1
|
Lowry AJ, Liang P, Song M, Wan Y, Pei ZM, Yang H, Zhang Y. TMEM16 and OSCA/TMEM63 proteins share a conserved potential to permeate ions and phospholipids. eLife 2024; 13:RP96957. [PMID: 39495104 PMCID: PMC11534332 DOI: 10.7554/elife.96957] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2024] Open
Abstract
The calcium-activated TMEM16 proteins and the mechanosensitive/osmolarity-activated OSCA/TMEM63 proteins belong to the Transmembrane Channel/Scramblase (TCS) superfamily. Within the superfamily, OSCA/TMEM63 proteins, as well as TMEM16A and TMEM16B, are thought to function solely as ion channels. However, most TMEM16 members, including TMEM16F, maintain an additional function as scramblases, rapidly exchanging phospholipids between leaflets of the membrane. Although recent studies have advanced our understanding of TCS structure-function relationships, the molecular determinants of TCS ion and lipid permeation remain unclear. Here, we show that single mutations along the transmembrane helix (TM) 4/6 interface allow non-scrambling TCS members to permeate phospholipids. In particular, this study highlights the key role of TM 4 in controlling TCS ion and lipid permeation and offers novel insights into the evolution of the TCS superfamily, suggesting that, like TMEM16s, the OSCA/TMEM63 family maintains a conserved potential to permeate ions and phospholipids.
Collapse
Affiliation(s)
- Augustus J Lowry
- Department of Biochemistry, Duke University School of MedicineDurhamUnited States
| | - Pengfei Liang
- Department of Biochemistry, Duke University School of MedicineDurhamUnited States
| | - Mo Song
- Institute of Molecular Physiology, Shenzhen Bay LaboratoryGuangdongChina
| | - Yuichun Wan
- Department of Biochemistry, Duke University School of MedicineDurhamUnited States
| | - Zhen-Ming Pei
- Department of Biology, Duke UniversityDurhamUnited States
| | - Huanghe Yang
- Department of Biochemistry, Duke University School of MedicineDurhamUnited States
- Department of Neurobiology, Duke University School of MedicineDurhamUnited States
| | - Yang Zhang
- Department of Biochemistry, Duke University School of MedicineDurhamUnited States
- Institute of Molecular Physiology, Shenzhen Bay LaboratoryGuangdongChina
| |
Collapse
|
2
|
Lian Z, Luo W, Liu J, Wang J, Chai W, Wang Y, Sethi S, Ma X. Analysis of ANO6, HAPLN1, and EDIL3 Polymorphisms in Patients with Ankylosing Spondylitis in a Chinese Han Population: A Case-Control Study. Genet Test Mol Biomarkers 2024; 28:385-392. [PMID: 39358671 DOI: 10.1089/gtmb.2023.0569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/04/2024] Open
Abstract
Background: Earlier research has demonstrated a genetic basis for the susceptibility to ankylosing spondylitis (AS) and the severity of AS. By employing a genome-wide association study, recent work has established a correlation between the susceptibility to AS and the ANO6, HAPLN, and EDIL3 genes in a Western study population-though alternative studies have not corroborated these findings. This study aims to examine the effects of ANO6, HAPLN1, and EDIL3 polymorphisms on the susceptibility and severity of AS among the predominantly Chinese Han population. Methods: The study involved the collection of blood samples from 497 patients with AS and 498 nonrelated healthy individuals. All participants in the study were human leukocyte antigen (HLA) HLA-B27 positive and of Han Chinese descent. Illness severity was the criteria used for classifying patients with AS. Thirteen tagSNPs in ANO6, HAPLN1, and EDIL3 were chosen and then subjected to genetic typing. Analysis was conducted on the occurrence rates of various genotypes and alleles between the control group and patients with varying AS severity. Results: Following Bonferroni correction, it was found that the rs4768085 and rs17095830 single nucleotide polymorphism (SNPs) in ANO6 were related to the susceptibility to AS. Further, the rs6869296 SNP in HAPLN1 and the rs2301071 SNP between EDIL3 and HAPLN1 were also related to AS susceptibility. Regarding AS severity, the rs4768085, rs2897868, rs7965430, and rs11182965 SNPs in ANO6 were found to be associated. Conclusions: Among the Han population in China, the ANO6 and HAPLN1 genes are related to the susceptibility to AS; the ANO6 gene is also associated with the severity of AS.
Collapse
Affiliation(s)
- Zijian Lian
- Department of Orthopaedics, Tianjin Hospital, Tianjin, China
| | - Wei Luo
- Department of Orthopaedics, Tianjin Hospital, Tianjin, China
| | - Jun Liu
- Department of Orthopaedics, Tianjin Hospital, Tianjin, China
| | - Jing Wang
- Department of Orthopaedics, Tianjin Hospital, Tianjin, China
| | - Wei Chai
- Department of Orthopaedics, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Yan Wang
- Department of Orthopaedics, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Sahil Sethi
- Department of Orthopaedics, University of Chicago Hospital, Chicago, Illinois, USA
| | - Xinlong Ma
- Department of Orthopaedics, Tianjin Hospital, Tianjin, China
| |
Collapse
|
3
|
Wang H, Zhao W, Wang D, Chen J. ANO6 (TMEM16F) inhibits gastrointestinal stromal tumor growth and induces ferroptosis. Open Med (Wars) 2024; 19:20240941. [PMID: 38756246 PMCID: PMC11097043 DOI: 10.1515/med-2024-0941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 02/21/2024] [Accepted: 03/05/2024] [Indexed: 05/18/2024] Open
Abstract
Herein, we elucidate the potential role of ANO6 (TMEM16F) in gastrointestinal stromal tumors (GISTs). ANO6 expression in GIST and adjacent normal tissues was determined using reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and western blotting. Cell proliferation, apoptosis, and pyroptosis were examined utilizing 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide, terminal deoxynucleotidyl transferase dUTP Nick-End Labeling staining, and flow cytometry. In addition, the total iron and Fe2+ levels were assessed. IL-18 and IL-1β levels were also evaluated. Lipid reactive oxygen species (ROS), cystine (Cys), glutathione (GSH), and glutathione peroxidase 4 (GPX4) levels were evaluated using appropriate kits. Ferroptotic markers, including Ptgs2, Chac1, SLC7A11, and SLC3A2, were analyzed by RT-qPCR, western blotting, and immunohistochemistry. ANO6 expression decreased in GIST tissues. ANO6-plasmid inhibits proliferation, induces apoptosis, and promotes pyroptosis in GIST-T1 and GIST-T1 IR cells. The ANO6-plasmid induced ferroptosis, as confirmed by enhanced lipid ROS levels, increased intracellular concentrations of total iron and Fe2+, promoted Ptgs2 and Chac1 expression, reduced Cys, GSH, and GPX4 levels, and downregulated SLC7A11 and SLC3A2 expression after in vitro and in vivo treatment with ANO6-plasmid. Moreover, the ANO6-plasmid inhibited GIST growth in vivo. Therefore, ANO6 may be a promising therapeutic target for blocking the development of GIST via the induction of apoptosis, pyroptosis, and ferroptosis.
Collapse
Affiliation(s)
- Hao Wang
- School of Public Health, Nanjing Medical University, Nanjing211166, China
- Department of Gastrointestinal Surgery, The Yangzhou School of Clinical Medicine of Nanjing Medical University, Yangzhou225001, China
| | - Wei Zhao
- Department of Gastrointestinal Surgery, The Yangzhou School of Clinical Medicine of Nanjing Medical University, Yangzhou225001, China
| | - Daorong Wang
- Department of Gastrointestinal Surgery, The Yangzhou School of Clinical Medicine of Nanjing Medical University, Yangzhou225001, China
- General Surgery Institute of Yangzhou, Yangzhou University, Yangzhou225001, China
- Yangzhou Key Laboratory of Basic and Clinical Transformation of Digestive and Metabolic Diseases, Yangzhou225001, China
| | - Jin Chen
- The Key Laboratory of Modern Toxicology, Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Jiangning District, Nanjing211166, China
- Jiangsu Province Engineering Research Center of Antibody Drug, Key Laboratory of Antibody Technique of National Health Commission, Nanjing Medical University, Nanjing211166, China
| |
Collapse
|
4
|
Dewdney B, Ursich L, Fletcher EV, Johns TG. Anoctamins and Calcium Signalling: An Obstacle to EGFR Targeted Therapy in Glioblastoma? Cancers (Basel) 2022; 14:cancers14235932. [PMID: 36497413 PMCID: PMC9740065 DOI: 10.3390/cancers14235932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 11/28/2022] [Accepted: 11/28/2022] [Indexed: 12/05/2022] Open
Abstract
Glioblastoma is the most common form of high-grade glioma in adults and has a poor survival rate with very limited treatment options. There have been no significant advancements in glioblastoma treatment in over 30 years. Epidermal growth factor receptor is upregulated in most glioblastoma tumours and, therefore, has been a drug target in recent targeted therapy clinical trials. However, while many inhibitors and antibodies for epidermal growth factor receptor have demonstrated promising anti-tumour effects in preclinical models, they have failed to improve outcomes for glioblastoma patients in clinical trials. This is likely due to the highly plastic nature of glioblastoma tumours, which results in therapeutic resistance. Ion channels are instrumental in the development of many cancers and may regulate cellular plasticity in glioblastoma. This review will explore the potential involvement of a class of calcium-activated chloride channels called anoctamins in brain cancer. We will also discuss the integrated role of calcium channels and anoctamins in regulating calcium-mediated signalling pathways, such as epidermal growth factor signalling, to promote brain cancer cell growth and migration.
Collapse
Affiliation(s)
- Brittany Dewdney
- Cancer Centre, Telethon Kids Institute, Perth, WA 6009, Australia
- Centre for Child Health Research, University of Western Australia, Perth, WA 6009, Australia
- Correspondence: ; Tel.: +61-8-6319-1023
| | - Lauren Ursich
- Cancer Centre, Telethon Kids Institute, Perth, WA 6009, Australia
- School of Biomedical Sciences, University of Western Australia, Perth, WA 6009, Australia
| | - Emily V. Fletcher
- Cancer Centre, Telethon Kids Institute, Perth, WA 6009, Australia
- Centre for Child Health Research, University of Western Australia, Perth, WA 6009, Australia
| | - Terrance G. Johns
- Cancer Centre, Telethon Kids Institute, Perth, WA 6009, Australia
- Centre for Child Health Research, University of Western Australia, Perth, WA 6009, Australia
| |
Collapse
|
5
|
Li B, Fan Q, Zheng L, Liu P, Fang N. Significance of Anoctamin 6 in progression and prognostic prediction of gastric adenocarcinoma. Histol Histopathol 2022; 37:1007-1017. [PMID: 35548923 DOI: 10.14670/hh-18-469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
BACKGROUND Gastric cancer is one of the most lethal malignancies worldwide with surgery as the only curative therapy. However, postoperative overall survival of gastric cancer is far from satisfactory although significant improvement has been made in adjuvant therapies. Gastric cancer is characterized as highly heterogeneous and illustrating the molecular mechanisms is invaluable for both identification of novel prognostic biomarkers and development of therapeutic drugs. Here we aimed to investigate the participation of Anoctamin 6 (ANO6) in gastric adenocarcinoma. METHODS Immunohistochemical (IHC) staining was used to explore the expression pattern of ANO6 in tumor tissues from gastric adenocarcinoma patients (n=108). Clinicopathological data was subjected to Kaplan-Meier survival and Cox multivariate analyses to evaluate prognostic predictors. Overexpression and silencing procedures were performed on gastric cancer cell lines to investigate the functional mechanisms of ANO6 in regulating tumor development. RESULTS Higher ANO6 expression showed a positive correlation with advanced tumor stage of gastric cancer. Univariate and multivariate analyses revealed that ANO6 was an independent prognostic factor for overall survival of gastric cancer. An in vitro study demonstrated that ANO6 can promote cell proliferation while silencing ANO6 significantly downregulated cell viability. CONCLUSION High ANO6 expression in gastric cancer indicates poor clinical outcomes, and ANO6 may act as a potential target for novel therapy development targeting gastric cancer.
Collapse
Affiliation(s)
- Bin Li
- Department of Gastroenterology, Third Affiliated Hospital of Nanchang University (Nanchang First Hospital), Nanchang, Jiangxi Province, China
| | - Qiong Fan
- Department of Gastroenterology, Third Affiliated Hospital of Nanchang University (Nanchang First Hospital), Nanchang, Jiangxi Province, China
| | - Li Zheng
- Department of Gastroenterology, Third Affiliated Hospital of Nanchang University (Nanchang First Hospital), Nanchang, Jiangxi Province, China
| | - Peng Liu
- Department of Gastroenterology, Third Affiliated Hospital of Nanchang University (Nanchang First Hospital), Nanchang, Jiangxi Province, China
| | - Nian Fang
- Department of Gastroenterology, Third Affiliated Hospital of Nanchang University (Nanchang First Hospital), Nanchang, Jiangxi Province, China.
| |
Collapse
|
6
|
Piraino LR, Benoit DSW, DeLouise LA. Optimizing Soluble Cues for Salivary Gland Tissue Mimetics Using a Design of Experiments (DoE) Approach. Cells 2022; 11:1962. [PMID: 35741092 PMCID: PMC9222211 DOI: 10.3390/cells11121962] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/14/2022] [Accepted: 06/14/2022] [Indexed: 02/01/2023] Open
Abstract
The development of therapies to prevent or treat salivary gland dysfunction has been limited by a lack of functional in vitro models. Specifically, critical markers of salivary gland secretory phenotype downregulate rapidly ex vivo. Here, we utilize a salivary gland tissue chip model to conduct a design of experiments (DoE) approach to test combinations of seven soluble cues that were previously shown to maintain or improve salivary gland cell function. This approach uses statistical techniques to improve efficiency and accuracy of combinations of factors. The DoE-designed culture conditions improve markers of salivary gland function. Data show that the EGFR inhibitor, EKI-785, maintains relative mRNA expression of Mist1, a key acinar cell transcription factor, while FGF10 and neurturin promote mRNA expression of Aqp5 and Tmem16a, channel proteins involved in secretion. Mist1 mRNA expression correlates with increased secretory function, including calcium signaling and mucin (PAS-AB) staining. Overall, this study demonstrates that media conditions can be efficiently optimized to support secretory function in vitro using a DoE approach.
Collapse
Affiliation(s)
- Lindsay R. Piraino
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14627, USA; (L.R.P.); (D.S.W.B.)
| | - Danielle S. W. Benoit
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14627, USA; (L.R.P.); (D.S.W.B.)
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY 14642, USA
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14642, USA
- Materials Science Program, University of Rochester, Rochester, NY 14627, USA
- Department of Chemical Engineering, University of Rochester, Rochester, NY 14627, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Lisa A. DeLouise
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14627, USA; (L.R.P.); (D.S.W.B.)
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
- Materials Science Program, University of Rochester, Rochester, NY 14627, USA
- Department of Dermatology, University of Rochester Medical Center, Rochester, NY 14642, USA
| |
Collapse
|
7
|
Behuria HG, Dash S, Sahu SK. Phospholipid Scramblases: Role in Cancer Progression and Anticancer Therapeutics. Front Genet 2022; 13:875894. [PMID: 35422844 PMCID: PMC9002267 DOI: 10.3389/fgene.2022.875894] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 03/11/2022] [Indexed: 11/13/2022] Open
Abstract
Phospholipid scramblases (PLSCRs) that catalyze rapid mixing of plasma membrane lipids result in surface exposure of phosphatidyl serine (PS), a lipid normally residing to the inner plasma membrane leaflet. PS exposure provides a chemotactic eat-me signal for phagocytes resulting in non-inflammatory clearance of apoptotic cells by efferocytosis. However, metastatic tumor cells escape efferocytosis through alteration of tumor microenvironment and apoptotic signaling. Tumor cells exhibit altered membrane features, high constitutive PS exposure, low drug permeability and increased multidrug resistance through clonal evolution. PLSCRs are transcriptionally up-regulated in tumor cells leading to plasma membrane remodeling and aberrant PS exposure on cell surface. In addition, PLSCRs interact with multiple cellular components to modulate cancer progression and survival. While PLSCRs and PS exposed on tumor cells are novel drug targets, many exogenous molecules that catalyze lipid scrambling on tumor plasma membrane are potent anticancer therapeutic molecules. In this review, we provide a comprehensive analysis of scramblase mediated signaling events, membrane alteration specific to tumor development and possible therapeutic implications of scramblases and PS exposure.
Collapse
Affiliation(s)
- Himadri Gourav Behuria
- Laboratory of Molecular Membrane Biology, Department of Biotechnology, Maharaja Sriram Chandra Bhanjadeo University, Baripada, India
| | - Sabyasachi Dash
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Santosh Kumar Sahu
- Laboratory of Molecular Membrane Biology, Department of Biotechnology, Maharaja Sriram Chandra Bhanjadeo University, Baripada, India
| |
Collapse
|
8
|
Luo S, Wang H, Bai L, Chen Y, Chen S, Gao K, Wang H, Wu S, Song H, Ma K, Liu M, Yao F, Fang Y, Xiao Q. Activation of TMEM16A Ca 2+-activated Cl - channels by ROCK1/moesin promotes breast cancer metastasis. J Adv Res 2021; 33:253-264. [PMID: 34603794 PMCID: PMC8463928 DOI: 10.1016/j.jare.2021.03.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 01/28/2021] [Accepted: 03/13/2021] [Indexed: 12/01/2022] Open
Abstract
Introduction Transmembrane protein 16A (TMEM16A) is a Ca2+-activated chloride channel that plays a role in cancer cell proliferation, migration, invasion, and metastasis. However, whether TMEM16A contributes to breast cancer metastasis remains unknown. Objective In this study, we investigated whether TMEM16A channel activation by ROCK1/moesin promotes breast cancer metastasis. Methods Wound healing assays and transwell migration and invasion assays were performed to study the migration and invasion of MCF-7 and T47D breast cancer cells. Western blotting was performed to evaluate the protein expression, and whole-cell patch clamp recordings were used to record TMEM16A Cl− currents. A mouse model of breast cancer lung metastasis was generated by injecting MCF-7 cells via the tail vein. Metastatic nodules in the lung were assessed by hematoxylin and eosin staining. Lymph node metastasis, overall survival, and metastasis-free survival of breast cancer patients were assessed using immunohistochemistry and The Cancer Genome Atlas dataset. Results TMEM16A activation promoted breast cancer cell migration and invasion in vitro as well as breast cancer metastasis in mice. Patients with breast cancer who had higher TMEM16A levels showed greater lymph node metastasis and shorter survival. Mechanistically, TMEM16A promoted migration and invasion by activating EGFR/STAT3/ROCK1 signaling, and the role of the TMEM16A channel activity was important in this respect. ROCK1 activation by RhoA enhanced the TMEM16A channel activity via the phosphorylation of moesin at T558. The cooperative action of TMEM16A and ROCK1 was supported through clinical findings indicating that breast cancer patients with high levels of TMEM16A/ROCK1 expression showed greater lymph node metastasis and poor survival. Conclusion Our findings revealed a novel mechanism underlying TMEM16A-mediated breast cancer metastasis, in which ROCK1 increased TMEM16A channel activity via moesin phosphorylation and the increase in TMEM16A channel activities promoted cell migration and invasion. TMEM16A inhibition may be a novel strategy for treating breast cancer metastasis.
Collapse
Key Words
- Cl− channel
- EGFR, epidermal growth factor receptor
- ER, estrogen receptor
- FBS, fetal bovine serum
- H&E, hematoxylin and eosin
- HNSCC, head and neck squamous cell carcinoma
- IHC, immunohistochemical
- MFS, metastasis-free survival
- Metastasis
- Moesin
- OS, overall survival
- PR, progesterone receptor
- ROCK1
- ROCK1, Rho-associated, coiled-coil containing protein kinase 1
- STAT3, signal transducers and activators of transcription 3
- TCGA, The Cancer Genome Atlas
- TMEM16A
- shRNAs, small hairpin RNAs
Collapse
Affiliation(s)
- Shuya Luo
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Hui Wang
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Lichuan Bai
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Yiwen Chen
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Si Chen
- Department of Microbial and Biochemical Pharmacy, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Kuan Gao
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Huijie Wang
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Shuwei Wu
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Hanbin Song
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Ke Ma
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Mei Liu
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Fan Yao
- Department of Breast Surgery and Surgical Oncology, Research Unit of General Surgery, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Yue Fang
- Department of Microbial and Biochemical Pharmacy, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Qinghuan Xiao
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China
| |
Collapse
|
9
|
Platelets and extracellular vesicles and their cross talk with cancer. Blood 2021; 137:3192-3200. [PMID: 33940593 DOI: 10.1182/blood.2019004119] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 11/16/2020] [Indexed: 02/06/2023] Open
Abstract
Platelets play significant and varied roles in cancer progression, as detailed throughout this review series, via direct interactions with cancer cells and by long-range indirect interactions mediated by platelet releasates. Microvesicles (MVs; also referred to as microparticles) released from activated platelets have emerged as major contributors to the platelet-cancer nexus. Interactions of platelet-derived MVs (PMVs) with cancer cells can promote disease progression through multiple mechanisms, but PMVs also harbor antitumor functions. This complex relationship derives from PMVs' binding to both cancer cells and nontransformed cells in the tumor microenvironment and transferring platelet-derived contents to the target cell, each of which can have stimulatory or modulatory effects. MVs are extracellular vesicles of heterogeneous size, ranging from 100 nm to 1 µm in diameter, shed by living cells during the outward budding of the plasma membrane, entrapping local cytosolic contents in an apparently stochastic manner. Hence, PMVs are encapsulated by a lipid bilayer harboring surface proteins and lipids mirroring the platelet exterior, with internal components including platelet-derived mature messenger RNAs, pre-mRNAs, microRNAs, and other noncoding RNAs, proteins, second messengers, and mitochondria. Each of these elements engages in established and putative PMV functions in cancer. In addition, PMVs contribute to cancer comorbidities because of their roles in coagulation and thrombosis and via interactions with inflammatory cells. However, separating the effects of PMVs from those of platelets in cancer contexts continues to be a major hurdle. This review summarizes our emerging understanding of the complex roles of PMVs in the development and progression of cancer and cancer comorbidities.
Collapse
|
10
|
Chu Y, Guo H, Zhang Y, Qiao R. Procoagulant platelets: Generation, characteristics, and therapeutic target. J Clin Lab Anal 2021; 35:e23750. [PMID: 33709517 PMCID: PMC8128296 DOI: 10.1002/jcla.23750] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 02/17/2021] [Accepted: 02/20/2021] [Indexed: 12/19/2022] Open
Abstract
Platelets play a pivotal role in hemostasis. Activated platelets are classified into two groups, according to their agonist response: aggregating and procoagulant platelets. Aggregating platelets consist of activated integrin αIIbβ3 and stretch out pseudopods to further attract platelets to the site of injury by connecting with fibrinogen. They mainly gather in the core of the thrombus and perform a secretory function, such as releasing adenosine diphosphate (ADP). Procoagulant platelets promote the formation of thrombin and fibrin by interacting with coagulation factors and can thus be considered as the connector between primary and secondary hemostasis. In addition to their functions in blood coagulation, procoagulant platelets play a proinflammatory role by releasing platelet microparticles and inorganic polyphosphate. Considering these important functions of procoagulant platelets, this subpopulation warrants detailed study to analyze their potential in preventing human diseases. This review summarizes the generation and important characteristics of procoagulant platelets, as well as their potential for preventing the adverse effects associated with current antiplatelet therapies.
Collapse
Affiliation(s)
- Yaxin Chu
- The Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
| | - Han Guo
- The Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
| | - Yuncong Zhang
- The Department of Laboratory Medicine, Peking University International Hospital, Beijing, China
| | - Rui Qiao
- The Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
| |
Collapse
|
11
|
Grigoriev VV. [Calcium-activated chloride channels: structure, properties, role in physiological and pathological processes]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2021; 67:17-33. [PMID: 33645519 DOI: 10.18097/pbmc20216701017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Ca2+-activated chloride channels (CaCC) are a class of intracellular calcium activated chloride channels that mediate numerous physiological functions. In 2008, the molecular structure of CaCC was determined. CaCC are formed by the protein known as anoctamine 1 (ANO1 or TMEM16A). CaCC mediates the secretion of Cl- in secretory epithelia, such as the airways, salivary glands, intestines, renal tubules, and sweat glands. The presence of CaCC has also been recognized in the vascular muscles, smooth muscles of the respiratory tract, which control vascular tone and hypersensitivity of the respiratory tract. TMEM16A is activated in many cancers; it is believed that TMEM16A is involved in carcinogenesis. TMEM16A is also involved in cancer cells proliferation. The role of TMEM16A in the mechanisms of hypertension, asthma, cystic fibrosis, nociception, and dysfunction of the gastrointestinal tract has been determined. In addition to TMEM16A, its isoforms are involved in other physiological and pathophysiological processes. TMEM16B (or ANO2) is involved in the sense of smell, while ANO6 works like scramblase, and its mutation causes a rare bleeding disorder, known as Scott syndrome. ANO5 is associated with muscle and bone diseases. TMEM16A interacts with various cellular signaling pathways including: epidermal growth factor receptor (EGFR), mitogen-activated protein kinases (MAPK), calmodulin (CaM) kinases, transforming growth factor TGF-β. The review summarizes existing information on known natural and synthetic compounds that can block/modulate CaCC currents and their effect on some pathologies in which CaCC is involved.
Collapse
Affiliation(s)
- V V Grigoriev
- Institute of Physiologically Active Compounds of the Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
12
|
Chen W, Gu M, Gao C, Chen B, Yang J, Xie X, Wang X, Sun J, Wang J. The Prognostic Value and Mechanisms of TMEM16A in Human Cancer. Front Mol Biosci 2021; 8:542156. [PMID: 33681289 PMCID: PMC7930745 DOI: 10.3389/fmolb.2021.542156] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 01/11/2021] [Indexed: 12/24/2022] Open
Abstract
As a calcium ion-dependent chloride channel transmembrane protein 16A (TMEM16A) locates on the cell membrane. Numerous research results have shown that TMEM16A is abnormally expressed in many cancers. Mechanically, TMEM16A participates in cancer proliferation and migration by affecting the MAPK and CAMK signaling pathways. Additionally, it is well documented that TMEM16A exerts a regulative impact on the hyperplasia of cancer cells by interacting with EGFR in head and neck squamous cell carcinoma (HNSCC), an epithelial growth factor receptor in head and neck squamous cell carcinoma respectively. Meanwhile, as an EGFR activator, TMEM16A is considered as an oncogene or a tumor-promoting factor. More and more experimental data showed that down-regulation of TMEM16A or gene targeted therapy may be an effective treatment for cancer. This review summarized its role in various cancers and research advances related to its clinical application included treatment and diagnosis.
Collapse
Affiliation(s)
- Wenjian Chen
- Anhui Province Children's Hospital Affiliated to Anhui Medical University, Hefei, China.,School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, China
| | - Meng Gu
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, China
| | - Chaobing Gao
- Department of Otorhinolaryngology, Head and Neck Surgery, The First Affiliated Hospital of AnHui Medical University, Hefei, China
| | - Bangjie Chen
- First Clinical Medical College of Anhui Medical University, Hefei, China
| | - Junfa Yang
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, China
| | - Xiaoli Xie
- Anhui Medicine Centralized Procurement Service Center, Hefei, China
| | - Xinyi Wang
- First Clinical Medical College of Anhui Medical University, Hefei, China
| | - Jun Sun
- Anhui Province Children's Hospital Affiliated to Anhui Medical University, Hefei, China
| | - Jinian Wang
- Department of Neurosurgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
13
|
Liang P, Yang H. Molecular underpinning of intracellular pH regulation on TMEM16F. J Gen Physiol 2021; 153:e202012704. [PMID: 33346788 PMCID: PMC7754671 DOI: 10.1085/jgp.202012704] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 10/29/2020] [Accepted: 11/24/2020] [Indexed: 12/14/2022] Open
Abstract
TMEM16F, a dual-function phospholipid scramblase and ion channel, is important in blood coagulation, skeleton development, HIV infection, and cell fusion. Despite advances in understanding its structure and activation mechanism, how TMEM16F is regulated by intracellular factors remains largely elusive. Here we report that TMEM16F lipid scrambling and ion channel activities are strongly influenced by intracellular pH (pHi). We found that low pHi attenuates, whereas high pHi potentiates, TMEM16F channel and scramblase activation under physiological concentrations of intracellular Ca2+ ([Ca2+]i). We further demonstrate that TMEM16F pHi sensitivity depends on [Ca2+]i and exhibits a bell-shaped relationship with [Ca2+]i: TMEM16F channel activation becomes increasingly pHi sensitive from resting [Ca2+]i to micromolar [Ca2+]i, but when [Ca2+]i increases beyond 15 µM, pHi sensitivity gradually diminishes. The mutation of a Ca2+-binding residue that markedly reduces TMEM16F Ca2+ sensitivity (E667Q) maintains the bell-shaped relationship between pHi sensitivity and Ca2+ but causes a dramatic shift of the peak [Ca2+]i from 15 µM to 3 mM. Our biophysical characterizations thus pinpoint that the pHi regulatory effects on TMEM16F stem from the competition between Ca2+ and protons for the primary Ca2+-binding residues in the pore. Within the physiological [Ca2+]i range, the protonation state of the primary Ca2+-binding sites influences Ca2+ binding and regulates TMEM16F activation. Our findings thus uncover a regulatory mechanism of TMEM16F by pHi and shine light on our understanding of the pathophysiological roles of TMEM16F in diseases with dysregulated pHi, including cancer.
Collapse
Affiliation(s)
- Pengfei Liang
- Department of Biochemistry, Duke University Medical Center, Durham, NC
| | - Huanghe Yang
- Department of Biochemistry, Duke University Medical Center, Durham, NC
- Department of Neurobiology, Duke University Medical Center, Durham, NC
| |
Collapse
|
14
|
Crottès D, Jan LY. The multifaceted role of TMEM16A in cancer. Cell Calcium 2019; 82:102050. [PMID: 31279157 PMCID: PMC6711484 DOI: 10.1016/j.ceca.2019.06.004] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 06/13/2019] [Accepted: 06/13/2019] [Indexed: 12/30/2022]
Abstract
The calcium-activated chloride channel TMEM16A is intimately linked to cancers. Over decades, TMEM16A over-expression and contribution to prognosis have been widely studied for multiple cancers strengthening the idea that TMEM16A could be a valuable biomarker and a promising therapeutic target. Surprisingly, from the survey of the literature, it appears that TMEM16A has been involved in multiple cancer-related functions and a large number of molecular targets of TMEM16A have been proposed. Thus, TMEM16A appears to be an ion channel with a multifaceted role in cancers. In this review, we summarize the latest development regarding TMEM16A contribution to cancers. We will survey TMEM16A contribution in cancer prognosis, the origins of its over-expression in cancer cells, the multiple biological functions and molecular pathways regulated by TMEM16A. Then, we will consider the question regarding the molecular mechanism of TMEM16A in cancers and the possible basis for the multifaceted role of TMEM16A in cancers.
Collapse
Affiliation(s)
- David Crottès
- Departments of Physiology, Biochemistry, and Biophysics, Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Lily Yeh Jan
- Departments of Physiology, Biochemistry, and Biophysics, Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA, 94143, USA.
| |
Collapse
|
15
|
Xuan ZB, Wang YJ, Xie J. ANO6 promotes cell proliferation and invasion in glioma through regulating the ERK signaling pathway. Onco Targets Ther 2019; 12:6721-6731. [PMID: 31692479 PMCID: PMC6708391 DOI: 10.2147/ott.s211725] [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] [Received: 04/10/2019] [Accepted: 06/11/2019] [Indexed: 12/19/2022] Open
Abstract
Purpose Anoctamin6 (ANO6) plays a crucial role in several cancers, whereas the specific role of ANO6 in glioblastoma is unclear. Methods Kaplan-Meier survival analysis was used to analysis the correlation between ANO6 and survival rate of patients with glioblastoma. Univariate Cox regression analysis was used to analysis the correlation among ANO6 expression level,and age, gender, WHO and overall survival rate. Immunohistocemical technique, RT-PCR and western blot were used to dected the ANO6 expression. CCK8, colony formation and transwell were used to detected cell viability, cell proliferation and cell invasion in glioblastoma cells transfected with sh-ANO6 and ANO6 overexpression. In addition, after SHG-44 cells trasfected with ANO6 overexpression were ERK inhibitor (PD98059), CCK8, colony formation and transwell were used to detected cell viability, cell proliferation and cell invasion. Western blot was used to detected ERK protein level and the phosphorylation level of ERK in T89G and U87MG cells tranfected wih sh-ANO6. Results The results indicated that the ANO6 expression level was significantly associated with patients' age and tumor stage. Univariate Cox regression analysis showed that the ANO6 expression level, age, gender and tumor stage were not related to the overall survival rate. ANO6 inhibition significantly suppressed the viability, invasion and the ability of colony formation in glioma cells, while ANO6 overexpression led to the opposite results in SHG-44 cells. ANO6 knockdown strongly inhibits the phosphorylation level and nuclear translocation of extracellular signal-regulated kinase (ERK) protein to inhibit ERK signaling. ERK inhibitor significantly decreased the cell proliferation and invasion in SHG-44 cells transfected with sh-ANO6. Conclusion This study revealed that ANO6 activited ERK signaling pathway through promoting the nuclear translocation of ERK to increase the proliferation and invasion of glioblastoma cells.
Collapse
Affiliation(s)
- Zhao-Bo Xuan
- Department of Neurosurgery, The First Hospital Affiliated to Jiamusi University, Jiamusi City, Heilongjiang Province 154002, People's Republic of China
| | - Ye-Ji Wang
- Department of Neurosurgery, Shanxian Haijiya Hospital, Heze City, Shandong Province 274300, People's Republic of China
| | - Jun Xie
- Department of Neurosurgery, Tongchuan People's Hospital, Tongchuan City, Shaanxi Province 727000, People's Republic of China
| |
Collapse
|
16
|
Morishita K, Watanabe K, Ichijo H. Cell volume regulation in cancer cell migration driven by osmotic water flow. Cancer Sci 2019; 110:2337-2347. [PMID: 31120184 PMCID: PMC6676112 DOI: 10.1111/cas.14079] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 05/17/2019] [Accepted: 05/21/2019] [Indexed: 12/12/2022] Open
Abstract
Cancer metastasis is the most frequent cause of death for patients with cancer. The main current treatment for cancer metastasis is chemotherapy targeting cancer cells’ ability to proliferate. However, some types of cancer cells show resistance to chemotherapy. Recently, cancer cell migration has become the subject of interest as a novel target of cancer therapy. Cell migration requires many factors, such as the cytoskeleton, cell‐matrix adhesion and cell volume regulation. Here, we focus on cell volume regulation and the role of ion/water transport systems in cell migration. Transport proteins, such as ion channels, ion carriers, and aquaporins, are indispensable for cell volume regulation under steady‐state conditions and during exposure to osmotic stress. Studies from the last ~25 years have revealed that cell volume regulation also plays an important role in the process of cell migration. Water flow in accordance with localized osmotic gradients generated by ion transport contributes to the driving force for cell migration. Moreover, it has been reported that metastatic cancer cells have higher expression of these transport proteins than nonmetastatic cancer cells. Thus, ion/water transport proteins involved in cell volume regulation and cell migration could be novel therapeutic targets for cancer metastasis. In this review, after presenting the importance of ion/water transport systems in cell volume regulation, we discuss the roles of transport proteins in a pathophysiological context, especially in the context of cancer cell migration.
Collapse
Affiliation(s)
- Kazuhiro Morishita
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Kengo Watanabe
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Hidenori Ichijo
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
17
|
Zeng X, Pan D, Wu H, Chen H, Yuan W, Zhou J, Shen Z, Chen S. Transcriptional activation of ANO1 promotes gastric cancer progression. Biochem Biophys Res Commun 2019; 512:131-136. [PMID: 30871776 DOI: 10.1016/j.bbrc.2019.03.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 03/01/2019] [Indexed: 02/06/2023]
Abstract
The prognosis of gastric cancer (GC) remains poor due to local invasion and distal metastasis. The GC-related molecular mechanisms underlying invasion and metastasis are not well understood. In this study, we investigated the functional role of ANO1 in GC progression. We found that ANO1 is overexpressed in GC tissues and correlated with GC tumor-node-metastasis stage. Knockdown of ANO1 significantly inhibited GC cell migration and invasion in vitro, and loss of ANO1 resulted in inhibition of tumor metastasis in vivo. Mechanistically, SP1 increased ANO1 transcription, recruited MLL1 to the ANO1 promoter region, facilitated H3K4 trimethylation, and subsequently promoted ANO1 expression. Together, our findings provide a mechanistic assessment of ANO1 overexpression, which represents a GC progression-related molecule and a potentially valuable target for future research.
Collapse
Affiliation(s)
- Xiaoqing Zeng
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, PR China
| | - Duyi Pan
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, PR China
| | - Hao Wu
- Key Laboratory of Glycoconjugate Research Ministry of Public Health, School of Basic Medical Sciences, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, PR China
| | - Hao Chen
- Department of General Surgery, Zhongshan Hospital of Fudan University, Shanghai, 200032, PR China
| | - Wei Yuan
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, PR China
| | - Ji Zhou
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, PR China
| | - Zhenbin Shen
- Department of General Surgery, Zhongshan Hospital of Fudan University, Shanghai, 200032, PR China.
| | - Shiyao Chen
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, PR China; Endoscopy Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, PR China.
| |
Collapse
|
18
|
Pethő Z, Najder K, Bulk E, Schwab A. Mechanosensitive ion channels push cancer progression. Cell Calcium 2019; 80:79-90. [PMID: 30991298 DOI: 10.1016/j.ceca.2019.03.007] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 03/26/2019] [Accepted: 03/26/2019] [Indexed: 02/07/2023]
Abstract
In many cases, the mechanical properties of a tumor are different from those of the host tissue. Mechanical cues regulate cancer development by affecting both tumor cells and their microenvironment, by altering cell migration, proliferation, extracellular matrix remodeling and metastatic spread. Cancer cells sense mechanical stimuli such as tissue stiffness, shear stress, tissue pressure of the extracellular space (outside-in mechanosensation). These mechanical cues are transduced into a cellular response (e. g. cell migration and proliferation; inside-in mechanotransduction) or to a response affecting the microenvironment (e. g. inducing a fibrosis or building up growth-induced pressure; inside-out mechanotransduction). These processes heavily rely on mechanosensitive membrane proteins, prominently ion channels. Mechanosensitive ion channels are involved in the Ca2+-signaling of the tumor and stroma cells, both directly, by mediating Ca2+ influx (e. g. Piezo and TRP channels), or indirectly, by maintaining the electrochemical gradient necessary for Ca2+ influx (e. g. K2P, KCa channels). This review aims to discuss the diverse roles of mechanosenstive ion channels in cancer progression, especially those involved in Ca2+-signaling, by pinpointing their functional relevance in tumor pathophysiology.
Collapse
Affiliation(s)
- Zoltán Pethő
- Institut für Physiologie II, Robert-Koch-Str. 27b, 48149 Münster, Germany.
| | - Karolina Najder
- Institut für Physiologie II, Robert-Koch-Str. 27b, 48149 Münster, Germany
| | - Etmar Bulk
- Institut für Physiologie II, Robert-Koch-Str. 27b, 48149 Münster, Germany
| | - Albrecht Schwab
- Institut für Physiologie II, Robert-Koch-Str. 27b, 48149 Münster, Germany
| |
Collapse
|
19
|
Kunzelmann K, Ousingsawat J, Benedetto R, Cabrita I, Schreiber R. Contribution of Anoctamins to Cell Survival and Cell Death. Cancers (Basel) 2019; 11:E382. [PMID: 30893776 PMCID: PMC6468699 DOI: 10.3390/cancers11030382] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 03/13/2019] [Accepted: 03/16/2019] [Indexed: 02/07/2023] Open
Abstract
Before anoctamins (TMEM16 proteins) were identified as a family of Ca2+-activated chloride channels and phospholipid scramblases, the founding member anoctamin 1 (ANO1, TMEM16A) was known as DOG1, a marker protein for gastrointestinal stromal tumors (GIST). Meanwhile, ANO1 has been examined in more detail, and the role of ANO1 in cell proliferation and the development of different types of malignomas is now well established. While ANO5, ANO7, and ANO9 may also be relevant for growth of cancers, evidence has been provided for a role of ANO6 (TMEM16F) in regulated cell death. The cellular mechanisms by which anoctamins control cell proliferation and cell death, respectively, are just emerging; however, the pronounced effects of anoctamins on intracellular Ca2+ levels are likely to play a significant role. Recent results suggest that some anoctamins control membrane exocytosis by setting Ca2+i levels near the plasma membrane, and/or by controlling the intracellular Cl- concentration. Exocytosis and increased membrane trafficking induced by ANO1 and ANO6 may enhance membrane expression of other chloride channels, such as CFTR and volume activated chloride channels (VRAC). Notably, ANO6-induced phospholipid scrambling with exposure of phosphatidylserine is pivotal for the sheddase function of disintegrin and metalloproteinase (ADAM). This may support cell death and tumorigenic activity of IL-6 by inducing IL-6 trans-signaling. The reported anticancer effects of the anthelminthic drug niclosamide are probably related to the potent inhibitory effect on ANO1, apart from inducing cell cycle arrest through the Let-7d/CDC34 axis. On the contrary, pronounced activation of ANO6 due to a large increase in intracellular calcium, activation of phospholipase A2 or lipid peroxidation, can lead to ferroptotic death of cancer cells. It therefore appears reasonable to search for both inhibitors and potent activators of TMEM16 in order to interfere with cancer growth and metastasis.
Collapse
Affiliation(s)
- Karl Kunzelmann
- Institut für Physiologie, Universität Regensburg, Universitätsstraße 31, D-93053 Regensburg, Germany.
| | - Jiraporn Ousingsawat
- Institut für Physiologie, Universität Regensburg, Universitätsstraße 31, D-93053 Regensburg, Germany.
| | - Roberta Benedetto
- Institut für Physiologie, Universität Regensburg, Universitätsstraße 31, D-93053 Regensburg, Germany.
| | - Ines Cabrita
- Institut für Physiologie, Universität Regensburg, Universitätsstraße 31, D-93053 Regensburg, Germany.
| | - Rainer Schreiber
- Institut für Physiologie, Universität Regensburg, Universitätsstraße 31, D-93053 Regensburg, Germany.
| |
Collapse
|
20
|
Ji Q, Guo S, Wang X, Pang C, Zhan Y, Chen Y, An H. Recent advances in TMEM16A: Structure, function, and disease. J Cell Physiol 2018; 234:7856-7873. [PMID: 30515811 DOI: 10.1002/jcp.27865] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 11/13/2018] [Indexed: 12/13/2022]
Abstract
TMEM16A (also known as anoctamin 1, ANO1) is the molecular basis of the calcium-activated chloride channels, with ten transmembrane segments. Recently, atomic structures of the transmembrane domains of mouse TMEM16A (mTMEM16A) were determined by single-particle electron cryomicroscopy. This gives us a solid ground to discuss the electrophysiological properties and functions of TMEM16A. TMEM16A is reported to be dually regulated by Ca2+ and voltage. In addition, the dysfunction of TMEM16A has been found to be involved in many diseases including cystic fibrosis, various cancers, hypertension, and gastrointestinal motility disorders. TMEM16A is overexpressed in many cancers, including gastrointestinal stromal tumors, gastric cancer, head and neck squamous cell carcinoma (HNSCC), colon cancer, pancreatic ductal adenocarcinoma, and esophageal cancer. Furthermore, overexpression of TMEM16A is related to the occurrence, proliferation, and migration of tumor cells. To date, several studies have shown that many natural compounds and synthetic compounds have regulatory effects on TMEM16A. These small molecule compounds might be novel drugs for the treatment of diseases caused by TMEM16A dysfunction in the future. In addition, recent studies have shown that TMEM16A plays different roles in different diseases through different signal transduction pathways. This review discusses the topology, electrophysiological properties, modulators and functions of TMEM16A in mediates nociception, gastrointestinal dysfunction, hypertension, and cancer and focuses on multiple regulatory mechanisms regarding TMEM16A.
Collapse
Affiliation(s)
- Qiushuang Ji
- Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Sciences, Hebei University of Technology, Tianjin, China
| | - Shuai Guo
- Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Sciences, Hebei University of Technology, Tianjin, China
| | - Xuzhao Wang
- Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Sciences, Hebei University of Technology, Tianjin, China
| | - Chunli Pang
- Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Sciences, Hebei University of Technology, Tianjin, China
| | - Yong Zhan
- Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Sciences, Hebei University of Technology, Tianjin, China
| | - Yafei Chen
- Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Sciences, Hebei University of Technology, Tianjin, China
| | - Hailong An
- Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Sciences, Hebei University of Technology, Tianjin, China
| |
Collapse
|
21
|
Guo R, Huang X, Jin X, Yang J. [Diltiazem inhibits proliferation and motility of hepatocellular cells in vitro by downregulating calcium-activated chloride channel TMEM16A]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2018; 38:818-823. [PMID: 33168514 DOI: 10.3969/j.issn.1673-4254.2018.07.08] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
OBJECTIVE To assess the inhibitory effect of diltiazem, a calcium channel inhibitor, on the proliferation and mobility of human hepatocellular carcinoma cells in vitro and explore the possible mechanism. METHODS Two human hepatocellular carcinoma cell lines, MHCC97H and 7402, were treated with different concentrations (0-400 μmol/L) of diltiazem for 12, 24, or 48 h, and the changes in the cell proliferation and mobility were observed with MTT assay and wound healing assay, respectively. The changes in the expressions of calcium-activated chloride channel TMEM16A at mRNA and protein levels in the treated cells were detected using RT-PCR and immunocytochemistry. RESULTS Treatment with diltiazem obviously inhibited the proliferation and suppressed the mobility of MHCC97H and 7402 cells in a time- and concentration-dependent manner (P < 0.05). Treatment with 100 μmol/L diltiazem for 24 h significantly inhibited the proliferation of MHCC97H cells and down-regulated the mRNA and protein levels of TMEM16A. In 7402 cells, diltiazem treatment at 50 μmol/L for 48 h resulted in the most significant inhibitory effect on the cell proliferation and TMEM16A expressions. CONCLUSIONS Diltiazem can transiently inhibit the invasion of hepatocellular carcinoma cells in vitro possibly by down-regulating the expression of TMEM16A at both the mRNA and protein levels.
Collapse
Affiliation(s)
- Rui Guo
- Department of Pathology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Xiaozhong Huang
- Department of Pathology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Xueyuan Jin
- Department of Pathology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Jun Yang
- Department of Pathology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| |
Collapse
|
22
|
Stewart SE, Ashkenazi A, Williamson A, Rubinsztein DC, Moreau K. Transbilayer phospholipid movement facilitates the translocation of annexin across membranes. J Cell Sci 2018; 131:jcs217034. [PMID: 29930080 PMCID: PMC6080606 DOI: 10.1242/jcs.217034] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 06/06/2018] [Indexed: 02/03/2023] Open
Abstract
Annexins are cytosolic phospholipid-binding proteins that can be found on the outer leaflet of the plasma membrane. The extracellular functions of annexin include modulating fibrinolysis activity and cell migration. Despite having well-described extracellular functions, the mechanism of annexin transport from the cytoplasmic inner leaflet to the extracellular outer leaflet of the plasma membrane remains unclear. Here, we show that the transbilayer movement of phospholipids facilitates the transport of annexins A2 and A5 across membranes in cells and in liposomes. We identified TMEM16F (also known as anoctamin-6, ANO6) as a lipid scramblase required for transport of these annexins to the outer leaflet of the plasma membrane. This work reveals a mechanism for annexin translocation across membranes which depends on plasma membrane phospholipid remodelling.
Collapse
Affiliation(s)
- Sarah E Stewart
- University of Cambridge, Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, Cambridge, CB2 0QQ, UK
| | - Avraham Ashkenazi
- University of Cambridge, Department of Medical Genetics, Cambridge Institute for Medical Research, Wellcome/MRC Building, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0XY, UK
| | - Athena Williamson
- University of Cambridge, Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, Cambridge, CB2 0QQ, UK
| | - David C Rubinsztein
- University of Cambridge, Department of Medical Genetics, Cambridge Institute for Medical Research, Wellcome/MRC Building, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0XY, UK
- UK Dementia Research Institute, Cambridge Biomedical Campus, Hills Road, Cambridge, UK
| | - Kevin Moreau
- University of Cambridge, Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, Cambridge, CB2 0QQ, UK
| |
Collapse
|
23
|
Wang L, Zhao XY, Zhu JS, Chen NW, Fan HN, Yang W, Guo JH. CCR7 regulates ANO6 to promote migration of pancreatic ductal adenocarcinoma cells via the ERK signaling pathway. Oncol Lett 2018; 16:2599-2605. [PMID: 30013654 DOI: 10.3892/ol.2018.8962] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 11/11/2017] [Indexed: 12/26/2022] Open
Abstract
The increase in migratory ability of pancreatic ductal adenocarcinoma cells is a key event in the development of metastasis to the lymph nodes and distant organs. Although the C-C motif chemokine receptor 7 (CCR7) and its ligand, C-C motif chemokine ligand 21 (CCL21), have been revealed to serve an important role in tumor migration, their precise roles and potential underlying mechanisms remain largely unknown. The present study revealed that overexpression of CCR7 significantly promoted BxPC-3 cell migration, accompanied by the induction of anoctamin 6 (ANO6) expression, indicating that ANO6 is a downstream target of CCR7 signaling. Furthermore, the level of phosphorylated extracellular signal-regulated kinase (ERK) was significantly increased in CCR7-overexpressing BxPC-3 cells, indicating that ERK may be a potential mediator of CCR7-regulated ANO6 expression in BxPC-3 cells. To characterize the receptor-mediated pathway, a specific ERK inhibitor, U0126, was used, which reduced BxPC-3 cell migration and the expression of ANO6. In summary, the results of the present study demonstrate that CCR7 promoted BxPC-3 cell migration by regulating ANO6 expression perhaps via activation of the ERK signaling pathway.
Collapse
Affiliation(s)
- Long Wang
- Department of Gastroenterology, Shanghai Jiao Tong University Affiliated Shanghai Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Xiang-Yun Zhao
- Department of Gastroenterology, Shanghai Jiao Tong University Affiliated Shanghai Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Jin-Shui Zhu
- Department of Gastroenterology, Shanghai Jiao Tong University Affiliated Shanghai Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Ni-Wei Chen
- Department of Gastroenterology, Shanghai Jiao Tong University Affiliated Shanghai Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Hui-Ning Fan
- Department of Gastroenterology, Shanghai Jiao Tong University Affiliated Shanghai Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Wei Yang
- Department of Laboratory, Shanghai Jiao Tong University Affiliated Shanghai Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Jing-Hui Guo
- Department of Gastroenterology, Shanghai Jiao Tong University Affiliated Shanghai Sixth People's Hospital, Shanghai 200233, P.R. China
| |
Collapse
|
24
|
Wang H, Zou L, Ma K, Yu J, Wu H, Wei M, Xiao Q. Cell-specific mechanisms of TMEM16A Ca 2+-activated chloride channel in cancer. Mol Cancer 2017; 16:152. [PMID: 28893247 PMCID: PMC5594453 DOI: 10.1186/s12943-017-0720-x] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 09/01/2017] [Indexed: 02/08/2023] Open
Abstract
TMEM16A (known as anoctamin 1) Ca2+-activated chloride channel is overexpressed in many tumors. TMEM16A overexpression can be caused by gene amplification in many tumors harboring 11q13 amplification. TMEM16A expression is also controlled in many cancer cells via transcriptional regulation, epigenetic regulation and microRNAs. In addition, TMEM16A activates different signaling pathways in different cancers, e.g. the EGFR and CAMKII signaling in breast cancer, the p38 and ERK1/2 signaling in hepatoma, the Ras-Raf-MEK-ERK1/2 signaling in head and neck squamous cell carcinoma and bladder cancer, and the NFκB signaling in glioma. Furthermore, TMEM16A overexpression has been reported to promote, inhibit, or produce no effects on cell proliferation and migration in different cancer cells. Since TMEM16A exerts different roles in different cancer cells via activation of distinct signaling pathways, we try to develop the idea that TMEM16A regulates cancer cell proliferation and migration in a cell-dependent mechanism. The cell-specific role of TMEM16A may depend on the cellular environment that is predetermined by TMEM16A overexpression mechanisms specific for a particular cancer type. TMEM16A may exert its cell-specific role via its associated protein networks, phosphorylation by different kinases, and involvement of different signaling pathways. In addition, we discuss the role of TMEM16A channel activity in cancer, and its clinical use as a prognostic and predictive marker in different cancers. This review highlights the cell-type specific mechanisms of TMEM16A in cancer, and envisions the promising use of TMEM16A inhibitors as a potential treatment for TMEM16A-overexpressing cancers.
Collapse
Affiliation(s)
- Hui Wang
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110122 China
| | - Liang Zou
- Department of Anesthesiology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021 China
| | - Ke Ma
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110122 China
| | - Jiankun Yu
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110122 China
| | - Huizhe Wu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122 China
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122 China
| | - Qinghuan Xiao
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110122 China
| |
Collapse
|
25
|
Chang Z, Cai C, Han D, Gao Y, Li Q, Feng L, Zhang W, Zheng J, Jin J, Zhang H, Wei Q. Anoctamin5 regulates cell migration and invasion in thyroid cancer. Int J Oncol 2017; 51:1311-1319. [PMID: 28902351 DOI: 10.3892/ijo.2017.4113] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 08/25/2017] [Indexed: 11/05/2022] Open
Abstract
Anoctamin/TMEM16 family members have recently been identified as novel calcium-activated chloride channels, and dysregulation of many family members participates in tumorigenesis and progression. However, the exact role of anoctamin5 (ANO5), one member of this family, in thyroid cancer is still not clarified. In this study, we firstly found that the expression levels of ANO5 was significantly downregulated in thyroid cancer compared to adjacent normal tissue by mining the public GEO database. Subsequently, we further demonstrated that the expression levels of ANO5 was significantly downregulated in 69.5% (57/82) clinical thyroid cancer tissues using real-time PCR assay. Moreover, western blot assay also showed that ANO5 was downregulated in papillary thyroid cancer and follicular thyroid cancer compared to adjacent noncancerous tissues. Furthermore, some biological and functional in vitro experiments proved that ANO5 knockdown promotes thyroid cancer cell migration and invasion but overexpression of ANO5 inhibits these phenotypes. By analyzing gene set enrichment, we found that lower ANO5 expression was positively associated with JAK/STAT3 signaling pathway. Collectively downregulation of ANO5 promotes thyroid cancer cell migration and invasion by affecting JAK/STAT3 pathway.
Collapse
Affiliation(s)
- Zhengyan Chang
- Department of Pathology, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, P.R. China
| | - Chunmiao Cai
- Department of Pathology, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, P.R. China
| | - Dongyan Han
- Department of Pathology, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, P.R. China
| | - Yaohui Gao
- Department of Pathology, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, P.R. China
| | - Qianyu Li
- Department of Pathology, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, P.R. China
| | - Lijin Feng
- Department of Pathology, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, P.R. China
| | - Wei Zhang
- Department of Pathology, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, P.R. China
| | - Jiayi Zheng
- Department of Pathology, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, P.R. China
| | - Jiaoying Jin
- Department of Pathology, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, P.R. China
| | - Huizhen Zhang
- Department of Pathology, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai 200233, P.R. China
| | - Qing Wei
- Department of Pathology, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, P.R. China
| |
Collapse
|
26
|
Murana E, Pagani F, Basilico B, Sundukova M, Batti L, Di Angelantonio S, Cortese B, Grimaldi A, Francioso A, Heppenstall P, Bregestovski P, Limatola C, Ragozzino D. ATP release during cell swelling activates a Ca 2+-dependent Cl - current by autocrine mechanism in mouse hippocampal microglia. Sci Rep 2017. [PMID: 28646166 PMCID: PMC5482828 DOI: 10.1038/s41598-017-04452-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Microglia cells, resident immune cells of the brain, survey brain parenchyma by dynamically extending and retracting their processes. Cl− channels, activated in the cellular response to stretch/swelling, take part in several functions deeply connected with microglia physiology, including cell shape changes, proliferation, differentiation and migration. However, the molecular identity and functional properties of these Cl− channels are largely unknown. We investigated the properties of swelling-activated currents in microglial from acute hippocampal slices of Cx3cr1+/GFP mice by whole-cell patch-clamp and imaging techniques. The exposure of cells to a mild hypotonic medium, caused an outward rectifying current, developing in 5–10 minutes and reverting upon stimulus washout. This current, required for microglia ability to extend processes towards a damage signal, was carried mainly by Cl− ions and dependent on intracellular Ca2+. Moreover, it involved swelling-induced ATP release. We identified a purine-dependent mechanism, likely constituting an amplification pathway of current activation: under hypotonic conditions, ATP release triggered the Ca2+-dependent activation of anionic channels by autocrine purine receptors stimulation. Our study on native microglia describes for the first time the functional properties of stretch/swelling-activated currents, representing a key element in microglia ability to monitor the brain parenchyma.
Collapse
Affiliation(s)
- E Murana
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | - F Pagani
- Istituto Italiano di Tecnologia, CLNS@Sapienza, Rome, Italy.
| | - B Basilico
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | - M Sundukova
- Mouse Biology Unit, EMBL, Monterotondo, Italy
| | - L Batti
- Mouse Biology Unit, EMBL, Monterotondo, Italy
| | - S Di Angelantonio
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy.,Istituto Italiano di Tecnologia, CLNS@Sapienza, Rome, Italy
| | - B Cortese
- CNR NANOTEC - Istituto di Nanotecnologia, Department of Physics, University Sapienza, Rome, Italy
| | - A Grimaldi
- Istituto Italiano di Tecnologia, CLNS@Sapienza, Rome, Italy
| | - A Francioso
- Department of Biochemistry, "Sapienza" University of Rome, Rome, Italy
| | | | - P Bregestovski
- Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes, Marseille, France
| | - C Limatola
- IRCCS Neuromed, Via Atinese, Pozzilli, Italy.,Pasteur Institute - Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | - D Ragozzino
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy.,IRCCS Neuromed, Via Atinese, Pozzilli, Italy
| |
Collapse
|
27
|
Cao Q, Liu F, Ji K, Liu N, He Y, Zhang W, Wang L. MicroRNA-381 inhibits the metastasis of gastric cancer by targeting TMEM16A expression. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2017; 36:29. [PMID: 28193228 PMCID: PMC5307754 DOI: 10.1186/s13046-017-0499-z] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 02/07/2017] [Indexed: 12/27/2022]
Abstract
Background MicroRNA-381 (miR-381) has been reported to play suppressive or promoting roles in different malignancies. However, the expression level, biological function, and underlying mechanisms of miR-381 in gastric cancer remain poorly understood. Our previous study indicated that transmembrane protein 16A (TMEM16A) contributed to migration and invasion of gastric cancer and predicted poor prognosis. In this study, we found that miR-381 inhibited the metastasis of gastric cancer through targeting TMEM16A expression. Methods MiR-381 expression was analyzed using bioinformatic software on open microarray datasets from the Gene Expression Omnibus (GEO) and confirmed by quantitative RT-PCR (qRT-PCR) in human gastric cancer tissues and cell lines. Cell proliferation was investigated using MTT and cell count assays, and cell migration and invasion abilities were evaluated by transwell assay. Xenograft nude mouse models were used to observe tumor growth and pulmonary metastasis. Luciferase reporter assay, western blot, enzyme-linked immunosorbent assay (ELISA) and immunohistochemistry were employed to explore the mechanisms of the effect of miR-381 on gastric cancer cells. Results MiR-381 was significantly down-regulated in gastric cancer tissues and cell lines. Low expression of miR-381 was negatively related to lymph node metastasis, advanced tumor stage and poor prognosis. MiR-381 decreased gastric cancer cell proliferation, migration and invasion in vitro and in vivo. TMEM16A was identified as a direct target of miR-381 and the expression of miR-381 was inversely correlated with TMEM16A expression in gastric cancer tissues. Combination analysis of miR-381 and TMEM16A revealed the improved prognostic accuracy for gastric cancer patients. Moreover, miR-381 inhibited TGF-β signaling pathway and down-regulated epithelial–mesenchymal transition (EMT) phenotype partially by mediating TMEM16A. Conclusions MiR-381 may function as a tumor suppressor by directly targeting TMEM16A and regulating TGF-β pathway and EMT process in the development of progression of gastric cancer. MiR-381/TMEM16A may be a novel therapeutic candidate target in gastric cancer treatment. Electronic supplementary material The online version of this article (doi:10.1186/s13046-017-0499-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Qinghua Cao
- Department of Pathology, The first affiliated hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Fang Liu
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Kaiyuan Ji
- Cancer Research Insitute, Southern Medical University, Guangzhou, 510515, China
| | - Ni Liu
- Department of Pathology, The first affiliated hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Yuan He
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine and Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Wenhui Zhang
- Department of Pathology, The first affiliated hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Liantang Wang
- Department of Pathology, The first affiliated hospital of Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|
28
|
KCa3.1 (IK) modulates pancreatic cancer cell migration, invasion and proliferation: anomalous effects on TRAM-34. Pflugers Arch 2016; 468:1865-1875. [DOI: 10.1007/s00424-016-1891-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 10/05/2016] [Accepted: 10/06/2016] [Indexed: 12/30/2022]
|
29
|
Abstract
Activation of ion channels and pores are essential steps during regulated cell death. Channels and pores participate in execution of apoptosis, necroptosis and other forms of caspase-independent cell death. Within the program of regulated cell death, these channels are strategically located. Ion channels can shrink cells and drive them towards apoptosis, resulting in silent, i.e. immunologically unrecognized cell death. Alternatively, activation of channels can induce cell swelling, disintegration of the cell membrane, and highly immunogenic necrotic cell death. The underlying cell death pathways are not strictly separated as identical stimuli may induce cell shrinkage and apoptosis when applied at low strength, but may also cause cell swelling at pronounced stimulation, resulting in regulated necrosis. Nevertheless, the precise role of ion channels during regulated cell death is far from being understood, as identical channels may support regulated death in some cell types, but may cause cell proliferation, cancer development, and metastasis in others. Along this line, the phospholipid scramblase and Cl(-)/nonselective channel anoctamin 6 (ANO6) shows interesting features, as it participates in apoptotic cell death during lower levels of activation, thereby inducing cell shrinkage. At strong activation, e.g. by stimulation of purinergic P2Y7 receptors, it participates in pore formation, causes massive membrane blebbing, cell swelling, and membrane disintegration. The LRRC8 proteins deserve much attention as they were found to have a major role in volume regulation, apoptotic cell shrinkage and resistance towards anticancer drugs.
Collapse
Affiliation(s)
- Karl Kunzelmann
- Institut für Physiologie, Universität Regensburg, Universitätsstraße 31, 93053, Regensburg, Germany.
| |
Collapse
|
30
|
Liu F, Cao QH, Lu DJ, Luo B, Lu XF, Luo RC, Wang XG. TMEM16A overexpression contributes to tumor invasion and poor prognosis of human gastric cancer through TGF-β signaling. Oncotarget 2016; 6:11585-99. [PMID: 25839162 PMCID: PMC4484478 DOI: 10.18632/oncotarget.3412] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 02/19/2015] [Indexed: 01/14/2023] Open
Abstract
TMEM16A is a newly identified calcium activated chloride channel, and has been reported to be overexpressed by various solid malignant cancers to promote proliferation and invasion, yet little is known about its role in gastric cancer(GC). Therefore, we investigated the role of TMEM16A in GC and its clinical significance by a retrospective analysis of 367 GC patients, and in vitro study was performed for validation and underlying molecular mechanism. TMEM16A was significantly upregulated and amplified in GC tissues, and its overexpression was positively correlated with disease stage, negatively with patient survival and identified as an independent prognostic factor for patient outcome. A negative correlation between TMEM16A and E-cadherin was found in 367 GC specimens. TMEM16A silencing significantly decreased calcium activated chloride currents, impaired TGF-β secretion, reduced E-cadherin expression, and inhibited the migration and invasion without affecting proliferation of GC cells (AGS and BGC-823). Supplement of TGF-β reverted the effects of TMEM16A silencing on E-cadherin expression, cell migration and invasion. In conclusion, TMEM16A promotes invasion and metastasis in GC, and might be a novel prognostic biomarker and potential therapeutic target in the treatment of GC.
Collapse
Affiliation(s)
- Fang Liu
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.,Faculty of Forensic Medicine, ZhongShan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China.,Cancer Center and Traditional Chinese Medicine-Integrated Hospital, Southern Medical University, Guangzhou 510315, China
| | - Qing-Hua Cao
- Department of Pathology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - De-Jian Lu
- Faculty of Forensic Medicine, ZhongShan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Bin Luo
- Faculty of Forensic Medicine, ZhongShan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Xiao-Fang Lu
- Department of Pathology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Rong-Cheng Luo
- Cancer Center and Traditional Chinese Medicine-Integrated Hospital, Southern Medical University, Guangzhou 510315, China
| | - Xiao-Guang Wang
- Faculty of Forensic Medicine, ZhongShan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| |
Collapse
|
31
|
Modulating Ca²⁺ signals: a common theme for TMEM16, Ist2, and TMC. Pflugers Arch 2015; 468:475-90. [PMID: 26700940 DOI: 10.1007/s00424-015-1767-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 11/24/2015] [Accepted: 11/26/2015] [Indexed: 12/21/2022]
Abstract
Since the discovery of TMEM16A (anoctamin 1, ANO1) as Ca(2+)-activated Cl(-) channel, the protein was found to serve different physiological functions, depending on the type of tissue. Subsequent reports on other members of the anoctamin family demonstrated a broad range of yet poorly understood properties. Compromised anoctamin function is causing a wide range of diseases, such as hearing loss (ANO2), bleeding disorder (ANO6), ataxia and dystonia (ANO3, 10), persistent borrelia and mycobacteria infection (ANO10), skeletal syndromes like gnathodiaphyseal dysplasia and limb girdle muscle dystrophy (ANO5), and cancer (ANO1, 6, 7). Animal models demonstrate CF-like airway disease, asthma, and intestinal hyposecretion (ANO1). Although present data indicate that ANO1 is a Ca(2+)-activated Cl(-) channel, it remains unclear whether all anoctamins form plasma membrane-localized or intracellular chloride channels. We find Ca(2+)-activated Cl(-) currents appearing by expression of most anoctamin paralogs, including the Nectria haematococca homologue nhTMEM16 and the yeast homologue Ist2. As recent studies show a role of anoctamins, Ist2, and the related transmembrane channel-like (TMC) proteins for intracellular Ca(2+) signaling, we will discuss the role of these proteins in generating compartmentalized Ca(2+) signals, which may give a hint as to the broad range of cellular functions of anoctamins.
Collapse
|
32
|
Sui Y, Wu F, Lv J, Li H, Li X, Du Z, Sun M, Zheng Y, Yang L, Zhong L, Zhang X, Zhang G. Identification of the Novel TMEM16A Inhibitor Dehydroandrographolide and Its Anticancer Activity on SW620 Cells. PLoS One 2015; 10:e0144715. [PMID: 26657333 PMCID: PMC4686118 DOI: 10.1371/journal.pone.0144715] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 11/23/2015] [Indexed: 11/18/2022] Open
Abstract
TMEM16A, a calcium-activated chloride channel (CaCC), is highly amplified and expressed in human cancers and is involved in the growth and metastasis of some malignancies. Inhibition of TMEM16A represents a novel pharmaceutical approach for the treatment of cancers and metastases. The purpose of this study is to identify a new TMEM16A inhibitor, investigate the effects of this inhibitor on the proliferation and metastasis of TMEM16A-amplified SW620 cells, and to elucidate the underlying molecular mechanism in vitro. We identified a novel small-molecule TMEM16A inhibitor dehydroandrographolide (DP). By using patch clamp electrophysiology, we showed that DP inhibited TMEM16A chloride currents in Fisher rat thyroid (FRT) cells that were transfected stably with human TMEM16A and in TMEM16A-overexpressed SW620 cells but did not alter cystic fibrosis transmembrane conductance regulator (CFTR) chloride currents. Further functional studies showed that DP suppressed the proliferation of SW620 cells in a dose- and time-dependent manner using MTT assays. Moreover, DP significantly inhibited migration and invasion of SW620 cells as detected by wound-healing and transwell assays. Further mechanistic study demonstrated that knockdown of human TMEM16A decreased the inhibitory effect of DP on the proliferation of SW620 cells and that TMEM16A-dependent cells (SW620 and HCT116) were more sensitive to DP than TMEM16A-independent cells (SW480 and HCT8). In addition, we found that treatment of SW620 cells with DP led to a decrease in TMEM16A protein levels but had no effect on TMEM16A mRNA levels. The current work reveals that DP, a novel TMEM16A inhibitor, exerts its anticancer activity on SW620 cells partly through a TMEM16A-dependent mechanism, which may introduce a new targeting approach for an antitumour therapy in TMEM16A-amplified cancers.
Collapse
Affiliation(s)
- Yujie Sui
- Key Laboratory for Molecular and Chemical Genetics of Critical Human Diseases of Jilin Province, Jilin University Bethune Second Hospital, Changchun, P. R. China
| | - Fei Wu
- Department of Gynecology and Obstetrics, Jilin University Bethune Second Hospital, Changchun, P. R. China
| | - Junfeng Lv
- Department of Radiology, Jilin University Bethune Second Hospital, Changchun, P. R. China
| | - Hongxia Li
- Department of Dermatology, Jilin University Bethune First Hospital, Changchun, P. R. China
| | - Xin Li
- Key Laboratory for Molecular and Chemical Genetics of Critical Human Diseases of Jilin Province, Jilin University Bethune Second Hospital, Changchun, P. R. China
| | - Zhenwu Du
- Key Laboratory for Molecular and Chemical Genetics of Critical Human Diseases of Jilin Province, Jilin University Bethune Second Hospital, Changchun, P. R. China
| | - Meiyan Sun
- Key Laboratory for Molecular and Chemical Genetics of Critical Human Diseases of Jilin Province, Jilin University Bethune Second Hospital, Changchun, P. R. China
| | - Yuhao Zheng
- Key Laboratory for Molecular and Chemical Genetics of Critical Human Diseases of Jilin Province, Jilin University Bethune Second Hospital, Changchun, P. R. China
| | - Longfei Yang
- Key Laboratory for Molecular and Chemical Genetics of Critical Human Diseases of Jilin Province, Jilin University Bethune Second Hospital, Changchun, P. R. China
| | - Lili Zhong
- Key Laboratory for Molecular and Chemical Genetics of Critical Human Diseases of Jilin Province, Jilin University Bethune Second Hospital, Changchun, P. R. China
| | - Xingyi Zhang
- Department of Thoracic Surgery, Jilin University Bethune Second Hospital, Changchun, P. R. China
- * E-mail: (XYZ), (GZZ)
| | - Guizhen Zhang
- Key Laboratory for Molecular and Chemical Genetics of Critical Human Diseases of Jilin Province, Jilin University Bethune Second Hospital, Changchun, P. R. China
- * E-mail: (XYZ), (GZZ)
| |
Collapse
|
33
|
Hoffmann EK, Sørensen BH, Sauter DPR, Lambert IH. Role of volume-regulated and calcium-activated anion channels in cell volume homeostasis, cancer and drug resistance. Channels (Austin) 2015; 9:380-96. [PMID: 26569161 DOI: 10.1080/19336950.2015.1089007] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
Volume-regulated channels for anions (VRAC) / organic osmolytes (VSOAC) play essential roles in cell volume regulation and other cellular functions, e.g. proliferation, cell migration and apoptosis. LRRC8A, which belongs to the leucine rich-repeat containing protein family, was recently shown to be an essential component of both VRAC and VSOAC. Reduced VRAC and VSOAC activities are seen in drug resistant cancer cells. ANO1 is a calcium-activated chloride channel expressed on the plasma membrane of e.g., secretory epithelia. ANO1 is amplified and highly expressed in a large number of carcinomas. The gene, encoding for ANO1, maps to a region on chromosome 11 (11q13) that is frequently amplified in cancer cells. Knockdown of ANO1 impairs cell proliferation and cell migration in several cancer cells. Below we summarize the basic biophysical properties of VRAC, VSOAC and ANO1 and their most important cellular functions as well as their role in cancer and drug resistance.
Collapse
Affiliation(s)
- Else K Hoffmann
- a Department of Biology ; Section for Cell Biology and Physiology; University of Copenhagen ; Copenhagen , Denmark
| | - Belinda H Sørensen
- a Department of Biology ; Section for Cell Biology and Physiology; University of Copenhagen ; Copenhagen , Denmark
| | - Daniel P R Sauter
- a Department of Biology ; Section for Cell Biology and Physiology; University of Copenhagen ; Copenhagen , Denmark
| | - Ian H Lambert
- a Department of Biology ; Section for Cell Biology and Physiology; University of Copenhagen ; Copenhagen , Denmark
| |
Collapse
|
34
|
Clinical significance of Anoctamin-1 gene at 11q13 in the development and progression of head and neck squamous cell carcinomas. Sci Rep 2015; 5:15698. [PMID: 26498851 PMCID: PMC4620505 DOI: 10.1038/srep15698] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 09/10/2015] [Indexed: 02/06/2023] Open
Abstract
This study investigates the clinical significance of Anoctamin-1 gene mapping at 11q13 amplicon in both the development and progression of head and neck squamous cell carcinomas (HNSCC). ANO1 protein expression was evaluated by immunohistochemistry in a cohort of 372 surgically treated HNSCC patients and also in 35 laryngeal precancerous lesions. ANO1 gene amplification was determined by real-time PCR in all the laryngeal premalignancies and 60 of the HNSCCs, and molecular data correlated with clinical outcome. ANO1 gene amplification was frequently detected in both premalignant lesions (63%) and HNSCC tumours (58%), whereas concomitant ANO1 expression occurred at a much lower frequency (20 and 22%). Interestingly, laryngeal dysplasias harbouring ANO1 gene amplification showed a higher risk of malignant transformation (HR = 3.62; 95% CI 0.79-16.57; P = 0.097; Cox regression). ANO1 expression and gene amplification showed no significant associations with clinicopathological parameters in HNSCC. However, remarkably ANO1 expression differentially influenced patient survival depending on the tumour site. Collectively, this study provides original evidence demonstrating the distinctive impact of ANO1 expression on HNSCC prognosis depending on the tumour site.
Collapse
|
35
|
Seo Y, Park J, Kim M, Lee HK, Kim JH, Jeong JH, Namkung W. Inhibition of ANO1/TMEM16A Chloride Channel by Idebenone and Its Cytotoxicity to Cancer Cell Lines. PLoS One 2015. [PMID: 26196390 PMCID: PMC4511415 DOI: 10.1371/journal.pone.0133656] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The expression levels of anoctamin 1 (ANO1, TMEM16A), a calcium-activated chloride channel (CaCC), are significantly increased in several tumors, and inhibition of ANO1 is known to reduce cell proliferation and migration. Here, we performed cell-based screening of a collection of natural products and drug-like compounds to identify inhibitors of ANO1. As a result of the screening, idebenone, miconazole and plumbagin were identified as novel ANO1 inhibitors. Electrophysiological studies showed that idebenone, a synthetic analog of coenzyme Q10, completely blocked ANO1 activity in FRT cells expressing ANO1 without any effect on intracellular calcium signaling and CFTR, a cAMP-regulated chloride channel. The CaCC activities in PC-3 and CFPAC-1 cells expressing abundant endogenous ANO1 were strongly blocked by idebenone. Idebenone inhibited cell proliferation and induced apoptosis in PC-3 and CFPAC-1 cells, but not in A549 cells, which do not express ANO1. These data suggest that idebenone, a novel ANO1 inhibitor, has potential for use in cancer therapy.
Collapse
Affiliation(s)
- Yohan Seo
- Department of Integrated OMICS for Biomedical Science, Graduate School, Yonsei University, Seoul, 120–749, Korea
| | - Jinhong Park
- Department of Integrated OMICS for Biomedical Science, Graduate School, Yonsei University, Seoul, 120–749, Korea
| | - Minseo Kim
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, 406–840, Korea
| | - Ho K. Lee
- Department of Integrated OMICS for Biomedical Science, Graduate School, Yonsei University, Seoul, 120–749, Korea
| | - Jin-Hee Kim
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, 406–840, Korea
| | - Jin-Hyun Jeong
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, 406–840, Korea
| | - Wan Namkung
- Department of Integrated OMICS for Biomedical Science, Graduate School, Yonsei University, Seoul, 120–749, Korea
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, 406–840, Korea
- * E-mail:
| |
Collapse
|
36
|
Leblanc N, Forrest AS, Ayon RJ, Wiwchar M, Angermann JE, Pritchard HAT, Singer CA, Valencik ML, Britton F, Greenwood IA. Molecular and functional significance of Ca(2+)-activated Cl(-) channels in pulmonary arterial smooth muscle. Pulm Circ 2015; 5:244-68. [PMID: 26064450 DOI: 10.1086/680189] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 07/22/2014] [Indexed: 12/31/2022] Open
Abstract
Increased peripheral resistance of small distal pulmonary arteries is a hallmark signature of pulmonary hypertension (PH) and is believed to be the consequence of enhanced vasoconstriction to agonists, thickening of the arterial wall due to remodeling, and increased thrombosis. The elevation in arterial tone in PH is attributable, at least in part, to smooth muscle cells of PH patients being more depolarized and displaying higher intracellular Ca(2+) levels than cells from normal subjects. It is now clear that downregulation of voltage-dependent K(+) channels (e.g., Kv1.5) and increased expression and activity of voltage-dependent (Cav1.2) and voltage-independent (e.g., canonical and vanilloid transient receptor potential [TRPC and TRPV]) Ca(2+) channels play an important role in the functional remodeling of pulmonary arteries in PH. This review focuses on an anion-permeable channel that is now considered a novel excitatory mechanism in the systemic and pulmonary circulations. It is permeable to Cl(-) and is activated by a rise in intracellular Ca(2+) concentration (Ca(2+)-activated Cl(-) channel, or CaCC). The first section outlines the biophysical and pharmacological properties of the channel and ends with a description of the molecular candidate genes postulated to encode for CaCCs, with particular emphasis on the bestrophin and the newly discovered TMEM16 and anoctamin families of genes. The second section provides a review of the various sources of Ca(2+) activating CaCCs, which include stimulation by mobilization from intracellular Ca(2+) stores and Ca(2+) entry through voltage-dependent and voltage-independent Ca(2+) channels. The third and final section summarizes recent findings that suggest a potentially important role for CaCCs and the gene TMEM16A in PH.
Collapse
Affiliation(s)
- Normand Leblanc
- Department of Pharmacology, University of Nevada School of Medicine, Reno, Nevada, USA
| | - Abigail S Forrest
- Department of Pharmacology, University of Nevada School of Medicine, Reno, Nevada, USA
| | - Ramon J Ayon
- Department of Medicine, University of Illinois, Chicago, Illinois, USA
| | - Michael Wiwchar
- Department of Pharmacology, University of Nevada School of Medicine, Reno, Nevada, USA
| | - Jeff E Angermann
- School of Community Health Sciences, University of Nevada, Reno, Nevada, USA
| | - Harry A T Pritchard
- Vascular Biology Research Centre, Institute of Cardiovascular and Cell Sciences, St. George's University of London, London, United Kingdom
| | - Cherie A Singer
- Department of Pharmacology, University of Nevada School of Medicine, Reno, Nevada, USA
| | - Maria L Valencik
- Department of Biochemistry and Molecular Biology, University of Nevada School of Medicine, Reno, Nevada, USA
| | - Fiona Britton
- Department of Physiology, School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Iain A Greenwood
- Vascular Biology Research Centre, Institute of Cardiovascular and Cell Sciences, St. George's University of London, London, United Kingdom
| |
Collapse
|
37
|
Ousingsawat J, Wanitchakool P, Kmit A, Romao AM, Jantarajit W, Schreiber R, Kunzelmann K. Anoctamin 6 mediates effects essential for innate immunity downstream of P2X7 receptors in macrophages. Nat Commun 2015; 6:6245. [PMID: 25651887 DOI: 10.1038/ncomms7245] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2014] [Accepted: 01/08/2015] [Indexed: 12/18/2022] Open
Abstract
Purinergic P2X7 receptors (P2X7R) are fundamental to innate immune response. In macrophages, transient stimulation of P2X7R activates several transport mechanisms and induces the scrambling of phospholipids with subsequent membrane blebbing and apoptosis. These processes support phagocytosis and subsequent killing of phagocytosed bacteria. Here we demonstrate that the stimulation of P2X7 receptors activates anoctamin 6 (ANO6, TMEM16F), a protein that functions as Ca(2+) dependent phospholipid scramblase and Ca(2+)-activated Cl(-) channel. Inhibition or knockdown of ANO6 attenuates ATP-induced cell shrinkage, cell migration and phospholipid scrambling. In mouse macrophages, Ano6 produces large ion currents by stimulation of P2X7 receptors and contributes to ATP-induced membrane blebbing and apoptosis, which is largely reduced in macrophages from Ano6-/- mice. ANO6 supports bacterial phagocytosis and killing by mouse and human THP-1 macrophages. Our data demonstrate that anoctamin 6 is an essential component of the immune defense by macrophages.
Collapse
Affiliation(s)
- Jiraporn Ousingsawat
- Physiological Institute, University of Regensburg, Universitätsstraße 31, D-93053 Regensburg, Germany
| | - Podchanart Wanitchakool
- Physiological Institute, University of Regensburg, Universitätsstraße 31, D-93053 Regensburg, Germany
| | - Arthur Kmit
- Physiological Institute, University of Regensburg, Universitätsstraße 31, D-93053 Regensburg, Germany
| | - Ana M Romao
- Physiological Institute, University of Regensburg, Universitätsstraße 31, D-93053 Regensburg, Germany
| | - Walailak Jantarajit
- Physiological Institute, University of Regensburg, Universitätsstraße 31, D-93053 Regensburg, Germany
| | - Rainer Schreiber
- Physiological Institute, University of Regensburg, Universitätsstraße 31, D-93053 Regensburg, Germany
| | - Karl Kunzelmann
- Physiological Institute, University of Regensburg, Universitätsstraße 31, D-93053 Regensburg, Germany
| |
Collapse
|
38
|
Mrkonjić S, Garcia-Elias A, Pardo-Pastor C, Bazellières E, Trepat X, Vriens J, Ghosh D, Voets T, Vicente R, Valverde MA. TRPV4 participates in the establishment of trailing adhesions and directional persistence of migrating cells. Pflugers Arch 2015; 467:2107-19. [PMID: 25559845 DOI: 10.1007/s00424-014-1679-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 12/17/2014] [Accepted: 12/17/2014] [Indexed: 12/21/2022]
Abstract
Calcium signaling participates in different cellular processes leading to cell migration. TRPV4, a non-selective cation channel that responds to mechano-osmotic stimulation and heat, is also involved in cell migration. However, the mechanistic involvement of TRPV4 in cell migration is currently unknown. We now report that expression of the mutant channel TRPV4-(121)AAWAA (lacking the phosphoinositide-binding site (121)KRWRK(125) and the response to physiological stimuli) altered HEK293 cell migration. Altered migration patterns included periods of fast and persistent motion followed by periods of stalling and turning, and the extension of multiple long cellular protrusions. TRPV4-WT overexpressing cells showed almost complete loss of directionality with frequent turns, no progression, and absence of long protrusions. Traction microscopy revealed higher tractions forces in the tail of TRPV4-(121)AAWAA than in TRPV4-WT expressing cells. These results are consistent with a defective and augmented tail retraction in TRPV4-(121)AAWAA- and TRPV4-WT-expressing cells, respectively. The activity of calpain, a protease implicated in focal adhesion (FA) disassembly, was decreased in TRPV4-(121)AAWAA compared with TRPV4-WT-expressing cells. Consistently, larger focal adhesions were seen in TRPV4-(121)AAWAA compared with TRPV4-WT-expressing HEK293 cells, a result that was also reproduced in T47D and U87 cells. Similarly, overexpression of the pore-dead mutant TRPV4-M680D resumed the TRPV4-(121)AAWAA phenotype presenting larger FA. The migratory phenotype obtained in HEK293 cells overexpressing TRPV4-(121)AAWAA was mimicked by knocking-down TRPC1, a cationic channel that participates in cell migration. Together, our results point to the participation of TRPV4 in the dynamics of trailing adhesions, a function that may require the interplay of TRPV4 with other cation channels or proteins present at the FA sites.
Collapse
Affiliation(s)
- Sanela Mrkonjić
- Laboratory of Molecular Physiology and Channelopathies, Dept. of Experimental and Health Sciences, Universitat Pompeu Fabra, C/ Dr. Aiguader 88, Barcelona, 08003, Spain
| | - Anna Garcia-Elias
- Laboratory of Molecular Physiology and Channelopathies, Dept. of Experimental and Health Sciences, Universitat Pompeu Fabra, C/ Dr. Aiguader 88, Barcelona, 08003, Spain
| | - Carlos Pardo-Pastor
- Laboratory of Molecular Physiology and Channelopathies, Dept. of Experimental and Health Sciences, Universitat Pompeu Fabra, C/ Dr. Aiguader 88, Barcelona, 08003, Spain
| | - Elsa Bazellières
- Institute for Bioengineering of Catalonia, Barcelona, Barcelona, Spain
- Facultat de Medicina, Universitat de Barcelona, and Ciber Enfermedades Respiratorias, Barcelona, Barcelona, Spain
| | - Xavier Trepat
- Institute for Bioengineering of Catalonia, Barcelona, Barcelona, Spain
- Facultat de Medicina, Universitat de Barcelona, and Ciber Enfermedades Respiratorias, Barcelona, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Barcelona, Spain
| | - Joris Vriens
- Laboratory of Ion Channels and TRP Research Platform Leuven, KU Leuven, Leuven, Belgium
| | - Debapriya Ghosh
- Laboratory of Ion Channels and TRP Research Platform Leuven, KU Leuven, Leuven, Belgium
| | - Thomas Voets
- Laboratory of Ion Channels and TRP Research Platform Leuven, KU Leuven, Leuven, Belgium
| | - Rubén Vicente
- Laboratory of Molecular Physiology and Channelopathies, Dept. of Experimental and Health Sciences, Universitat Pompeu Fabra, C/ Dr. Aiguader 88, Barcelona, 08003, Spain
| | - Miguel A Valverde
- Laboratory of Molecular Physiology and Channelopathies, Dept. of Experimental and Health Sciences, Universitat Pompeu Fabra, C/ Dr. Aiguader 88, Barcelona, 08003, Spain.
| |
Collapse
|
39
|
Kodigepalli KM, Bowers K, Sharp A, Nanjundan M. Roles and regulation of phospholipid scramblases. FEBS Lett 2014; 589:3-14. [PMID: 25479087 DOI: 10.1016/j.febslet.2014.11.036] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Revised: 11/22/2014] [Accepted: 11/24/2014] [Indexed: 02/06/2023]
Abstract
Phospholipid scramblase activity is involved in the collapse of phospholipid (PL) asymmetry at the plasma membrane leading to externalization of phosphatidylserine. This activity is crucial for initiation of the blood coagulation cascade and for recognition/elimination of apoptotic cells by macrophages. Efforts to identify gene products associated with this activity led to the characterization of PL scramblase (PLSCR) and XKR family members which contribute to phosphatidylserine exposure in response to apoptotic stimuli. Meanwhile, TMEM16 family members were identified to externalize phosphatidylserine in response to elevated calcium in Scott syndrome platelets, which is critical for activation of the coagulation cascade. Herein, we report their mechanisms of gene regulation, molecular functions independent of their scrambling activity, and their potential roles in pathogenic conditions.
Collapse
Affiliation(s)
- Karthik M Kodigepalli
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL 33620, United States
| | - Kiah Bowers
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL 33620, United States
| | - Arielle Sharp
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL 33620, United States
| | - Meera Nanjundan
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL 33620, United States.
| |
Collapse
|
40
|
Stock C, Schwab A. Ion channels and transporters in metastasis. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2014; 1848:2638-46. [PMID: 25445667 DOI: 10.1016/j.bbamem.2014.11.012] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2014] [Revised: 11/03/2014] [Accepted: 11/07/2014] [Indexed: 12/23/2022]
Abstract
An elaborate interplay between ion channels and transporters, components of the cytoskeleton, adhesion molecules, and signaling cascades provides the basis for each major step of the metastatic cascade. Ion channels and transporters contribute to cell motility by letting through or transporting ions essential for local Ca2+, pH and--in cooperation with water permeable aquaporins--volume homeostasis. Moreover, in addition to the actual ion transport they, or their auxiliary subunits, can display non-conducting activities. They can exert kinase activity in order to phosphorylate cytoskeletal constituents or their associates. They can become part of signaling processes by permeating Ca2+, by generating local pH-nanodomains or by being final downstream effectors. A number of channels and transporters are found at focal adhesions, interacting directly or indirectly with proteins of the extracellular matrix, with integrins or with components of the cytoskeleton. We also include the role of aquaporins in cell motility. They drive the outgrowth of lamellipodia/invadopodia or control the number of β1 integrins in the plasma membrane. The multitude of interacting ion channels and transporters (called transportome) including the associated signaling events holds great potential as therapeutic target(s) for anticancer agents that are aimed at preventing metastasis. This article is part of a Special Issue entitled: Membrane channels and transporters in cancers.
Collapse
Affiliation(s)
- Christian Stock
- Department of Gastroenterology, Hannover Medical School, Hannover, Germany.
| | - Albrecht Schwab
- Institute of Physiology II, University of Münster, Robert-Koch-Str. 27b, D-48149 Münster, Germany
| |
Collapse
|
41
|
TMEM16 proteins: unknown structure and confusing functions. J Mol Biol 2014; 427:94-105. [PMID: 25451786 DOI: 10.1016/j.jmb.2014.09.028] [Citation(s) in RCA: 106] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Revised: 09/24/2014] [Accepted: 09/29/2014] [Indexed: 11/23/2022]
Abstract
The TMEM16 family of membrane proteins, also known as anoctamins, plays key roles in a variety of physiological functions that range from ion transport to phospholipid scrambling and to regulating other ion channels. The first two family members to be functionally characterized, TMEM16A (ANO1) and TMEM16B (ANO2), form Ca(2+)-activated Cl(-) channels and are important for transepithelial ion transport, olfaction, phototransduction, smooth muscle contraction, nociception, cell proliferation and control of neuronal excitability. The roles of other family members, such as TMEM16C (ANO3), TMEM16D (ANO4), TMEM16F (ANO6), TMEM16G (ANO7) and TMEM16J (ANO9), remain poorly understood and controversial. These homologues were reported to be phospholipid scramblases, ion channels, to have both functions or to be regulatory subunits of other channels. Mutations in TMEM16F cause Scott syndrome, a bleeding disorder caused by impaired Ca(2+)-dependent externalization of phosphatidylserine in activated platelets, suggesting that this homologue might be a scramblase. However, overexpression of TMEM16F has also been associated with a remarkable number of different ion channel types, raising the possibility that this protein might be involved in both ion and lipid transports. The recent identification of an ancestral TMEM16 homologue with intrinsic channel and scramblase activities supports this hypothesis. Thus, the TMEM16 family might have diverged in two or three different subclasses, channels, scramblases and dual-function channel/scramblases. The structural bases and functional implication of such a functional diversity within a single protein family remain to be elucidated and the links between TMEM16 functions and human physiology and pathologies need to be investigated.
Collapse
|
42
|
Matsuba S, Niwa S, Muraki K, Kanatsuka S, Nakazono Y, Hatano N, Fujii M, Zhan P, Suzuki T, Ohya S. Downregulation of Ca2+-activated Cl- channel TMEM16A by the inhibition of histone deacetylase in TMEM16A-expressing cancer cells. J Pharmacol Exp Ther 2014; 351:510-8. [PMID: 25232193 DOI: 10.1124/jpet.114.217315] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The Ca(2+)-activated Cl(-) channel transmembrane proteins with unknown function 16 A (TMEM16A; also known as anoctamin 1 or discovered on gastrointestinal stromal tumor 1) plays an important role in facilitating the cell growth and metastasis of TMEM16A-expressing cancer cells. Histone deacetylase (HDAC) inhibitors (HDACi) are useful agents for cancer therapy, but it remains unclear whether ion channels are epigenetically regulated by them. Using real-time polymerase chain reaction, Western blot analysis, and whole-cell patch-clamp assays, we found a significant decrease in TMEM16A expression and its functional activity was induced by the vorinostat, a pan-HDACi in TMEM16A-expressing human cancer cell lines, the prostatic cancer cell line PC-3, and the breast cancer cell line YMB-1. TMEM16A downregulation was not induced by the chemotherapy drug paclitaxel in either cell type. Pharmacologic blockade of HDAC3 by 1 μM T247 [N-(2-aminophenyl)-4-[1-(2-thiophen-3-ylethyl)-1H-[1],[2],[3]triazol-4-yl]benzamide], a HDAC3-selective HDACi, elicited a large decrease in TMEM16A expression and functional activity in both cell types, and pharmacologic blockade of HDAC2 by AATB [4-(acetylamino)-N-[2-amino-5-(2-thienyl)phenyl]-benzamide; 300 nM] elicited partial inhibition of TMEM16A expression (∼40%) in both. Pharmacologic blockade of HDAC1 or HDAC6 did not elicit any significant change in TMEM16A expression, respectively. In addition, inhibition of HDAC3 induced by small interfering RNA elicited a large decrease in TMEM16A transcripts in both cell types. Taken together, in malignancies with a frequent gene amplification of TMEM16A, HDAC3 inhibition may exert suppressive effects on cancer cell viability via downregulation of TMEM16A.
Collapse
Affiliation(s)
- Sayo Matsuba
- Department of Pharmacology, Division of Pathological Sciences, Kyoto Pharmaceutical University, Kyoto, Japan (S.M., S.N., S.K., Y.N., M.F., S.O.); Laboratory of Cellular Pharmacology, School of Pharmacy, Aichi-Gakuin University, Nagoya, Japan (K.M., N.H.); and Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan (P.Z., T.S.)
| | - Satomi Niwa
- Department of Pharmacology, Division of Pathological Sciences, Kyoto Pharmaceutical University, Kyoto, Japan (S.M., S.N., S.K., Y.N., M.F., S.O.); Laboratory of Cellular Pharmacology, School of Pharmacy, Aichi-Gakuin University, Nagoya, Japan (K.M., N.H.); and Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan (P.Z., T.S.)
| | - Katsuhiko Muraki
- Department of Pharmacology, Division of Pathological Sciences, Kyoto Pharmaceutical University, Kyoto, Japan (S.M., S.N., S.K., Y.N., M.F., S.O.); Laboratory of Cellular Pharmacology, School of Pharmacy, Aichi-Gakuin University, Nagoya, Japan (K.M., N.H.); and Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan (P.Z., T.S.)
| | - Saki Kanatsuka
- Department of Pharmacology, Division of Pathological Sciences, Kyoto Pharmaceutical University, Kyoto, Japan (S.M., S.N., S.K., Y.N., M.F., S.O.); Laboratory of Cellular Pharmacology, School of Pharmacy, Aichi-Gakuin University, Nagoya, Japan (K.M., N.H.); and Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan (P.Z., T.S.)
| | - Yurika Nakazono
- Department of Pharmacology, Division of Pathological Sciences, Kyoto Pharmaceutical University, Kyoto, Japan (S.M., S.N., S.K., Y.N., M.F., S.O.); Laboratory of Cellular Pharmacology, School of Pharmacy, Aichi-Gakuin University, Nagoya, Japan (K.M., N.H.); and Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan (P.Z., T.S.)
| | - Noriyuki Hatano
- Department of Pharmacology, Division of Pathological Sciences, Kyoto Pharmaceutical University, Kyoto, Japan (S.M., S.N., S.K., Y.N., M.F., S.O.); Laboratory of Cellular Pharmacology, School of Pharmacy, Aichi-Gakuin University, Nagoya, Japan (K.M., N.H.); and Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan (P.Z., T.S.)
| | - Masanori Fujii
- Department of Pharmacology, Division of Pathological Sciences, Kyoto Pharmaceutical University, Kyoto, Japan (S.M., S.N., S.K., Y.N., M.F., S.O.); Laboratory of Cellular Pharmacology, School of Pharmacy, Aichi-Gakuin University, Nagoya, Japan (K.M., N.H.); and Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan (P.Z., T.S.)
| | - Peng Zhan
- Department of Pharmacology, Division of Pathological Sciences, Kyoto Pharmaceutical University, Kyoto, Japan (S.M., S.N., S.K., Y.N., M.F., S.O.); Laboratory of Cellular Pharmacology, School of Pharmacy, Aichi-Gakuin University, Nagoya, Japan (K.M., N.H.); and Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan (P.Z., T.S.)
| | - Takayoshi Suzuki
- Department of Pharmacology, Division of Pathological Sciences, Kyoto Pharmaceutical University, Kyoto, Japan (S.M., S.N., S.K., Y.N., M.F., S.O.); Laboratory of Cellular Pharmacology, School of Pharmacy, Aichi-Gakuin University, Nagoya, Japan (K.M., N.H.); and Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan (P.Z., T.S.)
| | - Susumu Ohya
- Department of Pharmacology, Division of Pathological Sciences, Kyoto Pharmaceutical University, Kyoto, Japan (S.M., S.N., S.K., Y.N., M.F., S.O.); Laboratory of Cellular Pharmacology, School of Pharmacy, Aichi-Gakuin University, Nagoya, Japan (K.M., N.H.); and Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan (P.Z., T.S.)
| |
Collapse
|
43
|
Sauter DRP, Novak I, Pedersen SF, Larsen EH, Hoffmann EK. ANO1 (TMEM16A) in pancreatic ductal adenocarcinoma (PDAC). Pflugers Arch 2014; 467:1495-1508. [PMID: 25163766 PMCID: PMC4464647 DOI: 10.1007/s00424-014-1598-8] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Revised: 07/22/2014] [Accepted: 08/11/2014] [Indexed: 12/31/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has one of the worst survival rates of all cancers. ANO1 (TMEM16A) is a recently identified Ca2+-activated Cl− channel (CaCC) that is upregulated in several tumors. Although ANO1 was subject to extensive studies in the recent years, its pathophysiological function has only been poorly understood. The aim of the present study is to establish the significance of ANO1 in PDAC behavior and demarcate its roles in PDAC from those of the volume-regulated anion channel (VRAC). We performed qPCR and Western blot measurements on different PDAC cell lines (Panc-1, Mia PaCa 2, Capan-1, AsPC-1, BxPC-3) and compared the results to those obtained in a human pancreatic ductal epithelium (HPDE) cell line. All cancer cell lines showed an upregulation of ANO1 on mRNA and protein levels. Whole-cell patch-clamp recordings identified large Ca2+ and voltage-dependent Cl− currents in PDAC cells. Using siRNA knockdown of ANO1 and three ANO1 inhibitors (T16Ainh-A01, CaCCinh-A01, and NS3728), we found that ANO1 is the main constituent of CaCC current in PDAC cells. We further characterized these three inhibitors and found that they had unspecific effects on the free intracellular calcium concentration. Functional studies on PDAC behavior showed that surprisingly inhibition of ANO1 did not influence cellular proliferation. On the other hand, we found ANO1 channel to be pivotal in PDAC cell migration as assessed in wound healing experiments.
Collapse
Affiliation(s)
- D R P Sauter
- Section for Cell and Developmental Biology, Department of Biology, University of Copenhagen, August Krogh Building, Universitetsparken 13, 2100, Copenhagen Ø, Denmark.
| | - I Novak
- Section for Molecular Integrative Physiology, Department of Biology, University of Copenhagen, August Krogh Building, Universitetsparken 13, 2100 Copenhagen Ø, Denmark
| | - S F Pedersen
- Section for Cell and Developmental Biology, Department of Biology, University of Copenhagen, August Krogh Building, Universitetsparken 13, 2100, Copenhagen Ø, Denmark
| | - E H Larsen
- Section for Molecular Integrative Physiology, Department of Biology, University of Copenhagen, August Krogh Building, Universitetsparken 13, 2100 Copenhagen Ø, Denmark
| | - E K Hoffmann
- Section for Cell and Developmental Biology, Department of Biology, University of Copenhagen, August Krogh Building, Universitetsparken 13, 2100, Copenhagen Ø, Denmark
| |
Collapse
|
44
|
Nielsen N, Lindemann O, Schwab A. TRP channels and STIM/ORAI proteins: sensors and effectors of cancer and stroma cell migration. Br J Pharmacol 2014; 171:5524-40. [PMID: 24724725 DOI: 10.1111/bph.12721] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Revised: 03/24/2014] [Accepted: 04/03/2014] [Indexed: 01/05/2023] Open
Abstract
UNLABELLED Cancer cells are strongly influenced by host cells within the tumour stroma and vice versa. This leads to the development of a tumour microenvironment with distinct physical and chemical properties that are permissive for tumour progression. The ability to migrate plays a central role in this mutual interaction. Migration of cancer cells is considered as a prerequisite for tumour metastasis and the migration of host stromal cells is required for reaching the tumour site. Increasing evidence suggests that transient receptor potential (TRP) channels and STIM/ORAI proteins affect key calcium-dependent mechanisms implicated in both cancer and stroma cell migration. These include, among others, cytoskeletal remodelling, growth factor/cytokine signalling and production, and adaptation to tumour microenvironmental properties such as hypoxia and oxidative stress. In this review, we will summarize the current knowledge regarding TRP channels and STIM/ORAI proteins in cancer and stroma cell migration. We focus on how TRP channel or STIM/ORAI-mediated Ca(2+) signalling directly or indirectly influences cancer and stroma cell migration by affecting the above listed mechanisms. LINKED ARTICLES This article is part of a themed section on Cytoskeleton, Extracellular Matrix, Cell Migration, Wound Healing and Related Topics. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2014.171.issue-24.
Collapse
Affiliation(s)
- N Nielsen
- Institute of Physiology II, University of Münster, Münster, Germany
| | | | | |
Collapse
|
45
|
Shiwarski DJ, Shao C, Bill A, Kim J, Xiao D, Bertrand CA, Seethala RS, Sano D, Myers JN, Ha P, Grandis J, Gaither LA, Puthenveedu MA, Duvvuri U. To "grow" or "go": TMEM16A expression as a switch between tumor growth and metastasis in SCCHN. Clin Cancer Res 2014; 20:4673-88. [PMID: 24919570 DOI: 10.1158/1078-0432.ccr-14-0363] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
PURPOSE Tumor metastasis is the leading cause of death in patients with cancer. However, the mechanisms that underlie metastatic progression remain unclear. We examined TMEM16A (ANO1) expression as a key factor shifting tumors between growth and metastasis. EXPERIMENTAL DESIGN We evaluated 26 pairs of primary and metastatic lymph node (LN) tissue from patients with squamous cell carcinoma of the head and neck (SCCHN) for differential expression of TMEM16A. In addition, we identified mechanisms by which TMEM16A expression influences tumor cell motility via proteomic screens of cell lines and in vivo mouse studies of metastasis. RESULTS Compared with primary tumors, TMEM16A expression decreases in metastatic LNs of patients with SCCHN. Stable reduction of TMEM16A expression enhances cell motility and increases metastases while decreasing tumor proliferation in an orthotopic mouse model. Evaluation of human tumor tissues suggests an epigenetic mechanism for decreasing TMEM16A expression through promoter methylation that correlated with a transition between an epithelial and a mesenchymal phenotype. These effects of TMEM16A expression on tumor cell size and epithelial-to-mesenchymal transition (EMT) required the amino acid residue serine 970 (S970); however, mutation of S970 to alanine does not disrupt the proliferative advantages of TMEM16A overexpression. Furthermore, S970 mediates the association of TMEM16A with Radixin, an actin-scaffolding protein implicated in EMT. CONCLUSIONS Together, our results identify TMEM16A, an eight transmembrane domain Ca2+-activated Cl- channel, as a primary driver of the "Grow" or "Go" model for cancer progression, in which TMEM16A expression acts to balance tumor proliferation and metastasis via its promoter methylation.
Collapse
Affiliation(s)
- Daniel J Shiwarski
- VA Pittsburgh Health System, Pittsburgh, Pennsylvania. Department of Otolaryngology, University of Pittsburgh, Pittsburgh, Pennsylvania. Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | - Chunbo Shao
- Department of Otolaryngology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Anke Bill
- Department of Developmental and Molecular Pathways, Novartis Institute for Biomedical Research Inc., Cambridge, Massachusetts
| | - Jean Kim
- VA Pittsburgh Health System, Pittsburgh, Pennsylvania. Department of Otolaryngology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Dong Xiao
- VA Pittsburgh Health System, Pittsburgh, Pennsylvania. Department of Otolaryngology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | - Raja S Seethala
- Pathology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Daisuke Sano
- Yokohama City University, School of Medicine, Yokohama, Japan
| | - Jeffery N Myers
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Patrick Ha
- Department of Otolaryngology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jennifer Grandis
- VA Pittsburgh Health System, Pittsburgh, Pennsylvania. Department of Otolaryngology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - L Alex Gaither
- Department of Developmental and Molecular Pathways, Novartis Institute for Biomedical Research Inc., Cambridge, Massachusetts
| | | | - Umamaheswar Duvvuri
- VA Pittsburgh Health System, Pittsburgh, Pennsylvania. Department of Otolaryngology, University of Pittsburgh, Pittsburgh, Pennsylvania.
| |
Collapse
|
46
|
Hoffmann EK, Holm NB, Lambert IH. Functions of volume-sensitive and calcium-activated chloride channels. IUBMB Life 2014; 66:257-67. [PMID: 24771413 DOI: 10.1002/iub.1266] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Revised: 03/31/2014] [Accepted: 03/31/2014] [Indexed: 01/23/2023]
Abstract
The review describes molecular and functional properties of the volume regulated anion channel and Ca(2+)-dependent Cl(-) channels belonging to the anoctamin family with emphasis on physiological importance of these channels in regulation of cell volume, cell migration, cell proliferation, and programmed cell death. Finally, we discuss the role of Cl(-) channels in various diseases.
Collapse
Affiliation(s)
- Else Kay Hoffmann
- Department of Biology, University of Copenhagen, 13 Universitetsparken, Copenhagen Ø, Denmark
| | | | | |
Collapse
|
47
|
Qu Z, Yao W, Yao R, Liu X, Yu K, Hartzell C. The Ca(2+) -activated Cl(-) channel, ANO1 (TMEM16A), is a double-edged sword in cell proliferation and tumorigenesis. Cancer Med 2014; 3:453-61. [PMID: 24639373 PMCID: PMC4101736 DOI: 10.1002/cam4.232] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2013] [Revised: 02/13/2014] [Accepted: 02/21/2014] [Indexed: 01/11/2023] Open
Abstract
Since anoctamin 1 ANO1 (TMEM16A) was found to be a molecular component of Ca2+-activated Cl− channels, its role in tumorigenesis has gained attention at a fast pace. ANO1 overexpression frequently occurs in the cancer tissues along with 11q13 chromosome amplification. Poor prognosis of many types of cancers has been closely correlated with ANO1 gene amplification and protein overexpression. ANO1 is now considered an excellent biomarker for certain cancers. Recent research suggests that it is the channel function of ANO1 that is involved in the tumorigenesis. However, how the overexpression of the functional ANO1 causes malignant transformation of tissues via signaling pathways, for example, MAPK remains to be investigated. Clarification of the reasons in future will avail to make ANO1 as a target for cancer treatment.
Collapse
Affiliation(s)
- Zhiqiang Qu
- Medical Research Center, Affiliated Hospital, Qingdao University, Qingdao, 266555, China; State Key Physiological Discipline, Qingdao University, Qingdao, 266555, China
| | | | | | | | | | | |
Collapse
|
48
|
Kunzelmann K, Nilius B, Owsianik G, Schreiber R, Ousingsawat J, Sirianant L, Wanitchakool P, Bevers EM, Heemskerk JWM. Molecular functions of anoctamin 6 (TMEM16F): a chloride channel, cation channel, or phospholipid scramblase? Pflugers Arch 2014; 466:407-14. [PMID: 23748496 DOI: 10.1007/s00424-013-1305-1] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Revised: 05/27/2013] [Accepted: 05/28/2013] [Indexed: 10/26/2022]
Abstract
Anoctamin 6 (Ano6; TMEM16F gene) is a ubiquitous protein; the expression of which is defective in patients with Scott syndrome, an inherited bleeding disorder based on defective scrambling of plasma membrane phospholipids. For Ano6, quite diverse functions have been described: (1) it can form an outwardly rectifying, Ca(2+)-dependent and a volume-regulated Cl(-) channel; (2) it was claimed to be a Ca(2+)-regulated nonselective cation channel permeable for Ca(2+); (3) it was shown to be essential for Ca(2+)-mediated scrambling of membrane phospholipids; and (4) it can regulate cell blebbing and microparticle shedding. Deficiency of Ano6 in blood cells from Scott patients or Ano6 null mice appears to affect all of these cell responses. Furthermore, Ano6 deficiency in mice impairs the mineralization of osteoblasts, resulting in reduced skeletal development. These diverse results have been obtained under different experimental conditions, which may explain some of the contradictions. This review therefore aims to summarize the currently available information on the diverse roles of Ano6 and tries to clear up some of the existing controversies.
Collapse
Affiliation(s)
- Karl Kunzelmann
- Institut für Physiologie, Universität Regensburg, Universitätsstraße 31, 93053, Regensburg, Germany,
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Schwab A, Stock C. Ion channels and transporters in tumour cell migration and invasion. Philos Trans R Soc Lond B Biol Sci 2014; 369:20130102. [PMID: 24493750 DOI: 10.1098/rstb.2013.0102] [Citation(s) in RCA: 101] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Cell migration is a central component of the metastatic cascade requiring a concerted action of ion channels and transporters (migration-associated transportome), cytoskeletal elements and signalling cascades. Ion transport proteins and aquaporins contribute to tumour cell migration and invasion among other things by inducing local volume changes and/or by modulating Ca(2+) and H(+) signalling. Targeting cell migration therapeutically bears great clinical potential, because it is a prerequisite for metastasis. Ion transport proteins appear to be attractive candidate target proteins for this purpose because they are easily accessible as membrane proteins and often overexpressed or activated in cancer. Importantly, a number of clinically widely used drugs are available whose anticipated efficacy as anti-tumour drugs, however, has now only begun to be evaluated.
Collapse
Affiliation(s)
- Albrecht Schwab
- Institut für Physiologie II, Westfälische Wilhelms-Universität Münster, , Robert-Koch-Strasse 27b, Münster 48149, Germany
| | | |
Collapse
|
50
|
Anoctamin 6 differs from VRAC and VSOAC but is involved in apoptosis and supports volume regulation in the presence of Ca2+. Pflugers Arch 2014; 466:1899-910. [PMID: 24419539 PMCID: PMC4159566 DOI: 10.1007/s00424-013-1428-4] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Revised: 12/17/2013] [Accepted: 12/19/2013] [Indexed: 10/25/2022]
Abstract
Anoctamin 6 (ANO6), also known as TMEM16F, has been shown to be a calcium-activated anion channel with delayed calcium activation. The cellular function of ANO6 is under debate, and different groups have come to different conclusions about ANO6's physiological role. Although it is now quite well established that ANO6 is distinct from the volume-regulated anion channel, it is still unclear whether ANO6 or other anoctamins can be activated by cell swelling. In this study, we suggest that ANO1, ANO6, and ANO10 do not contribute to the volume-activated current in ANO-overexpressing HEK293 cells. Furthermore, knock-down of ANO6 in Ehrlich ascites tumor cells (EATC) and Ehrlich-Lettre ascites (ELA) did not decrease but instead significantly increased swelling-activated membrane currents. Knock-down of ANO6 in EATC did not reduce regulatory volume decrease (RVD) in the absence of extracellular calcium, whereas it significantly reduced RVD in the presence of calcium. Interestingly, we found that knock-down of ANO6 in ELA cells resulted in a decrease in cisplatin-induced caspase-3 activity, confirming earlier findings that ANO6 is involved in apoptosis. Finally, knock-down of ANO1 and ANO6 did not affect the volume-sensitive release of taurine in ELA cells. Thus, our data provide evidence that ANO6 cannot be activated directly by cell swelling unless Ca(2+) is present. We also conclude that ANO6 carries a current during RVD, provided extracellular calcium is present. Thus, swelling activation of ANO6 requires the presence of free calcium.
Collapse
|