1
|
Rostom MM, Rashwan AA, Sotiropoulou CD, Hozayen SZ, Abdelhamid AM, Abdelhalim MM, Eltahtawy O, Emara HM, Elemam NM, Kontos CK, Youness RA. MIAT: A pivotal oncogenic long noncoding RNA tunning the hallmarks of solid malignancies. Transl Oncol 2025; 54:102329. [PMID: 40014977 PMCID: PMC11910686 DOI: 10.1016/j.tranon.2025.102329] [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: 10/22/2024] [Revised: 01/06/2025] [Accepted: 02/13/2025] [Indexed: 03/01/2025] Open
Abstract
Long non-coding RNAs (LncRNAs) have emerged as intriguing players in cellular regulation, challenging the traditional view of non-coding RNAs as mere "dark genome". Non-coding DNA makes up most of the human genome and plays a pivotal role in cancer development. These RNA molecules, which do not code for proteins, have captivated researchers with their diverse and crucial roles in gene regulation, chromatin dynamics, and other cellular processes. In several physiological and pathological circumstances, lncRNAs serve critical functions. This review will tackle the complex function of the lncRNA myocardial infarction-associated transcript (MIAT) in various solid malignancies. A special emphasis would be directed on the correlation between cancer patients' clinicopathological features and the expression profile of MIAT. MIAT is a oncogenic regulator in many malignant tumors, where it can control the growth, invasion, metastasis, and resistance to death of cells. As a result, MIAT is thought to be a possible biomarker and therapeutic target for cancer patients. The biological functions, mechanisms and potential clinical implications of MIAT during carcinogenesis and finally the current possible therapeutic approaches targeting MIAT are also outlined in this review.
Collapse
Affiliation(s)
- Monica M Rostom
- Pharmacology and Toxicology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo, 11835, Cairo, Egypt
| | - Alaa A Rashwan
- Biotechnology Graduate Program, School of Sciences and Engineering, The American University in Cairo (AUC), 11835, Cairo, Egypt
| | - Christina D Sotiropoulou
- Department of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, 15701, Athens, Greece
| | - Sama Z Hozayen
- Molecular Biology and Biochemistry Department, Molecular Genetics Research Team (MGRT), Faculty of Biotechnology, German International University (GIU), 11835, Cairo, Egypt
| | | | - Miriam Mokhtar Abdelhalim
- Molecular Biology and Biochemistry Department, Molecular Genetics Research Team (MGRT), Faculty of Biotechnology, German International University (GIU), 11835, Cairo, Egypt
| | - Omar Eltahtawy
- Molecular Biology and Biochemistry Department, Molecular Genetics Research Team (MGRT), Faculty of Biotechnology, German International University (GIU), 11835, Cairo, Egypt
| | - Hadir M Emara
- Molecular Biology and Biochemistry Department, Molecular Genetics Research Team (MGRT), Faculty of Biotechnology, German International University (GIU), 11835, Cairo, Egypt; Department of Nanotechnology, School of Sciences and Engineering, The American University in Cairo (AUC), 11835, Cairo, Egypt
| | - Noha M Elemam
- Clinical Sciences Department, College of Medicine, University of Sharjah, 27272, Sharjah, UAE; Research Institute for Medical and Health Sciences, University of Sharjah, 27272, Sharjah, UAE
| | - Christos K Kontos
- Department of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, 15701, Athens, Greece
| | - Rana A Youness
- Molecular Biology and Biochemistry Department, Molecular Genetics Research Team (MGRT), Faculty of Biotechnology, German International University (GIU), 11835, Cairo, Egypt.
| |
Collapse
|
2
|
Dadgar-Zankbar L, Elahi Z, Shariati A, Khaledi A, Razavi S, Khoshbayan A. Exploring the role of Fusobacterium nucleatum in colorectal cancer: implications for tumor proliferation and chemoresistance. Cell Commun Signal 2024; 22:547. [PMID: 39548531 PMCID: PMC11566256 DOI: 10.1186/s12964-024-01909-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 10/24/2024] [Indexed: 11/18/2024] Open
Abstract
Fusobacterium nucleatum (Fn) has been extensively studied for its connection to colorectal cancer (CRC) and its potential role in chemotherapy resistance. Studies indicate that Fn is commonly found in CRC tissues and is associated with unfavorable prognosis and treatment failure. It has been shown that Fn promotes chemoresistance by affecting autophagy, a cellular process that helps cells survive under stressful conditions. Additionally, Fn targets specific signaling pathways that activate particular microRNAs and modulate the response to chemotherapy. Understanding the current molecular mechanisms and investigating the importance of Fn-inducing chemoresistance could provide valuable insights for developing novel therapies. This review surveys the role of Fn in tumor proliferation, metastasis, and chemoresistance in CRC, focusing on its effects on the tumor microenvironment, gene expression, and resistance to conventional chemotherapy drugs. It also discusses the therapeutic implications of targeting Fn in CRC treatment and highlights the need for further research.
Collapse
Affiliation(s)
- Leila Dadgar-Zankbar
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Elahi
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Vice Chancellery of Education and Research, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | - Aref Shariati
- Infectious Diseases Research Center (IDRC), Arak University of Medical Sciences, Arak, Iran
| | - Azad Khaledi
- Infectious Diseases Research Center, Kashan University of Medical Sciences, Kashan, Iran
- Department of Microbiology and Immunology, School of Medicine, Kashan University of Medical Sciences, P.O. Box: 87155.111, Kashan, 87154, Iran
| | - Shabnam Razavi
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Microbial Biotechnology Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Amin Khoshbayan
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Microbial Biotechnology Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
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
|
4
|
Yang X, Cui L, Liu Z, Li Y, Wu X, Tian R, Jia C, Ren C, Mou Y, Song X. TMEM16A inhibits autophagy and promotes the invasion of hypopharyngeal squamous cell carcinoma through mTOR pathway. Carcinogenesis 2024; 45:569-581. [PMID: 38470063 DOI: 10.1093/carcin/bgae020] [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: 09/06/2023] [Revised: 02/15/2024] [Accepted: 03/11/2024] [Indexed: 03/13/2024] Open
Abstract
Previous studies have indicated that transmembrane protein 16A (TMEM16A) plays a crucial role in the pathogenesis and progression of various tumors by influencing multiple signaling pathways. However, the role of TMEM16A in regulating autophagy via the mammalian target of rapamycin (mTOR) pathway and its impact on the development of hypopharyngeal squamous cell carcinoma (HSCC) remain unclear. Immunohistochemistry and western blotting were used to assess the expression of TMEM16A in HSCC tissues and metastatic lymph nodes. Manipulation of TMEM16A expression levels was achieved in the FaDu cell line through overexpression or knockdown, followed by assessment of its biological effects using cell colony formation, wound healing, transwell and invasion assays. Additionally, apoptosis and autophagy-related proteins, as well as autophagosome formation, were evaluated through western blotting, transmission electron microscopy and immunofluorescence following TMEM16A knockdown or overexpression in FaDu cells. Our study revealed significantly elevated levels of TMEM16A in both HSCC tissues and metastatic lymph nodes compared with normal tissues. In vitro experiments demonstrated that silencing TMEM16A led to a notable suppression of HSCC cell proliferation, invasion and migration. Furthermore, TMEM16A silencing effectively inhibited tumor growth in xenografted mice. Subsequent investigations indicated that knockdown of TMEM16A in HSCC cells could suppress mTOR activation, thereby triggering autophagic cell death by upregulating sequestosome-1 (SQSTM1/P62) and microtubule-associated protein light chain 3 II (LC3II). This study highlights the crucial role of TMEM16A in modulating autophagy in HSCC, suggesting its potential as a therapeutic target for the treatment of this malignancy.
Collapse
Affiliation(s)
- Xin Yang
- Department of Otorhinolaryngology Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong Province, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong Province, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, Shandong Province, China
| | - Limei Cui
- Department of Otorhinolaryngology Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong Province, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong Province, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, Shandong Province, China
| | - Zhonglu Liu
- Department of Otorhinolaryngology Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong Province, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong Province, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, Shandong Province, China
| | - Yumei Li
- Department of Otorhinolaryngology Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong Province, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong Province, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, Shandong Province, China
| | - Xinxin Wu
- Department of Otorhinolaryngology Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong Province, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong Province, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, Shandong Province, China
| | - Ruxian Tian
- Department of Otorhinolaryngology Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong Province, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong Province, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, Shandong Province, China
| | - Chuanliang Jia
- Department of Otorhinolaryngology Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong Province, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong Province, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, Shandong Province, China
| | - Chao Ren
- Department of Otorhinolaryngology Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong Province, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong Province, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, Shandong Province, China
- Shandong Provincial Innovation and Practice Base for Postdoctors, Yantai Yuhuangding Hospital, Yantai, Shandong Province, China
| | - Yakui Mou
- Department of Otorhinolaryngology Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong Province, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong Province, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, Shandong Province, China
| | - Xicheng Song
- Department of Otorhinolaryngology Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong Province, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong Province, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, Shandong Province, China
| |
Collapse
|
5
|
Arreola J, López-Romero AE, Huerta M, Guzmán-Hernández ML, Pérez-Cornejo P. Insights into the function and regulation of the calcium-activated chloride channel TMEM16A. Cell Calcium 2024; 121:102891. [PMID: 38772195 DOI: 10.1016/j.ceca.2024.102891] [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: 02/27/2024] [Revised: 04/23/2024] [Accepted: 04/23/2024] [Indexed: 05/23/2024]
Abstract
The TMEM16A channel, a member of the TMEM16 protein family comprising chloride (Cl-) channels and lipid scramblases, is activated by the free intracellular Ca2+ increments produced by inositol 1,4,5-trisphosphate (IP3)-induced Ca2+ release after GqPCRs or Ca2+ entry through cationic channels. It is a ubiquitous transmembrane protein that participates in multiple physiological functions essential to mammals' lives. TMEM16A structure contains two identical 10-segment monomers joined at their transmembrane segment 10. Each monomer harbours one independent hourglass-shaped pore gated by Ca2+ ligation to an orthosteric site adjacent to the pore and controlled by two gates. The orthosteric site is created by assembling negatively charged glutamate side chains near the pore´s cytosolic end. When empty, this site generates an electrostatic barrier that controls channel rectification. In addition, an isoleucine-triad forms a hydrophobic gate at the boundary of the cytosolic vestibule and the inner side of the neck. When the cytosolic Ca2+ rises, one or two Ca2+ ions bind to the orthosteric site in a voltage (V)-dependent manner, thus neutralising the electrostatic barrier and triggering an allosteric gating mechanism propagating via transmembrane segment 6 to the hydrophobic gate. These coordinated events lead to pore opening, allowing the Cl- flux to ensure the physiological response. The Ca2+-dependent function of TMEM16A is highly regulated. Anions with higher permeability than Cl- facilitate V dependence by increasing the Ca2+ sensitivity, intracellular protons can replace Ca2+ and induce channel opening, and phosphatidylinositol 4,5-bisphosphate bound to four cytosolic sites likely maintains Ca2+ sensitivity. Additional regulation is afforded by cytosolic proteins, most likely by phosphorylation and protein-protein interaction mechanisms.
Collapse
Affiliation(s)
- Jorge Arreola
- Jorge Arreola, Physics Institute of Universidad Autónoma de San Luis Potosí. Av. Parque Chapultepec 1570, Privadas del Pedregal, 78295 San Luis Potosí, SLP., Mexico.
| | - Ana Elena López-Romero
- Jorge Arreola, Physics Institute of Universidad Autónoma de San Luis Potosí. Av. Parque Chapultepec 1570, Privadas del Pedregal, 78295 San Luis Potosí, SLP., Mexico
| | - Miriam Huerta
- Jorge Arreola, Physics Institute of Universidad Autónoma de San Luis Potosí. Av. Parque Chapultepec 1570, Privadas del Pedregal, 78295 San Luis Potosí, SLP., Mexico
| | - María Luisa Guzmán-Hernández
- Catedrática CONAHCYT, Department of Physiology and Biophysics, School of Medicine, Universidad Autónoma de San Luis Potosí. Ave. V. Carranza 2905, Los Filtros, San Luis Potosí, SLP 78210, Mexico
| | - Patricia Pérez-Cornejo
- Department of Physiology and Biophysics, School of Medicine, Universidad Autónoma de San Luis Potosí. Ave. V. Carranza 2905, Los Filtros, San Luis Potosí, SLP 78210, Mexico
| |
Collapse
|
6
|
Orang A, Marri S, McKinnon RA, Petersen J, Michael MZ. Restricting Colorectal Cancer Cell Metabolism with Metformin: An Integrated Transcriptomics Study. Cancers (Basel) 2024; 16:2055. [PMID: 38893174 PMCID: PMC11171104 DOI: 10.3390/cancers16112055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 05/13/2024] [Accepted: 05/18/2024] [Indexed: 06/21/2024] Open
Abstract
BACKGROUND Metformin is a first-line therapy for type 2 diabetes as it disrupts cellular metabolism. Despite the association between metformin and lower cancer incidence, the anti-tumour activity of the drug in colorectal cancer (CRC) is incompletely understood. This study identifies underlying molecular mechanisms by which metformin slows colorectal cancer cell proliferation by investigating metformin-associated microRNA (miRNA) and target gene pairs implicated in signalling pathways. METHODS The present study analysed changes in miRNAs and the coding transcriptome in CRC cells treated with a sublethal dose of metformin, followed by the contextual validation of potential miRNA-target gene pairs. RESULTS Analyses of small RNA and transcriptome sequencing data revealed 104 miRNAs and 1221 mRNAs to be differentially expressed in CRC cells treated with metformin for 72 h. Interaction networks between differentially expressed miRNAs and putative target mRNAs were identified. Differentially expressed genes were mainly implicated in metabolism and signalling processes, such as the PI3K-Akt and MAPK/ERK pathways. Further validation of potential miRNA-target mRNA pairs revealed that metformin induced miR-2110 and miR-132-3p to target PIK3R3 and, consequently, regulate CRC cell proliferation, cell cycle progression and the PI3K-Akt signalling pathway. Metformin also induced miR-222-3p and miR-589-3p, which directly target STMN1 to inhibit CRC cell proliferation and cell cycle progression. CONCLUSIONS This study identified novel changes in the coding transcriptome and small non-coding RNAs associated with metformin treatment of CRC cells. Integration of these datasets highlighted underlying mechanisms by which metformin impedes cell proliferation in CRC. Importantly, it identified the post-transcriptional regulation of specific genes that impact both metabolism and cell proliferation.
Collapse
Affiliation(s)
- Ayla Orang
- Flinders Health and Medical Research Institute, Flinders University, Bedford Park, SA 5042, Australia; (A.O.); (S.M.); (R.A.M.); (J.P.)
| | - Shashikanth Marri
- Flinders Health and Medical Research Institute, Flinders University, Bedford Park, SA 5042, Australia; (A.O.); (S.M.); (R.A.M.); (J.P.)
| | - Ross A. McKinnon
- Flinders Health and Medical Research Institute, Flinders University, Bedford Park, SA 5042, Australia; (A.O.); (S.M.); (R.A.M.); (J.P.)
| | - Janni Petersen
- Flinders Health and Medical Research Institute, Flinders University, Bedford Park, SA 5042, Australia; (A.O.); (S.M.); (R.A.M.); (J.P.)
- Nutrition and Metabolism, South Australia Health and Medical Research Institute, Adelaide, SA 5000, Australia
| | - Michael Z. Michael
- Flinders Health and Medical Research Institute, Flinders University, Bedford Park, SA 5042, Australia; (A.O.); (S.M.); (R.A.M.); (J.P.)
- Department of Gastroenterology and Hepatology, Flinders Medical Centre, Bedford Park, SA 5042, Australia
- Flinders Centre for Innovation in Cancer, Flinders Medical Centre, Bedford Park, SA 5042, Australia
| |
Collapse
|
7
|
Li X, Wang Y, Zhang L, Yao S, Liu Q, Jin H, Tuo B. The role of anoctamin 1 in liver disease. J Cell Mol Med 2024; 28:e18320. [PMID: 38685684 PMCID: PMC11058335 DOI: 10.1111/jcmm.18320] [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: 11/08/2023] [Revised: 03/21/2024] [Accepted: 04/03/2024] [Indexed: 05/02/2024] Open
Abstract
Liver diseases include all types of viral hepatitis, alcoholic liver disease (ALD), nonalcoholic fatty liver disease (NAFLD), cirrhosis, liver failure (LF) and hepatocellular carcinoma (HCC). Liver disease is now one of the leading causes of disease and death worldwide, which compels us to better understand the mechanisms involved in the development of liver diseases. Anoctamin 1 (ANO1), a calcium-activated chloride channel (CaCC), plays an important role in epithelial cell secretion, proliferation and migration. ANO1 plays a key role in transcriptional regulation as well as in many signalling pathways. It is involved in the genesis, development, progression and/or metastasis of several tumours and other diseases including liver diseases. This paper reviews the role and molecular mechanisms of ANO1 in the development of various liver diseases, aiming to provide a reference for further research on the role of ANO1 in liver diseases and to contribute to the improvement of therapeutic strategies for liver diseases by regulating ANO1.
Collapse
Affiliation(s)
- Xin Li
- Department of Gastroenterology, Digestive Disease HospitalAffiliated Hospital of Zunyi Medical UniversityZunyiChina
| | - Yongfeng Wang
- Department of Gastroenterology, Digestive Disease HospitalAffiliated Hospital of Zunyi Medical UniversityZunyiChina
| | - Li Zhang
- Department of Gastroenterology, Digestive Disease HospitalAffiliated Hospital of Zunyi Medical UniversityZunyiChina
| | - Shun Yao
- Department of Gastroenterology, Digestive Disease HospitalAffiliated Hospital of Zunyi Medical UniversityZunyiChina
| | - Qian Liu
- Department of Gastroenterology, Digestive Disease HospitalAffiliated Hospital of Zunyi Medical UniversityZunyiChina
| | - Hai Jin
- Department of Gastroenterology, Digestive Disease HospitalAffiliated Hospital of Zunyi Medical UniversityZunyiChina
- The Collaborative Innovation Center of Tissue Damage Repair and Regenerative Medicine of Zunyi Medical UniversityZunyiChina
| | - Biguang Tuo
- Department of Gastroenterology, Digestive Disease HospitalAffiliated Hospital of Zunyi Medical UniversityZunyiChina
- The Collaborative Innovation Center of Tissue Damage Repair and Regenerative Medicine of Zunyi Medical UniversityZunyiChina
| |
Collapse
|
8
|
An SX, Yu ZJ, Fu C, Wei MJ, Shen LH. Biological factors driving colorectal cancer metastasis. World J Gastrointest Oncol 2024; 16:259-272. [PMID: 38425391 PMCID: PMC10900157 DOI: 10.4251/wjgo.v16.i2.259] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 12/23/2023] [Accepted: 01/08/2024] [Indexed: 02/02/2024] Open
Abstract
Approximately 20% of colorectal cancer (CRC) patients present with metastasis at diagnosis. Among Stage I-III CRC patients who undergo surgical resection, 18% typically suffer from distal metastasis within the first three years following initial treatment. The median survival duration after the diagnosis of metastatic CRC (mCRC) is only 9 mo. mCRC is traditionally considered to be an advanced stage malignancy or is thought to be caused by incomplete resection of tumor tissue, allowing cancer cells to spread from primary to distant organs; however, increasing evidence suggests that the mCRC process can begin early in tumor development. CRC patients present with high heterogeneity and diverse cancer phenotypes that are classified on the basis of molecular and morphological alterations. Different genomic and nongenomic events can induce subclone diversity, which leads to cancer and metastasis. Throughout the course of mCRC, metastatic cascades are associated with invasive cancer cell migration through the circulatory system, extravasation, distal seeding, dormancy, and reactivation, with each step requiring specific molecular functions. However, cancer cells presenting neoantigens can be recognized and eliminated by the immune system. In this review, we explain the biological factors that drive CRC metastasis, namely, genomic instability, epigenetic instability, the metastatic cascade, the cancer-immunity cycle, and external lifestyle factors. Despite remarkable progress in CRC research, the role of molecular classification in therapeutic intervention remains unclear. This review shows the driving factors of mCRC which may help in identifying potential candidate biomarkers that can improve the diagnosis and early detection of mCRC cases.
Collapse
Affiliation(s)
- Shuai-Xing An
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, Liaoning Province, China
- Liaoning Key Laboratory of Molecular Targeted Antitumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors (China Medical University), Ministry of Education, Shenyang 110122, Liaoning Province, China
- BD Department, Greenpine Pharma Group Co., Ltd, Tianjin 300020, China
| | - Zhao-Jin Yu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, Liaoning Province, China
- Liaoning Key Laboratory of Molecular Targeted Antitumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors (China Medical University), Ministry of Education, Shenyang 110122, Liaoning Province, China
| | - Chen Fu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, Liaoning Province, China
- Liaoning Key Laboratory of Molecular Targeted Antitumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors (China Medical University), Ministry of Education, Shenyang 110122, Liaoning Province, China
| | - Min-Jie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, Liaoning Province, China
- Liaoning Key Laboratory of Molecular Targeted Antitumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors (China Medical University), Ministry of Education, Shenyang 110122, Liaoning Province, China
| | - Long-Hai Shen
- Center of Oncology, Genertec Liaoyou Gem Flower Hospital, PanJin 124010, Liaoning Province, China
| |
Collapse
|
9
|
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: 1] [Impact Index Per Article: 1.0] [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
|
10
|
Li S, Wang Z, Geng R, Zhang W, Wan H, Kang X, Guo S. TMEM16A ion channel: A novel target for cancer treatment. Life Sci 2023; 331:122034. [PMID: 37611692 DOI: 10.1016/j.lfs.2023.122034] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/13/2023] [Accepted: 08/18/2023] [Indexed: 08/25/2023]
Abstract
Cancer draws attention owing to the high morbidity and mortality. It is urgent to develop safe and effective cancer therapeutics. The calcium-activated chloride channel TMEM16A is widely distributed in various tissues and regulates physiological functions. TMEM16A is abnormally expressed in several cancers and associate with tumorigenesis, metastasis, and prognosis. Knockdown or inhibition of TMEM16A in cancer cells significantly inhibits cancer development. Therefore, TMEM16A is considered as a biomarker and therapeutic target for some cancers. This work reviews the cancers associated with TMEM16A. Then, the molecular mechanism of TMEM16A overexpression in cancer was analyzed, and the possible signal transduction mechanism of TMEM16A regulating cancer development was summarized. Finally, TMEM16A inhibitors with anticancer effect and their anticancer mechanism were concluded. We hope to provide new ideas for pharmacological studies on TMEM16A in cancer.
Collapse
Affiliation(s)
- Shuting Li
- School of Life Sciences, Hebei University, Baoding 071002, Hebei, China
| | - Zhichen Wang
- School of Life Sciences, Hebei University, Baoding 071002, Hebei, China
| | - Ruili Geng
- School of Life Sciences, Hebei University, Baoding 071002, Hebei, China
| | - Weiwei Zhang
- School of Basic Medical Sciences, Hebei University, Baoding 071002, Hebei, China
| | - Haifu Wan
- School of Life Sciences, Hebei University, Baoding 071002, Hebei, China; Institute of Life Sciences and Green Development, Hebei University, Baoding 071002, Hebei, China
| | - Xianjiang Kang
- School of Life Sciences, Hebei University, Baoding 071002, Hebei, China; Institute of Life Sciences and Green Development, Hebei University, Baoding 071002, Hebei, China.
| | - Shuai Guo
- School of Life Sciences, Hebei University, Baoding 071002, Hebei, China; Institute of Life Sciences and Green Development, Hebei University, Baoding 071002, Hebei, China.
| |
Collapse
|
11
|
Ascrizzi S, Arillotta GM, Grillone K, Caridà G, Signorelli S, Ali A, Romeo C, Tassone P, Tagliaferri P. Lynch Syndrome Biopathology and Treatment: The Potential Role of microRNAs in Clinical Practice. Cancers (Basel) 2023; 15:3930. [PMID: 37568746 PMCID: PMC10417124 DOI: 10.3390/cancers15153930] [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: 06/20/2023] [Revised: 07/27/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023] Open
Abstract
Lynch syndrome (LS), also known as Hereditary Non-Polyposis Colorectal Cancer (HNPCC), is an autosomal dominant cancer syndrome which causes about 2-3% of cases of colorectal carcinoma. The development of LS is due to the genetic and epigenetic inactivation of genes involved in the DNA mismatch repair (MMR) system, causing an epiphenomenon known as microsatellite instability (MSI). Despite the fact that the genetics of the vast majority of MSI-positive (MSI+) cancers can be explained, the etiology of this specific subset is still poorly understood. As a possible new mechanism, it has been recently demonstrated that the overexpression of certain microRNAs (miRNAs, miRs), such as miR-155, miR-21, miR-137, can induce MSI or modulate the expression of the genes involved in LS pathogenesis. MiRNAs are small RNA molecules that regulate gene expression at the post-transcriptional level by playing a critical role in the modulation of key oncogenic pathways. Increasing evidence of the link between MSI and miRNAs in LS prompted a deeper investigation into the miRNome involved in these diseases. In this regard, in this study, we discuss the emerging role of miRNAs as crucial players in the onset and progression of LS as well as their potential use as disease biomarkers and therapeutic targets in the current view of precision medicine.
Collapse
Affiliation(s)
- Serena Ascrizzi
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (S.A.); (G.M.A.); (K.G.); (G.C.); (S.S.); (A.A.); (C.R.); (P.T.)
| | - Grazia Maria Arillotta
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (S.A.); (G.M.A.); (K.G.); (G.C.); (S.S.); (A.A.); (C.R.); (P.T.)
| | - Katia Grillone
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (S.A.); (G.M.A.); (K.G.); (G.C.); (S.S.); (A.A.); (C.R.); (P.T.)
| | - Giulio Caridà
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (S.A.); (G.M.A.); (K.G.); (G.C.); (S.S.); (A.A.); (C.R.); (P.T.)
| | - Stefania Signorelli
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (S.A.); (G.M.A.); (K.G.); (G.C.); (S.S.); (A.A.); (C.R.); (P.T.)
| | - Asad Ali
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (S.A.); (G.M.A.); (K.G.); (G.C.); (S.S.); (A.A.); (C.R.); (P.T.)
| | - Caterina Romeo
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (S.A.); (G.M.A.); (K.G.); (G.C.); (S.S.); (A.A.); (C.R.); (P.T.)
| | - Pierfrancesco Tassone
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (S.A.); (G.M.A.); (K.G.); (G.C.); (S.S.); (A.A.); (C.R.); (P.T.)
- Medical Oncology and Translational Medical Oncology Units, University Hospital Renato Dulbecco, 88100 Catanzaro, Italy
| | - Pierosandro Tagliaferri
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (S.A.); (G.M.A.); (K.G.); (G.C.); (S.S.); (A.A.); (C.R.); (P.T.)
- Medical Oncology and Translational Medical Oncology Units, University Hospital Renato Dulbecco, 88100 Catanzaro, Italy
| |
Collapse
|
12
|
Aseervatham J. Dynamic Role of Exosome microRNAs in Cancer Cell Signaling and Their Emerging Role as Noninvasive Biomarkers. BIOLOGY 2023; 12:biology12050710. [PMID: 37237523 DOI: 10.3390/biology12050710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/26/2023] [Accepted: 05/10/2023] [Indexed: 05/28/2023]
Abstract
Exosomes are extracellular vesicles that originate from endosomes and are released by all cells irrespective of their origin or type. They play an important role in cell communication and can act in an autocrine, endocrine, or paracrine fashion. They are 40-150 nm in diameter and have a similar composition to the cell of origin. An exosome released by a particular cell is unique since it carries information about the state of the cell in pathological conditions such as cancer. miRNAs carried by cancer-derived exosomes play a multifaceted role by taking part in cell proliferation, invasion, metastasis, epithelial-mesenchymal transition, angiogenesis, apoptosis, and immune evasion. Depending on the type of miRNA that it carries as its cargo, it can render cells chemo- or radiosensitive or resistant and can also act as a tumor suppressor. Since the composition of exosomes is affected by the cellular state, stress, and changes in the environment, they can be used as diagnostic or prognostic biomarkers. Their unique ability to cross biological barriers makes them an excellent choice as vehicles for drug delivery. Because of their easy availability and stability, they can be used to replace cancer biopsies, which are invasive and expensive. Exosomes can also be used to follow the progression of diseases and monitor treatment strategies. A better understanding of the roles and functions of exosomal miRNA can be used to develop noninvasive, innovative, and novel treatments for cancer.
Collapse
Affiliation(s)
- Jaya Aseervatham
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
13
|
Wu Y, Li W, Chen X, Wang H, Su S, Xu Y, Deng X, Yang T, Wei M, Li L, Liu Y, Yang J, Li W. DOG1 as a novel antibody-drug conjugate target for the treatment of multiple gastrointestinal tumors and liver metastasis. Front Immunol 2023; 14:1051506. [PMID: 36776873 PMCID: PMC9909470 DOI: 10.3389/fimmu.2023.1051506] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 01/09/2023] [Indexed: 01/27/2023] Open
Abstract
Discovered On Gastrointestinal stromal tumors protein 1 (DOG1), a major calcium-activated chloride channel, has been used as a common diagnostic marker for gastrointestinal stromal tumors. However, the therapeutic application of DOG1 was not well defined. Here, we aim to investigate its potential as a therapeutic target for an antibody-drug conjugate (ADC) in various cancers of the alimentary tract and metastasis. The DOG1 expression profile was determined among TCGA samples and tissue microarrays. High levels of DOG1 expression were ubiquitously observed in multiple cancer samples from the alimentary tract determined by TCGA samples and tissue microarrays. Circulating tumor cells isolated from metastatic colon cancer patients were also positive for DOG1 expression. The mechanisms of anti-DOG1 antibody were investigated by dual-luciferase reporter assay. The anti-DOG1 antibody could inhibit proliferation and metastasis via p53 signaling in limited cancer cell lines. The anti-DOG1 antibody was conjugated with a microtubule inhibitor DM4, to construct a new anti-DOG1-DM4-ADC to strengthen its activity. The anti-DOG1-DM4-ADC showed cytotoxicity at the nanomolar level in vitro. In the murine xenograft tumor models, treatment of anti-DOG1-DM4-ADC achieved a significant tumor growth inhibition rate. Our study indicates that anti-DOG1-DM4-ADC may be promising therapeutic molecules for DOG1-positive alimentary tract tumors and may be effective in inhibiting recurrence after curative resection of liver metastases of colorectal origin.
Collapse
Affiliation(s)
- Yangping Wu
- Targeted Tracer Research and Development Laboratory, Department of Respiratory and Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Wenting Li
- State Key Laboratory of Biotherapy and Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xiangzheng Chen
- Department of Liver Surgery & Liver Transplantation, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Haichuan Wang
- Department of Liver Surgery & Liver Transplantation, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Siyuan Su
- Department of Chemistry, University of Illinois Chicago, Chicago, IL, United States
| | - Ying Xu
- Targeted Tracer Research and Development Laboratory, Department of Respiratory and Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xiangbing Deng
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Tinghan Yang
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Mingtian Wei
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Li Li
- Institute of Clinical Pathology, West China Hospital of Sichuan University, Chengdu, China
| | - Yixin Liu
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Jinliang Yang
- State Key Laboratory of Biotherapy and Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China,*Correspondence: Weimin Li, ; Jinliang Yang,
| | - Weimin Li
- Targeted Tracer Research and Development Laboratory, Department of Respiratory and Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China,*Correspondence: Weimin Li, ; Jinliang Yang,
| |
Collapse
|
14
|
Zhou H, Chen Y, Xiao Y, Wu Q, Li H, Li Y, Su G, Ke L, Wu J, Li J. Evaluation of the ability of fatty acid metabolism signature to predict response to neoadjuvant chemoradiotherapy and prognosis of patients with locally advanced rectal cancer. Front Immunol 2022; 13:1050721. [PMID: 36505493 PMCID: PMC9729334 DOI: 10.3389/fimmu.2022.1050721] [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: 09/22/2022] [Accepted: 10/26/2022] [Indexed: 11/25/2022] Open
Abstract
Neoadjuvant chemoradiotherapy (nCRT) is widely used to treat patients with locally advanced rectal cancer (LARC), and treatment responses vary. Fatty acid metabolism (FAM) is closely associated with carcinogenesis and cancer progression. In this study, we investigated the vital role of FAM on the gut microbiome and metabolism in the context of cancer. We screened 34 disease-free survival (DFS)-related, FAM-related, and radiosensitivity-related genes based on the Gene Expression Omnibus database. Subsequently, we developed a five-gene FAM-related signature using the least absolute shrinkage and selection operator Cox regression model. The FAM-related signature was also validated in external validation from Fujian Cancer Hospital for predicting nCRT response, DFS, and overall survival (OS). Notably, patients with a low-risk score were associated with pathological complete response and better DFS and OS outcomes. A comprehensive evaluation of the tumor microenvironment based on the FAM-related signature revealed that patients with high-risk scores were closely associated with activating type I interferon response and inflammation-promoting functions. In conclusion, our findings indicate the potential ability of FAM to predict nCRT response and the prognosis of DFS and OS in patients with LARC.
Collapse
Affiliation(s)
- Han Zhou
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Yanping Chen
- Department of Clinical Pathology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Yu Xiao
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Qian Wu
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Hui Li
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Yi Li
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Guangjian Su
- Department of Clinical Laboratory, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Longfeng Ke
- Department of Clinical Pathology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Junxin Wu
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China,*Correspondence: Jinluan Li, ; Junxin Wu,
| | - Jinluan Li
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China,*Correspondence: Jinluan Li, ; Junxin Wu,
| |
Collapse
|
15
|
Volovat SR, Augustin I, Zob D, Boboc D, Amurariti F, Volovat C, Stefanescu C, Stolniceanu CR, Ciocoiu M, Dumitras EA, Danciu M, Apostol DGC, Drug V, Shurbaji SA, Coca LG, Leon F, Iftene A, Herghelegiu PC. Use of Personalized Biomarkers in Metastatic Colorectal Cancer and the Impact of AI. Cancers (Basel) 2022; 14:4834. [PMID: 36230757 PMCID: PMC9562853 DOI: 10.3390/cancers14194834] [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: 07/10/2022] [Revised: 09/18/2022] [Accepted: 09/29/2022] [Indexed: 12/09/2022] Open
Abstract
Colorectal cancer is a major cause of cancer-related death worldwide and is correlated with genetic and epigenetic alterations in the colonic epithelium. Genetic changes play a major role in the pathophysiology of colorectal cancer through the development of gene mutations, but recent research has shown an important role for epigenetic alterations. In this review, we try to describe the current knowledge about epigenetic alterations, including DNA methylation and histone modifications, as well as the role of non-coding RNAs as epigenetic regulators and the prognostic and predictive biomarkers in metastatic colorectal disease that can allow increases in the effectiveness of treatments. Additionally, the intestinal microbiota's composition can be an important biomarker for the response to strategies based on the immunotherapy of CRC. The identification of biomarkers in mCRC can be enhanced by developing artificial intelligence programs. We present the actual models that implement AI technology as a bridge connecting ncRNAs with tumors and conducted some experiments to improve the quality of the model used as well as the speed of the model that provides answers to users. In order to carry out this task, we implemented six algorithms: the naive Bayes classifier, the random forest classifier, the decision tree classifier, gradient boosted trees, logistic regression and SVM.
Collapse
Affiliation(s)
- Simona-Ruxandra Volovat
- Department of Medical Oncology-Radiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iasi, Romania
| | - Iolanda Augustin
- Department of Medical Oncology, AI.Trestioreanu Institute of Oncology, 022328 Bucharest, Romania
| | - Daniela Zob
- Department of Medical Oncology, AI.Trestioreanu Institute of Oncology, 022328 Bucharest, Romania
| | - Diana Boboc
- Department of Medical Oncology-Radiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iasi, Romania
| | - Florin Amurariti
- Department of Medical Oncology-Radiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iasi, Romania
| | - Constantin Volovat
- Department of Medical Oncology, “Euroclinic” Center of Oncology, 2 Vasile Conta Str., 700106 Iasi, Romania
| | - Cipriana Stefanescu
- Department of Biophysics and Medical Physics-Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iasi, Romania
| | - Cati Raluca Stolniceanu
- Department of Biophysics and Medical Physics-Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iasi, Romania
| | - Manuela Ciocoiu
- Department of Pathophysiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Eduard Alexandru Dumitras
- Department of Pathophysiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Department of Anesthesiology and Intensive Care, Regional Institute of Oncology, 700115 Iasi, Romania
| | - Mihai Danciu
- Pathology Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | | | - Vasile Drug
- Department of Gastroenterology, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iasi, Romania
- Gastroenterology Clinic, Institute of Gastroenterology and Hepatology, ‘St. Spiridon’ Clinical Hospital, 700115 Iasi, Romania
| | - Sinziana Al Shurbaji
- Gastroenterology Clinic, Institute of Gastroenterology and Hepatology, ‘St. Spiridon’ Clinical Hospital, 700115 Iasi, Romania
| | - Lucia-Georgiana Coca
- Faculty of Computer Science, Alexandru Ioan Cuza University, 700115 Iasi, Romania
| | - Florin Leon
- Faculty of Automatic Control and Computer Engineering, Gheorghe Asachi Technical University, 700115 Iasi, Romania
| | - Adrian Iftene
- Faculty of Computer Science, Alexandru Ioan Cuza University, 700115 Iasi, Romania
| | - Paul-Corneliu Herghelegiu
- Faculty of Automatic Control and Computer Engineering, Gheorghe Asachi Technical University, 700115 Iasi, Romania
| |
Collapse
|
16
|
Pu Y, Wei J, Wu Y, Zhao K, Wu Y, Wu S, Yang X, Xing C. THUMPD3-AS1 facilitates cell growth and aggressiveness by the miR-218-5p/SKAP1 axis in colorectal cancer. Cell Biochem Biophys 2022; 80:483-494. [PMID: 35538197 DOI: 10.1007/s12013-022-01074-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 04/22/2022] [Indexed: 12/11/2022]
Abstract
BACKGROUND Colorectal cancer (CRC) is a malignant cancer with a high mortality. Accumulating studies have revealed that mRNAs involved in ceRNA (competing endogenous RNA) network are implicated in the tumorigenesis and development of CRC. Here, we aimed to elucidate the ceRNA network involving Src kinase associated phosphoprotein 1 (SKAP1) in the biological characteristics of CRC. METHODS Expression levels of genes in colon adenocarcinoma (COAD) samples and prognosis of COAD patients were predicted using publicly available online tool. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), clony formation and Transwell assays were conducted to test the biological functions of SKAP1 and THUMPD3 antisense RNA 1 (THUMPD3-AS1) in CRC cells. Western blot was used to measure the protein levels of SKAP1. Gene expression in CRC cells was detected by reverse transcription quantitative polymerase chain reaction (RT-qPCR). The interaction between miR-218-5p and THUMPD3-AS1 (or SKAP1) was verified by RNA pulldown and luciferase reporter assays. RESULTS SKAP1 was upregulated in COAD tissues and CRC cells and it reflected a poor prognosis in patients with COAD. SKAP1 knockdown inhibited CRC (HT-29 and HCT-116) cell proliferation, migration and invasion. Mechanistically, THUMPD3-AS1 acted as a ceRNA to sponge miR-218-5p and subsequently upregulated SKAP1 expression in CRC cells. SKAP1 overexpression reversed the suppressive effect of THUMPD3-AS1 knockdown on proliferation, migration and invision of CRC cells. CONCLUSIONS THUMPD3-AS1 promotes CRC cell growth and aggressiveness by regulating the miR-218-5p/SKAP1 axis.
Collapse
Affiliation(s)
- Yuwei Pu
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China
| | - Jinrong Wei
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China
| | - Yong Wu
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China
| | - Kui Zhao
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China
| | - Yongyou Wu
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China
| | - Shu Wu
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China
| | - Xiaodong Yang
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China.
| | - Chungen Xing
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China.
| |
Collapse
|
17
|
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
|
18
|
TMEM16A as a potential treatment target for head and neck cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2022; 41:196. [PMID: 35668455 PMCID: PMC9172006 DOI: 10.1186/s13046-022-02405-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 05/26/2022] [Indexed: 01/02/2023]
Abstract
Transmembrane protein 16A (TMEM16A) forms a plasma membrane-localized Ca2+-activated Cl- channel. Its gene has been mapped to an area on chromosome 11q13, which is amplified in head and neck squamous cell carcinoma (HNSCC). In HNSCC, TMEM16A overexpression is associated with not only high tumor grade, metastasis, low survival, and poor prognosis, but also deterioration of clinical outcomes following platinum-based chemotherapy. Recent study revealed the interaction between TMEM16A and transforming growth factor-β (TGF-β) has an indirect crosstalk in clarifying the mechanism of TMEM16A-induced epithelial-mesenchymal transition. Moreover, human papillomavirus (HPV) infection can modulate TMEM16A expression along with epidermal growth factor receptor (EGFR), whose phosphorylation has been reported as a potential co-biomarker of HPV-positive cancers. Considering that EGFR forms a functional complex with TMEM16A and is a co-biomarker of HPV, there may be crosstalk between TMEM16A expression and HPV-induced HNSCC. EGFR activation can induce programmed death ligand 1 (PD-L1) synthesis via activation of the nuclear factor kappa B pathway and JAK/STAT3 pathway. Here, we describe an interplay among EGFR, PD-L1, and TMEM16A. Combination therapy using TMEM16A and PD-L1 inhibitors may improve the survival rate of HNSCC patients, especially those resistant to anti-EGFR inhibitor treatment. To the best of our knowledge, this is the first review to propose a biological validation that combines immune checkpoint inhibition with TMEM16A inhibition.
Collapse
|
19
|
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
|
20
|
Zhou H, Liu Z, Wang Y, Wen X, Amador EH, Yuan L, Ran X, Xiong L, Ran Y, Chen W, Wen Y. Colorectal liver metastasis: molecular mechanism and interventional therapy. Signal Transduct Target Ther 2022; 7:70. [PMID: 35246503 PMCID: PMC8897452 DOI: 10.1038/s41392-022-00922-2] [Citation(s) in RCA: 139] [Impact Index Per Article: 46.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 01/25/2022] [Accepted: 02/09/2022] [Indexed: 02/08/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most frequently occurring malignancy tumors with a high morbidity additionally, CRC patients may develop liver metastasis, which is the major cause of death. Despite significant advances in diagnostic and therapeutic techniques, the survival rate of colorectal liver metastasis (CRLM) patients remains very low. CRLM, as a complex cascade reaction process involving multiple factors and procedures, has complex and diverse molecular mechanisms. In this review, we summarize the mechanisms/pathophysiology, diagnosis, treatment of CRLM. We also focus on an overview of the recent advances in understanding the molecular basis of CRLM with a special emphasis on tumor microenvironment and promise of newer targeted therapies for CRLM, further improving the prognosis of CRLM patients.
Collapse
Affiliation(s)
- Hui Zhou
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan Province, China
| | - Zhongtao Liu
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan Province, China
| | - Yongxiang Wang
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan Province, China
| | - Xiaoyong Wen
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan Province, China
| | - Eric H Amador
- Department of Physics, The University of Texas, Arlington, TX, 76019, USA
| | - Liqin Yuan
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan Province, China
| | - Xin Ran
- Department of Dermatovenereology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Li Xiong
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan Province, China.
| | - Yuping Ran
- Department of Dermatovenereology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Wei Chen
- Department of Physics, The University of Texas, Arlington, TX, 76019, USA.
- Medical Technology Research Centre, Chelmsford Campus, Anglia Ruskin University, Chelmsford, CM1 1SQ, UK.
| | - Yu Wen
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan Province, China.
| |
Collapse
|
21
|
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
|
22
|
Ion Channel Involvement in Tumor Drug Resistance. J Pers Med 2022; 12:jpm12020210. [PMID: 35207698 PMCID: PMC8878471 DOI: 10.3390/jpm12020210] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 01/28/2022] [Accepted: 02/02/2022] [Indexed: 11/30/2022] Open
Abstract
Over 90% of deaths in cancer patients are attributed to tumor drug resistance. Resistance to therapeutic agents can be due to an innate property of cancer cells or can be acquired during chemotherapy. In recent years, it has become increasingly clear that regulation of membrane ion channels is an important mechanism in the development of chemoresistance. Here, we review the contribution of ion channels in drug resistance of various types of cancers, evaluating their potential in clinical management. Several molecular mechanisms have been proposed, including evasion of apoptosis, cell cycle arrest, decreased drug accumulation in cancer cells, and activation of alternative escape pathways such as autophagy. Each of these mechanisms leads to a reduction of the therapeutic efficacy of administered drugs, causing more difficulty in cancer treatment. Thus, targeting ion channels might represent a good option for adjuvant therapies in order to counteract chemoresistance development.
Collapse
|
23
|
Chen J, Wang H, Peng F, Qiao H, Liu L, Wang L, Shang B. Ano1 is a Prognostic Biomarker That is Correlated with Immune Infiltration in Colorectal Cancer. Int J Gen Med 2022; 15:1547-1564. [PMID: 35210827 PMCID: PMC8858027 DOI: 10.2147/ijgm.s348296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 01/21/2022] [Indexed: 11/23/2022] Open
Affiliation(s)
- Jun Chen
- Laboratory Animal Center, Dalian Medical University, Dalian, 116044, Liaoning Province, People’s Republic of China
| | - Hongli Wang
- Cardiology Department, The Second Hospital of Dalian Medical University, Dalian, 116023, Liaoning Province, People’s Republic of China
| | - Fang Peng
- Pathology Department, The Second Hospital of Dalian Medical University, Dalian, 116023, Liaoning Province, People’s Republic of China
| | - Haiyan Qiao
- Laboratory Animal Center, Dalian Medical University, Dalian, 116044, Liaoning Province, People’s Republic of China
| | - Linfeng Liu
- Laboratory Animal Center, Dalian Medical University, Dalian, 116044, Liaoning Province, People’s Republic of China
| | - Liang Wang
- Laboratory Animal Center, Dalian Medical University, Dalian, 116044, Liaoning Province, People’s Republic of China
| | - Bingbing Shang
- Emergency Department, The Second Hospital of Dalian Medical University, Dalian, 116023, Liaoning Province, People’s Republic of China
- Correspondence: Bingbing Shang; Liang Wang, Tel +86-17709875175; +86-13332225676, Email ;
| |
Collapse
|
24
|
Liu W, Lin W, Yu L. Long non-coding RNA muscleblind like splicing regulator 1 antisense RNA 1 (LncRNA MBNL1-AS1) promotes the progression of acute myocardial infarction by regulating the microRNA-132-3p/SRY-related high-mobility-group box 4 (SOX4) axis. Bioengineered 2022; 13:1424-1435. [PMID: 34978261 PMCID: PMC8805923 DOI: 10.1080/21655979.2021.2018974] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 12/10/2021] [Indexed: 12/18/2022] Open
Abstract
Long non-coding RNA muscleblind like splicing regulator 1 antisense RNA 1 (LncRNA MBNL1-AS1) exerts vital role in various physiological processes. However, its functions in acute myocardial infarction (AMI) are not elucidated. AMI model was constructed using Wistar rats and it was found that LncRNA MBNL1-AS1 was upregulated in AMI model according to the quantitative real-time polymerase chain reaction (qRT-PCR) results. The left ventricular systolic pressure (LVSP), left ventricular end diastolic pressure (LVEDP) and maximum rate of rise/fall of left ventricle pressure (±dp/dt max) were detected through hemodynamics test, which showed that knockdown of MBNL1-AS1 improved cardiac function in AMI model. Next, the myocardial infarction area was estimated by triphenyltetrazole chloride (TTC) staining, and the levels of cardiac troponin I (cTn-I) and creatine kinase-MB (CK-MB) were detected by enzyme-linked immunosorbent assay (ELISA) kit. The results revealed that silencing MBLN1-AS1 alleviated myocardial injury in AMI model. Additionally, MBNL1-AS1 knockdown inhibited apoptosis of myocardial cells and reduced the expression of apoptotic proteins. According to DIANA database and luciferase reporter assay, miR-132-3p was the direct target of MBNL1-AS1 and was negatively regulated by MBNL1-AS1. Furthermore, Targetscan database predicted that SRY-related high-mobility-group box 4 (SOX4) was the direct target of miR-132-3p and was regulated by MBNL1-AS1 through miR-132-3p. Moreover, overexpression of SOX4 partially eliminated effects of MBNL1-AS1 on myocardial cells. In conclusion, this investigation for the first time revealed that LncRNA MBNL1-AS1 was the potential target for treating AMI and expounded the underlying mechanisms of it.
Collapse
Affiliation(s)
- Weifeng Liu
- Department of Cardiology, Yantai Yuhuangding Hospital, Qingdao Medical College, Qingdao University, Yantai, China
| | - Wenyuan Lin
- Department of Cardiology, Yantai Yuhuangding Hospital, Qingdao Medical College, Qingdao University, Yantai, China
| | - Liangliang Yu
- Department of Cardiology, Yantai Yuhuangding Hospital, Qingdao Medical College, Qingdao University, Yantai, China
| |
Collapse
|
25
|
Bai W, Liu M, Xiao Q. The diverse roles of TMEM16A Ca 2+-activated Cl - channels in inflammation. J Adv Res 2021; 33:53-68. [PMID: 34603778 PMCID: PMC8463915 DOI: 10.1016/j.jare.2021.01.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 12/17/2020] [Accepted: 01/24/2021] [Indexed: 12/14/2022] Open
Abstract
Background Transmembrane protein 16A (TMEM16A) Ca2+-activated Cl- channels have diverse physiological functions, such as epithelial secretion of Cl- and fluid and sensation of pain. Recent studies have demonstrated that TMEM16A contributes to the pathogenesis of infectious and non-infectious inflammatory diseases. However, the role of TMEM16A in inflammation has not been clearly elucidated. Aim of review In this review, we aimed to provide comprehensive information regarding the roles of TMEM16A in inflammation by summarizing the mechanisms underlying TMEM16A expression and activation under inflammatory conditions, in addition to exploring the diverse inflammatory signaling pathways activated by TMEM16A. This review attempts to develop the idea that TMEM16A plays a diverse role in inflammatory processes and contributes to inflammatory diseases in a cellular environment-dependent manner. Key scientific concepts of review Multiple inflammatory mediators, including cytokines (e.g., interleukin (IL)-4, IL-13, IL-6), histamine, bradykinin, and ATP/UTP, as well as bacterial and viral infections, promote TMEM16A expression and/or activity under inflammatory conditions. In addition, TMEM16A activates diverse inflammatory signaling pathways, including the IP3R-mediated Ca2+ signaling pathway, the NF-κB signaling pathway, and the ERK signaling pathway, and contributes to the pathogenesis of many inflammatory diseases. These diseases include airway inflammatory diseases, lipopolysaccharide-induced intestinal epithelial barrier dysfunction, acute pancreatitis, and steatohepatitis. TMEM16A also plays multiple roles in inflammatory processes by increasing vascular permeability and leukocyte adhesion, promoting inflammatory cytokine release, and sensing inflammation-induced pain. Furthermore, TMEM16A plays its diverse pathological roles in different inflammatory diseases depending on the disease severity, proliferating status of the cells, and its interacting partners. We herein propose cellular environment-dependent mechanisms that explain the diverse roles of TMEM16A in inflammation.
Collapse
Affiliation(s)
- Weiliang Bai
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Mei Liu
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Qinghuan Xiao
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China
| |
Collapse
|
26
|
Wang T, Wang H, Yang F, Gao K, Luo S, Bai L, Ma K, Liu M, Wu S, Wang H, Chen Z, Xiao Q. Honokiol inhibits proliferation of colorectal cancer cells by targeting anoctamin 1/TMEM16A Ca 2+ -activated Cl - channels. Br J Pharmacol 2021; 178:4137-4154. [PMID: 34192810 DOI: 10.1111/bph.15606] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 05/25/2021] [Accepted: 06/06/2021] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND AND PURPOSE Ca2+ -activated Cl- channels (Ano1 channels) contribute to the pathogenesis of colorectal cancer. Honokiol is known to inhibit cell proliferation and tumour growth in colorectal cancer. However, the molecular target of honokiol remains unclear. This study aimed to investigate whether honokiol inhibited cell proliferation of colorectal cancer by targeting Ano1 channels. EXPERIMENTAL APPROACH Patch-clamp techniques were performed to study the effect of honokiol on Ca2+ -activated Cl- currents in HEK293 cells overexpressing Ano1- or Ano2-containing plasmids or in human colorectal carcinoma SW620 cells. Site-directed mutagenesis was used to identify the critical residues for honokiol-induced Ano1 inhibition. Proliferation of SW620 cells or human intestinal epithelial NCM460 cells by CCK-8 assays. KEY RESULTS Honokiol blocked Ano1 currents in Ano1-overexpressing HEK293 cells and SW620 cells. Honokiol more potently inhibited Ano1 currents than Ano2 currents. Three amino acids (R429, K430 and N435) were critical for honokiol-induced Ano1 inhibition. The R429A/K430L/N435G mutation reduced the sensitivity of Ano1 to honokiol. Honokiol inhibited SW620 cell proliferation, and this effect was reduced by Ano1-shRNAs. Furthermore, Ano1 overexpression promoted proliferation in NCM460 cells with low Ano1 endogenous expression and resulted in an increased sensitivity to honokiol. Overexpression of the R429A/K430L/N435G mutation reduced WT Ano1-induced increase in the sensitivity of NCM460 cells to honokiol. CONCLUSION AND IMPLICATIONS We identified a new anticancer mechanism of honokiol, through the inhibition of cell proliferation, by targeting Ano1 Ca2+ -activated Cl- channels.
Collapse
Affiliation(s)
- Tianyu Wang
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Hui Wang
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Fan Yang
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Kuan Gao
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Shuya Luo
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Lichuan Bai
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Ke Ma
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Mei Liu
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Shuwei Wu
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Huijie Wang
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Zaixing Chen
- Pharmaceutical Teaching and Experimental Center, School of Pharmacy, China Medical University, Shenyang, China
| | - Qinghuan Xiao
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| |
Collapse
|
27
|
Liu Y, Liu Z, Wang K. The Ca 2+-activated chloride channel ANO1/TMEM16A: An emerging therapeutic target for epithelium-originated diseases? Acta Pharm Sin B 2021; 11:1412-1433. [PMID: 34221860 PMCID: PMC8245819 DOI: 10.1016/j.apsb.2020.12.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/19/2020] [Accepted: 09/14/2020] [Indexed: 02/07/2023] Open
Abstract
Anoctamin 1 (ANO1) or TMEM16A gene encodes a member of Ca2+ activated Cl– channels (CaCCs) that are critical for physiological functions, such as epithelial secretion, smooth muscle contraction and sensory signal transduction. The attraction and interest in ANO1/TMEM16A arise from a decade long investigations that abnormal expression or dysfunction of ANO1 is involved in many pathological phenotypes and diseases, including asthma, neuropathic pain, hypertension and cancer. However, the lack of specific modulators of ANO1 has impeded the efforts to validate ANO1 as a therapeutic target. This review focuses on the recent progress made in understanding of the pathophysiological functions of CaCC ANO1 and the current modulators used as pharmacological tools, hopefully illustrating a broad spectrum of ANO1 channelopathy and a path forward for this target validation.
Collapse
Key Words
- ANO1
- ANO1, anoctamin-1
- ASM, airway smooth muscle
- Ang II, angiotensin II
- BBB, blood–brain barrier
- CAMK, Ca2+/calmodulin-dependent protein kinase
- CF, cystic fibrosis
- CFTR, cystic fibrosis transmembrane conductance regulator
- Ca2+-activated Cl– channels (CaCCs)
- CaCCinh-A01
- CaCCs, Ca2+ activated chloride channels
- Cancer
- Cystic fibrosis
- DRG, dorsal root ganglion
- Drug target
- EGFR, epidermal growth factor receptor
- ENaC, epithelial sodium channels
- ER, endoplasmic reticulum
- ESCC, esophageal squamous cell carcinoma
- FRT, fisher rat thyroid
- GI, gastrointestinal
- GIST, gastrointestinal stromal tumor
- GPCR, G-protein coupled receptor
- HNSCC, head and neck squamous cell carcinoma
- HTS, high-throughput screening
- ICC, interstitial cells of Cajal
- IPAH, idiopathic pulmonary arterial hypertension
- MAPK, mitogen-activated protein kinase
- NF-κB, nuclear factor κB
- PAH, pulmonary arterial hypertension
- PAR2, protease activated receptor 2
- PASMC, pulmonary artery smooth muscle cells
- PIP2, phosphatidylinositol 4,5-bisphosphate
- PKD, polycystic kidney disease
- T16Ainh-A01
- TGF-β, transforming growth factor-β
- TMEM16A
- VGCC, voltage gated calcium channel
- VRAC, volume regulated anion channel
- VSMC, vascular smooth muscle cells
- YFP, yellow fluorescent protein
Collapse
Affiliation(s)
- Yani Liu
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, Qingdao 266073, China
- Institute of Innovative Drugs, Qingdao University, Qingdao 266021, China
| | - Zongtao Liu
- Department of Clinical Laboratory, Qingdao Third People's Hospital, Qingdao 266041, China
| | - KeWei Wang
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, Qingdao 266073, China
- Institute of Innovative Drugs, Qingdao University, Qingdao 266021, China
- Corresponding authors.
| |
Collapse
|
28
|
Wang H, Wang T, Zhang Z, Fan Y, Zhang L, Gao K, Luo S, Xiao Q, Sun C. Simvastatin inhibits oral squamous cell carcinoma by targeting TMEM16A Ca 2+-activated chloride channel. J Cancer Res Clin Oncol 2021; 147:1699-1711. [PMID: 33755783 DOI: 10.1007/s00432-021-03575-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 02/18/2021] [Indexed: 12/17/2022]
Abstract
PURPOSE Ca2+-activated chloride channel TMEM16A has been found to be overexpressed in many cancers including head and neck squamous cell carcinoma (HNSCC). Nevertheless, the role of TMEM16A in oral squamous cell carcinoma (OSCC) remains unclear. Although simvastatin is known to produce anti-tumor effect, the mechanisms by which simvastatin inhibits cancer remain unclear. METHODS In this study, we explored the role of TMEM16A expression in human OSCC tissues using both TCGA dataset and immunohistochemistry. CCK-8 assay was applied to evaluate cell proliferation. Patch clamp technique was applied to record TMEM16A Cl- currents. RESULTS We found that high TMEM16A expression is related with large tumor size, lymph node metastasis, and poor clinical outcome in patients with OSCC. In addition, TMEM16A overexpression could promote cell proliferation, and inhibition of TMEM16A channel activities could suppress cell proliferation in OSCC cells. Furthermore, simvastatin could suppress TMEM16A channel activities, and inhibited cell proliferation in OSCC cells via TMEM16A. CONCLUSION Our findings identify a novel anti-tumor mechanism of simvastatin by targeting TMEM16A. Simvastatin may represent an innovative strategy for treating OSCC with high TMEM16A expression.
Collapse
Affiliation(s)
- Hechen Wang
- Liaoning Provincial Key Laboratory of Oral Diseases, Department of Oromaxillofacial-Head and Neck Surgery, School and Hospital of Stomatology, China Medical University, 117 Nanjing Bei Jie, Heping District, Shenyang,, 110002, Liaoning, China.,Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning Province, China
| | - Tianyu Wang
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning Province, China
| | - Zeying Zhang
- Liaoning Provincial Key Laboratory of Oral Diseases, Department of Oromaxillofacial-Head and Neck Surgery, School and Hospital of Stomatology, China Medical University, 117 Nanjing Bei Jie, Heping District, Shenyang,, 110002, Liaoning, China
| | - Yu Fan
- Liaoning Provincial Key Laboratory of Oral Diseases, Department of Pathology, School and Hospital of Stomatology, China Medical University, Shenyang, China
| | - Lan Zhang
- Liaoning Provincial Key Laboratory of Oral Diseases, Hospital Infection Management Office, School and Hospital of Stomatology, China Medical University, Shenyang, China
| | - Kuan Gao
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning Province, China
| | - Shuya Luo
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning Province, China
| | - Qinghuan Xiao
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning Province, China.
| | - Changfu Sun
- Liaoning Provincial Key Laboratory of Oral Diseases, Department of Oromaxillofacial-Head and Neck Surgery, School and Hospital of Stomatology, China Medical University, 117 Nanjing Bei Jie, Heping District, Shenyang,, 110002, Liaoning, China.
| |
Collapse
|
29
|
Zheng G, Zhang G, Zhao Y, Zheng Z. Screening of miRNAs as Prognostic Biomarkers for Colon Adenocarcinoma and Biological Function Analysis of Their Target Genes. Front Oncol 2021; 11:560136. [PMID: 33816220 PMCID: PMC8017316 DOI: 10.3389/fonc.2021.560136] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 01/28/2021] [Indexed: 12/24/2022] Open
Abstract
We constructed a prognostic risk model for colon adenocarcinoma (COAD) using microRNAs (miRNAs) as biomarkers. Clinical data of patients with COADs and miRNA-seq data were from TCGA, and the differential expression of miRNAs (carcinoma vs. para-carcinoma tissues) was assessed using R software. COAD data were randomly divided into Training and Testing Sets. A linear prognostic risk model was constructed using Cox regression analysis based on the Training Set. Patients were classified as high-risk or low-risk according to the score of the prognostic model. Survival analysis and receiver operating characteristic (ROC) curves were used to evaluate model performance. The gene targets in the prognostic model were identified and their biological functions were analyzed. Analysis of COAD and normal cell lines using qPCR was used to verify the model. There were 134 up-regulated and 140 down-regulated miRNAs. We used the Training Set to develop a prognostic model based on the expression of seven miRNAs. ROC analysis indicated this model had acceptable prediction accuracy (area under the curve=0.784). Kaplan-Meier survival analysis showed that overall survival was worse in the high-risk group. Cox regression analysis showed that the 7-miRNA Risk Score was an independent prognostic factor. The 2,863 predicted target genes were mainly enriched in the MAPK, PI3K-AKT, proteoglycans in cancer, and mTOR signaling pathways. For unknown reasons, expression of these miRNAs in cancerous and normal cells differed somewhat from model predictions. Regardless, the 7-miRNA Risk Score can be used to predict COAD prognosis and may help to guide clinical treatment.
Collapse
Affiliation(s)
- Guoliang Zheng
- Department of Gastric Surgery, Cancer Hospital of China Medical University (Liaoning Cancer Hospital and Institute), Shenyang, China
| | - GuoJun Zhang
- Department of Pathophysiology, College of Basic Medicine Science, China Medical University, Shenyang, China
| | - Yan Zhao
- Department of Gastric Surgery, Cancer Hospital of China Medical University (Liaoning Cancer Hospital and Institute), Shenyang, China
| | - Zhichao Zheng
- Department of Gastric Surgery, Cancer Hospital of China Medical University (Liaoning Cancer Hospital and Institute), Shenyang, China
| |
Collapse
|
30
|
Zhang W, Wang Z, Cai G, Huang P. Circ_DOCK1 regulates USP11 through miR-132-3p to control colorectal cancer progression. World J Surg Oncol 2021; 19:67. [PMID: 33685455 PMCID: PMC7941900 DOI: 10.1186/s12957-021-02173-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 02/18/2021] [Indexed: 12/12/2022] Open
Abstract
Background Circular RNAs (circRNAs) take part in colorectal cancer malignancies. CircRNA dedicator of cytokinesis 1 (circ_DOCK1) is involved in colorectal cancer progression, but the mechanism underlying this circRNA that takes part in colorectal cancer development remains largely undetermined. Methods Tumor and normal para-cancerous tissues were collected from 42 colorectal cancer patients. Human colorectal cancer cell lines (HCT116 and SW480) were used for the experiments in vitro. Circ_DOCK1, microRNA (miR)-132-3p, and ubiquitin-specific protease 11 (USP11) levels were measured through quantitative real-time polymerase chain reaction and Western blotting. Cell growth, metastasis, and apoptosis were investigated via colony formation, 5-ethynyl-2′-deoxyuridine (EdU) staining, MTT, flow cytometry, Western blotting, and transwell analyses. The target association was evaluated via dual-luciferase reporter analysis, RNA pull-down, and immunoprecipitation (RIP). Xenograft assay was performed using HCT116 cells. USP11 and Ki67 levels in tumor tissues were detected via immunohistochemistry. Results Circ_DOCK1 expression was enhanced in colorectal cancer tissues and cells. Silencing circ_DOCK1 repressed cell growth, migration, and invasion, and facilitated apoptosis. Circ_DOCK1 sponged miR-132-3p, and miR-132-3p silence mitigated the effect of circ_DOCK1 interference on cell growth, metastasis, and apoptosis. MiR-132-3p targeted USP11, and circ_DOCK1 could regulate USP11 level by miR-132-3p. MiR-132-3p suppressed cell growth, metastasis, and apoptosis, and USP11 attenuated these effects. Knockdown of circ_DOCK1 decreased colorectal cancer cell xenograft tumor growth. Conclusion Circ_DOCK1 interference suppressed cell growth and metastasis, and increased apoptosis of colorectal cancer via decreasing USP11 by increasing miR-132-3p. Supplementary Information The online version contains supplementary material available at 10.1186/s12957-021-02173-x.
Collapse
Affiliation(s)
- Weitong Zhang
- Department of Anorectal Surgery, Hainan General Hospital, No. 19 Xiuhua Road, Haikou, 570311, Hainan, China
| | - Zhenfen Wang
- Department of Anorectal Surgery, Hainan General Hospital, No. 19 Xiuhua Road, Haikou, 570311, Hainan, China
| | - Guohao Cai
- Department of Anorectal Surgery, Hainan General Hospital, No. 19 Xiuhua Road, Haikou, 570311, Hainan, China
| | - Ping Huang
- Department of Anorectal Surgery, Hainan General Hospital, No. 19 Xiuhua Road, Haikou, 570311, Hainan, China.
| |
Collapse
|
31
|
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
|
32
|
Chen W, Gu M, Gao C, Chen B, Yang J, Xie X, Wang X, Sun J, Wang J. The Prognostic Value and Mechanisms of TMEM16A in Human Cancer. Front Mol Biosci 2021; 8:542156. [PMID: 33681289 PMCID: PMC7930745 DOI: 10.3389/fmolb.2021.542156] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 01/11/2021] [Indexed: 12/24/2022] Open
Abstract
As a calcium ion-dependent chloride channel transmembrane protein 16A (TMEM16A) locates on the cell membrane. Numerous research results have shown that TMEM16A is abnormally expressed in many cancers. Mechanically, TMEM16A participates in cancer proliferation and migration by affecting the MAPK and CAMK signaling pathways. Additionally, it is well documented that TMEM16A exerts a regulative impact on the hyperplasia of cancer cells by interacting with EGFR in head and neck squamous cell carcinoma (HNSCC), an epithelial growth factor receptor in head and neck squamous cell carcinoma respectively. Meanwhile, as an EGFR activator, TMEM16A is considered as an oncogene or a tumor-promoting factor. More and more experimental data showed that down-regulation of TMEM16A or gene targeted therapy may be an effective treatment for cancer. This review summarized its role in various cancers and research advances related to its clinical application included treatment and diagnosis.
Collapse
Affiliation(s)
- Wenjian Chen
- Anhui Province Children's Hospital Affiliated to Anhui Medical University, Hefei, China.,School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, China
| | - Meng Gu
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, China
| | - Chaobing Gao
- Department of Otorhinolaryngology, Head and Neck Surgery, The First Affiliated Hospital of AnHui Medical University, Hefei, China
| | - Bangjie Chen
- First Clinical Medical College of Anhui Medical University, Hefei, China
| | - Junfa Yang
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, China
| | - Xiaoli Xie
- Anhui Medicine Centralized Procurement Service Center, Hefei, China
| | - Xinyi Wang
- First Clinical Medical College of Anhui Medical University, Hefei, China
| | - Jun Sun
- Anhui Province Children's Hospital Affiliated to Anhui Medical University, Hefei, China
| | - Jinian Wang
- Department of Neurosurgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
33
|
Dos Santos IL, Penna KGBD, Dos Santos Carneiro MA, Libera LSD, Ramos JEP, Saddi VA. Tissue micro-RNAs associated with colorectal cancer prognosis: a systematic review. Mol Biol Rep 2021; 48:1853-1867. [PMID: 33598796 DOI: 10.1007/s11033-020-06075-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 12/10/2020] [Indexed: 01/05/2023]
Abstract
Colorectal cancer (CRC) is a multifactorial disease commonly diagnosed worldwide, with high mortality rates. Several studies demonstrate important associations between differential expression of micro-RNAs (miRs) and the prognosis of CRC. The present study aimed to identify differentially expressed tissue miRs associated with prognostic factors in CRC patients, through a systematic review of the Literature. Using the PubMed database, Cochrane Library and Web of Science, studies published in English evaluating miRs differentially expressed in tumor tissue and significantly associated with the prognostic aspects of CRC were selected. All the included studies used RT-PCR (Taqman or SYBR Green) for miR expression analysis and the period of publication was from 2009 to 2018. A total of 115 articles accomplished the inclusion criteria and were included in the review. The studies investigated the expression of 100 different miRs associated with prognostic aspects in colorectal cancer patients. The most frequent oncogenic miRs investigated were miR-21, miR-181a, miR-182, miR-183, miR-210 and miR-224 and the hyperexpression of these miRs was associated with distant metastasis, lymph node metastasis and worse survival in patients with CRC. The most frequent tumor suppressor miRs were miR-126, miR-199b and miR-22 and the hypoexpression of these miRs was associated with distant metastasis, worse prognosis and a higher risk of disease relapse (worse disease-free survival). Specific tissue miRs are shown to be promising prognostic biomarkers in patients with CRC, given their strong association with the prognostic aspects of these tumors, however, new studies are necessary to establish the sensibility and specificity of the individual miRs in order to use them in clinical practice.
Collapse
Affiliation(s)
- Igor Lopes Dos Santos
- Programa de Mestrado em Ciências Ambientais e Saúde da Pontifícia Universidade Católica de Goiás, Laboratório de Genética e Biodiversidade, Escola de Ciências Médicas, Farmacêuticas e Biomédicas da Pontifícia Universidade Católica de Goiás, Área IV, Praça Universitária, 1440, Setor Leste Universitário, Goiânia, GO, 74605-010, Brazil.
| | - Karlla Greick Batista Dias Penna
- Programa de Mestrado em Ciências Ambientais e Saúde da Pontifícia Universidade Católica de Goiás, Laboratório de Genética e Biodiversidade, Escola de Ciências Médicas, Farmacêuticas e Biomédicas da Pontifícia Universidade Católica de Goiás, Área IV, Praça Universitária, 1440, Setor Leste Universitário, Goiânia, GO, 74605-010, Brazil
| | | | | | - Jéssica Enocencio Porto Ramos
- Programa de Mestrado em Ciências Ambientais e Saúde da Pontifícia Universidade Católica de Goiás, Laboratório de Genética e Biodiversidade, Escola de Ciências Médicas, Farmacêuticas e Biomédicas da Pontifícia Universidade Católica de Goiás, Área IV, Praça Universitária, 1440, Setor Leste Universitário, Goiânia, GO, 74605-010, Brazil
| | - Vera Aparecida Saddi
- Programa de Mestrado em Ciências Ambientais e Saúde da Pontifícia Universidade Católica de Goiás, Laboratório de Genética e Biodiversidade, Escola de Ciências Médicas, Farmacêuticas e Biomédicas da Pontifícia Universidade Católica de Goiás, Área IV, Praça Universitária, 1440, Setor Leste Universitário, Goiânia, GO, 74605-010, Brazil
| |
Collapse
|
34
|
Cui X, Jing X, Liu J, Bi X, Wu X. miR‑132 is upregulated in polycystic ovarian syndrome and inhibits granulosa cells viability by targeting Foxa1. Mol Med Rep 2020; 22:5155-5162. [PMID: 33174054 PMCID: PMC7646966 DOI: 10.3892/mmr.2020.11590] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 09/15/2020] [Indexed: 11/24/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is one of the most common endocrine metabolic disorders characterized by hyperandrogenism, polycystic ovaries and ovulatory dysfunction. Several studies have suggested that the aberrant expression of microRNAs (miRNAs/miRs) plays an important role in the pathogenesis of PCOS; however, the role and underlying mechanisms of miR-132 in the development of PCOS remain unclear. In the present study, the expression of miR-132 in granulosa cells (GCs) derived from 26 patients with PCOS and 30 healthy controls was detected by reverse transcription-quantitative PCR (RT-qPCR). The apoptosis of GCs was examined using a TUNEL assay. The human ovarian granulosa-like tumor cell line, KGN, was cultured for Cell Counting Kit-8 assays following the overexpression or knockdown of miR-132. TargetScan was applied to identify the potential targets of miR-132, which was further verified by a luciferase assay, RT-qPCR and western blotting. The expression of miR-132 was decreased in GCs from patients with PCOS. Moreover, the GCs of patients with PCOS exhibited significantly increased apoptotic nuclei. Furthermore, the overexpression of miR-132 inhibited the viability of KGN cells. In addition, the results verified that miR-132 directly targeted forkhead box protein A1 (Foxa1), the knockdown of which suppressed KGN cell viability. On the whole, the findings of the present study demonstrated that miR-132 inhibited cell viability and induced apoptosis by directly interacting with Foxa1. Thus, miR-132 may be a potential target for the treatment of patients with PCOS.
Collapse
Affiliation(s)
- Xiangrong Cui
- Reproductive Medicine Center, Shanxi Women and Infants Hospital, Affiliate of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Xuan Jing
- Department of Clinical Laboratory, Shanxi Provincial People's Hospital, Affiliate of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Junfen Liu
- Reproductive Medicine Center, Shanxi Women and Infants Hospital, Affiliate of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Xingyu Bi
- Reproductive Medicine Center, Shanxi Women and Infants Hospital, Affiliate of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Xueqing Wu
- Reproductive Medicine Center, Shanxi Women and Infants Hospital, Affiliate of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| |
Collapse
|
35
|
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
|
36
|
How Dysregulated Ion Channels and Transporters Take a Hand in Esophageal, Liver, and Colorectal Cancer. Rev Physiol Biochem Pharmacol 2020; 181:129-222. [PMID: 32875386 DOI: 10.1007/112_2020_41] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Over the last two decades, the understanding of how dysregulated ion channels and transporters are involved in carcinogenesis and tumor growth and progression, including invasiveness and metastasis, has been increasing exponentially. The present review specifies virtually all ion channels and transporters whose faulty expression or regulation contributes to esophageal, hepatocellular, and colorectal cancer. The variety reaches from Ca2+, K+, Na+, and Cl- channels over divalent metal transporters, Na+ or Cl- coupled Ca2+, HCO3- and H+ exchangers to monocarboxylate carriers and organic anion and cation transporters. In several cases, the underlying mechanisms by which these ion channels/transporters are interwoven with malignancies have been fully or at least partially unveiled. Ca2+, Akt/NF-κB, and Ca2+- or pH-dependent Wnt/β-catenin signaling emerge as cross points through which ion channels/transporters interfere with gene expression, modulate cell proliferation, trigger epithelial-to-mesenchymal transition, and promote cell motility and metastasis. Also miRs, lncRNAs, and DNA methylation represent potential links between the misexpression of genes encoding for ion channels/transporters, their malfunctioning, and cancer. The knowledge of all these molecular interactions has provided the basis for therapeutic strategies and approaches, some of which will be broached in this review.
Collapse
|
37
|
Non-coding RNAS and colorectal cancer liver metastasis. Mol Cell Biochem 2020; 475:151-159. [PMID: 32767228 DOI: 10.1007/s11010-020-03867-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 07/31/2020] [Indexed: 02/08/2023]
Abstract
More than 50% of colorectal cancer (CRC) deaths are attributed to metastasis, and the liver is the most common distant metastatic site of CRC. The molecular mechanisms underlying CRC liver metastasis are very complicated and remain largely unknown. Accumulated evidence has shown that non-coding RNAs (NcRNAs) play critical roles in tumor development and progression. Here we reviewed the roles and underlying mechanisms of NcRNAs in CRC liver metastasis.
Collapse
|
38
|
Liu Z, Zhao W, Ren Y, Liu C, Liu X, Xiao J. Comprehensive analysis of the long non-coding RNA-associated competitive endogenous RNA network reveals novel prognostic biomarkers in Wilms' tumor. Oncol Lett 2020; 19:3731-3742. [PMID: 32382326 PMCID: PMC7202313 DOI: 10.3892/ol.2020.11500] [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: 05/30/2019] [Accepted: 01/15/2020] [Indexed: 11/06/2022] Open
Abstract
Wilms' tumor (WT) is one of the most common types of renal carcinoma in children. The aim of the present study was to construct a competitive endogenous RNA (ceRNA) regulation network and explore novel prognostic biomarkers for WT. The expression profiles were downloaded from The Cancer Genome Atlas database to identify differentially expressed RNAs (DERNAs). Based on the interactions between microRNAs (miRNAs) and mRNAs/long non-coding RNAs (lncRNAs), a ceRNA network was constructed. Functional enrichment analyses were subsequently conducted to explore the functions of the ceRNA-associated DEmRNAs. Survival analysis was performed to screen for prognosis-associated RNAs and the χ2 test was used to assess the associations between prognosis-associated RNA expression and histology classification/clinical staging. The present study identified 1,784 lncRNAs, 114 miRNAs and 3,337 mRNAs, which were abnormally expressed in WT compared with that in normal samples. By prediction, pairing and network analysis, a ceRNA network consisting of 38 DElncRNAs, 18 DEmiRNAs and 99 DEmRNAs was established. These DEmRNAs were significantly enriched in pathways associated with the occurrence and development of WT. By combining the expression data with survival analysis, seven prognosis-associated RNAs were identified (P<0.05). Of these seven RNAs, two (zinc finger and BTB domain containing 4; and deleted in lymphocytic leukemia 2) were significantly associated with clinical staging and histology classification. Lastly, the expression levels of the seven RNAs were verified in the Gene Expression Omnibus database. The present study revealed that 7 RNAs might be considered as novel prognostic biomarkers and potential treatment targets for therapy in WT. In addition, the ceRNA regulation network could provide novel strategies for further studies on lncRNAs and miRNAs in WT.
Collapse
Affiliation(s)
- Zifeng Liu
- Department of Clinical Data Center, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Wenbo Zhao
- Department of Nephrology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Yuqing Ren
- Tianpeng Technology Co., Ltd, Guangzhou, Guangdong 510600, P.R. China
| | - Chang Liu
- Department of Clinical Data Center, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Xun Liu
- Department of Nephrology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Jian Xiao
- Department of Medical Oncology, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510655, P.R. China
| |
Collapse
|
39
|
Yang Y, Meng WJ, Wang ZQ. MicroRNAs in Colon and Rectal Cancer - Novel Biomarkers from Diagnosis to Therapy. Endocr Metab Immune Disord Drug Targets 2020; 20:1211-1226. [PMID: 32370729 DOI: 10.2174/1871530320666200506075219] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 01/31/2020] [Accepted: 02/01/2020] [Indexed: 02/05/2023]
Abstract
Colorectal cancer (CRC) is one of the most common cancers and a significant cause of tumor- related deaths worldwide. Traditional biomarkers, such as CEA and CA199, are not sensitive enough to provide useful information for early diagnosis and treatment and are rather used to track the clinical progression of the disease. There is growing evidence that microRNAs (miRNA) are potentially superior to traditional biomarkers as promising non-invasive biomarkers for the timely diagnosis and prediction of prognosis or treatment response in the management of CRC. In this review, the latest studies on the dysregulation of miRNAs expression in CRC and the potential for miRNAs to serve as biomarkers were collected. Given the limitations of miRNA, as discussed in this paper, its clinical applications as a diagnostic biomarker should be limited to use in combination with other biomarkers. Further research is necessary to elucidate the clinical applications of miRNA in therapy for CRC.
Collapse
Affiliation(s)
- Ying Yang
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wen-Jian Meng
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zi-Qiang Wang
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
40
|
Maimaitiming A, Wusiman A, Aimudula A, Kuerban X, Su P. MicroRNA-152 Inhibits Cell Proliferation, Migration, and Invasion in Breast Cancer. Oncol Res 2020; 28:13-19. [PMID: 30982494 PMCID: PMC7851537 DOI: 10.3727/096504019x15519249902838] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The aim of the present study was to investigate the roles of microRNA-152 (miR-152) in the initiation and progression of breast cancer. The expression level of miR-152 was detected in human breast cancer tissue and a panel of human breast cancer cell lines using qRT-PCR. Results found that miR-152 expression was significantly downregulated in breast cancer tissue samples compared to adjacent noncancerous tissues as well as in breast cancer cell lines. Overexpression of miR-152 significantly suppressed breast cancer cell proliferation, migration, and invasion. Luciferase reporter assay results found that ROCK1 is a direct and functional target gene of miR-152 in breast cancer. In addition, downexpression of ROCK1 could inhibit breast cancer cell proliferation, migration, and invasion. These findings indicate that miR-152 inhibited breast cancer growth and metastasis through negative regulation of ROCK1 expression. These data suggest that miR-152/ROCK1 pathway may be a useful therapeutic target for breast cancer treatment.
Collapse
Affiliation(s)
- Adilijiang Maimaitiming
- *Department of Breast and Thyroid Surgery, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, P.R. China
| | - Ailijiang Wusiman
- †Department of Surgery, Hospital of Xinjiang Traditional Uyghur, Urumqi, P.R. China
| | - Abulajiang Aimudula
- †Department of Surgery, Hospital of Xinjiang Traditional Uyghur, Urumqi, P.R. China
| | - Xuekelaiti Kuerban
- *Department of Breast and Thyroid Surgery, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, P.R. China
| | - Pengcheng Su
- *Department of Breast and Thyroid Surgery, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, P.R. China
| |
Collapse
|
41
|
Sabit H, Cevik E, Tombuloglu H. Colorectal cancer: The epigenetic role of microbiome. World J Clin Cases 2019; 7:3683-3697. [PMID: 31799293 PMCID: PMC6887622 DOI: 10.12998/wjcc.v7.i22.3683] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 10/23/2019] [Accepted: 10/30/2019] [Indexed: 02/05/2023] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer in men (746000 cases per year) and the second most common cancer in women globally (614000 cases per year). The incidence rate of CRC in developed countries (737000 cases per year) is higher than that in less developed countries (624000 cases per year). CRC can arise from genetic causes such as chromosomal instability and microsatellite instability. Several etiologic factors underlie CRC including age, diet, and lifestyle. Gut microbiota represent a proven cause of the disease, where they play pivotal roles in modulating and reshaping the host epigenome. Several active microbial metabolites have been found to drive carcinogenesis, invasion, and metastasis via modifying both the methylation landscape along with histone structure in intestinal cells. Gut microbiota, in response to diet, can exert both beneficial and harmful functions in humans, according to the intestinal balance of number and types of these bacteria. Although the intestinal microbial community is diverse among individuals, these microbes cumulatively produce 100-fold more proteins than the human genome itself, which calls for further studies to elaborate on the complicated interaction between these microorganisms and intestinal cells. Therefore, understanding the exact role that gut microbiota play in inducing CRC will help attain reliable strategies to precisely diagnose and treat this fatal disease.
Collapse
Affiliation(s)
- Hussein Sabit
- Department of Genetics, Institute for Medical Research and Consultations, Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia
| | - Emre Cevik
- Department of Genetics, Institute for Medical Research and Consultations, Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia
| | - Huseyin Tombuloglu
- Department of Genetics, Institute for Medical Research and Consultations, Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia
| |
Collapse
|
42
|
Lu P, Xu M, Xiong Z, Zhou F, Wang L. Fusobacterium nucleatum prevents apoptosis in colorectal cancer cells via the ANO1 pathway. Cancer Manag Res 2019; 11:9057-9066. [PMID: 31802939 PMCID: PMC6829176 DOI: 10.2147/cmar.s185766] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 12/10/2018] [Indexed: 12/21/2022] Open
Abstract
Objective : Chemotherapy failure derived from drug resistance is the most important reason causing the recurrence in colorectal cancer patients. Therefore, it is necessary to shed light on the mechanism of chemotherapy resistance in colorectal cancer patients. Methods : We looked into the contribution of Fusobacterium nucleatum and ANO1 to chemoresistance in the human colorectal carcinoma cell lines. We silence and overexpress ANO1 in HCT116 and HT29 cells with lentivirus and siRNA knockdown technique in the absence or presence of F. nucleatum, oxaliplatin or 5-fluorouracil (5-FU). ANO1, p-pg, cleaved PARP, cleaved caspase-3, and EGFR expression was measured by Western blot. Cell apoptosis was measured by flow cytometry. Results : We found that F. nucleatum promoted ANO1 expression on colon cancer cells. Moreover, ANO1 prevent colon cancer apoptosis from oxaliplatin and 5-FU. Additionally, knockdown ANO1 expression could block F. nucleatum protective effects and increase the apoptosis effects induced by oxaliplatin and 5-FU. Therefore, F. nucleatum might be biologically involved in the development of colon cancer chemoresistance via ANO1 pathway. Conclusions : Taken together, our findings provide a valuable insight into clinical management and therapy, which may ameliorate colorectal cancer patient outcomes.
Collapse
Affiliation(s)
- Pei Lu
- Department of Clinical Laboratory, Shanghai No. 8 People's Hospital, Shanghai, China
| | - Minyi Xu
- Department of Clinical Laboratory, Shanghai No. 8 People's Hospital, Shanghai, China
| | - Zhongbo Xiong
- Department of Clinical Laboratory, Shanghai No. 8 People's Hospital, Shanghai, China
| | - Fangfang Zhou
- Department of Clinical Laboratory, Shanghai No. 8 People's Hospital, Shanghai, China
| | - Lei Wang
- Department of Clinical Laboratory, Shanghai No. 8 People's Hospital, Shanghai, China
| |
Collapse
|
43
|
Tian YQ, Fan ZJ, Liu S, Wu YJ, Liu SY. Value of microRNAs in diagnosis and prognosis of colorectal cancer. Shijie Huaren Xiaohua Zazhi 2019; 27:1278-1284. [DOI: 10.11569/wcjd.v27.i20.1278] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Some new treatment methods have been explored to delay the recurrence of colorectal cancer (CRC). Early diagnosis plays an important role in the improvement of curative effect. The conventional methods used to diagnose and monitor CRC are fecal occult blood test (FOBT) and colonoscopy. However, FOBT has an unsatisfactory sensitivity, while colonoscopy is expensive and invasive. As new biomarkers, microRNAs, which can be detected in CRC tissues, cells, and body fluid as tumor suppressors or oncogenes, can be used in early diagnosis, the monitoring of metastasis and treatment, as well prognostic evaluation of CRC. This article reviews the diagnostic and prognostic value of microRNAs in CRC.
Collapse
Affiliation(s)
- Ya-Qiong Tian
- Third Central Hospital of Tianjin, Tianjin Key Laboratory of Artificial Cell, Artificial Cell Engineering Technology Research Center of Public Health Ministry, Tianjin 300170, China
| | - Zhi-Juan Fan
- Third Central Hospital of Tianjin, Tianjin Key Laboratory of Artificial Cell, Artificial Cell Engineering Technology Research Center of Public Health Ministry, Tianjin 300170, China
| | - Shuang Liu
- Third Central Hospital of Tianjin, Tianjin Key Laboratory of Artificial Cell, Artificial Cell Engineering Technology Research Center of Public Health Ministry, Tianjin 300170, China
| | - Yu-Jing Wu
- Third Central Hospital of Tianjin, Tianjin Key Laboratory of Artificial Cell, Artificial Cell Engineering Technology Research Center of Public Health Ministry, Tianjin 300170, China
| | - Shu-Ye Liu
- Third Central Hospital of Tianjin, Tianjin Key Laboratory of Artificial Cell, Artificial Cell Engineering Technology Research Center of Public Health Ministry, Tianjin 300170, China
| |
Collapse
|
44
|
Anderson KJ, Cormier RT, Scott PM. Role of ion channels in gastrointestinal cancer. World J Gastroenterol 2019; 25:5732-5772. [PMID: 31636470 PMCID: PMC6801186 DOI: 10.3748/wjg.v25.i38.5732] [Citation(s) in RCA: 133] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 07/26/2019] [Accepted: 09/27/2019] [Indexed: 02/06/2023] Open
Abstract
In their seminal papers Hanahan and Weinberg described oncogenic processes a normal cell undergoes to be transformed into a cancer cell. The functions of ion channels in the gastrointestinal (GI) tract influence a variety of cellular processes, many of which overlap with these hallmarks of cancer. In this review we focus on the roles of the calcium (Ca2+), sodium (Na+), potassium (K+), chloride (Cl-) and zinc (Zn2+) transporters in GI cancer, with a special emphasis on the roles of the KCNQ1 K+ channel and CFTR Cl- channel in colorectal cancer (CRC). Ca2+ is a ubiquitous second messenger, serving as a signaling molecule for a variety of cellular processes such as control of the cell cycle, apoptosis, and migration. Various members of the TRP superfamily, including TRPM8, TRPM7, TRPM6 and TRPM2, have been implicated in GI cancers, especially through overexpression in pancreatic adenocarcinomas and down-regulation in colon cancer. Voltage-gated sodium channels (VGSCs) are classically associated with the initiation and conduction of action potentials in electrically excitable cells such as neurons and muscle cells. The VGSC NaV1.5 is abundantly expressed in human colorectal CRC cell lines as well as being highly expressed in primary CRC samples. Studies have demonstrated that conductance through NaV1.5 contributes significantly to CRC cell invasiveness and cancer progression. Zn2+ transporters of the ZIP/SLC39A and ZnT/SLC30A families are dysregulated in all major GI organ cancers, in particular, ZIP4 up-regulation in pancreatic cancer (PC). More than 70 K+ channel genes, clustered in four families, are found expressed in the GI tract, where they regulate a range of cellular processes, including gastrin secretion in the stomach and anion secretion and fluid balance in the intestinal tract. Several distinct types of K+ channels are found dysregulated in the GI tract. Notable are hERG1 upregulation in PC, gastric cancer (GC) and CRC, leading to enhanced cancer angiogenesis and invasion, and KCNQ1 down-regulation in CRC, where KCNQ1 expression is associated with enhanced disease-free survival in stage II, III, and IV disease. Cl- channels are critical for a range of cellular and tissue processes in the GI tract, especially fluid balance in the colon. Most notable is CFTR, whose deficiency leads to mucus blockage, microbial dysbiosis and inflammation in the intestinal tract. CFTR is a tumor suppressor in several GI cancers. Cystic fibrosis patients are at a significant risk for CRC and low levels of CFTR expression are associated with poor overall disease-free survival in sporadic CRC. Two other classes of chloride channels that are dysregulated in GI cancers are the chloride intracellular channels (CLIC1, 3 & 4) and the chloride channel accessory proteins (CLCA1,2,4). CLIC1 & 4 are upregulated in PC, GC, gallbladder cancer, and CRC, while the CLCA proteins have been reported to be down-regulated in CRC. In summary, it is clear, from the diverse influences of ion channels, that their aberrant expression and/or activity can contribute to malignant transformation and tumor progression. Further, because ion channels are often localized to the plasma membrane and subject to multiple layers of regulation, they represent promising clinical targets for therapeutic intervention including the repurposing of current drugs.
Collapse
Affiliation(s)
- Kyle J Anderson
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, United States
| | - Robert T Cormier
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, United States
| | - Patricia M Scott
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, United States
| |
Collapse
|
45
|
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
|
46
|
Wei XC, Lv ZH. MicroRNA-132 inhibits migration, invasion and epithelial-mesenchymal transition via TGFβ1/Smad2 signaling pathway in human bladder cancer. Onco Targets Ther 2019; 12:5937-5945. [PMID: 31413591 PMCID: PMC6662166 DOI: 10.2147/ott.s201731] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 03/25/2019] [Indexed: 01/01/2023] Open
Abstract
Background and aim: Increasing evidence shows that microRNAs play an important regulatory role in the development of several types of cancers. However, the role of microRNA-132 (miR-132) in human bladder cancer (BC) metastasis remains unclear. In this research, we aimed to investigate the effect of miR-132 on the cell migration and relate potential mechanism in BC. Methods: miR-132 expression level was assessed by quantitative real-time PCR (qRT-PCR) in 32 BC tissues and BC cell lines (T24). The function of miR-132 was evaluated by Transwell assay. Gene expression was determined by using qRT-PCR or Western blot. Results: The results showed that miR-132 had a lower expression in BC tissues than in adjacent normal tissues. At the same time, compared to human normal urethral epithelium cells, the expression level of miR-132 was downregulated in T24 cell lines. miR-132 overexpression significantly inhibited migration and invasion capacities in T24 cells, while downregulation of miR-132 expression strengthened such capacities. Compared with those transfected with miR-132 mimic, EMT-related markers and TGFβ1/Smad2 expression levels were higher in T24 cells transfected with miR-132 inhibitor. Moreover, EMT-related markers and Smad2 expression levels was obviously increased in BC tissues compared to the adjacent normal tissues. The correlation result indicated that the expression of miR-132 and Smad2 was reversed. Conclusion: In short, our results suggest that miR-132 may play a suppressive role in the metastasis of BC cells via TGFβ1/Smad2 signaling pathway.
Collapse
Affiliation(s)
- Xi Chao Wei
- Department of Urology, Jining Hospital of Traditional Chinese Medicine, Jining 272000, Shandong, People's Republic of China
| | - Zhong Hua Lv
- Department of Urology, Jining No. 1 People's Hospital, Jining 272011, Shandong, People's Republic of China
| |
Collapse
|
47
|
Wang H, Yao F, Luo S, Ma K, Liu M, Bai L, Chen S, Song C, Wang T, Du Q, Wu H, Wei M, Fang Y, Xiao Q. A mutual activation loop between the Ca 2+-activated chloride channel TMEM16A and EGFR/STAT3 signaling promotes breast cancer tumorigenesis. Cancer Lett 2019; 455:48-59. [PMID: 31042586 DOI: 10.1016/j.canlet.2019.04.027] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Revised: 04/19/2019] [Accepted: 04/25/2019] [Indexed: 10/26/2022]
Abstract
The Ca2+-activated chloride channel TMEM16A (anoctamin 1) is overexpressed in breast cancer. It remains unclear how TMEM16A overexpression plays a role in carcinogenesis in breast cancer. In this study, we found that high TMEM16A expression in combination with high EGFR or STAT3 expression was significantly associated with shorter overall survival in ER-positive breast cancer patients without tamoxifen treatment, and longer overall survival in patients with tamoxifen treatment. EGFR/STAT3 signaling activation by EGF promoted TMEM16A expression, and TMEM16A overexpression activated EGFR/STAT3 signaling in breast cancer cells. Both in vitro and in animal studies showed that TMEM16A overexpression promoted, and TMEM16A knockdown inhibited breast cancer cell proliferation and tumor growth. In addition, TMEM16A overexpression-induced cell proliferation was blocked by EGFR/STAT3 inhibitors, and TMEM16A knockdown reduced EGF-induced proliferation and tumorigenesis in breast cancer. Furthermore, inhibition of TMEM16A channel function effectively reduced breast cancer cell proliferation, especially in combination with EGFR inhibitors. Our findings identify a mutual activation loop between TMEM16A and EGFR/STAT3 signaling, which is important for breast cancer proliferation and growth. TMEM16A inhibition may represent a novel therapy for EGFR-expressing breast cancer.
Collapse
Affiliation(s)
- Hui Wang
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Fan Yao
- Department of Breast Surgery and Surgical Oncology, Research Unit of General Surgery, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Shuya Luo
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Ke Ma
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Mei Liu
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Lichuan Bai
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Si Chen
- Department of Microbial and Biochemical Pharmacy, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Chang Song
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Tianyu Wang
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Qiang Du
- Department of Microbial and Biochemical Pharmacy, School of Pharmacy, China Medical University, 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
| | - Yue Fang
- Department of Microbial and Biochemical Pharmacy, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Qinghuan Xiao
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China.
| |
Collapse
|
48
|
Liu Y, Zhang M. miR-132 Regulates Adriamycin Resistance in Colorectal Cancer Cells Through Targeting Extracellular Signal-Regulated Kinase 1. Cancer Biother Radiopharm 2019; 34:398-404. [PMID: 30939040 DOI: 10.1089/cbr.2018.2749] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Objective: Extracellular signal-regulated kinase 1 (ERK1) is an important signal transduction molecule in the ERK/mitogen-activated protein kinase pathway. miR-132 downregulation is associated with colorectal cancer (CRC). However, whether it is related to drug resistance remains poorly understood. Bioinformatics analysis demonstrated the targeting relationship between miR-132 and ERK1 3'-UTR. This study investigated the role of miR-132 in regulating ERK1 expression and affecting CRC cell proliferation, apoptosis, and adriamycin (ADM) resistance. Materials and Methods: Dual luciferase reporter gene assay was used to evaluate the targeted relationship between miR-132 and ERK1. ADM-resistant cell lines Lovo/ADM and SW480/ADM were established followed by analysis of miR-132 and ERK1 expression levels, and cell proliferation by cell counting kit-8 assay. The impact of ADM on cell proliferation and apoptosis was assessed by 5-bromodeoxyuridine (EdU) staining and flow cytometry, respectively. Lovo/ADM and SW480/ADM cells were cultured in vitro and divided into two groups, including miR-NC group and miR-132 mimic group. Results: There was a targeted regulatory relationship between miR-132 and ERK1 mRNA. The miR-132 expression was significantly lower, whereas ERK1 mRNA and protein expression levels were significantly higher in Lovo/ADM and SW480/ADM cells than those in Lovo and SW480 cells. Transfection of miR-132 mimic significantly reduced the expression of ERK1 in Lovo/ADM and SW480/ADM cells, enhanced cell apoptosis, and weakened cell proliferation. Conclusions: miR-132 reduction and ERK1 elevation are related to ADM resistance in CRC cells. Upregulation of miR-132 expression inhibits CRC cell proliferation, induces apoptosis, and reduces ADM resistance possibly by targeting ERK1 expression.
Collapse
Affiliation(s)
- Yong Liu
- 1Department of General Surgery, Yan'an People's Hospital, Yan'an, China
| | - Mei Zhang
- 2Department of Clinical Laboratory, The Fourth People's Hospital of Shaanxi, Xi'an, China
| |
Collapse
|
49
|
Wang C, Liu L, Zhu H, Zhang L, Wang R, Zhang Z, Huang J, Zhang S, Jian F, Ning C, Zhang L. MicroRNA expression profile of HCT-8 cells in the early phase of Cryptosporidium parvum infection. BMC Genomics 2019; 20:37. [PMID: 30642246 PMCID: PMC6332841 DOI: 10.1186/s12864-018-5410-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 12/26/2018] [Indexed: 12/20/2022] Open
Abstract
Background Cryptosporidium parvum is an important zoonotic parasitic disease worldwide, but the molecular mechanisms of the host–parasite interaction are not fully understood. Noncoding microRNAs (miRNAs) are considered key regulators of parasitic diseases. Therefore, we used microarray, qPCR, and bioinformatic analyses to investigate the intestinal epithelial miRNA expression profile after Cryptosporidium parvum infection. Results Twenty miRNAs were differentially expressed after infection (four upregulated and 16 downregulated). Further analysis of the differentially expressed miRNAs revealed that many important cellular responses were triggered by Cryptosporidium parvum infection, including cell apoptosis and the inflammatory and immune responses. Conclusions This study demonstrates for the first time that the miRNA expression profile of human intestinal epithelium cells is altered by C. parvum infection. This dysregulation of miRNA expression may contribute to the regulation of host biological processes in response to C. parvum infection, including cell apoptosis and the immune responses. These results provide new insight into the regulatory mechanisms of host miRNAs during cryptosporidiosis, which may offer potential targets for future C. parvum control strategies. Electronic supplementary material The online version of this article (10.1186/s12864-018-5410-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Chenrong Wang
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, People's Republic of China.,International Joint Research Laboratory for Zoonotic Diseases of Henan, Zhengzhou, 450002, People's Republic of China
| | - Limin Liu
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, People's Republic of China.,International Joint Research Laboratory for Zoonotic Diseases of Henan, Zhengzhou, 450002, People's Republic of China
| | - Huili Zhu
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, 453003, People's Republic of China
| | - Lu Zhang
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, People's Republic of China.,International Joint Research Laboratory for Zoonotic Diseases of Henan, Zhengzhou, 450002, People's Republic of China
| | - Rongjun Wang
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, People's Republic of China.,International Joint Research Laboratory for Zoonotic Diseases of Henan, Zhengzhou, 450002, People's Republic of China
| | - Zhenjie Zhang
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, People's Republic of China.,International Joint Research Laboratory for Zoonotic Diseases of Henan, Zhengzhou, 450002, People's Republic of China
| | - Jianying Huang
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, People's Republic of China.,International Joint Research Laboratory for Zoonotic Diseases of Henan, Zhengzhou, 450002, People's Republic of China
| | - Sumei Zhang
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, People's Republic of China.,International Joint Research Laboratory for Zoonotic Diseases of Henan, Zhengzhou, 450002, People's Republic of China
| | - Fuchun Jian
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, People's Republic of China.,International Joint Research Laboratory for Zoonotic Diseases of Henan, Zhengzhou, 450002, People's Republic of China
| | - Changshen Ning
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, People's Republic of China.,International Joint Research Laboratory for Zoonotic Diseases of Henan, Zhengzhou, 450002, People's Republic of China
| | - Longxian Zhang
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, People's Republic of China. .,International Joint Research Laboratory for Zoonotic Diseases of Henan, Zhengzhou, 450002, People's Republic of China.
| |
Collapse
|
50
|
Park YR, Lee ST, Kim SL, Zhu SM, Lee MR, Kim SH, Kim IH, Lee SO, Seo SY, Kim SW. Down-regulation of miR-9 promotes epithelial mesenchymal transition via regulating anoctamin-1 (ANO1) in CRC cells. Cancer Genet 2018; 231-232:22-31. [PMID: 30803553 DOI: 10.1016/j.cancergen.2018.12.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 09/21/2018] [Accepted: 12/23/2018] [Indexed: 12/19/2022]
Abstract
MicroRNA-9 (miR-9) has been reported to play a suppressive or promoting role according to cancer type. In this study, we investigated the effects of anoctamin-1 (ANO1) and miR-9 on colorectal cancer (CRC) cell proliferation, migration, and invasion and determined the underlying molecular mechanisms. Thirty-two paired CRC tissues and adjacent normal tissues were analyzed for ANO1 expression using quantitative real-time PCR (qRT-PCR). HCT116 cells were transiently transfected with miR-9 mimic, miR-9 inhibitor, or si-ANO1. Cell proliferation was determined by MTT, and flow cytometric analysis, while cell migration and invasion were assayed by trans-well migration and invasion assay in HCT116 cells. ANO1 was validated as a target of miR-9 using luciferase reporter assay and bioinformatics algorithms. We found that ANO1 expression was up-regulated in CRC tissues compared with adjacent normal tissues. ANO1 expression was associated with advanced tumor stage and lymph node metastasis, and there was an inverse relationship between miR-9 and ANO1 mRNA expression in CRC specimens, but no significant difference was found between miR-9 and ANO1 expression. ANO1 is a direct target of miR-9, and overexpression of miR-9 suppressed both mRNA and protein expression of ANO1 and inhibited cell proliferation, migration, and invasion of HCT116 cells. We also showed that overexpression of miR-9 suppressed expression of p-AKT, cyclin D1, and p-ERK in HCT116 cells. We conclude that miR-9 inhibits CRC cell proliferation, migration, and invasion by directly targeting ANO1, and miR-9/ANO1 could be a potential therapeutic target for CRC.
Collapse
Affiliation(s)
- Young Ran Park
- Department of Internal Medicine, Chonbuk National University Hospital, Chonbuk National University Medical School, 20 Geonji-ro, Deokjin-gu, Jeonju, Jeonbuk 54907, Republic of Korea; Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Republic of Korea
| | - Soo Teik Lee
- Department of Internal Medicine, Chonbuk National University Hospital, Chonbuk National University Medical School, 20 Geonji-ro, Deokjin-gu, Jeonju, Jeonbuk 54907, Republic of Korea; Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Republic of Korea
| | - Se Lim Kim
- Department of Internal Medicine, Chonbuk National University Hospital, Chonbuk National University Medical School, 20 Geonji-ro, Deokjin-gu, Jeonju, Jeonbuk 54907, Republic of Korea; Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Republic of Korea
| | - Shi Mao Zhu
- Department of Internal Medicine, Chonbuk National University Hospital, Chonbuk National University Medical School, 20 Geonji-ro, Deokjin-gu, Jeonju, Jeonbuk 54907, Republic of Korea; Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Republic of Korea
| | - Min Ro Lee
- Department of Surgery, Chonbuk National University Hospital, Chonbuk National University Medical School, Jeonju, Republic of Korea
| | - Seong Hun Kim
- Department of Internal Medicine, Chonbuk National University Hospital, Chonbuk National University Medical School, 20 Geonji-ro, Deokjin-gu, Jeonju, Jeonbuk 54907, Republic of Korea; Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Republic of Korea
| | - In Hee Kim
- Department of Internal Medicine, Chonbuk National University Hospital, Chonbuk National University Medical School, 20 Geonji-ro, Deokjin-gu, Jeonju, Jeonbuk 54907, Republic of Korea; Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Republic of Korea
| | - Seung Ok Lee
- Department of Internal Medicine, Chonbuk National University Hospital, Chonbuk National University Medical School, 20 Geonji-ro, Deokjin-gu, Jeonju, Jeonbuk 54907, Republic of Korea; Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Republic of Korea
| | - Seung Young Seo
- Department of Internal Medicine, Chonbuk National University Hospital, Chonbuk National University Medical School, 20 Geonji-ro, Deokjin-gu, Jeonju, Jeonbuk 54907, Republic of Korea; Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Republic of Korea
| | - Sang Wook Kim
- Department of Internal Medicine, Chonbuk National University Hospital, Chonbuk National University Medical School, 20 Geonji-ro, Deokjin-gu, Jeonju, Jeonbuk 54907, Republic of Korea; Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Republic of Korea.
| |
Collapse
|