1
|
Hashemi M, Gholamrezaie H, Ziyaei F, Asadi S, Naeini ZY, Salimian N, Enayat G, Sharifi N, Aliahmadi M, Rezaie YS, Khoushab S, Rahimzadeh P, Miri H, Abedi M, Farahani N, Taheriazam A, Nabavi N, Entezari M. Role of lncRNA PVT1 in the progression of urological cancers: Novel insights into signaling pathways and clinical opportunities. Cell Signal 2025; 131:111736. [PMID: 40081549 DOI: 10.1016/j.cellsig.2025.111736] [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: 07/31/2024] [Revised: 01/02/2025] [Accepted: 03/10/2025] [Indexed: 03/16/2025]
Abstract
Urologic malignancies, encompassing cancers of the kidney, bladder, and prostate, represent approximately 25 % of all cancer cases. Recent advances have enhanced our understanding of PVT1's crucial functions. Long noncoding RNAs influence both the onset and development of cancer, as well as epigenetic alterations. Recent findings have focused on PVT1's mechanism of action across several malignancies, particularly urologic cancers. Understanding the various functions of PVT1 linked to cancer is necessary for the development of cancer detection and treatment when PVT1 is dysregulated. Furthermore, recent advancements in genomic and epigenetic research have elucidated the complex regulatory networks that control PVT1 expression. Comprehending the intricate role of PVT1 Understanding the complex function of PVT1 in urologic cancers has substantial clinical implications. Here, we summarize some of the most recent findings about the carcinogenic effects of PVT1 signaling pathways and the possible treatment strategies for urological malignancies that target these pathways.
Collapse
Affiliation(s)
- Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Hamidreza Gholamrezaie
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Faezeh Ziyaei
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Saba Asadi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Zahra Yousefian Naeini
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Cellular and Molecular Biology,Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Niloufar Salimian
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Golnaz Enayat
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Nafiseh Sharifi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Melika Aliahmadi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Yasamin Soofi Rezaie
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Saloomeh Khoushab
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Payman Rahimzadeh
- Surgical Research Society (SRS), Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| | - Hossein Miri
- Faculty of Medicine, Tehran Medical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Maryam Abedi
- Department of Pathology, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Najma Farahani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, 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.
| | - Noushin Nabavi
- Independent Researcher, Victoria, British Columbia V8V 1P7, Canada
| | - Maliheh Entezari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
2
|
Vitkūnaitė E, Žymantaitė E, Mlynska A, Andrijec D, Limanovskaja K, Kaszynski G, Matulis D, Šakalys V, Jonušauskas L. Advancing 3D Spheroid Research through 3D Scaffolds Made by Two-Photon Polymerization. Bioengineering (Basel) 2024; 11:902. [PMID: 39329644 PMCID: PMC11429241 DOI: 10.3390/bioengineering11090902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/16/2024] [Accepted: 08/27/2024] [Indexed: 09/28/2024] Open
Abstract
Three-dimensional cancer cell cultures have been a valuable research model for developing new drug targets in the preclinical stage. However, there are still limitations to these in vitro models. Scaffold-based systems offer a promising approach to overcoming these challenges in cancer research. In this study, we show that two-photon polymerization (TPP)-assisted printing of scaffolds enhances 3D tumor cell culture formation without additional modifications. TPP is a perfect fit for this task, as it is an advanced 3D-printing technique combining a μm-level resolution with complete freedom in the design of the final structure. Additionally, it can use a wide array of materials, including biocompatible ones. We exploit these capabilities to fabricate scaffolds from two different biocompatible materials-PEGDA and OrmoClear. Cubic spheroid scaffolds with a more complex architecture were produced and tested. The biological evaluation showed that the human ovarian cancer cell lines SKOV3 and A2780 formed 3D cultures on printed scaffolds without a preference for the material. The gene expression evaluation showed that the A2780 cell line exhibited substantial changes in CDH1, CDH2, TWIST, COL1A1, and SMAD3 gene expression, while the SKOV3 cell line had slight changes in said gene expression. Our findings show how the scaffold architecture design impacts tumor cell culture 3D spheroid formation, especially for the A2780 cancer cell line.
Collapse
Affiliation(s)
- Eglė Vitkūnaitė
- Vital3D Technologies, Saulėtekio al. 15, LT-10224 Vilnius, Lithuania; (E.V.); (D.A.); (K.L.); (V.Š.)
| | - Eglė Žymantaitė
- Laboratory of Immunology, National Cancer Institute, P. Baublio g. 3B, LT-08406 Vilnius, Lithuania; (E.Ž.); (A.M.)
- Life Sciences Center, Vilnius University, Sauletekio 7, LT-10257 Vilnius, Lithuania
| | - Agata Mlynska
- Laboratory of Immunology, National Cancer Institute, P. Baublio g. 3B, LT-08406 Vilnius, Lithuania; (E.Ž.); (A.M.)
- Department of Chemistry and Bioengineering, Vilnius Gediminas Technical University, Saulėtekio al. 11, LT-10223 Vilnius, Lithuania
| | - Dovilė Andrijec
- Vital3D Technologies, Saulėtekio al. 15, LT-10224 Vilnius, Lithuania; (E.V.); (D.A.); (K.L.); (V.Š.)
| | - Karolina Limanovskaja
- Vital3D Technologies, Saulėtekio al. 15, LT-10224 Vilnius, Lithuania; (E.V.); (D.A.); (K.L.); (V.Š.)
| | - Grzegorz Kaszynski
- Vital3D Technologies, Saulėtekio al. 15, LT-10224 Vilnius, Lithuania; (E.V.); (D.A.); (K.L.); (V.Š.)
| | - Daumantas Matulis
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Sauletekio 7, LT-10257 Vilnius, Lithuania;
| | - Vidmantas Šakalys
- Vital3D Technologies, Saulėtekio al. 15, LT-10224 Vilnius, Lithuania; (E.V.); (D.A.); (K.L.); (V.Š.)
| | - Linas Jonušauskas
- Vital3D Technologies, Saulėtekio al. 15, LT-10224 Vilnius, Lithuania; (E.V.); (D.A.); (K.L.); (V.Š.)
| |
Collapse
|
3
|
Ou W, Qi Z, Liu N, Zhang J, Mi X, Song Y, Fang Y, Cui B, Hou J, Yuan Z. Elucidating the role of TWIST1 in ulcerative colitis: a comprehensive bioinformatics and machine learning approach. Front Genet 2024; 15:1296570. [PMID: 38510272 PMCID: PMC10952112 DOI: 10.3389/fgene.2024.1296570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 02/16/2024] [Indexed: 03/22/2024] Open
Abstract
Background: Ulcerative colitis (UC) is a common and progressive inflammatory bowel disease primarily affecting the colon and rectum. Prolonged inflammation can lead to colitis-associated colorectal cancer (CAC). While the exact cause of UC remains unknown, this study aims to investigate the role of the TWIST1 gene in UC. Methods: Second-generation sequencing data from adult UC patients were obtained from the Gene Expression Omnibus (GEO) database. Differentially expressed genes (DEGs) were identified, and characteristic genes were selected using machine learning and Lasso regression. The Receiver Operating Characteristic (ROC) curve assessed TWIST1's potential as a diagnostic factor (AUC score). Enriched pathways were analyzed, including Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Gene Set Variation Analysis (GSVA). Functional mechanisms of marker genes were predicted, considering immune cell infiltration and the competing endogenous RNA (ceRNA) network. Results: We found 530 DEGs, with 341 upregulated and 189 downregulated genes. TWIST1 emerged as one of four potential UC biomarkers via machine learning. TWIST1 expression significantly differed in two datasets, GSE193677 and GSE83687, suggesting its diagnostic potential (AUC = 0.717 in GSE193677, AUC = 0.897 in GSE83687). Enrichment analysis indicated DEGs associated with TWIST1 were involved in processes like leukocyte migration, humoral immune response, and cell chemotaxis. Immune cell infiltration analysis revealed higher rates of M0 macrophages and resting NK cells in the high TWIST1 expression group, while TWIST1 expression correlated positively with M2 macrophages and resting NK cell infiltration. We constructed a ceRNA regulatory network involving 1 mRNA, 7 miRNAs, and 32 long non-coding RNAs (lncRNAs) to explore TWIST1's regulatory mechanism. Conclusion: TWIST1 plays a significant role in UC and has potential as a diagnostic marker. This study sheds light on UC's molecular mechanisms and underscores TWIST1's importance in its progression. Further research is needed to validate these findings in diverse populations and investigate TWIST1 as a therapeutic target in UC.
Collapse
Affiliation(s)
- Wenjie Ou
- School of Clinical Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Zhaoxue Qi
- Department of Secretory Metabolism, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Ning Liu
- General Surgery of The First Clinical Hospital of Jilin Academy of Chinese Medicine Sciences, Changchun, Jilin, China
| | - Junzi Zhang
- School of Clinical Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Xuguang Mi
- Department of Central Laboratory, Jilin Provincial People’s Hospital, Changchun, Jilin, China
| | - Yuan Song
- Department of Gastroenterology, Jilin Provincial People’s Hospital, Changchun, Jilin, China
| | - Yanqiu Fang
- Department of Central Laboratory, Jilin Provincial People’s Hospital, Changchun, Jilin, China
| | - Baiying Cui
- School of Clinical Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Junjie Hou
- Department of Comprehensive Oncology, Jilin Provincial People’s Hospital, Changchun, Jilin, China
| | - Zhixin Yuan
- Department of Emergency Surgery, Jilin Provincial People’s Hospital, Changchun, Jilin, China
| |
Collapse
|
4
|
Lv T, Liu H, Mao L, Song Y, Liao L, Zhong K, Shuai B, Luo Y, Guo T, Huang W, Zhang S. Cancer-associated fibroblast-derived extracellular vesicles promote lymph node metastases in oral cavity squamous cell carcinoma by encapsulating ITGB1 and BMI1. BMC Cancer 2024; 24:113. [PMID: 38254031 PMCID: PMC10804601 DOI: 10.1186/s12885-024-11855-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 01/07/2024] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND Extracellular vesicles (EVs) have been revealed to facilitate the development of oral squamous cavity cell carcinoma (OCSCC), while its supporting role in lymph node metastases is under continuous investigation. This study aimed to examine the function of cancer-associated fibroblasts (CAF)-derived EVs (CAF-EVs) during lymph node metastasis in OCSCC and the mechanisms. METHODS CAF were isolated from OCSCC tissues of patients, and CAF-EVs were extracted and identified. EdU, colony formation, wound healing, and Transwell assays were performed. The OCSCC cells before and after CAF-EVs treatment were injected into mice to probe the effects of CAF-EVs on tumor growth and lymph node metastasis, respectively. The effect of CAF-EVs treatment on transcriptome changes in OCSCC cells was analyzed. Clinical data of patients with OCSCC were analyzed to determine the prognostic significance of the selected genes. Finally, loss-of-function assays were conducted to corroborate the involvement of polycomb complex protein BMI-1 (BMI1) and integrin beta1 (ITGB1). RESULTS CAF-EVs promoted the malignant behavior of OCSCC cells and accelerated tumor growth and lymph node metastasis in mice. CAF-EVs significantly increased the expression of BMI1 and ITGB1, and the expression of BMI1 and ITGB1 was negatively correlated with the overall survival and relapse-free survival of OCSCC patients. Knockdown of BMI1 or ITGB1 in OCSCC cells abated the promoting effects of CAF-EVs in vitro and in vivo. CONCLUSION CAF-EVs elicited the metastasis-promoting properties in OCSCC by elevating BMI1 and ITGB1, suggesting that BMI1 and ITGB1 could be potential biomarkers and therapeutic targets for OCSCC.
Collapse
Affiliation(s)
- Tianzhu Lv
- Guizhou Medical University, 550004, Guiyang, Guizhou, P.R. China
- China-British Joint Molecular Head and Neck Cancer Research Laboratory, Stomatological Hospital of Guizhou Medical University, 550004, Guiyang, Guizhou, P.R. China
| | - Hongjing Liu
- Comprehensive Emergency Department of Stomatology, Stomatological Hospital of Guizhou Medical University, 550004, Guiyang, Guizhou, P.R. China
| | - Ling Mao
- Guizhou Medical University, 550004, Guiyang, Guizhou, P.R. China
- China-British Joint Molecular Head and Neck Cancer Research Laboratory, Stomatological Hospital of Guizhou Medical University, 550004, Guiyang, Guizhou, P.R. China
| | - Yanrong Song
- Comprehensive Emergency Department of Stomatology, Stomatological Hospital of Guizhou Medical University, 550004, Guiyang, Guizhou, P.R. China
| | - Lili Liao
- Comprehensive Emergency Department of Stomatology, Stomatological Hospital of Guizhou Medical University, 550004, Guiyang, Guizhou, P.R. China
| | - Kun Zhong
- Comprehensive Emergency Department of Stomatology, Stomatological Hospital of Guizhou Medical University, 550004, Guiyang, Guizhou, P.R. China
| | - Binbin Shuai
- Comprehensive Emergency Department of Stomatology, Stomatological Hospital of Guizhou Medical University, 550004, Guiyang, Guizhou, P.R. China
| | - Yingkun Luo
- Comprehensive Emergency Department of Stomatology, Stomatological Hospital of Guizhou Medical University, 550004, Guiyang, Guizhou, P.R. China
| | - Tingting Guo
- Comprehensive Emergency Department of Stomatology, Stomatological Hospital of Guizhou Medical University, 550004, Guiyang, Guizhou, P.R. China
| | - Wentao Huang
- School of Savaid Stomatology, Hangzhou Medical College, 311399, Hangzhou, Zhejiang, P.R. China.
| | - Shenyingjie Zhang
- Medical Department, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), 310006, Hangzhou, Zhejiang, P.R. China
| |
Collapse
|
5
|
Githaka JM, Pirayeshfard L, Goping IS. Cancer invasion and metastasis: Insights from murine pubertal mammary gland morphogenesis. Biochim Biophys Acta Gen Subj 2023; 1867:130375. [PMID: 37150225 DOI: 10.1016/j.bbagen.2023.130375] [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: 12/20/2022] [Revised: 04/20/2023] [Accepted: 05/02/2023] [Indexed: 05/09/2023]
Abstract
Cancer invasion and metastasis accounts for the majority of cancer related mortality. A better understanding of the players that drive the aberrant invasion and migration of tumors cells will provide critical targets to inhibit metastasis. Postnatal pubertal mammary gland morphogenesis is characterized by highly proliferative, invasive, and migratory normal epithelial cells. Identifying the molecular regulators of pubertal gland development is a promising strategy since tumorigenesis and metastasis is postulated to be a consequence of aberrant reactivation of developmental stages. In this review, we summarize the pubertal morphogenesis regulators that are involved in cancer metastasis and revisit pubertal mammary gland transcriptome profiling to uncover both known and unknown metastasis genes. Our updated list of pubertal morphogenesis regulators shows that most are implicated in invasion and metastasis. This review highlights molecular linkages between development and metastasis and provides a guide for exploring novel metastatic drivers.
Collapse
Affiliation(s)
- John Maringa Githaka
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada.
| | - Leila Pirayeshfard
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Ing Swie Goping
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada; Department of Oncology, University of Alberta, Edmonton, AB T6G 2H7, Canada.
| |
Collapse
|
6
|
Quarshie JT, Fosu K, Offei NA, Sobo AK, Quaye O, Aikins AR. Cryptolepine Suppresses Colorectal Cancer Cell Proliferation, Stemness, and Metastatic Processes by Inhibiting WNT/β-Catenin Signaling. Pharmaceuticals (Basel) 2023; 16:1026. [PMID: 37513937 PMCID: PMC10383422 DOI: 10.3390/ph16071026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/25/2023] [Accepted: 07/04/2023] [Indexed: 07/30/2023] Open
Abstract
Colorectal cancer (CRC) is the third most frequent cancer and the second leading cause of cancer-related deaths globally. Evidence shows that over 90% of CRC cases are initiated by a deregulated Wingless Integrated Type-1 (WNT)/β-catenin signaling pathway. The WNT/β-catenin pathway also promotes CRC cell proliferation, stemness, and metastasis. Therefore, modulators of the WNT/β-catenin pathway may serve as promising regimens for CRC. This study investigated the effect of cryptolepine-a plant-derived compound-on the WNT/β-catenin pathway in CRC. Two CRC cell lines, COLO205 and DLD1, were treated with cryptolepine or XAV 939 (a WNT inhibitor) in the presence or absence of WNT3a (a WNT activator). Using a tetrazolium-based assay, cryptolepine was found to reduce cell viability in a dose- and time-dependent manner and was a more potent inhibitor of viability than XAV 939. RT-qPCR analyses showed that cryptolepine reverses WNT3a-induced expression of β-catenin, c-MYC, and WISP1, suggesting that cryptolepine inhibits WNT3a-mediated activation of WNT/β-catenin signaling. Cryptolepine also repressed WNT3a-induced OCT4 and CD133 expression and suppressed colony formation of the cells, indicating that cryptolepine inhibits the stemness of CRC cells. Additionally, cryptolepine inhibited WNT3a-induced epithelial-to-mesenchymal transition by reducing the expression of SNAI1 and TWIST1 genes. In a wound healing assay, cryptolepine was found to suppress cell migration under unstimulated and WNT3a-stimulated conditions. Moreover, cryptolepine downregulated WNT3a-induced expression of MMP2 and MMP9 genes, which are involved in cancer cell invasion. Altogether, cryptolepine suppresses CRC cell proliferation, stemness, and metastatic properties by inhibiting WNT3a-mediated activation of the WNT/β-catenin signaling pathway. These findings provide a rationale for considering cryptolepine as a potential WNT inhibitor in CRC.
Collapse
Affiliation(s)
- Jude Tetteh Quarshie
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), Department of Biochemistry Cell and Molecular Biology, University of Ghana, Accra P.O. Box LG 54, Ghana
| | - Kwadwo Fosu
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), Department of Biochemistry Cell and Molecular Biology, University of Ghana, Accra P.O. Box LG 54, Ghana
| | - Nicholas Awuku Offei
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), Department of Biochemistry Cell and Molecular Biology, University of Ghana, Accra P.O. Box LG 54, Ghana
| | - Augustine Kojo Sobo
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), Department of Biochemistry Cell and Molecular Biology, University of Ghana, Accra P.O. Box LG 54, Ghana
| | - Osbourne Quaye
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), Department of Biochemistry Cell and Molecular Biology, University of Ghana, Accra P.O. Box LG 54, Ghana
| | - Anastasia Rosebud Aikins
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), Department of Biochemistry Cell and Molecular Biology, University of Ghana, Accra P.O. Box LG 54, Ghana
| |
Collapse
|
7
|
The role of BMI1 in endometrial cancer and other cancers. Gene 2023; 856:147129. [PMID: 36563713 DOI: 10.1016/j.gene.2022.147129] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 11/11/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022]
Abstract
Endometrial cancer (EC) is the third leading gynecological malignancy, and its treatment remains challenging. B cell-specific Moloney murine leukemia virus integration site-1 (BMI1) is one of the core members of the polycomb group (PcG) family, which plays a promoting role in the occurrence and development of various tumors. Notably, BMI1 has been found to be frequently upregulated in endometrial cancer (EC) and promote the occurrence of EC through promoting epithelial-mesenchymal transition (EMT) and AKT/PI3K pathways. This review summarizes the structure and upstream regulatory mechanisms of BMI1 and its role in EC. In addition, we focused on the role of BMI1 in chemoradiotherapy resistance and summarized the current drugs that target BMI1.
Collapse
|
8
|
Zhang H, Wang Y, Chen X, Zhang A, Hou L, Hong J, Liu J, Liu Z, Yang P. Targeting epithelial cell-derived TWIST1 alleviates allergic asthma. Cell Signal 2023; 102:110552. [PMID: 36481410 DOI: 10.1016/j.cellsig.2022.110552] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 11/22/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022]
Abstract
It is well known that the T Helper (Th)2 bias plays a critical role in allergic asthma. Whereas the Th2 bias is maintained in the local tissues is uncertain. IL-33 is vital for the development of the Th2 polarization. TWIST-1 has an effect on regulating cellular functions. The aberrant activation of RAS sustains certain cellular activities. The aim of this study is to study the role of the interaction between activation of TWIST1 and RAS in inducing and maintaining Th2 polarization in allergic asthma. The epithelial cells of the airways (AEC) were isolated from the broncho-alveolar lavage fluids in patients with asthma. The mediators involved in the over-expression of IL-33 were determined by RNA sequencing. A mouse model was established to test the role of TWIST1 and RAS in developing allergic asthma. We observed a strong expression of TWIST1 in patients with allergic asthma that showed a positive correlation with asthmatic responses. TWIST1 favored the expression of the IL-33 in the AEC. Twist1-deficient AEC-carrying mice did not induce Th2 polarization in the airways. The expression TWIST1 in AECs was positively associated with RAS activation in AECs in patients with allergic asthma. The interaction between RAS and TWIST1 in AECs sustained airway allergic inflammation. Inhibition of TWIST1 or RAS prevented asthma-like inflammation in the mouse airways. In summary, the interaction between TWIST1 and RAS induces and maintains IL-33 expression in AECs to facilitate allergic inflammation in the respiratory tract. Inhibition of TWIST1 or RAS can prevent experimental allergic asthma.
Collapse
Affiliation(s)
- Huanping Zhang
- Department of Allergy Medicine, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Yanfen Wang
- Department of Pediatrics, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Xiaoxue Chen
- Department of Allergy Medicine, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Aizhi Zhang
- Department of Critical care medicine, Second Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Lijun Hou
- Department of Allergy Medicine, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Jingyi Hong
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Shenzhen, China; Institute of Allergy & Immunology of Shenzhen University School of Medicine, Shenzhen, China; State Key Laboratory of Respiratory Disease Allergy Shenzhen University Division, Shenzhen, China; Guangdong Provincial Standardization Allergen Engineering Research Center, Shenzhen, China; Shenzhen Key Laboratory of Allergy and Immunology, Shenzhen, China
| | - Jiangqi Liu
- Longgang ENT Hospital and Shenzhen ENT Institute, Shenzhen, China.
| | - Zhiqiang Liu
- Longgang ENT Hospital and Shenzhen ENT Institute, Shenzhen, China.
| | - Pingchang Yang
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Shenzhen, China; Institute of Allergy & Immunology of Shenzhen University School of Medicine, Shenzhen, China; State Key Laboratory of Respiratory Disease Allergy Shenzhen University Division, Shenzhen, China; Guangdong Provincial Standardization Allergen Engineering Research Center, Shenzhen, China; Shenzhen Key Laboratory of Allergy and Immunology, Shenzhen, China.
| |
Collapse
|
9
|
Izadpanah MH, Forghanifard MM. TWIST1 Plays Role in Expression of Stemness State Markers in ESCC. Genes (Basel) 2022; 13:genes13122369. [PMID: 36553636 PMCID: PMC9777594 DOI: 10.3390/genes13122369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/07/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Stemness markers play critical roles in the maintenance of key properties of embryonic stem cells (ESCs), including the pluripotency, stemness state, and self-renewal capacities, as well as cell fate decision. Some of these features are present in cancer stem cells (CSCs). TWIST1, as a bHLH transcription factor oncogene, is involved in the epithelial-mesenchymal transition (EMT) process in both embryonic and cancer development. Our aim in this study was to investigate the functional correlation between TWIST1 and the involved genes in the process of CSCs self-renewal in human esophageal squamous cell carcinoma (ESCC) line KYSE-30. METHODS TWIST1 overexpression was enforced in the ESCC KYSE-30 cells using retroviral vector containing the specific pruf-IRES-GFP-hTWIST1 sequence. Following RNA extraction and cDNA synthesis, the mRNA expression profile of TWIST1 and the stem cell markers, including BMI1, CRIPTO1, DPPA2, KLF4, SOX2, NANOG, and MSI1, were assessed using relative comparative real-time PCR. RESULTS Ectopic expression of TWIST1 in KYSE-30 cells resulted in an increased expression of TWIST1 compared to control GFP cells by nearly 9-fold. Transduction of TWIST1-retroviral particles caused a significant enhancement in BMI1, CRIPTO1, DPPA2, KLF4, and SOX2 mRNA expression, approximately 4.5-, 3.2-, 5.5-, 3.5-, and 3.7-folds, respectively, whereas this increased TWIST1 expression caused no change in the mRNA expression of NANOG and MSI1 genes. CONCLUSIONS TWIST1 gene ectopic expression in KYSE-30 cells enhanced the level of cancer stem cell markers' mRNA expression. These results may emphasize the role of TWIST1 in the self-renewal process and may corroborate the involvement of TWIST1 in the stemness state capacity of ESCC cell line KYSE-30, as well as its potential as a therapeutic target.
Collapse
Affiliation(s)
- Mohammad Hossein Izadpanah
- Division of Human Genetics, Immunology Research Center, Avicenna Research Institute, Mashhad University of Medical Sciences, Mashhad 9196773117, Iran
| | - Mohammad Mahdi Forghanifard
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan 3671637849, Iran
- Correspondence: or ; Tel.: +98-912-711-6027
| |
Collapse
|
10
|
Chen S, Li H, Chen S, Wang B, Zhang K. BMI1 promotes the proliferation and inhibits autophagy of breast cancer cells by activating COPZ1. Clin Transl Oncol 2022; 24:2166-2174. [PMID: 35789980 DOI: 10.1007/s12094-022-02869-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 06/01/2022] [Indexed: 06/15/2023]
Abstract
PURPOSE This study was designed to explore the role of COPZ1 in breast cancer as well as discuss its specific reaction mechanism. METHODS With the help of RT-qPCR and western blot, the expression of BMI1 and COPZ1 were measured. Then, the proliferation, colony formation and apoptosis were evaluated by CCK-8, colony formation and TUNEL assays, separately. Luciferase reporter assay and ChIP were applied to assess the relative activity of COPZ1 promoter as well as its binding with BMI1. Moreover, western blot was utilized to measure the expression of proliferation-, apoptosis- and autophagy-related proteins. RESULTS According to GEPIA2 database, COPZ1 was upregulated in breast cancer tissues and was associated with the poor prognosis (P = 0.03). Results obtained from RT-qPCR and western blot verified that COPZ1 expression was greatly increased at both mRNA and protein levels in breast cancer cells as compared to control cells (P < 0.05 or P < 0.001). COPZ1 knockdown inhibited the proliferation, induced the autophagy and promoted the apoptosis of breast cancer cells. HumanTFDB predicted the binding sites of BMI1 and COPZ1. The increased relative luciferase activity of COPZ1 promoter following BMI1 overexpression (P < 0.001) and the binding of BMI1 with COPZ1 promoter indicated that BMI1 could activate COPZ1. Further experiments suggested that the effects of COPZ1 knockdown on the proliferation, apoptosis and autophagy of breast cancer cells were reversed by BMI1 overexpression, implying that BMI1 promoted the proliferation and repressed the autophagy of breast cancer cells via activating COPZ1. CONCLUSIONS To sum up, BMI1 exhibited promotive effects on the malignant progression of breast cancer through the activation of COPZ1. These findings might offer a preliminary theoretical basis for COPZ1 participation in autophagy in breast cancer cells.
Collapse
Affiliation(s)
- Shuming Chen
- Department of General Surgery, 900 Hospital of the Joint Logistics Supports Force, Fuzhou, 350025, Fujian, China
| | - He Li
- Department of General Surgery, 900 Hospital of the Joint Logistics Supports Force, Fuzhou, 350025, Fujian, China
| | - Siyu Chen
- Department of Oncology, 900 Hospital of the Joint Logistics Supports Force, No.156 West Second Ring North Road, Gulou District, Fuzhou, 350025, Fujian, China
| | - Bing Wang
- Department of General Surgery, 900 Hospital of the Joint Logistics Supports Force, Fuzhou, 350025, Fujian, China
| | - Kaixiang Zhang
- Department of Oncology, 900 Hospital of the Joint Logistics Supports Force, No.156 West Second Ring North Road, Gulou District, Fuzhou, 350025, Fujian, China.
| |
Collapse
|
11
|
Yun H, Han GH, Kim J, Chung J, Kim J, Cho H. NANOG
regulates epithelial–mesenchymal transition via
AMPK
/
mTOR
signalling pathway in ovarian cancer
SKOV
‐3 and
A2780
cells. J Cell Mol Med 2022; 26:5277-5291. [PMID: 36114703 PMCID: PMC9575063 DOI: 10.1111/jcmm.17557] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 08/26/2022] [Accepted: 09/02/2022] [Indexed: 11/28/2022] Open
Abstract
NANOG engages with tumour initiation and metastasis by regulating the epithelial–mesenchymal transition (EMT) in epithelial ovarian cancer (EOC). However, its role in association with pAMPKα, and its clinical significance in EOC have not been elucidated even though AMPK is known to degrade NANOG in various human cancers. Hence, we investigated the role of pAMPKα and its association with NANOG as potential prognostic biomarkers in EOC. Both NANOG and pAMPKα expression were significantly overexpressed in EOCs comparing nonadjacent normal epithelial tissues, benign tissues, and borderline tumours. NANOG overexpression was significantly associated with poor disease‐free survival (DFS) and overall survival (OS), whereas pAMPKα overexpression was associated with good DFS and OS. Importantly, multivariate analysis revealed that the combination of high NANOG and low pAMPKα expression was a poor independent prognostic factor for DFS and was associated with platinum resistance. In ovarian cancer cell lines, siRNA‐mediated NANOG knockdown diminished migration and invasion properties by regulating the EMT process via the AMPK/mTOR signalling pathway. Furthermore, treatment with AMPK activator suppressed expression of stemness factors such as NANOG, Oct4 and Sox2. Collectively, these findings established that the combination of high NANOG and low pAMPKα expression was associated with EOC progression and platinum resistance, suggesting a potential prognostic biomarker for clinical management in EOC patients.
Collapse
Affiliation(s)
- Hee Yun
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital Yonsei University College of Medicine Seoul Korea
| | - Gwan Hee Han
- Department of Obstetrics and Gynecology Kyung Hee University Hospital at Gangdong Seoul Korea
| | - Julie Kim
- Weill Cornell Medical College New York New York USA
| | - Joon‐Yong Chung
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute National Institutes of Health Bethesda Maryland USA
| | - Jae‐Hoon Kim
- Department of Obstetrics and Gynecology Yonsei University College of Medicine Seoul Korea
- Institute of Women's Life Medical Science Yonsei University College of Medicine Seoul Korea
| | - Hanbyoul Cho
- Department of Obstetrics and Gynecology Yonsei University College of Medicine Seoul Korea
- Institute of Women's Life Medical Science Yonsei University College of Medicine Seoul Korea
| |
Collapse
|
12
|
Xu J, Li L, Shi P, Cui H, Yang L. The Crucial Roles of Bmi-1 in Cancer: Implications in Pathogenesis, Metastasis, Drug Resistance, and Targeted Therapies. Int J Mol Sci 2022; 23:ijms23158231. [PMID: 35897796 PMCID: PMC9367737 DOI: 10.3390/ijms23158231] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/22/2022] [Accepted: 07/23/2022] [Indexed: 12/01/2022] Open
Abstract
B-cell-specific Moloney murine leukemia virus integration region 1 (Bmi-1, also known as RNF51 or PCGF4) is one of the important members of the PcG gene family, and is involved in regulating cell proliferation, differentiation and senescence, and maintaining the self-renewal of stem cells. Many studies in recent years have emphasized the role of Bmi-1 in the occurrence and development of tumors. In fact, Bmi-1 has multiple functions in cancer biology and is closely related to many classical molecules, including Akt, c-MYC, Pten, etc. This review summarizes the regulatory mechanisms of Bmi-1 in multiple pathways, and the interaction of Bmi-1 with noncoding RNAs. In particular, we focus on the pathological processes of Bmi-1 in cancer, and explore the clinical relevance of Bmi-1 in cancer biomarkers and prognosis, as well as its implications for chemoresistance and radioresistance. In conclusion, we summarize the role of Bmi-1 in tumor progression, reveal the pathophysiological process and molecular mechanism of Bmi-1 in tumors, and provide useful information for tumor diagnosis, treatment, and prognosis.
Collapse
Affiliation(s)
- Jie Xu
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400716, China; (J.X.); (L.L.); (P.S.)
- Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, China
| | - Lin Li
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400716, China; (J.X.); (L.L.); (P.S.)
| | - Pengfei Shi
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400716, China; (J.X.); (L.L.); (P.S.)
- Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, China
| | - Hongjuan Cui
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400716, China; (J.X.); (L.L.); (P.S.)
- Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, China
- Correspondence: (H.C.); (L.Y.)
| | - Liqun Yang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400716, China; (J.X.); (L.L.); (P.S.)
- Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, China
- Correspondence: (H.C.); (L.Y.)
| |
Collapse
|
13
|
Shao J, Feng Q, Jiang W, Yang Y, Liu Z, Li L, Yang W, Zou Y. E3 ubiquitin ligase RBX1 drives the metastasis of triple negative breast cancer through a FBXO45-TWIST1-dependent degradation mechanism. Aging (Albany NY) 2022; 14:5493-5510. [PMID: 35802537 PMCID: PMC9320552 DOI: 10.18632/aging.204163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 06/23/2022] [Indexed: 11/25/2022]
Abstract
Triple-negative breast cancer (TNBC) patients are at high risk of recurrence and metastasis in the early stages, although receiving standard treatment. However, the underlying mechanism of TNBC remains unclear. Here, the critical effect of E3 ubiquitin ligase RBX1 in the metastasis of TNBC was reported for the first time. We discovered that RBX1 expression was evidently raised in the tissues of TNBC. Our clinical research displayed that high RBX1 expression was markedly related to poor distant invasion and survival. Functional analysis exhibited that RBX1 facilitated metastasis of TNBC cells through increasing EMT. Furthermore, we demonstrated that RBX1 knockdown increased the levels of the Twist family bHLH transcription factor 1 (TWIST1), is a significant regulator in the EMT process in some cancers. It can be observed an evident positive correlation between the TWIST1 and RBX1 levels, further confirming that EMT induced by RBX1 in TNBC cells is determined by TWIST1. Mechanistically, RBX1 modulates the expression of TWIST1 via modulating FBXO45, directly binding to FBXO45, and facilitating its degradation and ubiquitination. Briefly, our findings confirm that RBX1 is probably a new biomarker of TNBC carcinogenesis, thus suggesting that targeting the RBX1/FBXO45/TWIST1 axis may be an underlying strategy for TNBC treatment.
Collapse
Affiliation(s)
- Jun Shao
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Qian Feng
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Weifan Jiang
- Department of Urology Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Yuting Yang
- Department of Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Zhiqiang Liu
- Department of Neurology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang 330006, Jiangxi Province, China
| | - Liang Li
- Emergency Department, Jiangxi Maternal and Child Health Hospital, Nanchang 330006, Jiangxi Province, China
| | - Wenlong Yang
- Department of Infectious Diseases, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Yufeng Zou
- Department of Breast Surgery, The Third Hospital of Nanchang, Jiangxi Provincial-Key-Laboratory for Breast Diseases, Nanchang 330006, Jiangxi Province, China.,Department of Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
| |
Collapse
|
14
|
Huang H, Huo Z, Jiao J, Ji W, Huang J, Bian Z, Xu B, Shao J, Sun J. HOXC6 impacts epithelial-mesenchymal transition and the immune microenvironment through gene transcription in gliomas. Cancer Cell Int 2022; 22:170. [PMID: 35488304 PMCID: PMC9052479 DOI: 10.1186/s12935-022-02589-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 04/15/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Gliomas are the most common primary malignant tumours of the central nervous system (CNS). To improve the prognosis of glioma, it is necessary to identify molecular markers that may be useful for glioma therapy. HOXC6, an important transcription factor, is involved in multiple cancers. However, the role of HOXC6 in gliomas is not clear. METHODS Bioinformatic and IHC analyses of collected samples (n = 299) were performed to detect HOXC6 expression and the correlation between HOXC6 expression and clinicopathological features of gliomas. We collected clinical information from 177 to 299 patient samples and estimated the prognostic value of HOXC6. Moreover, cell proliferation assays were performed. We performed Gene Ontology (GO) analysis and gene set enrichment analysis (GSEA) based on ChIP-seq and public datasets to explore the biological characteristics of HOXC6 in gliomas. RNA-seq was conducted to verify the relationship between HOXC6 expression levels and epithelial-mesenchymal transition (EMT) biomarkers. Furthermore, the tumour purity, stromal and immune scores were evaluated. The relationship between HOXC6 expression and infiltrating immune cell populations and immune checkpoint proteins was also researched. RESULTS HOXC6 was overexpressed and related to the clinicopathological features of gliomas. In addition, knockdown of HOXC6 inhibited the proliferation of glioma cells. Furthermore, increased HOXC6 expression was associated with clinical progression. The biological role of HOXC6 in gliomas was primarily associated with EMT and the immune microenvironment in gliomas. High HOXC6 expression was related to high infiltration by immune cells, a low tumour purity score, a high stromal score, a high immune score and the expression of a variety of immune checkpoint genes, including PD-L1, B7-H3 and CLTA-4. CONCLUSIONS These results indicated that HOXC6 might be a key factor in promoting tumorigenesis and glioma progression by regulating the EMT signalling pathway and might represent a novel immune therapeutic target in gliomas.
Collapse
Affiliation(s)
- Hui Huang
- Department of Neurosurgery, Wuxi People's Hospital Affiliated to Nanjing Medical University, No. 299 Qing Yang Road, 214023, Wuxi, Jiangsu, China
| | - Zhengyuan Huo
- Department of Neurosurgery, Wuxi People's Hospital Affiliated to Nanjing Medical University, No. 299 Qing Yang Road, 214023, Wuxi, Jiangsu, China
| | - Jiantong Jiao
- Department of Neurosurgery, Wuxi People's Hospital Affiliated to Nanjing Medical University, No. 299 Qing Yang Road, 214023, Wuxi, Jiangsu, China
| | - Wei Ji
- Department of Neurosurgery, Wuxi People's Hospital Affiliated to Nanjing Medical University, No. 299 Qing Yang Road, 214023, Wuxi, Jiangsu, China
| | - Jin Huang
- Department of Neurosurgery, Wuxi People's Hospital Affiliated to Nanjing Medical University, No. 299 Qing Yang Road, 214023, Wuxi, Jiangsu, China
| | - Zheng Bian
- Department of Neurosurgery, Wuxi People's Hospital Affiliated to Nanjing Medical University, No. 299 Qing Yang Road, 214023, Wuxi, Jiangsu, China
| | - Bin Xu
- Department of Neurosurgery, Wuxi People's Hospital Affiliated to Nanjing Medical University, No. 299 Qing Yang Road, 214023, Wuxi, Jiangsu, China
| | - Junfei Shao
- Department of Neurosurgery, Wuxi People's Hospital Affiliated to Nanjing Medical University, No. 299 Qing Yang Road, 214023, Wuxi, Jiangsu, China.
| | - Jun Sun
- Department of Neurosurgery, Wuxi People's Hospital Affiliated to Nanjing Medical University, No. 299 Qing Yang Road, 214023, Wuxi, Jiangsu, China.
| |
Collapse
|
15
|
Zhu X, Ye F, Hao S, Yu Q, Wang Y, Lou W, Zhao K, Li H. MiR-1297 and MiR-26a-5p Inhibit Cell Progression of Keratinocytes in Cholesteatoma Depending on the Regulation of BMI1. BIOTECHNOL BIOPROC E 2022. [DOI: 10.1007/s12257-021-0178-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|
16
|
TWIST1-EP300 Expedites Gastric Cancer Cell Resistance to Apatinib by Activating the Expression of COL1A2. Anal Cell Pathol (Amst) 2022; 2022:5374262. [PMID: 35242497 PMCID: PMC8888114 DOI: 10.1155/2022/5374262] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 01/27/2022] [Indexed: 11/18/2022] Open
Abstract
The association between collagen type I alpha (COL1A) and chemoresistance has been verified in cancers. However, the specific role of COL1A2 in gastric cancer (GC) cell resistance to apatinib, a highly selective small-molecule inhibitor of vascular endothelial growth factor receptor 2, has not been investigated before. The purpose of this study was to explore the potential factors associated with COL1A2 regulation on GC cell apatinib resistance in vitro. With the aid of the Oncomine database and integrated bioinformatics methods, we identified COL1A2 overexpression in GC and its prognostic value. Mechanistically, the COL1A2 promoter has a distinct H3K27ac modification site and that E1A binding protein p300 (EP300) and twist family bHLH transcription factor 1 (TWIST1) can bind to the COL1A2 promoter, which in turn transcriptionally activated COL1A2 expression. In addition, overexpression of COL1A2 significantly promoted resistance to apatinib in GC cells, but knockdown of EP300 or TWIST1 remarkably inhibited COL1A2 expression and promoted sensitivity of GC cells to apatinib. Our findings demonstrated that the combination of EP300 and TWIST1 has a synergistically regulatory effect on COL1A2 expression, thus contributing to apatinib resistance in GC cells.
Collapse
|
17
|
Cao HJ, Zhou W, Xian XL, Sun SJ, Ding PJ, Tian CY, Tian FL, Jiang CH, Fu TT, Zhao S, Dai JY. A Mixture of Baicalein, Wogonin, and Oroxylin-A Inhibits EMT in the A549 Cell Line via the PI3K/AKT-TWIST1-Glycolysis Pathway. Front Pharmacol 2022; 12:821485. [PMID: 35222014 PMCID: PMC8864075 DOI: 10.3389/fphar.2021.821485] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 12/31/2021] [Indexed: 12/27/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is a worldwide disease with a high morbidity and mortality rate, which is most derived from its metastasis. Some studies show that the epithelial-mesenchymal transition (EMT) process promotes lung cancer cell migration and invasion, leading to NSCLC metastasis. Total flavonoid aglycones extract (TFAE) isolated from Scutellaria baicalensis was reported to inhibit tumor growth and induce apoptosis. In this study, we found that baicalein, wogonin, and oroxylin-A were the active compounds of TFAE. After reconstructing with these three compounds [baicalein (65.8%), wogonin (21.2%), and oroxylin-A (13.0%)], the reconstructed TFAE (reTFAE) inhibited the EMT process of A549 cells. Then, bioinformatic technology was employed to elucidate the potential pharmacodynamic mechanism network of reTFAE. We identified the relationship between reTFAE and PI3K/Akt signaling pathways, with TWIST1 as the key protein. LY294002, the inhibitor of the PI3K/Akt signaling pathway, and knock-down TWIST1 could significantly enhance the efficacy of reTFAE, with increasing expression of epithelial markers and decreasing expression of mesenchymal markers in A549 cells at the same time. Furthermore, stable isotope dimethyl-labeled proteomics technology was conducted to complement the follow-up mechanism that the EMT-inhibition process may be realized through the glycolysis pathway. In conclusion, we claim that TWIST1-targeted flavonoids could provide a new strategy to inhibit EMT progress for the treatment of NSCLC.
Collapse
Affiliation(s)
- Hui-Juan Cao
- Traditional Chinese Medicine College, North China University of Science and Technology, Tangshan, China
| | - Wei Zhou
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Xiao-Le Xian
- Traditional Chinese Medicine College, North China University of Science and Technology, Tangshan, China
| | - Shu-Jun Sun
- School of Biology and Food Engineering, Fuyang Normal University, Fuyang, China
| | - Pei-Jie Ding
- Traditional Chinese Medicine College, North China University of Science and Technology, Tangshan, China
| | - Chun-Yu Tian
- Traditional Chinese Medicine College, North China University of Science and Technology, Tangshan, China
| | - Fu-Ling Tian
- Traditional Chinese Medicine College, North China University of Science and Technology, Tangshan, China
| | - Chun-Hua Jiang
- Traditional Chinese Medicine College, North China University of Science and Technology, Tangshan, China
| | - Ting-Ting Fu
- Traditional Chinese Medicine College, North China University of Science and Technology, Tangshan, China
| | - Shu Zhao
- Traditional Chinese Medicine College, North China University of Science and Technology, Tangshan, China
| | - Jian-Ye Dai
- School of Pharmacy, Lanzhou University, Lanzhou, China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
- Collaborative Innovation Center for Northwestern Chinese Medicine, Lanzhou University, Lanzhou, China
| |
Collapse
|
18
|
Esparza-López J, Longoria O, De La Cruz-Escobar EN, Garibay-Díaz JC, León-Rodríguez E, De Jesús Ibarra-Sánchez M. Paclitaxel resistance is mediated by NF-κB on mesenchymal primary breast cancer cells. Oncol Lett 2022; 23:50. [PMID: 34992683 PMCID: PMC8721864 DOI: 10.3892/ol.2021.13168] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 11/09/2021] [Indexed: 12/16/2022] Open
Abstract
Paclitaxel has been used widely to treat breast cancer and other types of cancer. However, resistance is a major cause of failure for treatment and results in cancer progression. The present study investigated the association between paclitaxel resistance and the mesenchymal phenotype, using a model of primary breast cancer cells and employing four different cultures, two with an epithelial phenotype (MBCDF and MBCD17) and two with a mesenchymal phenotype (MBCDF-D5 and MBCD3). Epithelial-mesenchymal markers were evaluated by western blotting; MBCDF and MBCD17 cells expressed E-cadherin, SNAIL, Slug, and Twist, low levels of N-cadherin, but not vimentin. MBCDF-D5 and MBCD3 cells expressed N-cadherin, vimentin, and higher levels of SNAIL, and low levels of E-cadherin, Slug, and Twist. Cell viability was evaluated using a crystal violet assay after paclitaxel treatment; primary breast cancer cells with mesenchymal phenotype were resistant to paclitaxel compared with the epithelial primary breast cancer cells. Furthermore, using western blotting, it was revealed that mesenchymal cells had elevated levels of nuclear factor-κΒ (NF-κB) p65 and IκB kinase (IKK). Additionally, it was demonstrated that paclitaxel-induced degradation of the inhibitor of NF-κB, activation of NF-κB in a dose-dependent manner, and Bcl-2 and Bcl-xL upregulation. Finally, employing western blotting and crystal violet assays, the effects of the proteasome inhibitor ALLN were assessed. ALLN inhibited paclitaxel-induced NF-κB activation and restored the sensitivity to paclitaxel. Together, these data suggest that targeting the NF-κB/IKK axis might be a promising strategy to overcome paclitaxel resistance.
Collapse
Affiliation(s)
- José Esparza-López
- Biochemistry Unit, Salvador Zubirán National Institute of Health Sciences and Nutrition, Mexico City 14080, Mexico.,Research Support Network, National Autonomous University of Mexico-Salvador Zubirán National Institute of Health Sciences and Nutrition, Mexico City 14080, Mexico
| | - Ossian Longoria
- Hematology and Oncology Department, Salvador Zubirán National Institute of Health Sciences and Nutrition, Mexico City 14080, Mexico
| | | | - Julio Cesar Garibay-Díaz
- Hematology and Oncology Department, Salvador Zubirán National Institute of Health Sciences and Nutrition, Mexico City 14080, Mexico
| | - Eucario León-Rodríguez
- Hematology and Oncology Department, Salvador Zubirán National Institute of Health Sciences and Nutrition, Mexico City 14080, Mexico
| | | |
Collapse
|
19
|
Wu Y, Ma Z, Zhang Y, Zhang M, Zhang W, Zhang M, Shi X, Li W, Liu W. Cyclophilin A regulates A549 cells apoptosis via stabilizing Twist1 protein. J Cell Sci 2021; 135:273668. [PMID: 34881782 DOI: 10.1242/jcs.259018] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 11/23/2021] [Indexed: 11/20/2022] Open
Abstract
Cyclophilin A (CypA) is an essential member of the immunophilin family. As an intracellular target of immunosuppressive drug cyclosporin A (CsA) or a peptidyl-prolyl cis/trans isomerase (PPIase), it catalyzes the cis-trans isomerization of proline amidic peptide bonds, through which, it regulates a variety of biological processes, such as intracellular signaling, transcription, and apoptosis. In this study, we found that intracellular CypA enhanced Twist1 phosphorylation at Ser68 and inhibited apoptosis in A549 cells. Mechanistically, CypA could mediate the phosphorylation of Twist1 at Ser68 via p38 MAPK, which inhibited its ubiquitination-mediated degradation. In addition, CypA increased Twist-p65 interaction and nuclear accumulation, which regulated Twist1-dependent expression of CDH1 and CDH2. Our findings collectively indicated the role of CypA in Twist1-mediated A549 cells apoptosis through stabilizing Twist1 protein.
Collapse
Affiliation(s)
- Yaru Wu
- College of Life Sciences, Henan Agricultural University, Zhengzhou, China
| | - Zhenling Ma
- College of Life Sciences, Henan Agricultural University, Zhengzhou, China
| | - Yanyan Zhang
- College of Life Sciences, Henan Agricultural University, Zhengzhou, China
| | - Min Zhang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| | - Wenwen Zhang
- College of Life Sciences, Henan Agricultural University, Zhengzhou, China
| | - Menghao Zhang
- College of Life Sciences, Henan Agricultural University, Zhengzhou, China
| | - Xixi Shi
- College of Life Sciences, Henan Agricultural University, Zhengzhou, China
| | - Wenqing Li
- College of Life Sciences, Henan Agricultural University, Zhengzhou, China
| | - Wei Liu
- College of Life Sciences, Henan Agricultural University, Zhengzhou, China
| |
Collapse
|
20
|
Resveratrol Contrasts LPA-Induced Ovarian Cancer Cell Migration and Platinum Resistance by Rescuing Hedgehog-Mediated Autophagy. Cells 2021; 10:cells10113213. [PMID: 34831435 PMCID: PMC8625920 DOI: 10.3390/cells10113213] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/12/2021] [Accepted: 11/14/2021] [Indexed: 01/01/2023] Open
Abstract
Background: Ovarian cancer progression and invasiveness are promoted by a range of soluble factors released by cancer cells and stromal cells within the tumor microenvironment. Our previous studies demonstrated that resveratrol (RV), a nutraceutical and caloric restriction mimetic with tumor-suppressive properties, counteracts cancer cell motility induced by stromal IL-6 by upregulating autophagy. Lysophosphatidic acid (LPA), a bioactive phospholipid that shows elevated levels in the tumor microenvironment and the ascites of ovarian cancers, stimulates the growth and tissue invasion of cancer cells. Whether LPA elicits these effects by inhibiting autophagy and through which pathway and whether RV can counteract the same remain obscure. Aims: To investigate the molecular pathways involved in LPA-induced ovarian cancer malignancy, particularly focusing on the role of autophagy, and the ability of RV to counteract LPA activity. Results: LPA stimulated while RV inhibited ovarian cancer cell migration. Transcriptomic and bioinformatic analyses showed an opposite regulation by LPA and RV of genes linked to epithelial-to-mesenchymal transition (EMT) and autophagy with involvement of the PI3K-AKT, JAK-STAT and Hedgehog (Hh) pathways. LPA upregulated the Hh and EMT members GLI1, BMI-1, SNAIL-1 and TWIST1 and inhibited autophagy, while RV did the opposite. Similar to the inhibitors of the Hh pathway, RV inhibited LPA-induced cancer cell migration and 3D growth of ovarian cancer cells. BMI-1 silencing prevented LPA-induced EMT, restored autophagy and hampered cell migration, resembling the effects of RV. TCGA data analyses indicated that patients with low expression of Hh/EMT-related genes together with active autophagy flux tended to have a better prognosis and this correlates with a more effective response to platinum therapy. In in vitro 3D spheroids, LPA upregulated BMI-1, downregulated autophagy and inhibited platinum toxicity while RV and Hh inhibitors restored autophagy and favored BAX-mediated cell death in response to platinum. Conclusions: By inhibiting the Hh pathway and restoration of autophagy, RV counteracts LPA-induced malignancy, supporting its inclusion in the therapy of ovarian cancer for limiting metastasis and chemoresistance.
Collapse
|
21
|
Koyanagi S. Chrono-Pharmaceutical Approaches to Optimize Dosing Regimens Based on the Circadian Clock Machinery. Biol Pharm Bull 2021; 44:1577-1584. [PMID: 34719634 DOI: 10.1248/bpb.b21-00476] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Daily rhythmic variations in biological functions affect the efficacy and/or toxicity of drugs: a large number of drugs cannot be expected to exhibit the same potency at different administration times. The "circadian clock" is an endogenous timing system that broadly regulates metabolism, physiology and behavior. In mammals, this clock governs the oscillatory expression of the majority of genes with a period length of approximately 24 h. Genetic studies have revealed that molecular components of the circadian clock regulate the expression of genes responsible for the sensitivity to drugs and their disposition. The circadian control of pharmacodynamics and pharmacokinetics enables 'chrono-pharmaceutical' applications, namely drug administration at appropriate times of day to optimize the therapeutic index (efficacy vs. toxicity). On the other hand, a variety of pathological conditions also exhibit marked day-night changes in symptom intensity. Currently, novel therapeutic approaches are facilitated by the development of chemical compound targeted to key proteins that cause circadian exacerbation of disease events. This review presents an overview of the current understanding of the role of the circadian biological clock in regulating drug efficacy and disease conditions, and also describes the importance of identifying the difference in the circadian machinery between diurnal and nocturnal animals to select the most appropriate times of day to administer drugs in humans.
Collapse
|
22
|
Ogino T, Matsunaga N, Tanaka T, Tanihara T, Terajima H, Yoshitane H, Fukada Y, Tsuruta A, Koyanagi S, Ohdo S. Post-transcriptional repression of circadian component CLOCK regulates cancer-stemness in murine breast cancer cells. eLife 2021; 10:66155. [PMID: 33890571 PMCID: PMC8102063 DOI: 10.7554/elife.66155] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 04/22/2021] [Indexed: 12/18/2022] Open
Abstract
Disruption of the circadian clock machinery in cancer cells is implicated in tumor malignancy. Studies on cancer therapy reveal the presence of heterogeneous cells, including breast cancer stem-like cells (BCSCs), in breast tumors. BCSCs are often characterized by high aldehyde dehydrogenase (ALDH) activity, associated with the malignancy of cancers. In this study, we demonstrated the negative regulation of ALDH activity by the major circadian component CLOCK in murine breast cancer 4T1 cells. The expression of CLOCK was repressed in high-ALDH-activity 4T1, and enhancement of CLOCK expression abrogated their stemness properties, such as tumorigenicity and invasive potential. Furthermore, reduced expression of CLOCK in high-ALDH-activity 4T1 was post-transcriptionally regulated by microRNA: miR-182. Knockout of miR-182 restored the expression of CLOCK, resulted in preventing tumor growth. Our findings suggest that increased expression of CLOCK in BCSCs by targeting post-transcriptional regulation overcame stemness-related malignancy and may be a novel strategy for breast cancer treatments.
Collapse
Affiliation(s)
- Takashi Ogino
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Naoya Matsunaga
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan.,Department of Glocal Healthcare Science, Faculty of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Takahiro Tanaka
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Tomohito Tanihara
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Hideki Terajima
- Department of Biological Sciences, School of Science, The University of Tokyo, Tokyo, Japan
| | - Hikari Yoshitane
- Department of Biological Sciences, School of Science, The University of Tokyo, Tokyo, Japan
| | - Yoshitaka Fukada
- Department of Biological Sciences, School of Science, The University of Tokyo, Tokyo, Japan
| | - Akito Tsuruta
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Satoru Koyanagi
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan.,Department of Glocal Healthcare Science, Faculty of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Shigehiro Ohdo
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
23
|
Chen C, Mao X, Cheng C, Jiao Y, Zhou Y, Ren T, Wu Z, Lv Z, Sun X, Guo W. miR-135a Reduces Osteosarcoma Pulmonary Metastasis by Targeting Both BMI1 and KLF4. Front Oncol 2021; 11:620295. [PMID: 33828977 PMCID: PMC8019936 DOI: 10.3389/fonc.2021.620295] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 03/08/2021] [Indexed: 12/13/2022] Open
Abstract
Because of the modest response rate after surgery and chemotherapy, treatment of osteosarcoma (OS) remains challenging due to tumor recurrence and metastasis. miR-135a has been reported to act as an anticarcinogenic regulator of several cancers. However, its expression and function in osteosarcoma remain largely unknown. Here, we reported that abridged miR-135a expression in OS cells and tissues, and its expression is inversely correlated with the expression of BMI1 and KLF4, which are described as oncogenes in several cancers. Ectopic expression of miR-135a inhibited cell invasion and expression of BMI1 and KLF4 in OS cells. In vivo investigation confirmed that miR-135a acts as a tumor suppressor in OS to inhibit tumor growth and lung metastasis in xenograft nude mice. BMI1 and KLF4 were revealed to be direct targets of miR-135a, and miR-135a had a similar effect as the combination of si-BMI1 and si-KLF4 on inhibiting tumor progression and the expression of BMI1 and KLF4 in vivo. Altogether, our results demonstrate that the targeting of BMI1/KLF4 with miR-135a may provide an applicable strategy for exploring novel therapeutic approaches for OS.
Collapse
Affiliation(s)
- Chenglong Chen
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, China.,Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, China
| | - Xingjia Mao
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Caitong Cheng
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Yurui Jiao
- Endocrinology Research Center, Xiangya Hospital Central South University, Changsha, China
| | - Yi Zhou
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Tingting Ren
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, China.,Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, China
| | - Zhuangzhuang Wu
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Zhi Lv
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Xiaojuan Sun
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Wei Guo
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, China.,Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, China
| |
Collapse
|
24
|
Chen D, Cheng L, Cao H, Liu W. Role of microRNA-381 in bladder cancer growth and metastasis with the involvement of BMI1 and the Rho/ROCK axis. BMC Urol 2021; 21:5. [PMID: 33407350 PMCID: PMC7789167 DOI: 10.1186/s12894-020-00775-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 12/21/2020] [Indexed: 12/24/2022] Open
Abstract
Background Emerging evidence has noted the important participation of microRNAs (miRNAs) in several human diseases including cancer. This research was launched to probe the function of miR-381 in bladder cancer (BCa) progression. Methods Twenty-eight patients with primary BCa were included in this study. Cancer tissues and the adjacent normal tissues were obtained. Aberrantly expressed miRNAs in BCa tissues were analyzed using miRNA microarrays. miR-381 expression in the bladder and paired tumor tissues, and in BCa and normal cell lines was determined. The target relationship between miR-381 and BMI1 was predicted online and validated through a luciferase assay. Gain-of-functions of miR-381 and BMI1 were performed to identify their functions on BCa cell behaviors as well as tumor growth in vivo. The involvement of the Rho/ROCK signaling was identified. Results miR-381 was poor regulated in BCa tissues and cells (all p < 0.05). A higher miR-381 level indicated a better prognosis of patients with BCa. Artificial up-regulation of miR-381 inhibited proliferation, invasion, migration, resistance to apoptosis, and tumor formation ability of BCa T24 and RT4 cells (all p < 0.05). miR-381 was found to directly bind to BMI1 and was negatively correlated with BMI1 expression. Overexpression of BMI1 partially blocked the tumor suppressing roles of miR-381 in cell malignancy and tumor growth (all p < 0.05). In addition, miR-381 led to decreased RhoA phosphorylation and ROCK2 activation, which were also reversed by BMI1 (all p < 0.05). Artificial inhibition of the Rho/ROCK signaling blocked the functions of BMI1 in cell growth and metastasis (all p < 0.05). Conclusion The study evidenced that miR-381 may act as a beneficiary biomarker in BCa patients. Up-regulation of miR-381 suppresses BCa development both in vivo and in vitro through BMI1 down-regulation and the Rho/ROCK inactivation.
Collapse
Affiliation(s)
- Dayin Chen
- Department of Urology, The First Affiliated Hospital of Jiamusi University, No. 348, Dexiang Street, Jiamusi, 154002, Heilongjiang, People's Republic of China
| | - Liang Cheng
- Department of Urology, The First Affiliated Hospital of Jiamusi University, No. 348, Dexiang Street, Jiamusi, 154002, Heilongjiang, People's Republic of China
| | - Huifeng Cao
- Department of Urology, The First Affiliated Hospital of Jiamusi University, No. 348, Dexiang Street, Jiamusi, 154002, Heilongjiang, People's Republic of China.
| | - Wensi Liu
- Department of Urology, The First Affiliated Hospital of Jiamusi University, No. 348, Dexiang Street, Jiamusi, 154002, Heilongjiang, People's Republic of China
| |
Collapse
|
25
|
MicroRNA-361-5p Inhibits Tumorigenesis and the EMT of HCC by Targeting Twist1. BIOMED RESEARCH INTERNATIONAL 2020; 2020:8891876. [PMID: 33381597 PMCID: PMC7762665 DOI: 10.1155/2020/8891876] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 11/17/2020] [Accepted: 12/03/2020] [Indexed: 02/08/2023]
Abstract
MicroRNA-361-5p (miR-361-5p) is a tumor suppressor miRNA that is dysregulated in several types of human cancer. However, the functional significance of miR-361-5p in hepatocellular carcinoma (HCC) is unclear. This study explored the biological function of miR-361-5p in regulating the progression of HCC and the underlying molecular mechanism. RT-qPCR analysis showed that miR-361-5p was downregulated in HCC tissues and cell lines. Functional analysis revealed that miR-361-5p acted as a tumor suppressor, inhibiting cell proliferation, migration, and invasion in HCC cell lines. Bioinformatics analyses identified Twist1 as a direct target of miR-361-5p, which was validated by dual-luciferase reporter assays, RT-qPCR, and western blotting. Rescue experiments indicated that Twist1 may mediate the tumor-suppressive effect of miR-361-5p in HCC cells, and this was supported by the effect of miR-361-5p on inhibiting the epithelial-mesenchymal transition (EMT) by targeting Twist1. This study is the first to suggest that miR-361-5p inhibits tumorigenesis and EMT in HCC by targeting Twist1. These findings are valuable for the diagnosis and clinical management of HCC.
Collapse
|
26
|
Xue D, Zhou X, Qiu J. Emerging role of NRF2 in ROS-mediated tumor chemoresistance. Biomed Pharmacother 2020; 131:110676. [PMID: 32858502 DOI: 10.1016/j.biopha.2020.110676] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 08/16/2020] [Accepted: 08/20/2020] [Indexed: 12/24/2022] Open
Abstract
Chemoresistance is a central cause for the tumor management failure. Cancer cells disrupt the redox homeostasis through reactive oxygen species (ROS) regulatory mechanisms, leading to tumor progression and chemoresistance. The transcription factor nuclear factor erythroid 2-related factor 2 (NRF2) is a master regulator of neutralizing cellular ROS and restoring redox balance. Understanding the role of NRF2 in ROS-mediated chemoresistance can be helpful in the development of chemotherapy strategies with better efficiency. In this review, we sum up the roles of ROS in the development of chemoresistance to classical chemotherapy agents including cisplatin, 5-fluorouracil, gemcitabine, oxaliplatin, paclitaxel, and doxorubicin, and how to overcome ROS-mediated tumor chemoresistance by targeting NRF2. Finally, we propose that targeting NRF2 might be a promising strategy to resist ROS-driven chemoresistance and acquire better efficacy in cancer treatment.
Collapse
Affiliation(s)
- Danfeng Xue
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Xiongming Zhou
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Jiaxuan Qiu
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
27
|
Pourjafar M, Samadi P, Karami M, Najafi R. Assessment of clinicopathological and prognostic relevance of BMI-1 in patients with colorectal cancer: A meta-analysis. Biotechnol Appl Biochem 2020; 68:1313-1322. [PMID: 33086431 DOI: 10.1002/bab.2053] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 10/15/2020] [Indexed: 12/13/2022]
Abstract
B-cell-specific Moloney leukemia virus insertion site 1 (BMI-1) is one of the stemness markers. The prognostic and clinicopathological effects of BMI-1 expression in colorectal cancer (CRC) have been in dispute with different studies. Eligible studies were retrieved from international databases up to December 2019. Studies with a relationship between the clinicopathological and prognostic value of CRC patients with BMI-1 expression were selected. The correlations in the random-effect model were evaluated using the hazard ratios, odds ratio, and 95% confidence intervals (CIs). A total of nine studies comprising Asian cases (seven studies) and European cases (two studies) covering 1,294 samples of CRC were included for this meta-analysis. The analysis suggested that in Asian cases, increased expression of BMI-1 was associated with poor overall survival (OS) and death-free survival, whereas in European populations, high expression of BMI-1 was associated with better OS. Also, overexpression of BMI-1 in the Asian population was associated with the tumor size, distant metastasis, and patient's gender and age. Results suggested that high expression of BMI-1 can be involved in the progression and invasion of CRC, and so its inhibitor-based therapies could be used to prevent the progression of CRC.
Collapse
Affiliation(s)
- Mona Pourjafar
- Student Research Committee, Hamadan University of Medical Sciences, Hamadan, Iran.,Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Pouria Samadi
- Student Research Committee, Hamadan University of Medical Sciences, Hamadan, Iran.,Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Manoochehr Karami
- Research Center for Health Sciences, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Rezvan Najafi
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
28
|
Establishment of Acquired Cisplatin Resistance in Ovarian Cancer Cell Lines Characterized by Enriched Metastatic Properties with Increased Twist Expression. Int J Mol Sci 2020; 21:ijms21207613. [PMID: 33076245 PMCID: PMC7589258 DOI: 10.3390/ijms21207613] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 10/11/2020] [Accepted: 10/12/2020] [Indexed: 12/16/2022] Open
Abstract
Ovarian cancer (OC) is the most lethal of the gynecologic cancers, and platinum-based treatment is a part of the standard first-line chemotherapy regimen. However, rapid development of acquired cisplatin resistance remains the main cause of treatment failure, and the underlying mechanism of resistance in OC treatment remains poorly understood. Faced with this problem, our aim in this study was to generate cisplatin-resistant (CisR) OC cell models in vitro and investigate the role of epithelial–mesenchymal transition (EMT) transcription factor Twist on acquired cisplatin resistance in OC cell models. To achieve this aim, OC cell lines OV-90 and SKOV-3 were exposed to cisplatin using pulse dosing and stepwise dose escalation methods for a duration of eight months, and a total of four CisR sublines were generated, two for each cell line. The acquired cisplatin resistance was confirmed by determination of 50% inhibitory concentration (IC50) and clonogenic survival assay. Furthermore, the CisR cells were studied to assess their respective characteristics of metastasis, EMT phenotype, DNA repair and endoplasmic reticulum stress-mediated cell death. We found the IC50 of CisR cells to cisplatin was 3–5 times higher than parental cells. The expression of Twist and metastatic ability of CisR cells were significantly greater than those of sensitive cells. The CisR cells displayed an EMT phenotype with decreased epithelial cell marker E-cadherin and increased mesenchymal proteins N-cadherin and vimentin. We observed that CisR cells showed significantly higher expression of DNA repair proteins, X-ray repair cross-complementing protein 1 (XRCC1) and poly (ADP-ribose) polymerases 1 (PARP1), with significantly reduced endoplasmic reticulum (ER) stress-mediated cell death. Moreover, Twist knockdown reduced metastatic ability of CisR cells by suppressing EMT, DNA repair and inducing ER stress-induced cell death. In conclusion, we highlighted the utilization of an acquired cisplatin resistance model to identify the potential role of Twist as a therapeutic target to reverse acquired cisplatin resistance in OC.
Collapse
|
29
|
Zhang H, Lu B. The Roles of ceRNAs-Mediated Autophagy in Cancer Chemoresistance and Metastasis. Cancers (Basel) 2020; 12:cancers12102926. [PMID: 33050642 PMCID: PMC7600306 DOI: 10.3390/cancers12102926] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 09/28/2020] [Accepted: 10/07/2020] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Chemoresistance and metastasis are the main causes of treatment failure in cancers. Autophagy contribute to the survival and metastasis of cancer cells. Competing endogenous RNA (ceRNA), particularly long non-coding RNAs and circular RNA (circRNA), can bridge the interplay between autophagy and chemoresistance or metastasis in cancers via sponging miRNAs. This review aims to discuss on the function of ceRNA-mediated autophagy in the process of metastasis and chemoresistance in cancers. ceRNA network can sequester the targeted miRNA expression to indirectly upregulate the expression of autophagy-related genes, and thereof participate in autophagy-mediated chemoresistance and metastasis. Our clarification of the mechanism of autophagy regulation in metastasis and chemoresistance may greatly improve the efficacy of chemotherapy and survival in cancer patients. The combination of the tissue-specific miRNA delivery and selective autophagy inhibitors, such as hydroxychloroquine, is attractive to treat cancer patients in the future. Abstract Chemoresistance and metastasis are the main causes of treatment failure and unfavorable outcome in cancers. There is a pressing need to reveal their mechanisms and to discover novel therapy targets. Autophagy is composed of a cascade of steps controlled by different autophagy-related genes (ATGs). Accumulating evidence suggests that dysregulated autophagy contributes to chemoresistance and metastasis via competing endogenous RNA (ceRNA) networks including lncRNAs and circRNAs. ceRNAs sequester the targeted miRNA expression to indirectly upregulate ATGs expression, and thereof participate in autophagy-mediated chemoresistance and metastasis. Here, we attempt to summarize the roles of ceRNAs in cancer chemoresistance and metastasis through autophagy regulation.
Collapse
Affiliation(s)
- Huilin Zhang
- Department of Surgical Pathology, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou 310002, Zhejiang Province, China;
| | - Bingjian Lu
- Department of Surgical Pathology and Center for Uterine Cancer Diagnosis & Therapy Research of Zhejiang Province, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou 310002, Zhejiang Province, China
- Correspondence: ; Tel.: +86-571-89991702
| |
Collapse
|
30
|
Bmi1 Severs as a Potential Tumor-Initiating Cell Marker and Therapeutic Target in Esophageal Squamous Cell Carcinoma. Stem Cells Int 2020; 2020:8877577. [PMID: 32884573 PMCID: PMC7455816 DOI: 10.1155/2020/8877577] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/06/2020] [Accepted: 06/10/2020] [Indexed: 02/06/2023] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is a frequent malignant tumor with low 5-year overall survival. Targeting ESCC tumor-initiating cells (TICs) may provide a new research avenue to achieve better therapeutic effects of ESCC. However, the identity and characteristics of ESCC TICs remain poorly understood. Through genetic lineage tracing approach, we found that a group of Moloney murine leukemia virus insertion site 1- (Bmi1-) expressing cell populations present in the invasive front of the esophageal epithelium, providing a continuous flow of tumor cells for ESCC. Subsequently, we found that ablation of Bmi1+ cells from mice with ESCC led to inhibition of tumor growth. In addition, our results demonstrated that PTC-209, an inhibitor of Bmi1, was able to inhibit ESCC progression when combined with cisplatin. In summary, our data suggest that Bmi1+ cells serve as TICs in ESCC.
Collapse
|
31
|
The Interplay of Tumor Stroma and Translational Factors in Endometrial Cancer. Cancers (Basel) 2020; 12:cancers12082074. [PMID: 32726992 PMCID: PMC7463731 DOI: 10.3390/cancers12082074] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 07/19/2020] [Accepted: 07/22/2020] [Indexed: 12/21/2022] Open
Abstract
Endometrial cancer (EC) is a common gynecologic malignancy which continues to have a poor prognosis in advanced stages due to current therapeutic limitations. A significant mechanism of chemoresistance in EC has been shown to also be the enhancement of epithelial to mesenchymal transition (EMT) and the subsequent obtainment of stem cell-like characteristics of EC. Current evidence on EMT in EC however fails to explain the relationship leading to an EMT signaling enhancement. Our review therefore focuses on understanding eukaryotic translation initiation factors (eIFs) as key regulators of the translational process in enhancing EMT and subsequently impacting higher chemoresistance of EC. We identified pathways connected to the development of a microenvironment for EMT, inducers of the process specifically related to estrogen receptors as well as their interplay with eIFs. In the future, investigation elucidating the translational biology of EC in EMT may therefore focus on the signaling between protein kinase RNA-like ER kinase (PERK) and eIF2alpha as well as eIF3B.
Collapse
|
32
|
Wechman SL, Emdad L, Sarkar D, Das SK, Fisher PB. Vascular mimicry: Triggers, molecular interactions and in vivo models. Adv Cancer Res 2020; 148:27-67. [PMID: 32723566 DOI: 10.1016/bs.acr.2020.06.001] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Vascular mimicry is induced by a wide array of genes with functions related to cancer stemness, hypoxia, angiogenesis and autophagy. Vascular mimicry competent (VM-competent) cells that form de novo blood vessels are common in solid tumors facilitating tumor cell survival and metastasis. VM-competent cells display increased levels of vascular mimicry selecting for stem-like cells in an O2-gradient-dependent manner in deeply hypoxic tumor regions, while also aiding in maintaining tumor cell metabolism and stemness. Three of the principal drivers of vascular mimicry are EphA2, Nodal and HIF-1α, however, directly or indirectly many of these molecules affect VE-Cadherin (VE-Cad), which forms gap-junctions to bind angiogenic blood vessels together. During vascular mimicry, the endothelial-like functions of VM-competent cancer stem cells co-opt VE-Cad to bind cancer cells together to create cancer cell-derived blood conducting vessels. This process potentially compensates for the lack of access to blood and nutrient in avascular tumors, simultaneously providing nutrients and enhancing cancer invasion and metastasis. Current evidence also supports that vascular mimicry promotes cancer malignancy and metastasis due to the cooperation of oncogenic signaling molecules driving cancer stemness and autophagy. While a number of currently used cancer therapeutics are effective inhibitors of vascular mimicry, developing a new class of vascular mimicry specific inhibitors could allow for the treatment of angiogenesis-resistant tumors, inhibit cancer metastasis and improve patient survival. In this review, we describe the principal vascular mimicry pathways in addition to emphasizing the roles of hypoxia, autophagy and select proangiogenic oncogenes in this process.
Collapse
Affiliation(s)
- Stephen L Wechman
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - Luni Emdad
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - Devanand Sarkar
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - Swadesh K Das
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - Paul B Fisher
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States.
| |
Collapse
|
33
|
Wang SC, Sun HL, Hsu YH, Liu SH, Lii CK, Tsai CH, Liu KL, Huang CS, Li CC. α-Linolenic acid inhibits the migration of human triple-negative breast cancer cells by attenuating Twist1 expression and suppressing Twist1-mediated epithelial-mesenchymal transition. Biochem Pharmacol 2020; 180:114152. [PMID: 32679125 DOI: 10.1016/j.bcp.2020.114152] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 07/10/2020] [Accepted: 07/11/2020] [Indexed: 12/21/2022]
Abstract
α-Linolenic acid (ALA), an essential fatty acid, has anticancer activity in breast cancer, but the mechanism of its effects in triple-negative breast cancer (TNBC) remains unclear. We investigated the effect of ALA on Twist1, which is required to initiate epithelial-mesenchymal transition (EMT) and promotes tumor metastasis, and Twist1-mediated migration in MDA-MB231, MDA-MB468 and Hs578T cells. Twist1 protein was constitutively expressed in these TNBC cells, particularly MDA-MB-231 cells. Treatment with 100 μM ALA and Twist1 siRNA markedly decreased the Twist1 protein level and cell migration. Moreover, ALA transiently attenuated the nuclear accumulation of STAT3α as well as Twist1 mRNA expression. Treatment with ALA significantly attenuated the phosphorylation of JNK, ERK and Akt and decreased the phosphorylation of Twist1 at serine 68 in MDA-MB-231 cells. ALA accelerated Twist1 degradation in the presence of cycloheximide, whereas the ubiquitination and degradation of Twist1 by ALA was suppressed by MG-132. Pretreatment with ALA mimicked Twist1 siRNA, increased the protein expression of epithelial markers such as E-cadherin, and decreased the protein expression of mesenchymal markers including Twist1, Snail2, N-cadherin, vimentin, and fibronectin. Our findings suggest that ALA can be used not only to abolish EMT but also to suppress Twist1-mediated migration in TNBC cells.
Collapse
Affiliation(s)
- Shih-Chung Wang
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Changhua Christian Children's Hospital, Changhua, Taiwan
| | - Hai-Lun Sun
- Department of Pediatrics, Chung Shan Medical University Hospital, Taichung, Taiwan; School of Medicine, Chung Shan Medical University, Taichung, Taiwan; Immunology Research Center, Chung Shan Medical University, Taichung, Taiwan
| | - Yi-Hsuan Hsu
- Department of Nutrition, Chung Shan Medical University, Taichung, Taiwan
| | - Shu-Hui Liu
- Department of Nutrition, Chung Shan Medical University, Taichung, Taiwan
| | - Chong-Kuei Lii
- Department of Nutrition, China Medical University, Taichung, Taiwan; Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan
| | - Chia-Han Tsai
- Department of Nutrition, Chung Shan Medical University, Taichung, Taiwan
| | - Kai-Li Liu
- Department of Nutrition, Chung Shan Medical University, Taichung, Taiwan; Department of Nutrition, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chin-Shiu Huang
- Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan
| | - Chien-Chun Li
- Department of Nutrition, Chung Shan Medical University, Taichung, Taiwan; Department of Nutrition, Chung Shan Medical University Hospital, Taichung, Taiwan.
| |
Collapse
|
34
|
Liu Q, Li Q, Zhu S, Yi Y, Cao Q. B lymphoma Moloney murine leukemia virus insertion region 1: An oncogenic mediator in prostate cancer. Asian J Androl 2020; 21:224-232. [PMID: 29862993 PMCID: PMC6498728 DOI: 10.4103/aja.aja_38_18] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
B lymphoma Moloney murine leukemia virus insertion region 1 (BMI1), a core member of polycomb repressive complex 1 (PRC1), has been intensely investigated in the field of cancer epigenetics for decades. Widely known as a critical regulator in cellular physiology, BMI1 is essential in self-renewal and differentiation in different lineages of stem cells. BMI1 also plays a significant role in cancer etiology for its involvement in pathological progress such as epithelial–mesenchymal transition (EMT) and cancer stem cell maintenance, propagation, and differentiation. Importantly, overexpression of BMI1 is predictive for drug resistance, tumor recurrence, and eventual therapy failure of various cancer subtypes, which renders the pharmacological targeting at BMI1 as a novel and promising therapeutic approach. The study on prostate cancer, a prevalent hormone-related cancer among men, has promoted enormous research advancements in cancer genetics and epigenetics. This review summarizes the role of BMI1 as an oncogenic and epigenetic regulator in tumor initiation, progression, and relapse of prostate cancer.
Collapse
Affiliation(s)
- Qipeng Liu
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX 77030, USA.,Xiangya School of Medicine, Central South University, Changsha 410008, China
| | - Qiaqia Li
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX 77030, USA.,Xiangya School of Medicine, Central South University, Changsha 410008, China
| | - Sen Zhu
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Yang Yi
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX 77030, USA.,Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou 510080, China.,Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Qi Cao
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX 77030, USA.,Houston Methodist Cancer Center, Houston Methodist Research Institute, Houston, TX 77030, USA.,Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY 10065, USA
| |
Collapse
|
35
|
Anjitha R, Antony A, Shilpa O, Anupama KP, Mallikarjunaiah S, Gurushankara HP. Malathion induced cancer-linked gene expression in human lymphocytes. ENVIRONMENTAL RESEARCH 2020; 182:109131. [PMID: 32069766 DOI: 10.1016/j.envres.2020.109131] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 01/08/2020] [Accepted: 01/09/2020] [Indexed: 06/10/2023]
Abstract
BACKGROUND Malathion is the most widely used organophosphate pesticide in agriculture. Increasing cancer incidence in agricultural workers and their children links to the exposure of malathion. Identification of genes involved in the process of carcinogenesis is essential for exploring the role of malathion. The alteration in gene expression by malathion in human lymphocytes has not been explored yet, although hematological malignancies are rampant in humans. OBJECTIVE This study investigates the malathion induced expression of cancer associated genes in human lymphocytes. METHODS Human lymphocyte viability and colony-forming ability were analyzed in malathion treated and control groups. Gene expression profile in control and malathion treated human lymphocytes were performed using a microarray platform. The genes which have significant functions and those involved in different pathways were analyzed using the DAVID database. Differential gene expression upon malathion exposure was validated by quantitative real-time (qRT)-PCR. RESULTS Malathion caused a concentration-dependent reduction in human lymphocyte viability. At low concentration (50 μg/mL) of malathion treatment, human lymphocytes were viable indicating that low concentration of malathion is not cytotoxic and induces the colony formation. Total of 659 genes (15%) were up regulated and 3729 genes (85%) were down regulated in malathion treated human lymphocytes. About 57 cancer associated genes related to the growth and differentiation of B and T cells, immunoglobulin production, haematopoiesis, tumor suppression, oncogenes and signal transduction pathways like MAPK and RAS were induced by malathion. CONCLUSION This study evidences the carcinogenic nature of malathion. Low concentration of this pesticide is not cytotoxic and induces differentially regulated genes in human lymphocytes, which are involved in the initiation, progression, and pathogenesis of cancer.
Collapse
Affiliation(s)
- Ramakrishnan Anjitha
- Department of Zoology, School of Biological Sciences, Central University of Kerala, Periya, 671 320, Kasaragod, Kerala, India
| | - Anet Antony
- Department of Zoology, School of Biological Sciences, Central University of Kerala, Periya, 671 320, Kasaragod, Kerala, India
| | - Olakkaran Shilpa
- Department of Zoology, School of Biological Sciences, Central University of Kerala, Periya, 671 320, Kasaragod, Kerala, India
| | - Kizhakke P Anupama
- Department of Zoology, School of Biological Sciences, Central University of Kerala, Periya, 671 320, Kasaragod, Kerala, India
| | - Shanthala Mallikarjunaiah
- Center for Applied Genetics, Department of Studies in Zoology, Bangalore University, Jnanabharathi, Bengaluru, 560 056, Karnataka, India
| | - Hunasanahally P Gurushankara
- Department of Zoology, School of Biological Sciences, Central University of Kerala, Periya, 671 320, Kasaragod, Kerala, India.
| |
Collapse
|
36
|
Jiang N, Niu G, Pan YH, Pan W, Zhang MF, Zhang CZ, Shen H. CBX4 transcriptionally suppresses KLF6 via interaction with HDAC1 to exert oncogenic activities in clear cell renal cell carcinoma. EBioMedicine 2020; 53:102692. [PMID: 32113161 PMCID: PMC7044754 DOI: 10.1016/j.ebiom.2020.102692] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 02/12/2020] [Accepted: 02/12/2020] [Indexed: 12/19/2022] Open
Abstract
Background Dysregulation of polycomb chromobox (CBX) proteins that mediate epigenetic gene silencing contributes to the progression of human cancers. Yet their roles in clear cell renal cell carcinoma (ccRCC) remain to be explored. Methods The expression of CBX4 and its clinical significance were determined by qRT-PCR, western blot, immunohistochemistry and statistical analyses. The biological function of CBX4 in ccRCC tumor growth and metastasis and the underlying mechanism were investigated using in vitro and in vivo models. Findings CBX4 exerts oncogenic activities in ccRCC via interaction with HDAC1 to transcriptionally suppress tumor suppressor KLF6. CBX4 expression is increased in ccRCC and correlated with poor prognosis in two independent cohorts containing 840 patients. High CBX4 expression is significantly associated with Fuhrman grade and tumor lymph node invasion. CBX4 overexpression promotes tumor growth and metastasis, whereas CBX4 knockdown results in the opposite phenotypes. Mechanistically, CBX4 downregulates KLF6 via repressing the transcriptional activity of its promoter. Further studies show that CBX4 physically binds to HDAC1 to maintain its localization on the KLF6 promoter. Ectopic expression of KLF6 or disruption of CBX4-HDAC1 interaction attenuates CBX4-mediated cell growth and migration. Furthermore, CBX4 depletion markedly enhances the histone deacetylase inhibitor (HDACi)-induced cell apoptosis and suppression of tumor growth. Interpretation Our data suggest CBX4 as an oncogene with prognostic potential in ccRCC. The newly identified CBX4/HDAC1/KLF6 axis may represent a potential therapeutic target for the clinical intervention of ccRCC.
Collapse
Affiliation(s)
- Nan Jiang
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Gang Niu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Ying-Hua Pan
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510600, China
| | - Wenwei Pan
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Mei-Fang Zhang
- Department of Pathology, Sun Yat-sen University Cancer Center, China; State Key Laboratory of Oncology in South China, China; Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Chris Zhiyi Zhang
- Department of Pathology, Sun Yat-sen University Cancer Center, China; State Key Laboratory of Oncology in South China, China; Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China; Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes and MOE Key Laboratory of Tumor Molecular Biology, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China.
| | - Huimin Shen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China.
| |
Collapse
|
37
|
Bmi-1 Immunohistochemical Expression in Endometrial Carcinoma is Correlated with Prognostic Activity. ACTA ACUST UNITED AC 2020; 56:medicina56020072. [PMID: 32059385 PMCID: PMC7074093 DOI: 10.3390/medicina56020072] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 02/03/2020] [Accepted: 02/10/2020] [Indexed: 12/14/2022]
Abstract
Background and objectives: B-lymphoma Mo-MLV insertion region 1 (Bmi-1) is a stem cell factor that is overexpressed in various human cancer tissues. It has been implicated in cancer cell proliferation, cell invasion, distant metastasis, and chemosensitivity, and is associated with patient survival. Several reports have also identified Bmi-1 protein overexpression in endometrial carcinoma; however, the relationship between Bmi-1 expression and its significance as a clinicopathological parameter is still insufficiently understood. Accordingly, the present study aimed to clarify whether immunohistochemical staining for Bmi-1 in human endometrial carcinoma and normal endometrial tissues can be used as a prognostic and cell proliferation marker. Materials and Methods: Bmi-1 expression was assessed in endometrioid carcinoma (grade 1–3) and normal endometrial tissues (in the proliferative and secretory phases) by immunohistochemistry; protein expression was evaluated using the nuclear labeling index (%) in the hot spot. Furthermore, we examined other independent prognostic and proliferation markers, including the protein levels of Ki-67, p53, and cyclin A utilizing semi-serial sections of endometrial carcinoma tissues. Results: The expression of the Bmi-1 protein was significantly higher in all grades of endometrial carcinoma than in the secretory phase of normal tissues. Moreover, Bmi-1 levels tended to be higher in G2 and G3 tissues than in G1 tissue, without reaching significance. Bmi-1 expression showed no notable differences among International Federation of Gynecology and Obstetrics (FIGO) stages in endometrial carcinoma. Furthermore, we observed a significant positive relationship between Bmi-1 and Ki-67, cyclin A, or p53 by Spearman’s rank correlation test, implying that high Bmi-1 expression can be an independent prognostic marker in endometrial carcinoma. Conclusions: Our study suggests that Bmi-1 levels in endometrial carcinoma tissues may be useful as a reliable proliferation and prognostic biomarker. Recently, the promise of anti-Bmi-1 strategies for the treatment of endometrial carcinoma has been detected. Our results provide fundamental data regarding this anti-Bmi-1 strategy.
Collapse
|
38
|
Liu T, Zhao X, Zheng X, Zheng Y, Dong X, Zhao N, Liao S, Sun B. The EMT transcription factor, Twist1, as a novel therapeutic target for pulmonary sarcomatoid carcinomas. Int J Oncol 2020; 56:750-760. [PMID: 32124963 PMCID: PMC7010216 DOI: 10.3892/ijo.2020.4972] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 12/12/2019] [Indexed: 12/25/2022] Open
Abstract
Pulmonary sarcomatoid carcinomas (PSCs) are a rare subtype of non‑small‑cell lung cancer and are typically biphasic neoplasms. No effective treatment for PSCs is currently available in clinical practice. The expression of the epithelial‑mesenchymal transition (EMT) transcription factors, Twist1, Slug and Snail, as well as the EMT phenotype and vasculogenic mimicry (VM) were analysed in 41 PSC and 79 pulmonary squamous carcinoma (PSCC) samples. Compared with the PSCCs, the PSCs exhibited an EMT phenotype and VM, and they also exhibited an increased expression of the Twist1, Slug, Snail and VM markers. Twist1 expression was associated with metastasis and TNM stage. Twist1‑positive patients exhibited a poorer prognosis for overall survival (OS) than those with Twist1‑negative PSCs. Transforming growth factor β1 (TGFβ1) was used to induce an EMT transition in a PSCC cell line. SK‑MES‑1 cells treated with TGFβ1 exhibited an increased expression of Twist1. The EMT phenotype, VM and increased migratory and invasive abilities were induced following TGFβ1 treatment. Importantly, in cells treated with TGFβ1, the EMT phenotype was reversed, VM marker expression was decreased, and the migratory and invasive ability of the PSCC cell line was decreased following Twist1 knockdown. Collectively, this study provides a new perspective of Twist1 in the aggressiveness of PSCs. The identification of Twist1 as an independent marker of poor prognoses may lead to the development of novel strategies for improving the treatment of patients with PSC.
Collapse
Affiliation(s)
- Tieju Liu
- Department of Pathology, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Xiulan Zhao
- Department of Pathology, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Xu Zheng
- Department of Pathology, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Yanjun Zheng
- Department of Pathology, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Xueyi Dong
- Department of Pathology, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Nan Zhao
- Department of Pathology, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Shihan Liao
- Department of Pathology, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Baocun Sun
- Department of Pathology, Tianjin Medical University, Tianjin 300070, P.R. China
| |
Collapse
|
39
|
Zhu S, Zhao D, Li C, Li Q, Jiang W, Liu Q, Wang R, Fazli L, Li Y, Zhang L, Yi Y, Meng Q, Wang W, Wang G, Zhang M, Zu X, Zhao W, Deng T, Yu J, Dong X, Chen K, Cao Q. BMI1 is directly regulated by androgen receptor to promote castration-resistance in prostate cancer. Oncogene 2020; 39:17-29. [PMID: 31462713 PMCID: PMC7386438 DOI: 10.1038/s41388-019-0966-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 04/29/2019] [Accepted: 05/28/2019] [Indexed: 12/16/2022]
Abstract
B lymphoma Mo-MLV insertion region 1 (BMI1) has been reported to be an oncoprotein. BMI1 represses tumor suppressors to promote cell proliferation, epithelial-mesenchymal transition (EMT), and cancer progression. Although it is known that the expression of BMI1 is increased in many cancer types, the mechanism of BMI1 upregulation is not yet clear. We performed integrative analysis for 3 sets of prostate cancer (PCa) genomic data, and found that BMI1 and androgen receptor (AR) were positively correlated, suggesting that AR might regulate BMI1. Next, we showed that dihydrotestosterone (DHT) upregulated both mRNA and protein levels of BMI1 and that BMI1 was increased in castration-resistant prostate cancer (CRPC) from both human patients and a mouse xenograph model. We further identified an AR binding site in the promoter/enhancer region of BMI1, and confirmed BMI1 as the direct target of AR using gene-editing technology. We also demonstrated that high expression of BMI1 is critical for the development of castration-resistance. Our data also suggest that BMI1-specific inhibitors could be an effective treatment of CRPC.
Collapse
Affiliation(s)
- Sen Zhu
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Dongyu Zhao
- Center for Bioinformatics and Computational Biology, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, Cornell University, New York, NY, 10065, USA
| | - Chao Li
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
- Department of Urology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Qiaqia Li
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
- Xiangya School of Medicine, Central South University, Changsha, 410008, China
| | - Weihua Jiang
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Qipeng Liu
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Xiangya School of Medicine, Central South University, Changsha, 410008, China
| | - Rui Wang
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Ladan Fazli
- Vancouver Prostate Centre, Vancouver General Hospital, Vancouver, BC, V6H 3Z6, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, V6H 3Z6, Canada
| | - Yinan Li
- Vancouver Prostate Centre, Vancouver General Hospital, Vancouver, BC, V6H 3Z6, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, V6H 3Z6, Canada
| | - Lili Zhang
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Yang Yi
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Qingshu Meng
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Wanyi Wang
- Center for Research Design & Analysis, Office of Research and Sponsored Programs, Texas Woman's University, Houston, TX, 77030, USA
| | - Guangyu Wang
- Center for Bioinformatics and Computational Biology, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, Cornell University, New York, NY, 10065, USA
| | - Min Zhang
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, Cornell University, New York, NY, 10065, USA
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA
| | - Xiongbing Zu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Wei Zhao
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Tuo Deng
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital and Key Laboratory of Diabetes Immunology, Central South University, Ministry of Education, Changsha, 410011, China
| | - Jindan Yu
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Xuesen Dong
- Vancouver Prostate Centre, Vancouver General Hospital, Vancouver, BC, V6H 3Z6, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, V6H 3Z6, Canada
| | - Kaifu Chen
- Center for Bioinformatics and Computational Biology, Houston Methodist Research Institute, Houston, TX, 77030, USA.
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, Cornell University, New York, NY, 10065, USA.
| | - Qi Cao
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, 77030, USA.
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA.
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA.
- Houston Methodist Cancer Center, Houston Methodist Research Institute, Houston, TX, 77030, USA.
| |
Collapse
|
40
|
Flores-Ramírez I, Baranda-Avila N, Langley E. Breast Cancer Stem Cells and Sex Steroid Hormones. Curr Stem Cell Res Ther 2019; 14:398-404. [PMID: 30095060 DOI: 10.2174/1574888x13666180810121415] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Revised: 07/13/2018] [Accepted: 07/17/2018] [Indexed: 12/14/2022]
Abstract
Breast cancer stem cells (BCSCs) are a small population of tumor-initiating cells that express stem cell-associated markers. In recent years, their properties and mechanisms of regulation have become the focus of intense research due to their intrinsic resistance to conventional cancer therapies. This review describes breast cancer stem cell origin, signaling pathways involved in self-renewal, such as Wnt, Notch and Hedgehog, biomarkers linked to stemness, and the role of sex steroid hormones in BCSC regulation.
Collapse
Affiliation(s)
- Iván Flores-Ramírez
- Programa de Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México, CDMX, México.,Departamento de Investigacion Basica, Instituto Nacional de Cancerologia, Av. San Fernando No. 22, Col. Seccion XVI, Tlalpan 14080, CDMX, Mexico
| | - Noemi Baranda-Avila
- Departamento de Investigacion Basica, Instituto Nacional de Cancerologia, Av. San Fernando No. 22, Col. Seccion XVI, Tlalpan 14080, CDMX, Mexico
| | - Elizabeth Langley
- Departamento de Investigacion Basica, Instituto Nacional de Cancerologia, Av. San Fernando No. 22, Col. Seccion XVI, Tlalpan 14080, CDMX, Mexico
| |
Collapse
|
41
|
Jen WP, Chen HM, Lin YS, Chern Y, Lee YC. Twist1 Plays an Anti-apoptotic Role in Mutant Huntingtin Expression Striatal Progenitor Cells. Mol Neurobiol 2019; 57:1688-1703. [PMID: 31813126 DOI: 10.1007/s12035-019-01836-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Accepted: 11/12/2019] [Indexed: 10/25/2022]
Abstract
The Twist basic helix-loop-helix transcription factor 1 (Twist1) has been implicated in embryogenesis and carcinogenesis, due to its effects on cell proliferation and anti-apoptosis signaling. Interestingly, a connection between Twist1 and neurotoxicity was recently made in mutant huntingtin (mHtt)-expressing primary cortical neurons; however, the role of Twist1 in Huntington's disease (HD)-affected striatal neurons remains undescribed. In this study, we evaluated the expression and function of Twist1 in the R6/2 HD mouse model, which expresses the polyQ-expanded N-terminal portion of human HTT protein, and a pair of striatal progenitor cell lines (STHdhQ109 and STHdhQ7), which express polyQ-expanded or non-expanded full-length mouse Htt. We further probed upstream signaling events and Twist1 anti-apoptotic function in the striatal progenitor cell lines. Twist1 was increased in mHtt-expressing striatal progenitor cells (STHdhQ109) and was correlated with disease progression in striatum and cortex brain regions of R6/2 mice. In the cell model, downregulation of Twist1 induced death of STHdhQ109 cells but had no effect on wild-type striatal progenitor cells (STHdhQ7). Twist1 knockdown stimulated caspase-3 activation and apoptosis. Furthermore, we found that signal transducer and activator of transcription 3 (STAT3) were increased in HD striatal progenitor cells and acted as an upstream regulator of Twist1. As such, inhibition of STAT3 induced apoptosis in HD striatal progenitor cells. Our results suggest that mHtt upregulates STAT3 to induce Twist1 expression. Upregulated Twist1 inhibits apoptosis, which may protect striatal cells from death during disease progression. Thus, we propose that Twist1 might play a protective role against striatal degeneration in HD.
Collapse
Affiliation(s)
- Wei-Ping Jen
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, 11490, Taiwan.,Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, 11529, Taiwan.,Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529, Taiwan
| | - Hui-Mei Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529, Taiwan
| | - Yow-Sien Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529, Taiwan
| | - Yijuang Chern
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, 11490, Taiwan.,Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529, Taiwan
| | - Yi-Ching Lee
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, 11490, Taiwan. .,Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, 11529, Taiwan.
| |
Collapse
|
42
|
Aravindan N, Jain D, Somasundaram DB, Herman TS, Aravindan S. Cancer stem cells in neuroblastoma therapy resistance. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2019; 2:948-967. [PMID: 31867574 PMCID: PMC6924637 DOI: 10.20517/cdr.2019.72] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Neuroblastoma (NB) is the most common cancer of infancy and accounts for nearly one tenth of pediatric cancer deaths. This mortality rate has been attributed to the > 50% frequency of relapse despite intensive, multimodal clinical therapy in patients with progressive NB. Given the disease’s heterogeneity and developed resistance, attaining a cure after relapse of progressive NB is highly challenging. A rapid decrease in the timeline between successive recurrences is likely due to the ongoing acquisition of genetic rearrangements in undifferentiated NB-cancer stem cells (CSCs). In this review, we present the current understanding of NB-CSCs, their intrinsic role in tumorigenesis, their function in disease progression, and their influence on acquired therapy resistance and tumor evolution. In particular, this review focus on the intrinsic involvement of stem cells and signaling in the genesis of NB, the function of pre-existing CSCs in NB progression and therapy response, the formation and influence of induced CSCs (iCSCs) in drug resistance and tumor evolution, and the development of a CSC-targeted therapeutic approach.
Collapse
Affiliation(s)
- Natarajan Aravindan
- Department of Radiation Oncology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.,Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.,Department of Anesthesiology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Drishti Jain
- Department of Radiation Oncology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Dinesh Babu Somasundaram
- Department of Radiation Oncology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Terence S Herman
- Department of Radiation Oncology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.,Stephenson Cancer Center, Oklahoma City, OK 73104, USA
| | | |
Collapse
|
43
|
Jin K, Wang S, Zhang Y, Xia M, Mo Y, Li X, Li G, Zeng Z, Xiong W, He Y. Long non-coding RNA PVT1 interacts with MYC and its downstream molecules to synergistically promote tumorigenesis. Cell Mol Life Sci 2019; 76:4275-4289. [PMID: 31309249 PMCID: PMC6803569 DOI: 10.1007/s00018-019-03222-1] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 06/22/2019] [Accepted: 07/05/2019] [Indexed: 02/06/2023]
Abstract
Numerous studies have shown that non-coding RNAs play crucial roles in the development and progression of various tumor cells. Plasmacytoma variant translocation 1 (PVT1) mainly encodes a long non-coding RNA (lncRNA) and is located on chromosome 8q24.21, which constitutes a fragile site for genetic aberrations. PVT1 is well-known for its interaction with its neighbor MYC, which is a qualified oncogene that plays a vital role in tumorigenesis. In the past several decades, increasing attention has been paid to the interaction mechanism between PVT1 and MYC, which will benefit the clinical treatment and prognosis of patients. In this review, we summarize the coamplification of PVT1 and MYC in cancer, the positive feedback mechanism, and the latest promoter competition mechanism of PVT1 and MYC, as well as how PVT1 participates in the downstream signaling pathway of c-Myc by regulating key molecules. We also briefly describe the treatment prospects and research directions of PVT1 and MYC.
Collapse
Affiliation(s)
- Ke Jin
- NHC Key Laboratory of Carcinogenesis (Central South University) and Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shufei Wang
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Yazhuo Zhang
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Mengfang Xia
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Yongzhen Mo
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Xiaoling Li
- NHC Key Laboratory of Carcinogenesis (Central South University) and Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Guiyuan Li
- NHC Key Laboratory of Carcinogenesis (Central South University) and Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhaoyang Zeng
- NHC Key Laboratory of Carcinogenesis (Central South University) and Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wei Xiong
- NHC Key Laboratory of Carcinogenesis (Central South University) and Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Yi He
- NHC Key Laboratory of Carcinogenesis (Central South University) and Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.
| |
Collapse
|
44
|
Xiao Q, Zhao XY, Jiang RC, Chen XH, Zhu X, Chen KF, Chen SY, Zhang XL, Qin Y, Liu YH, Luo JD. Increased expression of Sonic hedgehog restores diabetic endothelial progenitor cells and improves cardiac repair after acute myocardial infarction in diabetic mice. Int J Mol Med 2019; 44:1091-1105. [PMID: 31524224 PMCID: PMC6657988 DOI: 10.3892/ijmm.2019.4277] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Accepted: 07/03/2019] [Indexed: 12/19/2022] Open
Abstract
Damaged endothelial progenitor cells (EPCs) are associated with poor prognosis in diabetic myocardial infarction (DMI). Our previous studies revealed that an impaired Sonic hedgehog (Shh) pathway contributes to insufficient function in diabetic EPCs; however, the roles of the Shh pathway in diabetic EPC apoptosis under basal and hypoxic/ischemic conditions remain unknown. Therefore, the present study investigated whether Shh revitalized diabetic EPCs and consequently improved the deteriorative status of DMI. For this purpose, streptozotocin injection was used in male C57/BL6 mice to induce type-1 diabetes, and diabetic EPCs were isolated from the bone marrow. Apoptosis, cell function, and protein expression were investigated in EPCs in vitro. Mouse hearts were injected with adenovirus Shh-modified diabetic EPCs (DM-EPCShh) or control DM-EPCNull immediately after coronary artery ligation in vivo. Cardiac function, capillary numbers, fibrosis, and cell apoptosis were then detected. First, the in vitro results demonstrated that the apoptosis of diabetic EPCs was reduced following treatment with Shh protein for 24 h, under normal or hypoxic conditions. BMI1 proto-oncogene (Bmi1), an antiapoptotic protein found in several cells, was reduced in diabetic EPCs under normal or hypoxic conditions, but was upregulated after Shh protein stimulation. When Bmi1-siRNA was administered, the antiapoptotic effect of Shh protein was significantly reversed. In addition, p53, a Bmi1-targeted gene, was demonstrated to mediate the antiapoptotic effect of the Shh/Bmi1 pathway in diabetic EPCs. The Shh/Bmi1/p53 axis also enhanced the diabetic EPC function. In vivo, Shh-modified diabetic EPCs exhibited increased EPC retention and decreased apoptosis at 3 days post-DMI. At 14 days post-DMI, these cells presented enhanced capillary density, reduced myocardial fibrosis and improved cardiac function. In conclusion, the present results demonstrated that the Shh pathway restored diabetic EPCs through the Shh/Bmi1/p53 axis, suppressed myocardial apoptosis and improved myocardial angiogenesis, thus reducing cardiac fibrosis and finally restoring myocardial repair and cardiac function in DMI. Thus, the Shh pathway may serve as a potential target for autologous cell therapy in diabetic myocardial ischemia.
Collapse
Affiliation(s)
- Qing Xiao
- Key Laboratory of Molecular Target and Clinical Pharmacology, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 511436, P.R. China
| | - Xiao-Ya Zhao
- Key Laboratory of Molecular Target and Clinical Pharmacology, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 511436, P.R. China
| | - Ru-Chao Jiang
- Department of Pharmacology, Guangzhou Medical University, Guangzhou, Guangdong 511436, P.R. China
| | - Xiu-Hui Chen
- Department of Pharmacology, Guangzhou Medical University, Guangzhou, Guangdong 511436, P.R. China
| | - Xiang Zhu
- Department of Pharmacology, Guangzhou Medical University, Guangzhou, Guangdong 511436, P.R. China
| | - Kai-Feng Chen
- Department of Pharmacology, Guangzhou Medical University, Guangzhou, Guangdong 511436, P.R. China
| | - Sheng-Ying Chen
- Department of Pharmacology, Guangzhou Medical University, Guangzhou, Guangdong 511436, P.R. China
| | - Xiao-Ling Zhang
- Maternal and Children Hospital of Guangdong Province, Guangzhou, Guangdong 510260, P.R. China
| | - Yuan Qin
- Department of Pharmacology, Guangzhou Medical University, Guangzhou, Guangdong 511436, P.R. China
| | - Ying-Hua Liu
- Department of Pharmacology, Guangzhou Medical University, Guangzhou, Guangdong 511436, P.R. China
| | - Jian-Dong Luo
- Department of Pharmacology, Guangzhou Medical University, Guangzhou, Guangdong 511436, P.R. China
| |
Collapse
|
45
|
He J, Lee HJ, Saha S, Ruan D, Guo H, Chan CH. Inhibition of USP2 eliminates cancer stem cells and enhances TNBC responsiveness to chemotherapy. Cell Death Dis 2019; 10:285. [PMID: 30918246 PMCID: PMC6437220 DOI: 10.1038/s41419-019-1512-6] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 02/28/2019] [Accepted: 03/08/2019] [Indexed: 01/17/2023]
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer that harbors enriched cancer stem cell (CSC) populations in tumors. Conventional chemotherapy is a standard treatment for TNBC, but it spares the CSC populations, which cause tumor recurrence and progression. Therefore, identification of the core molecular pathway that controls CSC activity and expansion is essential for developing effective therapeutics for TNBC. In this study, we identify that USP2 deubiquitinating enzyme is upregulated in CSCs and is a novel regulator of CSCs. Genetic and pharmacological targeting of USP2 substantially inhibits the self-renewal, expansion and chemoresistance of CSCs. We show that USP2 maintains the CSC population by activating self-renewing factor Bmi1 and epithelial-mesenchymal transition through Twist upregulation. Mechanistically, USP2 promotes Twist stabilization by removing β-TrCP-mediated ubiquitination of Twist. Animal studies indicate that pharmacological inhibition of USP2 suppresses tumor progression and sensitizes tumor responses to chemotherapy in TNBC. Furthermore, the histological analyses reveal a positive correlation between USP2 upregulation and lymph node metastasis. Our findings together demonstrate a previously unrecognized role of USP2 in mediating Twist activation and CSC enrichment, suggesting that targeting USP2 is a novel therapeutic strategy to tackle TNBC.
Collapse
Affiliation(s)
- Jiabei He
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Hong-Jen Lee
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY, 11794, USA.,Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Suchandrima Saha
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY, 11794, USA.,Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Diane Ruan
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Hua Guo
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Chia-Hsin Chan
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY, 11794, USA. .,Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY, 11794, USA.
| |
Collapse
|
46
|
Wang J, Ji H, Zhu Q, Yu X, Du J, Jiang Z. Co-inhibition of BMI1 and Mel18 enhances chemosensitivity of esophageal squamous cell carcinoma in vitro and in vivo. Oncol Lett 2019; 17:5012-5022. [PMID: 31186712 PMCID: PMC6507449 DOI: 10.3892/ol.2019.10160] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 02/20/2019] [Indexed: 12/18/2022] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) accounts for almost 90% of esophageal cancer cases and is the sixth most common cause of cancer-associated mortality worldwide. Cisplatin is the standard therapeutic reagent for ESCC; however, chemoresistance frequently occurs after a few weeks, which leads to ESCC recurrence. Aberrant expression of B lymphoma Mo-MLV insertion region 1 homolog (BMI1) has been reported to activate multiple growth-regulatory pathways, induce antiapoptotic abilities in numerous types of cancer cells and promote chemoresistance. However, to the best of our knowledge, the role of BMI1 in cisplatin-resistant ESCC, and the interaction between BMI1 and its homologue melanoma nuclear protein 18 (Mel18) remain unknown. The present study identified that knockdown of BMI1 promoted cytotoxic effects of cisplatin, and co-inhibition of Mel18 and BMI1 enhanced cisplatin-induced apoptosis and cytotoxicity. Inhibition of BMI1 and Mel18 also suppressed the expression of c-Myc. Furthermore, this combined inhibition sensitized esophageal xenograft tumors to cisplatin to a greater extent compared with BMI1 inhibition alone. In summary, the current study demonstrated that inhibition of BMI1 and Mel18 could increase the sensitivity of esophageal cancer cells to cisplatin via inhibition of c-Myc. Therefore, combined targeting of BMI1 and Mel18 may serve as a promising therapeutic strategy for sensitizing ESCC to chemotherapy.
Collapse
Affiliation(s)
- Jiansong Wang
- Department of Thoracic Surgery, Weifang Medical University, Weifang, Shandong 261031, P.R. China
| | - Huaijun Ji
- Department of Thoracic Surgery, Weihai Municipal Hospital, Weihai, Shandong 264200, P.R. China
| | - Qiang Zhu
- Department of Thoracic Surgery, Qianfoshan Hospital Affiliated to Shandong University, Jinan, Shandong 250014, P.R. China
| | - Xinshuang Yu
- Department of Radiation Oncology, Qianfoshan Hospital Affiliated to Shandong University, Jinan, Shandong 250014, P.R. China
| | - Juan Du
- Central Laboratory, Qianfoshan Hospital Affiliated to Shandong University, Jinan, Shandong 250014, P.R. China
| | - Zhongmin Jiang
- Department of Thoracic Surgery, Qianfoshan Hospital Affiliated to Shandong University, Jinan, Shandong 250014, P.R. China
| |
Collapse
|
47
|
Xie ZC, Huang JC, Zhang LJ, Gan BL, Wen DY, Chen G, Li SH, Yan HB. Exploration of the diagnostic value and molecular mechanism of miR‑1 in prostate cancer: A study based on meta‑analyses and bioinformatics. Mol Med Rep 2018; 18:5630-5646. [PMID: 30365107 PMCID: PMC6236292 DOI: 10.3892/mmr.2018.9598] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Accepted: 09/24/2018] [Indexed: 02/07/2023] Open
Abstract
Prostate cancer (PCa) remains a principal issue to be addressed in male cancer-associated mortality. Therefore, the present study aimed to examine the clinical value and associated molecular mechanism of microRNA (miR)-1 in PCa. A meta-analysis was conducted to evaluate the diagnosis of miR-1 in PCa via Gene Expression Omnibus and ArrayExpress datasets, The Cancer Genome Atlas miR-1 expression data and published literature. It was identified that expression of miR-1 was significantly downregulated in PCa. Decreased miR-1 expression possessed moderate diagnostic value, with area under the curve, sensitivity, specificity and odds ratio values at 0.73, 0.77, 0.57 and 4.60, respectively. Using bioinformatics methods, it was revealed that a number of pathways, including the ‘androgen receptor signaling pathway’, ‘androgen receptor activity’, ‘transcription factor binding’ and ‘protein processing in the endoplasmic reticulum’, were important in PCa. A total of seven hub genes, including phosphoribosylaminoimidazole carboxylase and phosphoribosylaminoimidazolesuccin ocarboxamide synthase (PAICS), cadherin 1 (CDH1), SRC proto-oncogene, non-receptor tyrosine kinase, twist family bHLH transcription factor 1 (TWIST1), ZW10 interacting kinetochore protein (ZWINT), PCNA clamp associated factor (KIAA0101) and androgen receptor, among which, five (PAICS, CDH1, TWIST1, ZWINT and KIAA0101) were significantly upregulated and negatively correlated with miR-1, were identified as key miR-1 target genes in PCa. Additionally, it was investigated whether miR-1 and its hub genes were associated with clinical features, including age, tumor status, residual tumor, lymph node metastasis, pathological T stage and prostate specific antigen level. Collectively the results suggest that miR-1 may be involved in the progression of PCa, and consequently be a promising diagnostic marker. The ‘androgen receptor signaling pathway’, ‘androgen receptor activity’, ‘transcription factor binding’ and ‘protein processing in the endoplasmic reticulum’ may be crucial interactive pathways in PCa. Furthermore, PAICS, CDH1, TWIST1, ZWINT and KIAA0101 may serve as crucial miR-1 target genes in PCa.
Collapse
Affiliation(s)
- Zu-Cheng Xie
- Department of Urological Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Jia-Cheng Huang
- Department of Urological Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Li-Jie Zhang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Bin-Liang Gan
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Dong-Yue Wen
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Gang Chen
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Sheng-Hua Li
- Department of Urological Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Hai-Biao Yan
- Department of Urological Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| |
Collapse
|
48
|
Xu L, Wei B, Hui H, Sun Y, Liu Y, Yu X, Dai J. Positive feedback loop of lncRNA LINC01296/miR-598/Twist1 promotes non-small cell lung cancer tumorigenesis. J Cell Physiol 2018; 234:4563-4571. [PMID: 30240003 DOI: 10.1002/jcp.27235] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Accepted: 07/24/2018] [Indexed: 12/11/2022]
Abstract
Emerging evidence has illustrated the vital roles of long noncoding RNAs (lncRNAs) in human cancers. However, the role of lncRNAs in non-small cell lung cancer (NSCLC) is still elusive and poorly understood. In the current study, our team conducted extensive experiments to identify the role of long intergenic nonprotein coding (LINC01296) on NSCLC tumorigenesis. The results illustrated that the elevated LINC01296 expression in NSCLC tissue specimens and cell lines were closely correlated with the poor prognosis of patients with NSCLC. Functional studies revealed that LINC01296 knockdown silenced by small interfering RNAs inhibited proliferation, accelerated apoptosis in vitro, and impaired tumor growth in vivo. Mechanical studies showed that INC01296 harbored miR-598, acting as a microRNA "sponge." Besides, miR-598 targeted the 3'-UTR of Twist1. Interestingly, transcription factor Twist1 could bind with the promoter of INC01296 and activate its transcriptional level. In summary, we conclude that INC01296/miR-598/Twist1 constitutes a positive feedback loop to promote the tumorigenesis of NSCLC, providing a novel insight and a valuable therapeutic strategy.
Collapse
Affiliation(s)
- Lijuan Xu
- Department of Oncology, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Bin Wei
- Department of Oncology, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Hongxia Hui
- Department of Oncology, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Yuan Sun
- Department of Oncology, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Yangqing Liu
- Department of Oncology, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Xiaojuan Yu
- Department of Oncology, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Jian Dai
- Department of Orthopedics, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| |
Collapse
|
49
|
Kim M, Lee S, Park WH, Suh DH, Kim K, Kim YB, No JH. Silencing Bmi1 expression suppresses cancer stemness and enhances chemosensitivity in endometrial cancer cells. Biomed Pharmacother 2018; 108:584-589. [PMID: 30243092 DOI: 10.1016/j.biopha.2018.09.041] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 09/03/2018] [Accepted: 09/07/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Bmi1, a polycomb group gene, is essential for self-renewal of stem cells and is frequently upregulated in various cancer cells. We aimed to investigate the effect of Bmi1 silencing on cancer stemness and chemosensitivity in endometrial cancer using targeted siRNA approach in HEC1A and Ishikawa cells. METHODS Cell viability after treatment with Bmi1 siRNA was assessed using the MTT assay, and cell apoptosis was visualized using the TdT-mediated dUTP nick-end labeling (TUNEL) method. Western blotting, migration assays and invasion assays were performed to detect changes in the stem-like properties of cancer cells. To evaluate the anticancer effect of Bmi1 silencing, HEC1A and Ishikawa cells were treated with 100 nM Bmi1 siRNA and/or 40 μM cisplatin. RESULTS In the MTT assay, compared to control, viability of HEC1A and Ishikawa cells significantly decreased after Bmi1 siRNA treatment in a dose-dependent manner. Bmi1 silencing using siRNA increased the expression of cleaved caspase-3 and cleaved poly adenosine diphosphate-ribose polymerase polymerase (PARP) as observed in the western blot analysis. Apoptosis significantly increased in the HEC1A and Ishikawa cells treated with 100 nM Bmi1 siRNA for 48 h than in the control cells in TUNEL assay. SOX2 and Oct4 expression decreased in the HEC1A and Ishikawa cells treated with Bmi1 siRNA, while E-cadherin expression increased. Further, migratory and invasive properties were significantly inhibited by Bmi1 siRNA treatment in both cell lines. Notably, viability of HEC1A and Ishikawa cells decreased more when they were concurrently treated with Bmi1 siRNA and cisplatin compared to when they were treated with Bmi1 siRNA or cisplatin alone. CONCLUSION Bmi1 silencing suppresses cancer stemness in HEC1A and Ishikawa cells. Concurrent treatment with Bmi1 siRNA and cisplatin resulted in additive anticancer effect with a cell line-specific pattern, which was higher than that shown by cisplatin treatment alone.
Collapse
Affiliation(s)
- Miseon Kim
- Department of Obstetrics and Gynecology, CHA Gangnam Medical Center, CHA University School of Medicine, Seoul, Republic of Korea
| | - Seul Lee
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Wook Ha Park
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Dong Hoon Suh
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Kidong Kim
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Yong Beom Kim
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Jae Hong No
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea.
| |
Collapse
|
50
|
Ma J, Wang L, Li J, Zhang G, Tao H, Li X, Sun D, Hu Y. Swainsonine Inhibits Invasion and the EMT Process in Esophageal Carcinoma Cells by Targeting Twist1. Oncol Res 2018; 26:1207-1213. [PMID: 28899457 PMCID: PMC7844710 DOI: 10.3727/096504017x15046134836575] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Esophageal cancer is a common gastrointestinal cancer, with a very high mortality rate in patients with metastasis. Swainsonine, a cytotoxic fungal alkaloid, has been shown to inhibit cell growth in esophageal cancer. In the present study, we explored the effects of swainsonine on cell invasion and metastasis in esophageal cancer cells. Human esophageal carcinoma cells were treated with different doses of swainsonine, and then cell viability, invasion, and apoptosis were measured. The mRNA and protein expressions of Twist1, apoptosis- and EMT-related factors, and PI3K/AKT pathway factors were detected by qRT-PCR and Western blot. Swainsonine had no effect on esophageal cancer cell viability and apoptosis, but it significantly decreased cell invasion in a dose-dependent manner. Swainsonine increased the expression of E-cadherin but decreased the expression of N-cadherin, vimentin, ZEB1, and snail in a dose-dependent manner, thereby inhibiting EMT. Last, we found that swainsonine inhibits cell invasion and EMT in the esophageal carcinoma cells by downregulation of Twist1 and deactivation of the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Junxun Ma
- Department of Medical Oncology, Chinese PLA General Hospital, Beijing, P.R. China
| | - Lijie Wang
- Department of Medical Oncology, Chinese PLA General Hospital, Beijing, P.R. China
| | - Jinyu Li
- Department of Medical Oncology, Chinese PLA General Hospital, Beijing, P.R. China
| | - Guoqing Zhang
- Department of Medical Oncology, Chinese PLA General Hospital, Beijing, P.R. China
| | - Haitao Tao
- Department of Medical Oncology, Chinese PLA General Hospital, Beijing, P.R. China
| | - Xiaoyan Li
- Department of Medical Oncology, Chinese PLA General Hospital, Beijing, P.R. China
| | - Danyang Sun
- Department of Medical Oncology, Chinese PLA General Hospital, Beijing, P.R. China
| | - Yi Hu
- Department of Medical Oncology, Chinese PLA General Hospital, Beijing, P.R. China
| |
Collapse
|