1
|
Huang Y, Huang S, Li Q, Zhang H, Xiao W, Chen Y. miR-338-3p Targets SIRT6 to Inhibit Liver Cancer Malignancy and Paclitaxel Resistance. Drug Dev Res 2025; 86:e70089. [PMID: 40258128 DOI: 10.1002/ddr.70089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 02/14/2025] [Accepted: 04/08/2025] [Indexed: 04/23/2025]
Abstract
For patients with liver cancer, a widespread and lethal tumor on a global scale, chemotherapy and immunotherapy are often the top choices. Paclitaxel, a widely administered chemotherapy drug, faces the dual issues of poor tumor response rates and the rapid onset of chemoresistance. This study delves into the functions of SIRT6 and miR-338-3p in malignancy and paclitaxel resistance of liver cancer cells. Bioinformatics and qRT-PCR were engaged to predict and examine expression profiles of SIRT6 and miR-338-3p in liver cancer tissues and cell lines. A paclitaxel-resistant cell line (MHCC97-PTX) was established for dissecting cellular responses to drug treatment. CCK-8 and colony formation tests measured cell vitality and proliferation, respectively. Flow cytometry assessed apoptotic cell death, and the paclitaxel IC50 values were derived for each group. We utilized online tools to predict miR-338-3p as an upstream regulator of SIRT6, and a dual-luciferase reporter assay verified their direct interaction. SIRT6 is abundantly expressed in liver cancer tissues and cells. SIRT6 knockdown decreased cell vitality and proliferation while promoting apoptosis and paclitaxel sensitivity. miR-338-3p, an upstream regulator of SIRT6 in liver cancer cells, binds to SIRT6 and downregulates its expression, modulating cell malignancy and drug resistance. The duo of miR-338-3p and SIRT6 can drive the aggressiveness and chemoresistance of liver cancer, emerging as hopeful candidates for biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Yiyuan Huang
- Department of Hepatobiliary Surgery, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, China
| | - Sunhui Huang
- Department of Hepatobiliary Surgery, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, China
| | - Quan Li
- Department of Hepatobiliary Surgery, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, China
| | - Hongchang Zhang
- Department of Hepatobiliary Surgery, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, China
| | - Wei Xiao
- Department of Hepatobiliary Surgery, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, China
| | - Yunhui Chen
- Department of Hepatobiliary Surgery, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, China
| |
Collapse
|
2
|
Lapkina E, Zinchenko I, Kutcenko V, Bondar E, Kirichenko A, Yamskikh I, Palkina N, Ruksha T. MiR-204-5p overexpression abrogates Dacarbazine-induced senescence in melanoma cells in vivo. Noncoding RNA Res 2025; 10:130-139. [PMID: 39385998 PMCID: PMC11462174 DOI: 10.1016/j.ncrna.2024.09.009] [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: 04/20/2024] [Revised: 09/16/2024] [Accepted: 09/16/2024] [Indexed: 10/12/2024] Open
Abstract
Cancer cell drug resistance hinders significantly therapeutic modalities in oncology. Dacarbazine is chemotherapeutic agent traditionally used for melanoma treatment although it's effectiveness insufficient. In the present study we performed NGS-based transcriptomic profiling of B16 melanoma tumors after Dacarbazine treatment in vivo. Whole transcriptome sequencing revealed 34 differentially expressed genes most of them associated with drug resistance and apoptosis evading. In accordance to bionformatic analysis, 6 signaling cascades: "D-Amino acid metabolism", "NF-kappa B signaling pathway", "Phosphatidylinositol signaling system", "P53 signaling pathway", "IL-17 signaling pathway" and "Bile secretion" were enriched by differentially expressed genes. Next we provided a combined treatment by Dacarbazine and miR-204-5p mimic as miR-204-5p was considered previously implicated in cancer drug resistance. This approach lead to an increase of miR-204-5p expression in B16 melanoma cells in vivo that was accompanied by subsequent decrease in the expression of miR-204-5p target genes - BCL2 and SIRT1 in the primary tumors. MiR-204-5p overexpression with Dacarbazine application resulted in increased the weight, and volume of primary tumors and diminished the proportion of β-Galactosidase expression in melanoma B16-bearing mice. Taking together, our study revealed that although miR-204-5p showed antiproliferative capacities in vitro, it's mimic in combination with Dacarbazine is able to potentiate tumor growth triggering probably a switch from senescent to proliferative phenotype of malignant cells.
Collapse
Affiliation(s)
- Ekaterina Lapkina
- Department of Pathophysiology, Krasnoyarsk State Medical University, Krasnoyarsk, Russia
| | - Ivan Zinchenko
- Department of Pathophysiology, Krasnoyarsk State Medical University, Krasnoyarsk, Russia
| | - Viktoriya Kutcenko
- Department of Pathophysiology, Krasnoyarsk State Medical University, Krasnoyarsk, Russia
| | - Eugeniya Bondar
- Department of Genomics and Bioinformatics, Institute of Fundamental Biology and Biotechnology, Siberian Federal University, Krasnoyarsk, Russia
- Laboratory of Forest Genomics, Institute of Fundamental Biology and Biotechnology, Siberian Federal University, Krasnoyarsk, Russia
- Laboratory of Genomic Research and Biotechnology, Federal Research Center “Krasnoyarsk Science Center” Siberian Branch, Russian Academy of Science, Krasnoyarsk, Russia
| | - Andrey Kirichenko
- Department of Pathological Anatomy, Krasnoyarsk State Medical University, Krasnoyarsk, Russia
| | - Irina Yamskikh
- Department of Genomics and Bioinformatics, Institute of Fundamental Biology and Biotechnology, Siberian Federal University, Krasnoyarsk, Russia
- Laboratory of Forest Genomics, Institute of Fundamental Biology and Biotechnology, Siberian Federal University, Krasnoyarsk, Russia
- Laboratory of Genomic Research and Biotechnology, Federal Research Center “Krasnoyarsk Science Center” Siberian Branch, Russian Academy of Science, Krasnoyarsk, Russia
| | - Nadezhda Palkina
- Department of Pathophysiology, Krasnoyarsk State Medical University, Krasnoyarsk, Russia
| | - Tatiana Ruksha
- Department of Pathophysiology, Krasnoyarsk State Medical University, Krasnoyarsk, Russia
| |
Collapse
|
3
|
Poullennec KG, Jnoff E, Abendroth J, Bhuma N, Calmiano M, Calmus L, Cardenas A, Courade JP, Delatour C, Hall A, de Haro T, Delker SL, Demaude T, Gaikwad N, Ghavate D, Gholap AR, Kierkowicz M, Le Mestre R, Van Hijfte N, Verheijden S, Vernerova K, De Wever V, Waghmode N. Discovery of UCB9386: A Potent, Selective, and Brain-Penetrant Nuak1 Inhibitor Suitable for In Vivo Pharmacological Studies. J Med Chem 2024; 67:20879-20910. [PMID: 39588908 DOI: 10.1021/acs.jmedchem.4c01237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2024]
Abstract
Nuak1 (NUAK family SnF1-like kinase-1) is a serine-threonine kinase and a member of the AMPK family. Interest in Nuak1 has increased over the years due to the role it plays in several biological processes, from tumor cell invasion and proliferation to Tau stabilization. Nuak1 is expressed in many cancer cell lines and many reports describe this target as an oncogene, the inhibition of which is hypothesized to be valuable for treating various cancer types including glioma. We report here the discovery of Nuak1 inhibitors originating from HTS hit 9 with excellent selectivity and the subsequent medicinal chemistry optimization program, supported by structural information from the first crystal structures of a Nuak1 chimeric protein which provided insights into the binding modes of our compounds. These efforts yielded a nanomolar cell potent, highly selective and brain penetrant Nuak1 inhibitor UCB9386 (56) suitable for in vivo pharmacological studies for central nervous system (CNS) disorders.
Collapse
Affiliation(s)
| | - Eric Jnoff
- Chemin du Foriest, UCB, 1420 Braine l'Alleud, Belgium
| | - Jan Abendroth
- UCB Seattle, Bainbridge Island, Washington 98110, United States
| | - Naresh Bhuma
- Illumina Centre, 19 Granta Park, Great Abingdon CB21 6DF, United Kingdom
| | | | - Laurent Calmus
- NovAliX, 240 Parc d'Affaires des Portes, 27100 Val-de-Reuil, France
| | | | | | | | - Adrian Hall
- Chemin du Foriest, UCB, 1420 Braine l'Alleud, Belgium
| | | | | | | | - Nilesh Gaikwad
- Sai Life Sciences Ltd, IKP Knowledge Park, Genome Valley, Turkapally, Hyderabad 500078, India
| | - Dnyaneshwar Ghavate
- Sai Life Sciences Ltd, IKP Knowledge Park, Genome Valley, Turkapally, Hyderabad 500078, India
| | - Atul R Gholap
- Sai Life Sciences Ltd, IKP Knowledge Park, Genome Valley, Turkapally, Hyderabad 500078, India
| | | | - Régis Le Mestre
- Minoryx Therapeutics, Avenue Jean Mermoz 32, 6041 Charleroi, Belgium
| | | | - Simon Verheijden
- Janssen Research and Development, Antwerpseweg 15, 2340 Beerse, Belgium
| | | | | | | |
Collapse
|
4
|
Skalka GL, Whyte D, Lubawska D, Murphy DJ. NUAK: never underestimate a kinase. Essays Biochem 2024; 68:295-307. [PMID: 38939918 DOI: 10.1042/ebc20240005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 06/11/2024] [Accepted: 06/18/2024] [Indexed: 06/29/2024]
Abstract
NUAK1 and NUAK2 belong to a family of kinases related to the catalytic α-subunits of the AMP-activated protein kinase (AMPK) complexes. Despite canonical activation by the tumour suppressor kinase LKB1, both NUAKs exhibit a spectrum of activities that favour tumour development and progression. Here, we review similarities in structure and function of the NUAKs, their regulation at gene, transcript and protein level, and discuss their phosphorylation of specific downstream targets in the context of the signal transduction pathways and biological activities regulated by each or both NUAKs.
Collapse
Affiliation(s)
- George L Skalka
- School of Cancer Sciences, University of Glasgow, Glasgow, U.K
- CRUK Scotland Institute, Garscube Estate, Glasgow G61 1BD, U.K
| | - Declan Whyte
- CRUK Scotland Institute, Garscube Estate, Glasgow G61 1BD, U.K
| | | | - Daniel J Murphy
- School of Cancer Sciences, University of Glasgow, Glasgow, U.K
- CRUK Scotland Institute, Garscube Estate, Glasgow G61 1BD, U.K
| |
Collapse
|
5
|
Chen L, He Y, Lan J, Li Z, Gu D, Nie W, Zhang T, Ding Y. Advancements in nano drug delivery system for liver cancer therapy based on mitochondria-targeting. Biomed Pharmacother 2024; 180:117520. [PMID: 39395257 DOI: 10.1016/j.biopha.2024.117520] [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: 07/10/2024] [Revised: 09/26/2024] [Accepted: 10/04/2024] [Indexed: 10/14/2024] Open
Abstract
Based on poor efficacy and non-specific toxic side effects of conventional drug therapy for liver cancer, nano-based drug delivery system (NDDS) offers the advantage of drug targeting delivery. Subcellular targeting of nanomedicines on this basis enables more precise and effective termination of tumor cells. Mitochondria, as the crucial cell powerhouse, possesses distinctive physical and chemical properties in hepatoma cells different from that in hepatic cells, and controls apoptosis, tumor metastasis, and cellular drug resistance in hepatoma cells through metabolism and dynamics, which serves as a good choice for drug targeting delivery. Thus, mitochondria-targeting NDDS have become a recent research focus, showcasing the design of cationic nanoparticles, metal nanoparticles, mitochondrial peptide modification and so on. Although many studies have shown good results regarding anti-tumor efficacy, it is a long way to go before the successful translation of clinical application. Based on these, we summarized the specificity and importance of mitochondria in hepatoma cells, and reviewed the current mitochondria-targeting NDDS for liver cancer therapy, aiming to provide a better understanding for current development process, strengths and weaknesses of mitochondria-targeting NDDS as well as informing subsequent improvements and developments.
Collapse
Affiliation(s)
- Lixia Chen
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yitian He
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jinshuai Lan
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; The MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Zhe Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; The MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Donghao Gu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Wenlong Nie
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Tong Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; The MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Yue Ding
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; The MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
6
|
Li J, Sun X, Lv M, Han Z, Zhong X, Zhang W, Hu R, Feng W, Ma M, Huang Q, Zhou X. ncRNA-mediated SOX4 overexpression correlates with unfavorable outcomes and immune infiltration in hepatocellular carcinoma. BMC Gastroenterol 2024; 24:265. [PMID: 39143462 PMCID: PMC11323613 DOI: 10.1186/s12876-024-03346-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 08/01/2024] [Indexed: 08/16/2024] Open
Abstract
BACKGROUND The activity and number of immune cells in the tumor microenvironment are closely related to the overall survival of patients with hepatocellular carcinoma (HCC). The sex-determining region Y-box 4 (SOX4) gene is abnormally expressed in various tumor tissues and is critical for tumor development. However, the correlation between SOX4 expression in HCC and tumor immunity is unclear. METHODS SOX4 expression was explored using data from The Cancer Genome Atlas, and UALCAN databases. Real-time reverse transcription quantitative and western blotting were used to analyze SOX4 expression in several liver cancer cell lines. Additionally, correlations among SOX4 expression, cancer immune characteristics, and infiltrated immune cell gene marker sets in patients with HCC were analyzed using data from the Tumor Immune Estimation Resource, Gene Expression Profiling Interactive Analysis, and Tumor-Immune System Interactions databases. Moreover, we evaluated SOX4 expression in HCC tissues and the correlation of SOX4 expression with survival rate. Subsequently, noncoding RNAs (ncRNAs) responsible for SOX4 overexpression were identified using expression, correlation, and survival analyses. RESULTS SOX4 expression was significantly upregulated in HCC and correlated with a poor prognosis. Additionally, SOX4 upregulation in HCC positively correlated with immune cell infiltration, several biomarkers of immune cells, and immune checkpoint expression. Finally, the MCM3AP-AS1/hsa-miR-204-5p axis was identified as the most likely upstream ncRNA-related pathway for SOX4 in HCC. These results indicated that ncRNA-mediated upregulation of SOX4 correlated with the immune infiltration level and poor prognosis in HCC. Our findings provide new directions for the development of novel immunotherapeutic targets for HCC.
Collapse
Affiliation(s)
- Jing Li
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
- Department of Liver Disease, The fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, China
| | - Xinfeng Sun
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
- Department of Liver Disease, The fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Minling Lv
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
- Department of Liver Disease, The fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Zhiyi Han
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
- Department of Liver Disease, The fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Xin Zhong
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
- Department of Liver Disease, The fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Wei Zhang
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
- Department of Liver Disease, The fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Rui Hu
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
- Department of Liver Disease, The fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Wenxing Feng
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
- Department of Liver Disease, The fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Mengqing Ma
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
- Department of Liver Disease, The fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Qi Huang
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
- Department of Liver Disease, The fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Xiaozhou Zhou
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China.
- Department of Liver Disease, The fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China.
| |
Collapse
|
7
|
Cao L, Lin G, Fan D, Weng K, Chen Y, Wang J, Li P, Zheng C, Huang C, Xie J. NUAK1 activates STAT5/GLI1/SOX2 signaling to enhance cancer cell expansion and drives chemoresistance in gastric cancer. Cell Rep 2024; 43:114446. [PMID: 38996065 DOI: 10.1016/j.celrep.2024.114446] [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: 09/12/2023] [Revised: 06/02/2024] [Accepted: 06/20/2024] [Indexed: 07/14/2024] Open
Abstract
The gene encoding the NUAK family kinase 1 (NUAK1) is frequently amplified and its expression is upregulated, activating oncogenic signaling in various cancers. However, little is known about its role in gastric cancer (GC). We investigate the mechanistic links among NUAK1, Hedgehog signaling, and tumorigenesis in GC. NUAK1 overexpression is validated in local and public GC cohorts. Patient-derived xenograft and transgenic mouse models demonstrate that NUAK1 depletion or inhibition dramatically ameliorates gastric tumorigenesis. NUAK1 upregulates GLI1 expression by activating STAT5-mediated transcription and stabilizing GLI1 protein. NUAK1 depletion or inhibition impairs cancer cell expansion, tumor formation, and chemotherapy resistance in in vitro and in vivo models. Clinicopathological analysis confirms that upregulated NUAK1 expression correlates with poor prognosis and chemotherapy resistance in human GC. Our findings demonstrate that the signaling axis NUAK1/STAT5/GLI1 promotes cancer cell expansion and tumorigenesis and indicate that NUAK1 is an attractive therapeutic target and prognostic factor in GC.
Collapse
Affiliation(s)
- Longlong Cao
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, Fujian, China.
| | - Guangtan Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, Fujian, China
| | - Denghui Fan
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, Fujian, China
| | - Kai Weng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, Fujian, China
| | - Yujing Chen
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, Fujian, China
| | - Jiabin Wang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, Fujian, China
| | - Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, Fujian, China
| | - Chaohui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, Fujian, China
| | - Changming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, Fujian, China.
| | - Jianwei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, Fujian, China.
| |
Collapse
|
8
|
Hashemi M, Daneii P, Asadalizadeh M, Tabari K, Matinahmadi A, Bidoki SS, Motlagh YSM, Jafari AM, Ghorbani A, Dehghanpour A, Nabavi N, Tan SC, Rashidi M, Taheriazam A, Entezari M, Goharrizi MASB. Epigenetic regulation of hepatocellular carcinoma progression: MicroRNAs as therapeutic, diagnostic and prognostic factors. Int J Biochem Cell Biol 2024; 170:106566. [PMID: 38513802 DOI: 10.1016/j.biocel.2024.106566] [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: 06/30/2023] [Revised: 01/28/2024] [Accepted: 03/19/2024] [Indexed: 03/23/2024]
Abstract
Hepatocellular carcinoma (HCC), a significant challenge for public healthcare systems in developed Western countries including the USA, Canada, and the UK, is influenced by different risk factors including hepatitis virus infections, alcoholism, and smoking. The disruption in the balance of microRNAs (miRNAs) plays a vital function in tumorigenesis, given their function as regulators in numerous signaling networks. These miRNAs, which are mature and active in the cytoplasm, work by reducing the expression of target genes through their impact on mRNAs. MiRNAs are particularly significant in HCC as they regulate key aspects of the tumor, like proliferation and invasion. Additionally, during treatment phases such as chemotherapy and radiotherapy, the levels of miRNAs are key determinants. Pre-clinical experiments have demonstrated that altered miRNA expression contributes to HCC development, metastasis, drug resistance, and radio-resistance, highlighting related molecular pathways and processes like MMPs, EMT, apoptosis, and autophagy. Furthermore, the regulatory role of miRNAs in HCC extends beyond their immediate function, as they are also influenced by other epigenetic factors like lncRNAs and circular RNAs (circRNAs), as discussed in recent reviews. Applying these discoveries in predicting the prognosis of HCC could mark a significant advancement in the therapy of this disease.
Collapse
Affiliation(s)
- Mehrdad Hashemi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Pouria Daneii
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mahya Asadalizadeh
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Kiana Tabari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Arash Matinahmadi
- Department of Cellular and Molecular Biology, Nicolaus Copernicus University, Torun, Poland
| | - Seyed Shahabadin Bidoki
- Faculty of medicine, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | | | - Ali Moghadas Jafari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Amin Ghorbani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Amir Dehghanpour
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Noushin Nabavi
- Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, V6H3Z6, Vancouver, BC, Canada
| | - Shing Cheng Tan
- UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Mohsen Rashidi
- Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Maliheh Entezari
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | | |
Collapse
|
9
|
Bakhsh T, Alhazmi S, Farsi A, Yusuf AS, Alharthi A, Qahl SH, Alghamdi MA, Alzahrani FA, Elgaddar OH, Ibrahim MA, Bahieldin A. Molecular detection of exosomal miRNAs of blood serum for prognosis of colorectal cancer. Sci Rep 2024; 14:8902. [PMID: 38632250 PMCID: PMC11024162 DOI: 10.1038/s41598-024-58536-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Accepted: 04/01/2024] [Indexed: 04/19/2024] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer affecting people. The discovery of new, non-invasive, specific, and sensitive molecular biomarkers for CRC may assist in the diagnosis and support therapeutic decision making. Exosomal miRNAs have been demonstrated in carcinogenesis and CRC development, which makes these miRNAs strong biomarkers for CRC. Deep sequencing allows a robust high-throughput informatics investigation of the types and abundance of exosomal miRNAs. Thus, exosomal miRNAs can be efficiently examined as diagnostic biomarkers for disease screening. In the present study, a number of 660 mature miRNAs were detected in patients diagnosed with CRC at different stages. Of which, 29 miRNAs were differentially expressed in CRC patients compared with healthy controls. Twenty-nine miRNAs with high abundance levels were further selected for subsequent analysis. These miRNAs were either highly up-regulated (e.g., let-7a-5p, let-7c-5p, let-7f-5p, let-7d-3p, miR-423-5p, miR-3184-5p, and miR-584) or down-regulated (e.g., miR-30a-5p, miR-99-5p, miR-150-5p, miR-26-5p and miR-204-5p). These miRNAs influence critical genes in CRC, leading to either tumor growth or suppression. Most of the reported diagnostic exosomal miRNAs were shown to be circulating in blood serum. The latter is a novel miRNA that was found in exosomal profile of blood serum. Some of the predicted target genes of highly expressed miRNAs participate in several cancer pathways, including CRC pathway. These target genes include tumor suppressor genes, oncogenes and DNA repair genes. Main focus was given to multiple critical signaling cross-talking pathways including transforming growth factor β (TGFβ) signaling pathways that are directly linked to CRC. In conclusion, we recommend further analysis in order to experimentally confirm exact relationships between selected differentially expressed miRNAs and their predicted target genes and downstream functional consequences.
Collapse
Affiliation(s)
- Tahani Bakhsh
- Department of Biology, College of Science, University of Jeddah, Jeddah, Saudi Arabia.
| | - Safiah Alhazmi
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, 21589, Jeddah, Saudi Arabia
- Immunology Unit, King Fahad Medical Research Centre, King Abdulaziz University, 80200, Jedaah, Saudi Arabia
- Neuroscience and Geroscience Research Unit, King Fahad Medical Research Centre, King Abdulaziz University, 80200, Jeddah, Saudi Arabia
- Central lab of biological Sciences, Faculty of Sciences, King Abdulaziz University, 80200, Jeddah, Saudi Arabia
| | - Ali Farsi
- Department of Surgery, Faculty of Medicine, King Abdulaziz University, 21589, Jeddah, Saudi Arabia
| | - Abdulaziz S Yusuf
- Department of Biochemistry, Faculty of science, Stem Cell Unit, King Fahad Center for Medical Research, King Abdulaziz University, 21589, Jeddah, Saudi Arabia
- Medical Laboratory Sciences Department, Fakeeh College for Medical Sciences, 21461, Jeddah, Saudi Arabia
| | - Amani Alharthi
- Department of Biology, College of Science Al-Zulfi, Majmaah University, 11952, Majmaah, Saudi Arabia
| | - Safa H Qahl
- Department of Biology, College of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Maha Ali Alghamdi
- Department of Biotechnology, College of Science, Taif University, Taif, Saudi Arabia
| | - Faisal A Alzahrani
- Department of Biochemistry, Faculty of science, Stem Cell Unit, King Fahad Center for Medical Research, King Abdulaziz University, 21589, Jeddah, Saudi Arabia
| | - Ola H Elgaddar
- Department of Chemical Pathology, Alexandria University, Alexandria, Egypt
| | - Mohanad A Ibrahim
- Data Science Program, King Abdullah International Medical Research Center, 11481, Riyadh, Saudi Arabia
| | - Ahmed Bahieldin
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, 21589, Jeddah, Saudi Arabia.
| |
Collapse
|
10
|
Sabaghi F, Sadat SY, Mirsaeedi Z, Salahi A, Vazifehshenas S, Kesh NZ, Balavar M, Ghoraeian P. The Role of Long Noncoding RNAs in Progression of Leukemia: Based on Chromosomal Location. Microrna 2024; 13:14-32. [PMID: 38275047 DOI: 10.2174/0122115366265540231201065341] [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: 07/11/2023] [Revised: 08/29/2023] [Accepted: 10/12/2023] [Indexed: 01/27/2024]
Abstract
Long non-coding RNA [LncRNA] dysregulation has been seen in many human cancers, including several kinds of leukemia, which is still a fatal disease with a poor prognosis. LncRNAs have been demonstrated to function as tumor suppressors or oncogenes in leukemia. This study covers current research findings on the role of lncRNAs in the prognosis and diagnosis of leukemia. Based on recent results, several lncRNAs are emerging as biomarkers for the prognosis, diagnosis, and even treatment outcome prediction of leukemia and have been shown to play critical roles in controlling leukemia cell activities, such as proliferation, cell death, metastasis, and drug resistance. As a result, lncRNA profiles may have superior predictive and diagnostic potential in leukemia. Accordingly, this review concentrates on the significance of lncRNAs in leukemia progression based on their chromosomal position.
Collapse
Affiliation(s)
- Fatemeh Sabaghi
- Department of Molecular cell biology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Saina Yousefi Sadat
- Department of Microbiology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Zohreh Mirsaeedi
- Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Aref Salahi
- Department of Molecular cell biology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Sara Vazifehshenas
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Neda Zahmat Kesh
- Department of Genetics, Zanjan Branch Islamic Azad University, Zanjan, Iran
| | - Mahdieh Balavar
- Department of Genetics, Falavarjan Branch Islamic Azad University, Falavarjan, Iran
| | - Pegah Ghoraeian
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| |
Collapse
|
11
|
Yang F, Bian Z, Xu P, Sun S, Huang Z. MicroRNA-204-5p: A pivotal tumor suppressor. Cancer Med 2023; 12:3185-3200. [PMID: 35908280 PMCID: PMC9939231 DOI: 10.1002/cam4.5077] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 06/08/2022] [Accepted: 07/03/2022] [Indexed: 12/15/2022] Open
Abstract
MicroRNAs (miRNAs) are a class of non-coding single-stranded RNA molecules with a length of approximately 18-25 nt nucleotides that regulate gene expression post-transcriptionally. MiR-204-5p originates from the sixth intron of the transient receptor potential cation channel subfamily M member 3 (TRPM3) gene. MiR-204-5p is frequently downregulated in various cancer types and is related to the clinicopathological characteristics and prognosis of cancer patients. So far, many studies have determined that miR-204-5p functions as a tumor suppressor for its extensive and powerful capacity to inhibit tumor proliferation, metastasis, autophagy, and chemoresistance in multiple cancer types. MiR-204-5p appears to be a promising prognostic biomarker and a therapeutic target for human cancers. This review summarized the latest advances on the role of miR-204-5p in human cancers.
Collapse
Affiliation(s)
- Fan Yang
- Wuxi Cancer InstituteAffiliated Hospital of Jiangnan UniversityWuxiJiangsuChina
- Laboratory of Cancer Epigenetics, Wuxi School of MedicineJiangnan UniversityWuxiJiangsuChina
| | - Zehua Bian
- Wuxi Cancer InstituteAffiliated Hospital of Jiangnan UniversityWuxiJiangsuChina
- Laboratory of Cancer Epigenetics, Wuxi School of MedicineJiangnan UniversityWuxiJiangsuChina
| | - Peiwen Xu
- Wuxi Cancer InstituteAffiliated Hospital of Jiangnan UniversityWuxiJiangsuChina
- Laboratory of Cancer Epigenetics, Wuxi School of MedicineJiangnan UniversityWuxiJiangsuChina
| | - Shengbai Sun
- Wuxi Cancer InstituteAffiliated Hospital of Jiangnan UniversityWuxiJiangsuChina
- Laboratory of Cancer Epigenetics, Wuxi School of MedicineJiangnan UniversityWuxiJiangsuChina
| | - Zhaohui Huang
- Wuxi Cancer InstituteAffiliated Hospital of Jiangnan UniversityWuxiJiangsuChina
- Laboratory of Cancer Epigenetics, Wuxi School of MedicineJiangnan UniversityWuxiJiangsuChina
| |
Collapse
|
12
|
Wang K, Lin X. Circular RNA circMTO1 suppressed the progression of renal cell carcinoma progression by sponging miR-211 and miR-204. Mamm Genome 2022; 33:517-524. [PMID: 35092479 DOI: 10.1007/s00335-022-09944-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 01/11/2022] [Indexed: 12/01/2022]
Abstract
Despite considerable improvements in renal cell carcinoma (RCC) diagnostic and therapeutic strategy, the clinical prognosis of patients is far from satisfactory due to its recurrence and metastasis. Here, we attempted to explore the role of circMTO1 in RCC progression, and the underlying mechanism was further elucidated. We first detected the expression of circMTO1 in 90 pairs of RCC tissues and adjacent normal tissues using qRT-PCR. Besides, circMTO1, miR-211, miR-204 and KLF6 expression levels in RCC cells were also measured using qRT-PCR. MTT assay, cell migration, flow cytometry analysis, qRT-PCR and western blotting analysis were applied to evaluating the effect of circMTO1 in RCC cells. The bioinformatics analysis and the rescue experiment were devoted to the underlying mechanism. The results demonstrated CircMTO1 expression was significantly down-regulated in RCC tissues and cell lines. Besides, CircMTO1 inhibited cell proliferation, migration and invasion, induced apoptosis in RCC cells. In addition, CircMTO1 serves as a sponge for miR-211 and miR-204, KLF6 is a direct target of miR-211 and miR-204. Furthermore, circMTO1 and KLF6 overexpression rescued the suppression of miR-211/204 in RCC cell proliferation. In short, circMTO1 repressed RCC progression by regulating KLF6 via sponging miR-211 and miR-204, which may provide new idea of diagnosis and treatment in renal cell carcinoma.
Collapse
Affiliation(s)
- Ke Wang
- Department of Urology, The First People's Hospital of Xianyang, Xianyang, 712000, Shaanxi Province, China
| | - Xiaofeng Lin
- Department of Urology, People's Hospital of Wuqi County, Wuqi County Middle Street, Yan'an City, 717600, Shaanxi Province, China.
| |
Collapse
|
13
|
Seo MS, Jung KH, Kim K, Lee JE, Han BS, Ko S, Kim JH, Hong S, Lee SH, Hong SS. Discovery of a novel NUAK1 inhibitor against pancreatic cancer. Biomed Pharmacother 2022; 152:113241. [PMID: 35691157 DOI: 10.1016/j.biopha.2022.113241] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 05/31/2022] [Accepted: 06/02/2022] [Indexed: 12/24/2022] Open
Abstract
The novel (nua) kinase family 1 (NUAK1) is an AMPK-related kinase and its expression is associated with tumor malignancy and poor prognosis in several types of cancer, suggesting its potential as a target for cancer therapy. Therefore, the development of NUAK1-targeting inhibitors could improve therapeutic outcomes in cancer. We synthesized KI-301670, a novel NUAK1 inhibitor, and assessed its anticancer effects and mechanism of action in pancreatic cancer. It effectively inhibited pancreatic cancer growth and proliferation, and induced cell cycle arrest, markedly G0/G1 arrest, by increasing the expression of p27 and decreasing expression of p-Rb and E2F1. Additionally, the apoptotic effect of KI-301670 was observed by an increase in cleaved PARP, TUNEL-positive cells, and annexin V cell population, as well as the release of cytochrome c via the loss of mitochondrial membrane potential. KI-301670 inhibited the migration and invasion of pancreatic cancer cells. Mechanistically, KI-301670 effectively inhibited the PI3K/AKT pathway in pancreatic cancer cells. Furthermore, it significantly attenuated tumor growth in a mouse xenograft tumor model. Our results demonstrate that a novel NUAK1 inhibitor, KI-301670, exerts anti-tumor effects by directly suppressing cancer cell growth by affecting the PI3K/AKT pathway, suggesting that it could be a novel therapeutic candidate for pancreatic cancer treatment.
Collapse
Affiliation(s)
- Myeong-Seong Seo
- Department of Medicine, College of Medicine, and Program in Biomedical Science & Engineering, Inha University, 3-ga, Sinheung-dong, Jung-gu, Incheon 22332, South Korea
| | - Kyung Hee Jung
- Department of Medicine, College of Medicine, and Program in Biomedical Science & Engineering, Inha University, 3-ga, Sinheung-dong, Jung-gu, Incheon 22332, South Korea
| | - Kewon Kim
- Center for Catalytic Hydrocarbon Functionalization, Institute of Basic Science (IBS) and Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, South Korea
| | - Ji Eun Lee
- Department of Medicine, College of Medicine, and Program in Biomedical Science & Engineering, Inha University, 3-ga, Sinheung-dong, Jung-gu, Incheon 22332, South Korea
| | - Beom Seok Han
- Department of Medicine, College of Medicine, and Program in Biomedical Science & Engineering, Inha University, 3-ga, Sinheung-dong, Jung-gu, Incheon 22332, South Korea
| | - Soyeon Ko
- Department of Medicine, College of Medicine, and Program in Biomedical Science & Engineering, Inha University, 3-ga, Sinheung-dong, Jung-gu, Incheon 22332, South Korea
| | - Jae Ho Kim
- Chemical Kinomics Research Center, Institute of Science and Technology, Seoul 02792, South Korea
| | - Sungwoo Hong
- Center for Catalytic Hydrocarbon Functionalization, Institute of Basic Science (IBS) and Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, South Korea.
| | - So Ha Lee
- Chemical Kinomics Research Center, Institute of Science and Technology, Seoul 02792, South Korea.
| | - Soon-Sun Hong
- Department of Medicine, College of Medicine, and Program in Biomedical Science & Engineering, Inha University, 3-ga, Sinheung-dong, Jung-gu, Incheon 22332, South Korea.
| |
Collapse
|
14
|
miR-622 Counteracts the NUAK1-Induced Gastric Cancer Cell Proliferation and the Antioxidative Stress. DISEASE MARKERS 2022; 2022:9616764. [PMID: 35872695 PMCID: PMC9303142 DOI: 10.1155/2022/9616764] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/25/2022] [Accepted: 06/28/2022] [Indexed: 01/10/2023]
Abstract
Background Gastric cancer (GC), a highly prevalent gastric cancer, has high-risk mortality. Thus, investigating strategies to counteract its growth is important to provide theoretical guidance for its prevention and treatment. It has been pointed out that abnormal expression of microRNAs (miRNAs) serves as noninvasive biomarkers for GC. This present study probed into the role of miR-622 and the NUAK family SNF1-like kinase 1 (NUAK1). Methods Five mRNA datasets (GSE64916, GSE118916, GSE122401, GSE158662, and GSE159721) and one miRNA dataset (GSE128720) from the Gene Expression of Omnibus (GEO) database were used to analyze the differentially expressed miRNAs and mRNA in GC and noncancer samples. Further, western blot, real-time quantitative PCR (qRT-PCR), reactive oxygen species (ROS) assay kit experiments, and wound healing assay, together with in vivo experiments, were performed. Results miR-622 was downregulated, and NUAK1 was upregulated in GC, and NUAK1 was a potential target of miR-622. Knocking down NUAK1 decreased GC cell proliferation and migration but increased oxidative stress in vitro and inhibited the development of tumor in vivo, while miR-622 acted to suppress the action of NUAK1 through the miR-622/NUAK1/p-protein kinase B (Akt) axis, thereby inhibiting the occurrence of GC. Conclusion miR-622 and NUAK1 demonstrated potential for being targets and biomarkers for GC treatment.
Collapse
|
15
|
Effect and Role of miR-196b in Ectopic Pregnancy. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:7797484. [PMID: 35265305 PMCID: PMC8901340 DOI: 10.1155/2022/7797484] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 01/25/2022] [Accepted: 02/02/2022] [Indexed: 11/30/2022]
Abstract
Ectopic pregnancy (EP) is associated with significant morbidity and mortality, but the molecular mechanism of this condition is still unclear. miR-196b, a hot research direction for the past few years, participates in the occurrence of various diseases but whether it plays a regulatory role in EP is still unclear. This research was set to investigate the expression and potential value of miR-196b in EP. qRT-PCR was utilized to determine the relative expression of miR-196b in peripheral blood of EP patients and to observe the expression changes of miR-196b before and after treatment. Correlation analysis of miR-196b with HCG and progesterone was performed. Logistic regression analysis was applied to independent risk factors affecting EP patients. TargetScan was utilized to predict the downstream target genes of miR-196b, and GO and KEGG analysis was carried out using the R language pack. qRT-PCR showed that miR-196b expression in peripheral blood of EP patients was lower than that of normal people. miR-196b expression in patients after treatment was notably higher than that before treatment. In addition, correlation analysis showed that miR-196b was positively correlated with the expression of HCG, progesterone, and estradiol. Risk factor analysis revealed that abortion history, pelvic inflammatory disease history, lower abdominal surgery history, and miR-196b were independent risk factors for EP, and the AUC of the combined ROC curve was 0.899. GO function enrichment and KEGG signal pathway enrichment found 10 potential functions and 2 potential signal pathways of miR-196b. miR-196b is expressed in EP patients, is differentially expressed according to the change in EP condition, and is expected to become a promising index for clinical diagnosis of EP.
Collapse
|
16
|
Zhang Y, Wang ZY, Jing HS, Zhang HD, Yan HX, Fan JX, Zhai B. A pre‑clinical model combining cryopreservation technique with precision‑cut slice culture method to assess the in vitro drug response of hepatocellular carcinoma. Int J Mol Med 2022; 49:51. [PMID: 35179217 PMCID: PMC8904079 DOI: 10.3892/ijmm.2022.5107] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 01/28/2022] [Indexed: 11/22/2022] Open
Abstract
Models considering hepatocellular carcinoma (HCC) complexity cannot be accurately replicated in routine cell lines or animal models. We aimed to evaluate the practicality of tissue slice culture by combining it with a cryopreservation technique. We prepared 0.3-mm-thick tissue slices by a microtome and maintained their cell viability using a cryopreservation technique. Slices were cultured individually in the presence or absence of regorafenib (REG) for 72 h. Alterations in morphology and gene expression were assessed by histological and genetic analysis. Overall viability was also analyzed in tissue slices by CCK-8 quantification assay and fluorescent staining. Tissue morphology and cell viability were evaluated to quantify drug effects. Histological and genetic analyses showed that no significant alterations in morphology and gene expression were induced by the vitrification-based cryopreservation method. The viability of warmed HCC tissues was up to 90% of the fresh tissues. The viability and proliferation could be retained for at least four days in the filter culture system. The positive drug responses in precision-cut slice culture in vitro were evaluated by tissue morphology and cell viability. In summary, the successful application of precision-cut HCC slice culture combined with a cryopreservation technique in a systematic drug screening demonstrates the feasibility and utility of slice culture method for assessing drug response.
Collapse
Affiliation(s)
- Yuan Zhang
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Zhen-Yu Wang
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Hong-Shu Jing
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Hong-Dan Zhang
- Shanghai Celliver Biotechnology Co. Ltd., Shanghai 200120, P.R. China
| | - He-Xin Yan
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Jian-Xia Fan
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Bo Zhai
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| |
Collapse
|
17
|
Wang C, Tan R, Peng L, Zhang J. Relationship between miR-204 and ANGPTL2 expression and diagnosis, pathological stage, and prognosis in patients with colon cancer. Transl Cancer Res 2022; 10:3788-3796. [PMID: 35116678 PMCID: PMC8798759 DOI: 10.21037/tcr-21-1385] [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: 06/04/2021] [Accepted: 08/12/2021] [Indexed: 12/19/2022]
Abstract
Background Angiopoietin-like protein 2 (ANGPTL2) is linked to various tumors. MicroRNA-204 (miR-204) is associated with colorectal cancer (CRC). Bioinformatic analysis has demonstrated a targeting relationship between miR-204 and ANGPTL2. The present study aimed to investigate the role of miR-204 in the proliferation and apoptosis of colorectal tumor cells. Methods Colorectal tumor tissues were collected. Normal colon mucosa was used as a control. The relationship between miR-204 and ANGPTL2 expression and tumor stage and prognosis was analyzed. The dual-luciferase reporter assay confirmed targeted regulation between miR-204 and ANGPTL2. SW480 cells were allocated to the miR-NC group and the miR-204 mimic group, followed by apoptotic analysis using flow cytometry and cellular proliferation analysis using EdU staining. Results Compared with normal colonic mucosa, miR-204 expression was decreased in colorectal tumor tissues and ANGPTL2 expression was increased, which correlated with TNM staging. The prognosis of patients with low miR-204 expression and high ANGPTL2 expression was worse than for patients with high miR-204 expression and low ANGPTL2 expression. The dual-luciferase reporter assay confirmed a targeting regulation relationship between miR-204 and ANGPTL2. Transfection of miR-204 mimic significantly inhibited the expression of ANGPTL2 and cell proliferation in SW480 cells and promoted apoptosis. Conclusions Downregulating miR-204 expression plays a vital role in upregulating ANGPTL2 expression and promoting the pathogenesis of CRC. MiR-204 is able to hinder the proliferation of colorectal tumor cells and encourage apoptosis by targeting the inhibition of ANGPTL2 expression.
Collapse
Affiliation(s)
- Chenliang Wang
- Department of Pathology, First People's Hospital of Jiujiang City, Jiujiang, China
| | - Rongfei Tan
- Laboratory Department, Ruichang Maternal and Child Health Hospital, Jiujiang, China
| | - Lizi Peng
- Department of Pathology, First People's Hospital of Jiujiang City, Jiujiang, China
| | - Jing Zhang
- Department of Pathology, First People's Hospital of Jiujiang City, Jiujiang, China
| |
Collapse
|
18
|
Dashti F, Mirazimi SMA, Rabiei N, Fathazam R, Rabiei N, Piroozmand H, Vosough M, Rahimian N, Hamblin MR, Mirzaei H. The role of non-coding RNAs in chemotherapy for gastrointestinal cancers. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 26:892-926. [PMID: 34760336 PMCID: PMC8551789 DOI: 10.1016/j.omtn.2021.10.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Gastrointestinal (GI) cancers, including colorectal, gastric, hepatic, esophageal, and pancreatic tumors, are responsible for large numbers of deaths around the world. Chemotherapy is the most common approach used to treat advanced GI cancer. However, chemoresistance has emerged as a critical challenge that prevents successful tumor elimination, leading to metastasis and recurrence. Chemoresistance mechanisms are complex, and many factors and pathways are involved. Among these factors, non-coding RNAs (ncRNAs) are critical regulators of GI tumor development and subsequently can induce resistance to chemotherapy. This occurs because ncRNAs can target multiple signaling pathways, affect downstream genes, and modulate proliferation, apoptosis, tumor cell migration, and autophagy. ncRNAs can also induce cancer stem cell features and affect the epithelial-mesenchymal transition. Thus, ncRNAs could possibly act as new targets in chemotherapy combinations to treat GI cancer and to predict treatment response.
Collapse
Affiliation(s)
- Fatemeh Dashti
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Seyed Mohammad Ali Mirazimi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Nikta Rabiei
- School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Fathazam
- School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negin Rabiei
- School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Haleh Piroozmand
- Faculty of Veterinary Sciences, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Neda Rahimian
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Michael R. Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa
- Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
19
|
Revealing the role of miRNA-489 as a new onco-suppressor factor in different cancers based on pre-clinical and clinical evidence. Int J Biol Macromol 2021; 191:727-737. [PMID: 34562537 DOI: 10.1016/j.ijbiomac.2021.09.089] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 09/13/2021] [Accepted: 09/14/2021] [Indexed: 01/17/2023]
Abstract
Recently, microRNAs (miRNAs) have shown to be potential therapeutic, diagnostic and prognostic targets in disease therapy. These endogenous non-coding RNAs contribute to regulation of different cellular events that are necessary for maintaining physiological condition. Dysregulation of miRNAs is correlated with development of various pathological events such as neurological disorders, cardiovascular diseases, and cancer. miRNA-489 is a new emerging miRNA and studies are extensively investigating its role in pathological conditions. Herein, potential function of miRNA-489 as tumor-suppressor in various cancers is described. miRNA-489 is able to sensitize cancer cells into chemotherapy by disrupting molecular pathways involved in cancer growth such as PI3K/Akt, and induction of apoptosis. The PROX1 and SUZ12 as oncogenic pathways, are affected by miRNA-489 in suppressing metastasis of cancer cells. Wnt/β-catenin as an oncogenic factor ensuring growth and malignancy of tumors is inhibited via miRNA-489 function. For enhancing drug sensitivity of tumors, restoring miRNA-489 expression is a promising strategy. The lncRNAs can modulate miRNA-489 expression in tumors and studies about circRNA role in miRNA-489 modulation should be performed. The expression level of miRNA-489 is a diagnostic tool for tumor detection. Besides, down-regulation of miRNA-489 in tumors provides unfavorable prognosis.
Collapse
|
20
|
Qiu K, Song Y, Rao Y, Liu Q, Cheng D, Pang W, Ren J, Zhao Y. Diagnostic and Prognostic Value of MicroRNAs in Metastasis and Recurrence of Head and Neck Squamous Cell Carcinoma: A Systematic Review and Meta-Analysis. Front Oncol 2021; 11:711171. [PMID: 34646767 PMCID: PMC8503605 DOI: 10.3389/fonc.2021.711171] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 09/01/2021] [Indexed: 02/05/2023] Open
Abstract
MicroRNAs have been proven to make remarkable differences in the clinical behaviors of head and neck squamous cell carcinoma (HNSCC). This study aims to systematically analyze whether differential expression levels of microRNAs are related to recurrence or metastasis in patients with HNSCC. A comprehensive search of the PubMed, EMBASE, and CENTRAL was conducted up to July 24th, 2021. Data were collected and combined from studies reporting recurrence-free survival (RFS) of HNSCC patients with high microRNA expression compared to those with low expression. Besides, studies providing necessary data for evaluating the diagnostic value of microRNAs for detecting recurrence and metastasis based on their expression levels were also included and combined. The pooled hazard ratio (HR) value for the outcomes of RFS in 1,093 HNSCC samples from 10 studies was 2.51 (95%CI: 2.13–2.96). A sensitivity of 0.79 (95% CI: 0.72–0.85) and specificity of 0.77 (95%CI: 0.68–0.83) were observed in three studies, of which 93 patients with recurrence and 82 nonrecurrence controls were included, and the area under the curve (AUC) was 0.85 (95% CI: 0.81–0.88). Additionally, high diagnostic accuracy of microRNAs in detecting lymph node metastasis (LNM) was also reported. In conclusion, two panels of microRNAs showed the potential to predict recurrence or diagnose recurrence in HNSCC patients, respectively, which could facilitate prognosis prediction and diagnosis of clinical behaviors in HNSCC patients.
Collapse
Affiliation(s)
- Ke Qiu
- Department of Oto-Rhino-Laryngology, West China Hospital, Sichuan University, Chengdu, China
| | - Yao Song
- Department of Oto-Rhino-Laryngology, West China Hospital, Sichuan University, Chengdu, China
| | - Yufang Rao
- Department of Oto-Rhino-Laryngology, West China Hospital, Sichuan University, Chengdu, China
| | - Qiurui Liu
- Department of Oto-Rhino-Laryngology, West China Hospital, Sichuan University, Chengdu, China
| | - Danni Cheng
- Department of Oto-Rhino-Laryngology, West China Hospital, Sichuan University, Chengdu, China
| | - Wendu Pang
- Department of Oto-Rhino-Laryngology, West China Hospital, Sichuan University, Chengdu, China
| | - Jianjun Ren
- Department of Oto-Rhino-Laryngology, West China Hospital, Sichuan University, Chengdu, China.,West China Biomedical Big Data Center, West China Hospital/West China School of Medicine, Sichuan University, Chengdu, China.,Medical Big Data Center, Sichuan University, Chengdu, China
| | - Yu Zhao
- Department of Oto-Rhino-Laryngology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
21
|
Chen S, Wu H, Zhu L, Jiang M, Wei S, Luo J, Liu A. MiR-199b-5p Promotes Gastric Cancer Progression by Regulating HHIP Expression. Front Oncol 2021; 11:728393. [PMID: 34532291 PMCID: PMC8438221 DOI: 10.3389/fonc.2021.728393] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 08/03/2021] [Indexed: 12/29/2022] Open
Abstract
Objectives Gastric cancer (GC) is one of the most common malignant tumors. More and more evidences support the role of microRNAs (miRNAs) in tumor progression. However, the role of miRNAs in human GC remains largely unknown. Methods Based on the published gastric cancer expression profile data, combined with bioinformatics analysis, potential miRNAs in the process of GC were screened. The expression of miR-199b-5p in GC cells and patients’ plasma was detected by RT-PCR. The effects of miR-199b-5p on GC in vitro were detected by EdU proliferation assay, colony formation assay, Transwell assay and wound healing assay. Western blot was used to detect epithelial-mesenchymal transition (EMT) related proteins. The subcutaneous tumorigenesis model and metastatic tumor model of mice were used to study its effect in vivo. Bioinformatics and Dual luciferase reporter assay were used to verify the effect of miR-199b-5p and its target gene. Results Through bioinformatics analysis, we screened a novel miRNA miR-199b-5p that was significantly up-regulated in GC tissue and associated with poor prognosis of GC patients. RT-PCR results showed that its expression was also up-regulated in GC cell lines and patients’ plasma. MiR-199b-5p can significantly promote GC cell proliferation and migration in vitro and in vivo. Western blot showed that miR-199b-5p could promote the EMT process of GC. HHIP has been proved to be a target of miR-199b-5p, and the recovery of HHIP can weaken the effect of miR-199b-5p. Conclusion MiR-199b-5p may play an oncogene role in GC by targeting HHIP, suggesting that miR-199b-5p may be a potential therapeutic target for GC.
Collapse
Affiliation(s)
- Songda Chen
- Department of Endoscopy, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Huijie Wu
- Department of Endoscopy, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Lingyu Zhu
- Department of Endoscopy, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Mengjie Jiang
- Department of Endoscopy, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Shuli Wei
- Department of Gastroenterology, The 10th Affiliated Hospital of Guangxi Medical University, Qinzhou, China
| | - Jinhua Luo
- Department of Gastroenterology, The 10th Affiliated Hospital of Guangxi Medical University, Qinzhou, China
| | - Aiqun Liu
- Department of Endoscopy, Guangxi Medical University Cancer Hospital, Nanning, China
| |
Collapse
|
22
|
Identification of prognostic biomarkers related to the tumor microenvironment in thyroid carcinoma. Sci Rep 2021; 11:16239. [PMID: 34376710 PMCID: PMC8355328 DOI: 10.1038/s41598-021-90538-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 05/10/2021] [Indexed: 12/13/2022] Open
Abstract
Thyroid Carcinoma (THCA) is the most common endocrine tumor that is mainly treated using surgery and radiotherapy. In addition, immunotherapy is a recently developed treatment option that has played an essential role in the management of several types of tumors. However, few reports exist on the use of immunotherapy to treat THCA. The study downloaded the miRNA, mRNA and lncRNA data for THCA patients from the TCGA database ( https://portal.gdc.cancer.gov/ ). Thereafter, the tumor samples were divided into cold and hot tumors, based on the immune score of the tumor microenvironment. Moreover, the differentially expressed lncRNAs and miRNAs were obtained. Finally, the study jointly constructed a ceRNA network through differential analysis of the mRNA data for cold and hot tumors. The study first assessed the level of immune infiltration in the THCA tumor microenvironment then divided the samples into cold and hot tumors, based on the immune score. Additionally, a total of 568 up-regulated and 412 down-regulated DEGs were screened by analyzing the differences between hot and cold tumors. Thereafter, the study examined the differentially expressed genes for lncRNA and miRNA. The results revealed 629 differentially expressed genes related to lncRNA and 114 associated with miRNA. Finally, a ceRNA network of the differentially expressed genes was constructed. The results showed a five-miRNA hubnet, i.e., hsa-mir-204, hsa-mir-128, hsa-mir-214, hsa-mir-150 and hsa-mir-338. The present study identified the immune-related mRNA, lncRNA and miRNA in THCA then constructed a ceRNA network. These results are therefore important as they provide more insights on the immune mechanisms in THCA. The findings also provides additional information for possible THCA immunotherapy.
Collapse
|
23
|
Zhang H, Wu X, Sui Z, Ma Z, Gong L, Meng B, Tang P, Yu Z. High-mobility group AT-hook 2 promotes growth and metastasis and is regulated by miR-204-5p in oesophageal squamous cell carcinoma. Eur J Clin Invest 2021; 51:e13563. [PMID: 33901298 DOI: 10.1111/eci.13563] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 03/22/2021] [Accepted: 03/25/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND To investigate the expression of high-mobility group AT-hook 2 (HMGA2) and miR-204-5p in oesophageal squamous cell carcinoma (ESCC) and their biological roles in ESCC development and progression. METHODS HMGA2 and miR-204-5p expression levels in ESCC tissues and cell lines were detected by qRT-PCR, Western blotting and immunohistochemical staining. ESCC cell lines were transfected with a small interfering RNA for HMGA2 and miR-204-5p mimic to downregulate and upregulate the expression levels of HMGA2 and miR-204-5p, respectively. The growth, migration and invasion abilities of ESCC cells were assessed by MTT, colony formation, wound-healing and Transwell assays, respectively. A luciferase reporter gene assay was used to determine whether the 3'-untranslated coding regions of HMGA2 could be directly bound by miR-204-5p. RESULTS HMGA2 expression was markedly upregulated (P < .001), while miR-204-5p expression was markedly downregulated (P = .003) in ESCC tissues compared with adjacent normal tissues. HMGA2 expression was correlated with tumour size, invasion depth, lymph node metastasis and tumour-node-metastasis stage (all P < .05) and was identified as an independent prognostic factor for ESCC patients. The expression levels of HMGA2 and miR-204-5p were negatively correlated (r2 = 0.609, P < .001). HMGA2 knockdown or miR-204-5p overexpression markedly inhibited ESCC cell growth, migration and invasion (P < .05). In addition, restoration of HMGA2 expression partly reversed the inhibitory effects of miR-204-5p overexpression on migration and invasion (P < .05). The luciferase reporter gene assay suggested that HMGA2 is a direct downstream target of miR-204-5p. CONCLUSION HMGA2 functions as an oncogene in the growth and metastasis of ESCC and is negatively regulated by miR-204-5p.
Collapse
Affiliation(s)
- Hongdian Zhang
- Department of Esophageal Cancer, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center of Cancer, Tianjin, China
| | - Xianxian Wu
- Department of Esophageal Cancer, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center of Cancer, Tianjin, China
| | - Zhilin Sui
- Department of Esophageal Cancer, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center of Cancer, Tianjin, China
| | - Zhao Ma
- Department of Esophageal Cancer, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center of Cancer, Tianjin, China
| | - Lei Gong
- Department of Esophageal Cancer, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center of Cancer, Tianjin, China
| | - Bin Meng
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Peng Tang
- Department of Esophageal Cancer, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center of Cancer, Tianjin, China
| | - Zhentao Yu
- Department of Esophageal Cancer, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center of Cancer, Tianjin, China.,Department of Thoracic Surgery, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| |
Collapse
|
24
|
Xu B, Qian Y, Hu C, Wang Y, Gao H, Yang J. Dexmedetomidine upregulates the expression of miR-493-5p, inhibiting growth and inducing the apoptosis of lung adenocarcinoma cells by targeting RASL11B. Biochem Cell Biol 2021; 99:457-464. [PMID: 34296954 DOI: 10.1139/bcb-2020-0267] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Numerous studies have indicated that microRNAs (miRNAs) play critical roles in the development and progression of cancer. However, how changes to the expression levels of miRNAs in response to dexmedetomidine affects the progression of lung cancer remains poorly understood. In this study, we treated the lung adenocarcinoma cell line-A549 with dexmedetomidine and then examined the changes to the expression levels of miRNAs. We found that one of the most significantly upregulated miRNAs was miR-493-5p, which has an important role in the growth and apoptosis of lung adenocarcinoma (LUAD) cells. In addition, bioinformatics searches and luciferase reporter assays revealed that miR-493-5p targets RASL11B, which has a high degree of similarity to RAS. Finally, database searches revealed that RASL11B is associated with survival of LUAD cells. In conclusion, dexmedetomidine causes changes to the expression levels of miRNAs in LUAD, including significant upregulation of miR-493-5p. MiR-493-5p targets RASL11B, thereby inhibiting cell growth and inducing apoptosis in LUAD.
Collapse
Affiliation(s)
- Bo Xu
- Department of Anesthesiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, China.,Department of Anesthesiology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yiling Qian
- Department of Anesthesiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, China
| | - Chunxiao Hu
- Department of Anesthesiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, China
| | - Yongsheng Wang
- Department of Respiratory Medicine, Drum Tower Hospital Affiliated to Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Hong Gao
- Department of Anesthesiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, China
| | - Jianping Yang
- Department of Anesthesiology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
25
|
Wang H, Wang H, Cui W, Zhang Q, Li J, Zhang Q. Enhanced expression of miR-889 forecasts an unfavorable prognosis and facilitates cell progression in hepatocellular carcinoma. Diagn Pathol 2021; 16:51. [PMID: 34116691 PMCID: PMC8196534 DOI: 10.1186/s13000-021-01111-5] [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: 09/10/2020] [Accepted: 05/27/2021] [Indexed: 02/06/2023] Open
Abstract
Background As a new type of molecular marker, microRNAs (miRNAs) can be used for early diagnosis and prognosis prediction of malignant tumors, and has broad clinical application prospects. This paper mainly studies the important role of miR-889 in the occurrence and development of hepatocellular carcinoma and the prognostic significance of miR-889 in hepatocellular carcinoma. Methods Quantitative real-time PCR analysis detected the expression levels of miR-889 in hepatocellular carcinoma tissues and cell lines. Kaplan-Meier curve and Cox regression analysis were used to explore the prognostic significance of miR-889 in hepatocellular carcinoma. The CCK-8 and Transwell assays assay were used to assess cell proliferation, migration, and invasion abilities ability. Results The expression of miR-889 in hepatocellular carcinoma tissues was significantly higher than that in adjacent tissues. Overexpression of miR-889 was significantly associated with TNM stage, hepatitis B virus infection, and cirrhosis. Patients with high miR-889 expression had shorter overall survival than those with low miR-889 expression. And functional studies in two hepatocellular carcinoma cell lines have shown that overexpression of miR-889 significantly promoted cell proliferation, migration, and invasion in vitro. Conclusions Overall, miR-889 was upregulated in hepatocellular carcinoma tissues and cell lines, and overexpression of miR-889 promoted cell proliferation, migration, and invasion in hepatocellular carcinoma cells. Based on our findings, high expression of miR-889 may promote the progression of hepatocellular carcinoma, and high expression of miR-889 is also forecasted for an unfavorable prognosis in hepatocellular carcinoma.
Collapse
Affiliation(s)
- He Wang
- Department of Interventional, Harbin Medical University Cancer Hospital, No.150 Haping Road, Harbin, Heilongjiang, 150081, Harbin, China
| | - Huiwen Wang
- Department of Interventional, Harbin Medical University Cancer Hospital, No.150 Haping Road, Harbin, Heilongjiang, 150081, Harbin, China
| | - Wenyu Cui
- School of Pharmacy, Harbin University of Commerce, 138 Tongda Street, Harbin, 150076, Heilongjiang, China
| | - Qiao Zhang
- School of Pharmacy, Harbin University of Commerce, 138 Tongda Street, Harbin, 150076, Heilongjiang, China.
| | - Jing Li
- School of Pharmacy, Harbin University of Commerce, 138 Tongda Street, Harbin, 150076, Heilongjiang, China
| | - Qi Zhang
- School of Pharmacy, Harbin University of Commerce, 138 Tongda Street, Harbin, 150076, Heilongjiang, China
| |
Collapse
|
26
|
Mo G, Zhang B, Jiang Q. Role of ARK5 in cancer and other diseases (Review). Exp Ther Med 2021; 22:697. [PMID: 33986861 PMCID: PMC8112134 DOI: 10.3892/etm.2021.10129] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Accepted: 06/20/2020] [Indexed: 12/14/2022] Open
Abstract
Malignant tumors are often exposed to hypoxic and glucose-starved microenvironments. AMP-activated protein kinase (AMPK) is an energy sensor that is stimulated during energy-deficient conditions and protects cells from hypoxic injury by regulating metabolism. AMPK-related protein kinase 5 (ARK5) is a member of the catalytic sub-unit of the AMPK family and has an important role in energy regulation and hypoxia. ARK5 is regulated by Akt and liver kinase B1 and is associated with numerous tumor-related molecules to exert the negative effects of tumors. Studies have revealed ARK5 overexpression in cases of tumor invasion and metastasis and a positive association with the degree of cancer cell malignancy, which is regarded as a key element in determining cancer prognosis. Furthermore, ARK5 downregulation improves drug sensitivity through the epithelial-mesenchymal transition pathway, indicating that it may be a potential therapeutic target. In other non-cancer conditions, ARK5 has various roles in neurodegenerative diseases (Alzheimer's and Huntington's disease), renal disorders (diabetic nephropathy and renal fibrosis) and physiological processes (striated muscle generation). In the present review, the upstream and downstream molecular pathways of ARK5 in cancer and other diseases are described and potential therapeutic strategies are discussed.
Collapse
Affiliation(s)
- Guoheng Mo
- Department of Neurosurgery, Queen Mary College of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Bohan Zhang
- First Clinical Medical College, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Qunguang Jiang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
27
|
Chen X, Xu Y, Jiang L, Tan Q. miRNA-218-5p increases cell sensitivity by inhibiting PRKDC activity in radiation-resistant lung carcinoma cells. Thorac Cancer 2021; 12:1549-1557. [PMID: 33759399 PMCID: PMC8107034 DOI: 10.1111/1759-7714.13939] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 03/01/2021] [Accepted: 03/02/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Non-small cell lung carcinoma (NSCLC) is a malignancy with the highest mortality rate. Currently, surgery combined with radiotherapy is the first choice in the clinical treatment of lung carcinoma (LC); however, long-term radiotherapy leads to radiation resistance in patients, resulting in treatment failure. METHODS In this study, a new microRNA-218-5p (miRNA-218-5p) was identified, and its function in LC was investigated. RESULTS Reverse transcription quantitative polymerase chain reaction (RT-qPCR) results revealed that miRNA-218-5p was downregulated in LC. Overexpression or inhibition of miRNA-218-5p in LC and targeted binding of protein kinase, DNA-activated, catalytic polypeptide (PRKDC) to miRNA-218-5p were confirmed by comprehensive bioinformatic analysis. Exosomes from A549 and H1299 cells were cocultured with miRNA-218-5p and then cotransfected into radiation-resistant A549R and H1299R cells; the proliferation of radiation-resistant LC cells was found to be effectively inhibited and apoptosis was induced. Overexpression of miRNA-218-5p and X-irradiation could enhance the radiosensitivity of LC cells. Exogenous miRNA-218-5p derived from A549 and H1299 cells could be transfected into radiation-resistant LC cells and could inhibit PRKDC expression, thus accelerating DNA damage, apoptosis, and radiation sensitization of LC cells. CONCLUSIONS miRNA-218-5p could induce apoptosis and enhance the radiosensitivity of LC cells through regulatory activities, thus suggesting its application as a potential target for LC treatment.
Collapse
Affiliation(s)
- Xiaoke Chen
- Shanghai Lung Cancer CenterShanghai Chest Hospital, Shanghai Jiao Tong UniversityShanghaiChina
| | - Yuanyuan Xu
- Shanghai Lung Cancer CenterShanghai Chest Hospital, Shanghai Jiao Tong UniversityShanghaiChina
| | - Long Jiang
- Shanghai Lung Cancer CenterShanghai Chest Hospital, Shanghai Jiao Tong UniversityShanghaiChina
| | - Qiang Tan
- Shanghai Lung Cancer CenterShanghai Chest Hospital, Shanghai Jiao Tong UniversityShanghaiChina
| |
Collapse
|
28
|
Ghafouri-Fard S, Honarmand Tamizkar K, Hussen BM, Taheri M. MicroRNA signature in liver cancer. Pathol Res Pract 2021; 219:153369. [PMID: 33626406 DOI: 10.1016/j.prp.2021.153369] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/31/2021] [Accepted: 02/02/2021] [Indexed: 12/24/2022]
Abstract
Liver cancer is the 7th utmost frequent neoplasm and the 4th principal source of cancer deaths. This malignancy is linked with several environmental and lifestyle-related factors emphasizing the role of epigenetics in its pathogenesis. MicroRNAs (miRNAs) have been regarded as potent epigenetic mechanisms partaking in the pathogenesis of liver cancer. Dysregulation of miRNAs has been related with poor outcome of patients with liver cancer. In the current manuscript, we provide a concise review of the results of recent studies about the role of miRNAs in the progression of liver cancer and their diagnostic and prognostic utility.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Bashdar Mahmud Hussen
- Pharmacognosy Department, College of Pharmacy, Hawler Medical University, Erbil, Iraq
| | - Mohammad Taheri
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
29
|
|
30
|
Zhao MM, Ge LY, Yang LF, Zheng HX, Chen G, Wu LZ, Shi SM, Wang N, Hang YP. LncRNA NEAT1/ miR-204/ NUAK1 Axis is a Potential Therapeutic Target for Non-Small Cell Lung Cancer. Cancer Manag Res 2020; 12:13357-13368. [PMID: 33402847 PMCID: PMC7778439 DOI: 10.2147/cmar.s277524] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 11/23/2020] [Indexed: 12/17/2022] Open
Abstract
Background Long non-coding RNA (lncRNA) is a key part of non-coding RNA, and more and more evidence has revealed that it plays a vital role in tumors. NEAT1 is a lncRNA discovered in the early stage. However, it is still unclear whether NEAT1 and miR-204 play a regulatory role in lung cancer (LC). This research aimed to determine the biological function of NEAT1/miR-204 in non-small cell lung cancer (NSCLC). Materials and Methods In order to research the function of NEAT1 in NSCLC, RT-PCR, Western blot, luciferase assay and RNA immunoprecipitation assay were used to determine the relationship between NEAT1, miR-204 and NUAK1. CCK8 test, cell migration and invasion test were used to explore the influence of NEAT1 on proliferation and metastasis of LC cells. Tumor allotransplantation was used to detect the influence of NEAT1 on the growth of LC. Results The results revealed that NEAT1 was obviously enhanced in LC cell lines. Further functional analysis showed that low expression of NEAT1 obviously suppressed the growth, migration and invasion of NSCLC and facilitated cell apoptosis. Determination of luciferase reporter gene revealed that miR-204 was the direct target of NEAT1 in LC. In addition, NUAK1 was called the direct target of miR-204, and miR-204/NUAK1 had saved the role of NEAT1 in NSCLC cells. Tumor allotransplantation experiments showed that knocking down NEAT1 could inhibit the growth of LC. Conclusion In summary, our results showed that the down-regulation of NEAT1 in NSCLC inhibited its growth, migration and invasion through the miR-204/NUAK1 axis.
Collapse
Affiliation(s)
- Ming-Ming Zhao
- Department of Respiratory and Critical Care Medicine, People's Hospital of Gaochun, Nanjing 211300, People's Republic of China
| | - Lin-Yang Ge
- Department of Respiratory and Critical Care Medicine, People's Hospital of Gaochun, Nanjing 211300, People's Republic of China
| | - Liang-Feng Yang
- Department of Respiratory and Critical Care Medicine, People's Hospital of Gaochun, Nanjing 211300, People's Republic of China
| | - Hai-Xia Zheng
- Department of Respiratory and Critical Care Medicine, People's Hospital of Gaochun, Nanjing 211300, People's Republic of China
| | - Gang Chen
- Department of Respiratory and Critical Care Medicine, People's Hospital of Gaochun, Nanjing 211300, People's Republic of China
| | - Li-Zheng Wu
- Department of Respiratory and Critical Care Medicine, People's Hospital of Gaochun, Nanjing 211300, People's Republic of China
| | - Shao-Ming Shi
- Department of Respiratory and Critical Care Medicine, People's Hospital of Gaochun, Nanjing 211300, People's Republic of China
| | - Nan Wang
- Department of Respiratory and Critical Care Medicine, People's Hospital of Gaochun, Nanjing 211300, People's Republic of China
| | - Yan-Ping Hang
- Department of Respiratory and Critical Care Medicine, People's Hospital of Gaochun, Nanjing 211300, People's Republic of China
| |
Collapse
|
31
|
Xu B, Mei J, Ji W, Huo Z, Bian Z, Jiao J, Li X, Sun J, Shao J. MicroRNAs involved in the EGFR pathway in glioblastoma. Biomed Pharmacother 2020; 134:111115. [PMID: 33341046 DOI: 10.1016/j.biopha.2020.111115] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 11/26/2020] [Accepted: 12/04/2020] [Indexed: 12/14/2022] Open
Abstract
Glioblastoma (GBM) is the most common primary malignant tumor in adults, and its morbidity and mortality are very high. Although progress has been achieved in the treatment of GBM, such as surgery, chemotherapy and radiotherapy, in recent years, the prognosis of patients with GBM has not improved significantly. MicroRNAs (miRNAs) are endogenous noncoding single-stranded RNAs consisting of approximately 20-22 nucleotides that regulate gene expression at the posttranscriptional level by binding to target protein-encoding mRNAs. Notably, miRNAs regulate various carcinogenic pathways, one of which is the epidermal growth factor receptor (EGFR) signaling pathway, which controls cell proliferation, invasion, migration, angiogenesis and apoptosis. In this review, we summarize the novel discoveries of roles for miRNAs targeting the factors in the EGFR signaling pathway in the occurrence and development of GBM. In addition, we describe their potential roles as biomarkers for the diagnosis and prognosis of GBM and for determining the treatment resistance of GBM and the efficacy of therapeutic drugs.
Collapse
Affiliation(s)
- Bin Xu
- Department of Neurosurgery, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, PR China.
| | - Jie Mei
- Department of Oncology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, PR China.
| | - Wei Ji
- Department of Neurosurgery, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, PR China.
| | - Zhengyuan Huo
- Department of Neurosurgery, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, PR China.
| | - Zheng Bian
- Department of Neurosurgery, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, PR China.
| | - Jiantong Jiao
- Department of Neurosurgery, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, PR China.
| | - Xiaoqing Li
- Department of Geriatrics, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, PR China.
| | - Jun Sun
- Department of Neurosurgery, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, PR China.
| | - Junfei Shao
- Department of Neurosurgery, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, PR China.
| |
Collapse
|
32
|
Ashrafizadeh M, Zarrabi A, Orouei S, Kiavash Hushmandi, Hakimi A, Amirhossein Zabolian, Daneshi S, Samarghandian S, Baradaran B, Najafi M. MicroRNA-mediated autophagy regulation in cancer therapy: The role in chemoresistance/chemosensitivity. Eur J Pharmacol 2020; 892:173660. [PMID: 33310181 DOI: 10.1016/j.ejphar.2020.173660] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 10/10/2020] [Accepted: 10/20/2020] [Indexed: 12/20/2022]
Abstract
Chemoresistance has doubled the effort needed to reach an effective treatment for cancer. Now, scientists should consider molecular pathways and mechanisms involved in chemoresistance to overcome cancer. Autophagy is a "self-digestion" mechanism in which potentially toxic and aged organelles and macromolecules are degraded. Increasing evidence has shown that autophagy possesses dual role in cancer cells (onco-suppressor or oncogene). So, it is vital to identify its role in cancer progression and malignancy. MicroRNAs (miRs) are epigenetic factors capable of modulation of autophagy in cancer cells. In the current review, we emphasize on the relationship between miRs and autophagy in cancer chemotherapy. Besides, we discuss upstream mediators of miR/autophagy axis in cancer chemotherapy including long non-coding RNAs, circular RNAs, Nrf2 c-Myc, and HIF-1α. At the final section, we provide a discussion about how anti-tumor compounds affect miR/autophagy axis in ensuring chemosensitivity. These topics are described in this review to show how autophagy inhibition/induction can lead to chemosensitivity/chemoresistance, and miRs are considered as key players in these discussions.
Collapse
Affiliation(s)
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, 34956, Istanbul, Turkey
| | - Sima Orouei
- Department of Genetics, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology & Zoonoses, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Azadeh Hakimi
- Department of Anatomical Sciences, School of Medicine, Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amirhossein Zabolian
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Salman Daneshi
- Department of Public Health, School of Health, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Saeed Samarghandian
- Department of Basic Medical Sciences, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Masoud Najafi
- Medical Technology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran; Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
33
|
Moghadam ER, Ang HL, Asnaf SE, Zabolian A, Saleki H, Yavari M, Esmaeili H, Zarrabi A, Ashrafizadeh M, Kumar AP. Broad-Spectrum Preclinical Antitumor Activity of Chrysin: Current Trends and Future Perspectives. Biomolecules 2020; 10:E1374. [PMID: 32992587 PMCID: PMC7600196 DOI: 10.3390/biom10101374] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 09/21/2020] [Accepted: 09/23/2020] [Indexed: 02/06/2023] Open
Abstract
Pharmacological profile of phytochemicals has attracted much attention to their use in disease therapy. Since cancer is a major problem for public health with high mortality and morbidity worldwide, experiments have focused on revealing the anti-tumor activity of natural products. Flavonoids comprise a large family of natural products with different categories. Chrysin is a hydroxylated flavonoid belonging to the flavone category. Chrysin has demonstrated great potential in treating different disorders, due to possessing biological and therapeutic activities, such as antioxidant, anti-inflammatory, hepatoprotective, neuroprotective, etc. Over recent years, the anti-tumor activity of chrysin has been investigated, and in the present review, we provide a mechanistic discussion of the inhibitory effect of chrysin on proliferation and invasion of different cancer cells. Molecular pathways, such as Notch1, microRNAs, signal transducer and activator of transcription 3 (STAT3), nuclear factor-kappaB (NF-κB), PI3K/Akt, MAPK, etc., as targets of chrysin are discussed. The efficiency of chrysin in promoting anti-tumor activity of chemotherapeutic agents and suppressing drug resistance is described. Moreover, poor bioavailability, as one of the drawbacks of chrysin, is improved using various nanocarriers, such as micelles, polymeric nanoparticles, etc. This updated review will provide a direction for further studies in evaluating the anti-tumor activity of chrysin.
Collapse
Affiliation(s)
- Ebrahim Rahmani Moghadam
- Department of Anatomical Sciences, School of Medicine, Shiraz University of Medical Sciences, Shiraz 7134814336, Iran;
| | - Hui Li Ang
- Cancer Science Institute of Singapore and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore;
| | - Sholeh Etehad Asnaf
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, North Tehran Branch, IslamicAzad University, Tehran 165115331, Iran;
| | - Amirhossein Zabolian
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran 1916893813, Iran; (A.Z.); (H.S.); (H.E.)
| | - Hossein Saleki
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran 1916893813, Iran; (A.Z.); (H.S.); (H.E.)
| | - Mohammad Yavari
- Nursing and Midwifery Department, Islamic Azad University, Tehran Medical Sciences Branch, Tehran 1916893813, Iran;
| | - Hossein Esmaeili
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran 1916893813, Iran; (A.Z.); (H.S.); (H.E.)
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, Istanbul 34956, Turkey
| | - Milad Ashrafizadeh
- Faculty of Veterinary Medicine, University of Tabriz, Tabriz 5166616471, Iran
| | - Alan Prem Kumar
- Cancer Science Institute of Singapore and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore;
| |
Collapse
|
34
|
Chen Y, Xie X, Wang C, Hu Y, Zhang H, Zhang L, Tu S, He Y, Li Y. Dual targeting of NUAK1 and ULK1 using the multitargeted inhibitor MRT68921 exerts potent antitumor activities. Cell Death Dis 2020; 11:712. [PMID: 32873786 PMCID: PMC7463258 DOI: 10.1038/s41419-020-02885-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 08/01/2020] [Accepted: 08/03/2020] [Indexed: 12/16/2022]
Abstract
Utilizing oxidative stress has recently been regarded as a potential strategy for tumor therapy. The NUAK family SNF1-like kinase 1 (NUAK1) is a critical component of the antioxidant defense system and is necessary for the survival of tumors. Therefore, NUAK1 is considered an attractive therapeutic target in cancer. However, antioxidant therapy induced elevated ROS levels to activate the Unc-51-like kinase 1 (ULK1) pathway to promote protective autophagy and ULK1-dependent mitophagy. Thus, the combined inhibition of NUAK1 and ULK1 showed a strong synergistic effect in different tumor types. Herein, the potential antitumor activities of a dual NUAK1/ULK1 inhibitor MRT68921 were evaluated in both tumor cell lines and animal models. MRT68921 significantly kills tumor cells by breaking the balance of oxidative stress signals. These results highlight the potential of MRT68921 as an effective agent for tumor therapy.
Collapse
Affiliation(s)
- Yiran Chen
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoling Xie
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Chunsheng Wang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yuxing Hu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Honghao Zhang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Lenghe Zhang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Sanfang Tu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yanjie He
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| | - Yuhua Li
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China. .,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China.
| |
Collapse
|
35
|
张 丽, 白 俊, 胡 雅, 周 丹, 郑 荃, 尹 崇, 牟 青, 李 洪. [MiR-204 inhibits invasion and metastasis of breast cancer cells by targeted regulation of HNRNPA2B1]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2020; 40:869-875. [PMID: 32895200 PMCID: PMC7321266 DOI: 10.12122/j.issn.1673-4254.2020.06.15] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Indexed: 12/14/2022]
Abstract
OBJECTIVE To investigate the effect of miR-204 on the invasion and metastasis of breast cancer by targeted regulation of HNRNPA2B1. METHODS The bioinformatics database was used to obtain data of the expressions of miR-204 in breast cancer patients and the survival rate of the patients. RT-qPCR was used to detect the expression of miR-204 in breast cancer cell lines. The expression vector GV369-miR-204 was used to overexpress miR-204 in MDA-MB-231 cells. Transwell assay was performed to detect the effect of miR-204 on the migration and invasion ability of the breast cancer cells. The key genes (hub genes) of miR-204 were determined by bioinformatics method. A dual luciferase assay was used to analyze the targeting relationship between miR-204 and HNRNPA2B1. The expression of HNRNPA2B1 in MDA-MB-231 cells after miR-204 overexpression was detected by Western blotting, and Transwell assay was used to examine the changes in the cell invasion ability. RESULTS The expression of miR-204 was decreased in both breast cancer tissues, and was significantly lower in breast cancer MDA-MB-231 cells than in MCF-10A cells (P < 0.05). The decreased expression of miR-204 was associated with poorer prognosis of breast cancer patients (P < 0.05). Upregulation of miR-204 in MDA-MB-231 cells significantly inhibited the invasion and migration of the cells (P < 0.05). Analysis of the data from the Starbase revealed that the expression of miR-204-5p was negatively correlated with the expression of HNRNPA2B1, and the expression of HNRNPA2B1 was increased in breast cancer patients (P < 0.05) in association with a poorer prognosis of the patients (P < 0.05). Dual luciferase assay demonstrated that miR-204 could bind to HNRNPA2B1 in a target-specific manner. Western blotting and Transwell assay showed that miR-204 significant inhibited the migration and invasion ability of breast cancer cells by targeting HNRNPA2B1 (P < 0.05). CONCLUSIONS miR-204 expression is decreased in breast cancer tissues and cells, and its overexpression can inhibit the invasion and metastasis of breast cancer cells by targeted regulation of HNRNPA2B1.
Collapse
Affiliation(s)
- 丽萍 张
- 潍坊医学院 病理学教研室, 山东 潍坊 261053Department of Pathology, Weifang Medical University, Weifang 261053, China
| | - 俊 白
- 潍坊医学院 病理学教研室, 山东 潍坊 261053Department of Pathology, Weifang Medical University, Weifang 261053, China
| | - 雅琼 胡
- 潍坊医学院 病理学教研室, 山东 潍坊 261053Department of Pathology, Weifang Medical University, Weifang 261053, China
| | - 丹丹 周
- 潍坊医学院 病理学教研室, 山东 潍坊 261053Department of Pathology, Weifang Medical University, Weifang 261053, China
| | - 荃 郑
- 潍坊医学院 病理学教研室, 山东 潍坊 261053Department of Pathology, Weifang Medical University, Weifang 261053, China
| | - 崇高 尹
- 潍坊医学院 护理学院, 山东 潍坊 261053College of Nursing, Weifang Medical University, Weifang 261053, China
| | - 青杰 牟
- 潍坊医学院 临床医学院, 山东 潍坊 261053Clinical Medical College, Weifang Medical University, Weifang 261053, China
| | - 洪利 李
- 潍坊医学院 医学研究实验中心, 山东 潍坊 261053Medicine Research Center, Weifang Medical University, Weifang 261053, China
| |
Collapse
|
36
|
Ala U. Competing Endogenous RNAs, Non-Coding RNAs and Diseases: An Intertwined Story. Cells 2020; 9:E1574. [PMID: 32605220 PMCID: PMC7407898 DOI: 10.3390/cells9071574] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/18/2020] [Accepted: 06/23/2020] [Indexed: 01/17/2023] Open
Abstract
MicroRNAs (miRNAs), a class of small non-coding RNA molecules, are responsible for RNA silencing and post-transcriptional regulation of gene expression. They can mediate a fine-tuned crosstalk among coding and non-coding RNA molecules sharing miRNA response elements (MREs). In a suitable environment, both coding and non-coding RNA molecules can be targeted by the same miRNAs and can indirectly regulate each other by competing for them. These RNAs, otherwise known as competing endogenous RNAs (ceRNAs), lead to an additional post-transcriptional regulatory layer, where non-coding RNAs can find new significance. The miRNA-mediated interplay among different types of RNA molecules has been observed in many different contexts. The analyses of ceRNA networks in cancer and other pathologies, as well as in other physiological conditions, provide new opportunities for interpreting omics data for the field of personalized medicine. The development of novel computational tools, providing putative predictions of ceRNA interactions, is a rapidly growing field of interest. In this review, I discuss and present the current knowledge of the ceRNA mechanism and its implications in a broad spectrum of different pathologies, such as cardiovascular or autoimmune diseases, cancers and neurodegenerative disorders.
Collapse
Affiliation(s)
- Ugo Ala
- Department of Veterinary Sciences, University of Turin, 10124 Turin, Italy
| |
Collapse
|
37
|
Goel A, Ward DG, Gordon NS, Abbotts B, Zeegers MP, Cheng KK, James ND, Bryan RT, Arnold R. Back-Splicing Transcript Isoforms (Circular RNAs) Affect Biologically Relevant Pathways and Offer an Additional Layer of Information to Stratify NMIBC Patients. Front Oncol 2020; 10:812. [PMID: 32670866 PMCID: PMC7326039 DOI: 10.3389/fonc.2020.00812] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 04/24/2020] [Indexed: 12/24/2022] Open
Abstract
Circularized transcript isoforms due to back-splicing are increasingly being reported in different tissues types and pathological states including cancer. Since these circular RNAs (circRNAs) are more stable than linear messenger RNA their identification and profiling in tumor tissue could aid in stratifying patients and may serve as biomarkers. In this study, we have investigated the relationship between circRNA expression and tumor grade in a cohort of 58, mostly non-muscle-invasive bladder cancer patients. From 4571 circRNAs detected, we identified 157 that were significantly differentially expressed between tumor grades relative to the linear transcript. We demonstrated that such grade-related differences can be identified in an independent cohort, and that a large fraction of circRNAs can be, in principle, detected in urine. The differentially expressed circRNAs cluster into subgroups according to their co-expression, subgroups which are enriched for DNA repair, cell cycle and intracellular signaling genes. Since one proposed function of circRNAs is to interfere with gene-regulation by acting as microRNA “sponges,” candidates which were differentially expressed between tumor grades were investigated for potential miRNA target sites. By investigating the circRNAs from bladder cancer related pathways we demonstrated that the expression of these pathways, the circRNAs, and their parental genes are often decoupled and do not correlate, yet that some circRNAs do not follow this tendency. The present study provides the next step for the comprehensive evaluation of this novel class of RNAs in the context of non-muscle-invasive bladder cancer. Intriguingly, despite their possible function as microRNA sponges, they potentially affect host mRNA levels at the transcriptional stage, as compared to post-transcriptional control by miRNAs. Our analysis indicates differences of their activity between bladder cancer tumor stages, and their relative expression levels may provide an additional layer of information for patient stratification.
Collapse
Affiliation(s)
- Anshita Goel
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Douglas G Ward
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Naheema S Gordon
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Ben Abbotts
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Maurice P Zeegers
- NUTRIM School for Nutrition and Translational Research in Metabolism & CAPHRI Care and Public Health Research Institute, Maastricht University, Maastricht, Netherlands
| | - K K Cheng
- Institute of Applied Health Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Nicholas D James
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Richard T Bryan
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Roland Arnold
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
38
|
Orlandella FM, Mariniello RM, Mirabelli P, De Stefano AE, Iervolino PLC, Lasorsa VA, Capasso M, Giannatiempo R, Rongo M, Incoronato M, Messina F, Salvatore M, Soricelli A, Salvatore G. miR-622 is a novel potential biomarker of breast carcinoma and impairs motility of breast cancer cells through targeting NUAK1 kinase. Br J Cancer 2020; 123:426-437. [PMID: 32418991 PMCID: PMC7403386 DOI: 10.1038/s41416-020-0884-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 04/03/2020] [Accepted: 04/24/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Aberrant expression of microRNAs (miR) has been proposed as non-invasive biomarkers for breast cancers. The aim of this study was to analyse the miR-622 level in the plasma and in tissues of breast cancer patients and to explore the role of miR-622 and its target, the NUAK1 kinase, in this context. METHODS miR-622 expression was analysed in plasma and in tissues samples of breast cancer patients by q-RT-PCR. Bioinformatics programs, luciferase assay, public dataset analysis and functional experiments were used to uncover the role of miR-622 and its target in breast cancer cells. RESULTS miR-622 is downregulated in plasma and in tissues of breast cancer patients respect to healthy controls and its downregulation is significantly associated with advanced grade and high Ki67 level. Modulation of miR-622 affects the motility phenotype of breast cancer cells. NUAK1 kinase is a functional target of miR-622, it is associated with poor clinical outcomes of breast cancer patients and is inversely correlated with miR-622 level. CONCLUSIONS miR-622/NUAK1 axis is deregulated in breast cancer patients and affects the motility phenotype of breast cancer cells. Importantly, miR-622 and NUAK1 hold promises as biomarkers and as targets for breast cancers.
Collapse
Affiliation(s)
| | - Raffaela Mariarosaria Mariniello
- Dipartimento di Scienze Motorie e del Benessere, Universita' degli Studi di Napoli "Parthenope", Via Medina 40, 80133, Naples, Italy.,CEINGE - Biotecnologie Avanzate S.c.a.r.l., Via Gaetano Salvatore 486, 80145, Naples, Italy
| | | | - Anna Elisa De Stefano
- Dipartimento di Scienze Motorie e del Benessere, Universita' degli Studi di Napoli "Parthenope", Via Medina 40, 80133, Naples, Italy.,CEINGE - Biotecnologie Avanzate S.c.a.r.l., Via Gaetano Salvatore 486, 80145, Naples, Italy
| | - Paola Lucia Chiara Iervolino
- CEINGE - Biotecnologie Avanzate S.c.a.r.l., Via Gaetano Salvatore 486, 80145, Naples, Italy.,Dipartimento di Scienze Biomediche Avanzate, Universita' "Federico II", Via Pansini 5, 80131, Napoli, Italy
| | - Vito Alessandro Lasorsa
- CEINGE - Biotecnologie Avanzate S.c.a.r.l., Via Gaetano Salvatore 486, 80145, Naples, Italy.,Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli "Federico II", Naples, Italy
| | - Mario Capasso
- IRCCS SDN, Via Emanuele Gianturco 113, 80143, Naples, Italy.,CEINGE - Biotecnologie Avanzate S.c.a.r.l., Via Gaetano Salvatore 486, 80145, Naples, Italy.,Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli "Federico II", Naples, Italy
| | | | - Maria Rongo
- IRCCS SDN, Via Emanuele Gianturco 113, 80143, Naples, Italy
| | | | | | | | - Andrea Soricelli
- IRCCS SDN, Via Emanuele Gianturco 113, 80143, Naples, Italy.,Dipartimento di Scienze Motorie e del Benessere, Universita' degli Studi di Napoli "Parthenope", Via Medina 40, 80133, Naples, Italy
| | - Giuliana Salvatore
- IRCCS SDN, Via Emanuele Gianturco 113, 80143, Naples, Italy. .,Dipartimento di Scienze Motorie e del Benessere, Universita' degli Studi di Napoli "Parthenope", Via Medina 40, 80133, Naples, Italy. .,CEINGE - Biotecnologie Avanzate S.c.a.r.l., Via Gaetano Salvatore 486, 80145, Naples, Italy.
| |
Collapse
|
39
|
Li HM, Yu YK, Liu Q, Wei XF, Zhang J, Zhang RX, Sun HB, Wang ZF, Xing WQ, Li Y. LncRNA SNHG1 Regulates the Progression of Esophageal Squamous Cell Cancer by the miR-204/HOXC8 Axis. Onco Targets Ther 2020; 13:757-767. [PMID: 32158227 PMCID: PMC6986417 DOI: 10.2147/ott.s224550] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 12/24/2019] [Indexed: 12/14/2022] Open
Abstract
Objective Long noncoding RNA small nucleolar RNA host gene 1 (SNHG1) has been reported to be aberrantly expressed and plays an important role in human cancers, including esophageal squamous cell cancer. However, the regulatory mechanism underlying SNHG1 in the progression of esophageal squamous cell cancer is poorly defined. Materials and Methods Fifty-three esophageal squamous cell cancer patients were recruited and overall survival was analyzed. EC9706 and KYSE150 cells were cultured for study in vitro. The expression levels of SNHG1, microRNA (miR)-204 and homeobox c8 (HOXC8) were detected by quantitative real-time polymerase chain reaction and Western blot. Cell cycle distribution, apoptosis, migration and invasion were determined by flow cytometry and transwell assays, respectively. The target interaction among SNHG1, miR-204 and HOXC8 was validated by luciferase reporter assay and RNA immunoprecipitation. Xenograft model was established to investigate the role of SNHG1 in vivo. Results High expression of SNHG1 was exhibited in esophageal squamous cell cancer and indicated poor outcomes of patients. SNHG1 silence led to cell cycle arrest at G0-G1 phase, inhibition of migration and invasion and increase of apoptosis. miR-204 was validated to sponge by SNHG1 and target HOXC8 in esophageal squamous cell cancer cells. miR-204 knockdown or HOXC8 restoration reversed the inhibitive role of SNHG1 silence in the progression of esophageal squamous cell cancer cells. Furthermore, inhibiting SNHG1 decreased xenograft tumor growth by regulating miR-204 and HOXC8. Conclusion SNHG1 knockdown suppresses migration and invasion but induces apoptosis of esophageal squamous cell cancer cells by increasing miR-204 and decreasing HOXC8.
Collapse
Affiliation(s)
- Hao Miao Li
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Yong Kui Yu
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Qi Liu
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Xiu Feng Wei
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Jun Zhang
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Rui Xiang Zhang
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Hai Bo Sun
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Zong Fei Wang
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Wen Qun Xing
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Yin Li
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China.,Department of Thoracic Surgery, The Cancer Hospital Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| |
Collapse
|