1
|
Huang Z, Iqbal Z, Zhao Z, Chen X, Mahmmod A, Liu J, Li W, Deng Z. TMEM16 proteins: Ca 2+‑activated chloride channels and phospholipid scramblases as potential drug targets (Review). Int J Mol Med 2024; 54:81. [PMID: 39092585 PMCID: PMC11315658 DOI: 10.3892/ijmm.2024.5405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 06/06/2024] [Indexed: 08/04/2024] Open
Abstract
TMEM16 proteins, which function as Ca2+‑activated Cl‑ channels are involved in regulating a wide variety of cellular pathways and functions. The modulators of Cl‑ channels can be used for the molecule‑based treatment of respiratory diseases, cystic fibrosis, tumors, cancer, osteoporosis and coronavirus disease 2019. The TMEM16 proteins link Ca2+ signaling, cellular electrical activity and lipid transport. Thus, deciphering these complex regulatory mechanisms may enable a more comprehensive understanding of the physiological functions of the TMEM16 proteins and assist in ascertaining the applicability of these proteins as potential pharmacological targets for the treatment of a range of diseases. The present review examined the structures, functions and characteristics of the different types of TMEM16 proteins, their association with the pathogenesis of various diseases and the applicability of TMEM16 modulator‑based treatment methods.
Collapse
Affiliation(s)
- Zeqi Huang
- Department of Hand and Foot Surgery, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen, Guangdong 518000, P.R. China
| | - Zoya Iqbal
- Department of Orthopaedics, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen, Guangdong 518000, P.R. China
| | - Zhe Zhao
- Department of Hand and Foot Surgery, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen, Guangdong 518000, P.R. China
| | - Xiaoqiang Chen
- Department of Hand and Foot Surgery, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen, Guangdong 518000, P.R. China
| | - Ayesha Mahmmod
- Faculty of Pharmacy, The University of Lahore, Lahore, Punjab 58240, Pakistan
| | - Jianquan Liu
- Department of Hand and Foot Surgery, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen, Guangdong 518000, P.R. China
| | - Wencui Li
- Department of Hand and Foot Surgery, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen, Guangdong 518000, P.R. China
| | - Zhiqin Deng
- Department of Hand and Foot Surgery, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen, Guangdong 518000, P.R. China
| |
Collapse
|
2
|
Li Y, Sanchez Triviño CA, Hernandez A, Mortal S, Spada F, Krivosheia I, Franco N, Spelat R, Cesselli D, Manini I, Skrap M, Menini A, Cesca F, Torre V. Mechanisms of Glioblastoma Replication: Ca2+ Flares and Cl- Currents. Mol Cancer Res 2024; 22:852-863. [PMID: 38820126 DOI: 10.1158/1541-7786.mcr-23-0934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 04/11/2024] [Accepted: 05/29/2024] [Indexed: 06/02/2024]
Abstract
Glioblastoma (GBM) is amongst the deadliest types of cancers, with no resolutive cure currently available. GBM cell proliferation in the patient's brain is a complex phenomenon controlled by multiple mechanisms. The aim of this study was to determine whether the ionic fluxes controlling cell duplication could represent a target for GBM therapy. In this work, we combined multi-channel Ca2+ and Cl- imaging, optical tweezers, electrophysiology, and immunohistochemistry to describe the role of ion fluxes in mediating the cell volume changes that accompany mitosis of U87 GBM cells. We identified three main steps: (i) in round GBM cells undergoing mitosis, during the transition from anaphase to telophase and cytokinesis, large Ca2+ flares occur, reaching values of 0.5 to 1 μmol/L; (ii) these Ca2+ flares activate Ca2+-dependent Cl- channels, allowing the entry of Cl- ions; and (iii) to maintain osmotic balance, GBM cells swell to complete mitosis. This sequence of steps was validated by electrophysiological experiments showing that Cl- channels are activated either directly or indirectly by Ca2+, and by additional live-cell imaging experiments. Cl- channel blockers with different molecular structures, such as niflumic acid and carbenoxolone, blocked GBM replication by arresting GBM cells in a round configuration. These results describe the central role of Ca2+ flares and Cl- fluxes during mitosis and show that inhibition of Ca2+-activated Cl- channels blocks GBM replication, opening the way to new approaches for the clinical treatment of GBM. Implications: Our work identifies ionic fluxes occurring during cell division as targets for devising novel therapies for glioblastoma treatment.
Collapse
Affiliation(s)
- Yunzhen Li
- Central Laboratory of the Medical Research Center, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang Province, China
- International School for Advanced Studies (SISSA), Trieste, Italy
| | | | - Andres Hernandez
- International School for Advanced Studies (SISSA), Trieste, Italy
| | - Simone Mortal
- International School for Advanced Studies (SISSA), Trieste, Italy
| | - Federica Spada
- International School for Advanced Studies (SISSA), Trieste, Italy
- Department of Life Sciences, University of Trieste, Trieste, Italy
- IOM-CNR, Trieste, Italy
| | - Ilona Krivosheia
- International School for Advanced Studies (SISSA), Trieste, Italy
- Department of Life Sciences, University of Trieste, Trieste, Italy
- IOM-CNR, Trieste, Italy
| | - Nicoletta Franco
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Renza Spelat
- International School for Advanced Studies (SISSA), Trieste, Italy
- IOM-CNR, Trieste, Italy
| | - Daniela Cesselli
- Department of Medicine, Institute of Pathology, University Hospital of Udine, University of Udine, Udine, Italy
| | - Ivana Manini
- Department of Medicine, Institute of Pathology, University Hospital of Udine, University of Udine, Udine, Italy
| | - Miran Skrap
- Neurosurgery Unit, Head-Neck and Neuroscience Department, University Hospital of Udine, Udine, Italy
| | - Anna Menini
- International School for Advanced Studies (SISSA), Trieste, Italy
| | - Fabrizia Cesca
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Vincent Torre
- International School for Advanced Studies (SISSA), Trieste, Italy
- IOM-CNR, Trieste, Italy
- BISS GlioGuard S.r.l., Trieste, Italy
- Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, P.R. China
| |
Collapse
|
3
|
Wang Y, Cao Y, Chen Y, Cheng H, Liu Z, Wang M, Feng Y, Fei B, Cui K, Huang Z. YWHAG promotes colorectal cancer progression by regulating the CTTN-Wnt/β-catenin signaling axis. Med Oncol 2024; 41:100. [PMID: 38538804 DOI: 10.1007/s12032-024-02349-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 02/27/2024] [Indexed: 05/31/2024]
Abstract
Colorectal cancer (CRC) ranks as the third most prevalent cancer type globally. Nevertheless, the fundamental mechanisms driving CRC progression remain ambiguous, and the prognosis for the majority of patients diagnosed at an advanced stage is dismal. YWHA/14-3-3 proteins serve as central nodes in several signaling pathways and are closely related to tumorigenesis and progression. However, their exact roles in CRC are still poorly elucidated. In this study, we revealed that YWHAG was the most significantly upregulated member of the YWHA/14-3-3 family in CRC tissues and was associated with a poor prognosis. Subsequent phenotypic experiments showed that YWHAG promoted the proliferation, migration, and invasion of CRC cells. Mechanistically, RNA-seq data showed that multiple signaling pathways, including Wnt and epithelial-mesenchymal transition, were potentially regulated by YWHAG. CTTN was identified as a YWHAG-associated protein, and mediated its tumor-promoting functions by activating the Wnt/β-catenin signaling in CRC cells. In summary, our data indicate that YWHAG facilitates the proliferation, migration, and invasion of CRC cells by modulating the CTTN-Wnt/β-catenin signaling pathway, which offers a novel perspective for the treatment of CRC.
Collapse
Affiliation(s)
- Yuanben Wang
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, 214062, Jiangsu, China
- Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Yulin Cao
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, 214062, Jiangsu, China
- Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Ying Chen
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, 214062, Jiangsu, China
- Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Han Cheng
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, 214062, Jiangsu, China
- Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Zhiang Liu
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, 214062, Jiangsu, China
- Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Mengna Wang
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, 214062, Jiangsu, China
- Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Yuyang Feng
- Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Bojian Fei
- Department of Department of Gastrointestinal Surgery, Affiliated Hospital of Jiangnan University, 1000 He Feng Road, Wuxi, 214122, Jiangsu, China
| | - Kaisa Cui
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, 214062, Jiangsu, China
- Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Zhaohui Huang
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, 214062, Jiangsu, China.
- Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, China.
| |
Collapse
|
4
|
Arreola J, Pérez-Cornejo P, Segura-Covarrubias G, Corral-Fernández N, León-Aparicio D, Guzmán-Hernández ML. Function and Regulation of the Calcium-Activated Chloride Channel Anoctamin 1 (TMEM16A). Handb Exp Pharmacol 2024; 283:101-151. [PMID: 35768554 DOI: 10.1007/164_2022_592] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Various human tissues express the calcium-activated chloride channel Anoctamin 1 (ANO1), also known as TMEM16A. ANO1 allows the passive chloride flux that controls different physiological functions ranging from muscle contraction, fluid and hormone secretion, gastrointestinal motility, and electrical excitability. Overexpression of ANO1 is associated with pathological conditions such as hypertension and cancer. The molecular cloning of ANO1 has led to a surge in structural, functional, and physiological studies of the channel in several tissues. ANO1 is a homodimer channel harboring two pores - one in each monomer - that work independently. Each pore is activated by voltage-dependent binding of two intracellular calcium ions to a high-affinity-binding site. In addition, the binding of phosphatidylinositol 4,5-bisphosphate to sites scattered throughout the cytosolic side of the protein aids the calcium activation process. Furthermore, many pharmacological studies have established ANO1 as a target of promising compounds that could treat several illnesses. This chapter describes our current understanding of the physiological roles of ANO1 and its regulation under physiological conditions as well as new pharmacological compounds with potential therapeutic applications.
Collapse
Affiliation(s)
- Jorge Arreola
- Physics Institute, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico.
| | - Patricia Pérez-Cornejo
- Department of Physiology and Biophysics, School of Medicine of Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | - Guadalupe Segura-Covarrubias
- Physics Institute, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, OH, USA
| | - Nancy Corral-Fernández
- Department of Physiology and Biophysics, School of Medicine of Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | - Daniel León-Aparicio
- Physics Institute, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | | |
Collapse
|
5
|
Yoshie S, Murono S, Hazama A. Approach for Elucidating the Molecular Mechanism of Epithelial to Mesenchymal Transition in Fibrosis of Asthmatic Airway Remodeling Focusing on Cl - Channels. Int J Mol Sci 2023; 25:289. [PMID: 38203460 PMCID: PMC10779031 DOI: 10.3390/ijms25010289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/15/2023] [Accepted: 12/16/2023] [Indexed: 01/12/2024] Open
Abstract
Airway remodeling caused by asthma is characterized by structural changes of subepithelial fibrosis, goblet cell metaplasia, submucosal gland hyperplasia, smooth muscle cell hyperplasia, and angiogenesis, leading to symptoms such as dyspnea, which cause marked quality of life deterioration. In particular, fibrosis exacerbated by asthma progression is reportedly mediated by epithelial-mesenchymal transition (EMT). It is well known that the molecular mechanism of EMT in fibrosis of asthmatic airway remodeling is closely associated with several signaling pathways, including the TGF-β1/Smad, TGF-β1/non-Smad, and Wnt/β-catenin signaling pathways. However, the molecular mechanism of EMT in fibrosis of asthmatic airway remodeling has not yet been fully clarified. Given that Cl- transport through Cl- channels causes passive water flow and consequent changes in cell volume, these channels may be considered to play a key role in EMT, which is characterized by significant morphological changes. In the present article, we highlight how EMT, which causes fibrosis and carcinogenesis in various tissues, is strongly associated with activation or inactivation of Cl- channels and discuss whether Cl- channels can lead to elucidation of the molecular mechanism of EMT in fibrosis of asthmatic airway remodeling.
Collapse
Affiliation(s)
- Susumu Yoshie
- Department of Cellular and Integrative Physiology, Graduate School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Shigeyuki Murono
- Department of Otolaryngology Head and Neck Surgery, Graduate School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Akihiro Hazama
- Department of Cellular and Integrative Physiology, Graduate School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
| |
Collapse
|
6
|
Wang Z, Choi K. Pharmacological modulation of chloride channels as a therapeutic strategy for neurological disorders. Front Physiol 2023; 14:1122444. [PMID: 36935741 PMCID: PMC10017882 DOI: 10.3389/fphys.2023.1122444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 02/09/2023] [Indexed: 03/06/2023] Open
Abstract
Chloride homeostasis is critical in the physiological functions of the central nervous system (CNS). Its concentration is precisely regulated by multiple ion-transporting proteins such as chloride channels and transporters that are widely distributed in the brain cells, including neurons and glia. Unlike ion transporters, chloride channels provide rapid responses to efficiently regulate ion flux. Some of chloride channels are also permeable to selected organic anions such as glutamate and γ-aminobutyric acid, suggesting neuroexcitatory and neuroinhibitory functions while gating. Dysregulated chloride channels are implicated in neurological disorders, e.g., ischemia and neuroinflammation. Modulation of chloride homeostasis through chloride channels has been suggested as a potential therapeutic approach for neurological disorders. The drug design for CNS diseases is challenging because it requires the therapeutics to traverse the blood-brain-barrier. Small molecules are a well-established modality with better cell permeability due to their lower molecular weight and flexibility for structure optimization compared to biologics. In this article, we describe the important roles of chloride homeostasis in each type of brain cells and introduce selected chloride channels identified in the CNS. We then discuss the contribution of their dysregulations towards the pathogenesis of neurological disorders, emphasizing the potential of targeting chloride channels as a therapeutic strategy for CNS disease treatment. Along with this literature survey, we summarize the small molecules that modulate chloride channels and propose the potential strategy of optimizing existing drugs to brain-penetrants to support future CNS drug discovery.
Collapse
|
7
|
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
|
8
|
Interactions between 14-3-3 Proteins and Actin Cytoskeleton and Its Regulation by microRNAs and Long Non-Coding RNAs in Cancer. ENDOCRINES 2022. [DOI: 10.3390/endocrines3040057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
14-3-3s are a family of structurally similar proteins that bind to phosphoserine or phosphothreonine residues, forming the central signaling hub that coordinates or integrates various cellular functions, thereby controlling many pathways important in cancer, cell motility, cell death, cytoskeletal remodeling, neuro-degenerative disorders and many more. Their targets are present in all cellular compartments, and when they bind to proteins they alter their subcellular localization, stability, and molecular interactions with other proteins. Changes in environmental conditions that result in altered homeostasis trigger the interaction between 14-3-3 and other proteins to retrieve or rescue homeostasis. In circumstances where these regulatory proteins are dysregulated, it leads to pathological conditions. Therefore, deeper understanding is needed on how 14-3-3 proteins bind, and how these proteins are regulated or modified. This will help to detect disease in early stages or design inhibitors to block certain pathways. Recently, more research has been devoted to identifying the role of MicroRNAs, and long non-coding RNAs, which play an important role in regulating gene expression. Although there are many reviews on the role of 14-3-3 proteins in cancer, they do not provide a holistic view of the changes in the cell, which is the focus of this review. The unique feature of the review is that it not only focuses on how the 14-3-3 subunits associate and dissociate with their binding and regulatory proteins, but also includes the role of micro-RNAs and long non-coding RNAs and how they regulate 14-3-3 isoforms. The highlight of the review is that it focuses on the role of 14-3-3, actin, actin binding proteins and Rho GTPases in cancer, and how this complex is important for cell migration and invasion. Finally, the reader is provided with super-resolution high-clarity images of each subunit of the 14-3-3 protein family, further depicting their distribution in HeLa cells to illustrate their interactions in a cancer cell.
Collapse
|
9
|
Guo S, Zhang L, Li N. ANO1: More Than Just Calcium-Activated Chloride Channel in Cancer. Front Oncol 2022; 12:922838. [PMID: 35734591 PMCID: PMC9207239 DOI: 10.3389/fonc.2022.922838] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 05/09/2022] [Indexed: 11/22/2022] Open
Abstract
ANO1, a calcium-activated chloride channel (CACC), is also known as transmembrane protein 16A (TMEM16A). It plays a vital role in the occurrence, development, metastasis, proliferation, and apoptosis of various malignant tumors. This article reviews the mechanism of ANO1 involved in the replication, proliferation, invasion and apoptosis of various malignant tumors. Various molecules and Stimuli control the expression of ANO1, and the regulatory mechanism of ANO1 is different in tumor cells. To explore the mechanism of ANO1 overexpression and activation of tumor cells by studying the different effects of ANO1. Current studies have shown that ANO1 expression is controlled by 11q13 gene amplification and may also exert cell-specific effects through its interconnected protein network, phosphorylation of different kinases, and signaling pathways. At the same time, ANO1 also resists tumor apoptosis and promotes tumor immune escape. ANO1 can be used as a promising biomarker for detecting certain malignant tumors. Further studies on the channels and the mechanism of protein activity of ANO1 are needed. Finally, the latest inhibitors of ANO1 are summarized, which provides the research direction for the tumor-promoting mechanism of ANO1.
Collapse
Affiliation(s)
- Saisai Guo
- Department of Oncology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Linna Zhang
- Department of Oncology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Na Li
- Department of Oncology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
10
|
Li H, Yu Z, Wang H, Wang N, Sun X, Yang S, Hua X, Liu Z. Role of ANO1 in tumors and tumor immunity. J Cancer Res Clin Oncol 2022; 148:2045-2068. [PMID: 35471604 DOI: 10.1007/s00432-022-04004-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 03/29/2022] [Indexed: 12/24/2022]
Abstract
Dysregulation of gene amplification, cell-signaling-pathway transduction, epigenetic and transcriptional regulation, and protein interactions drives tumor-cell proliferation and invasion, while ion channels also play an important role in the generation and development of tumor cells. Overexpression of Ca2+-activated Cl- channel anoctamin 1 (ANO1) is shown in numerous cancer types and correlates with poor prognosis. However, the mechanisms involved in ANO1-mediated malignant cellular transformation and the role of ANO1 in tumor immunity remain unknown. In this review, we discuss recent studies to determine the role of ANO1 in tumorigenesis and provide novel insights into the role of ANO1 in the context of tumor immunity. Furthermore, we analyze the roles and potential mechanisms of ANO1 in different types of cancers, and provide novel notions for the role of ANO1 in the tumor microenvironment and for potential use of ANO1 in clinical applications. Our review shows that ANO1 is involved in tumor immunity and microenvironment, and may, therefore, be an effective biomarker and therapeutic drug target.
Collapse
Affiliation(s)
- Haini Li
- Department of Gastroenterology, Qingdao Sixth People's Hospital, Qingdao, 266001, China
| | - Zongxue Yu
- Department of Endocrinology, Affiliated Qingdao Third People's Hospital, Qingdao University, Qingdao, 266001, China
| | - Haiyan Wang
- Department of Clinical Laboratory, Affiliated Qingdao Third People's Hospital, Qingdao University, Qingdao, 266021, China
| | - Ning Wang
- Department of Clinical Laboratory, Affiliated Qingdao Third People's Hospital, Qingdao University, Qingdao, 266021, China
| | - Xueguo Sun
- Department of Gastroenterology, Qingdao University Affiliated Hospital, Qingdao, 266001, China
| | - Shengmei Yang
- Department of Gynecology, Qingdao University Affiliated Hospital, Qingdao, 266001, China
| | - Xu Hua
- Department of Clinical Laboratory, Affiliated Qingdao Third People's Hospital, Qingdao University, Qingdao, 266021, China
| | - Zongtao Liu
- Department of Clinical Laboratory, Affiliated Qingdao Third People's Hospital, Qingdao University, Qingdao, 266021, China.
| |
Collapse
|
11
|
Lee YS, Kwon O, Jeong GR, Noh J, Kim SE, Yi GS, Hwang EM, Park JY. Deficiency of TTYH1 Expression Reduces the Migration and Invasion of U2OS Human Osteosarcoma Cells. Life (Basel) 2022; 12:life12040530. [PMID: 35455021 PMCID: PMC9032734 DOI: 10.3390/life12040530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 03/31/2022] [Accepted: 04/01/2022] [Indexed: 11/29/2022] Open
Abstract
The Tweety homolog (TTYH) chloride channel family is involved in oncogenic processes including cell proliferation, invasion, and colonization of cancers. Among the TTYH family, TTYH1 is highly expressed in several cancer cells, such as glioma, breast, and gastric cancer cells. However, the role of TTYH1 in the progression of osteosarcoma remains unknown. Here, we report that deficient TTYH1 expression results in the inhibition of the migration and invasion of U2OS human osteosarcoma cells. We found that TTYH1 was endogenously expressed at both mRNA and protein levels in U2OS cells and that these channels were located at the plasma membrane of the cells. Moreover, we found that silencing of the TTYH1 with small interfering RNA (siRNA) resulted in a decrease in the migration and invasion of U2OS cells, while the proliferation of the cells was not affected. Additionally, treatment with TTYH1 siRNA significantly suppressed the mRNA expression of epithelial−mesenchymal transition (EMT)-regulated transcription factors such as Zinc E-Box Binding Homeobox 1 (ZEB1) and SNAIL. Most importantly, the expression of matrix metalloproteinase (MMP)-2, MPP-9, and N-cadherin was dramatically reduced following the silencing of TTYH1. Taken together, our findings suggest that silencing of TTYH1 expression reduces migration and invasion of U2OS cells and that TTYH1 may act as a potential molecular target for osteosarcoma treatment.
Collapse
Affiliation(s)
- Young-Sun Lee
- School of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul 02841, Korea; (Y.-S.L.); (O.K.); (G.-R.J.); (J.N.); (S.E.K.)
- Center for Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea;
| | - Osung Kwon
- School of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul 02841, Korea; (Y.-S.L.); (O.K.); (G.-R.J.); (J.N.); (S.E.K.)
| | - Geuk-Rae Jeong
- School of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul 02841, Korea; (Y.-S.L.); (O.K.); (G.-R.J.); (J.N.); (S.E.K.)
| | - Junyeol Noh
- School of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul 02841, Korea; (Y.-S.L.); (O.K.); (G.-R.J.); (J.N.); (S.E.K.)
| | - Sung Eun Kim
- School of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul 02841, Korea; (Y.-S.L.); (O.K.); (G.-R.J.); (J.N.); (S.E.K.)
| | - Gwan-Su Yi
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea;
| | - Eun Mi Hwang
- Center for Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea;
| | - Jae-Yong Park
- School of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul 02841, Korea; (Y.-S.L.); (O.K.); (G.-R.J.); (J.N.); (S.E.K.)
- Correspondence:
| |
Collapse
|
12
|
Caratti B, Fidan M, Caratti G, Breitenecker K, Engler M, Kazemitash N, Traut R, Wittig R, Casanova E, Ahmadian MR, Tuckermann JP, Moll HP, Cirstea IC. The glucocorticoid receptor associates with RAS complexes to inhibit cell proliferation and tumor growth. Sci Signal 2022; 15:eabm4452. [PMID: 35316097 DOI: 10.1126/scisignal.abm4452] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Mutations that activate members of the RAS family of GTPases are associated with various cancers and drive tumor growth. The glucocorticoid receptor (GR), a member of the nuclear receptor family, has been proposed to interact with and inhibit the activation of components of the PI3K-AKT and MAPK pathways downstream of RAS. In the absence of activating ligands, we found that GR was present in cytoplasmic KRAS-containing complexes and inhibited the activation of wild-type and oncogenic KRAS in mouse embryonic fibroblasts and human lung cancer A549 cells. The DNA binding domain of GR was involved in the interaction with KRAS, but GR-dependent inhibition of RAS activation did not depend on the nuclear translocation of GR. The addition of ligand released GR-dependent inhibition of RAS, AKT, the MAPK p38, and the MAPKK MEK. CRISPR-Cas9-mediated deletion of GR in A549 cells enhanced tumor growth in xenografts in mice. Patient samples of non-small cell lung carcinomas showed lower expression of NR3C1, the gene encoding GR, compared to adjacent normal tissues and lower NR3C1 expression correlated with a worse disease outcome. These results suggest that glucocorticoids prevent the ability of GR to limit tumor growth by inhibiting RAS activation, which has potential implications for the use of glucocorticoids in patients with cancer.
Collapse
Affiliation(s)
- Bozhena Caratti
- Institute of Comparative Molecular Endocrinology, University of Ulm, Helmholtzstrasse 8/1, 89081 Ulm, Germany
| | - Miray Fidan
- Institute of Comparative Molecular Endocrinology, University of Ulm, Helmholtzstrasse 8/1, 89081 Ulm, Germany
| | - Giorgio Caratti
- Institute of Comparative Molecular Endocrinology, University of Ulm, Helmholtzstrasse 8/1, 89081 Ulm, Germany
| | - Kristina Breitenecker
- Department of Pharmacology, Center of Physiology and Pharmacology, Comprehensive Cancer Center (CCC), Medical University of Vienna, Vienna 1090, Austria
| | - Melanie Engler
- Institute of Comparative Molecular Endocrinology, University of Ulm, Helmholtzstrasse 8/1, 89081 Ulm, Germany
| | - Naser Kazemitash
- Institute of Comparative Molecular Endocrinology, University of Ulm, Helmholtzstrasse 8/1, 89081 Ulm, Germany
| | - Rebecca Traut
- Institute of Comparative Molecular Endocrinology, University of Ulm, Helmholtzstrasse 8/1, 89081 Ulm, Germany
| | - Rainer Wittig
- Institute for Laser Technologies in Medicine and Metrology (ILM), University of Ulm, Helmholtzstrasse 12, 89081 Ulm, Germany
| | - Emilio Casanova
- Department of Pharmacology, Center of Physiology and Pharmacology, Comprehensive Cancer Center (CCC), Medical University of Vienna, Vienna 1090, Austria
| | - Mohammad Reza Ahmadian
- Institute of Biochemistry and Molecular Biology II, Medical Faculty of the Heinrich Heine University, Universitätsstraße 1, Building 22.03.05, 40225 Düsseldorf, Germany
| | - Jan P Tuckermann
- Institute of Comparative Molecular Endocrinology, University of Ulm, Helmholtzstrasse 8/1, 89081 Ulm, Germany
| | - Herwig P Moll
- Department of Pharmacology, Center of Physiology and Pharmacology, Comprehensive Cancer Center (CCC), Medical University of Vienna, Vienna 1090, Austria
| | - Ion Cristian Cirstea
- Institute of Comparative Molecular Endocrinology, University of Ulm, Helmholtzstrasse 8/1, 89081 Ulm, Germany
| |
Collapse
|
13
|
Chloride Channels and Transporters: Roles beyond Classical Cellular Homeostatic pH or Ion Balance in Cancers. Cancers (Basel) 2022; 14:cancers14040856. [PMID: 35205604 PMCID: PMC8870652 DOI: 10.3390/cancers14040856] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/28/2022] [Accepted: 02/06/2022] [Indexed: 01/04/2023] Open
Abstract
Simple Summary Roles of chloride-associated transporters have been raised in various cancers. Although complicated ion movements, crosstalk among channels/transporters through homeostatic electric regulation, difficulties with experimental implementation such as activity measurement of intracellular location were disturbed to verify the precise modulation of channels/transporters, recently defined cancerous function and communication with tumor microenvironment of chloride channels/transporters should be highlighted beyond classical homeostatic ion balance. Chloride-associated transporters as membrane-associated components of chloride movement, regulations of transmembrane member 16A, calcium-activated chloride channel regulators, transmembrane member 206, chloride intracellular channels, voltage-gated chloride channels, cystic fibrosis transmembrane conductance regulator, voltage-dependent anion channel, volume-regulated anion channel, and chloride-bicarbonate exchangers are discussed. Abstract The canonical roles of chloride channels and chloride-associated transporters have been physiologically determined; these roles include the maintenance of membrane potential, pH balance, and volume regulation and subsequent cellular functions such as autophagy and cellular proliferative processes. However, chloride channels/transporters also play other roles, beyond these classical function, in cancerous tissues and under specific conditions. Here, we focused on the chloride channel-associated cancers and present recent advances in understanding the environments of various types of cancer caused by the participation of many chloride channel or transporters families and discuss the challenges and potential targets for cancer treatment. The modulation of chloride channels/transporters might promote new aspect of cancer treatment strategies.
Collapse
|
14
|
Pulzová LB, Roška J, Kalman M, Kliment J, Slávik P, Smolková B, Goffa E, Jurkovičová D, Kulcsár Ľ, Lešková K, Bujdák P, Mego M, Bhide MR, Plank L, Chovanec M. Screening for the Key Proteins Associated with Rete Testis Invasion in Clinical Stage I Seminoma via Label-Free Quantitative Mass Spectrometry. Cancers (Basel) 2021; 13:cancers13215573. [PMID: 34771736 PMCID: PMC8583098 DOI: 10.3390/cancers13215573] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 10/11/2021] [Accepted: 10/13/2021] [Indexed: 12/12/2022] Open
Abstract
Rete testis invasion (RTI) is an unfavourable prognostic factor for the risk of relapse in clinical stage I (CS I) seminoma patients. Notably, no evidence of difference in the proteome of RTI-positive vs. -negative CS I seminomas has been reported yet. Here, a quantitative proteomic approach was used to investigate RTI-associated proteins. 64 proteins were differentially expressed in RTI-positive compared to -negative CS I seminomas. Of them, 14-3-3γ, ezrin, filamin A, Parkinsonism-associated deglycase 7 (PARK7), vimentin and vinculin, were validated in CS I seminoma patient cohort. As shown by multivariate analysis controlling for clinical confounders, PARK7 and filamin A expression lowered the risk of RTI, while 14-3-3γ expression increased it. Therefore, we suggest that in real clinical biopsy specimens, the expression level of these proteins may reflect prognosis in CS I seminoma patients.
Collapse
Affiliation(s)
- Lucia Borszéková Pulzová
- Biomedical Research Center, Department of Genetics, Cancer Research Institute, Slovak Academy of Sciences, Dúbravská cesta 9, 845 05 Bratislava, Slovakia; (L.B.P.); (J.R.); (E.G.); (D.J.); (Ľ.K.); (M.M.)
| | - Jan Roška
- Biomedical Research Center, Department of Genetics, Cancer Research Institute, Slovak Academy of Sciences, Dúbravská cesta 9, 845 05 Bratislava, Slovakia; (L.B.P.); (J.R.); (E.G.); (D.J.); (Ľ.K.); (M.M.)
| | - Michal Kalman
- Department of Pathological Anatomy, Jessenius Faculty of Medicine and University Hospital in Martin, Comenius University, Malá Hora 4A, 036 01 Martin, Slovakia; (M.K.); (P.S.); (K.L.); (L.P.)
| | - Ján Kliment
- Clinic of Urology, Jessenius Faculty of Medicine and University Hospital in Martin, Comenius University, Malá Hora 4A, 036 01 Martin, Slovakia;
| | - Pavol Slávik
- Department of Pathological Anatomy, Jessenius Faculty of Medicine and University Hospital in Martin, Comenius University, Malá Hora 4A, 036 01 Martin, Slovakia; (M.K.); (P.S.); (K.L.); (L.P.)
| | - Božena Smolková
- Biomedical Research Center, Department of Molecular Oncology, Cancer Research Institute, Slovak Academy of Sciences, Dúbravská cesta 9, 845 05 Bratislava, Slovakia;
| | - Eduard Goffa
- Biomedical Research Center, Department of Genetics, Cancer Research Institute, Slovak Academy of Sciences, Dúbravská cesta 9, 845 05 Bratislava, Slovakia; (L.B.P.); (J.R.); (E.G.); (D.J.); (Ľ.K.); (M.M.)
| | - Dana Jurkovičová
- Biomedical Research Center, Department of Genetics, Cancer Research Institute, Slovak Academy of Sciences, Dúbravská cesta 9, 845 05 Bratislava, Slovakia; (L.B.P.); (J.R.); (E.G.); (D.J.); (Ľ.K.); (M.M.)
| | - Ľudovít Kulcsár
- Biomedical Research Center, Department of Genetics, Cancer Research Institute, Slovak Academy of Sciences, Dúbravská cesta 9, 845 05 Bratislava, Slovakia; (L.B.P.); (J.R.); (E.G.); (D.J.); (Ľ.K.); (M.M.)
| | - Katarína Lešková
- Department of Pathological Anatomy, Jessenius Faculty of Medicine and University Hospital in Martin, Comenius University, Malá Hora 4A, 036 01 Martin, Slovakia; (M.K.); (P.S.); (K.L.); (L.P.)
| | - Peter Bujdák
- Department of Urology, Faculty of Medicine, Comenius University, 813 72 Bratislava, Slovakia;
| | - Michal Mego
- Biomedical Research Center, Department of Genetics, Cancer Research Institute, Slovak Academy of Sciences, Dúbravská cesta 9, 845 05 Bratislava, Slovakia; (L.B.P.); (J.R.); (E.G.); (D.J.); (Ľ.K.); (M.M.)
- 2nd Department of Oncology, Faculty of Medicine, Comenius University and National Cancer Institute, Klenová 1, 833 10 Bratislava, Slovakia
| | - Mangesh R. Bhide
- Department of Microbiology and Immunology, University of Veterinary Medicine, Komenského 73, 041 81 Košice, Slovakia;
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dúbravská cesta 9, 845 05 Bratislava, Slovakia
| | - Lukáš Plank
- Department of Pathological Anatomy, Jessenius Faculty of Medicine and University Hospital in Martin, Comenius University, Malá Hora 4A, 036 01 Martin, Slovakia; (M.K.); (P.S.); (K.L.); (L.P.)
| | - Miroslav Chovanec
- Biomedical Research Center, Department of Genetics, Cancer Research Institute, Slovak Academy of Sciences, Dúbravská cesta 9, 845 05 Bratislava, Slovakia; (L.B.P.); (J.R.); (E.G.); (D.J.); (Ľ.K.); (M.M.)
- Correspondence:
| |
Collapse
|
15
|
Sharma A, Ramena GT, Elble RC. Advances in Intracellular Calcium Signaling Reveal Untapped Targets for Cancer Therapy. Biomedicines 2021; 9:1077. [PMID: 34572262 PMCID: PMC8466575 DOI: 10.3390/biomedicines9091077] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/15/2021] [Accepted: 07/18/2021] [Indexed: 02/07/2023] Open
Abstract
Intracellular Ca2+ distribution is a tightly regulated process. Numerous Ca2+ chelating, storage, and transport mechanisms are required to maintain normal cellular physiology. Ca2+-binding proteins, mainly calmodulin and calbindins, sequester free intracellular Ca2+ ions and apportion or transport them to signaling hubs needing the cations. Ca2+ channels, ATP-driven pumps, and exchangers assist the binding proteins in transferring the ions to and from appropriate cellular compartments. Some, such as the endoplasmic reticulum, mitochondria, and lysosomes, act as Ca2+ repositories. Cellular Ca2+ homeostasis is inefficient without the active contribution of these organelles. Moreover, certain key cellular processes also rely on inter-organellar Ca2+ signaling. This review attempts to encapsulate the structure, function, and regulation of major intracellular Ca2+ buffers, sensors, channels, and signaling molecules before highlighting how cancer cells manipulate them to survive and thrive. The spotlight is then shifted to the slow pace of translating such research findings into anticancer therapeutics. We use the PubMed database to highlight current clinical studies that target intracellular Ca2+ signaling. Drug repurposing and improving the delivery of small molecule therapeutics are further discussed as promising strategies for speeding therapeutic development in this area.
Collapse
Affiliation(s)
- Aarushi Sharma
- Department of Pharmacology and Simmons Cancer Institute, Southern Illinois University School of Medicine, Springfield, IL 62702, USA;
| | - Grace T. Ramena
- Department of Aquaculture, University of Arkansas, Pine Bluff, AR 71601, USA;
| | - Randolph C. Elble
- Department of Pharmacology and Simmons Cancer Institute, Southern Illinois University School of Medicine, Springfield, IL 62702, USA;
| |
Collapse
|
16
|
Li H, Yang Q, Huo S, Du Z, Wu F, Zhao H, Chen S, Yang L, Ma Z, Sui Y. Expression of TMEM16A in Colorectal Cancer and Its Correlation With Clinical and Pathological Parameters. Front Oncol 2021; 11:652262. [PMID: 33816307 PMCID: PMC8017291 DOI: 10.3389/fonc.2021.652262] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 02/22/2021] [Indexed: 12/12/2022] Open
Abstract
TMEM16A is a recently identified calcium-activated chloride channel (CaCC) and its overexpression contributes to tumorigenesis and progression in several human malignancies. However, little is known about expression of TMEM16A and its clinical significance in colorectal cancer (CRC). TMEM16A mRNA expression was determined by quantitative real time-PCR (qRT-PCR) in 67 CRC tissues and 24 para-carcinoma tissues. TMEM16A protein expression was performed by immunohistochemistry in 80 CRC tissues. The correlation between TMEM16A expression and clinicopathological parameters, and known genes and proteins involved in CRC was analyzed. The results showed that TMEM16A mRNA expression was frequently detected in 51 CRC tissues (76%), whereas TMEM16A protein expression was determined at a relatively lower frequency (26%). TMEM16A mRNA expression in tumor tissues was higher than its expression in normal para-carcinoma tissues (P < 0.05). TMEM16A mRNA expression was significantly correlated with TNM stage (p = 0.039) and status of lymph node metastasis (p = 0.047). In addition, there was a strong positive correlation between TMEM16A mRNA expression and MSH2 protein. More importantly, TMEM16A protein expression was positively associated with KRAS mutation, and negatively correlated with mutant p53 protein. Logistic regression analysis demonstrated that TMEM16A mRNA expression was an important independent predictive factor of lymph node metastasis (OR = 16.38, CI: 1.91-140.27, p = 0.01). TMEM16A mRNA and protein expression was not significantly related with patient survival. Our findings provide original evidence demonstrating TMEM16A mRNA expression can be a novel predictive marker of lymph node metastasis and TMEM16A protein expression may be an important regulator of tumor proliferation and metastasis in CRC.
Collapse
Affiliation(s)
- Hongxia Li
- Department of Dermatology, First Hospital of Jilin University, Changchun, China
| | - Qiwei Yang
- Key Laboratory for Molecular and Chemical Genetics of Critical Human Diseases of Jilin Province, Second Hospital of Jilin University, Changchun, China
| | - Sibo Huo
- Department of Gastrointestinal Nutrition and Hernia Surgery, Second Hospital of Jilin University, Changchun, China
- Department of General Surgery, Qian Wei Hospital of Jilin Province, Changchun, China
| | - Zhenwu Du
- Key Laboratory for Molecular and Chemical Genetics of Critical Human Diseases of Jilin Province, Second Hospital of Jilin University, Changchun, China
- Department of Orthopedics, Second Hospital of Jilin University, Changchun, China
| | - Fei Wu
- Department of Gynecology and Obstetrics, Second Hospital of Jilin University, Changchun, China
| | - Haiyue Zhao
- Center of Reproductive Medicine and Center of Prenatal Diagnosis, First Hospital of Jilin University, Changchun, China
| | - Shifan Chen
- Department of Pathology, Second Hospital of Jilin University, Changchun, China
| | - Longfei Yang
- Key Laboratory for Molecular and Chemical Genetics of Critical Human Diseases of Jilin Province, Second Hospital of Jilin University, Changchun, China
| | - Zhiming Ma
- Department of Gastrointestinal Nutrition and Hernia Surgery, Second Hospital of Jilin University, Changchun, China
| | - Yujie Sui
- Key Laboratory for Molecular and Chemical Genetics of Critical Human Diseases of Jilin Province, Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
17
|
The Groovy TMEM16 Family: Molecular Mechanisms of Lipid Scrambling and Ion Conduction. J Mol Biol 2021; 433:166941. [PMID: 33741412 DOI: 10.1016/j.jmb.2021.166941] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/09/2021] [Accepted: 03/09/2021] [Indexed: 12/28/2022]
Abstract
The TMEM16 family of membrane proteins displays a remarkable functional dichotomy - while some family members function as Ca2+-activated anion channels, the majority of characterized TMEM16 homologs are Ca2+-activated lipid scramblases, which catalyze the exchange of phospholipids between the two membrane leaflets. Furthermore, some TMEM16 scramblases can also function as channels. Due to their involvement in important physiological processes, the family has been actively studied ever since their molecular identity was unraveled. In this review, we will summarize the recent advances in the field and how they influenced our view of TMEM16 family function and evolution. Structural, functional and computational studies reveal how relatively small rearrangements in the permeation pathway are responsible for the observed functional duality: while TMEM16 scramblases can adopt both ion- and lipid conductive conformations, TMEM16 channels can only populate the former. Recent data further provides the molecular details of a stepwise activation mechanism, which is initiated by Ca2+ binding and modulated by various cellular factors, including lipids. TMEM16 function and the surrounding membrane properties are inextricably intertwined, with the protein inducing bilayer deformations associated with scrambling, while the surrounding lipids modulate TMEM16 conformation and activity.
Collapse
|
18
|
Filippou A, Pehkonen H, Karhemo PR, Väänänen J, Nieminen AI, Klefström J, Grénman R, Mäkitie AA, Joensuu H, Monni O. ANO1 Expression Orchestrates p27Kip1/MCL1-Mediated Signaling in Head and Neck Squamous Cell Carcinoma. Cancers (Basel) 2021; 13:cancers13051170. [PMID: 33803266 PMCID: PMC7967175 DOI: 10.3390/cancers13051170] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/01/2021] [Accepted: 03/05/2021] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Our aim was to elucidate the molecular mechanisms of how ANO1 contributes to oncogenic processes in squamous cell carcinoma of the head and neck (HNSCC). We explored transcriptional programs influenced by ANO1 knockdown in patient-derived UT-SCC cell lines with 11q13 amplification and ANO1 overexpression. ANO1 depletion led to downregulation of broad pro-survival BCL2 family protein members, including MCL1, and simultaneously induced upregulation of the cell cycle inhibitor p27Kip1 and its redistribution from the cytoplasm into the nucleus in the studied HNSCC cells. Gene set enrichment analysis highlighted pathways associated with perturbed cell cycle and apoptosis in the ANO1-depleted samples. Silencing of ANO1 and application of an ANO1-targeting small-molecule inhibitor led to ANO1 degradation and reduction of cell viability. These findings suggest that ANO1 has drug target potential that deserves further evaluation in preclinical in vivo models. Abstract Head and neck squamous cell carcinoma (HNSCC) is a heterogeneous group of tumors that derive from the mucosal epithelium of the upper aerodigestive tract and present high mortality rate. Lack of efficient targeted-therapies and biomarkers towards patients’ stratification are caveats in the disease treatment. Anoctamin 1 (ANO1) gene is amplified in 30% of HNSCC cases. Evidence suggests involvement of ANO1 in proliferation, migration, and evasion of apoptosis; however, the exact mechanisms remain elusive. Aim of this study was to unravel the ANO1-dependent transcriptional programs and expand the existing knowledge of ANO1 contribution to oncogenesis and drug response in HNSCC. We cultured two HNSCC cell lines established from primary tumors harboring amplification and high expression of ANO1 in three-dimensional collagen. Differential expression analysis of ANO1-depleted HNSCC cells demonstrated downregulation of MCL1 and simultaneous upregulation of p27Kip1 expression. Suppressing ANO1 expression led to redistribution of p27Kip1 from the cytoplasm to the nucleus and associated with a cell cycle arrested phenotype. ANO1 silencing or pharmacological inhibition resulted in reduction of cell viability and ANO1 protein levels, as well as suppression of pro-survival BCL2 family proteins. Collectively, these data provide insights of ANO1 involvement in HNSCC carcinogenesis and support the rationale that ANO1 is an actionable drug target.
Collapse
Affiliation(s)
- Artemis Filippou
- Applied Tumor Genomics Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland; (A.F.); (H.P.); (P.-R.K.); (J.V.)
| | - Henna Pehkonen
- Applied Tumor Genomics Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland; (A.F.); (H.P.); (P.-R.K.); (J.V.)
| | - Piia-Riitta Karhemo
- Applied Tumor Genomics Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland; (A.F.); (H.P.); (P.-R.K.); (J.V.)
| | - Juho Väänänen
- Applied Tumor Genomics Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland; (A.F.); (H.P.); (P.-R.K.); (J.V.)
| | - Anni I. Nieminen
- Translational Cancer Medicine Research Program and Medicum, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland;
| | - Juha Klefström
- Finnish Cancer Institute, FICAN South Helsinki University Hospital, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland;
| | - Reidar Grénman
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Turku and Turku University Hospital, 20520 Turku, Finland;
| | - Antti A. Mäkitie
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Helsinki and Helsinki University Hospital, 00130 Helsinki, Finland;
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
| | - Heikki Joensuu
- Department of Oncology, University of Helsinki and Helsinki University Hospital, 00290 Helsinki, Finland;
| | - Outi Monni
- Applied Tumor Genomics Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland; (A.F.); (H.P.); (P.-R.K.); (J.V.)
- Department of Oncology, Clinicum, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
- Correspondence: ; Tel.: +358-407639302
| |
Collapse
|
19
|
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
|
20
|
ANO1 regulates the maintenance of stemness in glioblastoma stem cells by stabilizing EGFRvIII. Oncogene 2021; 40:1490-1502. [PMID: 33452454 DOI: 10.1038/s41388-020-01612-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 11/29/2020] [Accepted: 12/10/2020] [Indexed: 01/27/2023]
Abstract
Glioblastoma multiforme (GBM) or glioblastoma is the most deadly malignant brain tumor in adults. GBM is difficult to treat mainly due to the presence of glioblastoma stem cells (GSCs). Epidermal growth factor receptor variant III (EGFRvIII) has been linked to stemness and malignancy of GSCs; however, the regulatory mechanism of EGFRvIII is largely unknown. Here, we demonstrated that Anoctamin-1 (ANO1), a Ca2+-activated Cl- channel, interacts with EGFRvIII, increases its protein stability, and supports the maintenance of stemness and tumor progression in GSCs. Specifically, shRNA-mediated knockdown and pharmacological inhibition of ANO1 suppressed the self-renewal, invasion activities, and expression of EGFRvIII and related stem cell factors, including NOTCH1, nestin, and SOX2 in GSCs. Conversely, ANO1 overexpression enhanced the above phenomena. Mechanistically, ANO1 protected EGFRvIII from proteasomal degradation by directly binding to it. ANO1 knockdown significantly increased survival in mice and strongly suppressed local invasion of GSCs in an in vivo intracranial mouse model. Collectively, these results suggest that ANO1 plays a crucial role in the maintenance of stemness and invasiveness of GSCs by regulating the expression of EGFRvIII and related signaling molecules, and can be considered a promising therapeutic target for GBM treatment.
Collapse
|
21
|
Choi SH, Ryu S, Sim K, Song C, Shin I, Kim SS, Lee YS, Park JY, Sim T. Anti-glioma effects of 2-aminothiophene-3-carboxamide derivatives, ANO1 channel blockers. Eur J Med Chem 2020; 208:112688. [PMID: 32906067 DOI: 10.1016/j.ejmech.2020.112688] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 07/15/2020] [Accepted: 07/21/2020] [Indexed: 12/22/2022]
Abstract
Anoctamin1 (ANO1), a calcium-activated chloride ion channel (CaCC), is associated with various physiological functions including cancer progression and metastasis/invasion. ANO1 has been considered as a promising target for cancer therapeutics as ANO1 is over-expressed in a variety of cancers including glioblastoma (GBM) and inhibition of ANO1 has been reported to suppress cell proliferation, migration and invasion in GBM. GBM is one of the most common and aggressive cancers with poor prognosis with median survival for 15 months. Lack of effective treatment options against GBM emphasizes urgent necessity of effective GBM therapeutics. In an effort to discover potent and selective ANO1 inhibitors capable of inhibiting GBM cells, we have designed and synthesized a series of new 2-aminothiophene-3-carboxamide derivatives and performed SAR studies using both fluorescent cellular membrane potential assay and whole-cell patch-clamp recording. We observed that among these substances, 9c and 10q strongly suppress ANO1 channel activities and possess remarkable selectivity over ANO2. Unique structural feature of 10q, a cyclopentane-fused thiophene-3-carboxamide derivative, is the presence of benzoylthiourea functionality which dramatically contributes to activity. Both 9c and 10q suppress more strongly proliferation of GBM cells than four reference compounds including 3, Ani-9 and are also capable of inhibiting much more strongly colony formation than reference compounds in both 2D colony formation assay and 3D soft agar assay using U251 glioma cells. In addition, 9c and 10q suppress far more strongly migration/invasion of GBM cells than reference compounds. We, for the first time, found that the combination of ANO1 inhibitor (9c or 3) and temozolomide (TMZ) brings about remarkable synergistic effects in suppressing proliferation of GBM cells. Our study may provide an insight into designing selective and potent ANO1 inhibitors aiming at GBM treatment.
Collapse
Affiliation(s)
- Seung-Hye Choi
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - SeongShick Ryu
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Kyoungmi Sim
- School of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul, 02841, Republic of Korea
| | - Chiman Song
- Chemical Kinomics Research Center, Korea Institute of Science and Technology, 5 Hwarangro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea; Department of Agricultural Biotechnology, College of Agriculture and Life Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Injae Shin
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea; Severance Biomedical Science Institute, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Seong-Seop Kim
- School of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul, 02841, Republic of Korea
| | - Young-Sun Lee
- School of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul, 02841, Republic of Korea
| | - Jae-Yong Park
- School of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul, 02841, Republic of Korea
| | - Taebo Sim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea; Chemical Kinomics Research Center, Korea Institute of Science and Technology, 5 Hwarangro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea; Severance Biomedical Science Institute, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.
| |
Collapse
|
22
|
Dulin NO. Calcium-Activated Chloride Channel ANO1/TMEM16A: Regulation of Expression and Signaling. Front Physiol 2020; 11:590262. [PMID: 33250781 PMCID: PMC7674831 DOI: 10.3389/fphys.2020.590262] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 10/13/2020] [Indexed: 01/11/2023] Open
Abstract
Anoctamin-1 (ANO1), also known as TMEM16A, is the most studied member of anoctamin family of calcium-activated chloride channels with diverse cellular functions. ANO1 controls multiple cell functions including cell proliferation, survival, migration, contraction, secretion, and neuronal excitation. This review summarizes the current knowledge of the cellular mechanisms governing the regulation of ANO1 expression and of ANO1-mediated intracellular signaling. This includes the stimuli and mechanisms controlling ANO1 expression, agonists and processes that activate ANO1, and signal transduction mediated by ANO1. The major conclusion is that this field is poorly understood, remains highly controversial, and requires extensive and rigorous further investigation.
Collapse
Affiliation(s)
- Nickolai O Dulin
- Department of Medicine, The University of Chicago, Chicago, IL, United States
| |
Collapse
|
23
|
Abstract
14-3-3 proteins are mostly expressed in the brain and are closely involved in numerous brain functions and various brain disorders. Among the isotypes of the 14-3-3 proteins, 14-3-3γ is mainly expressed in neurons and is highly produced during brain development, which could indicate that it has a significance in neural development. Furthermore, the distinctive levels of temporally and locally regulated 14-3-3γ expression in various brain disorders suggest that it could play a substantial role in brain plasticity of the diseased states. In this review, we introduce the various brain disorders reported to be involved with 14-3-3γ, and summarize the changes of 14-3-3γ expression in each brain disease. We also discuss the potential of 14-3-3γ for treatment and the importance of research on specific 14-3-3 isotypes for an effective therapeutic approach.
Collapse
Affiliation(s)
- Eunsil Cho
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul 02708, Korea
| | - Jae-Yong Park
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul 02708, Korea
| |
Collapse
|
24
|
Takayasu T, Kurisu K, Esquenazi Y, Ballester LY. Ion Channels and Their Role in the Pathophysiology of Gliomas. Mol Cancer Ther 2020; 19:1959-1969. [PMID: 33008831 PMCID: PMC7577395 DOI: 10.1158/1535-7163.mct-19-0929] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 03/24/2020] [Accepted: 08/06/2020] [Indexed: 01/10/2023]
Abstract
Malignant gliomas are the most common primary central nervous system tumors and their prognosis is very poor. In recent years, ion channels have been demonstrated to play important roles in tumor pathophysiology such as regulation of gene expression, cell migration, and cell proliferation. In this review, we summarize the current knowledge on the role of ion channels on the development and progression of gliomas. Cell volume changes through the regulation of ion flux, accompanied by water flux, are essential for migration and invasion. Signaling pathways affected by ion channel activity play roles in cell survival and cell proliferation. Moreover, ion channels are involved in glioma-related seizures, sensitivity to chemotherapy, and tumor metabolism. Ion channels are potential targets for the treatment of these lethal tumors. Despite our increased understanding of the contributions of ion channels to glioma biology, this field remains poorly studied. This review summarizes the current literature on this important topic.
Collapse
Affiliation(s)
- Takeshi Takayasu
- Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center at Houston, Houston, Texas
- Department of Neurosurgery, Institute of Biomedical and Health Sciences, Hiroshima University, Higashihiroshima, Hiroshima, Japan
| | - Kaoru Kurisu
- Department of Neurosurgery, Institute of Biomedical and Health Sciences, Hiroshima University, Higashihiroshima, Hiroshima, Japan
| | - Yoshua Esquenazi
- Vivian L. Smith Department of Neurosurgery, The University of Texas Health Science Center at Houston, Medical School, Houston, Texas.
- Memorial Hermann Hospital-TMC, Houston, Texas
| | - Leomar Y Ballester
- Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center at Houston, Houston, Texas.
- Vivian L. Smith Department of Neurosurgery, The University of Texas Health Science Center at Houston, Medical School, Houston, Texas
- Memorial Hermann Hospital-TMC, Houston, Texas
| |
Collapse
|
25
|
Anderson G. Glioblastoma chemoresistance: roles of the mitochondrial melatonergic pathway. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2020; 3:334-355. [PMID: 35582450 PMCID: PMC8992488 DOI: 10.20517/cdr.2020.17] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 04/18/2020] [Accepted: 04/24/2020] [Indexed: 12/14/2022]
Abstract
Treatment-resistance is common in glioblastoma (GBM) and the glioblastoma stem-like cells (GSC) from which they arise. Current treatment options are generally regarded as very poor and this arises from a poor conceptualization of the biological underpinnings of GBM/GSC and of the plasticity that these cells are capable of utilizing in response to different treatments. A number of studies indicate melatonin to have utility in the management of GBM/GSC, both per se and when adjunctive to chemotherapy. Recent work shows melatonin to be produced in mitochondria, with the mitochondrial melatonergic pathway proposed to be a crucial factor in driving the wide array of changes in intra- and inter-cellular processes, as well as receptors that can be evident in the cells of the GBM/GSC microenvironment. Variations in the enzymatic conversion of N-acetylserotonin (NAS) to melatonin may be especially important in GSC, as NAS can activate the tyrosine receptor kinase B to increase GSC survival and proliferation. Consequently, variations in the NAS/melatonin ratio may have contrasting effects on GBM/GSC survival. It is proposed that mitochondrial communication across cell types in the tumour microenvironment is strongly driven by the need to carefully control the mitochondrial melatonergic pathways across cell types, with a number of intra- and inter-cellular processes occurring as a consequence of the need to carefully regulate the NAS/melatonin ratio. This better integrates previously disparate data on GBM/GSC as well as providing clear future research and treatment options.
Collapse
Affiliation(s)
- George Anderson
- CRC Scotland & London, Eccleston Square, London SW1V 1PG, UK
| |
Collapse
|
26
|
Suppression of CaMKIIβ Inhibits ANO1-Mediated Glioblastoma Progression. Cells 2020; 9:cells9051079. [PMID: 32357567 PMCID: PMC7290681 DOI: 10.3390/cells9051079] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/22/2020] [Accepted: 04/22/2020] [Indexed: 12/18/2022] Open
Abstract
ANO1, a Ca2+-activated chloride channel, is highly expressed in glioblastoma cells and its surface expression is involved in their migration and invasion. However, the regulation of ANO1 surface expression in glioblastoma cells is largely unknown. In this study, we found that Ca2+/Calmodulin-dependent protein kinase II (CaMKII) β specifically enhances the surface expression and channel activity of ANO1 in U251 glioblastoma cells. When KN-93, a CaMKII inhibitor, was used to treat U251 cells, the surface expression and channel activity of ANO1 were significantly reduced. Only CaMKIIβ, among the four CaMKII isoforms, increased the surface expression and channel activity of ANO1 in a heterologous expression system. Additionally, gene silencing of CaMKIIβ suppressed the surface expression and channel activity of ANO1 in U251 cells. Moreover, gene silencing of CaMKIIβ or ANO1 prominently reduced the migration and invasion of U251 and U87 MG glioblastoma cells. We thus conclude that CaMKIIβ plays a specific role in the surface expression of ANO1 and in the ANO1-mediated tumorigenic properties of glioblastoma cells, such as migration and invasion.
Collapse
|
27
|
Zhang C, Liu J, Han Z, Cui X, Peng D, Xing Y. Inhibition of TMEM16A suppresses growth and induces apoptosis in hepatocellular carcinoma. Int J Clin Oncol 2020; 25:1145-1154. [PMID: 32240440 DOI: 10.1007/s10147-020-01653-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 03/03/2020] [Indexed: 01/24/2023]
Abstract
BACKGROUND Increase of the Ca2+-activated chloride channel TMEM16A is contribute to tumorigenesis. However, the expression level of TMEM16A and its underlying molecular mechanism for TMEM16Apromotingliver carcinogenesis is remains unknown. METHODS In the present study, the expression of TMEM16A in hepatocellular carcinoma (HCC) tissues were measured by quantitative reverse-transcription polymerase chain reaction (qRT-PCR), Western blot and immunohistochemical. Cell proliferation was detected using CCK-8, EdU staining and colony formation methods. Flow cytometry was carried out for detecting cell cycle distribution and apoptosis rate. Migration and invasion abilities were analyzed using transwell and wound healing assay. Western blot method was performed to analyze protein expression. RESULTS Here, we found TMEM16A was significantly increased in HCC tissues, and a higher TMEM16A expression levels were detected in larger tumor size, higher tumor grade, with distant metastasis and poor differentiation. Moreover, overexpression of TMEM16A promoted HCC growth, migration and invasion, and suppressed apoptosis in vitro and in vivo. Knockdown of TMEM16A inhibited HCC growth, migration and invasion, and induced apoptosis in vitro and in vivo. Furthermore, TMEM16A regulated PI3K/AKT-MAKP signaling pathway. CONCLUSION Our data indicate that TMEM16A may represent a novel biomarker of HCC and may be a potential therapeutic target for diagnosis and therapy.
Collapse
Affiliation(s)
- Chuantao Zhang
- Department of Respiration, Hospital of Chengdu university of Traditional Chinese Medicine, Chengdu, 610072, Sichuan, China
| | - Jianxiang Liu
- Key Laboratory of Medicinal Biotechnology, Guilin Medical University, Guilin, 541004, Guangxi, China
| | - Zhiyi Han
- Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital, NO.1, Fuhua Road, Futian District, Shenzhen, 518033, Guangdong, China
| | - Xiang Cui
- Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital, NO.1, Fuhua Road, Futian District, Shenzhen, 518033, Guangdong, China
| | - Deti Peng
- Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital, NO.1, Fuhua Road, Futian District, Shenzhen, 518033, Guangdong, China
| | - Yufeng Xing
- Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital, NO.1, Fuhua Road, Futian District, Shenzhen, 518033, Guangdong, China.
| |
Collapse
|
28
|
Cho C, Lee S, Kim A, Yarishkin O, Ryoo K, Lee Y, Jung H, Yang E, Lee DY, Lee B, Kim H, Oh U, Im H, Hwang EM, Park J. TMEM16A expression in cholinergic neurons of the medial habenula mediates anxiety-related behaviors. EMBO Rep 2020; 21:e48097. [PMID: 31782602 PMCID: PMC7001509 DOI: 10.15252/embr.201948097] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 11/13/2019] [Accepted: 11/14/2019] [Indexed: 11/09/2022] Open
Abstract
TMEM16A, a Ca2+ -activated Cl- channel, is known to modulate the excitability of various types of cells; however, its function in central neurons is largely unknown. Here, we show the specific expression of TMEM16A in the medial habenula (mHb) via RNAscope in situ hybridization, immunohistochemistry, and electrophysiology. When TMEM16A is ablated in the mHb cholinergic neurons (TMEM16A cKO mice), the slope of after-hyperpolarization of spontaneous action potentials decreases and the firing frequency is reduced. Reduced mHb activity also decreases the activity of the interpeduncular nucleus (IPN). Moreover, TMEM16A cKO mice display anxiogenic behaviors and deficits in social interaction without despair-like phenotypes or cognitive dysfunctions. Finally, chemogenetic inhibition of mHb cholinergic neurons using the DREADD (Designer Receptors Exclusively Activated by Designer Drugs) approach reveals similar behavioral phenotypes to those of TMEM16A cKO mice. We conclude that TMEM16A plays a key role in anxiety-related behaviors regulated by mHb cholinergic neurons and could be a potential therapeutic target against anxiety-related disorders.
Collapse
Affiliation(s)
- Chang‐Hoon Cho
- School of Biosystems and Biomedical SciencesCollege of Health SciencesKorea UniversitySeoulKorea
| | - Sangjoon Lee
- Convergence Research Center for Diagnosis, Treatment and Care System of DementiaKISTSeoulKorea
- Department of Pharmacology and Biomedical SciencesSeoul National University College of MedicineSeoulKorea
- Center for NeuroscienceBrain Science InstituteKorea Institute of Science and Technology (KIST)SeoulKorea
| | - Ajung Kim
- Center for Functional ConnectomicsKISTSeoulKorea
- KHU‐KIST Department of Converging Science and TechnologyGraduate SchoolKyung Hee UniversitySeoulKorea
| | | | - Kanghyun Ryoo
- School of Biosystems and Biomedical SciencesCollege of Health SciencesKorea UniversitySeoulKorea
| | - Young‐Sun Lee
- School of Biosystems and Biomedical SciencesCollege of Health SciencesKorea UniversitySeoulKorea
| | - Hyun‐Gug Jung
- School of Biosystems and Biomedical SciencesCollege of Health SciencesKorea UniversitySeoulKorea
- Center for Functional ConnectomicsKISTSeoulKorea
| | - Esther Yang
- Department of AnatomyCollege of MedicineKorea UniversitySeoulKorea
| | - Da Yong Lee
- Center for Functional ConnectomicsKISTSeoulKorea
| | - Byeongjun Lee
- Sensory Research CenterCRIBrain Science InstituteKorea Institute of Science and TechnologySeoulKorea
| | - Hyun Kim
- Department of AnatomyCollege of MedicineKorea UniversitySeoulKorea
| | - Uhtaek Oh
- Sensory Research CenterCRIBrain Science InstituteKorea Institute of Science and TechnologySeoulKorea
| | - Heh‐In Im
- Convergence Research Center for Diagnosis, Treatment and Care System of DementiaKISTSeoulKorea
- Center for NeuroscienceBrain Science InstituteKorea Institute of Science and Technology (KIST)SeoulKorea
- Division of Bio‐Medical Science & TechnologyKIST SchoolKorea University of Science and TechnologySeoulKorea
| | - Eun Mi Hwang
- Center for Functional ConnectomicsKISTSeoulKorea
- KHU‐KIST Department of Converging Science and TechnologyGraduate SchoolKyung Hee UniversitySeoulKorea
- Division of Bio‐Medical Science & TechnologyKIST SchoolKorea University of Science and TechnologySeoulKorea
| | - Jae‐Yong Park
- School of Biosystems and Biomedical SciencesCollege of Health SciencesKorea UniversitySeoulKorea
| |
Collapse
|
29
|
Liu P, Kong L, Liang K, Wu Y, Jin H, Song B, Tan X. Identification of dissociation factors in pancreatic Cancer using a mass spectrometry-based proteomic approach. BMC Cancer 2020; 20:45. [PMID: 31959150 PMCID: PMC6971861 DOI: 10.1186/s12885-020-6522-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 01/07/2020] [Indexed: 01/06/2023] Open
Abstract
Backgroud Pancreatic cancer is a highly malignant tumor of the digestive system. This secretome of pancreatic cancer is key to its progression and metastasis. But different methods of protein extraction affect the final results. In other words, the real secretion of proteins in cancer cells has been changed. Based on mass spectrometry, we analyze the secretome from the serum-containing and serum-free medium, using different protein pretreatment methods. This study aims to identify dissociation factors in pancreatic cancer. Methods In this study, pancreatic cancer cells were cultured in serum-containing or serum-free medium, and the corresponding supernatants were extracted as samples. Subsequently, the above samples were separated by size exclusion chromatography (SEC), and peptide segments were identified by LC-MS/MS. The final results were identified via the hamster secreted protein database and a public database. Results Although the number of identified proteins in the serum-free medium group was high, the real secretion of proteins in pancreatic cancer cells was changed. There were six significant secreted proteins in the serum-containing medium group. Survival analysis via the TCGA database suggested that patients with higher expression levels of YWHAG showed a worse overall survival rate than those with lower YWHAG expression. Conclusions Our study demonstrated the results in the serum-containing medium group were more similar to the real secretome of pancreatic cancer cells. YWHAG could be used as a prognostic indicator for pancreatic cancer.
Collapse
Affiliation(s)
- Peng Liu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Lingming Kong
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Keke Liang
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Yunhao Wu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Haoyi Jin
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Bing Song
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, China.,Cardiff Institute of Tissue Engineering and Repair, School of Dentistry, Cardiff University, Cardiff, CF14 4XY, UK
| | - Xiaodong Tan
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| |
Collapse
|
30
|
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
|
31
|
Kim JY, Youn HY, Choi J, Baek SK, Kwon SY, Eun BK, Park JY, Oh KH. Anoctamin-1 affects the migration and invasion of anaplastic thyroid carcinoma cells. Anim Cells Syst (Seoul) 2019; 23:294-301. [PMID: 31489251 PMCID: PMC6711113 DOI: 10.1080/19768354.2019.1614981] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 04/27/2019] [Accepted: 05/01/2019] [Indexed: 11/11/2022] Open
Abstract
Anaplastic thyroid carcinoma (ATC) is a rare malignancy with very poor prognosis. The exact cause underlying its strong aggressive nature is not clear. Here, we discovered the elevated expression of anoctamin-1 (ANO1; Ca2+-activated Cl− channels) in advanced-stage ATC tissue. Using different ATC cell lines, the degree of expression of ANO1 was found to be related to the degree of ATC cell invasion by quantitative reverse transcription polymerase chain reaction and western blotting. Suppression of ANO1 activity either by selective inhibitor (T16Ainh-A01) or by siRNA significantly attenuated the migration and invasion of ATC cells. In conclusion, ANO1 appears to increase the ability of ATC cells to invade and migrate. Our results also suggest that the expression of ANO1 in patients with ATC may be helpful in predicting the prognosis of ATC.
Collapse
Affiliation(s)
- Jae-Young Kim
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Hwa Young Youn
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - June Choi
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University College of Medicine, Seoul, Republic of Korea
| | - Seung Kuk Baek
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University College of Medicine, Seoul, Republic of Korea
| | - Soon Young Kwon
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University College of Medicine, Seoul, Republic of Korea
| | - Bok Kee Eun
- Core-Laboratory for Convergent Translational Research, Korea University College of Medicine, Seoul, Republic of Korea
| | - Jae-Yong Park
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul, Republic of Korea
| | - Kyoung Ho Oh
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
32
|
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: 67] [Impact Index Per Article: 11.2] [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
|
33
|
Lu CS, Lee YN, Wang SW, Wu YJ, Su CH, Hsieh CL, Tien TY, Wang BJ, Chen MC, Chen CW, Yeh HI. KC21 Peptide Inhibits Angiogenesis and Attenuates Hypoxia-Induced Retinopathy. J Cardiovasc Transl Res 2019; 12:366-377. [PMID: 30790141 PMCID: PMC6707963 DOI: 10.1007/s12265-019-09865-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 01/20/2019] [Indexed: 12/31/2022]
Abstract
Desmogleins (Dsg2) are the major components of desmosomes. Dsg2 has five extracellular tandem cadherin domains (EC1-EC5) for cell-cell interaction. We had previously confirmed the Dsg2 antibody and its epitope (named KC21) derived from EC2 domain suppressing epithelial-mesenchymal transition and invasion in human cancer cell lines. Here, we screened six peptide fragments derived from EC2 domain and found that KR20, the parental peptide of KC21, was the most potent one on suppressing endothelial colony-forming cell (ECFC) tube-like structure formation. KC21 peptide also attenuated migration but did not disrupt viability and proliferation of ECFCs, consistent with the function to inhibit VEGF-mediated activation of p38 MAPK but not AKT and ERK. Animal studies showed that KC21 peptides suppressed capillary growth in Matrigel implant assay and inhibited oxygen-induced retinal neovascularization. The effects were comparable to bevacizumab (Bev). In conclusion, KC21 peptide is an angiogenic inhibitor potentially useful for treating angiogenesis-related diseases.
Collapse
Affiliation(s)
- Chi-Sheng Lu
- Departments of Medical Research, Mackay Memorial Hospital, Taipei, 10449, Taiwan.,Virginia Contract Research Organization Co., Ltd, Taipei, 11491, Taiwan
| | - Yi-Nan Lee
- Departments of Medical Research, Mackay Memorial Hospital, Taipei, 10449, Taiwan
| | - Shin-Wei Wang
- Department of Medicine, Mackay Medical College, New Taipei City, 25245, Taiwan
| | - Yih-Jer Wu
- Departments of Medical Research, Mackay Memorial Hospital, Taipei, 10449, Taiwan.,Department of Medicine, Mackay Medical College, New Taipei City, 25245, Taiwan
| | - Cheng-Huang Su
- Departments of Medical Research, Mackay Memorial Hospital, Taipei, 10449, Taiwan
| | - Chin-Ling Hsieh
- Departments of Medical Research, Mackay Memorial Hospital, Taipei, 10449, Taiwan
| | - Ting Yi Tien
- Departments of Medical Research, Mackay Memorial Hospital, Taipei, 10449, Taiwan
| | - Bo-Jeng Wang
- Department of Medicine, Mackay Medical College, New Taipei City, 25245, Taiwan
| | - Min-Che Chen
- Asclepiumm Taiwan Co., Ltd, New Taipei City, 25160, Taiwan
| | - Chun-Wei Chen
- Asclepiumm Taiwan Co., Ltd, New Taipei City, 25160, Taiwan
| | - Hung-I Yeh
- Departments of Medical Research, Mackay Memorial Hospital, Taipei, 10449, Taiwan. .,Department of Medicine, Mackay Medical College, New Taipei City, 25245, Taiwan. .,Departments of Medical Research and Internal Medicine, Mackay Memorial Hospital, No. 92, Sec. 2, Zhongshan N. Rd, Taipei City, 10449, Taiwan.
| |
Collapse
|
34
|
Kim DE, Cho CH, Sim KM, Kwon O, Hwang EM, Kim HW, Park JY. 14-3-3γ Haploinsufficient Mice Display Hyperactive and Stress-sensitive Behaviors. Exp Neurobiol 2019; 28:43-53. [PMID: 30853823 PMCID: PMC6401549 DOI: 10.5607/en.2019.28.1.43] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 01/16/2019] [Accepted: 01/18/2019] [Indexed: 02/03/2023] Open
Abstract
14-3-3γ plays diverse roles in different aspects of cellular processes. Especially in the brain where 14-3-3γ is enriched, it has been reported to be involved in neurological and psychiatric diseases (e.g. Williams-Beuren syndrome and Creutzfeldt-Jakob disease). However, behavioral abnormalities related to 14-3-3γ deficiency are largely unknown. Here, by using 14-3-3γ deficient mice, we found that homozygous knockout mice were prenatally lethal, and heterozygous mice showed developmental delay relative to wild-type littermate mice. In addition, in behavioral analyses, we found that 14-3-3γ heterozygote mice display hyperactive and depressive-like behavior along with more sensitive responses to acute stress than littermate control mice. These results suggest that 14-3-3γ levels may be involved in the developmental manifestation of related neuropsychiatric diseases. In addition, 14-3-3γ heterozygote mice may be a potential model to study the molecular pathophysiology of neuropsychiatric symptoms.
Collapse
Affiliation(s)
- Do Eon Kim
- College of Life Sciences, Sejong University, Seoul 05006, Korea
| | - Chang-Hoon Cho
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul 02708, Korea
| | - Kyoung Mi Sim
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul 02708, Korea
| | - Osung Kwon
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul 02708, Korea
| | - Eun Mi Hwang
- Center for Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
| | - Hyung-Wook Kim
- College of Life Sciences, Sejong University, Seoul 05006, Korea
| | - Jae-Yong Park
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul 02708, Korea
| |
Collapse
|
35
|
Ryu J, Kim DG, Lee YS, Bae Y, Kim A, Park N, Hwang EM, Park JY. Surface expression of TTYH2 is attenuated by direct interaction with β-COP. BMB Rep 2019. [PMID: 30670146 PMCID: PMC6675250 DOI: 10.5483/bmbrep.2019.52.7.188] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
TTYH2 is a calcium-activated, inwardly rectifying anion channel that has been shown to be related to renal cancer and colon cancer. Based on the topological prediction, TTYH2 protein has five transmembrane domains with the extracellular N-terminus and the cytoplasmic C-terminus. In the present study, we identified a vesicle transport protein, β-COP, as a novel specific binding partner of TTYH2 by yeast two-hybrid screening using a human brain cDNA library with the C-terminal region of TTYH2 (TTYH2-C) as a bait. Using in vitro and in vivo binding assays, we confirmed the protein-protein interactions between TTYH2 and β-COP. We also found that the surface expression and activity of TTYH2 were decreased by co-expression with β-COP in the heterologous expression system. In addition, β-COP associated with TTYH2 in a native condition at a human colon cancer cell line, LoVo cells. The over-expression of β-COP in the LoVo cells led to a dramatic decrease in the surface expression and activity of endogenous TTYH2. Collectively, these data suggested that β-COP plays a critical role in the trafficking of the TTYH2 channel to the plasma membrane.
Collapse
Affiliation(s)
- Jiwon Ryu
- School of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul 02841, Korea
| | - Dong-Gyu Kim
- Korea Institute of Science and Technology (KIST), Center for Functional Connectomics, Seoul 02792, Korea
| | - Young-Sun Lee
- School of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul 02841, Korea
| | - Yeonju Bae
- School of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul 02841, Korea
| | - Ajung Kim
- Korea Institute of Science and Technology (KIST), Center for Functional Connectomics, Seoul 02792, Korea; KHU-KIST Department of Converging Science and Technology, Graduate School, Kyung Hee University, Seoul 02447, Korea
| | - Nammi Park
- School of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul 02841, Korea
| | - Eun Mi Hwang
- Korea Institute of Science and Technology (KIST), Center for Functional Connectomics, Seoul 02792, Korea; KHU-KIST Department of Converging Science and Technology, Graduate School, Kyung Hee University, Seoul 02447, Korea
| | - Jae-Yong Park
- School of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul 02841, Korea
| |
Collapse
|
36
|
Lu G, Shi W, Zheng H. Inhibition of STAT6/Anoctamin-1 Activation Suppresses Proliferation and Invasion of Gastric Cancer Cells. Cancer Biother Radiopharm 2018; 33:3-7. [PMID: 29466035 DOI: 10.1089/cbr.2017.2287] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Gastric carcinoma is the most popular cancer worldwide. Anoctamin-1 is a calcium-activated channel and highly expressed in various tumors. A previous study indicated that suppressed Anoctamin-1 expression decreased cancer cell proliferation or migration. As a signal transduction and transcription activator, STAT6 is a novel agonist for Anoctamin-1 promoter. However, its role in tumor cell proliferation or migration remains unclear. Therefore, this study aimed to suppress STAT6 and Anoctamin-1 protein expression in gastric cancer cells to test the inhibitory effects on gastric cancer cell migration or invasion. MATERIALS AND METHODS MTT colorimetry was used to test cell proliferation. Western blot was used to measure STAT6 and Anoctamin-1 expression before and after small interfering RNA (siRNA) treatment. A scratch assay was performed to measure cell migration, followed by Transwell chamber assay analysis of cell invasion. RESULTS After STAT6 siRNA interference, the expression of STAT6 and Anoctamin-1 was significantly decreased in the gastric carcinoma cell line. Anoctamin-1 siRNA interference only decreased its protein expression, but not STAT6 protein expression. Interference of STAT6 or Anoctamin-1 reduced their protein expression and inhibited proliferation, migration, or invasion of gastric cancer cells. CONCLUSIONS Inhibition of STAT6/Anoctamin-1 activation decreased proliferation, migration, or invasion of gastric cancer cells, suggesting that the STAT6/Anoctamin-1 pathway might be a novel target for treating gastric cancer.
Collapse
Affiliation(s)
- Guohao Lu
- 1 Department of Emergency, The People's Hospital of Guangxi Zhuang Autonomous Region , Nanning, China
| | - Wanling Shi
- 2 Department of Ultrasound, The People's Hospital of Guangxi Zhuang Autonomous Region , Nanning, China
| | - Hongyu Zheng
- 2 Department of Ultrasound, The People's Hospital of Guangxi Zhuang Autonomous Region , Nanning, China
| |
Collapse
|
37
|
Bill A, Alex Gaither L. The Mechanistic Role of the Calcium-Activated Chloride Channel ANO1 in Tumor Growth and Signaling. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 966:1-14. [PMID: 28293832 DOI: 10.1007/5584_2016_201] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Multiple studies have described the high expression and amplification of Anoctamin 1 (ANO1) in various cancers, including, but not limited to breast cancer, head and neck cancer, gastrointestinal stromal tumors and glioblastoma. ANO1 has been demonstrated to be critical for tumor growth in breast and head and neck cancers through its regulation of EGFR signaling and pathway modulators like MAPK and protein kinase B. However, the discovery of ANO1 as a calcium activated chloride channel came as a surprise to the field and has given rise to many questions. How does a chloride channel promote oncogenesis? Is the chloride channel function of ANO1 important for its role in cancer? Does ANO1 exhibits chloride-independent functions in cancer cells? This review summarizes the current understanding of ANO1's function in cancer, provides a synopsis of the findings addressing the open questions in the field and gives an outlook on the promising future of ANO1 as a potential therapeutic target for the treatment of various cancers.
Collapse
Affiliation(s)
- Anke Bill
- Novartis Institutes for Biomedical Research, Cambridge, MA, 02139, USA
| | | |
Collapse
|
38
|
Inhibition of ANO1/TMEM16A induces apoptosis in human prostate carcinoma cells by activating TNF-α signaling. Cell Death Dis 2018; 9:703. [PMID: 29899325 PMCID: PMC5999606 DOI: 10.1038/s41419-018-0735-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Revised: 04/23/2018] [Accepted: 05/21/2018] [Indexed: 12/16/2022]
Abstract
Overexpression of the Ca2+-activated chloride channel ANO1/TMEM16A is implicated in tumorigenesis, and inhibition of ANO1 overexpression suppresses xenograft tumor growth and invasiveness. However, the underlying molecular mechanism for ANO1 inhibition in suppression of tumorigenesis remains unknown. Here, we show that silencing or inhibition of endogenous ANO1 inhibits cell growth, induces apoptosis and upregulates TNF-α expression in prostate cancer PC-3 cells. Enhancement of TNF-α signaling by ANO1 knockdown leads to upregulation of phosphorylated Fas-associated protein with death domain and caspase activation. Furthermore, silencing of ANO1 inhibits growth of PC-3 xenograft tumors in nude mice and induces apoptosis in tumors via upregulation of TNF-α signaling. Taken together, our findings provide mechanistic insight into promoting apoptosis in prostate cancer cells by ANO1 inhibition through upregulation of TNF-α signaling.
Collapse
|
39
|
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: 108] [Impact Index Per Article: 13.5] [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
|