1
|
Dekker PM, Boeren S, Saccenti E, Hettinga KA. Network analysis of the proteome and peptidome sheds light on human milk as a biological system. Sci Rep 2024; 14:7569. [PMID: 38555284 PMCID: PMC10981717 DOI: 10.1038/s41598-024-58127-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 03/26/2024] [Indexed: 04/02/2024] Open
Abstract
Proteins and peptides found in human milk have bioactive potential to benefit the newborn and support healthy development. Research has been carried out on the health benefits of proteins and peptides, but many questions still need to be answered about the nature of these components, how they are formed, and how they end up in the milk. This study explored and elucidated the complexity of the human milk proteome and peptidome. Proteins and peptides were analyzed with non-targeted nanoLC-Orbitrap-MS/MS in a selection of 297 milk samples from the CHILD Cohort Study. Protein and peptide abundances were determined, and a network was inferred using Gaussian graphical modeling (GGM), allowing an investigation of direct associations. This study showed that signatures of (1) specific mechanisms of transport of different groups of proteins, (2) proteolytic degradation by proteases and aminopeptidases, and (3) coagulation and complement activation are present in human milk. These results show the value of an integrated approach in evaluating large-scale omics data sets and provide valuable information for studies that aim to associate protein or peptide profiles from biofluids such as milk with specific physiological characteristics.
Collapse
Affiliation(s)
- Pieter M Dekker
- Food Quality and Design Group, Wageningen University and Research, Wageningen, 6708 WE, The Netherlands
- Laboratory of Biochemistry, Wageningen University and Research, Wageningen, 6708 WE, The Netherlands
| | - Sjef Boeren
- Laboratory of Biochemistry, Wageningen University and Research, Wageningen, 6708 WE, The Netherlands
| | - Edoardo Saccenti
- Laboratory of Systems and Synthetic Biology, Wageningen University and Research, Wageningen, 6708 WE, The Netherlands
| | - Kasper A Hettinga
- Food Quality and Design Group, Wageningen University and Research, Wageningen, 6708 WE, The Netherlands.
| |
Collapse
|
2
|
Shuai Y, Zhang H, Liu C, Wang J, Jiang Y, Sun J, Gao X, Bo X, Xiao X, Liao X, Huang C, Chen H, Jiang G. CLIC3 interacts with NAT10 to inhibit N4-acetylcytidine modification of p21 mRNA and promote bladder cancer progression. Cell Death Dis 2024; 15:9. [PMID: 38182571 PMCID: PMC10770081 DOI: 10.1038/s41419-023-06373-z] [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: 08/06/2023] [Revised: 11/27/2023] [Accepted: 12/05/2023] [Indexed: 01/07/2024]
Abstract
Chromatin accessibility plays important roles in revealing the regulatory networks of gene expression, while its application in bladder cancer is yet to be fully elucidated. Chloride intracellular channel 3 (CLIC3) protein has been reported to be associated with the progression of some tumors, whereas the specific mechanism of CLIC3 in tumor remains unclear. Here, we screened for key genes in bladder cancer through the identification of transcription factor binding site clustered region (TFCR) on the basis of chromatin accessibility and TF motif. CLIC3 was identified by joint profiling of chromatin accessibility data with TCGA database. Clinically, CLIC3 expression was significantly elevated in bladder cancer and was negatively correlated with patient survival. CLIC3 promoted the proliferation of bladder cancer cells by reducing p21 expression in vitro and in vivo. Mechanistically, CLIC3 interacted with NAT10 and inhibited the function of NAT10, resulting in the downregulation of ac4C modification and stability of p21 mRNA. Overall, these findings uncover an novel mechanism of mRNA ac4C modification and CLIC3 may act as a potential therapeutic target for bladder cancer.
Collapse
Affiliation(s)
- Yujun Shuai
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hui Zhang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Changhao Liu
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, China
| | - Junting Wang
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, China
| | - Yangkai Jiang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jiayin Sun
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xincheng Gao
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiaochen Bo
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, China
| | - Xingyuan Xiao
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Xin Liao
- Department of General Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Chao Huang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Hebing Chen
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, China.
| | - Guosong Jiang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
3
|
Ott E, Hoff S, Indorf L, Ditengou FA, Müller J, Renschler G, Lienkamp SS, Kramer-Zucker A, Bergmann C, Epting D. A novel role for the chloride intracellular channel protein Clic5 in ciliary function. Sci Rep 2023; 13:17647. [PMID: 37848494 PMCID: PMC10582032 DOI: 10.1038/s41598-023-44235-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 10/05/2023] [Indexed: 10/19/2023] Open
Abstract
CLIC5 belongs to a family of ion channels with six members reported so far. In vertebrates, the CLIC5 gene encodes two different isoforms, CLIC5A and CLIC5B. In addition to its ion channel activity, there is evidence for further functions of CLIC5A, such as the remodeling of the actin cytoskeleton during the formation of a functional glomerulus in the vertebrate kidney. However, its specific role is still incompletely understood and a specific functional role for CLIC5B has not been described yet. Here we report our findings on the differential expression and functions of Clic5a and Clic5b during zebrafish kidney development. Whole-mount in situ hybridization studies revealed specific expression of clic5a in the eye and pronephric glomerulus, and clic5b is expressed in the gut, liver and the pronephric tubules. Clic5 immunostainings revealed that Clic5b is localized in the cilia. Whereas knockdown of Clic5a resulted in leakiness of the glomerular filtration barrier, Clic5b deficient embryos displayed defective ciliogenesis, leading to ciliopathy-associated phenotypes such as ventral body curvature, otolith deposition defects, altered left-right asymmetry and formation of hydrocephalus and pronephric cysts. In addition, Clic5 deficiency resulted in dysregulation of cilia-dependent Wnt signalling pathway components. Mechanistically, we identified a Clic5-dependent activation of the membrane-cytoskeletal linker proteins Ezrin/Radixin/Moesin (ERM) in the pronephric tubules of zebrafish. In conclusion, our in vivo data demonstrates a novel role for Clic5 in regulating essential ciliary functions and identified Clic5 as a positive regulator of ERM phosphorylation.
Collapse
Affiliation(s)
- Elisabeth Ott
- Department of Medicine IV, Faculty of Medicine, Medical Center-University of Freiburg, 79106, Freiburg, Germany
| | - Sylvia Hoff
- Department of Medicine IV, Faculty of Medicine, Medical Center-University of Freiburg, 79106, Freiburg, Germany
| | - Lara Indorf
- Department of Medicine IV, Faculty of Medicine, Medical Center-University of Freiburg, 79106, Freiburg, Germany
| | - Franck Anicet Ditengou
- Bio Imaging Core Light Microscopy (BiMiC), Medical Faculty-Institute for Disease Modeling and Targeted Medicine (IMITATE), 79106, Freiburg, Germany
| | - Julius Müller
- Limbach Genetics, Medizinische Genetik Mainz, 55128, Mainz, Germany
| | - Gina Renschler
- Limbach Genetics, Medizinische Genetik Mainz, 55128, Mainz, Germany
| | - Soeren S Lienkamp
- Department of Medicine IV, Faculty of Medicine, Medical Center-University of Freiburg, 79106, Freiburg, Germany
- Center for Biological Signaling Studies (BIOSS), 79104, Freiburg, Germany
| | - Albrecht Kramer-Zucker
- Department of Medicine IV, Faculty of Medicine, Medical Center-University of Freiburg, 79106, Freiburg, Germany
| | - Carsten Bergmann
- Department of Medicine IV, Faculty of Medicine, Medical Center-University of Freiburg, 79106, Freiburg, Germany
- Limbach Genetics, Medizinische Genetik Mainz, 55128, Mainz, Germany
| | - Daniel Epting
- Department of Medicine IV, Faculty of Medicine, Medical Center-University of Freiburg, 79106, Freiburg, Germany.
| |
Collapse
|
4
|
Sanchez VC, Craig‐Lucas A, Cataisson C, Carofino BL, Yuspa SH. Crosstalk between tumor and stroma modifies CLIC4 cargo in extracellular vesicles. JOURNAL OF EXTRACELLULAR BIOLOGY 2023; 2:e118. [PMID: 38264628 PMCID: PMC10803055 DOI: 10.1002/jex2.118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 09/08/2023] [Accepted: 09/11/2023] [Indexed: 01/25/2024]
Abstract
Mouse models of breast cancer have revealed that tumor-bearing hosts must express the oxidoreductase CLIC4 to develop lung metastases. In the absence of host CLIC4, primary tumors grow but the lung premetastatic niche is defective for metastatic seeding. Primary breast cancer cells release EVs that incorporate CLIC4 as cargo and circulate in plasma of wildtype tumor-bearing hosts. CLIC4-deficient breast cancer cells also form tumors in wildtype hosts and release EVs in plasma, but these EVs lack CLIC4, suggesting that the tumor is the source of the plasma-derived EVs that carry CLIC4 as cargo. Paradoxically, circulating EVs are also devoid of CLIC4 when CLIC4-expressing primary tumors are grown in CLIC4 knockout hosts. Thus, the incorporation of CLIC4 (and perhaps other factors) as EV cargo released from tumors involves specific signals from the surrounding stroma determined by its genetic composition. Since CLIC4 is also detected in circulating EVs from human breast cancer patients, future studies will address its association with disease.
Collapse
Affiliation(s)
- Vanesa C. Sanchez
- Center for Drug Evaluation and ResearchU.S. Food and Drug AdministrationSilver SpringMarylandUSA
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Alayna Craig‐Lucas
- Department of SurgeryLehigh Valley Health NetworkAllentownPennsylvaniaUSA
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Christophe Cataisson
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Brandi L. Carofino
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Stuart H. Yuspa
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer InstituteNational Institutes of HealthBethesdaMarylandUSA
| |
Collapse
|
5
|
Kleinjan ML, Mao DY, Naiche LA, Joshi JC, Gupta A, Jesse JJ, Shaye DD, Mehta D, Kitajewski J. CLIC4 Regulates Endothelial Barrier Control by Mediating PAR1 Signaling via RhoA. Arterioscler Thromb Vasc Biol 2023; 43:1441-1454. [PMID: 37317855 PMCID: PMC10527476 DOI: 10.1161/atvbaha.123.319206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 05/30/2023] [Indexed: 06/16/2023]
Abstract
BACKGROUND Endothelial CLICs (chloride intracellular channel proteins) CLIC1 and CLIC4 are required for the GPCRs (G-protein-coupled receptors) S1PR1 (sphingosine-1-phosphate receptor 1) and S1PR3 to activate the small GTPases Rac1 (Ras-related C3 botulinum toxin substrate 1) and RhoA (Ras homolog family member A). To determine whether CLIC1 and CLIC4 function in additional endothelial GPCR pathways, we evaluated CLIC function in thrombin signaling via the thrombin-regulated PAR1 (protease-activated receptor 1) and downstream effector RhoA. METHODS We assessed the ability of CLIC1 and CLIC4 to relocalize to cell membranes in response to thrombin in human umbilical vein endothelial cells (HUVEC). We examined CLIC1 and CLIC4 function in HUVEC by knocking down expression of each CLIC protein and compared thrombin-mediated RhoA or Rac1 activation, ERM (ezrin/radixin/moesin) phosphorylation, and endothelial barrier modulation in control and CLIC knockdown HUVEC. We generated a conditional murine allele of Clic4 and examined PAR1-mediated lung microvascular permeability and retinal angiogenesis in mice with endothelial-specific loss of Clic4. RESULTS Thrombin promoted relocalization of CLIC4, but not CLIC1, to HUVEC membranes. Knockdown of CLIC4 in HUVEC reduced thrombin-mediated RhoA activation, ERM phosphorylation, and endothelial barrier disruption. Knockdown of CLIC1 did not reduce thrombin-mediated RhoA activity but prolonged the RhoA and endothelial barrier response to thrombin. Endothelial-specific deletion of Clic4 in mice reduced lung edema and microvascular permeability induced by PAR1 activating peptide. CONCLUSIONS CLIC4 is a critical effector of endothelial PAR1 signaling and is required to regulate RhoA-mediated endothelial barrier disruption in cultured endothelial cells and murine lung endothelium. CLIC1 was not critical for thrombin-mediated barrier disruption but contributed to the barrier recovery phase after thrombin treatment.
Collapse
Affiliation(s)
- Matthew L. Kleinjan
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, USA
| | - De Yu Mao
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, USA
| | - L. A. Naiche
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, USA
| | - Jagdish C. Joshi
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL, USA
| | - Ahana Gupta
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, USA
| | - Jordan J. Jesse
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, USA
| | - Daniel D. Shaye
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, USA
| | - Dolly Mehta
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL, USA
| | - Jan Kitajewski
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, USA
- University of Illinois Cancer Center, Chicago, IL, USA
| |
Collapse
|
6
|
Chen GL, Li J, Zhang J, Zeng B. To Be or Not to Be an Ion Channel: Cryo-EM Structures Have a Say. Cells 2023; 12:1870. [PMID: 37508534 PMCID: PMC10378246 DOI: 10.3390/cells12141870] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 07/13/2023] [Accepted: 07/16/2023] [Indexed: 07/30/2023] Open
Abstract
Ion channels are the second largest class of drug targets after G protein-coupled receptors. In addition to well-recognized ones like voltage-gated Na/K/Ca channels in the heart and neurons, novel ion channels are continuously discovered in both excitable and non-excitable cells and demonstrated to play important roles in many physiological processes and diseases such as developmental disorders, neurodegenerative diseases, and cancer. However, in the field of ion channel discovery, there are an unignorable number of published studies that are unsolid and misleading. Despite being the gold standard of a functional assay for ion channels, electrophysiological recordings are often accompanied by electrical noise, leak conductance, and background currents of the membrane system. These unwanted signals, if not treated properly, lead to the mischaracterization of proteins with seemingly unusual ion-conducting properties. In the recent ten years, the technical revolution of cryo-electron microscopy (cryo-EM) has greatly advanced our understanding of the structures and gating mechanisms of various ion channels and also raised concerns about the pore-forming ability of some previously identified channel proteins. In this review, we summarize cryo-EM findings on ion channels with molecular identities recognized or disputed in recent ten years and discuss current knowledge of proposed channel proteins awaiting cryo-EM analyses. We also present a classification of ion channels according to their architectures and evolutionary relationships and discuss the possibility and strategy of identifying more ion channels by analyzing structures of transmembrane proteins of unknown function. We propose that cross-validation by electrophysiological and structural analyses should be essentially required for determining molecular identities of novel ion channels.
Collapse
Affiliation(s)
- Gui-Lan Chen
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, China
| | - Jian Li
- College of Pharmaceutical Sciences, Gannan Medical University, Ganzhou 341000, China
| | - Jin Zhang
- School of Basic Medical Sciences, Nanchang University, Nanchang 330031, China
| | - Bo Zeng
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, China
| |
Collapse
|
7
|
Hossain KR, Turkewitz DR, Holt SA, Le Brun AP, Valenzuela SM. Sterol Structural Features' Impact on the Spontaneous Membrane Insertion of CLIC1 into Artificial Lipid Membranes. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023; 39:3286-3300. [PMID: 36821411 DOI: 10.1021/acs.langmuir.2c03129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Background: A membrane protein interaction with lipids shows distinct specificity in terms of the sterol structure. The structure of the sterol's polar headgroup, steroidal rings, and aliphatic side chains have all been shown to influence protein membrane interactions, including the initial binding and subsequent oligomerization to form functional channels. Previous studies have provided some insights into the regulatory role that cholesterol plays in the spontaneous membrane insertion of the chloride intracellular ion channel protein, CLIC1. However, the manner in which cholesterol interacts with CLIC1 is yet largely unknown. Method: In this study, the CLIC1 interaction with different lipid:sterol monolayers was studied using the Langmuir trough and neutron reflectometry in order to investigate the structural features of cholesterol essential for the spontaneous membrane insertion of the CLIC1 protein. Molecular docking simulations were also performed to study the binding affinities between CLIC1 and the different sterol molecules. Results: This study, for the first time, highlights the vital role of the free sterol 3β-OH group as an essential structural requirement for the interaction of CLIC1 with cholesterol. Furthermore, the presence of additional hydroxyl groups, methylation of the sterol skeleton, and the structure of the sterol alkyl side chain have also been shown to modulate the magnitude of CLIC1 interaction with sterols and hence their spontaneous membrane insertion. This study also reports the ability of CLIC1 to interact with other naturally existing sterol molecules. General Significance: Like the sterol molecules, CLIC proteins are evolutionarily conserved with almost all vertebrates expressing six CLIC proteins (CLIC1-6), and CLIC-like proteins are also present in invertebrates and have also been reported in plants. This discovery of CLIC1 protein interaction with other natural sterols and the sterol structural requirements for CLIC membrane insertion provide key information to explore the feasibility of exploiting these properties for therapeutic and prophylactic purposes.
Collapse
Affiliation(s)
- Khondker R Hossain
- School of Life Sciences, University of Technology Sydney, Sydney, New South Wales 2007, Australia
- Australian Centre for Neutron Scattering, Australian Nuclear Science and Technology Organisation (ANSTO), Lucas Heights, New South Wales 2234, Australia
| | - Daniel R Turkewitz
- School of Life Sciences, University of Technology Sydney, Sydney, New South Wales 2007, Australia
| | - Stephen A Holt
- Australian Centre for Neutron Scattering, Australian Nuclear Science and Technology Organisation (ANSTO), Lucas Heights, New South Wales 2234, Australia
| | - Anton P Le Brun
- Australian Centre for Neutron Scattering, Australian Nuclear Science and Technology Organisation (ANSTO), Lucas Heights, New South Wales 2234, Australia
| | - Stella M Valenzuela
- School of Life Sciences, University of Technology Sydney, Sydney, New South Wales 2007, Australia
- Institute for Biomedical Materials and Devices (IBMD), University of Technology Sydney, Sydney, New South Wales 2007, Australia
- ARC Research Hub for Integrated Device for End-User Analysis at Low-Levels (IDEAL Hub), Faculty of Science, University of Technology Sydney, , Sydney, New South Wales 2007, Australia
| |
Collapse
|
8
|
Structure-based discovery and in vitro validation of inhibitors of chloride intracellular channel 4 protein. Comput Struct Biotechnol J 2022; 21:688-701. [PMID: 36659928 PMCID: PMC9826898 DOI: 10.1016/j.csbj.2022.12.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 12/22/2022] [Accepted: 12/23/2022] [Indexed: 12/25/2022] Open
Abstract
The use of computer-aided methods have continued to propel accelerated drug discovery across various disease models, interestingly allowing the specific inhibition of pathogenic targets. Chloride Intracellular Channel Protein 4 (CLIC4) is a novel class of intracellular ion channel highly implicated in tumor and vascular biology. It regulates cell proliferation, apoptosis and angiogenesis; and is involved in multiple pathologic signaling pathways. Absence of specific inhibitors however impedes its advancement to translational research. Here, we integrate structural bioinformatics and experimental research approaches for the discovery and validation of small-molecule inhibitors of CLIC4. High-affinity allosteric binders were identified from a library of 1615 Food and Drug Administration (FDA)-approved drugs via a high-performance computing-powered blind-docking approach, resulting in the selection of amphotericin B and rapamycin. NMR assays confirmed the binding and conformational disruptive effects of both drugs while they also reversed stress-induced membrane translocation of CLIC4 and inhibited endothelial cell migration. Structural and dynamics simulation studies further revealed that the inhibitory mechanisms of these compounds were hinged on the allosteric modulation of the catalytic glutathione (GSH)-like site loop and the extended catalytic β loop which may elicit interference with the catalytic activities of CLIC4. Structure-based insights from this study provide the basis for the selective targeting of CLIC4 to treat the associated pathologies.
Collapse
Key Words
- A9C, 9-Anthracenecarboxylic acid
- AMPhB, Amphotericin B
- Ad, Adenovirus
- Allosteric inhibition
- Bad, BCL2 associated agonist of cell death
- Bcl-2, B-cell lymphoma 2
- Bcl-xL, B-cell lymphoma-extra large
- CDK, Cyclin-dependent kinases
- CLIC, Chloride intracellular channel protein
- Chloride intracellular channel protein 4
- Computational high-throughput screening
- DAPI, 4′,6-diamidino-2-phenylindole
- DIDS, 4,4′-Diisothiocyano-2,2′-stilbenedisulfonic acid
- DMSO, Dimethyl sulfoxide
- DOPE, Discrete optimized protein energy
- GPU, Graphics Processing Unit
- GSH-like catalytic site
- GST, glutathione S-transferases
- GUI, Graphical User Interface
- HEPES, (4-(2-hydroxyethyl)− 1-piperazineethanesulfonic acid;
- HIF, Hypoxia-inducible factor
- HSQC, Heteronuclear single quantum coherence spectroscopy
- HUVEC, Human umbilical vein endothelial cells
- IKKβ, Inhibitor of nuclear kappa-B-kinase subunit beta
- JNK, c-Jun N-terminal kinase
- MKK6, Mitogen-activated protein kinase kinase-6
- MOI, Multiplicity of infection
- NF-κB, Nuclear factor kappa-light-chain-enhancer of activated B cells
- NMR, Nuclear magnetic resonance
- NPT, The constant-temperature, constant-pressure ensemble
- NaCL, Sodium chloride
- Nuclear magnetic resonance
- PAH, Pulmonary arterial hypertension
- RAPA, Rapamycin
- SASA, Solvent accessible surface area
- SEK1, Dual specificity mitogen-activated protein kinase kinase 4
- Smad, Suppressor of Mothers against Decapentaplegic
- Structure-based drug discovery
- TEV, Tobacco etch virus
- TIP3P, Transferable intermolecular potential 3 P
- TROSY, Transverse relaxation optimized spectroscopy
- UCSF, University of California, San Francisco
- VEGF, Vascular endothelial growth factor
- p38, Mitogen activated protein kinases
Collapse
|
9
|
Heckman CA, Ademuyiwa OM, Cayer ML. How filopodia respond to calcium in the absence of a calcium-binding structural protein: non-channel functions of TRP. Cell Commun Signal 2022; 20:130. [PMID: 36028898 PMCID: PMC9414478 DOI: 10.1186/s12964-022-00927-y] [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: 12/25/2021] [Accepted: 06/21/2022] [Indexed: 11/18/2022] Open
Abstract
Background For many cell types, directional locomotion depends on their maintaining filopodia at the leading edge. Filopodia lack any Ca2+-binding structural protein but respond to store-operated Ca2+ entry (SOCE). Methods SOCE was induced by first replacing the medium with Ca2+-free salt solution with cyclopiazonic acid (CPA). This lowers Ca2+ in the ER and causes stromal interacting molecule (STIM) to be translocated to the cell surface. After this priming step, CPA was washed out, and Ca2+ influx restored by addition of extracellular Ca2+. Intracellular Ca2+ levels were measured by calcium orange fluorescence. Regulatory mechanisms were identified by pharmacological treatments. Proteins mediating SOCE were localized by immunofluorescence and analyzed after image processing. Results Depletion of the ER Ca2+ increased filopodia prevalence briefly, followed by a spontaneous decline that was blocked by inhibitors of endocytosis. Intracellular Ca2+ increased continuously for ~ 50 min. STIM and a transient receptor potential canonical (TRPC) protein were found in separate compartments, but an aquaporin unrelated to SOCE was present in both. STIM1- and TRPC1-bearing vesicles were trafficked on microtubules. During depletion, STIM1 migrated to the surface where it coincided with Orai in punctae, as expected. TRPC1 was partially colocalized with Vamp2, a rapidly releasable pool marker, and with phospholipases (PLCs). TRPC1 retreated to internal compartments during ER depletion. Replenishment of extracellular Ca2+ altered the STIM1 distribution, which came to resemble that of untreated cells. Vamp2 and TRPC1 underwent exocytosis and became homogeneously distributed on the cell surface. This was accompanied by an increased prevalence of filopodia, which was blocked by inhibitors of TRPC1/4/5 and endocytosis. Conclusions Because the media were devoid of ligands that activate receptors during depletion and Ca2+ replenishment, we could attribute filopodia extension to SOCE. We propose that the Orai current stimulates exocytosis of TRPC-bearing vesicles, and that Ca2+ influx through TRPC inhibits PLC activity. This allows regeneration of the substrate, phosphatidylinositol 4,5 bisphosphate (PIP2), a platform for assembling proteins, e. g. Enabled and IRSp53. TRPC contact with PLC is required but is broken by TRPC dissemination. This explains how STIM1 regulates the cell’s ability to orient itself in response to attractive or repulsive cues. Video Abstract
Supplementary Information The online version contains supplementary material available at 10.1186/s12964-022-00927-y.
Collapse
Affiliation(s)
- C A Heckman
- Department of Biological Sciences, 217 Life Science Building, Bowling Green State University, Bowling Green, OH, 43403-0001, USA.
| | - O M Ademuyiwa
- Department of Biological Sciences, 217 Life Science Building, Bowling Green State University, Bowling Green, OH, 43403-0001, USA
| | - M L Cayer
- Center for Microscopy and Microanalysis, Bowling Green State University, Bowling Green, OH, 43403, USA
| |
Collapse
|
10
|
Sanchez VC, Yang HH, Craig-Lucas A, Dubois W, Carofino BL, Lack J, Dwyer JE, Simpson RM, Cataisson C, Lee MP, Luo J, Hunter KW, Yuspa SH. Host CLIC4 expression in the tumor microenvironment is essential for breast cancer metastatic competence. PLoS Genet 2022; 18:e1010271. [PMID: 35727842 PMCID: PMC9249210 DOI: 10.1371/journal.pgen.1010271] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 07/01/2022] [Accepted: 05/22/2022] [Indexed: 11/18/2022] Open
Abstract
The TGF-β-regulated Chloride Intracellular Channel 4 (CLIC4) is an essential participant in the formation of breast cancer stroma. Here, we used data available from the TCGA and METABRIC datasets to show that CLIC4 expression was higher in breast cancers from younger women and those with early-stage metastatic disease. Elevated CLIC4 predicted poor outcome in breast cancer patients and was linked to the TGF-β pathway. However, these associations did not reveal the underlying biological contribution of CLIC4 to breast cancer progression. Constitutive ablation of host Clic4 in two murine metastatic breast cancer models nearly eliminated lung metastases without reducing primary tumor weight, while tumor cells ablated of Clic4 retained metastatic capability in wildtype hosts. Thus, CLIC4 was required for host metastatic competence. Pre- and post-metastatic proteomic analysis identified circulating pro-metastatic soluble factors that differed in tumor-bearing CLIC4-deficient and wildtype hosts. Vascular abnormalities and necrosis increased in primary tumors from CLIC4-deficient hosts. Transcriptional profiles of both primary tumors and pre-metastatic lungs of tumor-bearing CLIC4-deficient hosts were consistent with a microenvironment where inflammatory pathways were elevated. Altogether, CLIC4 expression in human breast cancers may serve as a prognostic biomarker; therapeutic targeting of CLIC4 could reduce primary tumor viability and host metastatic competence.
Collapse
Affiliation(s)
- Vanesa C. Sanchez
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Howard H. Yang
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Alayna Craig-Lucas
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Wendy Dubois
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Brandi L. Carofino
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Justin Lack
- NIAID Collaborative Bioinformatics Resource (NCBR), National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, Maryland, United States of America
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, Maryland, United States of America
| | - Jennifer E. Dwyer
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - R. Mark Simpson
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Christophe Cataisson
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Max P. Lee
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Ji Luo
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Kent W. Hunter
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Stuart H. Yuspa
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Center, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
11
|
Reibring CG, El Shahawy M, Hallberg K, Harfe BD, Linde A, Gritli-Linde A. Loss of BMP2 and BMP4 Signaling in the Dental Epithelium Causes Defective Enamel Maturation and Aberrant Development of Ameloblasts. Int J Mol Sci 2022; 23:6095. [PMID: 35682776 PMCID: PMC9180982 DOI: 10.3390/ijms23116095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/18/2022] [Accepted: 05/25/2022] [Indexed: 12/10/2022] Open
Abstract
BMP signaling is crucial for differentiation of secretory ameloblasts, the cells that secrete enamel matrix. However, whether BMP signaling is required for differentiation of maturation-stage ameloblasts (MA), which are instrumental for enamel maturation into hard tissue, is hitherto unknown. To address this, we used an in vivo genetic approach which revealed that combined deactivation of the Bmp2 and Bmp4 genes in the murine dental epithelium causes development of dysmorphic and dysfunctional MA. These fail to exhibit a ruffled apical plasma membrane and to reabsorb enamel matrix proteins, leading to enamel defects mimicking hypomaturation amelogenesis imperfecta. Furthermore, subsets of mutant MA underwent pathological single or collective cell migration away from the ameloblast layer, forming cysts and/or exuberant tumor-like and gland-like structures. Massive apoptosis in the adjacent stratum intermedium and the abnormal cell-cell contacts and cell-matrix adhesion of MA may contribute to this aberrant behavior. The mutant MA also exhibited severely diminished tissue non-specific alkaline phosphatase activity, revealing that this enzyme's activity in MA crucially depends on BMP2 and BMP4 inputs. Our findings show that combined BMP2 and BMP4 signaling is crucial for survival of the stratum intermedium and for proper development and function of MA to ensure normal enamel maturation.
Collapse
Affiliation(s)
- Claes-Göran Reibring
- Department of Oral Biochemistry, Institute of Odontology, Sahlgrenska Academy at the University of Gothenburg, SE-40530 Göteborg, Sweden; (C.-G.R.); (M.E.S.); (K.H.); (A.L.)
| | - Maha El Shahawy
- Department of Oral Biochemistry, Institute of Odontology, Sahlgrenska Academy at the University of Gothenburg, SE-40530 Göteborg, Sweden; (C.-G.R.); (M.E.S.); (K.H.); (A.L.)
- Department of Oral Biology, Faculty of Dentistry, Minia University, Minia 61511, Egypt
| | - Kristina Hallberg
- Department of Oral Biochemistry, Institute of Odontology, Sahlgrenska Academy at the University of Gothenburg, SE-40530 Göteborg, Sweden; (C.-G.R.); (M.E.S.); (K.H.); (A.L.)
| | - Brian D. Harfe
- Department of Molecular Genetics and Microbiology Genetics Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA;
| | - Anders Linde
- Department of Oral Biochemistry, Institute of Odontology, Sahlgrenska Academy at the University of Gothenburg, SE-40530 Göteborg, Sweden; (C.-G.R.); (M.E.S.); (K.H.); (A.L.)
| | - Amel Gritli-Linde
- Department of Oral Biochemistry, Institute of Odontology, Sahlgrenska Academy at the University of Gothenburg, SE-40530 Göteborg, Sweden; (C.-G.R.); (M.E.S.); (K.H.); (A.L.)
| |
Collapse
|
12
|
Turkewitz DR, Moghaddasi S, Alghalayini A, D'Amario C, Ali HM, Wallach M, Valenzuela SM. Comparative study of His- and Non-His-tagged CLIC proteins, reveals changes in their enzymatic activity. Biochem Biophys Rep 2021; 26:101015. [PMID: 34036185 PMCID: PMC8138732 DOI: 10.1016/j.bbrep.2021.101015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 04/15/2021] [Accepted: 05/02/2021] [Indexed: 10/29/2022] Open
Abstract
The chloride intracellular ion channel protein (CLIC) family are a unique set of ion channels that can exist as soluble and integral membrane proteins. New evidence has emerged that demonstrates CLICs' possess oxidoreductase enzymatic activity and may function as either membrane-spanning ion channels or as globular enzymes. To further characterize the enzymatic profile of members of the CLIC family and to expand our understanding of their functions, we expressed and purified recombinant CLIC1, CLIC3, and a non-functional CLIC1-Cys24A mutant using a Histidine tag, bacterial protein expression system. We demonstrate that the presence of the six-polyhistidine tag at the amino terminus of the proteins led to a decrease in their oxidoreductase enzymatic activity compared to their non-His-tagged counterparts, when assessed using 2-hydroxyethyl disulfide as a substrate. These results strongly suggest the six-polyhistidine tag alters CLIC's structure at the N-terminus, which also contains the enzyme active site. It also raises the need for caution in use of His-tagged proteins when assessing oxidoreductase protein enzymatic function.
Collapse
Affiliation(s)
- Daniel R. Turkewitz
- School of Life Sciences, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Saba Moghaddasi
- School of Life Sciences, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Amani Alghalayini
- School of Life Sciences, University of Technology Sydney, Sydney, NSW, 2007, Australia
- ARC Research Hub for Integrated Device for End-user Analysis at Low-levels (IDEAL), Faculty of Science, University of Technology Sydney, NSW, 2007, Australia
| | - Claudia D'Amario
- School of Life Sciences, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Hala M. Ali
- School of Life Sciences, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Michael Wallach
- School of Life Sciences, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Stella M. Valenzuela
- School of Life Sciences, University of Technology Sydney, Sydney, NSW, 2007, Australia
- ARC Research Hub for Integrated Device for End-user Analysis at Low-levels (IDEAL), Faculty of Science, University of Technology Sydney, NSW, 2007, Australia
| |
Collapse
|
13
|
Wang J, Luo J, Sun Z, Sun F, Kong Z, Yu J. Identification of MTHFD2 as a novel prognosis biomarker in esophageal carcinoma patients based on transcriptomic data and methylation profiling. Medicine (Baltimore) 2020; 99:e22194. [PMID: 32925794 PMCID: PMC7489726 DOI: 10.1097/md.0000000000022194] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
DNA methylation is an important epigenetic regulatory mechanism in esophageal carcinoma (EC) and is associated with genomic instability and carcinogenesis. In the present study, we aimed to identify tumor biomarkers for predicting prognosis of EC patients.We downloaded mRNA expression profiles and DNA methylation profiles associated with EC from the Gene Expression Omnibus database. Differentially expressed and differentially methylated genes between tumor tissues and adjacent normal tissue samples were identified. Functional enrichment analyses were performed, followed by the construction of protein-protein interaction networks. Data were validated based on methylation profiles from The Cancer Genome Atlas. Candidate genes were further verified according to survival analysis and Cox regression analysis.We uncovered multiple genes with differential expression or methylation in tumor samples compared with normal samples. After taking the intersection of 3 differential gene sets, we obtained a total of 232 overlapping genes. Functional enrichment analysis revealed that these genes are related to pathways such as "glutathione metabolism," "p53 signaling pathway," and "focal adhesion." Furthermore, 8 hub genes with inversed expression and methylation correlation were identified as candidate genes. The abnormal expression levels of MSN, PELI1, and MTHFD2 were correlated with overall survival times in EC patients (P < .05). Only MTHFD2 was significantly associated with a pathologic stage according to univariate analysis (P = .037) and multivariate analysis (P = .043).Our study identified several novel EC biomarkers with prognostic value by integrated analysis of transcriptomic data and methylation profiles. MTHFD2 could serve as an independent biomarker for predicting prognosis and pathological stages of EC.
Collapse
Affiliation(s)
- Jianlin Wang
- Department of Radiotherapy, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University
- Center for Medical Physics, Nanjing Medical University, Changzhou, Jiangsu Province, China
| | - Judong Luo
- Department of Radiotherapy, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University
| | - Zhiqiang Sun
- Department of Radiotherapy, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University
| | - Fei Sun
- Department of Radiotherapy, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University
| | - Ze Kong
- Department of Radiotherapy, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University
| | - Jingping Yu
- Department of Radiotherapy, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University
- Center for Medical Physics, Nanjing Medical University, Changzhou, Jiangsu Province, China
| |
Collapse
|
14
|
Buechner M, Yang Z, Al-Hashimi H. A Series of Tubes: The C. elegans Excretory Canal Cell as a Model for Tubule Development. J Dev Biol 2020; 8:jdb8030017. [PMID: 32906663 PMCID: PMC7557474 DOI: 10.3390/jdb8030017] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 08/31/2020] [Accepted: 09/02/2020] [Indexed: 12/25/2022] Open
Abstract
Formation and regulation of properly sized epithelial tubes is essential for multicellular life. The excretory canal cell of C. elegans provides a powerful model for investigating the integration of the cytoskeleton, intracellular transport, and organismal physiology to regulate the developmental processes of tube extension, lumen formation, and lumen diameter regulation in a narrow single cell. Multiple studies have provided new understanding of actin and intermediate filament cytoskeletal elements, vesicle transport, and the role of vacuolar ATPase in determining tube size. Most of the genes discovered have clear homologues in humans, with implications for understanding these processes in mammalian tissues such as Schwann cells, renal tubules, and brain vasculature. The results of several new genetic screens are described that provide a host of new targets for future studies in this informative structure.
Collapse
Affiliation(s)
- Matthew Buechner
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS 66045, USA;
- Correspondence:
| | - Zhe Yang
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS 66045, USA;
| | | |
Collapse
|
15
|
Llorente García I, Marsh M. A biophysical perspective on receptor-mediated virus entry with a focus on HIV. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2020; 1862:183158. [PMID: 31863725 PMCID: PMC7156917 DOI: 10.1016/j.bbamem.2019.183158] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 12/12/2019] [Accepted: 12/13/2019] [Indexed: 12/14/2022]
Abstract
As part of their entry and infection strategy, viruses interact with specific receptor molecules expressed on the surface of target cells. The efficiency and kinetics of the virus-receptor interactions required for a virus to productively infect a cell is determined by the biophysical properties of the receptors, which are in turn influenced by the receptors' plasma membrane (PM) environments. Currently, little is known about the biophysical properties of these receptor molecules or their engagement during virus binding and entry. Here we review virus-receptor interactions focusing on the human immunodeficiency virus type 1 (HIV), the etiological agent of acquired immunodeficiency syndrome (AIDS), as a model system. HIV is one of the best characterised enveloped viruses, with the identity, roles and structure of the key molecules required for infection well established. We review current knowledge of receptor-mediated HIV entry, addressing the properties of the HIV cell-surface receptors, the techniques used to measure these properties, and the macromolecular interactions and events required for virus entry. We discuss some of the key biophysical principles underlying receptor-mediated virus entry and attempt to interpret the available data in the context of biophysical mechanisms. We also highlight crucial outstanding questions and consider how new tools might be applied to advance understanding of the biophysical properties of viral receptors and the dynamic events leading to virus entry.
Collapse
Affiliation(s)
| | - Mark Marsh
- Medical Research Council Laboratory for Molecular Cell Biology, University College London, London, UK
| |
Collapse
|
16
|
Argenzio E, Innocenti M. The chloride intracellular channel protein CLIC4 inhibits filopodium formation induced by constitutively active mutants of formin mDia2. FEBS Lett 2020; 594:1750-1758. [PMID: 32145706 DOI: 10.1002/1873-3468.13766] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 02/14/2020] [Accepted: 03/02/2020] [Indexed: 11/07/2022]
Abstract
Chloride intracellular channel 4 (CLIC4) functions in diverse actin-dependent processes. Upon Rho activation, CLIC4 reversibly translocates from the cytosol to the plasma membrane to regulate cell adhesion and migration. At the plasma membrane, CLIC4 counters the formation of filopodia, which requires actin assembly by the formin mammalian Diaphanous (mDia)2. To this end, mDia2 must be activated through conversion from the closed to the open conformation. Thus, CLIC4 could harness the activation or the open conformation of mDia2 to inhibit filopodium formation. Here, we find that CLIC4 silencing enhances the filopodia induced by two constitutively active mDia2 mutants. Furthermore, we report that CLIC4 binds the actin-regulatory region of mDia2 in vitro. These results suggest that CLIC4 modulates the activity of the open conformation of mDia2, shedding new light into how cells may control filopodia.
Collapse
Affiliation(s)
- Elisabetta Argenzio
- Division of Cell Biology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Metello Innocenti
- Heidelberg University Biochemistry Center (BZH), Heidelberg University, Germany
| |
Collapse
|
17
|
Abstract
In children with normal hearing, inflammatory disorders caused by infections of the middle ear (otitis media) are the most common ear illnesses. Many of older adults experience some level of hearing loss. Several factors can lead to either a partial loss or the total inability to hear (deafness) including exposure to noise, a hereditary predisposition, chronic infections, traumas, medications, and aging.
Collapse
|
18
|
Uretmen Kagiali ZC, Saner N, Akdag M, Sanal E, Degirmenci BS, Mollaoglu G, Ozlu N. CLIC4 and CLIC1 bridge plasma membrane and cortical actin network for a successful cytokinesis. Life Sci Alliance 2019; 3:3/2/e201900558. [PMID: 31879279 PMCID: PMC6933522 DOI: 10.26508/lsa.201900558] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 12/20/2019] [Accepted: 12/20/2019] [Indexed: 11/24/2022] Open
Abstract
CLIC members are required for the progression of cytokinesis by coupling the plasma membrane and cortical actin network at the cleavage furrow and polar cortex. CLIC4 and CLIC1 are members of the well-conserved chloride intracellular channel proteins (CLICs) structurally related to glutathione-S-transferases. Here, we report new roles of CLICs in cytokinesis. At the onset of cytokinesis, CLIC4 accumulates at the cleavage furrow and later localizes to the midbody in a RhoA-dependent manner. The cell cycle–dependent localization of CLIC4 is abolished when its glutathione S-transferase activity–related residues (C35A and F37D) are mutated. Ezrin, anillin, and ALIX are identified as interaction partners of CLIC4 at the cleavage furrow and midbody. Strikingly, CLIC4 facilitates the activation of ezrin at the cleavage furrow and reciprocally inhibition of ezrin activation diminishes the translocation of CLIC4 to the cleavage furrow. Furthermore, knockouts of CLIC4and CLIC1 cause abnormal blebbing at the polar cortex and regression of the cleavage furrow at late cytokinesis leading to multinucleated cells. We conclude that CLIC4 and CLIC1 function together with ezrin where they bridge plasma membrane and actin cytoskeleton at the polar cortex and cleavage furrow to promote cortical stability and successful completion of cytokinesis in mammalian cells.
Collapse
Affiliation(s)
| | - Nazan Saner
- Department of Molecular Biology and Genetics, Koç University, Istanbul, Turkey
| | - Mehmet Akdag
- Department of Molecular Biology and Genetics, Koç University, Istanbul, Turkey
| | - Erdem Sanal
- Department of Molecular Biology and Genetics, Koç University, Istanbul, Turkey
| | | | - Gurkan Mollaoglu
- Department of Molecular Biology and Genetics, Koç University, Istanbul, Turkey
| | - Nurhan Ozlu
- Department of Molecular Biology and Genetics, Koç University, Istanbul, Turkey .,Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey
| |
Collapse
|
19
|
Ueno Y, Ozaki S, Umakoshi A, Yano H, Choudhury ME, Abe N, Sumida Y, Kuwabara J, Uchida R, Islam A, Ogawa K, Ishimaru K, Yorozuya T, Kunieda T, Watanabe Y, Takada Y, Tanaka J. Chloride intracellular channel protein 2 in cancer and non-cancer human tissues: relationship with tight junctions. Tissue Barriers 2019; 7:1593775. [PMID: 30929599 PMCID: PMC6592591 DOI: 10.1080/21688370.2019.1593775] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Chloride intracellular channel protein 2 (CLIC2) belongs to the CLIC family of conserved metazoan proteins. Although CLICs have been identified as chloride channels, they are currently considered multifunctional proteins. CLIC2 is the least studied family member. We investigated CLIC2 expression and localization in human hepatocellular carcinoma, metastatic colorectal cancer in the liver, and colorectal cancer. Significant expression of mRNAs encoding CLIC1, 2, 4, and 5 were found in the human tissues, but only CLIC2 was predominantly expressed in non-cancer tissues surrounding cancer masses. Fibrotic or dysfunctional (aspartate aminotransferase ≥40) non-cancer liver tissues and advanced stage HCC tissues expressed low levels of CLIC2. Endothelial cells lining blood vessels but not lymphatic vessels in non-cancer tissues expressed CLIC2 as well as high levels of the tight junction proteins claudins 1 and 5, occludin, and ZO-1. Most endothelial cells in blood vessels in cancer tissues had very low expressions of CLIC2 and tight junction proteins. CD31+/CD45− endothelial cells isolated from non-cancer tissues expressed mRNAs encoding CLIC2, claudin 1, occludin and ZO-1, while similar cell fractions from cancer tissues had very low expressions of these molecules. Knockdown of CLIC2 expression in human umbilical vein endothelial cells (HUVECs) allowed human cancer cells to transmigrate through a HUVEC monolayer. These results suggest that CLIC2 may be involved in the formation and/or maintenance of tight junctions and that cancer tissue vasculature lacks CLIC2 and tight junctions, which allows the intravasation of cancer cells necessary for hematogenous metastasis.
Collapse
Affiliation(s)
- Yoshitomo Ueno
- a Department of Hepato-biliary Pancreatic Surgery and Breast Surgery, Graduate School of Medicine , Ehime University , Toon , Ehime , Japan
| | - Saya Ozaki
- b Department of Neurosurgery, Graduate School of Medicine , Ehime University , Toon , Ehime , Japan
| | - Akihiro Umakoshi
- c Department of Molecular and Cellular Physiology, Graduate School of Medicine , Ehime University , Toon , Ehime , Japan
| | - Hajime Yano
- c Department of Molecular and Cellular Physiology, Graduate School of Medicine , Ehime University , Toon , Ehime , Japan
| | - Mohammed E Choudhury
- c Department of Molecular and Cellular Physiology, Graduate School of Medicine , Ehime University , Toon , Ehime , Japan
| | - Naoki Abe
- d Department of Anesthesia and Perioperative Medicine, Graduate School of Medicine , Ehime University , Toon , Ehime , Japan
| | - Yutaro Sumida
- c Department of Molecular and Cellular Physiology, Graduate School of Medicine , Ehime University , Toon , Ehime , Japan
| | - Jun Kuwabara
- e Department of Gastrointestinal Surgery and Surgical Oncology, Graduate School of Medicine , Ehime University , Toon , Ehime , Japan
| | - Rina Uchida
- c Department of Molecular and Cellular Physiology, Graduate School of Medicine , Ehime University , Toon , Ehime , Japan
| | - Afsana Islam
- c Department of Molecular and Cellular Physiology, Graduate School of Medicine , Ehime University , Toon , Ehime , Japan
| | - Kohei Ogawa
- a Department of Hepato-biliary Pancreatic Surgery and Breast Surgery, Graduate School of Medicine , Ehime University , Toon , Ehime , Japan
| | - Kei Ishimaru
- e Department of Gastrointestinal Surgery and Surgical Oncology, Graduate School of Medicine , Ehime University , Toon , Ehime , Japan
| | - Toshihiro Yorozuya
- d Department of Anesthesia and Perioperative Medicine, Graduate School of Medicine , Ehime University , Toon , Ehime , Japan
| | - Takeharu Kunieda
- b Department of Neurosurgery, Graduate School of Medicine , Ehime University , Toon , Ehime , Japan
| | - Yuji Watanabe
- e Department of Gastrointestinal Surgery and Surgical Oncology, Graduate School of Medicine , Ehime University , Toon , Ehime , Japan
| | - Yasutsugu Takada
- a Department of Hepato-biliary Pancreatic Surgery and Breast Surgery, Graduate School of Medicine , Ehime University , Toon , Ehime , Japan
| | - Junya Tanaka
- c Department of Molecular and Cellular Physiology, Graduate School of Medicine , Ehime University , Toon , Ehime , Japan
| |
Collapse
|
20
|
Sun S, Adyshev D, Dudek S, Paul A, McColloch A, Cho M. Cholesterol-dependent Modulation of Stem Cell Biomechanics: Application to Adipogenesis. J Biomech Eng 2019; 141:2729412. [PMID: 30901381 DOI: 10.1115/1.4043253] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Indexed: 11/08/2022]
Abstract
Cell mechanics has been shown to regulate stem cell differentiation. We have previously reported that altered cell stiffness of mesenchymal stem cells can delay or facilitate biochemically directed differentiation. One of the factors that can affect the cell stiffness is cholesterol. However, the effect of cholesterol on differentiation of human mesenchymal stem cells (hMSCs) remains elusive. In this paper, we demonstrate that cholesterol is involved in the modulation of the cell stiffness and subsequent adipogenic differentiation. Rapid cytoskeletal actin reorganization was evident and correlated with the cell's Young's modulus measured using atomic force microscopy (AFM). In addition, the level of membrane-bound cholesterol was found to increase during adipogenic differentiation and inversely varied with the cell stiffness. Furthermore, cholesterol played a key role in the regulation of the cell morphology and biomechanics, suggesting its crucial involvement in mechanotransduction. To better understand the underlying mechanisms, we investigated the effect of cholesterol on the membrane-cytoskeleton linker proteins (ezrin and moesin). Cholesterol depletion was found to up-regulate the ezrin expression which promoted cell spreading, increased Young's modulus, and hindered adipogenesis. In contrast, cholesterol enrichment increased the moesin expression, decreased Young's modulus, and induced cell rounding and facilitated adipogenesis. Taken together, cholesterol appears to regulate the stem cell mechanics and adipogenesis through the membrane-associated linker proteins.
Collapse
Affiliation(s)
- Shan Sun
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL 60607
| | - Djanybek Adyshev
- Department of Medicine, University of Illinois at Chicago, Chicago, IL 60607
| | - Steve Dudek
- Department of Medicine, University of Illinois at Chicago, Chicago, IL 60607
| | - Amit Paul
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL 60607
| | - Andrew McColloch
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76019
| | - Michael Cho
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76019
| |
Collapse
|
21
|
SREBP-1a-stimulated lipid synthesis is required for macrophage phagocytosis downstream of TLR4-directed mTORC1. Proc Natl Acad Sci U S A 2018; 115:E12228-E12234. [PMID: 30530672 PMCID: PMC6310840 DOI: 10.1073/pnas.1813458115] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
There is a growing appreciation for a fundamental connection between lipid metabolism and the immune response. Macrophage phagocytosis is a signature innate immune response to pathogen exposure, and cytoplasmic membrane expansion is required to engulf the phagocytic target. The sterol regulatory element binding proteins (SREBPs) are key transcriptional regulatory proteins that sense the intracellular lipid environment and modulate expression of key genes of fatty acid and cholesterol metabolism to maintain lipid homeostasis. In this study, we show that TLR4-dependent stimulation of macrophage phagocytosis requires mTORC1-directed SREBP-1a-dependent lipid synthesis. We also show that the phagocytic defect in macrophages from SREBP-1a-deficient mice results from decreased interaction between membrane lipid rafts and the actin cytoskeleton, presumably due to reduced accumulation of newly synthesized fatty acyl chains within major membrane phospholipids. We show that mTORC1-deficient macrophages also have a phagocytic block downstream from TLR4 signaling, and, interestingly, the reduced level of phagocytosis in both SREBP-1a- and mTORC1-deficient macrophages can be restored by ectopic SREBP-1a expression. Taken together, these observations indicate SREBP-1a is a major downstream effector of TLR4-mTORC1 directed interactions between membrane lipid rafts and the actin cytoskeleton that are required for pathogen-stimulated phagocytosis in macrophages.
Collapse
|
22
|
Heredero-Bermejo I, Varberg JM, Charvat R, Jacobs K, Garbuz T, Sullivan WJ, Arrizabalaga G. TgDrpC, an atypical dynamin-related protein in Toxoplasma gondii, is associated with vesicular transport factors and parasite division. Mol Microbiol 2018; 111:46-64. [PMID: 30362624 DOI: 10.1111/mmi.14138] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/21/2018] [Indexed: 01/01/2023]
Abstract
Dynamin-related proteins (Drps) are involved in diverse processes such as organelle division and vesicle trafficking. The intracellular parasite Toxoplasma gondii possesses three distinct Drps. TgDrpC, whose function remains unresolved, is unusual in that it lacks a conserved GTPase Effector Domain, which is typically required for function. Here, we show that TgDrpC localizes to cytoplasmic puncta; however, in dividing parasites, TgDrpC redistributes to the growing edge of the daughter cells. By conditional knockdown, we determined that loss of TgDrpC stalls division and leads to rapid deterioration of multiple organelles and the IMC. We also show that TgDrpC interacts with proteins that exhibit homology to those involved in vesicle transport, including members of the adaptor complex 2. Two of these proteins, a homolog of the adaptor protein 2 (AP-2) complex subunit alpha-1 and a homolog of the ezrin-radixin-moesin (ERM) family proteins, localize to puncta and associate with the daughter cells. Consistent with the association with vesicle transport proteins, re-distribution of TgDrpC to the IMC during division is dependent on post-Golgi trafficking. Together, these results support that TgDrpC contributes to vesicle trafficking and is critical for stability of parasite organelles and division.
Collapse
Affiliation(s)
- Irene Heredero-Bermejo
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Joseph M Varberg
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Robert Charvat
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Kylie Jacobs
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Tamila Garbuz
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - William J Sullivan
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.,Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Gustavo Arrizabalaga
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.,Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| |
Collapse
|
23
|
Argenzio E, Klarenbeek J, Kedziora KM, Nahidiazar L, Isogai T, Perrakis A, Jalink K, Moolenaar WH, Innocenti M. Profilin binding couples chloride intracellular channel protein CLIC4 to RhoA-mDia2 signaling and filopodium formation. J Biol Chem 2018; 293:19161-19176. [PMID: 30381396 PMCID: PMC6302171 DOI: 10.1074/jbc.ra118.002779] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 10/26/2018] [Indexed: 12/31/2022] Open
Abstract
Chloride intracellular channel 4 (CLIC4) is a cytosolic protein implicated in diverse actin-based processes, including integrin trafficking, cell adhesion, and tubulogenesis. CLIC4 is rapidly recruited to the plasma membrane by RhoA-activating agonists and then partly colocalizes with β1 integrins. Agonist-induced CLIC4 translocation depends on actin polymerization and requires conserved residues that make up a putative binding groove. However, the mechanism and significance of CLIC4 trafficking have been elusive. Here, we show that RhoA activation by either lysophosphatidic acid (LPA) or epidermal growth factor is necessary and sufficient for CLIC4 translocation to the plasma membrane and involves regulation by the RhoA effector mDia2, a driver of actin polymerization and filopodium formation. We found that CLIC4 binds the G-actin–binding protein profilin-1 via the same residues that are required for CLIC4 trafficking. Consistently, shRNA-induced profilin-1 silencing impaired agonist-induced CLIC4 trafficking and the formation of mDia2-dependent filopodia. Conversely, CLIC4 knockdown increased filopodium formation in an integrin-dependent manner, a phenotype rescued by wild-type CLIC4 but not by the trafficking-incompetent mutant CLIC4(C35A). Furthermore, CLIC4 accelerated LPA-induced filopodium retraction. We conclude that through profilin-1 binding, CLIC4 functions in a RhoA–mDia2–regulated signaling network to integrate cortical actin assembly and membrane protrusion. We propose that agonist-induced CLIC4 translocation provides a feedback mechanism that counteracts formin-driven filopodium formation.
Collapse
Affiliation(s)
| | | | | | | | | | - Anastassis Perrakis
- Biochemistry, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | | | | | | |
Collapse
|
24
|
Liu YR, Ortiz-Bonilla CJ, Lee YF. Extracellular Vesicles in Bladder Cancer: Biomarkers and Beyond. Int J Mol Sci 2018; 19:E2822. [PMID: 30231589 PMCID: PMC6165150 DOI: 10.3390/ijms19092822] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 09/12/2018] [Accepted: 09/15/2018] [Indexed: 12/14/2022] Open
Abstract
Tumor-derived extracellular vesicles (TEVs) are membrane-bound, nanosized vesicles released by cancer cells and taken up by cells in the tumor microenvironment to modulate the molecular makeup and behavior of recipient cells. In this report, we summarize the pivotal roles of TEVs involved in bladder cancer (BC) development, progression and treatment resistance through transferring their bioactive cargos, including proteins and nucleic acids. We also report on the molecular profiling of TEV cargos derived from urine and blood of BC patients as non-invasive disease biomarkers. The current hurdles in EV research and plausible solutions are discussed.
Collapse
Affiliation(s)
- Yu-Ru Liu
- Department of Urology, University of Rochester Medical Center, Rochester, NY 14642, USA.
| | - Carlos J Ortiz-Bonilla
- Department of Urology, University of Rochester Medical Center, Rochester, NY 14642, USA.
- Department of Pathology and Lab Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA.
| | - Yi-Fen Lee
- Department of Urology, University of Rochester Medical Center, Rochester, NY 14642, USA.
- Department of Pathology and Lab Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA.
| |
Collapse
|
25
|
Thuringer D, Chanteloup G, Winckler P, Garrido C. The vesicular transfer of CLIC1 from glioblastoma to microvascular endothelial cells requires TRPM7. Oncotarget 2018; 9:33302-33311. [PMID: 30279961 PMCID: PMC6161795 DOI: 10.18632/oncotarget.26048] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 08/16/2018] [Indexed: 01/01/2023] Open
Abstract
Chloride intracellular channel 1 (CLIC1) is highly expressed and secreted by human glioblastoma cells and cell lines such as U87, initiating cell migration and tumor growth. Here, we examined whether CLIC1 could be transferred to human primary microvascular endothelial cells (HMEC). We previously reported that the oncogenic microRNA, miR-5096, increased the release of extracellular vesicles (EVs) by which it increased its own transfer from U87 to surrounding cells. Thus, we also examined its effect on the CLIC1 transfer. In homotypic cultures, miR-5096 did not increase the expression of CLIC1 in U87 nor in HMEC. However, the endothelial CLIC1 level increased after exposure to EVs released by U87, and even more by miR-5096-loaded U87. The EVs-transferred CLIC1 was active in HMEC, promoting endothelial sprouting in matrigel. Cell exposure to EVs induced cytosolic Ca2+ spikes which were dependent on the transient receptor potential melastatin member 7 (TRPM7). TRPM7 silencing prevented Ca2+ spikes and the subsequent CLIC1 delivery into HMEC. Our data suggest that the vesicular transfer of CLIC1 between cells requires TRMP7 expression in recipient endothelial cells. How the vesicular transfer of CLIC1 is modulated in cancer therapy is a future challenge.
Collapse
Affiliation(s)
- Dominique Thuringer
- INSERM U1231, Laboratory of Excellence Ligue Nationale contre le Cancer, 21000 Dijon, France.,Université de Bourgogne Franche Comté, 21000 Dijon, France
| | - Gaetan Chanteloup
- INSERM U1231, Laboratory of Excellence Ligue Nationale contre le Cancer, 21000 Dijon, France.,Université de Bourgogne Franche Comté, 21000 Dijon, France
| | - Pascale Winckler
- AgroSup Dijon, PAM UMR, DImaCell Imaging Facility, Université de Bourgogne Franche Comté, 21000 Dijon, France
| | - Carmen Garrido
- INSERM U1231, Laboratory of Excellence Ligue Nationale contre le Cancer, 21000 Dijon, France.,Université de Bourgogne Franche Comté, 21000 Dijon, France.,Centre Georges François Leclerc (CGFL), 21000 Dijon, France
| |
Collapse
|
26
|
Lee J, McKinney KQ, Pavlopoulos AJ, Niu M, Kang JW, Oh JW, Kim KP, Hwang S. Altered Proteome of Extracellular Vesicles Derived from Bladder Cancer Patients Urine. Mol Cells 2018; 41:179-187. [PMID: 29562735 PMCID: PMC5881091 DOI: 10.14348/molcells.2018.2110] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 12/06/2017] [Accepted: 12/20/2017] [Indexed: 12/29/2022] Open
Abstract
Proteomic analysis of extracellular vesicles (EVs) from biological fluid is a powerful approach to discover potential biomarkers for human diseases including cancers, as EV secreted to biological fluids are originated from the affected tissue. In order to investigate significant molecules related to the pathogenesis of bladder cancer, EVs were isolated from patient urine which was analyzed by mass spectrometry based proteomics. Comparison of the EV proteome to the whole urine proteome demonstrated an increased number of protein identification in EV. Comparative MS analyses of urinary EV from control subjects and bladder cancer patients identified a total of 1,222 proteins. Statistical analyses provided 56 proteins significantly increased in bladder cancer urine, including proteins for which expression levels varied by cancer stage (P-value < 0.05). While urine represents a valuable, noninvasive specimen for biomarker discovery in urologic cancers, there is a high degree of intra- and inter-individual variability in urine samples. The enrichment of urinary EV demonstrated its capability and applicability of providing a focused identification of biologically relevant proteins in urological diseases.
Collapse
Affiliation(s)
- Jingyun Lee
- Proteomics Laboratory for Clinical and Translational Research, Carolinas HealthCare System, Charlotte, NC 28203,
United States
| | - Kimberly Q. McKinney
- Proteomics Laboratory for Clinical and Translational Research, Carolinas HealthCare System, Charlotte, NC 28203,
United States
| | - Antonis J. Pavlopoulos
- Proteomics Laboratory for Clinical and Translational Research, Carolinas HealthCare System, Charlotte, NC 28203,
United States
| | - Meng Niu
- Proteomics Laboratory for Clinical and Translational Research, Carolinas HealthCare System, Charlotte, NC 28203,
United States
| | - Jung Won Kang
- Omics Core Laboratory, Research Institute, National Cancer Center, Goyang 10408,
Korea
| | - Jae Won Oh
- Department of Applied Chemistry, The Institute of Natural Science, Global Center for Pharmaceutical Ingredient Materials, Kyung Hee University, Yongin 17104,
Korea
| | - Kwang Pyo Kim
- Department of Applied Chemistry, The Institute of Natural Science, Global Center for Pharmaceutical Ingredient Materials, Kyung Hee University, Yongin 17104,
Korea
| | - Sunil Hwang
- Proteomics Laboratory for Clinical and Translational Research, Carolinas HealthCare System, Charlotte, NC 28203,
United States
| |
Collapse
|
27
|
Stakaitytė G, Nwogu N, Lippiat JD, Blair GE, Poterlowicz K, Boyne JR, Macdonald A, Mankouri J, Whitehouse A. The cellular chloride channels CLIC1 and CLIC4 contribute to virus-mediated cell motility. J Biol Chem 2018; 293:4582-4590. [PMID: 29462791 PMCID: PMC5868249 DOI: 10.1074/jbc.ra117.001343] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Indexed: 12/11/2022] Open
Abstract
Ion channels regulate many aspects of cell physiology, including cell proliferation, motility, and migration, and aberrant expression and activity of ion channels is associated with various stages of tumor development, with K+ and Cl− channels now being considered the most active during tumorigenesis. Accordingly, emerging in vitro and preclinical studies have revealed that pharmacological manipulation of ion channel activity offers protection against several cancers. Merkel cell polyomavirus (MCPyV) is a major cause of Merkel cell carcinoma (MCC), primarily because of the expression of two early regulatory proteins termed small and large tumor antigens (ST and LT, respectively). Several molecular mechanisms have been attributed to MCPyV-mediated cancer formation but, thus far, no studies have investigated any potential link to cellular ion channels. Here we demonstrate that Cl− channel modulation can reduce MCPyV ST-induced cell motility and invasiveness. Proteomic analysis revealed that MCPyV ST up-regulates two Cl− channels, CLIC1 and CLIC4, which when silenced, inhibit MCPyV ST-induced motility and invasiveness, implicating their function as critical to MCPyV-induced metastatic processes. Consistent with these data, we confirmed that CLIC1 and CLIC4 are up-regulated in primary MCPyV-positive MCC patient samples. We therefore, for the first time, implicate cellular ion channels as a key host cell factor contributing to virus-mediated cellular transformation. Given the intense interest in ion channel modulating drugs for human disease. This highlights CLIC1 and CLIC4 activity as potential targets for MCPyV-induced MCC.
Collapse
Affiliation(s)
- Gabrielė Stakaitytė
- From the School of Molecular and Cellular Biology.,Astbury Centre for Structural Molecular Biology
| | - Nnenna Nwogu
- From the School of Molecular and Cellular Biology.,Astbury Centre for Structural Molecular Biology
| | - Jonathan D Lippiat
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, United Kingdom and
| | - G Eric Blair
- From the School of Molecular and Cellular Biology
| | - Krzysztof Poterlowicz
- Centre for Skin Sciences, School of Chemistry and Biosciences, Faculty of Life Sciences, University of Bradford, Bradford BD7 1DP, United Kingdom
| | - James R Boyne
- Centre for Skin Sciences, School of Chemistry and Biosciences, Faculty of Life Sciences, University of Bradford, Bradford BD7 1DP, United Kingdom
| | - Andrew Macdonald
- From the School of Molecular and Cellular Biology.,Astbury Centre for Structural Molecular Biology
| | - Jamel Mankouri
- From the School of Molecular and Cellular Biology, .,Astbury Centre for Structural Molecular Biology
| | - Adrian Whitehouse
- From the School of Molecular and Cellular Biology, .,Astbury Centre for Structural Molecular Biology
| |
Collapse
|
28
|
Barros FBA, Assao A, Garcia NG, Nonogaki S, Carvalho AL, Soares FA, Kowalski LP, Oliveira DT. Moesin expression by tumor cells is an unfavorable prognostic biomarker for oral cancer. BMC Cancer 2018; 18:53. [PMID: 29310601 PMCID: PMC5759236 DOI: 10.1186/s12885-017-3914-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 12/14/2017] [Indexed: 12/30/2022] Open
Abstract
Background Moesin is a member of the ERM (ezrin, radixin and moesin) proteins that participate in cell migration and tumor invasion through transductional signals sent to actin filaments by glycoproteins, such as podoplanin. Methods This study aimed to evaluate the participation of moesin and podoplanin in the invasive tumor front of oral squamous cell carcinomas, and their influence on patients’ prognosis. Podoplanin and moesin immunoexpressions were evaluated by a semi-quantitative score method, based on the capture of 10 microscopic fields, at 400X magnification, in the invasive tumor front of oral squamous cell carcinomas. The association of moesin and podoplanin expression with clinicopathological variables was analyzed by the chi-square, or Fisher’s exact test. The 5 and 10 years survival rates were calculated by the Kaplan-Meier method and the survival curves were compared by using the log-rank test. Results The immunohistochemical expression of moesin in the invasive front of oral squamous cell carcinomas was predominantly strong, homogenously distributed on the membrane and in the cytoplasm of tumor cells. The expression of moesin was not associated with clinical, demographic and microscopic features of the patients. Otherwise, podoplanin expression by malignant epithelial cells was predominantly strong and significantly associated with radiotherapy (p = 0.004), muscular invasion (p = 0.006) and lymph node involvement (p = 0.013). Strong moesin expression was considered an unfavorable prognostic factor for patients with oral squamous cell carcinomas, clinical stage II and III (p = 0.024). Conclusions These results suggested that strong moesin expression by malignant cells may help to determine patients with oral squamous cell carcinoma and poor prognosis.
Collapse
Affiliation(s)
- Francisco Bárbara Abreu Barros
- Department of Stomatology, Area of Pathology, Bauru School of Dentistry, University of São Paulo, Alameda Octávio Pinheiro Brisolla, 9-75, Bauru, São Paulo, 17012-901, Brazil
| | - Agnes Assao
- Department of Stomatology, Area of Pathology, Bauru School of Dentistry, University of São Paulo, Alameda Octávio Pinheiro Brisolla, 9-75, Bauru, São Paulo, 17012-901, Brazil
| | - Natália Galvão Garcia
- Department of Stomatology, Area of Pathology, Bauru School of Dentistry, University of São Paulo, Alameda Octávio Pinheiro Brisolla, 9-75, Bauru, São Paulo, 17012-901, Brazil
| | - Suely Nonogaki
- Adolfo Lutz Institute, Pathology Division, São Paulo, Brazil
| | - André Lopes Carvalho
- Fundação Pio XII Institution - Cancer Hospital of Barretos, Barretos, São Paulo, Brazil
| | | | - Luiz Paulo Kowalski
- Department of Head and Neck Surgery and Otorhinolaringology, A.C.Camargo Cancer Center Hospital, São Paulo, Brazil
| | - Denise Tostes Oliveira
- Department of Stomatology, Area of Pathology, Bauru School of Dentistry, University of São Paulo, Alameda Octávio Pinheiro Brisolla, 9-75, Bauru, São Paulo, 17012-901, Brazil.
| |
Collapse
|
29
|
Wang G, Bi L, Wang G, Huang F, Lu M, Zhu K. Microarray analysis to identify the similarities and differences of pathogenesis between aortic occlusive disease and abdominal aortic aneurysm. Vascular 2017; 26:301-314. [PMID: 29087237 DOI: 10.1177/1708538117736695] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Objectives Expression profile of GSE57691 was analyzed to identify the similarities and differences between aortic occlusive disease and abdominal aortic aneurysm. Methods The expression profile of GSE57691 was downloaded from Gene Expression Omnibus database, including 20 small abdominal aortic aneurysm samples, 29 large abdominal aortic aneurysm samples, 9 aortic occlusive disease samples, and 10 control samples. Using the limma package in R, the differentially expressed genes were screened. Followed by enrichment analysis was performed for the differentially expressed genes using database for annotation, visualization, and integrated discovery online tool. Based on string online tool and Cytoscape software, protein-protein interaction network and module analyses were carried out. Moreover, integrated TF platform database and Cytoscape software were used for constructing transcriptional regulatory networks. Results As a result, 1757, 354, and 396 differentially expressed genes separately were identified in aortic occlusive disease, large abdominal aortic aneurysm, and small abdominal aortic aneurysm samples. UBB was significantly enriched in proteolysis related pathways with a high degree in three groups. SPARCL1 was another gene shared by these groups and regulated by NFIA, which had a high degree in transcriptional regulatory network. ACTB, a significant upregulated gene in abdominal aortic aneurysm samples, could be regulated by CLIC4, which was significantly enriched in cell motions. ACLY and NFIB were separately identified in aortic occlusive disease and small abdominal aortic aneurysm samples, and separately enriched in lipid metabolism and negative regulation of cell proliferation. Conclusions The downregulated UBB, NFIA, and SPARCL1 might play key roles in both aortic occlusive disease and abdominal aortic aneurysm, while the upregulated ACTB might only involve in abdominal aortic aneurysm. ACLY and NFIB were specifically involved in aortic occlusive disease and small abdominal aortic aneurysm separately.
Collapse
Affiliation(s)
- Guofu Wang
- Department of Vascular Surgery, Shaoxing Hospital of China Medical University, Shaoxing, China
| | - Lechang Bi
- Department of Vascular Surgery, Shaoxing Hospital of China Medical University, Shaoxing, China
| | - Gaofeng Wang
- Department of Vascular Surgery, Shaoxing Hospital of China Medical University, Shaoxing, China
| | - Feilai Huang
- Department of Vascular Surgery, Shaoxing Hospital of China Medical University, Shaoxing, China
| | - Mingjing Lu
- Department of Vascular Surgery, Shaoxing Hospital of China Medical University, Shaoxing, China
| | - Kai Zhu
- Department of Vascular Surgery, Shaoxing Hospital of China Medical University, Shaoxing, China
| |
Collapse
|
30
|
Norman J, Zanivan S. Chloride intracellular channel 3: A secreted pro-invasive oxidoreductase. Cell Cycle 2017; 16:1993-1994. [PMID: 28933594 PMCID: PMC5731412 DOI: 10.1080/15384101.2017.1377031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 09/15/2017] [Accepted: 08/30/2017] [Indexed: 10/29/2022] Open
Affiliation(s)
- Jim Norman
- Cancer Research UK Beatson Institute, Glasgow, UK
| | - Sara Zanivan
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
31
|
Vogl C, Butola T, Haag N, Hausrat TJ, Leitner MG, Moutschen M, Lefèbvre PP, Speckmann C, Garrett L, Becker L, Fuchs H, Hrabe de Angelis M, Nietzsche S, Kessels MM, Oliver D, Kneussel M, Kilimann MW, Strenzke N. The BEACH protein LRBA is required for hair bundle maintenance in cochlear hair cells and for hearing. EMBO Rep 2017; 18:2015-2029. [PMID: 28893864 DOI: 10.15252/embr.201643689] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 07/27/2017] [Accepted: 08/07/2017] [Indexed: 12/15/2022] Open
Abstract
Lipopolysaccharide-responsive beige-like anchor protein (LRBA) belongs to the enigmatic class of BEACH domain-containing proteins, which have been attributed various cellular functions, typically involving intracellular protein and membrane transport processes. Here, we show that LRBA deficiency in mice leads to progressive sensorineural hearing loss. In LRBA knockout mice, inner and outer hair cell stereociliary bundles initially develop normally, but then partially degenerate during the second postnatal week. LRBA deficiency is associated with a reduced abundance of radixin and Nherf2, two adaptor proteins, which are important for the mechanical stability of the basal taper region of stereocilia. Our data suggest that due to the loss of structural integrity of the central parts of the hair bundle, the hair cell receptor potential is reduced, resulting in a loss of cochlear sensitivity and functional loss of the fraction of spiral ganglion neurons with low spontaneous firing rates. Clinical data obtained from two human patients with protein-truncating nonsense or frameshift mutations suggest that LRBA deficiency may likewise cause syndromic sensorineural hearing impairment in humans, albeit less severe than in our mouse model.
Collapse
Affiliation(s)
- Christian Vogl
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
| | - Tanvi Butola
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany.,Synaptic Nanophysiology Group, Max-Planck-Institute for Biophysical Chemistry Göttingen, Göttingen, Germany
| | - Natja Haag
- Institute for Biochemistry I, University Hospital Jena, Jena, Germany
| | - Torben J Hausrat
- Department for Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Michael G Leitner
- Department of Physiology, Philipps University Marburg, Marburg, Germany
| | - Michel Moutschen
- Department of Immunology and Infectious Diseases, University of Liège CHU Liège, Liège, Belgium
| | - Philippe P Lefèbvre
- Department of Otorhinolaryngology, University of Liège CHU Liège, Liège, Belgium
| | - Carsten Speckmann
- Division of Pediatric Hematology and Oncology, Center for Chronic Immunodeficiency and Department of Pediatrics and Adolescent Medicine, Medical Centre, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Lillian Garrett
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München German Research Center for Environmental Health, Neuherberg, Germany.,Institute of Developmental Genetics, Helmholtz Zentrum München German Research Center for Environmental Health, Neuherberg, Germany
| | - Lore Becker
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München German Research Center for Environmental Health, Neuherberg, Germany
| | - Helmut Fuchs
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München German Research Center for Environmental Health, Neuherberg, Germany
| | - Martin Hrabe de Angelis
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München German Research Center for Environmental Health, Neuherberg, Germany.,Chair of Experimental Genetics, School of Life Science Weihenstephan, Technische Universität München, München, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany
| | | | - Michael M Kessels
- Institute for Biochemistry I, University Hospital Jena, Jena, Germany
| | - Dominik Oliver
- Department of Physiology, Philipps University Marburg, Marburg, Germany
| | - Matthias Kneussel
- Department for Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Manfred W Kilimann
- Institute for Auditory Neuroscience, University Medical Center Göttingen, Göttingen, Germany.,Department of Molecular Neurobiology, Max Planck Institute for Experimental Medicine, Göttingen, Germany
| | - Nicola Strenzke
- Auditory Systems Physiology Group Department of Otolaryngology University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
32
|
Jiang YY, Hou HT, Yang Q, Liu XC, He GW. Chloride Channels are Involved in the Development of Atrial Fibrillation - A Transcriptomic and proteomic Study. Sci Rep 2017; 7:10215. [PMID: 28860555 PMCID: PMC5579191 DOI: 10.1038/s41598-017-10590-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 08/11/2017] [Indexed: 11/23/2022] Open
Abstract
Electrical and structural remodeling processes are contributors to the self-perpetuating nature of atrial fibrillation (AF). However, their correlation has not been clarified. In this study, human atrial tissues from the patients with rheumatic mitral valve disease in either sinus rhythm or persistent AF were analyzed using a combined transcriptomic and proteomic approach. An up-regulation in chloride intracellular channel (CLIC) 1, 4, 5 and a rise in type IV collagen were revealed. Combined with the results from immunohistochemistry and electron microscope analysis, the distribution of type IV collagen and effects of fibrosis on myocyte membrane indicated the possible interaction between CLIC and type IV collagen, confirmed by protein structure prediction and co-immunoprecipitation. These results indicate that CLICs play an important role in the development of atrial fibrillation and that CLICs and structural type IV collagen may interact on each other to promote the development of AF in rheumatic mitral valve disease.
Collapse
Affiliation(s)
- Yi-Yao Jiang
- Department of Cardiovascular Surgery & Center for Basic Medical Research, TEDA International Cardiovascular Hospital, The Chinese Academy of Medical Sciences & Peking Union Medical College, & Nankai University, Tianjin, China.,The Affiliated Hospital of Hangzhou Normal University & Zhejiang University, Hangzhou, China
| | - Hai-Tao Hou
- Department of Cardiovascular Surgery & Center for Basic Medical Research, TEDA International Cardiovascular Hospital, The Chinese Academy of Medical Sciences & Peking Union Medical College, & Nankai University, Tianjin, China
| | - Qin Yang
- Department of Cardiovascular Surgery & Center for Basic Medical Research, TEDA International Cardiovascular Hospital, The Chinese Academy of Medical Sciences & Peking Union Medical College, & Nankai University, Tianjin, China
| | - Xiao-Cheng Liu
- Department of Cardiovascular Surgery & Center for Basic Medical Research, TEDA International Cardiovascular Hospital, The Chinese Academy of Medical Sciences & Peking Union Medical College, & Nankai University, Tianjin, China
| | - Guo-Wei He
- Department of Cardiovascular Surgery & Center for Basic Medical Research, TEDA International Cardiovascular Hospital, The Chinese Academy of Medical Sciences & Peking Union Medical College, & Nankai University, Tianjin, China. .,The Affiliated Hospital of Hangzhou Normal University & Zhejiang University, Hangzhou, China. .,Department of Surgery, Oregon Health and Science University, Portland, Oregon, USA.
| |
Collapse
|
33
|
Argenzio E, Moolenaar WH. Emerging biological roles of Cl- intracellular channel proteins. J Cell Sci 2017; 129:4165-4174. [PMID: 27852828 DOI: 10.1242/jcs.189795] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Cl- intracellular channels (CLICs) are a family of six evolutionary conserved cytosolic proteins that exist in both soluble and membrane-associated forms; however, their functions have long been elusive. Soluble CLICs adopt a glutathione S-transferase (GST)-fold, can induce ion currents in artificial membranes and show oxidoreductase activity in vitro, but there is no convincing evidence of CLICs having such activities in vivo. Recent studies have revealed a role for CLIC proteins in Rho-regulated cortical actin dynamics as well as vesicular trafficking and integrin recycling, the latter of which are under the control of Rab GTPases. In this Commentary, we discuss the emerging roles of CLIC proteins in these processes and the lessons learned from gene-targeting studies. We also highlight outstanding questions regarding the molecular function(s) of these important but still poorly understood proteins.
Collapse
Affiliation(s)
- Elisabetta Argenzio
- Division of Cell Biology, The Netherlands Cancer Institute, Amsterdam 1066CX, The Netherlands
| | - Wouter H Moolenaar
- Division of Cell Biology, The Netherlands Cancer Institute, Amsterdam 1066CX, The Netherlands
| |
Collapse
|
34
|
Brum AM, van der Leije CS, Schreuders-Koedam M, Verhoeven J, Janssen M, Dekkers DH, Demmers JA, Eijken M, van de Peppel J, van Leeuwen JP, van der Eerden BC. Identification of Chloride Intracellular Channel Protein 3 as a Novel Gene Affecting Human Bone Formation. JBMR Plus 2017; 1:16-26. [PMID: 30283877 PMCID: PMC6124162 DOI: 10.1002/jbm4.10003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 03/09/2017] [Indexed: 12/14/2022] Open
Abstract
Osteoporosis is a common skeletal disorder characterized by low bone mass leading to increased bone fragility and fracture susceptibility. The bone building cells, osteoblasts, are derived from mesenchymal stromal cells (MSCs); however, with increasing age osteogenic differentiation is diminished and more adipocytes are seen in the bone marrow, suggesting a shift in MSC lineage commitment. Identification of specific factors that stimulate osteoblast differentiation from human MSCs may deliver therapeutic targets to treat osteoporosis. The aim of this study was to identify novel genes involved in osteoblast differentiation of human bone marrow–derived MSCs (hMSCs). We identified the gene chloride intracellular channel protein 3 (CLIC3) to be strongly upregulated during MSC‐derived osteoblast differentiation. Lentiviral overexpression of CLIC3 in hMSCs caused a 60% increase of matrix mineralization. Conversely, knockdown of CLIC3 in hMSCs using two short‐hairpin RNAs (shRNAs) against CLIC3 resulted in a 69% to 76% reduction in CLIC3 mRNA expression, 53% to 37% less alkaline phosphatase (ALP) activity, and 78% to 88% less matrix mineralization compared to scrambled control. Next, we used an in vivo human bone formation model in which hMSCs lentivirally transduced with the CLIC3 overexpression construct were loaded onto a scaffold (hydroxyapatite‐tricalcium‐phosphate), implanted under the skin of NOD‐SCID mice, and analyzed for bone formation 8 weeks later. CLIC3 overexpression led to a 15‐fold increase in bone formation (0.33% versus 5.05% bone area relative to scaffold). Using a Clic3‐His‐tagged pull‐down assay and liquid chromatography–mass spectrometry (LS/MS)‐based proteomics analysis in lysates of osteogenically differentiated hMSCs, we showed that CLIC3 interacts with NIMA‐related kinase 9 (NEK9) and phosphatidylserine synthase 1 (PTDSS1) in vitro, and this finding was supported by immunofluorescent analysis. In addition, inhibition of NEK9 or PTDSS1 gene expression by shRNAs inhibited osteoblast differentiation and mineralization. In conclusion, we successfully identified CLIC3 to be a lineage‐specific gene regulating osteoblast differentiation and bone formation through its interaction with NEK9 and PTDSS1. © The Authors. JBMR Plus is published by Wiley Periodicals, Inc. on behalf of the American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Andrea M Brum
- Department of Internal Medicine School of Molecular Medicine Erasmus University Medical Center Rotterdam the Netherlands
| | - Cindy S van der Leije
- Department of Internal Medicine School of Molecular Medicine Erasmus University Medical Center Rotterdam the Netherlands
| | - Marijke Schreuders-Koedam
- Department of Internal Medicine School of Molecular Medicine Erasmus University Medical Center Rotterdam the Netherlands
| | - Jeroen Verhoeven
- Department of Internal Medicine School of Molecular Medicine Erasmus University Medical Center Rotterdam the Netherlands
| | | | - Dick Hw Dekkers
- Proteomics Center Erasmus University Medical Center Rotterdam The Netherlands
| | - Jeroen Aa Demmers
- Proteomics Center Erasmus University Medical Center Rotterdam The Netherlands
| | | | - Jeroen van de Peppel
- Department of Internal Medicine School of Molecular Medicine Erasmus University Medical Center Rotterdam the Netherlands
| | - Johannes Ptm van Leeuwen
- Department of Internal Medicine School of Molecular Medicine Erasmus University Medical Center Rotterdam the Netherlands
| | - Bram Cj van der Eerden
- Department of Internal Medicine School of Molecular Medicine Erasmus University Medical Center Rotterdam the Netherlands
| |
Collapse
|
35
|
Ulmasov B, Bruno J, Oshima K, Cheng YW, Holly SP, Parise LV, Egan TM, Edwards JC. CLIC1 null mice demonstrate a role for CLIC1 in macrophage superoxide production and tissue injury. Physiol Rep 2017; 5:e13169. [PMID: 28275112 PMCID: PMC5350177 DOI: 10.14814/phy2.13169] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 01/16/2017] [Accepted: 01/17/2017] [Indexed: 12/23/2022] Open
Abstract
We generated and studied CLIC1 null (C1KO) mice to investigate the physiological role of this protein. C1KO and matched wild-type (WT) mice were studied in two models of acute toxic tissue injury. CLIC1 expression is upregulated following acute injury of WT kidney and pancreas and is absent in C1KOs. Acute tissue injury is attenuated in the C1KOs and this correlates with an absence of the rise in tissue reactive oxygen species (ROS) that is seen in WT mice. Infiltration of injured tissue by inflammatory cells was comparable between WT and C1KOs. Absence of CLIC1 increased PMA-induced superoxide production by isolated peritoneal neutrophils but dramatically decreased PMA-induced superoxide production by peritoneal macrophages. CLIC1 is expressed in both neutrophils and macrophages in a peripheral pattern consistent with either plasma membrane or the cortical cytoskeleton in resting cells and redistributes away from the periphery following PMA stimulation in both cell types. Absence of CLIC1 had no effect on redistribution or dephosphorylation of Ezrin/ERM cytoskeleton in macrophages. Plasma membrane chloride conductance is altered in the absence of CLIC1, but not in a way that would be expected to block superoxide production. NADPH oxidase redistributes from an intracellular compartment to the plasma membrane when WT macrophages are stimulated to produce superoxide and this redistribution fails to occur in C1KO macrophages. We conclude that the role of CLIC1 in macrophage superoxide production is to support redistribution of NADPH oxidase to the plasma membrane, and not through major effects on ERM cytoskeleton or by acting as a plasma membrane chloride channel.
Collapse
Affiliation(s)
- Barbara Ulmasov
- Department of Internal Medicine, Saint Louis University, St. Louis, Missouri
| | - Jonathan Bruno
- Department of Internal Medicine, Saint Louis University, St. Louis, Missouri
- UNC Kidney Center, University of North Carolina, Chapel Hill, North Carolina
| | - Kiyoko Oshima
- Department of Pathology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Yao-Wen Cheng
- UNC Kidney Center, University of North Carolina, Chapel Hill, North Carolina
| | - Stephen P Holly
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, North Carolina
| | - Leslie V Parise
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, North Carolina
| | - Terrance M Egan
- Department of Pharmacology and Physiology, Saint Louis University, St. Louis, Missouri
| | - John C Edwards
- Department of Internal Medicine, Saint Louis University, St. Louis, Missouri
- UNC Kidney Center, University of North Carolina, Chapel Hill, North Carolina
- Department of Pharmacology and Physiology, Saint Louis University, St. Louis, Missouri
| |
Collapse
|
36
|
The Caenorhabditis elegans Excretory System: A Model for Tubulogenesis, Cell Fate Specification, and Plasticity. Genetics 2017; 203:35-63. [PMID: 27183565 DOI: 10.1534/genetics.116.189357] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 03/07/2016] [Indexed: 12/12/2022] Open
Abstract
The excretory system of the nematode Caenorhabditis elegans is a superb model of tubular organogenesis involving a minimum of cells. The system consists of just three unicellular tubes (canal, duct, and pore), a secretory gland, and two associated neurons. Just as in more complex organs, cells of the excretory system must first adopt specific identities and then coordinate diverse processes to form tubes of appropriate topology, shape, connectivity, and physiological function. The unicellular topology of excretory tubes, their varied and sometimes complex shapes, and the dynamic reprogramming of cell identity and remodeling of tube connectivity that occur during larval development are particularly fascinating features of this organ. The physiological roles of the excretory system in osmoregulation and other aspects of the animal's life cycle are only beginning to be explored. The cellular mechanisms and molecular pathways used to build and shape excretory tubes appear similar to those used in both unicellular and multicellular tubes in more complex organs, such as the vertebrate vascular system and kidney, making this simple organ system a useful model for understanding disease processes.
Collapse
|
37
|
Tavasoli M, Al-Momany A, Wang X, Li L, Edwards JC, Ballermann BJ. Both CLIC4 and CLIC5A activate ERM proteins in glomerular endothelium. Am J Physiol Renal Physiol 2016; 311:F945-F957. [PMID: 27582103 DOI: 10.1152/ajprenal.00353.2016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 08/25/2016] [Indexed: 01/17/2023] Open
Abstract
The chloride intracellular channel (CLIC) 5A is expressed at very high levels in renal glomeruli, in both endothelial cells (EC) and podocytes. CLIC5A stimulates Rac1- and phosphatidylinositol (4,5)-bisphosphate-dependent ERM (ezrin, radixin, moesin) activation. ERM proteins, in turn, function in lumen formation and in the development of actin-based cellular projections. In mice lacking CLIC5A, ERM phosphorylation is profoundly reduced in podocytes, but preserved in glomerular EC. Since glomerular EC also express CLIC4, we reasoned that, if CLIC4 activates ERM proteins like CLIC5A, then CLIC4 could compensate for the CLIC5A loss in glomerular EC. In glomeruli of CLIC5-deficient mice, CLIC4 expression was upregulated and colocalized with moesin and ezrin in glomerular EC, but not in podocytes. In cultured glomerular EC, CLIC4 silencing reduced ERM phosphorylation and cytoskeletal association, and expression of exogenous CLIC4 or CLIC5A rescued ERM de-phosphorylation due to CLIC4 silencing. In mice lacking either CLIC4 or CLIC5, ERM phosphorylation was retained in glomerular EC, but, in mice lacking both CLIC4 and CLIC5, glomerular EC ERM phosphorylation was profoundly reduced. Although glomerular EC fenestrae developed normally in dual CLIC4/CLIC5-deficient mice, the density of fenestrae declined substantially by 8 mo of age, along with the deposition of subendothelial electron-lucent material. The dual CLIC4/CLIC5-deficient mice developed spontaneous proteinuria, glomerular cell proliferation, and matrix deposition. Thus CLIC4 stimulates ERM activation and can compensate for CLIC5A in glomerular EC. The findings indicate that CLIC4/CLIC5A-mediated ERM activation is required for maintenance of the glomerular capillary architecture.
Collapse
Affiliation(s)
- Mahtab Tavasoli
- Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Abass Al-Momany
- Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada; and
| | - Xin Wang
- Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Laiji Li
- Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - John C Edwards
- Department of Internal Medicine, St. Louis University, St. Louis, Missouri
| | - Barbara J Ballermann
- Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada; .,Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada; and
| |
Collapse
|
38
|
Hare JE, Goodchild SC, Breit SN, Curmi PMG, Brown LJ. Interaction of Human Chloride Intracellular Channel Protein 1 (CLIC1) with Lipid Bilayers: A Fluorescence Study. Biochemistry 2016; 55:3825-33. [PMID: 27299171 DOI: 10.1021/acs.biochem.6b00080] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Chloride intracellular channel protein 1 (CLIC1) is very unusual as it adopts a soluble glutathione S-transferase-like canonical fold but can also autoinsert into lipid bilayers to form an ion channel. The conversion between these forms involves a large, but reversible, structural rearrangement of the CLIC1 module. The only identified environmental triggers controlling the metamorphic transition of CLIC1 are pH and oxidation. Until now, there have been no high-resolution structural data available for the CLIC1 integral membrane state, and consequently, a limited understanding of how CLIC1 unfolds and refolds across the bilayer to form a membrane protein with ion channel activity exists. Here we show that fluorescence spectroscopy can be used to establish the interaction and position of CLIC1 in a lipid bilayer. Our method employs a fluorescence energy transfer (FRET) approach between CLIC1 and a dansyl-labeled lipid analogue to probe the CLIC1-lipid interface. Under oxidizing conditions, a strong FRET signal between the single tryptophan residue of CLIC1 (Trp35) and the dansyl-lipid analogue was detected. When considering the proportion of CLIC1 interacting with the lipid bilayer, as estimated by fluorescence quenching experiments, the FRET distance between Trp35 and the dansyl moiety on the membrane surface was determined to be ∼15 Å. This FRET-detected interaction provides direct structural evidence that CLIC1 associates with membranes. The results presented support the current model of an oxidation-driven interaction of CLIC1 with lipid bilayers and also propose a membrane anchoring role for Trp35.
Collapse
Affiliation(s)
- Joanna E Hare
- Department of Chemistry and Biomolecular Sciences, Macquarie University , Sydney, New South Wales 2109, Australia
| | - Sophia C Goodchild
- Astbury Centre for Structural Molecular Biology and School of Molecular and Cellular Biology, University of Leeds , Leeds LS29JT, United Kingdom
| | - Samuel N Breit
- St Vincent's Centre for Applied Medical Research, St Vincent's Hospital , Sydney, New South Wales 2010, Australia
| | - Paul M G Curmi
- School of Physics, University of New South Wales , Sydney, New South Wales 2052, Australia
| | - Louise J Brown
- Department of Chemistry and Biomolecular Sciences, Macquarie University , Sydney, New South Wales 2109, Australia
| |
Collapse
|
39
|
Salao K, Jiang L, Li H, Tsai VWW, Husaini Y, Curmi PMG, Brown LJ, Brown DA, Breit SN. CLIC1 regulates dendritic cell antigen processing and presentation by modulating phagosome acidification and proteolysis. Biol Open 2016; 5:620-30. [PMID: 27113959 PMCID: PMC4874360 DOI: 10.1242/bio.018119] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Intracellular chloride channel protein 1 (CLIC1) participates in inflammatory processes by regulating macrophage phagosomal functions such as pH and proteolysis. Here, we sought to determine if CLIC1 can regulate adaptive immunity by actions on dendritic cells (DCs), the key professional antigen presenting cells. To do this, we first generated bone marrow-derived DCs (BMDCs) from germline CLIC1 gene-deleted (CLIC1−/−) and wild-type (CLIC1+/+) mice, then studied them in vitro and in vivo. We found phagocytosis triggered cytoplasmic CLIC1 translocation to the phagosomal membrane where it regulated phagosomal pH and proteolysis. Phagosomes from CLIC1−/− BMDCs displayed impaired acidification and proteolysis, which could be reproduced if CLIC1+/+, but not CLIC1−/− cells, were treated with IAA94, a CLIC family ion channel blocker. CLIC1−/− BMDC displayed reduced in vitro antigen processing and presentation of full-length myelin oligodendrocyte glycoprotein (MOG) and reduced MOG-induced experimental autoimmune encephalomyelitis. These data suggest that CLIC1 regulates DC phagosomal pH to ensure optimal processing of antigen for presentation to antigen-specific T-cells. Further, they indicate that CLIC1 is a novel therapeutic target to help reduce the adaptive immune response in autoimmune diseases. Summary: DC phagosomes from CLIC1−/− mice display impaired acidification and in vivo and in vitro antigen processing and presentation, revealing CLIC1−/− as a potential therapeutic target in reducing the adaptive immune response in autoimmune diseases.
Collapse
Affiliation(s)
- Kanin Salao
- St Vincent's Centre for Applied Medical Research, St. Vincent's Hospital and University of New South Wales, Sydney, New South Wales 2010, Australia
| | - Lele Jiang
- St Vincent's Centre for Applied Medical Research, St. Vincent's Hospital and University of New South Wales, Sydney, New South Wales 2010, Australia
| | - Hui Li
- St Vincent's Centre for Applied Medical Research, St. Vincent's Hospital and University of New South Wales, Sydney, New South Wales 2010, Australia
| | - Vicky W-W Tsai
- St Vincent's Centre for Applied Medical Research, St. Vincent's Hospital and University of New South Wales, Sydney, New South Wales 2010, Australia
| | - Yasmin Husaini
- St Vincent's Centre for Applied Medical Research, St. Vincent's Hospital and University of New South Wales, Sydney, New South Wales 2010, Australia
| | - Paul M G Curmi
- School of Physics, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Louise J Brown
- Department of Chemistry and Biomolecular Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - David A Brown
- St Vincent's Centre for Applied Medical Research, St. Vincent's Hospital and University of New South Wales, Sydney, New South Wales 2010, Australia
| | - Samuel N Breit
- St Vincent's Centre for Applied Medical Research, St. Vincent's Hospital and University of New South Wales, Sydney, New South Wales 2010, Australia
| |
Collapse
|
40
|
Tavasoli M, Li L, Al-Momany A, Zhu LF, Adam BA, Wang Z, Ballermann BJ. The chloride intracellular channel 5A stimulates podocyte Rac1, protecting against hypertension-induced glomerular injury. Kidney Int 2016; 89:833-47. [PMID: 26924049 DOI: 10.1016/j.kint.2016.01.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 07/02/2015] [Accepted: 10/15/2015] [Indexed: 11/16/2022]
Abstract
Glomerular capillary hypertension elicits podocyte remodeling and is a risk factor for the progression of glomerular disease. Ezrin, which links podocalyxin to actin in podocytes, is activated through the chloride intracellular channel 5A (CLIC5A)-dependent phosphatidylinositol 4,5 bisphosphate (PI[4,5]P2) accumulation. Because Rac1 is involved in podocyte actin remodeling and can promote PI[4,5]P2 production we determined whether CLIC5A-dependent PI[4,5]P2 generation and ezrin activation are mediated by Rac1. In COS7 cells, CLIC5A expression stimulated Rac1 but not Cdc42 or Rho activity. CLIC5A also stimulated phosphorylation of the Rac1 effector Pak1 in COS7 cells and in cultured mouse podocytes. CLIC5A-induced PI[4,5]P2 accumulation and Pak1 and ezrin phosphorylation were all Rac1 dependent. In DOCA/Salt hypertension, phosphorylated Pak increased in podocytes of wild-type, but not CLIC5-deficient mice. In DOCA/salt hypertensive mice lacking CLIC5, glomerular capillary microaneurysms were more frequent and albuminuria was greater than in wild-type mice. Thus, augmented hypertension-induced glomerular capillary injury in mice lacking CLIC5 results from abrogation of Rac1-dependent Pak and ezrin activation, perhaps reducing the tensile strength of the podocyte actin cytoskeleton.
Collapse
Affiliation(s)
- Mahtab Tavasoli
- Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Laiji Li
- Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Abass Al-Momany
- Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Lin-Fu Zhu
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Benjamin A Adam
- Department of Pathology and Laboratory Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Zhixiang Wang
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Barbara J Ballermann
- Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada; Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
41
|
McRobert EA, Bach LA. Ezrin contributes to impaired podocyte migration and adhesion caused by advanced glycation end products. Nephrology (Carlton) 2015; 21:13-20. [DOI: 10.1111/nep.12526] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/25/2015] [Indexed: 11/30/2022]
Affiliation(s)
| | - Leon A Bach
- Department of Medicine (Alfred); Monash University; Melbourne Victoria Australia
- Department of Endocrinology and Diabetes; Alfred Hospital; Melbourne Victoria Australia
| |
Collapse
|
42
|
Liu G, Honisch S, Liu G, Schmidt S, Alkahtani S, AlKahtane AA, Stournaras C, Lang F. Up-regulation of Orai1 expression and store operated Ca(2+) entry following activation of membrane androgen receptors in MCF-7 breast tumor cells. BMC Cancer 2015; 15:995. [PMID: 26690689 PMCID: PMC4687293 DOI: 10.1186/s12885-015-2014-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 12/15/2015] [Indexed: 12/20/2022] Open
Abstract
Background Membrane androgen receptors (mAR) are functionally expressed in a variety of tumor-cells including the breast tumor-cell line MCF-7. They are specifically activated by testosterone albumin conjugates (TAC). The mAR sensitive signaling includes activation of Ras-related C3 botulinum toxin substrate 1 (Rac1) and reorganization of the actin filament network. Signaling of tumor-cells may further involve up-regulation of pore forming Ca2+ channel protein Orai1, which accomplishes store operated Ca2+ entry (SOCE). This study explored the regulation of Orai1 abundance and SOCE by mAR. Methods Actin filaments were visualized utilizing confocal microscopy, Rac1 activity using GST-GBD assay, Orai1 transcript levels by RT-PCR and total protein abundance by western blotting, Orai1 abundance at the cell surface by confocal microscopy and FACS-analysis, cytosolic Ca2+ activity ([Ca2+]i) utilizing Fura-2-fluorescence, and SOCE from increase of [Ca2+]i following readdition of Ca2+ after store depletion with thapsigargin (1 μM). Results TAC treatment of MCF-7 cells was followed by Rac1 activation, actin polymerization, transient increase of Orai1transcript levels and protein abundance, and transient increase of SOCE. The transient increase of Orai1 protein abundance was abrogated by Rac1 inhibitor NSC23766 (50 μM) and by prevention of actin reorganization with cytochalasin B (1 μM). Conclusions mAR sensitive Rac1 activation and actin reorganization contribute to the regulation of Orai1 protein abundance and SOCE.
Collapse
Affiliation(s)
- Guilai Liu
- Department of Physiology, University of Tuebingen, Tuebingen, Germany.
| | - Sabina Honisch
- Department of Physiology, University of Tuebingen, Tuebingen, Germany.
| | - Guoxing Liu
- Department of Physiology, University of Tuebingen, Tuebingen, Germany.
| | - Sebastian Schmidt
- Department of Physiology, University of Tuebingen, Tuebingen, Germany.
| | - Saad Alkahtani
- Department of Biochemistry, University of Crete Medical School, Heraklion, Crete, Greece. .,Department of Zoology, Science College, King Saud University, Riyadh, Saudi Arabia.
| | - Abdullah A AlKahtane
- Department of Zoology, Science College, King Saud University, Riyadh, Saudi Arabia.
| | - Christos Stournaras
- Department of Physiology, University of Tuebingen, Tuebingen, Germany. .,Department of Biochemistry, University of Crete Medical School, Heraklion, Crete, Greece.
| | - Florian Lang
- Department of Physiology, University of Tuebingen, Tuebingen, Germany. .,Physiologisches Institut, der Universität Tübingen, Gmelinstr. 5, D-72076, Tübingen, Germany.
| |
Collapse
|
43
|
Elferink RPJO, Paulusma CC. MRP2 in cholestasis: Putting down the anchor. J Hepatol 2015; 63:1309-10. [PMID: 26434396 DOI: 10.1016/j.jhep.2015.09.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 09/29/2015] [Indexed: 12/04/2022]
Affiliation(s)
- Ronald P J Oude Elferink
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center Amsterdam, The Netherlands.
| | - Coen C Paulusma
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center Amsterdam, The Netherlands
| |
Collapse
|
44
|
Chigaev A. Does aberrant membrane transport contribute to poor outcome in adult acute myeloid leukemia? Front Pharmacol 2015; 6:134. [PMID: 26191006 PMCID: PMC4489100 DOI: 10.3389/fphar.2015.00134] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 06/15/2015] [Indexed: 12/31/2022] Open
Abstract
Acute myeloid leukemia in adults is a highly heterogeneous disease. Gene expression profiling performed using unsupervised algorithms can be used to distinguish specific groups of patients within a large patient cohort. The identified gene expression signatures can offer insights into underlying physiological mechanisms of disease pathogenesis. Here, the analysis of several related gene expression clusters associated with poor outcome, worst overall survival and highest rates of resistant disease and obtained from the patients at the time of diagnosis or from previously untreated individuals is presented. Surprisingly, these gene clusters appear to be enriched for genes corresponding to proteins involved in transport across membranes (transporters, carriers and channels). Several ideas describing the possible relationship of membrane transport activity and leukemic cell biology, including the "Warburg effect," the specific role of chloride ion transport, direct "import" of metabolic energy through uptake of creatine phosphate, and modification of the bone marrow niche microenvironment are discussed.
Collapse
Affiliation(s)
- Alexandre Chigaev
- Department of Pathology and Cancer Center, University of New Mexico Health Sciences Center, University of New Mexico Albuquerque, NM, USA
| |
Collapse
|
45
|
Parodi M, Pedrazzi M, Cantoni C, Averna M, Patrone M, Cavaletto M, Spertino S, Pende D, Balsamo M, Pietra G, Sivori S, Carlomagno S, Mingari MC, Moretta L, Sparatore B, Vitale M. Natural Killer (NK)/melanoma cell interaction induces NK-mediated release of chemotactic High Mobility Group Box-1 (HMGB1) capable of amplifying NK cell recruitment. Oncoimmunology 2015; 4:e1052353. [PMID: 26587323 DOI: 10.1080/2162402x.2015.1052353] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 05/12/2015] [Accepted: 05/13/2015] [Indexed: 02/06/2023] Open
Abstract
In this study we characterize a new mechanism by which Natural Killer (NK) cells may amplify their recruitment to tumors. We show that NK cells, upon interaction with melanoma cells, can release a chemotactic form of High Mobility Group Box-1 (HMGB1) protein capable of attracting additional activated NK cells. We first demonstrate that the engagement of different activating NK cell receptors, including those mainly involved in tumor cell recognition can induce the active release of HMGB1. Then we show that during NK-mediated tumor cell killing two HMGB1 forms are released, each displaying a specific electrophoretic mobility possibly corresponding to a different redox status. By the comparison of normal and perforin-defective NK cells (which are unable to kill target cells) we demonstrate that, in NK/melanoma cell co-cultures, NK cells specifically release an HMGB1 form that acts as chemoattractant, while dying tumor cells passively release a non-chemotactic HMGB1. Finally, we show that Receptor for Advanced Glycation End products is expressed by NK cells and mediates HMGB1-induced NK cell chemotaxis. Proteomic analysis of NK cells exposed to recombinant HMGB1 revealed that this molecule, besides inducing immediate chemotaxis, also promotes changes in the expression of proteins involved in the regulation of the cytoskeletal network. Importantly, these modifications could be associated with an increased motility of NK cells. Thus, our findings allow the definition of a previously unidentified mechanism used by NK cells to amplify their response to tumors, and provide additional clues for the emerging role of HMGB1 in immunomodulation and tumor immunity.
Collapse
Affiliation(s)
- Monica Parodi
- Department of Experimental Medicine (DIMES); University of Genova ; Genova, Italy
| | - Marco Pedrazzi
- Department of Experimental Medicine (DIMES); University of Genova ; Genova, Italy
| | - Claudia Cantoni
- Department of Experimental Medicine (DIMES); University of Genova ; Genova, Italy ; Center of Excellence for Biomedical Research (CEBR); University of Genova ; Genova, Italy ; Istituto Giannina Gaslini ; Genova, Italy
| | - Monica Averna
- Department of Experimental Medicine (DIMES); University of Genova ; Genova, Italy ; Center of Excellence for Biomedical Research (CEBR); University of Genova ; Genova, Italy
| | - Mauro Patrone
- Department of Sciences and Technological Innovation (DiSIT); University of Piemonte Orientale ; Alessandria, Italy
| | - Maria Cavaletto
- Department of Sciences and Technological Innovation (DiSIT); University of Piemonte Orientale ; Alessandria, Italy
| | - Stefano Spertino
- Department of Sciences and Technological Innovation (DiSIT); University of Piemonte Orientale ; Alessandria, Italy
| | | | - Mirna Balsamo
- Department of Experimental Medicine (DIMES); University of Genova ; Genova, Italy
| | - Gabriella Pietra
- Department of Experimental Medicine (DIMES); University of Genova ; Genova, Italy ; IRCCS AOU San Martino-IST ; Genova, Italy
| | - Simona Sivori
- Department of Experimental Medicine (DIMES); University of Genova ; Genova, Italy ; Center of Excellence for Biomedical Research (CEBR); University of Genova ; Genova, Italy
| | - Simona Carlomagno
- Department of Experimental Medicine (DIMES); University of Genova ; Genova, Italy
| | - Maria Cristina Mingari
- Department of Experimental Medicine (DIMES); University of Genova ; Genova, Italy ; Center of Excellence for Biomedical Research (CEBR); University of Genova ; Genova, Italy ; IRCCS AOU San Martino-IST ; Genova, Italy
| | | | - Bianca Sparatore
- Department of Experimental Medicine (DIMES); University of Genova ; Genova, Italy ; Center of Excellence for Biomedical Research (CEBR); University of Genova ; Genova, Italy
| | | |
Collapse
|
46
|
Al Khamici H, Brown LJ, Hossain KR, Hudson AL, Sinclair-Burton AA, Ng JPM, Daniel EL, Hare JE, Cornell BA, Curmi PMG, Davey MW, Valenzuela SM. Members of the chloride intracellular ion channel protein family demonstrate glutaredoxin-like enzymatic activity. PLoS One 2015; 10:e115699. [PMID: 25581026 PMCID: PMC4291220 DOI: 10.1371/journal.pone.0115699] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 11/26/2014] [Indexed: 01/07/2023] Open
Abstract
The Chloride Intracellular Ion Channel (CLIC) family consists of six evolutionarily conserved proteins in humans. Members of this family are unusual, existing as both monomeric soluble proteins and as integral membrane proteins where they function as chloride selective ion channels, however no function has previously been assigned to their soluble form. Structural studies have shown that in the soluble form, CLIC proteins adopt a glutathione S-transferase (GST) fold, however, they have an active site with a conserved glutaredoxin monothiol motif, similar to the omega class GSTs. We demonstrate that CLIC proteins have glutaredoxin-like glutathione-dependent oxidoreductase enzymatic activity. CLICs 1, 2 and 4 demonstrate typical glutaredoxin-like activity using 2-hydroxyethyl disulfide as a substrate. Mutagenesis experiments identify cysteine 24 as the catalytic cysteine residue in CLIC1, which is consistent with its structure. CLIC1 was shown to reduce sodium selenite and dehydroascorbate in a glutathione-dependent manner. Previous electrophysiological studies have shown that the drugs IAA-94 and A9C specifically block CLIC channel activity. These same compounds inhibit CLIC1 oxidoreductase activity. This work for the first time assigns a functional activity to the soluble form of the CLIC proteins. Our results demonstrate that the soluble form of the CLIC proteins has an enzymatic activity that is distinct from the channel activity of their integral membrane form. This CLIC enzymatic activity may be important for protecting the intracellular environment against oxidation. It is also likely that this enzymatic activity regulates the CLIC ion channel function.
Collapse
Affiliation(s)
- Heba Al Khamici
- School of Medical and Molecular Biosciences, University of Technology Sydney, Sydney, New South Wales 2007, Australia
| | - Louise J. Brown
- Department of Chemistry and Bimolecular Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Khondker R. Hossain
- School of Medical and Molecular Biosciences, University of Technology Sydney, Sydney, New South Wales 2007, Australia
- Bragg Institute, Australian Nuclear Science and Technology Organisation, Sydney, New South Wales 2234, Australia
| | - Amanda L. Hudson
- School of Medical and Molecular Biosciences, University of Technology Sydney, Sydney, New South Wales 2007, Australia
| | - Alxcia A. Sinclair-Burton
- School of Medical and Molecular Biosciences, University of Technology Sydney, Sydney, New South Wales 2007, Australia
| | - Jane Phui Mun Ng
- School of Medical and Molecular Biosciences, University of Technology Sydney, Sydney, New South Wales 2007, Australia
| | - Elizabeth L. Daniel
- Department of Chemistry and Bimolecular Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Joanna E. Hare
- Department of Chemistry and Bimolecular Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Bruce A. Cornell
- Surgical Diagnostics, Roseville, Sydney, New South Wales 2069, Australia
| | - Paul M. G. Curmi
- School of Physics, University of New South Wales, Sydney, New South Wales 2052, Australia
- Centre for Applied Medical Research, St Vincent's Hospital, Sydney, New South Wales 2010, Australia
| | - Mary W. Davey
- School of Medical and Molecular Biosciences, University of Technology Sydney, Sydney, New South Wales 2007, Australia
| | - Stella M. Valenzuela
- School of Medical and Molecular Biosciences, University of Technology Sydney, Sydney, New South Wales 2007, Australia
- Centre for Health Technologies, University of Technology Sydney, Sydney, New South Wales 2007, Australia
| |
Collapse
|
47
|
Nammalwar RC, Heil A, Gerke V. Ezrin interacts with the scaffold protein IQGAP1 and affects its cortical localization. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1853:2086-94. [PMID: 25554515 DOI: 10.1016/j.bbamcr.2014.12.026] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 12/18/2014] [Accepted: 12/19/2014] [Indexed: 10/24/2022]
Abstract
The cortical cytoskeleton constitutes an important subcellular structure that determines cell shape and regulates cell migration as well as membrane traffic to and from the plasma membrane. Many components of the cortical cytoskeleton have been identified including structural and scaffolding proteins, membrane-cytoskeleton linker proteins and signaling intermediates. We describe here an association of the membrane-F-actin linker protein ezrin with the scaffolding protein IQGAP1 that serves as a hub for concentrating different signaling complexes. Both, ezrin and IQGAP1 bind in a Ca²⁺-dependent manner to the EF hand protein S100P and complexes consisting of Ca²⁺-bound S100P, IQGAP1 and ezrin can be isolated by immunoprecipitation. Ezrin and IQGAP1 also interact in the absence of Ca²⁺, thus independent of S100P. Direct ezrin-IQGAP1 interaction can be shown with the purified proteins. It is mediated via the N-terminal FERM domain of ezrin and the IQ domain of IQGAP1, respectively. Ezrin and IQGAP1 colocalize in the submembraneous cytoskeleton and in cellular protrusions of human epithelial cells and knockdown of ezrin reduces the cortical localization of IQGAP1. Thus, ezrin appears to participate in recruiting IQGAP1 to the cell cortex thereby establishing a close connection between membrane-F-actin contacts and actin regulators that can be assembled by IQGAP1. This article is part of a Special Issue entitled: 13th European Symposium on Calcium.
Collapse
Affiliation(s)
- Rathangadhara Chakrapani Nammalwar
- Institute of Medical Biochemistry, Centre for Molecular Biology of Inflammation (ZMBE), Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Münster, Von-Esmarch-Str. 56, 48149 Münster, Germany
| | - Annika Heil
- Institute of Medical Biochemistry, Centre for Molecular Biology of Inflammation (ZMBE), Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Münster, Von-Esmarch-Str. 56, 48149 Münster, Germany
| | - Volker Gerke
- Institute of Medical Biochemistry, Centre for Molecular Biology of Inflammation (ZMBE), Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Münster, Von-Esmarch-Str. 56, 48149 Münster, Germany.
| |
Collapse
|
48
|
Takeshima H, Venturi E, Sitsapesan R. New and notable ion-channels in the sarcoplasmic/endoplasmic reticulum: do they support the process of intracellular Ca²⁺ release? J Physiol 2014; 593:3241-51. [PMID: 26228553 DOI: 10.1113/jphysiol.2014.281881] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 10/10/2014] [Indexed: 12/11/2022] Open
Abstract
Intracellular Ca(2+) release through ryanodine receptor (RyR) and inositol trisphosphate receptor (IP3 R) channels is supported by a complex network of additional proteins that are located in or near the Ca(2+) release sites. In this review, we focus, not on RyR/IP3 R, but on other ion-channels that are known to be present in the sarcoplasmic/endoplasmic reticulum (ER/SR) membranes. We review their putative physiological roles and the evidence suggesting that they may support the process of intracellular Ca(2+) release, either indirectly by manipulating ionic fluxes across the ER/SR membrane or by directly interacting with a Ca(2+) -release channel. These channels rarely receive scientific attention because of the general lack of information regarding their biochemical and/or electrophysiological characteristics makes it difficult to predict their physiological roles and their impact on SR Ca(2+) fluxes. We discuss the possible role of SR K(+) channels and, in parallel, detail the known biochemical and biophysical properties of the trimeric intracellular cation (TRIC) proteins and their possible biological and pathophysiological roles in ER/SR Ca(2+) release. We summarise what is known regarding Cl(-) channels in the ER/SR and the non-selective cation channels or putative 'Ca(2+) leak channels', including mitsugumin23 (MG23), pannexins, presenilins and the transient receptor potential (TRP) channels that are distributed across ER/SR membranes but which have not yet been fully characterised functionally.
Collapse
Affiliation(s)
- Hiroshi Takeshima
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, 606-8501, Japan
| | - Elisa Venturi
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | | |
Collapse
|
49
|
Al-Momany A, Li L, Alexander RT, Ballermann BJ. Clustered PI(4,5)P₂ accumulation and ezrin phosphorylation in response to CLIC5A. J Cell Sci 2014; 127:5164-78. [PMID: 25344252 DOI: 10.1242/jcs.147744] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
CLIC5A (encoded by CLIC5) is a component of the ezrin-NHERF2-podocalyxin complex in renal glomerular podocyte foot processes. We explored the mechanism(s) by which CLIC5A regulates ezrin function. In COS-7 cells, CLIC5A augmented ezrin phosphorylation without changing ezrin abundance, increased the association of ezrin with the cytoskeletal fraction and enhanced actin polymerization and the formation of cell surface projections. CLIC5A caused the phosphatidylinositol 4,5-bisphosphate [PI(4,5)P2] reporter RFP-PH-PLC to translocate from the cytosol to discrete plasma membrane clusters at the cell surface, where it colocalized with CLIC5A. Transiently expressed HA-PIP5Kα colocalized with GFP-CLIC5A and was pulled from cell lysates by GST-CLIC5A, and silencing of endogenous PIP5Kα abrogated CLIC5A-dependent ERM phosphorylation. N- and C-terminal deletion mutants of CLIC5A, which failed to associate with the plasma membrane, failed to colocalize with PIP5Kα, did not alter the abundance of PI(4,5)P2 plasma membrane clusters and failed to enhance ezrin phosphorylation. Relative to wild-type mice, in CLIC5-deficient mice, the phosphorylation of glomerular ezrin was diminished and the cytoskeletal association of both ezrin and NHERF2 was reduced. Therefore, the mechanism of CLIC5A action involves clustered plasma membrane PI(4,5)P2 accumulation through an interaction of CLIC5A with PI(4,5)P2-generating kinases, in turn facilitating ezrin activation and actin-dependent cell surface remodeling.
Collapse
Affiliation(s)
- Abass Al-Momany
- Department of Physiology, University of Alberta, Edmonton, AL T6G 2V2, Canada
| | - Laiji Li
- Department of Medicine (Nephrology), University of Alberta, Edmonton, AL T6G 2V2, Canada
| | - R Todd Alexander
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AL T6G 2V2, Canada
| | - Barbara J Ballermann
- Department of Physiology, University of Alberta, Edmonton, AL T6G 2V2, Canada Department of Medicine (Nephrology), University of Alberta, Edmonton, AL T6G 2V2, Canada
| |
Collapse
|
50
|
Seco CZ, Oonk AMM, Domínguez-Ruiz M, Draaisma JMT, Gandía M, Oostrik J, Neveling K, Kunst HPM, Hoefsloot LH, del Castillo I, Pennings RJE, Kremer H, Admiraal RJC, Schraders M. Progressive hearing loss and vestibular dysfunction caused by a homozygous nonsense mutation in CLIC5. Eur J Hum Genet 2014; 23:189-94. [PMID: 24781754 DOI: 10.1038/ejhg.2014.83] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Revised: 03/20/2014] [Accepted: 04/04/2014] [Indexed: 11/10/2022] Open
Abstract
In a consanguineous Turkish family diagnosed with autosomal recessive nonsyndromic hearing impairment (arNSHI), a homozygous region of 47.4 Mb was shared by the two affected siblings on chromosome 6p21.1-q15. This region contains 247 genes including the known deafness gene MYO6. No pathogenic variants were found in MYO6, neither with sequence analysis of the coding region and splice sites nor with mRNA analysis. Subsequent candidate gene evaluation revealed CLIC5 as an excellent candidate gene. The orthologous mouse gene is mutated in the jitterbug mutant that exhibits progressive hearing impairment and vestibular dysfunction. Mutation analysis of CLIC5 revealed a homozygous nonsense mutation c.96T>A (p.(Cys32Ter)) that segregated with the hearing loss. Further analysis of CLIC5 in 213 arNSHI patients from mostly Dutch and Spanish origin did not reveal any additional pathogenic variants. CLIC5 mutations are thus not a common cause of arNSHI in these populations. The hearing loss in the present family had an onset in early childhood and progressed from mild to severe or even profound before the second decade. Impaired hearing is accompanied by vestibular areflexia and in one of the patients with mild renal dysfunction. Although we demonstrate that CLIC5 is expressed in many other human tissues, no additional symptoms were observed in these patients. In conclusion, our results show that CLIC5 is a novel arNSHI gene involved in progressive hearing impairment, vestibular and possibly mild renal dysfunction in a family of Turkish origin.
Collapse
Affiliation(s)
- Celia Zazo Seco
- 1] Department of Otorhinolaryngology, Hearing & Genes, Radboud University Medical Center, Nijmegen, The Netherlands [2] Nijmegen Centre for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands [3] Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Anne M M Oonk
- 1] Department of Otorhinolaryngology, Hearing & Genes, Radboud University Medical Center, Nijmegen, The Netherlands [2] Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - María Domínguez-Ruiz
- 1] Servicio de Genética, Hospital Universitario Ramon y Cajal, IRYCIS, Madrid, Spain [2] Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Jos M T Draaisma
- Department of Pediatrics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marta Gandía
- 1] Servicio de Genética, Hospital Universitario Ramon y Cajal, IRYCIS, Madrid, Spain [2] Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Jaap Oostrik
- 1] Department of Otorhinolaryngology, Hearing & Genes, Radboud University Medical Center, Nijmegen, The Netherlands [2] Nijmegen Centre for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands [3] Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Kornelia Neveling
- 1] Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands [2] Institute for Genetic and Metabolic Disease, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Henricus P M Kunst
- 1] Department of Otorhinolaryngology, Hearing & Genes, Radboud University Medical Center, Nijmegen, The Netherlands [2] Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Lies H Hoefsloot
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ignacio del Castillo
- Servicio de Genética, Hospital Universitario Ramon y Cajal, IRYCIS, Madrid, Spain
| | - Ronald J E Pennings
- 1] Department of Otorhinolaryngology, Hearing & Genes, Radboud University Medical Center, Nijmegen, The Netherlands [2] Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Hannie Kremer
- 1] Department of Otorhinolaryngology, Hearing & Genes, Radboud University Medical Center, Nijmegen, The Netherlands [2] Nijmegen Centre for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands [3] Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands [4] Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ronald J C Admiraal
- 1] Department of Otorhinolaryngology, Hearing & Genes, Radboud University Medical Center, Nijmegen, The Netherlands [2] Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Margit Schraders
- 1] Department of Otorhinolaryngology, Hearing & Genes, Radboud University Medical Center, Nijmegen, The Netherlands [2] Nijmegen Centre for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands [3] Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|