOPS (homotypic fusion and protein sorting) complex is part of the machinery that promotes the progression from early to late endosomes and lysosomes by regulating the exchange of small GTPases. We report that an interaction between subunits of the HOPS complex and the ERM (ezrin, radixin, moesin) proteins is required for the delivery of EGF receptor (EGFR) to lysosomes. Inhibiting either ERM proteins or the HOPS complex leads to the accumulation of the EGFR into early endosomes, delaying its degradation. This impairment in EGFR trafficking observed in cells depleted of ERM proteins is due to a delay in the recruitment of Rab7 on endosomes. As a consequence, the maturation of endosomes is perturbed as reflected by an accumulation of hybrid compartments positive for both early and late endosomal markers. Thus, ERM proteins represent novel regulators of the HOPS complex in the early to late endosomal maturation.
Collapse
Affiliation(s)
- Dafne Chirivino
- Institut Curie-Unité Mixte de Recherche 144 (UMR144), Centre National de la Recherche Scientifique (CNRS)/Morphogenèse et Signalisation cellulaires, Paris, France
| | | | | | | | | | | | | |
Collapse
|
32
|
Wang L, Chen W, Zhao C, Huo R, Guo XJ, Lin M, Huang XY, Mao YD, Zhou ZM, Sha JH. The role of ezrin-associated protein network in human sperm capacitation. Asian J Androl 2010; 12:667-76. [PMID: 20711218 PMCID: PMC3739321 DOI: 10.1038/aja.2010.79] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2010] [Revised: 04/28/2010] [Accepted: 06/11/2010] [Indexed: 11/09/2022] Open
Abstract
Membrane modifications in sperm cells represent a key step in sperm capacitation; however, the molecular basis of these modifications is not fully understood. Ezrin is the best-studied member of the ezrin/radixin/merlin family. As a cross-linker between the cortical cytoskeleton and plasma membrane proteins, ezrin contributes to remodeling of the membrane surface structure. Furthermore, activated ezrin and the Rho dissociation inhibitor, RhoGDI, promote the formation of cortical cytoskeleton-polymerized actin through Rho activation. Thus, ezrin, actin, RhoGDI, Rho and plasma membrane proteins form a complicated network in vivo, which contributes to the assembly of the structure of the membrane surface. Previously, we showed that ezrin and RhoGDI1 are expressed in human testes. Thus, we sought to determine whether the ezrin-RhoGDI1-actin-membrane protein network has a role in human sperm capacitation. Our results by Western blot indicate that ezrin is activated by phosphorylation of the threonine567 residue during capacitation. Co-immunoprecipitation studies revealed that, during sperm capacitation, the interaction between ezrin and RhoGDI1 increases, and phosphostaining of two dimensional electrophoresis gels showed that RhoGDI1 is phosphorylated, suggesting that RhoGDI1 dissociates from RhoA and leads to actin polymerization on the sperm head. We speculate that activated ezrin interacts with polymerized actin and the glycosylated membrane protein cd44 after capacitation. Blocking sperm capacitation using ezrin- or actin-specific monoclonal antibodies decreases their acrosome reaction (AR) rate, but has no effect on the AR alone. Taken together, our results show that a network consisting of ezrin, RhoGDI1, RhoA, F-actin and membrane proteins functions to influence the modifications that occur on the membrane of the sperm head during human sperm capacitation.
Collapse
Affiliation(s)
- Lei Wang
- Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical University, Nanjing 210029, China
| | - Wen Chen
- Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical University, Nanjing 210029, China
| | - Chun Zhao
- Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical University, Nanjing 210029, China
| | - Ran Huo
- Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical University, Nanjing 210029, China
| | - Xue-Jiang Guo
- Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical University, Nanjing 210029, China
| | - Min Lin
- Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical University, Nanjing 210029, China
| | - Xiao-Yan Huang
- Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical University, Nanjing 210029, China
| | - Yun-Dong Mao
- Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical University, Nanjing 210029, China
- Department of Reproductive Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Zuo-Min Zhou
- Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical University, Nanjing 210029, China
| | - Jia-Hao Sha
- Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
33
|
|
34
|
Zhu L, Crothers J, Zhou R, Forte JG. A possible mechanism for ezrin to establish epithelial cell polarity. Am J Physiol Cell Physiol 2010; 299:C431-43. [PMID: 20505040 DOI: 10.1152/ajpcell.00090.2010] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Ezrin is an important membrane/actin cytoskeleton linker protein, especially in epithelia. Ezrin has two important binding domains: an NH(2)-terminal region that binds to plasma membrane and a COOH-terminal region that binds to F-actin only after a conformational activation by phosphorylation at Thr567 of ezrin. The present experiments were undertaken to investigate the detailed cellular changes in the time course of expression of ezrin-T567 mutants (nonphosphorylatable T567A and permanent phospho-mimic T567D) in parietal cells and to assess ezrin distribution and its influence on the elaborate membrane recruitment processes of these cells. T567A mutant and wild-type (WT) ezrin were consistently localized to the apical plasma membrane, even with overexpression. On the other hand, T567D went first to apical membrane at early times and low expression levels, then accumulated mainly at the basal surface after 24 h. Overexpression of WT or T567A led to incorporation of internal membranes to apical vacuoles, while overexpression of T567D led to large incorporation of apical and intracellular membranes (including H-K-ATPase) to the basal surface. Differences in polar distribution of ezrin suggest a role for the linker protein in promoting formation and plasticity of membrane surface projections, forming the basis for a novel theory for ezrin as an organizer and regulator of membrane recruitment. A model simulating the cellular distribution of ezrin and its associated membrane- and F-actin-binding forms is given to predict redistributions observed with phosphorylation and mutant overexpression, and it can easily be modified as more specific information regarding binding constants and specific sites becomes available.
Collapse
Affiliation(s)
- Lixin Zhu
- Department of Pediatrics, State University of New York at Buffalo, Buffalo, New York, USA
| | | | | | | |
Collapse
|
35
|
Ding X, Deng H, Wang D, Zhou J, Huang Y, Zhao X, Yu X, Wang M, Wang F, Ward T, Aikhionbare F, Yao X. Phospho-regulated ACAP4-Ezrin interaction is essential for histamine-stimulated parietal cell secretion. J Biol Chem 2010; 285:18769-80. [PMID: 20360010 DOI: 10.1074/jbc.m110.129007] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The ezrin-radixin-moesin proteins provide a regulated linkage between membrane proteins and the cortical cytoskeleton and also participate in signal transduction pathways. Ezrin is localized to the apical membrane of parietal cells and couples the protein kinase A activation cascade to the regulated HCl secretion. Our recent proteomic study revealed a protein complex of ezrin-ACAP4-ARF6 essential for volatile membrane remodeling (Fang, Z., Miao, Y., Ding, X., Deng, H., Liu, S., Wang, F., Zhou, R., Watson, C., Fu, C., Hu, Q., Lillard, J. W., Jr., Powell, M., Chen, Y., Forte, J. G., and Yao, X. (2006) Mol. Cell Proteomics 5, 1437-1449). However, knowledge of whether ACAP4 physically interacts with ezrin and how their interaction is integrated into membrane-cytoskeletal remodeling has remained elusive. Here we provide the first evidence that ezrin interacts with ACAP4 in a protein kinase A-mediated phosphorylation-dependent manner through the N-terminal 400 amino acids of ACAP4. ACAP4 locates in the cytoplasmic membrane in resting parietal cells but translocates to the apical plasma membrane upon histamine stimulation. ACAP4 was precipitated with ezrin from secreting but not resting parietal cell lysates, suggesting a phospho-regulated interaction. Indeed, this interaction is abolished by phosphatase treatment and validated by an in vitro reconstitution assay using phospho-mimicking ezrin(S66D). Importantly, ezrin specifies the apical distribution of ACAP4 in secreting parietal cells because either suppression of ezrin or overexpression of non-phosphorylatable ezrin prevents the apical localization of ACAP4. In addition, overexpressing GTPase-activating protein-deficient ACAP4 results in an inhibition of apical membrane-cytoskeletal remodeling and gastric acid secretion. Taken together, these results define a novel molecular mechanism linking ACAP4-ezrin interaction to polarized epithelial secretion.
Collapse
Affiliation(s)
- Xia Ding
- Anhui Key Laboratory of Cellular Dynamics and Chemical Biology, University of Science and Technology of China, Hefei 230027, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Affiliation(s)
- John G. Forte
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720;
| | - Lixin Zhu
- Department of Pediatrics, Digestive Disease and Nutrition Center, The State University of New York, Buffalo, New York 14214;
| |
Collapse
|
37
|
Internalization of NK cells into tumor cells requires ezrin and leads to programmed cell-in-cell death. Cell Res 2009; 19:1350-62. [PMID: 19786985 DOI: 10.1038/cr.2009.114] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Cytotoxic lymphocytes are key players in the orchestration of immune response and elimination of defective cells. We have previously reported that natural killer (NK) cells enter target tumor cells, leading to either target cell death or self-destruction within tumor cells. However, it has remained elusive as to the fate of NK cells after internalization and whether the heterotypic cell-in-cell process is different from that of the homotypic cell-in-cell event recently named entosis. Here, we show that NK cells undergo a cell-in-cell process with the ultimate fate of apoptosis within tumor cells and reveal that the internalization process requires the actin cytoskeletal regulator, ezrin. To visualize how NK cells enter into tumor cells, we carried out real-time dual color imaging analyses of NK cell internalization into tumor cells. Surprisingly, most NK cells commit to programmed cell death after their entry into tumor cells, which is distinctively different from entosis observed in the homotypic cell-in-cell process. The apoptotic cell death of the internalized NK cells was evident by activation of caspase 3 and DNA fragmentation. Furthermore, NK cell death after internalization is attenuated by the caspase inhibitor, Z-VAD-FMK, confirming apoptosis as the mode of NK cell death within tumor cells. To determine protein factors essential for the entry of NK cells into tumor cells, we carried out siRNA-based knockdown analysis and discovered a critical role of ezrin in NK cell internalization. Importantly, PKA-mediated phosphorylation of ezrin promotes the NK cell internalization process. Our findings suggest a novel regulatory mechanism by which ezrin governs NK cell internalization into tumor cells.
Collapse
|
38
|
Di Sole F, Babich V, Moe OW. The calcineurin homologous protein-1 increases Na(+)/H(+) -exchanger 3 trafficking via ezrin phosphorylation. J Am Soc Nephrol 2009; 20:1776-86. [PMID: 19556366 DOI: 10.1681/asn.2008121255] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
The Na(+)/H(+)-exchanger 3 (NHE3) is essential for regulation of Na(+) transport in the renal and intestinal epithelium. Although changes in cell surface abundance control NHE3 function, the molecular signals that regulate NHE3 surface expression are not well defined. We found that overexpression of the calcineurin homologous protein-1 (CHP1) in opossum kidney cells increased NHE3 transport activity, surface protein abundance, and ezrin phosphorylation. CHP1 knockdown by small interfering RNA had the opposite effects. Overexpression of wild-type ezrin increased both NHE3 transport activity and surface protein abundance, confirming that NHE3 is downstream of ezrin. Expression of a pseudophosphorylated ezrin enhanced these effects, whereas expression of an ezrin variant that could not be phosphorylated prevented the downstream effects on NHE3. Furthermore, CHP1 knockdown reversed the activation of NHE3 by wild-type ezrin but not by the pseudophosphorylated ezrin. Taken together, these results demonstrate that CHP1 increases NHE3 abundance and constitutive function in a manner dependent on ezrin phosphorylation.
Collapse
Affiliation(s)
- Francesca Di Sole
- Department of Internal Medicine, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390-8885, USA.
| | | | | |
Collapse
|
39
|
Wang F, Xia P, Wu F, Wang D, Wang W, Ward T, Liu Y, Aikhionbare F, Guo Z, Powell M, Liu B, Bi F, Shaw A, Zhu Z, Elmoselhi A, Fan D, Cover TL, Ding X, Yao X. Helicobacter pylori VacA disrupts apical membrane-cytoskeletal interactions in gastric parietal cells. J Biol Chem 2008; 283:26714-25. [PMID: 18625712 DOI: 10.1074/jbc.m800527200] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Helicobacter pylori persistently colonize the human stomach and have been linked to atrophic gastritis and gastric carcinoma. Although it is well known that H. pylori infection can result in hypochlorhydria, the molecular mechanisms underlying this phenomenon remain poorly understood. Here we show that VacA permeabilizes the apical membrane of gastric parietal cells and induces hypochlorhydria. The functional consequences of VacA infection on parietal cell physiology were studied using freshly isolated rabbit gastric glands and cultured parietal cells. Secretory activity of parietal cells was judged by an aminopyrine uptake assay and confocal microscopic examination. VacA permeabilization induces an influx of extracellular calcium, followed by activation of calpain and subsequent proteolysis of ezrin at Met(469)-Thr(470), which results in the liberation of ezrin from the apical membrane of the parietal cells. VacA treatment inhibits acid secretion by preventing the recruitment of H,K-ATPase-containing tubulovesicles to the apical membrane of gastric parietal cells. Electron microscopic examination revealed that VacA treatment disrupts the radial arrangement of actin filaments in apical microvilli due to the loss of ezrin integrity in parietal cells. Significantly, expression of calpain-resistant ezrin restored the functional activity of parietal cells in the presence of VacA. Proteolysis of ezrin in VacA-infected parietal cells is a novel mechanism underlying H. pylori-induced inhibition of acid secretion. Our results indicate that VacA disrupts the apical membrane-cytoskeletal interactions in gastric parietal cells and thereby causes hypochlorhydria.
Collapse
Affiliation(s)
- Fengsong Wang
- Morehouse School of Medicine, Atlanta, Georgia 30310, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Pagliocca A, Hegyi P, Venglovecz V, Rackstraw SA, Khan Z, Burdyga G, Wang TC, Dimaline R, Varro A, Dockray GJ. Identification of ezrin as a target of gastrin in immature mouse gastric parietal cells. Exp Physiol 2008; 93:1174-89. [PMID: 18567601 DOI: 10.1113/expphysiol.2008.042648] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The gastric acid-secreting parietal cell exhibits profound morphological changes on stimulation. Studies in gastrin null (Gas-KO) mice indicate that maturation of parietal cell function depends on the hormone gastrin acting at the G-protein-coupled cholecystokinin 2 receptor. The relevant cellular mechanisms are unknown. The application of differential mRNA display to samples of the gastric corpus of wild-type (C57BL/6) and Gas-KO mice identified the cytoskeletal linker protein, ezrin, as a previously unsuspected target of gastrin. Gastrin administered in vivo or added to gastric glands in vitro increased ezrin abundance in Gas-KO parietal cells. In parietal cells of cultured gastric glands from wild-type mice treated with gastrin, histamine or carbachol, ezrin was localized to vesicular structures resembling secretory canaliculi. In contrast, in cultured parietal cells from Gas-KO mice, ezrin was typically distributed in the cytosol, and this did not change after incubation with gastrin, histamine or carbachol. However, priming with gastrin for approximately 24 h, either in vivo prior to cell culture or by addition to cultured gastric glands, induced the capacity for secretagogue-stimulated localization of ezrin to large vesicular structures in Gas-KO mice. Similarly, in a functional assay based on measurement of intracellular pH, cultured parietal cells from Gas-KO mice were refractory to gastrin unless primed. The priming effect of gastrin was not attributable to the paracrine mediator histamine, but was prevented by inhibitors of protein kinase C and transactivation of the epidermal growth factor receptor. We conclude that in gastrin null mice there is reduced ezrin expression and a defect in ezrin subcellular distribution in gastric parietal cells, and that both can be reversed by priming with gastrin.
Collapse
Affiliation(s)
- Adelina Pagliocca
- Physiological Laboratory, University of Liverpool, Crown Street, Liverpool L69 3BX, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Zhu L, Hatakeyama J, Chen C, Shastri A, Poon K, Forte JG. Comparative study of ezrin phosphorylation among different tissues: more is good; too much is bad. Am J Physiol Cell Physiol 2008; 295:C192-202. [PMID: 18480298 DOI: 10.1152/ajpcell.00159.2008] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In a comparison of three different tissues, the membrane cytoskeleton linker protein ezrin was found to assume high levels of phosphorylation on threonine-567 (T567) in the brush border membranes of renal proximal tubule cells and small intestine enterocytes, in contrast to the apical canalicular membrane of gastric parietal cells. Together with an earlier observation that increased T567 phosphorylation is associated with more elaborate microvilli in parietal cells, this comparative study suggested a higher phosphorylation level requirement for the denser and more uniform distribution of microvilli at brush border surfaces. Using a kinase inhibitor, staurosporin, and metabolic inhibitor, sodium azide, relatively high turnover of ezrin T567 phosphorylation was observed in all three epithelia. Aiming to understand the role of phosphorylation turnover in these tissues, detergent extraction analysis of gastric glands and proximal tubules revealed that an increased phosphorylation on ezrin T567 greatly enhanced its association with F-actin, while ezrin-membrane interaction persisted regardless of the changes of phosphorylation level on ezrin T567. Finally, expression of Thr567Asp mutant ezrin, which mimics the phospho-ezrin state but does not allow turnover, caused aberrant growth of membrane projections in cultured proximal tubule cells, consistent with what had previously been observed in several cell lines and gastric parietal cells. These results fit into a model of surface plasticity, which posits that the turnover of phosphorylation on T567 empowers ezrin to relax and reposition membrane to the underlying cytoskeleton under varying conditions of filament growth or rapid membrane expansion (or depletion).
Collapse
Affiliation(s)
- Lixin Zhu
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720-3200, USA
| | | | | | | | | | | |
Collapse
|
42
|
Compartmentalized cAMP signalling in regulated exocytic processes in non-neuronal cells. Cell Signal 2007; 20:590-601. [PMID: 18061403 DOI: 10.1016/j.cellsig.2007.10.020] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2007] [Accepted: 10/16/2007] [Indexed: 01/04/2023]
Abstract
Cyclic adenosine monophosphate (cAMP) is a central second messenger controlling a plethora of vital functions. Studies of cAMP dynamics in living cells have revealed markedly inhomogeneous concentrations of the second messenger in different compartments. Moreover, cAMP effectors such as cAMP-dependent protein kinase (PKA) and cAMP-activated GTP-exchange factors (Epacs) are tethered to specific cellular sites. Both the tailoring of cAMP concentrations, and the activities of cAMP-dependent signalling systems at specific cellular locations are prerequisites for most, if not all, cAMP-dependent processes. This review focuses on the role of compartmentalized cAMP signalling in exocytic processes in non-neuronal cells. Particularly, the insertion of aquaporin-2 into the plasma membrane of renal principal cells as an example for a cAMP-dependent exocytic process in a non-secretory cell type, renin secretion from juxtaglomerular cells as a cAMP-triggered exocytosis from an endocrine cell, insulin release from pancreatic beta-cells as a Ca2+-mediated and cAMP-potentiated exocytic processes in an endocrine cell, and cAMP- or Ca2+ -triggered H+ secretion from gastric parietal cells as an exocytic process in an exocrine cell are discussed. The selected examples of cAMP-regulated exocytic pathways are reviewed with regard to key proteins involved: adenylyl cyclases, phosphodiesterases, PKA, A kinase anchoring proteins (AKAPs) and Epacs.
Collapse
|
43
|
Mettler SE, Ghayouri S, Christensen GP, Forte JG. Modulatory role of phosphoinositide 3-kinase in gastric acid secretion. Am J Physiol Gastrointest Liver Physiol 2007; 293:G532-43. [PMID: 17569740 DOI: 10.1152/ajpgi.00138.2007] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The gastric parietal cell is responsible for the secretion of HCl into the lumen of the stomach mainly due to stimulation by histamine via the cAMP pathway. However, the participation of several other receptors and pathways have been discovered to influence both stimulation and inhibition of acid secretion (e.g., cholinergic). Here we examine the role of phosphoinositide 3-kinase (PI3K) in the modulation of acid secretion. Treatment of isolated gastric glands and parietal cells with the PI3K inhibitor, LY294002 (LY), potentiated acid secretion in response to histamine to nearly the maximal secretion obtained with histamine plus phosphodiesterase inhibitors. As cAMP levels were elevated in response to histamine plus LY, but other means of elevating cAMP (e.g., forskolin, dbcAMP) were not influenced by LY, we posited that the effect might require activation of G-protein-coupled histamine H(2) receptors, possibly through the protein kinase B pathway (also known as Akt). Study of downstream effectors of PI3K showed that histaminergic stimulation increased Akt phosphorylation, which in turn was blocked by inhibition of PI3K. Expression studies showed that high expression of active Akt decreased acid secretion, whereas dominant-negative Akt increased acid secretion. Taken together, these data suggest stimulation with histamine increases the activity of PI3K leading to increased activity of Akt and decreased levels of cAMP in the parietal cell.
Collapse
Affiliation(s)
- S E Mettler
- Department of Molecular and Cell Biology, University of California-Berkeley, Berkeley, CA 94720-3200, USA
| | | | | | | |
Collapse
|
44
|
Liu Y, Wang D, Ding X, Deng H, Feng M, Yu X, Jiang K, Ward T, Guo Z, Forte J, Yao X. A mechanism of Munc18b-syntaxin 3-SNAP25 complex assembly in regulated epithelial secretion. FEBS Lett 2007; 581:4318-24. [PMID: 17716669 PMCID: PMC3690314 DOI: 10.1016/j.febslet.2007.07.083] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2007] [Revised: 07/01/2007] [Accepted: 07/31/2007] [Indexed: 01/14/2023]
Abstract
Syntaxin and Munc18 are essential for regulated exocytosis in all eukaryotes. It was shown that Munc18 inhibition of neuronal syntaxin 1 can be overcome by CDK5 phosphorylation, indicating that structural change disrupts the syntaxin-Munc18 interaction. Here, we show that this phosphorylation promotes the assembly of Munc18b-syntaxin 3-SNAP25 tripartite complex and membrane fusion machinery SNARE. Using siRNAs to screen for genes required for regulated epithelial secretion, we identified the requirements of CDK5 and Munc18b in cAMP-dependent gastric acid secretion. Biochemical characterization revealed that Munc18b bears a syntaxin 3-selective binding site located at its most C-terminal 53 amino acids. Significantly, the phosphorylation of Thr572 by CDK5 attenuates Munc18b-syntaxin 3 interaction and promotes formation of Munc18b-syntaxin 3-SNAP25 tripartite complex, leading to an assembly of functional Munc18b-syntaxin 3-SNAP25-VAMP2 membrane fusion machinery. Thus, our studies suggest a novel regulatory mechanism in which phosphorylation of Munc18b operates vesicle docking and fusion in regulated exocytosis.
Collapse
Affiliation(s)
- Ya Liu
- Cellular Dynamics, Hefei National Laboratory & University of Science & Technology of China, Hefei, CHINA 230027
| | - Dongmei Wang
- Cellular Dynamics, Hefei National Laboratory & University of Science & Technology of China, Hefei, CHINA 230027
- Department of Medicine, Beijing University of Chinese Medicine, Beijing, China 100029
| | - Xia Ding
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA 30310
| | - Hui Deng
- Cellular Dynamics, Hefei National Laboratory & University of Science & Technology of China, Hefei, CHINA 230027
| | - Mingye Feng
- Cellular Dynamics, Hefei National Laboratory & University of Science & Technology of China, Hefei, CHINA 230027
| | - Xue Yu
- Cellular Dynamics, Hefei National Laboratory & University of Science & Technology of China, Hefei, CHINA 230027
| | - Kai Jiang
- Cellular Dynamics, Hefei National Laboratory & University of Science & Technology of China, Hefei, CHINA 230027
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA 30310
| | - Tarsha Ward
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA 30310
| | - Zhen Guo
- Cellular Dynamics, Hefei National Laboratory & University of Science & Technology of China, Hefei, CHINA 230027
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA 30310
| | - John Forte
- Department of Molecular & Cell Biology, University of California, Berkeley, CA 94720, USA
| | - Xuebiao Yao
- Cellular Dynamics, Hefei National Laboratory & University of Science & Technology of China, Hefei, CHINA 230027
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA 30310
- All correspondence addressed to: Xuebiao Yao,
| |
Collapse
|
45
|
Liu D, Ge L, Wang F, Takahashi H, Wang D, Guo Z, Yoshimura SH, Ward T, Ding X, Takeyasu K, Yao X. Single-molecule detection of phosphorylation-induced plasticity changes during ezrin activation. FEBS Lett 2007; 581:3563-71. [PMID: 17628548 DOI: 10.1016/j.febslet.2007.06.071] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2007] [Revised: 06/05/2007] [Accepted: 06/26/2007] [Indexed: 11/21/2022]
Abstract
Ezrin-radixin-moesin protein family provides a regulated link between the cortical actin cytoskeleton and the plasma membrane. Phosphorylation of ezrin has been functionally linked to membrane dynamics and plasticity. Our recent study demonstrated that phosphorylation of the conserved T567 residue of ezrin alters the physiology of gastric parietal cells. However, the molecular mechanism of phosphorylation-induced ezrin activation has remained elusive. Here we use atomic force microscopy (AFM) to probe phosphorylation-mediated activation of ezrin in single molecules. The phospho-mimicking and non-phosphorylatable mutant ezrin proteins were generated and purified to homogeneity. Comparative analyses of two ezrin mutants by AFM demonstrate the unfolding of the N- and C-terminal domains upon the phospho-activation. To measure the physical force underlying the inter-domain contact during mechanical unfolding, we probed the defined region of ezrin using the N-terminal ezrin coated onto the AFM tip. Comparative force measurements indicate that T567 phosphorylation-induced unfolding of ezrin favors the inter-molecular association. Taken together, these results provide molecular illustration of phosphorylation elicited functional activation of ERM proteins and indicate that stimulus-induced protein conformational change can be used as a signaling mechanism orchestrating cellular dynamics.
Collapse
Affiliation(s)
- Dan Liu
- Division of Cellular Dynamics, Hefei National Laboratory for Physical Sciences, Hefei 230027, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Zhu L, Zhou R, Mettler S, Wu T, Abbas A, Delaney J, Forte JG. High turnover of ezrin T567 phosphorylation: conformation, activity, and cellular function. Am J Physiol Cell Physiol 2007; 293:C874-84. [PMID: 17553936 DOI: 10.1152/ajpcell.00111.2007] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In its dormant state, the membrane cytoskeletal linker protein ezrin takes on a NH(2) terminal-to-COOH terminal (N-C) binding conformation. In vitro evidence suggests that eliminating the N-C binding conformation by Thr(567) phosphorylation leads to ezrin activation. Here, we found for resting gastric parietal cells that the levels of ezrin phosphorylation on Thr(567) are low and can be increased to a small extent ( approximately 40%) by stimulating secretion via the cAMP pathway. Treatment of cells with protein phosphatase inhibitors led to a rapid, dramatic increase in Thr(567) phosphorylation by 400% over resting levels, prompting the hypothesis that ezrin activity is regulated by turnover of phosphorylation on Thr(567). In vitro and in vivo fluorescence resonance energy transfer analysis demonstrated that Thr(567) phosphorylation opens the N-C interaction. However, even in the closed conformation, ezrin localizes to membranes by an exposed NH(2) terminal binding site. Importantly, the opened phosphorylated form of ezrin more readily cosediments with F-actin and binds more tightly to membrane than the closed forms. Furthermore, fluorescence recovery after photobleaching analysis in live cells showed that the Thr567Asp mutant had longer recovery times than the wild type or the Thr567Ala mutant, indicating the Thr(567)-phosphorylated form of ezrin is tightly associated with F-actin and the membrane, restricting normal activity. These data demonstrate and emphasize the functional importance of reversible phosphorylation of ezrin on F-actin binding. A novel model is proposed whereby ezrin and closely associated kinase and phosphatase proteins represent a motor complex to maintain a dynamic relationship between the varying membrane surface area and filamentous actin length.
Collapse
Affiliation(s)
- Lixin Zhu
- 241 LSA, Dept. of Molecular and Cell Biology, Univ. of California, Berkeley, CA 94720-3200, USA
| | | | | | | | | | | | | |
Collapse
|
47
|
Tamma G, Procino G, Svelto M, Valenti G. Hypotonicity causes actin reorganization and recruitment of the actin-binding ERM protein moesin in membrane protrusions in collecting duct principal cells. Am J Physiol Cell Physiol 2007; 292:C1476-84. [PMID: 17428844 DOI: 10.1152/ajpcell.00375.2006] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Hypotonicity-induced cell swelling is characterized by a modification in cell architecture associated with actin cytoskeleton remodeling. The ezrin/radixin/moesin (ERM) family proteins are important signal transducers during actin reorganization regulated by the monomeric G proteins of the Rho family. We report here that in collecting duct CD8 cells hypotonicity-induced cell swelling resulted in deep actin reorganization, consisting of loss of stress fibers and formation of F-actin patches in membrane protrusions where the ERM protein moesin was recruited. Cell swelling increased the interaction between actin and moesin and induced the transition of moesin from an oligomeric to a monomeric functional conformation, characterized by both the COOH- and NH2-terminal domains being exposed. In this conformation, which is stabilized by phosphorylation of a conserved threonine in the COOH-terminal domain by PKC or Rho kinase, moesin can bind interacting proteins. Interestingly, hypotonic stress increased the amount of threonine-phosphorylated moesin, which was prevented by the PKC-α inhibitor Gö-6976 (50 nM). In contrast, the Rho kinase inhibitor Y-27632 (1 μM) did not affect the hypotonicity-induced increase in phosphorylated moesin. The present data represent the first evidence that hypotonicity-induced actin remodeling is associated with phosphorylated moesin recruitment at the cell border and interaction with actin.
Collapse
Affiliation(s)
- Grazia Tamma
- Dipartimento di Fisiologia Generale e Ambientale, Via Amendola 165/A, 70126 Bari, Italy
| | | | | | | |
Collapse
|
48
|
Fiévet B, Louvard D, Arpin M. ERM proteins in epithelial cell organization and functions. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2006; 1773:653-60. [PMID: 16904765 DOI: 10.1016/j.bbamcr.2006.06.013] [Citation(s) in RCA: 137] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2006] [Revised: 05/18/2006] [Accepted: 06/30/2006] [Indexed: 11/17/2022]
Abstract
ERM (Ezrin, Radixin, Moesin) proteins are membrane-cytoskeleton linkers that regulate the structure and the function of specific domains of the plasma membrane. ERM proteins are expressed in all metazoan analyzed so far. Genetic analysis of ERM protein functions has recently been performed simultaneously in three different organisms, mouse, Drosophila melanogaster and C. elegans. These studies have revealed a remarkable conservation of the protein functions through evolution. Moreover they have shed light on the crucial role these proteins play in various physiological processes that occur in epithelial cells.
Collapse
Affiliation(s)
- Bruno Fiévet
- UMR 144 CNRS-Institut Curie 26 rue d'Ulm, 75248 PARIS cedex 05, France
| | | | | |
Collapse
|
49
|
Fang Z, Miao Y, Ding X, Deng H, Liu S, Wang F, Zhou R, Watson C, Fu C, Hu Q, Lillard JW, Powell M, Chen Y, Forte JG, Yao X. Proteomic identification and functional characterization of a novel ARF6 GTPase-activating protein, ACAP4. Mol Cell Proteomics 2006; 5:1437-49. [PMID: 16737952 DOI: 10.1074/mcp.m600050-mcp200] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
ARF6 GTPase is a conserved regulator of membrane trafficking and actin-based cytoskeleton dynamics at the leading edge of migrating cells. A key determinant of ARF6 function is the lifetime of the GTP-bound active state, which is orchestrated by GTPase-activating protein (GAP) and GTP-GDP exchanging factor. However, very little is known about the molecular mechanisms underlying ARF6-mediated cell migration. To systematically analyze proteins that regulate ARF6 activity during cell migration, we performed a proteomic analysis of proteins selectively bound to active ARF6 using mass spectrometry and identified a novel ARF6-specific GAP, ACAP4. ACAP4 encodes 903 amino acids and contains two coiled coils, one pleckstrin homology domain, one GAP motif, and two ankyrin repeats. Our biochemical characterization demonstrated that ACAP4 has a phosphatidylinositol 4,5-bisphosphate-dependent GAP activity specific for ARF6. The co-localization of ACAP4 with ARF6 occurred in ruffling membranes formed upon AIF(4) and epidermal growth factor stimulation. ACAP4 overexpression limited the recruitment of ARF6 to the membrane ruffles in the absence of epidermal growth factor stimulation. Expression of GTP hydrolysis-resistant ARF6(Q67L) resulted in accumulations of ACAP4 and ARF6 in the cytoplasmic membrane, suggesting that GTP hydrolysis is required for the ARF6-dependent membrane remodeling. Significantly the depletion of ACAP4 by small interfering RNA or inhibition of ARF6 GTP hydrolysis by overexpressing GAP-deficient ACAP4 suppressed ARF6-dependent cell migration in wound healing, demonstrating the importance of ACAP4 in cell migration. Thus, our study sheds new light on the biological function of ARF6-mediated cell migration.
Collapse
Affiliation(s)
- Zhiyou Fang
- Laboratory of Cellular Dynamics, Hefei National Laboratory for Physical Sciences at Microscale, Hefei 230027, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|