1
|
Jiang F, Liu W, Zhou Y, Lin S, Zhang Q, Zhang W, Xue Y, Li C, Gao A, Shao M, Liao S, Ma T, Yu X. Bortezomib induces cell apoptosis and increases the efficacy of αPD-1 in BCR::ABL T315I mutation CML by targeting UBE2Q1. Int Immunopharmacol 2024; 143:113311. [PMID: 39454411 DOI: 10.1016/j.intimp.2024.113311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 09/12/2024] [Accepted: 09/30/2024] [Indexed: 10/28/2024]
Abstract
The BCR:ABL T315I mutation presents a significant challenge in the current management of Chronic Myeloid Leukemia (CML), highlighting the need to identify novel targets and drugs. In our study, we observed the elevated expression of UBE2Q1 in KBM5-T315I cells compared to KBM5 cells, where it interacted with DDX3, regulating its ubiquitination. Furthermore, we found that Bortezomib (BTZ) targeted UBE2Q1, reducing its protein level expression. Consequently, BTZ dose-dependently inhibited the growth vitality of KBM5-T315I cells, inducing increased ROS production, mitochondrial membrane potential collapse, cytochrome C release, and expression of apoptosis-related proteins. These events collectively induced apoptosis in KBM5-T315I cells. Moreover, BTZ enhanced the therapeutic effects of anti-PD-1 treatment. In NOD/SCID mice bearing KBM5-T315I cell line xenografts, BTZ administration (2 mg/kg, ip, every other day for 4 weeks) significantly inhibited the growth of KBM5-T315Iderived xenografts and extended survival. In conclusion, our study sheds new light on the BTZ-induced apoptosis mechanism, suggesting the potential of BTZ as a promising chemo-immunotherapy agent against BCR:ABL T315I mutation CML.
Collapse
MESH Headings
- Animals
- Bortezomib/pharmacology
- Bortezomib/therapeutic use
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Apoptosis/drug effects
- Fusion Proteins, bcr-abl/genetics
- Fusion Proteins, bcr-abl/metabolism
- Mice, SCID
- Cell Line, Tumor
- Mice, Inbred NOD
- Mice
- Mutation
- Ubiquitin-Conjugating Enzymes/genetics
- Ubiquitin-Conjugating Enzymes/metabolism
- Programmed Cell Death 1 Receptor/metabolism
- Programmed Cell Death 1 Receptor/antagonists & inhibitors
- Programmed Cell Death 1 Receptor/genetics
- Xenograft Model Antitumor Assays
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Drug Synergism
- Female
Collapse
Affiliation(s)
- Fengyu Jiang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Wenjie Liu
- Department of Hematology, the First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing 210029, PR China
| | - Yanyu Zhou
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Siwei Lin
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Qin Zhang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Wan Zhang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Yangyang Xue
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Cenming Li
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Anran Gao
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Miaomiao Shao
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Shanting Liao
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Tonghui Ma
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Xiaoxuan Yu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, PR China; State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, PR China.
| |
Collapse
|
2
|
Duan C, Lin X, Zou W, He Q, Wei F, Pan J, Liu C, Jin Y. Targeting DDX3X eliminates leukemia stem cells in chronic myeloid leukemia by blocking NT5DC2 mRNA translation. Oncogene 2024:10.1038/s41388-024-03215-w. [PMID: 39516658 DOI: 10.1038/s41388-024-03215-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 10/29/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024]
Abstract
Tyrosine kinase inhibitors (TKIs) are highly effective in the treatment of patients with chronic myeloid leukemia (CML), but fail to eliminate leukemia stem cells (LSCs), which can lead to disease relapse or progression. It is urgently need to identify the regulators specifically driving LSCs. In this study, we identified DEAD-box helicase 3 X-linked (DDX3X), a ubiquitously expressed RNA helicase, as a critical regulator for CML LSCs by using patient samples and BCR-ABL-driven CML mouse model. We found that DDX3X enhanced the survival, serially plating and long-term engraftment abilities of human primary CML CD34+ cells. Inhibition of DDX3X reduced leukemia burden, eradicated LSCs and extended the survival of CML mice. Mechanistically, we uncovered that DDX3X protein bound to 5'-Nucleotidase Domain Containing 2 (NT5DC2) mRNA and promoted its translation in CML cells. NT5DC2 was a functional mediator in DDX3X regulation of LSCs. Collectively, our findings provide new evidence for RNA helicase facilitating the translation of specific mRNA in LSCs. Targeting DDX3X may represent a promising therapeutic strategy for eradication of LSCs in CML patients.
Collapse
Affiliation(s)
- Chen Duan
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
- Institute of Tumor Pharmacology, College of Pharmacy, Jinan University, Guangzhou, China
| | - Xiaoying Lin
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
- Institute of Tumor Pharmacology, College of Pharmacy, Jinan University, Guangzhou, China
| | - Waiyi Zou
- Department of Hematology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qi He
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
- Institute of Tumor Pharmacology, College of Pharmacy, Jinan University, Guangzhou, China
| | - Fen Wei
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
- Institute of Tumor Pharmacology, College of Pharmacy, Jinan University, Guangzhou, China
| | - Jingxuan Pan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China.
| | - Chang Liu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China.
- Institute of Tumor Pharmacology, College of Pharmacy, Jinan University, Guangzhou, China.
| | - Yanli Jin
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China.
- Institute of Tumor Pharmacology, College of Pharmacy, Jinan University, Guangzhou, China.
| |
Collapse
|
3
|
Han Z, Aizezi A, Ma L, Su Y, Fan L, Liu J. The association between human papillomavirus and lung cancer: A Mendelian randomization study. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2024; 123:105646. [PMID: 39059733 DOI: 10.1016/j.meegid.2024.105646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 07/02/2024] [Accepted: 07/21/2024] [Indexed: 07/28/2024]
Abstract
BACKGROUND To investigate the causal relationship between human papillomavirus (HPV) and lung cancer, we conducted a study using the two-sample Mendelian randomization (TSMR). METHOD Data from genome-wide association studies (GWAS) were analyzed with HPV E7 Type 16 and HPV E7 Type 18 as exposure factors. The outcome variables included lung cancer, small cell lung cancer, adenocarcinoma and squamous cell lung cancer. Causality was estimated using inverse variance weighted (IVW), MR-Egger and weighted median methods. Heterogeneity testing, sensitivity analysis, and multiple validity analysis were also performed.. RESULTS The results showed that HPV E7 Type 16 infection was associated with a higher risk of squamous cell lung cancer (OR = 7.69; 95% CI:1.98-29.85; p = 0.0149). HPV E7 Type 18 infection significantly increased the risk of lung adenocarcinoma (OR = 0.71; 95% CI: 0.38-1.31; p = 0.0079) and lung cancer (OR = 7.69; 95% CI:1.98-29.85; p = 0.0292). No significant causal relationship was found between HPV E7 Type 16 and lung adenocarcinoma, lung cancer, or small cell lung carcinoma, and between HPV E7 Type 18 and squamous cell lung cancer or small cell lung carcinoma. CONCLUSIONS This study has revealed a causal relationship between HPV and lung cancers. Our findings provide valuable insights for further mechanistic and clinical studies on HPV-mediated cancer.
Collapse
Affiliation(s)
- Zhongcheng Han
- Department of Oncology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region 830002, PR China.
| | - Ayixiamuguli Aizezi
- Department of Oncology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region 830002, PR China
| | - Lili Ma
- Department of Oncology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region 830002, PR China
| | - Ying Su
- Department of Oncology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region 830002, PR China
| | - Lijuan Fan
- Department of Oncology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region 830002, PR China
| | - Jiang Liu
- Department of Oncology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region 830002, PR China
| |
Collapse
|
4
|
Benitez DA, Cumplido-Laso G, Olivera-Gómez M, Del Valle-Del Pino N, Díaz-Pizarro A, Mulero-Navarro S, Román-García A, Carvajal-Gonzalez JM. p53 Genetics and Biology in Lung Carcinomas: Insights, Implications and Clinical Applications. Biomedicines 2024; 12:1453. [PMID: 39062026 PMCID: PMC11274425 DOI: 10.3390/biomedicines12071453] [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: 05/16/2024] [Revised: 06/20/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024] Open
Abstract
The TP53 gene is renowned as a tumor suppressor, playing a pivotal role in overseeing the cell cycle, apoptosis, and maintaining genomic stability. Dysregulation of p53 often contributes to the initiation and progression of various cancers, including lung cancer (LC) subtypes. The review explores the intricate relationship between p53 and its role in the development and progression of LC. p53, a crucial tumor suppressor protein, exists in various isoforms, and understanding their distinct functions in LC is essential for advancing our knowledge of this deadly disease. This review aims to provide a comprehensive literature overview of p53, its relevance to LC, and potential clinical applications.
Collapse
Affiliation(s)
- Dixan A. Benitez
- Departamento de Bioquímica, Biología Molecular y Genética, Facultad de Ciencias, Universidad de Extremadura, 06006 Badajoz, Spain; (G.C.-L.); (M.O.-G.); (N.D.V.-D.P.); (A.D.-P.); (S.M.-N.); (A.R.-G.)
| | | | | | | | | | | | | | - Jose Maria Carvajal-Gonzalez
- Departamento de Bioquímica, Biología Molecular y Genética, Facultad de Ciencias, Universidad de Extremadura, 06006 Badajoz, Spain; (G.C.-L.); (M.O.-G.); (N.D.V.-D.P.); (A.D.-P.); (S.M.-N.); (A.R.-G.)
| |
Collapse
|
5
|
Zhang H, Mañán-Mejías PM, Miles HN, Putnam AA, MacGillivray LR, Ricke WA. DDX3X and Stress Granules: Emerging Players in Cancer and Drug Resistance. Cancers (Basel) 2024; 16:1131. [PMID: 38539466 PMCID: PMC10968774 DOI: 10.3390/cancers16061131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 03/09/2024] [Accepted: 03/11/2024] [Indexed: 05/02/2024] Open
Abstract
The DEAD (Asp-Glu-Ala-Asp)-box helicase 3 X-linked (DDX3X) protein participates in many aspects of mRNA metabolism and stress granule (SG) formation. DDX3X has also been associated with signal transduction and cell cycle regulation that are important in maintaining cellular homeostasis. Malfunctions of DDX3X have been implicated in multiple cancers, including brain cancer, leukemia, prostate cancer, and head and neck cancer. Recently, literature has reported SG-associated cancer drug resistance, which correlates with a negative disease prognosis. Based on the connections between DDX3X, SG formation, and cancer pathology, targeting DDX3X may be a promising direction for cancer therapeutics development. In this review, we describe the biological functions of DDX3X in terms of mRNA metabolism, signal transduction, and cell cycle regulation. Furthermore, we summarize the contributions of DDX3X in SG formation and cellular stress adaptation. Finally, we discuss the relationships of DDX3X, SG, and cancer drug resistance, and discuss the current research progress of several DDX3X inhibitors for cancer treatment.
Collapse
Affiliation(s)
- Han Zhang
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Paula M. Mañán-Mejías
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Hannah N. Miles
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Andrea A. Putnam
- Department of Biomolecular Chemistry, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | | | - William A. Ricke
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Urology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
- George M. O’Brien Urology Research Center of Excellence, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|
6
|
Aubry A, Pearson JD, Charish J, Yu T, Sivak JM, Xirodimas DP, Avet-Loiseau H, Corre J, Monnier PP, Bremner R. Deneddylation of ribosomal proteins promotes synergy between MLN4924 and chemotherapy to elicit complete therapeutic responses. Cell Rep 2023; 42:112925. [PMID: 37552601 DOI: 10.1016/j.celrep.2023.112925] [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: 11/14/2022] [Revised: 05/29/2023] [Accepted: 07/18/2023] [Indexed: 08/10/2023] Open
Abstract
The neddylation inhibitor MLN4924/Pevonedistat is in clinical trials for multiple cancers. Efficacy is generally attributed to cullin RING ligase (CRL) inhibition, but the contribution of non-CRL targets is unknown. Here, CRISPR screens map MLN4924-monotherapy sensitivity in retinoblastoma to a classic DNA damage-induced p53/E2F3/BAX-dependent death effector network, which synergizes with Nutlin3a or Navitoclax. In monotherapy-resistant cells, MLN4924 plus standard-of-care topotecan overcomes resistance, but reduces DNA damage, instead harnessing ribosomal protein nucleolar-expulsion to engage an RPL11/p21/MYCN/E2F3/p53/BAX synergy network that exhibits extensive cross-regulation. Strikingly, unneddylatable RPL11 substitutes for MLN4924 to perturb nucleolar function and enhance topotecan efficacy. Orthotopic tumors exhibit complete responses while preserving visual function. Moreover, MLN4924 plus melphalan deploy this DNA damage-independent strategy to synergistically kill multiple myeloma cells. Thus, MLN4924 synergizes with standard-of-care drugs to unlock a nucleolar death effector network across cancer types implying broad therapeutic relevance.
Collapse
Affiliation(s)
- Arthur Aubry
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, ON, Canada; Department of Lab Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada; Centre Hospitalo-universitaire (CHU) de Toulouse, Institut Universitaire du Cancer de Toulouse-Oncopole (IUCT-O), Université de Toulouse, UPS, Unité de Génomique du Myélome, Toulouse, France
| | - Joel D Pearson
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, ON, Canada
| | - Jason Charish
- Department of Ophthalmology and Vision Science, University of Toronto, Toronto, ON, Canada; Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Tao Yu
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, ON, Canada
| | - Jeremy M Sivak
- Department of Lab Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada; Department of Ophthalmology and Vision Science, University of Toronto, Toronto, ON, Canada; Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | | | - Hervé Avet-Loiseau
- Centre Hospitalo-universitaire (CHU) de Toulouse, Institut Universitaire du Cancer de Toulouse-Oncopole (IUCT-O), Université de Toulouse, UPS, Unité de Génomique du Myélome, Toulouse, France; Centre de Recherches en Cancérologie de Toulouse (CRCT), INSERM, Toulouse, France
| | - Jill Corre
- Centre Hospitalo-universitaire (CHU) de Toulouse, Institut Universitaire du Cancer de Toulouse-Oncopole (IUCT-O), Université de Toulouse, UPS, Unité de Génomique du Myélome, Toulouse, France; Centre de Recherches en Cancérologie de Toulouse (CRCT), INSERM, Toulouse, France
| | - Philippe P Monnier
- Department of Ophthalmology and Vision Science, University of Toronto, Toronto, ON, Canada; Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Rod Bremner
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, ON, Canada; Department of Lab Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada; Department of Ophthalmology and Vision Science, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
7
|
Mukherjee AG, Ramesh Wanjari U, Valsala Gopalakrishnan A, Jayaraj R, Katturajan R, Kannampuzha S, Murali R, Namachivayam A, Evan Prince S, Vellingiri B, Dey A, Renu K. HPV-associated cancers: insights into the mechanistic scenario and latest updates. Med Oncol 2023; 40:212. [PMID: 37358816 DOI: 10.1007/s12032-023-02085-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 06/03/2023] [Indexed: 06/27/2023]
Abstract
Cancer and related diseases are the second leading cause of death worldwide. The human papillomavirus (HPV) is an infectious agent that can be spread mainly through sexual contact and has been linked to several malignancies in both sexes. HPV is linked to almost all cases of cervical cancer. It is also linked to many head and neck cancer (HNC) cases, especially oropharyngeal cancer. Also, some HPV-related cancers, like vaginal, vulvar, penile, and anal cancers, are related to the anogenital area. Over the past few decades, testing for and preventing cervical cancer has improved, but anogenital cancers are still harder to confirm. HPV16 and HPV18 have been extensively researched due to their significant carcinogenic potential. The products of two early viral genes, E6 and E7, have been identified as playing crucial roles in cellular transformation, as emphasized by biological investigations. The complete characterization of numerous mechanisms employed by E6 and E7 in undermining the regulation of essential cellular processes has significantly contributed to our comprehension of HPV-induced cancer progression. This review focuses on the various types of cancers caused by HPV infection and also sheds light on the signaling cascades involved in the same.
Collapse
Affiliation(s)
- Anirban Goutam Mukherjee
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - Uddesh Ramesh Wanjari
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India.
| | - Rama Jayaraj
- Jindal Institute of Behavioral Sciences (JIBS), Jindal Global Institution of Eminence Deemed to Be University, 28, Sonipat, 131001, India
- Director of Clinical Sciences, Northern Territory Institute of Research and Training, Darwin, NT, 0909, Australia
| | - Ramkumar Katturajan
- Department of Biotechnology, School of Bio-Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - Sandra Kannampuzha
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - Reshma Murali
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - Arunraj Namachivayam
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - Sabina Evan Prince
- Department of Biotechnology, School of Bio-Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - Balachandar Vellingiri
- Stem cell and Regenerative Medicine/Translational Research, Department of Zoology, School of Basic Sciences, Central University of Punjab (CUPB), Bathinda, Punjab, 151401, India
| | - Abhijit Dey
- Department of Life Sciences, Presidency University, Kolkata, West Bengal, 700073, India
| | - Kaviyarasi Renu
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Institute of Medical and Technical Sciences, Saveetha Dental College & Hospitals, Saveetha University, Chennai, Tamil Nadu, 600077, India
| |
Collapse
|
8
|
Gao F, Hu K, Zheng P, Shi H, Ke X. Characteristics and prognosis of
rrDLBCL
with
TP53
mutations and a high‐risk subgroup represented by the co‐mutations of
DDX3X‐TP53. Cancer Med 2023; 12:10267-10279. [DOI: 10.1002/cam4.5756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 02/09/2023] [Accepted: 02/17/2023] [Indexed: 03/29/2023] Open
|
9
|
Nilius-Eliliwi V, Tembrink M, Gerding WM, Lubieniecki KP, Lubieniecka JM, Kankel S, Liehr T, Mika T, Dimopoulos F, Döhner K, Schroers R, Nguyen HHP, Vangala DB. Broad genomic workup including optical genome mapping uncovers a DDX3X: MLLT10 gene fusion in acute myeloid leukemia. Front Oncol 2022; 12:959243. [PMID: 36158701 PMCID: PMC9501710 DOI: 10.3389/fonc.2022.959243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 08/12/2022] [Indexed: 11/24/2022] Open
Abstract
In acute myeloid leukemia (AML), treatment decisions are currently made according to the risk classification of the European LeukemiaNet (ELN), which is based on genetic alterations. Recently, optical genome mapping (OGM) as a novel method proved to yield a genome-wide and detailed cytogenetic characterization at the time of diagnosis. A young female patient suffered from a rather unexpected aggressive disease course under FLT3 targeted therapy in combination with induction chemotherapy. By applying a “next-generation diagnostic workup“ strategy with OGM and whole-exome sequencing (WES), a DDX3X: MLLT10 gene fusion could be detected, otherwise missed by routine diagnostics. Furthermore, several aspects of lineage ambiguity not shown by standard diagnostics were unraveled such as deletions of SUZ12 and ARPP21, as well as T-cell receptor recombination. In summary, the detection of this particular gene fusion DDX3X: MLLT10 in a female AML patient and the findings of lineage ambiguity are potential explanations for the aggressive course of disease. Our study demonstrates that OGM can yield novel clinically significant results, including additional information helpful in disease monitoring and disease biology.
Collapse
Affiliation(s)
- Verena Nilius-Eliliwi
- Department of Medicine, Hematology and Oncology, Knappschaftskrankenhaus, Ruhr-University Bochum, Bochum, Germany
| | | | | | | | | | - Stefanie Kankel
- Jena University Hospital, Friedrich Schiller University, Institute of Human Genetics, Jena, Germany
| | - Thomas Liehr
- Jena University Hospital, Friedrich Schiller University, Institute of Human Genetics, Jena, Germany
| | - Thomas Mika
- Department of Medicine, Hematology and Oncology, Knappschaftskrankenhaus, Ruhr-University Bochum, Bochum, Germany
| | - Fotios Dimopoulos
- Department of Medicine, Hematology and Oncology, Knappschaftskrankenhaus, Ruhr-University Bochum, Bochum, Germany
| | - Konstanze Döhner
- Department of Internal Medicine III, University Hospital Ulm, Ulm, Germany
| | - Roland Schroers
- Department of Medicine, Hematology and Oncology, Knappschaftskrankenhaus, Ruhr-University Bochum, Bochum, Germany
| | | | - Deepak Ben Vangala
- Department of Medicine, Hematology and Oncology, Knappschaftskrankenhaus, Ruhr-University Bochum, Bochum, Germany
- *Correspondence: Deepak Ben Vangala,
| |
Collapse
|
10
|
Doneti R, Pasha A, Botlagunta M, Heena SK, Mutyala VVVP, Pawar SC. Molecular docking, synthesis, and biological evaluation of 7-azaindole-derivative (7AID) as novel anti-cancer agent and potent DDX3 inhibitor:-an in silico and in vitro approach. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 39:179. [PMID: 36048256 DOI: 10.1007/s12032-022-01826-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 08/15/2022] [Indexed: 11/30/2022]
Abstract
The DEAD-box helicase family member DDX3 is involved in many diseases, such as viral infection, inflammation, and cancer. Many studies in the last decade have revealed the role of DDX3 in tumorigenesis and metastasis. DDX3 has both tumour suppressor and oncogenic effect, in the present study we have evaluated the expression levels of DDX3 in cervical squamous cell carcinoma at mRNA level via real-time PCR and protein level via Immunohistochemistry. DDX3 has become a molecule of interest in cancer biology that promotes drug resistance by adaptive response inevitably leading to treatment failure. One approach to avoid the development of resistant to disease is to create novel drugs that target the overexpressed proteins, we designed and synthesized a novel 7-azaindole derivative (7-AID) compound, {5-[1H-pyrrolo (2, 3-b) pyridin-5-yl] pyridin-2-ol]} that could lodge within the adenosine-binding pocket of the DDX3 (PDB ID: 2I4I). The binding efficacy of 7-AID compound with DDX3 was analysed by molecular docking studies. 7-AID was found to interact with the key residues Tyr200 and Arg202 from the Q-motif rendered by π-interactions and hydrogen bonds within the binding pocket with good docking score - 7.99 kcal/mol. The cytotoxicity effect of 7-AID compound was evaluated using MTT assay on human cervical carcinoma cells (HeLa) and breast cancer cells (MCF-7 and MDA MB-231) and the compound shown effective inhibitory concentration (IC50) on Hela cells 16.96 µM/ml and 14.12 and 12.69 µM/ml on MCF-7 and MDA MB-231, respectively. Further, the in-vitro, in-vivo anti-cancer and anti-angiogenic assessment of 7-AID compound was evaluated on Hela cells using scratch wound-healing assay, DAPI staining, cell cycle analysis, immunoblotting, and chorioallontoic membrane assay. Furthermore, the inhibitory effect of derivative compound on DDX3 was investigated in HeLa, MCF-7, and MDA MB-231 cells at the mRNA and protein levels. The results showed that the 7-AID compound effectively inhibited DDX3 in a dose-dependent manner, and the findings suggest that the compound could be used as a potential DDX3 inhibitor.
Collapse
Affiliation(s)
- Ravinder Doneti
- Department of Genetics & Biotechnology, Osmania University, Hyderabad, Telangana, 500 007, India
| | - Akbar Pasha
- Department of Genetics & Biotechnology, Osmania University, Hyderabad, Telangana, 500 007, India
| | - Mahendran Botlagunta
- School of Biosciences Engineering and Technology, VIT Bhopal University, Bhopal, Madhya Pradesh, 466114, India
| | - S K Heena
- Department of Pathology, Osmania Medical College, Hyderabad, Telangana, 500095, India
| | | | - Smita C Pawar
- Department of Genetics & Biotechnology, Osmania University, Hyderabad, Telangana, 500 007, India.
| |
Collapse
|
11
|
Li L, Du W, Wang H, Zhao Y, Huang Z, Peng Y, Zeng S, Zhang G. Small-molecule MX-C2/3 suppresses non-small cell lung cancer progression via p53 activation. Chem Biol Interact 2022; 366:110142. [PMID: 36058261 DOI: 10.1016/j.cbi.2022.110142] [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: 07/22/2022] [Revised: 08/22/2022] [Accepted: 08/28/2022] [Indexed: 11/30/2022]
Abstract
p53 inactivation is a common feature in non-small cell lung cancer (NSCLC) resulting in NSCLC malignant transformation. Targeting serine 392 phosphorylation to restore p53 anticancer activity has proven to be an effective therapeutic strategy against NSCLC. A synthetic p53 activator, NA-17, has been developed that shows promise in preclinical models of NSCLC. However, NA-17 exhibits limited therapeutic efficacy in oncogene-driven tumors as well as relatively high toxicity to normal cells. It is possible that high efficiency and low toxicity p53 activators can be obtained by optimizing the leading molecule. Here, we performed high-throughput screening of compounds optimized based on NA-17 to identify new p53 activators. Two promising candidates named MX-C2 and MX-C3 were identified, both exhibited considerable therapeutic efficacy in oncogene-driven tumor models. Similar to NA-17, MX-C2/3 induced p53 activation via phosphorylating serine-392 without DNA damage. Both compounds showed broad antitumor activity in NSCLC cells and limited toxicity in normal cell lines. Moreover, MX-C2/3 suppressed tumor progression by arresting the cell cycle at G2/M phase, exhibiting a different mechanism of cell cycle arrest than NA-17. In addition, MX-C2/3 promoted the enrichment of p-p53 (s392) in mitochondria, leading to the conformational activation of Bak for cell apoptosis, which is consistent with NA-17. Finally, we demonstrated that MX-C2 significantly inhibited tumor growth without obvious systemic toxicity in oncogene-driven HCC-827 xenograft models. Collectively, we report two p53 activators with high-efficiency and low-toxicity that target p53 serine 392 phosphorylation for anticancer translational investigation.
Collapse
Affiliation(s)
- Liangping Li
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, China
| | - Wenqing Du
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, China
| | - Hui Wang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, China
| | - Yufei Zhao
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, China
| | - Zetian Huang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, China
| | - Yan Peng
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, China
| | - Shulan Zeng
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, China.
| | - Guohai Zhang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, China.
| |
Collapse
|
12
|
Secchi M, Lodola C, Garbelli A, Bione S, Maga G. DEAD-Box RNA Helicases DDX3X and DDX5 as Oncogenes or Oncosuppressors: A Network Perspective. Cancers (Basel) 2022; 14:cancers14153820. [PMID: 35954483 PMCID: PMC9367324 DOI: 10.3390/cancers14153820] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/01/2022] [Accepted: 08/04/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary The transformation of a normal cell into a cancerous one is caused by the deregulation of different metabolic pathways, involving a complex network of protein–protein interactions. The cellular enzymes DDX3X and DDX5 play important roles in the maintenance of normal cell metabolism, but their deregulation can accelerate tumor transformation. Both DDX3X and DDX5 interact with hundreds of different cellular proteins, and depending on the specific pathways in which they are involved, both proteins can either act as suppressors of cancer or as oncogenes. In this review, we summarize the current knowledge about the roles of DDX3X and DDX5 in different tumors. In addition, we present a list of interacting proteins and discuss the possible contribution of some of these protein–protein interactions in determining the roles of DDX3X and DDX5 in the process of cancer proliferation, also suggesting novel hypotheses for future studies. Abstract RNA helicases of the DEAD-box family are involved in several metabolic pathways, from transcription and translation to cell proliferation, innate immunity and stress response. Given their multiple roles, it is not surprising that their deregulation or mutation is linked to different pathological conditions, including cancer. However, while in some cases the loss of function of a given DEAD-box helicase promotes tumor transformation, indicating an oncosuppressive role, in other contexts the overexpression of the same enzyme favors cancer progression, thus acting as a typical oncogene. The roles of two well-characterized members of this family, DDX3X and DDX5, as both oncogenes and oncosuppressors have been documented in several cancer types. Understanding the interplay of the different cellular contexts, as defined by the molecular interaction networks of DDX3X and DDX5 in different tumors, with the cancer-specific roles played by these proteins could help to explain their apparently conflicting roles as cancer drivers or suppressors.
Collapse
|
13
|
Li L, Li P, Ma X, Zeng S, Peng Y, Zhang G. Therapeutic restoring p53 function with small molecule for oncogene-driven non-small cell lung cancer by targeting serine 392 phosphorylation. Biochem Pharmacol 2022; 203:115188. [PMID: 35902040 DOI: 10.1016/j.bcp.2022.115188] [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: 05/11/2022] [Revised: 07/05/2022] [Accepted: 07/20/2022] [Indexed: 12/01/2022]
Abstract
p53 inactivation by disabling its function is a hallmark in lung carcinomas, emphasizing the significance of restoring p53 function as an attractive therapeutic strategy. However, the clinical efficacy of existing p53 activators is limited due to their inability to effectively activate p53 within the tumors. Here, we established a p53 activator screening assay in EGFR-driven lung cancer cells and identified a small molecular, MX-C4, as a promising candidate. Using high throughput compound screening and combination analyses, we found that MX-C4 effectively promoted the phosphorylation of p53 at serine-392 (s392). It exhibited potent antitumor activity in a variety of cancer cell lines, but only limited toxicity to NCI-H1299 (p53-null) and normal cell lines such as LX2 and HL-7702. Overexpression of p53 in NCI-H1299 cells by a p53 expressing virus vector sensitized cells to MX-C4 treatment, suggesting a p53-dependent anticancer activity. Furthermore, we demonstrated that MX-C4 bound to p53 and exerted its anticancer activity through cell cycle arrest at G2/M phase and apoptosis induction. Mechanistic study indicated that p53 activation regulated cell cycle and cell survival related targets at protein levels. Moreover, p53 activation raised phospho-p53 translocation to mitochondria and subsequently reorganized the Bcl-xl-Bak complex, thus conformationally activating Bak and inducing apoptosis. It is noteworthy that MX-C4 could effectively activate p53 within the tumors in EGFR-driven xenograft models, where tumor was significantly suppressed without obvious toxicity. Our study identified a promising candidate for lung cancer therapy by restoring p53 function.
Collapse
Affiliation(s)
- Liangping Li
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Ministry of Science and Technology of China, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Pingping Li
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Ministry of Science and Technology of China, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Xuesong Ma
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Ministry of Science and Technology of China, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Shulan Zeng
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Ministry of Science and Technology of China, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China.
| | - Yan Peng
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Ministry of Science and Technology of China, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China.
| | - Guohai Zhang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Ministry of Science and Technology of China, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China.
| |
Collapse
|
14
|
Huang JY, Lin C, Tsai SCS, Lin FCF. Human Papillomavirus Is Associated With Adenocarcinoma of Lung: A Population-Based Cohort Study. Front Med (Lausanne) 2022; 9:932196. [PMID: 35847783 PMCID: PMC9279738 DOI: 10.3389/fmed.2022.932196] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 05/25/2022] [Indexed: 11/13/2022] Open
Abstract
Background Recent trends in the incidence of lung cancer have been reported despite the decreasing rate of smoking. Lung cancer is ranked among the top causes of cancer-related deaths. The ratio of adenocarcinoma to squamous cell carcinoma, as well as the ratio of women to men, is still increasing. Human papillomavirus (HPV) has been discovered in lung cancer tissues and blood specimens, particularly in Eastern countries. However, the association between HPV infection and lung adenocarcinoma remains unclear. Methods This population-based cohort study was conducted using data from Taiwan's single-payer national health insurance and cancer registry databases. Data on HPV infection, cancer, sex, age, comorbidities, urbanization, and occupation were collected. The cumulative incidence rates were generated using Kaplan–Meier curves and log-rank tests. COX regression analysis was used to estimate the hazard ratios of factors associated with cancer occurrence. We used data from 2007 and 2015. The cases were matched with sex and age in a 1:2 manner with 939,874 HPV+ and 1,879,748 HPV– individuals, respectively. Results The adjusted hazard ratios [95% confidence interval (CI)] for HPV infection in all lung cancers were 1.539 (1.436–1.649), male lung cancer 1.434 (1.312–1.566), female lung cancer 1.742 (1.557–1.948), squamous cell carcinoma (SCC) 1.092 (0.903–1.320), male SCC 1.092 (0.903–1.320), female SCC 0.949 (0.773–1.164), adenocarcinoma 1.714 (1.572–1.870), male adenocarcinoma 1.646 (1.458–1.858), and female adenocarcinoma 1.646 (1.458–1.858). The highest adjusted hazard ratio for lung cancer was chronic obstructive pulmonary disease (COPD) 1.799 (1.613–2.007), followed by male sex 1.567 (1.451–6.863) and HPV infection. The highest adjusted hazard ratio for adenocarcinoma was HPV infection 1.714 (1.572–1.870), followed by COPD 1.300 (1.102–1.533), and for SCC, male sex 5.645 (4.43–3.37), followed by COPD 2.528 (2.002–3.192). Conclusion Our study showed that HPV infection was associated with the occurrence of adenocarcinoma of the lung in both men and women but was not associated with SCC of the lung.
Collapse
Affiliation(s)
- Jing-Yang Huang
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Chuck Lin
- College of William and Mary, Williamsburg, VA, United States
| | - Stella Chin-Shaw Tsai
- Superintendents' Office, Tungs' Taichung Metro Harbor Hospital, Taichung, Taiwan
- National Chung Hsing University, Taichung, Taiwan
| | - Frank Cheau-Feng Lin
- Department of Thoracic Surgery, Chung Shan Medical University Hospital, Taichung, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- *Correspondence: Frank Cheau-Feng Lin
| |
Collapse
|
15
|
Wang J, Wang Y, Wang J, Zhang S, Yu Z, Zheng K, Fu Z, Wang C, Huang W, Chen J. DEAD-box helicase 56 functions as an oncogene promote cell proliferation and invasion in gastric cancer via the FOXO1/p21 Cip1/c-Myc signaling pathway. Bioengineered 2022; 13:13970-13985. [PMID: 35723050 PMCID: PMC9275944 DOI: 10.1080/21655979.2022.2084235] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
DEAD-box helicase (DDX) family exerts a critical effect on cancer initiation and progression through alternative splicing, transcription and ribosome biogenesis. Increasing evidence has demonstrated that DEAD-box helicase 56 (DDX56) is over-expressed in several cancers, which plays an oncogenic role. Till the present, the impact of DDX56 on gastric cancer (GC) remains unclear. We conducted high-throughput sequencing (RNA-seq) to demonstrate aberrant DDX56 levels within 10 GC and matched non-carcinoma tissue samples. DDX56 levels were detected through qRT-PCR, western blotting (WB) and immunochemical staining in GC patients. We conducted gain- and loss-of-function studies to examine DDX56's biological role in GC development. In vitro, we carried out 5‑Ethynyl‑2‑deoxyuridine (EdU), scratch, Transwell, and flow cytometry (FCM) assays for detecting GC cell growth, invasion, migration and apoptosis. Additionally, gene set enrichment analysis (GSEA), WB assay, and Encyclopedia of RNA Interactomes (ENCORI) were carried out for analyzing DDX56-regulated downstream genes and signaling pathways. In vivo, tumor xenograft experiment was performed for investigating how DDX56 affected GC development within BALB/c nude mice. Functionally, DDX56 knockdown arrested cell cycle at G1 phase, invasion and migration of AGS and MKN28 cells, and enhanced their apoptosis. Ectopic DDX56 expression enhanced the cell growth, migration and invasion, and inhibited apoptosis. Knockdown of DDX56 suppressed GC growth in the tumor models of BALB/c nude mice. Mechanistically, DDX56 post-transcriptionally suppressed FOXO1/p21 Cip1 protein expression, which could activate its downstream cyclin E1/CDK2/c-Myc signaling pathways. This sheds lights on the GC pathogenic mechanism and offers a potential anti-cancer therapeutic target.
Collapse
Affiliation(s)
- Jiancheng Wang
- Department of Gastrointestinal Gland Surgery, the First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China.,Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Ye Wang
- Department of Gastrointestinal Gland Surgery, the First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China.,Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Junfu Wang
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Siwen Zhang
- Department of Gastrointestinal Gland Surgery, the First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China.,Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Zhu Yu
- Department of Gastrointestinal Gland Surgery, the First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China.,Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Kaitian Zheng
- Department of Gastrointestinal Gland Surgery, the First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China.,Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Zhao Fu
- Department of Gastrointestinal Gland Surgery, the First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China.,Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Congjun Wang
- Department of Gastrointestinal Gland Surgery, the First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China.,Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Weijia Huang
- Department of Gastrointestinal Gland Surgery, the First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China.,Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Junqiang Chen
- Department of Gastrointestinal Gland Surgery, the First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China.,Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
16
|
High Risk-Human Papillomavirus in HNSCC: Present and Future Challenges for Epigenetic Therapies. Int J Mol Sci 2022; 23:ijms23073483. [PMID: 35408843 PMCID: PMC8998945 DOI: 10.3390/ijms23073483] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/16/2022] [Accepted: 03/18/2022] [Indexed: 02/01/2023] Open
Abstract
Head and Neck Squamous Cell Carcinoma (HNSCC) is a highly heterogeneous group of tumors characterized by an incidence of 650,000 new cases and 350,000 deaths per year worldwide and a male to female ratio of 3:1. The main risk factors are alcohol and tobacco consumption and Human Papillomavirus (HPV) infections. HNSCC cases are divided into two subgroups, the HPV-negative (HPV−) and the HPV-positive (HPV+) which have different clinicopathological and molecular profiles. However, patients are still treated with the same therapeutic regimens. It is thus of utmost importance to characterize the molecular mechanisms underlying these differences to find new biomarkers and novel therapeutic targets towards personalized therapies. Epigenetic alterations are a hallmark of cancer and can be exploited as both promising biomarkers and potential new targets. E6 and E7 HPV oncoviral proteins besides targeting p53 and pRb, impair the expression and the activity of several epigenetic regulators. While alterations in DNA methylation patterns have been well described in HPV+ and HPV− HNSCC, accurate histone post-translational modifications (hPTMs) characterization is still missing. Herein, we aim to provide an updated overview on the impact of HPV on the hPTMs landscape in HNSCC. Moreover, we will also discuss the sex and gender bias in HNSCC and how the epigenetic machinery could be involved in this process, and the importance of taking into account sex and/or gender also in this field.
Collapse
|
17
|
Pardeshi J, McCormack N, Gu L, Ryan CS, Schröder M. DDX3X functionally and physically interacts with Estrogen Receptor-alpha. BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2022; 1865:194787. [PMID: 35121200 DOI: 10.1016/j.bbagrm.2022.194787] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 11/19/2022]
Abstract
DEAD-box protein 3X (DDX3X) is a human DEAD-box protein with conventional roles in RNA metabolism and unconventional functions in signalling pathways that do not require its enzymatic activity. For example, DDX3X acts as a multifunctional adaptor molecule in anti-viral innate immune signalling pathways, where it interacts with and regulates the kinase IKB-kinase-epsilon (IIKKε). Interestingly, both DDX3X and IKKɛ have also independently been shown to act as breast cancer oncogenes. IKKɛ's oncogenic functions are likely multifactorial, but it was suggested to phosphorylate the transcription factor Estrogen receptor alpha (ERα) at Serine 167, which drives expression of Erα target genes in an estrogen-independent manner. In this study, we identified a novel physical interaction between DDX3X and ERα that positively regulates ERα activation. DDX3X knockdown in ER+ breast cancer cell lines resulted in reduced ERα phosphorylation, reduced Estrogen Response Element (ERE)-controlled reporter gene expression, decreased expression of ERα target genes, and decreased cell proliferation. Vice versa, overexpression of DDX3X resulted in enhanced ERα phosphorylation and activity. Furthermore, we provide evidence that DDX3X physically binds to ERα from co-immunoprecipitation and pulldown experiments. Based on our data, we propose that DDX3X acts as an adaptor to facilitate IKKε-mediated ERα activation, akin to the mechanism we previously elucidated for IKKε-mediated Interferon Regulatory factor 3 (IRF3) activation in innate immune signalling. In conclusion, our research provides a novel molecular mechanism that might contribute to the oncogenic effect of DDX3X in breast cancer, potentially linking it to the development of resistance against endocrine therapy.
Collapse
Affiliation(s)
- Jyotsna Pardeshi
- Biology Department, Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co. Kildare, Ireland
| | - Niamh McCormack
- Biology Department, Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co. Kildare, Ireland
| | - Lili Gu
- Biology Department, Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co. Kildare, Ireland
| | - Cathal S Ryan
- Biology Department, Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co. Kildare, Ireland
| | - Martina Schröder
- Biology Department, Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co. Kildare, Ireland.
| |
Collapse
|
18
|
Wang J, Yang F, Zhuang J, Huo Q, Li J, Xie N. TRIM58 Inactivates p53/p21 to Promote Chemoresistance via Ubiquitination of DDX3 in Breast Cancer. Int J Biochem Cell Biol 2021; 143:106140. [PMID: 34954155 DOI: 10.1016/j.biocel.2021.106140] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 12/18/2021] [Accepted: 12/20/2021] [Indexed: 02/07/2023]
Abstract
Chemotherapy resistance is that the most important reason behind of carcinoma treatment failure but the underlying molecular mechanisms are unclear. Members of the tripartite motifcontaining protein (TRIM) family play crucial roles in the carcinogenesis and development of resistance against chemotherapy. Herein, we first confirmed that TRIM58 is highly expressed in triple-negative breast cancer tissues and drug-resistant MCF7/ADR cells. Furthermore, TRIM58 knockdown resulted in increased sensitivity of MCF7/ADR cells toward doxorubicin in vitro and in vivo. In contrast, TRIM58 overexpression in breast cancer cells increased doxorubicin resistance. TRIM58 was found to interact with DDX3, a protein recently reported to modulate resistance against chemotherapy. We found that TRIM58 negatively regulates DDX3 expression downstream of the P53/P21 pathway, and that DDX3 is degraded by TRIM58-mediated ubiquitination. Knockdown of DDX3 reversed doxorubicin chemotherapy sensitivity induced by TRIM58 knockdown via the P53/P21 pathway.Our study reveals that TRIM58 mediates a novel mechanism underlying the development of resistance against chemotherapy in breast cancer and provides potential targets for developing novel therapeutic targets for breast cancer.
Collapse
Affiliation(s)
- Juan Wang
- Biobank, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China; University of South China, Hunan, 421001, China
| | - Fan Yang
- Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, 315000, China
| | - Jialang Zhuang
- Biobank, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China
| | - Qin Huo
- Biobank, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China
| | - Jiaying Li
- University of South China, Hunan, 421001, China
| | - Ni Xie
- Biobank, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China.
| |
Collapse
|
19
|
DEAD-Box RNA Helicases in Cell Cycle Control and Clinical Therapy. Cells 2021; 10:cells10061540. [PMID: 34207140 PMCID: PMC8234093 DOI: 10.3390/cells10061540] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/11/2021] [Accepted: 06/15/2021] [Indexed: 12/11/2022] Open
Abstract
Cell cycle is regulated through numerous signaling pathways that determine whether cells will proliferate, remain quiescent, arrest, or undergo apoptosis. Abnormal cell cycle regulation has been linked to many diseases. Thus, there is an urgent need to understand the diverse molecular mechanisms of how the cell cycle is controlled. RNA helicases constitute a large family of proteins with functions in all aspects of RNA metabolism, including unwinding or annealing of RNA molecules to regulate pre-mRNA, rRNA and miRNA processing, clamping protein complexes on RNA, or remodeling ribonucleoprotein complexes, to regulate gene expression. RNA helicases also regulate the activity of specific proteins through direct interaction. Abnormal expression of RNA helicases has been associated with different diseases, including cancer, neurological disorders, aging, and autosomal dominant polycystic kidney disease (ADPKD) via regulation of a diverse range of cellular processes such as cell proliferation, cell cycle arrest, and apoptosis. Recent studies showed that RNA helicases participate in the regulation of the cell cycle progression at each cell cycle phase, including G1-S transition, S phase, G2-M transition, mitosis, and cytokinesis. In this review, we discuss the essential roles and mechanisms of RNA helicases in the regulation of the cell cycle at different phases. For that, RNA helicases provide a rich source of targets for the development of therapeutic or prophylactic drugs. We also discuss the different targeting strategies against RNA helicases, the different types of compounds explored, the proposed inhibitory mechanisms of the compounds on specific RNA helicases, and the therapeutic potential of these compounds in the treatment of various disorders.
Collapse
|
20
|
Abstract
The DEAD-box helicase family member DDX3X (DBX, DDX3) functions in nearly all stages of RNA metabolism and participates in the progression of many diseases, including virus infection, inflammation, intellectual disabilities and cancer. Over two decades, many studies have gradually unveiled the role of DDX3X in tumorigenesis and tumour progression. In fact, DDX3X possesses numerous functions in cancer biology and is closely related to many well-known molecules. In this review, we describe the function of DDX3X in RNA metabolism, cellular stress response, innate immune response, metabolic stress response in pancreatic β cells and embryo development. Then, we focused on the role of DDX3X in cancer biology and systematically demonstrated its functions in various aspects of tumorigenesis and development. To provide a more intuitive understanding of the role of DDX3X in cancer, we summarized its functions and specific mechanisms in various types of cancer and presented its involvement in cancer-related signalling pathways.
Collapse
|
21
|
Wang Y, Li G, Deng M, Liu X, Huang W, Zhang Y, Liu M, Chen Y. The multifaceted functions of RNA helicases in the adaptive cellular response to hypoxia: From mechanisms to therapeutics. Pharmacol Ther 2020; 221:107783. [PMID: 33307143 DOI: 10.1016/j.pharmthera.2020.107783] [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: 09/27/2020] [Revised: 11/26/2020] [Accepted: 11/30/2020] [Indexed: 02/08/2023]
Abstract
Hypoxia is a hallmark of cancer. Hypoxia-inducible factor (HIF), a master player for sensing and adapting to hypoxia, profoundly influences genome instability, tumor progression and metastasis, metabolic reprogramming, and resistance to chemotherapies and radiotherapies. High levels and activity of HIF result in poor clinical outcomes in cancer patients. Thus, HIFs provide ideal therapeutic targets for cancers. However, HIF biology is sophisticated, and currently available HIF inhibitors have limited clinical utility owing to their low efficacy or side effects. RNA helicases, which are master players in cellular RNA metabolism, are usually highly expressed in tumors to meet the increased oncoprotein biosynthesis demand. Intriguingly, recent findings provide convincing evidence that RNA helicases are crucial for the adaptive cellular response to hypoxia via a mutual regulation with HIFs. More importantly, some RNA helicase inhibitors may suppress HIF signaling by blocking the translation of HIF-responsive genes. Therefore, RNA helicase inhibitors may work synergistically with HIF inhibitors in cancer to improve treatment efficacy. In this review, we discuss current knowledge of how cells sense and adapt to hypoxia through HIFs. However, our primary focus is on the multiple functions of RNA helicases in the adaptive response to hypoxia. We also highlight how these hypoxia-related RNA helicases can be exploited for anti-cancer therapeutics.
Collapse
Affiliation(s)
- Yijie Wang
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Guangqiang Li
- Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, Jinan University, Zhuhai, Guangdong 519000, China; Biomedical Translational Research Institute, Jinan University, Guangzhou, Guangdong 510632, China
| | - Mingxia Deng
- Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, Jinan University, Zhuhai, Guangdong 519000, China; Biomedical Translational Research Institute, Jinan University, Guangzhou, Guangdong 510632, China
| | - Xiong Liu
- School of Medicine, Jinan University, Guangzhou, Guangdong 510632, China
| | - Weixiao Huang
- School of Medicine, Jinan University, Guangzhou, Guangdong 510632, China
| | - Yao Zhang
- School of Medicine, Jinan University, Guangzhou, Guangdong 510632, China
| | - Min Liu
- Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, Shandong 250014, China
| | - Yan Chen
- Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, Jinan University, Zhuhai, Guangdong 519000, China; Biomedical Translational Research Institute, Jinan University, Guangzhou, Guangdong 510632, China; School of Medicine, Jinan University, Guangzhou, Guangdong 510632, China.
| |
Collapse
|
22
|
Hu Y, Ren S, He Y, Wang L, Chen C, Tang J, Liu W, Yu F. Possible Oncogenic Viruses Associated with Lung Cancer. Onco Targets Ther 2020; 13:10651-10666. [PMID: 33116642 PMCID: PMC7585805 DOI: 10.2147/ott.s263976] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 09/02/2020] [Indexed: 01/01/2023] Open
Abstract
Lung cancer is the most common cause of cancer death worldwide. Tobacco smoking is the most predominant etiology for lung cancer. However, only a small percentage of heavy smokers develop lung cancer, which suggests that other cofactors are required for lung carcinogenesis. Viruses have been central to modern cancer research and provide profound insights into cancer causes. Nevertheless, the role of virus in lung cancer is still unclear. In this article, we reviewed the possible oncogenic viruses associated with lung cancer.
Collapse
Affiliation(s)
- Yan Hu
- Department of Thoracic Surgery, The Thoracic Surgery Research Room, Second Xiangya Hospital, Central South University, Changsha 410011, People's Republic of China
| | - Siying Ren
- Department of Respiratory Medicine, Hunan Centre for Evidence-Based Medicine, Research Unit of Respiratory Diseases, Second Xiangya Hospital, Central South University, Changsha 410011, People's Republic of China
| | - Yu He
- Department of Thoracic Surgery, The Thoracic Surgery Research Room, Second Xiangya Hospital, Central South University, Changsha 410011, People's Republic of China
| | - Li Wang
- Department of Thoracic Surgery, The Thoracic Surgery Research Room, Second Xiangya Hospital, Central South University, Changsha 410011, People's Republic of China
| | - Chen Chen
- Department of Thoracic Surgery, The Thoracic Surgery Research Room, Second Xiangya Hospital, Central South University, Changsha 410011, People's Republic of China
| | - Jingqun Tang
- Department of Thoracic Surgery, The Thoracic Surgery Research Room, Second Xiangya Hospital, Central South University, Changsha 410011, People's Republic of China
| | - Wenliang Liu
- Department of Thoracic Surgery, The Thoracic Surgery Research Room, Second Xiangya Hospital, Central South University, Changsha 410011, People's Republic of China
| | - Fenglei Yu
- Department of Thoracic Surgery, The Thoracic Surgery Research Room, Second Xiangya Hospital, Central South University, Changsha 410011, People's Republic of China
| |
Collapse
|
23
|
Cardinal T, Bergeron KF, Soret R, Souchkova O, Faure C, Guillon A, Pilon N. Male-biased aganglionic megacolon in the TashT mouse model of Hirschsprung disease involves upregulation of p53 protein activity and Ddx3y gene expression. PLoS Genet 2020; 16:e1009008. [PMID: 32898154 PMCID: PMC7500598 DOI: 10.1371/journal.pgen.1009008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 09/18/2020] [Accepted: 07/23/2020] [Indexed: 02/07/2023] Open
Abstract
Hirschsprung disease (HSCR) is a complex genetic disorder of neural crest development resulting in incomplete formation of the enteric nervous system (ENS). This life-threatening neurocristopathy affects 1/5000 live births, with a currently unexplained male-biased ratio. To address this lack of knowledge, we took advantage of the TashT mutant mouse line, which is the only HSCR model to display a robust male bias. Our prior work revealed that the TashT insertional mutation perturbs a Chr.10 silencer-enriched non-coding region, leading to transcriptional dysregulation of hundreds of genes in neural crest-derived ENS progenitors of both sexes. Here, through sex-stratified transcriptome analyses and targeted overexpression in ENS progenitors, we show that male-biased ENS malformation in TashT embryos is not due to upregulation of Sry-the murine ortholog of a candidate gene for the HSCR male bias in humans-but instead involves upregulation of another Y-linked gene, Ddx3y. This discovery might be clinically relevant since we further found that the DDX3Y protein is also expressed in the ENS of a subset of male HSCR patients. Mechanistically, other data including chromosome conformation captured-based assays and CRISPR/Cas9-mediated deletions suggest that Ddx3y upregulation in male TashT ENS progenitors is due to increased transactivation by p53, which appears especially active in these cells yet without triggering apoptosis. Accordingly, in utero treatment of TashT embryos with the p53 inhibitor pifithrin-α decreased Ddx3y expression and abolished the otherwise more severe ENS defect in TashT males. Our data thus highlight novel pathogenic roles for p53 and DDX3Y during ENS formation in mice, a finding that might help to explain the intriguing male bias of HSCR in humans.
Collapse
Affiliation(s)
- Tatiana Cardinal
- Molecular Genetics of Development Laboratory, Département des Sciences Biologiques, Université du Québec à Montréal (UQAM), Montréal, Québec, Canada
- Centre d'excellence en recherche sur les maladies orphelines-Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montréal, Québec, Canada
| | - Karl-Frédérik Bergeron
- Centre d'excellence en recherche sur les maladies orphelines-Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montréal, Québec, Canada
- Lipid Metabolism Laboratory, Département des Sciences Biologiques, Université du Québec à Montréal (UQAM), Montréal, Québec, Canada
| | - Rodolphe Soret
- Molecular Genetics of Development Laboratory, Département des Sciences Biologiques, Université du Québec à Montréal (UQAM), Montréal, Québec, Canada
- Centre d'excellence en recherche sur les maladies orphelines-Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montréal, Québec, Canada
| | - Ouliana Souchkova
- Molecular Genetics of Development Laboratory, Département des Sciences Biologiques, Université du Québec à Montréal (UQAM), Montréal, Québec, Canada
- Centre d'excellence en recherche sur les maladies orphelines-Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montréal, Québec, Canada
| | - Christophe Faure
- Centre d'excellence en recherche sur les maladies orphelines-Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montréal, Québec, Canada
- Département de pédiatrie, Université de Montréal, Montréal, Québec, Canada
- Division de gastroentérologie, hépatologie et nutrition pédiatrique, Centre hospitalier universitaire Sainte-Justine, Montréal, Québec, Canada
| | - Amélina Guillon
- Molecular Genetics of Development Laboratory, Département des Sciences Biologiques, Université du Québec à Montréal (UQAM), Montréal, Québec, Canada
| | - Nicolas Pilon
- Molecular Genetics of Development Laboratory, Département des Sciences Biologiques, Université du Québec à Montréal (UQAM), Montréal, Québec, Canada
- Centre d'excellence en recherche sur les maladies orphelines-Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montréal, Québec, Canada
- Département de pédiatrie, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
24
|
Sun L, Zhong Y, Qiu W, Guo J, Gui L, Li M. MiR-26 regulates ddx3x expression in medaka (Oryzias latipes) gonads. Comp Biochem Physiol B Biochem Mol Biol 2020; 246-247:110456. [DOI: 10.1016/j.cbpb.2020.110456] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 05/07/2020] [Accepted: 05/11/2020] [Indexed: 10/24/2022]
|
25
|
DDX3X is Epigenetically Repressed in Renal Cell Carcinoma and Serves as a Prognostic Indicator and Therapeutic Target in Cancer Progression. Int J Mol Sci 2020; 21:ijms21082881. [PMID: 32326089 PMCID: PMC7215876 DOI: 10.3390/ijms21082881] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 04/16/2020] [Accepted: 04/17/2020] [Indexed: 12/17/2022] Open
Abstract
DEAD (Asp-Glu-Ala-Asp) box polypeptide 3, X-linked (DDX3X) is a member of the DEAD-box family of RNA helicases whose function has been revealed to be involved in RNA metabolism. Recent studies further indicate the abnormal expression in pan-cancers and the relevant biological effects on modulating cancer progression. However, DDX3X’s role in renal cell carcinoma (RCC) progression remains largely unknown. In this study, a medical informatics-based analysis using The Cancer Genome Atlas (TCGA) dataset was performed to evaluate clinical prognoses related to DDX3X. The results suggest that DDX3X is epigenetically repressed in tumor tissue and that lower DDX3X is correlated with the poor overall survival of RCC patients and high tumor size, lymph node metastasis, and distant metastasis (TNM staging system). Furthermore, knowledge-based transcriptomic analysis by Ingenuity Pathway Analysis (IPA) revealed that the SPINK1-metallothionein pathway is a top 1-repressed canonical signaling pathway by DDX3X. Furthermore, SPINK1 and the metallothionein gene family all serve as poor prognostic indicators, and the expression levels of those genes are inversely correlated with DDX3X in RCC. Furthermore, digoxin was identified via Connectivity Map analysis (L1000) for its capability to reverse gene signatures in patients with low DDX3X. Importantly, cancer cell proliferation and migration were decreased upon digoxin treatment in RCC cells. The results of this study indicate the significance of the DDX3Xlow/SPINK1high/metallothioneinhigh axis for predicting poor survival outcome in RCC patients and suggest digoxin as a precise and personalized compound for curing those patients with low DDX3X expression levels.
Collapse
|
26
|
Clinical and Prognostic Implications of P21 (WAF1/CIP1) Expression in Patients with Esophageal Cancer: A Systematic Review and Meta-Analysis. DISEASE MARKERS 2020; 2020:6520259. [PMID: 31998417 PMCID: PMC6970003 DOI: 10.1155/2020/6520259] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 10/22/2019] [Accepted: 11/15/2019] [Indexed: 02/06/2023]
Abstract
Background Previous studies have demonstrated that P21 (WAF1/CIP1) is a valuable prognostic factor in several malignant tumors. However, it is not known whether P21 can predict the prognosis in patients with esophageal cancer (EC). The aim of this research was to investigate the contribution of P21 expression to the clinicopathological characteristics and of EC. Methods A systematic review and meta-analysis of study focusing on P21 expression, clinicopathological characteristics, and clinical outcomes in patients with EC was performed using seven databases (PubMed, Embase, Web of Science, and four Chinese databases). Pooled hazard ratios and odds ratios were used to explore the association between P21 expression, clinicopathological characteristics, and outcomes in patients with EC. The heterogeneity of the studies was classified by the I 2 statistic. The sensitivity analysis was then utilized to assess the robustness of the results. Finally, the funnel plot and Begg's test were used to evaluate the publication bias. Results Forty-five studies with 3098 patients were eligible for inclusion in the meta-analysis. Thirty of these studies reported on clinicopathological characteristics and 15 on clinical outcomes. The pooled hazard ratio of 1.456 (95% confidence intervals 1.033-2.053, P = 0.032) for overall survival indicated that a low P21 expression level was an unfavorable prognostic factor for a clinical outcome in patients with EC. Furthermore, the pooled odds ratio confirmed an association between decreased P21 expression and poor clinicopathological characteristics, including differentiation, lymph node metastasis, invasion, and higher grade and clinical stage. Notably, high P21 expression was a significant predictor of a favorable response to chemotherapy. There was no evidence of publication bias. Conclusion Reduced P21 expression is associated with a poor outcome in patients with EC.
Collapse
|
27
|
Lin TC. DDX3X Multifunctionally Modulates Tumor Progression and Serves as a Prognostic Indicator to Predict Cancer Outcomes. Int J Mol Sci 2019; 21:ijms21010281. [PMID: 31906196 PMCID: PMC6982152 DOI: 10.3390/ijms21010281] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 12/27/2019] [Accepted: 12/28/2019] [Indexed: 12/22/2022] Open
Abstract
DEAD (Asp-Glu-Ala-Asp) box polypeptide 3, X-Linked (DDX3X), also known as DDX3, is one of the most widely studied and evolutionarily conserved members of the DEAD-box RNA helicase subfamily, and has been reported to participate in several cytosolic steps of mRNA metabolism. DDX3X facilitates the translation of specific targets via its helicase activity and regulates factors of the translation initiation complex. Emerging evidence illustrates the biological activities of DDX3X beyond its originally identified functions. The nonconventional regulatory effects include acting as a signaling adaptor molecule independent of enzymatic RNA remodeling, and DDX3X exhibits abnormal expression in cancers. DDX3X interacts with specific components to perform both oncogenic and tumor-suppressive roles in modulating tumor proliferation, migration, invasion, drug resistance, and cancer stemness in many types of cancers, indicating the need to unravel the associated molecular mechanisms. In this review article, we summarized and integrated current findings relevant to DDX3X in cancer research fields, cytokines and compounds modulating DDX3X's functions, and the released transcriptomic information and cancer patient clinical data from public databases. We found evidence for DDX3X having multiple impacts on cancer progression, and evaluated DDX3X expression levels in a pancancer panel and its associations with patient survival in each cancer-type cohort.
Collapse
Affiliation(s)
- Tsung-Chieh Lin
- Genomic Medicine Core Laboratory, Chang Gung Memorial Hospital, Linkou, Taiwan
| |
Collapse
|
28
|
Nguyen CN, Nguyen KVA, Eom S, Choi YJ, Kang LJ, Lee J, Kim C, Lee S, Lee SG, Lee JH. Regulation of p21 expression for anti-apoptotic activity of DDX3 against sanguinarine-induced cell death on intrinsic pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 65:153096. [PMID: 31568920 DOI: 10.1016/j.phymed.2019.153096] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Revised: 09/17/2019] [Accepted: 09/22/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND DDX3 plays a role in multicellular pathways, especially exerting an anti-apoptotic effect on extrinsic apoptosis. However, studies on the role of DDX3 in intrinsic apoptosis are lacking. PURPOSE In this study, we aimed to study the bio-function of DDX3 anti-apoptotic activity in the intrinsic pathway using HeLa cells treated with sanguinarine. STUDY DESIGN Screening of apoptosis-inducing agents found that sanguinarine was the most effective. After treatment with sanguinarine, cell viability, caspase-3 activity, and intrinsic gene expression were analyzed. FACS assays were used to analyze the effect of overexpression and knockdown of DDX3 to determine its role on intrinsic apoptosis. The relationship between DDX3 and the inhibition of p21 and apoptosis was investigated. RESULTS Sanguinarine was determined to be the most effective intrinsic apoptosis-inducing agent in HeLa cervical cancer cells. DDX3 upregulated anti-apoptotic gene expression (Bcl-xL, cyclin D1, cyclin E, and cyclin B1) and downregulated pro-apoptotic gene expression (caspase-3, Bax) after sanguinarine treatment. The apoptotic cell death rate increased from 8.74% (sanguinarine-treated control) to 17.6% after the knockdown of DDX3 but decreased to 5.29% after DDX3 overexpression. The results implied that p21 might be involved in the toxicity of sanguinarine to HeLa cells. Overexpression and knockdown of DDX3 under sanguinarine-treated conditions showed that DDX3 inhibited p21 expression in sanguinarine-treated HeLa cells. Notably, when we tested p21 expression among eight mutants located in the functional residues of DDX3 (S90A, S90E, T204A, T204E, GET, NEAD, LAT, and HRISR) under sanguinarine-treated conditions, only the S90E mutation in DDX3 had an effect on the inhibition of p21 expression and levels of pro-apoptotic genes (Bax and caspase-3) and anti-apoptotic genes (Bcl-xL, cyclin D1, cyclin E, and cyclin B1), as well as DDX3. CONCLUSION Taken together, the results suggest that the S90E residue is important for the regulation of p21 expression responsible for the anti-apoptotic activity of DDX3 in HeLa cells treated with sanguinarine. A model of the antiapoptotic function of DDX3 on sanguinarine-treated HeLa cells was proposed to understand the molecular mechanism of the intrinsic apoptosis inhibition in cervical cancer cells.
Collapse
Affiliation(s)
- Cam Ngoc Nguyen
- Department of Biotechnology, Chonnam National University, Address: #4-415, Gwangju 61886, Republic of Korea
| | - Khoa V A Nguyen
- Department of Biotechnology, Chonnam National University, Address: #4-415, Gwangju 61886, Republic of Korea
| | - Sanung Eom
- Department of Biotechnology, Chonnam National University, Address: #4-415, Gwangju 61886, Republic of Korea
| | - Yeo-Jin Choi
- Department of Biotechnology, Chonnam National University, Address: #4-415, Gwangju 61886, Republic of Korea
| | - Li-Jung Kang
- Department of Biotechnology, Chonnam National University, Address: #4-415, Gwangju 61886, Republic of Korea
| | - Jaeeun Lee
- Department of Biotechnology, Chonnam National University, Address: #4-415, Gwangju 61886, Republic of Korea
| | - Chaelin Kim
- Department of Biotechnology, Chonnam National University, Address: #4-415, Gwangju 61886, Republic of Korea
| | - Shinhui Lee
- Department of Biotechnology, Chonnam National University, Address: #4-415, Gwangju 61886, Republic of Korea
| | - Seong-Gene Lee
- Department of Biotechnology, Chonnam National University, Address: #4-415, Gwangju 61886, Republic of Korea.
| | - Jun-Ho Lee
- Department of Biotechnology, Chonnam National University, Address: #4-415, Gwangju 61886, Republic of Korea.
| |
Collapse
|
29
|
Cai Q, Cai J, Fang Y, Young KH. Epstein-Barr Virus-Positive Natural Killer/T-Cell Lymphoma. Front Oncol 2019; 9:386. [PMID: 31139570 PMCID: PMC6527808 DOI: 10.3389/fonc.2019.00386] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Accepted: 04/25/2019] [Indexed: 12/22/2022] Open
Abstract
Extranodal natural killer/T-cell lymphoma, nasal type (ENKL), is a rare malignancy of Non-Hodgkin lymphoma characterized by an aggressive clinical course and poor prognosis. It shows strong association with Epstein-Barr virus infection and occurs more commonly in Asia and Latin America. Various genetic alterations have been identified in ENKL by gene expression profiling and sequencing techniques. The frequent deletion of chromosome 6q21 was reported to lead to the silence of several tumor suppressor genes. Also, there have been novel genetic mutations that were recently uncovered and were found to frequently activate several oncogenic pathways, including the JAK/STAT, NF-κB, and MAPK pathways. Besides, we believe that deregulated single genes and epigenetic dysregulation might be relevant to the mechanism of this disease and thus, may have the potential to shed lights on the development of new therapeutic strategies. The consensus on the standard treatment for ENKL has not yet been currently established. For localized ENKL patients, radiotherapy with concurrent chemotherapy and sequential patterns of chemotherapy and radiotherapy are recommended as first-line therapy. As for advanced or relapsed/refractory ENKL patients, the application of non-anthracycline-containing regimens have significantly improved the clinical outcome, contributing to higher response rate, longer overall survival and progression-free survival. Hematopoietic stem cell transplantation is widely recommended for consolidation after a complete remission or partial remission has been achieved. The anti-programmed death 1 antibody, an immune checkpoint inhibitor, has demonstrated favorable results in treating relapsed or refractory ENKL. Of the current ENKL treatment, researchers are still striving to validate how radiotherapy and chemotherapy should be optimally combined and which of the non-anthracycline-containing regimens is superior. In this review, we summarize the main genetic alterations frequently found in ENKL and their role in providing new insights into the therapeutic targets of this disease, and highlight the recent findings regarding new biologic markers, novel therapeutic strategies applied to this intriguing neoplasm.
Collapse
Affiliation(s)
- Qingqing Cai
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jun Cai
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yu Fang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Ken H. Young
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
30
|
From the magic bullet to the magic target: exploiting the diverse roles of DDX3X in viral infections and tumorigenesis. Future Med Chem 2019; 11:1357-1381. [PMID: 30816053 DOI: 10.4155/fmc-2018-0451] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
DDX3X is an ATPase/RNA helicase of the DEAD-box family and one of the most multifaceted helicases known up to date, acting in RNA metabolism, cell cycle control, apoptosis, stress response and innate immunity. Depending on the virus or the viral cycle stage, DDX3X can act either in a proviral fashion or as an antiviral factor. Similarly, in different cancer types, it can act either as an oncogene or a tumor-suppressor gene. Accumulating evidence indicated that DDX3X can be considered a promising target for anticancer and antiviral chemotherapy, but also that its exploitation requires a deeper understanding of the molecular mechanisms underlying its dual role in cancer and viral infections. In this Review, we will summarize the known roles of DDX3X in different tumor types and viral infections, and the different inhibitors available, illustrating the possible advantages and potential caveats of their use as anticancer and antiviral drugs.
Collapse
|
31
|
Yang F, Fang E, Mei H, Chen Y, Li H, Li D, Song H, Wang J, Hong M, Xiao W, Wang X, Huang K, Zheng L, Tong Q. Cis-Acting circ-CTNNB1 Promotes β-Catenin Signaling and Cancer Progression via DDX3-Mediated Transactivation of YY1. Cancer Res 2018; 79:557-571. [PMID: 30563889 DOI: 10.1158/0008-5472.can-18-1559] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 09/27/2018] [Accepted: 12/11/2018] [Indexed: 11/16/2022]
Abstract
Circular RNAs (circRNA), a subclass of noncoding RNA characterized by covalently closed continuous loops, play emerging roles in tumorigenesis and aggressiveness. However, the functions and underlying mechanisms of circRNA in regulating Wnt/β-catenin signaling and cancer progression remain elusive. Here, we screen cis-acting circRNA generated by β-catenin (CTNNB1)/transcription factor 7-like 2 genes and identify one intronic circRNA derived from CTNNB1 (circ-CTNNB1) as a novel driver of cancer progression. Circ-CTNNB1 was predominantly expressed in the nucleus, upregulated in cancer tissues and cell lines, and associated with unfavorable outcomes in patients with cancer. Circ-CTNNB1 promoted β-catenin activation, growth, invasion, and metastasis in cancer cells. Circ-CTNNB1 bound DEAD-box polypeptide 3 (DDX3) to facilitate its physical interaction with transcription factor Yin Yang 1 (YY1), resulting in the transactivation of YY1 and transcriptional alteration of downstream genes associated with β-catenin activation and cancer progression. Preclinically, administration of lentivirus-mediated short hairpin RNA targeting circ-CTNNB1 or a cell-penetrating inhibitory peptide blocking the circ-CTNNB1-DDX3 interaction inhibited downstream gene expression, tumorigenesis, and aggressiveness in cancer cells. Taken together, these results demonstrate cis-acting circ-CTNNB1 as a mediator of β-catenin signaling and cancer progression through DDX3-mediated transactivation of YY1. SIGNIFICANCE: These findings reveal the oncogenic functions of a cis-acting circular RNA in β-catenin activation and cancer progression, with potential value as a therapeutic target for human cancers.
Collapse
Affiliation(s)
- Feng Yang
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P. R. China
| | - Erhu Fang
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P. R. China
| | - Hong Mei
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P. R. China
| | - Yajun Chen
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P. R. China
| | - Huanhuan Li
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P. R. China
| | - Dan Li
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P. R. China
| | - Huajie Song
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P. R. China
| | - Jianqun Wang
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P. R. China
| | - Mei Hong
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P. R. China
| | - Wenjing Xiao
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P. R. China
| | - Xiaojing Wang
- Clinical Center of Human Genomic Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P. R. China
| | - Kai Huang
- Clinical Center of Human Genomic Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P. R. China
| | - Liduan Zheng
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P. R. China. .,Clinical Center of Human Genomic Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P. R. China
| | - Qiangsong Tong
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P. R. China. .,Clinical Center of Human Genomic Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P. R. China
| |
Collapse
|
32
|
Zheng B, Liu F, Zeng L, Geng L, Ouyang X, Wang K, Huang Q. Overexpression of Pyruvate Kinase Type M2 (PKM2) Promotes Ovarian Cancer Cell Growth and Survival Via Regulation of Cell Cycle Progression Related with Upregulated CCND1 and Downregulated CDKN1A Expression. Med Sci Monit 2018; 24:3103-3112. [PMID: 29752805 PMCID: PMC5973491 DOI: 10.12659/msm.907490] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 10/31/2017] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Many findings have shown that pyruvate kinase type M2 (PKM2) plays crucial roles in regulating the occurrence and development of various human cancers; however, its roles in ovarian cancer oncogenesis remain to be determined. MATERIAL AND METHODS The expression intensity of PKM2 in ovarian cancer tissues was examined by immunohistochemistry (IHC), and was then correlated to patient clinicopathologic characteristics. The roles of PKM2 in ovarian cancer cell proliferation, growth, and survival were examined by CCK-8, colony forming, and flow cytometry assays. The potentially involved molecular were then investigated by Western blot analysis. RESULTS IHC results showed that PKM2 was overexpressed in 100 of 114 (87.7%) serous ovarian cancer tissues as compared with 50 cases of non-cancerous ovarian tissues, and was associated with tumor size ≥7.5 cm and <7.5 cm (p<0.05). Overexpression of PKM2 in SKOV3 and HEY ovarian cancer cells by transfection with PKM2 lentivirus vector led to increased cell proliferation, growth, and survival, which may be related with PKM2 being able to increase cell cycle progress: G1 stage decreased, whereas S stage significantly increased. In contrast, all functions of SKOV3 and HEY cells described above were reversed by knocked down PKM2 expression using siRNA. Further data showed that overexpressed PKM2 led to increased CCND1 and decreased CDKN1A expression, whereas underexpressed PKM2 led to decreased CCND1 and increased CDKN1A expression in ovarian cancer cells. CONCLUSIONS PKM2 may play important roles in ovarian cancer development and may be a treatment target for this cancer.
Collapse
Affiliation(s)
- Bin Zheng
- The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, P.R. China
| | - Fangfang Liu
- The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, P.R. China
| | - Li Zeng
- The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, P.R. China
| | - Li Geng
- The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, P.R. China
| | - Xiaojuan Ouyang
- The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, P.R. China
| | - Kai Wang
- The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, P.R. China
| | - Qiaojia Huang
- The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, P.R. China
- Department of Experimental Medicine, Fuzhou General Hospital, Fuzhou, Fujian, P.R. China
| |
Collapse
|
33
|
Wu CC, Wu DW, Lin YY, Lin PL, Lee H. Hepatitis B virus X protein represses LKB1 expression to promote tumor progression and poor postoperative outcome in hepatocellular carcinoma. Surgery 2018; 163:1040-1046. [PMID: 29475611 DOI: 10.1016/j.surg.2017.11.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 10/29/2017] [Accepted: 11/24/2017] [Indexed: 01/04/2023]
Abstract
BACKGROUND Hepatitis B virus X (HBx) protein plays critical roles in hepatitis B virus (HBV)-associated hepatocellular tumorigenesis through different molecular mechanisms, including inactivation of p53, a key transcription factor of liver kinase B1 (LKB1). We hypothesized that p53 inactivation by HBx protein could decrease LKB1 expression, thereby promoting tumor progression and poor outcomes in patients with HBV-associated hepatocellular carcinoma. METHODS Manipulation strategies for HBx protein and/or p53 were used to verify that loss of LKB1 could promote colony formation and invasiveness in HepG2 and Hep3B cells. The expressions of HBx protein and LKB1 in 93 hepatocellular carcinomas (HCC) were also evaluated by immunohistochemistry. Kaplan-Meier and Cox regression models were used to assess the prognostic value of both HBx protein and LKB1 proteins in patients with hepatocellular carcinoma. RESULTS Mechanistically, LKB1 expression was decreased at the transcriptional level after inactivation of p53 by HBx protein. Decreases in LKB1 expression were also associated with HBx protein-mediated colony formation and invasive capabilities. HBx protein, LKB1, and a combination of both proteins had prognostic significance for overall survival and relapse-free survival in our study population. CONCLUSION The results from cell line experiments and evaluation of patient prognosis according to expression of HBx protein and LKB1 in their HCC strongly support the hypothesis that decreases in LKB1 expression by HBx protein-mediated p53 inactivation may play an important role in HBV-associated hepatocellular tumorigenesis.
Collapse
Affiliation(s)
- Cheng-Chung Wu
- Department of General Surgery, Taichung Veterans General Hospital, Taichung, Taiwan
| | - De-Wei Wu
- Graduate Institute of Cancer Biology and Drug Discovery, Taipei Medical University, Taipei, Taiwan
| | - Ying-Yu Lin
- Graduate Institute of Cancer Biology and Drug Discovery, Taipei Medical University, Taipei, Taiwan
| | - Po-Lin Lin
- Graduate Institute of Cancer Biology and Drug Discovery, Taipei Medical University, Taipei, Taiwan
| | - Huei Lee
- Graduate Institute of Cancer Biology and Drug Discovery, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
34
|
Lin Y, Wang X, Yu Y, Liu W, Xie F, Ouyang X, Huang Q. Expression and prognostic significance of cyclin-dependent kinase inhibitor 1A in patients with resected gastric adenocarcinoma. Oncol Lett 2018; 14:7473-7482. [PMID: 29344191 DOI: 10.3892/ol.2017.7107] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 07/17/2017] [Indexed: 02/07/2023] Open
Abstract
Cyclin-dependent kinase inhibitor 1A (CDKN1A) is an important cell cycleregulator, and has been identified to exhibit aberrant expression in various types of cancer tissues. However, the association between CDKN1A expression level and prognosis in patients with resected gastric adenocarcinoma (RGA) requires additional elucidation. In the present study, the CDKN1A expression profile in RGA tissues obtained from 217 patients were analyzed using immunohistochemistry. Its prognostic significance was evaluated by using the χ2 test, Kaplan-Meier curves and the log-rank test, and a multivariate Cox model analysis, during a median follow-up time of 51 months. The results demonstrated that CDKN1A expression was significantly correlated with lymph node metastasis (LNM; P=0.001), recurrence (P<0.001) and overall survival (OS; P<0.001). In addition, the recurrence-free survival (RFS) and OS times were significantly shorter in patients with low CDKN1A expression compared with those with high CDKN1A expression (RFS, 20 months vs. 69 months, P<0.001; and OS, 32 months vs. 70 months, P<0.001, respectively). Multivariate analysis additionally confirmed that low CDKN1A expression was significantly correlated with an increased risk of LNM (P=0.001), recurrence (P<0.001) and mortality (P<0.001). Therefore, these data suggest that low expression of CDKN1A has independent prognostic significance indicative of tumor progression and poor survival in patients with RGA. Evaluation of CDKN1A expression may assist in determining prognosis in patients with RGA.
Collapse
Affiliation(s)
- Youdong Lin
- Department of Experimental Medicine, Fuzhou General Hospital, Fuzong Clinical College of Fujian Medical University, Fuzhou, Fujian 350025, P.R. China
| | - Xiaoting Wang
- Department of Experimental Medicine, Fuzhou General Hospital, Fuzong Clinical College of Fujian Medical University, Fuzhou, Fujian 350025, P.R. China
| | - Yinghao Yu
- Department of Pathology, Fuzhou General Hospital, Fuzong Clinical College of Fujian Medical University, Fuzhou, Fujian 350025, P.R. China
| | - Wei Liu
- Department of Pathology, Fuzhou General Hospital, Fuzong Clinical College of Fujian Medical University, Fuzhou, Fujian 350025, P.R. China
| | - Feilai Xie
- Department of Pathology, Fuzhou General Hospital, Fuzong Clinical College of Fujian Medical University, Fuzhou, Fujian 350025, P.R. China
| | - Xuenong Ouyang
- Department of Oncology, Fuzhou General Hospital, Fuzong Clinical College of Fujian Medical University, Fuzhou, Fujian 350025, P.R. China
| | - Qiaojia Huang
- Department of Experimental Medicine, Fuzhou General Hospital, Fuzong Clinical College of Fujian Medical University, Fuzhou, Fujian 350025, P.R. China
| |
Collapse
|
35
|
Tung MC, Lin PL, Cheng YW, Wu DW, Yeh SD, Chen CY, Lee H. Reduction of microRNA-184 by E6 oncoprotein confers cisplatin resistance in lung cancer via increasing Bcl-2. Oncotarget 2017; 7:32362-74. [PMID: 27083050 PMCID: PMC5078019 DOI: 10.18632/oncotarget.8708] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 03/28/2016] [Indexed: 12/04/2022] Open
Abstract
MicroRNA-184 suppresses cell growth and survival via targeting c-Myc and Bcl- 2. We recently reported that miR-184 promotes tumor progression in non-small cell lung cancer via targeting CDC25A and c-Myc. We here hypothesized that miR-184 could be down-regulated by E6 oncoprotein to confer cisplatin resistance in NSCLC. Human papillomavirus (HPV) 16-positive lung cancer TL-1 and cervical cancer SiHa cells compared with HPV16-negative TL-10 and C33A cells were enrolled for E6 manipulation. MiR-184 expression levels were increased by E6-knockdown in TL-1 and SiHa cells, but decreased by E6-overexpression in TL-10 and C33A cells. The MTT assay showed that the inhibition concentration of cisplatin yielding for 50% cell viability was dependent on miR-184 levels. Bcl-2 de-targeted by E6-mediated miR- 184 reduction was responsible for cisplatin resistance. Luciferase reporter assay and real- time PCR analysis indicated that the miR-184 promoter activity and its expression were modulated by E6 and/or p53 manipulation. Chromatin immunoprecipitation (ChIP) assay confirmed that p53 was bound onto the miR-184 promoter and its binding activity was modulated by E6 and/or p53 manipulation. Among patients, high miR184 and high Bcl-2 mRNA expression was more commonly occurred in E6- positive tumors than in E6-negative tumors. Fifty-nine out of 136 patients receiving cisplatin-based chemotherapy were available for the retrospective study. Patients with low-mR-184, E6-positive, high-Bcl-2 tumors, and both combinations were more prevalently occurred unfavorable response to cisplatin-based chemotherapy than their counterparts. In conclusion, a decrease in miR-184 level by E6 oncoprotein may predict unfavorable response to cisplatin-based chemotherapy in HPV-infected NSCLC patients via increasing Bcl-2 expression.
Collapse
Affiliation(s)
- Min-Che Tung
- Graduate Institute of Clinical Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Surgery, Tung's Taichung Metro-Harbor Hospital, Taichung, Taiwan
| | - Po-Lin Lin
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Ya-Wen Cheng
- Graduate Institute of Cancer Biology and Drug Discovery, Taipei Medical University, Taipei, Taiwan
| | - De-Wei Wu
- Graduate Institute of Cancer Biology and Drug Discovery, Taipei Medical University, Taipei, Taiwan
| | - Sauh-Der Yeh
- Graduate Institute of Clinical Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chi-Yi Chen
- Department of Surgery, Chung Shan Medical University, Taichung, Taiwan
| | - Huei Lee
- Graduate Institute of Cancer Biology and Drug Discovery, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
36
|
Wu DW, Lin PL, Cheng YW, Huang CC, Wang L, Lee H. DDX3 enhances oncogenic KRAS‑induced tumor invasion in colorectal cancer via the β‑catenin/ZEB1 axis. Oncotarget 2017; 7:22687-99. [PMID: 27007150 PMCID: PMC5008392 DOI: 10.18632/oncotarget.8143] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Accepted: 02/21/2016] [Indexed: 12/22/2022] Open
Abstract
DDX3 plays a dual role in colorectal cancer; however, the role and underlying mechanism of DDX3 in colorectal tumorigenesis remains unclear. Here, we provide evidence that DDX3 enhances oncogenic KRAS transcription via an increase in SP1 binding to its promoter. Accelerating oncogenic KRAS expression by DDX3 promotes the invasion capability via the ERK/PTEN/AKT/β-catenin cascade. Moreover, the β-catenin/ZEB1 axis is responsible for DDX3-induced cell invasiveness and xenograft lung tumor nodule formation. The xenograft lung tumor nodules induced by DDX3-overexpressing T84 stable clone were nearly suppressed by the inhibitor of AKT (perifosine) or β-catenin (XAV939). Among patients, high KRAS, positive nuclear β-catenin expression and high ZEB1 were more commonly occurred in high-DDX3 tumors than in low-DDX3 tumors. High-DDX3, high-KRAS, positive nuclear β-catenin tumors, and high-ZEB1 exhibited worse overall survival (OS) and relapse free survival (RFS) than their counterparts. In conclusion, DDX3 may play an oncogenic role to promote tumor growth and invasion in colon cancer cells via the β-catenin/ZEB1 axis due to increasing KRAS transcription. We therefore suggest that AKT or β-catenin may potentially act as a therapeutic target to improve tumor regression and outcomes in colorectal cancer patients who harbored high-DDX3 tumors.
Collapse
Affiliation(s)
- De-Wei Wu
- Graduate Institute of Cancer Biology and Drug Discovery, Taipei Medical University, Taipei, Taiwan
| | - Po-Lin Lin
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Ya-Wen Cheng
- Graduate Institute of Cancer Biology and Drug Discovery, Taipei Medical University, Taipei, Taiwan
| | - Chi-Chou Huang
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Surgery, Division of Colon and Rectum, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Lee Wang
- School of Public Health, Chung Shan Medical University, Taichung, Taiwan
| | - Huei Lee
- Graduate Institute of Cancer Biology and Drug Discovery, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
37
|
Lin PL, Wu TC, Wu DW, Wang L, Chen CY, Lee H. An increase in BAG-1 by PD-L1 confers resistance to tyrosine kinase inhibitor in non–small cell lung cancer via persistent activation of ERK signalling. Eur J Cancer 2017; 85:95-105. [DOI: 10.1016/j.ejca.2017.07.025] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Accepted: 07/14/2017] [Indexed: 01/08/2023]
|
38
|
Heerma van Voss MR, van Diest PJ, Raman V. Targeting RNA helicases in cancer: The translation trap. Biochim Biophys Acta Rev Cancer 2017; 1868:510-520. [PMID: 28965870 DOI: 10.1016/j.bbcan.2017.09.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 09/25/2017] [Accepted: 09/26/2017] [Indexed: 12/14/2022]
Abstract
Cancer cells are reliant on the cellular translational machinery for both global elevation of protein synthesis and the translation of specific mRNAs that promote tumor cell survival. Targeting translational control in cancer is therefore increasingly recognized as a promising therapeutic strategy. In this regard, DEAD/H box RNA helicases are a very interesting group of proteins, with several family members regulating mRNA translation in cancer cells. In this review, we delineate the mechanisms by which DEAD/H box proteins modulate oncogenic translation and how inhibition of these RNA helicases can be exploited for anti-cancer therapeutics.
Collapse
Affiliation(s)
- Marise R Heerma van Voss
- Department of Radiology and Radiological Sciences, Johns Hopkins University, School of Medicine, Baltimore, MD, USA; Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Paul J van Diest
- Department of Radiology and Radiological Sciences, Johns Hopkins University, School of Medicine, Baltimore, MD, USA; Department of Oncology, Johns Hopkins University, School of Medicine, MD, USA
| | - Venu Raman
- Department of Radiology and Radiological Sciences, Johns Hopkins University, School of Medicine, Baltimore, MD, USA; Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands; Department of Oncology, Johns Hopkins University, School of Medicine, MD, USA.
| |
Collapse
|
39
|
Lin PL, Cheng YM, Wu DW, Huang YJ, Lin HC, Chen CY, Lee H. A combination of anti-PD-L1 mAb plus Lm-LLO-E6 vaccine efficiently suppresses tumor growth and metastasis in HPV-infected cancers. Cancer Med 2017; 6:2052-2062. [PMID: 28795532 PMCID: PMC5603833 DOI: 10.1002/cam4.1143] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 06/18/2017] [Accepted: 06/21/2017] [Indexed: 01/08/2023] Open
Abstract
PD‐1/PD‐L1 immunotherapy is viewed as having clinical benefits in advanced cancers but is effective in only a few patients, suggesting that an efficient combination approach is needed to improve efficacy. Immunohistochemistry analysis indicated that PD‐L1 expression was correlated with the E6 expression in tumors from 122 lung cancer patients. The poorest survival occurred in PD‐L1‐positive/E6‐positive tumor. PD‐L1 expression was increased by the expression of E6, but not the E7, oncoprotein in lung and cervical cancer cells. PD‐L1 expression was responsible for E6‐mediated colony formation and soft agar growth. Therefore, PD‐L1 secreted from tumor cells may directly promote tumor progression, particularly in E6‐positive tumors. Immune deficiency nude mice were used to test the possibility that combining anti‐PD‐L1 mAb with Lm‐LLO‐E6 vaccine could have a higher antitumor activity compared with anti‐PD‐L1 mAb or Lm‐LLO‐E6 vaccine alone. A greater antitumor activity was obtained with anti‐PD‐L1 mAb + Lm‐LLO‐E6 vaccine than with anti‐PD‐L1 mAb or Lm‐LLO‐E6 alone in subcutaneous and metastatic tumors induced by TL‐1 and SiHa cells. The longest survival time for nude mice was observed in the anti‐PD‐L1 mAb + Lm‐LLO‐E6 vaccine group. In conclusion, an anti‐PD‐L1 mAb + Lm‐LLO‐E6 vaccine may be an efficient treatment for suppression of tumor growth and metastasis induced by HPV‐infected cells.
Collapse
Affiliation(s)
- Po-Lin Lin
- Graduate Institute of Cancer Biology and Drug Discovery, Taipei Medical University, Taipei, Taiwan
| | - Ya-Min Cheng
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, Tainan, Taiwan
| | - De-Wei Wu
- Graduate Institute of Cancer Biology and Drug Discovery, Taipei Medical University, Taipei, Taiwan
| | | | | | - Chih-Yi Chen
- Department of Surgery, Chung Shan Medical Hospital, Taichung, Taiwan
| | - Huei Lee
- Graduate Institute of Cancer Biology and Drug Discovery, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
40
|
Cai W, Xiong Chen Z, Rane G, Satendra Singh S, Choo Z, Wang C, Yuan Y, Zea Tan T, Arfuso F, Yap CT, Pongor LS, Yang H, Lee MB, Cher Goh B, Sethi G, Benoukraf T, Tergaonkar V, Prem Kumar A. Wanted DEAD/H or Alive: Helicases Winding Up in Cancers. J Natl Cancer Inst 2017; 109:2957323. [PMID: 28122908 DOI: 10.1093/jnci/djw278] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 09/08/2016] [Accepted: 10/20/2016] [Indexed: 12/23/2022] Open
Abstract
Cancer is one of the most studied areas of human biology over the past century. Despite having attracted much attention, hype, and investments, the search to find a cure for cancer remains an uphill battle. Recent discoveries that challenged the central dogma of molecular biology not only further increase the complexity but also demonstrate how various types of noncoding RNAs such as microRNA and long noncoding RNA, as well as their related processes such as RNA editing, are important in regulating gene expression. Parallel to this aspect, an increasing number of reports have focused on a family of proteins known as DEAD/H-box helicases involved in RNA metabolism, regulation of long and short noncoding RNAs, and novel roles as "editing helicases" and their association with cancers. This review summarizes recent findings on the roles of RNA helicases in various cancers, which are broadly classified into adult solid tumors, childhood solid tumors, leukemia, and cancer stem cells. The potential small molecule inhibitors of helicases and their therapeutic value are also discussed. In addition, analyzing next-generation sequencing data obtained from public portals and reviewing existing literature, we provide new insights on the potential of DEAD/H-box helicases to act as pharmacological drug targets in cancers.
Collapse
Affiliation(s)
- Wanpei Cai
- Affiliations of authors: Cancer Science Institute of Singapore, National University of Singapore, Singapore (WC, GR, SSS, CW, YY, TZT, HY, BCG, TB, APK); Departments of Pharmacology (WC, GR, SSS, CW, BCG, GS, APK), Physiology (ZXC, ZC, CTY), and Biochemistry (VT), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; KK Women's and Children's Hospital, Singapore (ZXC); Stem Cell and Cancer Biology Laboratory (FA), School of Biomedical Sciences (GS, APK), Curtin Health Innovation Research Institute, Curtin Medical School (APK), Curtin University, Perth, WA, Australia; National University Cancer Institute, National University Health System, Singapore (CTY, BCG, APK); 2 Department of Pediatrics, Semmelweis University, Budapest, Hungary (LSP); MTA TTK Lendület Cancer Biomarker Research Group, Research Centre for Natural Sciences, Budapest, Hungary (LSP); Department of Renal Medicine (MBL) and Department of Haematology-Oncology (BCG), National University Health System, Singapore; Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore (VT); Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia (VT); Department of Biological Sciences, University of North Texas, Denton, TX (APK)
| | - Zhi Xiong Chen
- Affiliations of authors: Cancer Science Institute of Singapore, National University of Singapore, Singapore (WC, GR, SSS, CW, YY, TZT, HY, BCG, TB, APK); Departments of Pharmacology (WC, GR, SSS, CW, BCG, GS, APK), Physiology (ZXC, ZC, CTY), and Biochemistry (VT), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; KK Women's and Children's Hospital, Singapore (ZXC); Stem Cell and Cancer Biology Laboratory (FA), School of Biomedical Sciences (GS, APK), Curtin Health Innovation Research Institute, Curtin Medical School (APK), Curtin University, Perth, WA, Australia; National University Cancer Institute, National University Health System, Singapore (CTY, BCG, APK); 2 Department of Pediatrics, Semmelweis University, Budapest, Hungary (LSP); MTA TTK Lendület Cancer Biomarker Research Group, Research Centre for Natural Sciences, Budapest, Hungary (LSP); Department of Renal Medicine (MBL) and Department of Haematology-Oncology (BCG), National University Health System, Singapore; Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore (VT); Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia (VT); Department of Biological Sciences, University of North Texas, Denton, TX (APK)
| | - Grishma Rane
- Affiliations of authors: Cancer Science Institute of Singapore, National University of Singapore, Singapore (WC, GR, SSS, CW, YY, TZT, HY, BCG, TB, APK); Departments of Pharmacology (WC, GR, SSS, CW, BCG, GS, APK), Physiology (ZXC, ZC, CTY), and Biochemistry (VT), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; KK Women's and Children's Hospital, Singapore (ZXC); Stem Cell and Cancer Biology Laboratory (FA), School of Biomedical Sciences (GS, APK), Curtin Health Innovation Research Institute, Curtin Medical School (APK), Curtin University, Perth, WA, Australia; National University Cancer Institute, National University Health System, Singapore (CTY, BCG, APK); 2 Department of Pediatrics, Semmelweis University, Budapest, Hungary (LSP); MTA TTK Lendület Cancer Biomarker Research Group, Research Centre for Natural Sciences, Budapest, Hungary (LSP); Department of Renal Medicine (MBL) and Department of Haematology-Oncology (BCG), National University Health System, Singapore; Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore (VT); Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia (VT); Department of Biological Sciences, University of North Texas, Denton, TX (APK)
| | - Shikha Satendra Singh
- Affiliations of authors: Cancer Science Institute of Singapore, National University of Singapore, Singapore (WC, GR, SSS, CW, YY, TZT, HY, BCG, TB, APK); Departments of Pharmacology (WC, GR, SSS, CW, BCG, GS, APK), Physiology (ZXC, ZC, CTY), and Biochemistry (VT), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; KK Women's and Children's Hospital, Singapore (ZXC); Stem Cell and Cancer Biology Laboratory (FA), School of Biomedical Sciences (GS, APK), Curtin Health Innovation Research Institute, Curtin Medical School (APK), Curtin University, Perth, WA, Australia; National University Cancer Institute, National University Health System, Singapore (CTY, BCG, APK); 2 Department of Pediatrics, Semmelweis University, Budapest, Hungary (LSP); MTA TTK Lendület Cancer Biomarker Research Group, Research Centre for Natural Sciences, Budapest, Hungary (LSP); Department of Renal Medicine (MBL) and Department of Haematology-Oncology (BCG), National University Health System, Singapore; Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore (VT); Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia (VT); Department of Biological Sciences, University of North Texas, Denton, TX (APK)
| | - Zhang'e Choo
- Affiliations of authors: Cancer Science Institute of Singapore, National University of Singapore, Singapore (WC, GR, SSS, CW, YY, TZT, HY, BCG, TB, APK); Departments of Pharmacology (WC, GR, SSS, CW, BCG, GS, APK), Physiology (ZXC, ZC, CTY), and Biochemistry (VT), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; KK Women's and Children's Hospital, Singapore (ZXC); Stem Cell and Cancer Biology Laboratory (FA), School of Biomedical Sciences (GS, APK), Curtin Health Innovation Research Institute, Curtin Medical School (APK), Curtin University, Perth, WA, Australia; National University Cancer Institute, National University Health System, Singapore (CTY, BCG, APK); 2 Department of Pediatrics, Semmelweis University, Budapest, Hungary (LSP); MTA TTK Lendület Cancer Biomarker Research Group, Research Centre for Natural Sciences, Budapest, Hungary (LSP); Department of Renal Medicine (MBL) and Department of Haematology-Oncology (BCG), National University Health System, Singapore; Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore (VT); Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia (VT); Department of Biological Sciences, University of North Texas, Denton, TX (APK)
| | - Chao Wang
- Affiliations of authors: Cancer Science Institute of Singapore, National University of Singapore, Singapore (WC, GR, SSS, CW, YY, TZT, HY, BCG, TB, APK); Departments of Pharmacology (WC, GR, SSS, CW, BCG, GS, APK), Physiology (ZXC, ZC, CTY), and Biochemistry (VT), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; KK Women's and Children's Hospital, Singapore (ZXC); Stem Cell and Cancer Biology Laboratory (FA), School of Biomedical Sciences (GS, APK), Curtin Health Innovation Research Institute, Curtin Medical School (APK), Curtin University, Perth, WA, Australia; National University Cancer Institute, National University Health System, Singapore (CTY, BCG, APK); 2 Department of Pediatrics, Semmelweis University, Budapest, Hungary (LSP); MTA TTK Lendület Cancer Biomarker Research Group, Research Centre for Natural Sciences, Budapest, Hungary (LSP); Department of Renal Medicine (MBL) and Department of Haematology-Oncology (BCG), National University Health System, Singapore; Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore (VT); Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia (VT); Department of Biological Sciences, University of North Texas, Denton, TX (APK)
| | - Yi Yuan
- Affiliations of authors: Cancer Science Institute of Singapore, National University of Singapore, Singapore (WC, GR, SSS, CW, YY, TZT, HY, BCG, TB, APK); Departments of Pharmacology (WC, GR, SSS, CW, BCG, GS, APK), Physiology (ZXC, ZC, CTY), and Biochemistry (VT), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; KK Women's and Children's Hospital, Singapore (ZXC); Stem Cell and Cancer Biology Laboratory (FA), School of Biomedical Sciences (GS, APK), Curtin Health Innovation Research Institute, Curtin Medical School (APK), Curtin University, Perth, WA, Australia; National University Cancer Institute, National University Health System, Singapore (CTY, BCG, APK); 2 Department of Pediatrics, Semmelweis University, Budapest, Hungary (LSP); MTA TTK Lendület Cancer Biomarker Research Group, Research Centre for Natural Sciences, Budapest, Hungary (LSP); Department of Renal Medicine (MBL) and Department of Haematology-Oncology (BCG), National University Health System, Singapore; Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore (VT); Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia (VT); Department of Biological Sciences, University of North Texas, Denton, TX (APK)
| | - Tuan Zea Tan
- Affiliations of authors: Cancer Science Institute of Singapore, National University of Singapore, Singapore (WC, GR, SSS, CW, YY, TZT, HY, BCG, TB, APK); Departments of Pharmacology (WC, GR, SSS, CW, BCG, GS, APK), Physiology (ZXC, ZC, CTY), and Biochemistry (VT), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; KK Women's and Children's Hospital, Singapore (ZXC); Stem Cell and Cancer Biology Laboratory (FA), School of Biomedical Sciences (GS, APK), Curtin Health Innovation Research Institute, Curtin Medical School (APK), Curtin University, Perth, WA, Australia; National University Cancer Institute, National University Health System, Singapore (CTY, BCG, APK); 2 Department of Pediatrics, Semmelweis University, Budapest, Hungary (LSP); MTA TTK Lendület Cancer Biomarker Research Group, Research Centre for Natural Sciences, Budapest, Hungary (LSP); Department of Renal Medicine (MBL) and Department of Haematology-Oncology (BCG), National University Health System, Singapore; Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore (VT); Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia (VT); Department of Biological Sciences, University of North Texas, Denton, TX (APK)
| | - Frank Arfuso
- Affiliations of authors: Cancer Science Institute of Singapore, National University of Singapore, Singapore (WC, GR, SSS, CW, YY, TZT, HY, BCG, TB, APK); Departments of Pharmacology (WC, GR, SSS, CW, BCG, GS, APK), Physiology (ZXC, ZC, CTY), and Biochemistry (VT), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; KK Women's and Children's Hospital, Singapore (ZXC); Stem Cell and Cancer Biology Laboratory (FA), School of Biomedical Sciences (GS, APK), Curtin Health Innovation Research Institute, Curtin Medical School (APK), Curtin University, Perth, WA, Australia; National University Cancer Institute, National University Health System, Singapore (CTY, BCG, APK); 2 Department of Pediatrics, Semmelweis University, Budapest, Hungary (LSP); MTA TTK Lendület Cancer Biomarker Research Group, Research Centre for Natural Sciences, Budapest, Hungary (LSP); Department of Renal Medicine (MBL) and Department of Haematology-Oncology (BCG), National University Health System, Singapore; Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore (VT); Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia (VT); Department of Biological Sciences, University of North Texas, Denton, TX (APK)
| | - Celestial T Yap
- Affiliations of authors: Cancer Science Institute of Singapore, National University of Singapore, Singapore (WC, GR, SSS, CW, YY, TZT, HY, BCG, TB, APK); Departments of Pharmacology (WC, GR, SSS, CW, BCG, GS, APK), Physiology (ZXC, ZC, CTY), and Biochemistry (VT), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; KK Women's and Children's Hospital, Singapore (ZXC); Stem Cell and Cancer Biology Laboratory (FA), School of Biomedical Sciences (GS, APK), Curtin Health Innovation Research Institute, Curtin Medical School (APK), Curtin University, Perth, WA, Australia; National University Cancer Institute, National University Health System, Singapore (CTY, BCG, APK); 2 Department of Pediatrics, Semmelweis University, Budapest, Hungary (LSP); MTA TTK Lendület Cancer Biomarker Research Group, Research Centre for Natural Sciences, Budapest, Hungary (LSP); Department of Renal Medicine (MBL) and Department of Haematology-Oncology (BCG), National University Health System, Singapore; Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore (VT); Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia (VT); Department of Biological Sciences, University of North Texas, Denton, TX (APK)
| | - Lorinc S Pongor
- Affiliations of authors: Cancer Science Institute of Singapore, National University of Singapore, Singapore (WC, GR, SSS, CW, YY, TZT, HY, BCG, TB, APK); Departments of Pharmacology (WC, GR, SSS, CW, BCG, GS, APK), Physiology (ZXC, ZC, CTY), and Biochemistry (VT), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; KK Women's and Children's Hospital, Singapore (ZXC); Stem Cell and Cancer Biology Laboratory (FA), School of Biomedical Sciences (GS, APK), Curtin Health Innovation Research Institute, Curtin Medical School (APK), Curtin University, Perth, WA, Australia; National University Cancer Institute, National University Health System, Singapore (CTY, BCG, APK); 2 Department of Pediatrics, Semmelweis University, Budapest, Hungary (LSP); MTA TTK Lendület Cancer Biomarker Research Group, Research Centre for Natural Sciences, Budapest, Hungary (LSP); Department of Renal Medicine (MBL) and Department of Haematology-Oncology (BCG), National University Health System, Singapore; Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore (VT); Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia (VT); Department of Biological Sciences, University of North Texas, Denton, TX (APK)
| | - Henry Yang
- Affiliations of authors: Cancer Science Institute of Singapore, National University of Singapore, Singapore (WC, GR, SSS, CW, YY, TZT, HY, BCG, TB, APK); Departments of Pharmacology (WC, GR, SSS, CW, BCG, GS, APK), Physiology (ZXC, ZC, CTY), and Biochemistry (VT), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; KK Women's and Children's Hospital, Singapore (ZXC); Stem Cell and Cancer Biology Laboratory (FA), School of Biomedical Sciences (GS, APK), Curtin Health Innovation Research Institute, Curtin Medical School (APK), Curtin University, Perth, WA, Australia; National University Cancer Institute, National University Health System, Singapore (CTY, BCG, APK); 2 Department of Pediatrics, Semmelweis University, Budapest, Hungary (LSP); MTA TTK Lendület Cancer Biomarker Research Group, Research Centre for Natural Sciences, Budapest, Hungary (LSP); Department of Renal Medicine (MBL) and Department of Haematology-Oncology (BCG), National University Health System, Singapore; Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore (VT); Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia (VT); Department of Biological Sciences, University of North Texas, Denton, TX (APK)
| | - Martin B Lee
- Affiliations of authors: Cancer Science Institute of Singapore, National University of Singapore, Singapore (WC, GR, SSS, CW, YY, TZT, HY, BCG, TB, APK); Departments of Pharmacology (WC, GR, SSS, CW, BCG, GS, APK), Physiology (ZXC, ZC, CTY), and Biochemistry (VT), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; KK Women's and Children's Hospital, Singapore (ZXC); Stem Cell and Cancer Biology Laboratory (FA), School of Biomedical Sciences (GS, APK), Curtin Health Innovation Research Institute, Curtin Medical School (APK), Curtin University, Perth, WA, Australia; National University Cancer Institute, National University Health System, Singapore (CTY, BCG, APK); 2 Department of Pediatrics, Semmelweis University, Budapest, Hungary (LSP); MTA TTK Lendület Cancer Biomarker Research Group, Research Centre for Natural Sciences, Budapest, Hungary (LSP); Department of Renal Medicine (MBL) and Department of Haematology-Oncology (BCG), National University Health System, Singapore; Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore (VT); Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia (VT); Department of Biological Sciences, University of North Texas, Denton, TX (APK)
| | - Boon Cher Goh
- Affiliations of authors: Cancer Science Institute of Singapore, National University of Singapore, Singapore (WC, GR, SSS, CW, YY, TZT, HY, BCG, TB, APK); Departments of Pharmacology (WC, GR, SSS, CW, BCG, GS, APK), Physiology (ZXC, ZC, CTY), and Biochemistry (VT), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; KK Women's and Children's Hospital, Singapore (ZXC); Stem Cell and Cancer Biology Laboratory (FA), School of Biomedical Sciences (GS, APK), Curtin Health Innovation Research Institute, Curtin Medical School (APK), Curtin University, Perth, WA, Australia; National University Cancer Institute, National University Health System, Singapore (CTY, BCG, APK); 2 Department of Pediatrics, Semmelweis University, Budapest, Hungary (LSP); MTA TTK Lendület Cancer Biomarker Research Group, Research Centre for Natural Sciences, Budapest, Hungary (LSP); Department of Renal Medicine (MBL) and Department of Haematology-Oncology (BCG), National University Health System, Singapore; Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore (VT); Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia (VT); Department of Biological Sciences, University of North Texas, Denton, TX (APK)
| | - Gautam Sethi
- Affiliations of authors: Cancer Science Institute of Singapore, National University of Singapore, Singapore (WC, GR, SSS, CW, YY, TZT, HY, BCG, TB, APK); Departments of Pharmacology (WC, GR, SSS, CW, BCG, GS, APK), Physiology (ZXC, ZC, CTY), and Biochemistry (VT), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; KK Women's and Children's Hospital, Singapore (ZXC); Stem Cell and Cancer Biology Laboratory (FA), School of Biomedical Sciences (GS, APK), Curtin Health Innovation Research Institute, Curtin Medical School (APK), Curtin University, Perth, WA, Australia; National University Cancer Institute, National University Health System, Singapore (CTY, BCG, APK); 2 Department of Pediatrics, Semmelweis University, Budapest, Hungary (LSP); MTA TTK Lendület Cancer Biomarker Research Group, Research Centre for Natural Sciences, Budapest, Hungary (LSP); Department of Renal Medicine (MBL) and Department of Haematology-Oncology (BCG), National University Health System, Singapore; Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore (VT); Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia (VT); Department of Biological Sciences, University of North Texas, Denton, TX (APK)
| | - Touati Benoukraf
- Affiliations of authors: Cancer Science Institute of Singapore, National University of Singapore, Singapore (WC, GR, SSS, CW, YY, TZT, HY, BCG, TB, APK); Departments of Pharmacology (WC, GR, SSS, CW, BCG, GS, APK), Physiology (ZXC, ZC, CTY), and Biochemistry (VT), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; KK Women's and Children's Hospital, Singapore (ZXC); Stem Cell and Cancer Biology Laboratory (FA), School of Biomedical Sciences (GS, APK), Curtin Health Innovation Research Institute, Curtin Medical School (APK), Curtin University, Perth, WA, Australia; National University Cancer Institute, National University Health System, Singapore (CTY, BCG, APK); 2 Department of Pediatrics, Semmelweis University, Budapest, Hungary (LSP); MTA TTK Lendület Cancer Biomarker Research Group, Research Centre for Natural Sciences, Budapest, Hungary (LSP); Department of Renal Medicine (MBL) and Department of Haematology-Oncology (BCG), National University Health System, Singapore; Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore (VT); Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia (VT); Department of Biological Sciences, University of North Texas, Denton, TX (APK)
| | - Vinay Tergaonkar
- Affiliations of authors: Cancer Science Institute of Singapore, National University of Singapore, Singapore (WC, GR, SSS, CW, YY, TZT, HY, BCG, TB, APK); Departments of Pharmacology (WC, GR, SSS, CW, BCG, GS, APK), Physiology (ZXC, ZC, CTY), and Biochemistry (VT), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; KK Women's and Children's Hospital, Singapore (ZXC); Stem Cell and Cancer Biology Laboratory (FA), School of Biomedical Sciences (GS, APK), Curtin Health Innovation Research Institute, Curtin Medical School (APK), Curtin University, Perth, WA, Australia; National University Cancer Institute, National University Health System, Singapore (CTY, BCG, APK); 2 Department of Pediatrics, Semmelweis University, Budapest, Hungary (LSP); MTA TTK Lendület Cancer Biomarker Research Group, Research Centre for Natural Sciences, Budapest, Hungary (LSP); Department of Renal Medicine (MBL) and Department of Haematology-Oncology (BCG), National University Health System, Singapore; Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore (VT); Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia (VT); Department of Biological Sciences, University of North Texas, Denton, TX (APK)
| | - Alan Prem Kumar
- Affiliations of authors: Cancer Science Institute of Singapore, National University of Singapore, Singapore (WC, GR, SSS, CW, YY, TZT, HY, BCG, TB, APK); Departments of Pharmacology (WC, GR, SSS, CW, BCG, GS, APK), Physiology (ZXC, ZC, CTY), and Biochemistry (VT), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; KK Women's and Children's Hospital, Singapore (ZXC); Stem Cell and Cancer Biology Laboratory (FA), School of Biomedical Sciences (GS, APK), Curtin Health Innovation Research Institute, Curtin Medical School (APK), Curtin University, Perth, WA, Australia; National University Cancer Institute, National University Health System, Singapore (CTY, BCG, APK); 2 Department of Pediatrics, Semmelweis University, Budapest, Hungary (LSP); MTA TTK Lendület Cancer Biomarker Research Group, Research Centre for Natural Sciences, Budapest, Hungary (LSP); Department of Renal Medicine (MBL) and Department of Haematology-Oncology (BCG), National University Health System, Singapore; Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore (VT); Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia (VT); Department of Biological Sciences, University of North Texas, Denton, TX (APK)
| |
Collapse
|
41
|
Wu YL, Hsu NY, Cheau-Feng Lin F, Lee H, Cheng YW. MiR-30c-2* negative regulated MTA-1 expression involved in metastasis and drug resistance of HPV-infected non-small cell lung cancer. Surgery 2016; 160:1591-1598. [PMID: 27506865 DOI: 10.1016/j.surg.2016.06.025] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 05/26/2016] [Accepted: 06/08/2016] [Indexed: 02/07/2023]
Abstract
BACKGROUND MiR-30c-2* is considered to be a tumor suppressor microRNA in various cancers and is associated with gemcitabine sensitivity of lung cancer cells. Downregulation of miR-30c-2* promotes tumor invasion via increased expression of metastasis-associated protein-1. We hypothesized that downregulated expression of miR-30c-2* was involved in human papillomavirus-associated lung tumorigenesis and drug resistance. METHODS We examined whether expression of human papillomavirus 16/18 oncoprotein and miR-30c-2*-associated genes could be linked to patient outcome by collecting 319 lung tumors from patients with non-small cell lung cancer to determine expression of human papillomavirus 16/18 E6 protein, miR-30c-2*, and miR-30c-2* downstream metastasis-associated protein-1 mRNA by immunohistochemical and real-time polymerase chain reaction analysis. RESULTS Our results showed that miR-30C-2* levels were increased 45-fold in the E6-knockdown TL-1 cells when compared with levels in the parental cells. More interestingly, metastasis-associated protein-1 expression correlated negatively with miR-30C-2* and positively with human papillomavirus 16 E6 protein expression in lung tumors from lung cancer patients. Metastasis-associated protein-1 expression levels in the tumor tissues correlated positively with tumor stage and nodal metastasis. Patients with high metastasis-associated protein-1 expression, and especially patients infected with human papillomavirus, experienced a poor clinical outcome, tumor recurrence, and a poor therapeutic response compared with those with low metastasis-associated protein-1 expression. CONCLUSION These results showed that miR-30C-2* and levels of downstream metastasis-associated protein-1 gene expression in the tumor tissues of patients could be useful in predicting clinical outcome and therapeutic response and in selecting useful therapeutic drugs for lung cancer patients, especially patients with human papillomavirus infection.
Collapse
Affiliation(s)
- Yi-Liang Wu
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan; School of Medicine, College of Medicine, Chung Shan Medical University, Taichung, Taiwan; Department of Surgery, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Nan-Yung Hsu
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan; Cancer Center, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan; Department of Surgery, Taipei Medical University Hospital, Taipei, Taiwan; Division of Thoracic Surgery, Ningbo Medical Center Lihuili Eastern Hospital, Ningbo, China
| | - Frank Cheau-Feng Lin
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan; Department of Surgery, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Huei Lee
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan; Cancer Center, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
| | - Ya-Wen Cheng
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan; Cancer Center, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
42
|
The DEAD-Box RNA Helicase DDX3 Interacts with NF-κB Subunit p65 and Suppresses p65-Mediated Transcription. PLoS One 2016; 11:e0164471. [PMID: 27736973 PMCID: PMC5063347 DOI: 10.1371/journal.pone.0164471] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Accepted: 09/26/2016] [Indexed: 11/29/2022] Open
Abstract
RNA helicase family members exhibit diverse cellular functions, including in transcription, pre-mRNA processing, RNA decay, ribosome biogenesis, RNA export and translation. The RNA helicase DEAD-box family member DDX3 has been characterized as a tumour-associated factor and a transcriptional co-activator/regulator. Here, we demonstrate that DDX3 interacts with the nuclear factor (NF)-κB subunit p65 and suppresses NF-κB (p65/p50)-mediated transcriptional activity. The downregulation of DDX3 by RNA interference induces the upregulation of NF-κB (p65/p50)-mediated transcription. The regulation of NF-κB (p65/p50)-mediated transcriptional activity was further confirmed by the expression levels of its downstream cytokines, such as IL-6 and IL-8. Moreover, the binding of the ATP-dependent RNA helicase domain of DDX3 to the N-terminal Rel homology domain (RHD) of p65 is involved in the inhibition of NF-κB-regulated gene transcription. In summary, the results suggest that DDX3 functions to suppress the transcriptional activity of the NF-κB subunit p65.
Collapse
|
43
|
Heerma van Voss MR, Vesuna F, Trumpi K, Brilliant J, Berlinicke C, de Leng W, Kranenburg O, Offerhaus GJ, Bürger H, van der Wall E, van Diest PJ, Raman V. Identification of the DEAD box RNA helicase DDX3 as a therapeutic target in colorectal cancer. Oncotarget 2016; 6:28312-26. [PMID: 26311743 PMCID: PMC4695062 DOI: 10.18632/oncotarget.4873] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 07/09/2015] [Indexed: 02/06/2023] Open
Abstract
Identifying druggable targets in the Wnt-signaling pathway can optimize colorectal cancer treatment. Recent studies have identified a member of the RNA helicase family DDX3 (DDX3X) as a multilevel activator of Wnt signaling in cells without activating mutations in the Wnt-signaling pathway. In this study, we evaluated whether DDX3 plays a role in the constitutively active Wnt pathway that drives colorectal cancer. We determined DDX3 expression levels in 303 colorectal cancers by immunohistochemistry. 39% of tumors overexpressed DDX3. High cytoplasmic DDX3 expression correlated with nuclear β-catenin expression, a marker of activated Wnt signaling. Functionally, we validated this finding in vitro and found that inhibition of DDX3 with siRNA resulted in reduced TCF4-reporter activity and lowered the mRNA expression levels of downstream TCF4-regulated genes. In addition, DDX3 knockdown in colorectal cancer cell lines reduced proliferation and caused a G1 arrest, supporting a potential oncogenic role of DDX3 in colorectal cancer. RK-33 is a small molecule inhibitor designed to bind to the ATP-binding site of DDX3. Treatment of colorectal cancer cell lines and patient-derived 3D cultures with RK-33 inhibited growth and promoted cell death with IC50 values ranging from 2.5 to 8 μM. The highest RK-33 sensitivity was observed in tumors with wild-type APC-status and a mutation in CTNNB1. Based on these results, we conclude that DDX3 has an oncogenic role in colorectal cancer. Inhibition of DDX3 with the small molecule inhibitor RK-33 causes inhibition of Wnt signaling and may therefore be a promising future treatment strategy for a subset of colorectal cancers.
Collapse
Affiliation(s)
- Marise R Heerma van Voss
- Department of Radiology and Radiological Science, Johns Hopkins University, School of Medicine, Baltimore, MD, USA.,Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Farhad Vesuna
- Department of Radiology and Radiological Science, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Kari Trumpi
- Department of Surgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Justin Brilliant
- Department of Radiology and Radiological Science, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Cynthia Berlinicke
- Wilmer Eye Institute, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Wendy de Leng
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Onno Kranenburg
- Department of Surgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | - G Johan Offerhaus
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Elsken van der Wall
- Department of Internal Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Paul J van Diest
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Venu Raman
- Department of Radiology and Radiological Science, Johns Hopkins University, School of Medicine, Baltimore, MD, USA.,Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands.,Department of Oncology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| |
Collapse
|
44
|
de Freitas AC, Gurgel AP, de Lima EG, de França São Marcos B, do Amaral CMM. Human papillomavirus and lung cancinogenesis: an overview. J Cancer Res Clin Oncol 2016; 142:2415-2427. [PMID: 27357515 DOI: 10.1007/s00432-016-2197-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 06/18/2016] [Indexed: 01/14/2023]
Abstract
Lung cancer is the most common cause of cancer deaths worldwide. Although tobacco smoking is considered to be the main risk factor and the most well-established risk factor for lung cancer, a number of patients who do not smoke have developed this disease. This number varies between 15 % to over one-half of lung cancer cases, and the deaths from lung cancer in non-smokers are increasing every year. There are many other agents that are thought to be etiological, including diesel exhaust exposure, metals, radiation, radon, hormonal factors, cooking oil, air pollution and infectious diseases, such as human papillomavirus (HPV). Studies in various parts of the world have detected HPV DNA at different rates in lung tumors. However, the role of HPV in lung cancer is still unclear. Thus, in this review, we investigated some molecular mechanisms of HPV protein activity in host cells, the entry of HPV into lung tissue and the possible route used by the virus to reach the lung cells.
Collapse
Affiliation(s)
- Antonio Carlos de Freitas
- Laboratory of Molecular Studies and Experimental Therapy, Department of Genetics, Federal University of Pernambuco, Cidade Universitária, Av Professor Moraes Rêgo S/N, Recife, Pernambuco, 50670-901, Brazil.
| | - Ana Pavla Gurgel
- Laboratory of Molecular Studies and Experimental Therapy, Department of Genetics, Federal University of Pernambuco, Cidade Universitária, Av Professor Moraes Rêgo S/N, Recife, Pernambuco, 50670-901, Brazil
| | - Elyda Golçalves de Lima
- Laboratory of Molecular Studies and Experimental Therapy, Department of Genetics, Federal University of Pernambuco, Cidade Universitária, Av Professor Moraes Rêgo S/N, Recife, Pernambuco, 50670-901, Brazil
| | - Bianca de França São Marcos
- Laboratory of Molecular Studies and Experimental Therapy, Department of Genetics, Federal University of Pernambuco, Cidade Universitária, Av Professor Moraes Rêgo S/N, Recife, Pernambuco, 50670-901, Brazil
| | - Carolina Maria Medeiros do Amaral
- Laboratory of Molecular Studies and Experimental Therapy, Department of Genetics, Federal University of Pernambuco, Cidade Universitária, Av Professor Moraes Rêgo S/N, Recife, Pernambuco, 50670-901, Brazil
| |
Collapse
|
45
|
Shen CC, Huang P. Role of DEAD box family in colorectal cancer. Shijie Huaren Xiaohua Zazhi 2016; 24:2811-2816. [DOI: 10.11569/wcjd.v24.i18.2811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer is one of the most common gastrointestinal tumors, posing a serious threat to human health. The DEAD box family plays an important role in RNA processing, such as transcription, pre-mRNA splicing and mRNA export and translation. Studies have shown that dysregulated expression of many RNA helicases exists in tumors, and some RNA helicases are involved in cell differentiation, cell cycle, apoptosis, oncogene expression and tumor drug resistance. This review aims to elucidate the research progress about the role of the DEAD box family in colorectal cancer.
Collapse
|
46
|
Li HK, Mai RT, Huang HD, Chou CH, Chang YA, Chang YW, You LR, Chen CM, Lee YHW. DDX3 Represses Stemness by Epigenetically Modulating Tumor-suppressive miRNAs in Hepatocellular Carcinoma. Sci Rep 2016; 6:28637. [PMID: 27344963 PMCID: PMC4921922 DOI: 10.1038/srep28637] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 06/06/2016] [Indexed: 12/16/2022] Open
Abstract
Studies indicate that the presence of cancer stem cells (CSCs) is responsible for poor prognosis of hepatocellular carcinoma (HCC) patients. In this study, the functional role of DDX3 in regulation of hepatic CSCs was investigated. Our results demonstrated that reduced DDX3 expression was not only inversely associated with tumor grade, but also predicted poor prognosis of HCC patients. Knockdown of DDX3 in HCC cell line HepG2 induced stemness gene signature followed by occurrence of self-renewal, chemoreisistance, EMT, migration as well as CSC expansion, and most importantly, DDX3 knockdown promotes tumorigenesis. Moreover, we found positive correlations between DDX3 level and expressions of tumor-suppressive miR-200b, miR-200c, miR-122 and miR-145, but not miR-10b and miR-519a, implying their involvement in DDX3 knockdown-induced CSC phenotypes. In addition, DDX3 reduction promoted up-regulation of DNA methyltransferase 3A (DNMT3A), while neither DNMT3B nor DNMT1 expression was affected. Enriched DNMT3A binding along with hypermethylation on promoters of these tumor-suppressive miRNAs reflected their transcriptional repressions in DDX3-knockdown cells. Furthermore, individual restoration of these tumor-suppressive miRNAs represses DDX3 knockdown-induced CSC phenotypes. In conclusion, our study suggested that DDX3 prevents generation of CSCs through epigenetically regulating a subset of tumor-suppressive miRNAs expressions, which strengthens tumor suppressor role of DDX3 in HCC.
Collapse
Affiliation(s)
- Hao-Kang Li
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Ru-Tsun Mai
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, National Yang-Ming University, Taipei, Taiwan.,Department of Biological Science and Technology, College of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan
| | - Hsien-Da Huang
- Department of Biological Science and Technology, College of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan.,Institute of Bioinformatics and Systems Biology, College of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan
| | - Chih-Hung Chou
- Department of Biological Science and Technology, College of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan.,Institute of Bioinformatics and Systems Biology, College of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan
| | - Yi-An Chang
- Department of Biological Science and Technology, College of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan.,Department of Medical Research, Mackay Memorial Hospital, Hsinchu, Taiwan
| | - Yao-Wen Chang
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Li-Ru You
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Chun-Ming Chen
- Department of Life Sciences and Institute of Genome Sciences, School of Life Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Yan-Hwa Wu Lee
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, National Yang-Ming University, Taipei, Taiwan.,Department of Biological Science and Technology, College of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan
| |
Collapse
|
47
|
Su CY, Lin TC, Lin YF, Chen MH, Lee CH, Wang HY, Lee YC, Liu YP, Chen CL, Hsiao M. DDX3 as a strongest prognosis marker and its downregulation promotes metastasis in colorectal cancer. Oncotarget 2016; 6:18602-12. [PMID: 26087195 PMCID: PMC4621913 DOI: 10.18632/oncotarget.4329] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 05/12/2015] [Indexed: 12/11/2022] Open
Abstract
Background Conflicting results regarding the role of DEAD-box polypeptide 3 (DDX3) are seen not only between cancer types but also within the same type of cancer. In this study, we aimed at clarifying the prognostic significance of DDX3 in patients of major cancer types through large cohort survival analysis and further investigated its effects on cancer progression. Methods Large cohort survival analysis of 7 cancer types, including colorectal cancer, breast cancer, lung cancer, head and neck cancer, liver cancer, glioblastoma, and ovarian cancer, was performed using public database at RNA level and was further confirmed by IHC analysis at protein level. Phenotype parameters of DDX3 knockdown colon cancer cells and the mechanism of DDX3 regulated cancer progression were investigated in vitro and in vivo. Results In large cohort survival analysis, DDX3 had a significant prognostic predictive power in colorectal cancer at both RNA and protein level. Patients with low DDX3 expression had poor prognosis and frequent distant metastasis. Knockdown of DDX3 enhanced the migration and invasion abilities of colon cancer cells and promoted tumor metastasis in vivo. Snail upregulation with decreased membranous E-cadherin expression and reduced cell aggregation were found after DDX3 downregulation. Conclusions Our study revealed the strong prognostic effect of DDX3 on colorectal cancer among seven major cancer types through larger cohort survival analysis at RNA and protein level. Low DDX3 expression promotes Snail/E-cadherin pathway mediated cancer metastasis and poor clinical outcome in colorectal cancer patients.
Collapse
Affiliation(s)
- Chia-Yi Su
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | | | - Yuan-Feng Lin
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ming-Huang Chen
- Division of Hematology and Oncology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chien-Hsin Lee
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Hsuan-Yao Wang
- Graduate Program of Molecular Pharmacology and Toxicology, School of Pharmacy, University of Southern California, Los Angeles, USA
| | - Yu-Chieh Lee
- Graduate Institute of Medical Sciences, Taipei Medical University, Taipei, Taiwan
| | - Yu-Peng Liu
- Department of Genome Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chi-Long Chen
- Department of Pathology, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan.,Department of Pathology, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Michael Hsiao
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
48
|
Valentin-Vega YA, Wang YD, Parker M, Patmore DM, Kanagaraj A, Moore J, Rusch M, Finkelstein D, Ellison DW, Gilbertson RJ, Zhang J, Kim HJ, Taylor JP. Cancer-associated DDX3X mutations drive stress granule assembly and impair global translation. Sci Rep 2016; 6:25996. [PMID: 27180681 PMCID: PMC4867597 DOI: 10.1038/srep25996] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 04/21/2016] [Indexed: 12/26/2022] Open
Abstract
DDX3X is a DEAD-box RNA helicase that has been implicated in multiple aspects of RNA metabolism including translation initiation and the assembly of stress granules (SGs). Recent genomic studies have reported recurrent DDX3X mutations in numerous tumors including medulloblastoma (MB), but the physiological impact of these mutations is poorly understood. Here we show that a consistent feature of MB-associated mutations is SG hyper-assembly and concomitant translation impairment. We used CLIP-seq to obtain a comprehensive assessment of DDX3X binding targets and ribosome profiling for high-resolution assessment of global translation. Surprisingly, mutant DDX3X expression caused broad inhibition of translation that impacted DDX3X targeted and non-targeted mRNAs alike. Assessment of translation efficiency with single-cell resolution revealed that SG hyper-assembly correlated precisely with impaired global translation. SG hyper-assembly and translation impairment driven by mutant DDX3X were rescued by a genetic approach that limited SG assembly and by deletion of the N-terminal low complexity domain within DDX3X. Thus, in addition to a primary defect at the level of translation initiation caused by DDX3X mutation, SG assembly itself contributes to global translation inhibition. This work provides mechanistic insights into the consequences of cancer-related DDX3X mutations, suggesting that globally reduced translation may provide a context-dependent survival advantage that must be considered as a possible contributor to tumorigenesis.
Collapse
Affiliation(s)
- Yasmine A. Valentin-Vega
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Yong-Dong Wang
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Matthew Parker
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Deanna M. Patmore
- Department of Oncology, Cambridge Cancer Centre, Cancer Research UK Cambridge Institute, Cambridge, UK
| | - Anderson Kanagaraj
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Jennifer Moore
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Michael Rusch
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - David Finkelstein
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - David W. Ellison
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Richard J. Gilbertson
- Department of Oncology, Cambridge Cancer Centre, Cancer Research UK Cambridge Institute, Cambridge, UK
| | - Jinghui Zhang
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Hong Joo Kim
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - J. Paul Taylor
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| |
Collapse
|
49
|
Abbas I, Verdin A, Escande F, Saint-Georges F, Cazier F, Mulliez P, Courcot D, Shirali P, Gosset P, Garçon G. In vitro short-term exposure to air pollution PM2.5-0.3 induced cell cycle alterations and genetic instability in a human lung cell coculture model. ENVIRONMENTAL RESEARCH 2016; 147:146-158. [PMID: 26874047 DOI: 10.1016/j.envres.2016.01.041] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 01/25/2016] [Accepted: 01/27/2016] [Indexed: 06/05/2023]
Abstract
Although its adverse health effects of air pollution particulate matter (PM2.5) are well-documented and often related to oxidative stress and pro-inflammatory response, recent evidence support the role of the remodeling of the airway epithelium involving the regulation of cell death processes. Hence, the overarching goals of the present study were to use an in vitro coculture model, based on human AM and L132 cells to study the possible alteration of TP53-RB gene signaling pathways (i.e. cell cycle phases, gene expression of TP53, BCL2, BAX, P21, CCND1, and RB, and protein concentrations of their active forms), and genetic instability (i.e. LOH and/or MSI) in the PM2.5-0.3-exposed coculture model. PM2.5-0.3 exposure of human AM from the coculture model induced marked cell cycle alterations after 24h, as shown by increased numbers of L132 cells in subG1 and S+G2 cell cycle phases, indicating apoptosis and proliferation. Accordingly, activation of the TP53-RB gene signaling pathways after the coculture model exposure to PM2.5-0.3 was reported in the L132 cells. Exposure of human AM from the coculture model to PM2.5-0.3 resulted in MS alterations in 3p chromosome multiple critical regions in L132 cell population. Hence, in vitro short-term exposure of the coculture model to PM2.5-0.3 induced cell cycle alterations relying on the sequential occurrence of molecular abnormalities from TP53-RB gene signaling pathway activation and genetic instability.
Collapse
Affiliation(s)
- Imane Abbas
- Université de Lille, Lille, France; EA4492-UCEIV, Université du Littoral-Côte d'Opale, Dunkerque, France; Lebanese Atomic Energy Commission - CNRS, Beirut, Lebanon
| | - Anthony Verdin
- Université de Lille, Lille, France; EA4492-UCEIV, Université du Littoral-Côte d'Opale, Dunkerque, France
| | - Fabienne Escande
- Centre de Biologie Pathologie, Centre Hospitalier Régional et Universitaire, Lille, France
| | - Françoise Saint-Georges
- Université de Lille, Lille, France; Groupement Hospitalier de l'Institut Catholique de Lille, Lille, France
| | - Fabrice Cazier
- Université de Lille, Lille, France; Centre Commun de Mesures, Université du Littoral-Côte d'Opale, Dunkerque, France
| | - Philippe Mulliez
- Université de Lille, Lille, France; Groupement Hospitalier de l'Institut Catholique de Lille, Lille, France
| | - Dominique Courcot
- Université de Lille, Lille, France; EA4492-UCEIV, Université du Littoral-Côte d'Opale, Dunkerque, France
| | - Pirouz Shirali
- Université de Lille, Lille, France; EA4492-UCEIV, Université du Littoral-Côte d'Opale, Dunkerque, France
| | - Pierre Gosset
- Université de Lille, Lille, France; Groupement Hospitalier de l'Institut Catholique de Lille, Lille, France
| | - Guillaume Garçon
- Université de Lille, Lille, France; EA4492-UCEIV, Université du Littoral-Côte d'Opale, Dunkerque, France; EA4483-IMPECS, Université de Lille 2, Lille, France.
| |
Collapse
|
50
|
Kotov AA, Olenkina OM, Kibanov MV, Olenina LV. RNA helicase Belle (DDX3) is essential for male germline stem cell maintenance and division in Drosophila. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:1093-105. [PMID: 26876306 DOI: 10.1016/j.bbamcr.2016.02.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 01/19/2016] [Accepted: 02/09/2016] [Indexed: 01/08/2023]
Abstract
The present study showed that RNA helicase Belle (DDX3) was required intrinsically for mitotic progression and survival of germline stem cells (GSCs) and spermatogonial cells in the Drosophila melanogaster testes. We found that deficiency of Belle in the male germline resulted in a strong germ cell loss phenotype. Early germ cells are lost through cell death, whereas somatic hub and cyst cell populations are maintained. The observed phenotype is related to that of the human Sertoli Cell-Only Syndrome caused by the loss of DBY (DDX3) expression in the human testes and results in a complete lack of germ cells with preservation of somatic Sertoli cells. We found the hallmarks of mitotic G2 delay in early germ cells of the larval testes of bel mutants. Both mitotic cyclins, A and B, are markedly reduced in the gonads of bel mutants. Transcription levels of cycB and cycA decrease significantly in the testes of hypomorph bel mutants. Overexpression of Cyclin B in the germline partially rescues germ cell survival, mitotic progression and fertility in the bel-RNAi knockdown testes. Taken together, these results suggest that a role of Belle in GSC maintenance and regulation of early germ cell divisions is associated with the expression control of mitotic cyclins.
Collapse
Affiliation(s)
- Alexei A Kotov
- Laboratory of Biochemical Genetics of Animals, Institute of Molecular Genetics, Russian Academy of Sciences, Kurchatov Sq. 2, Moscow 123182, Russia
| | - Oxana M Olenkina
- Laboratory of Biochemical Genetics of Animals, Institute of Molecular Genetics, Russian Academy of Sciences, Kurchatov Sq. 2, Moscow 123182, Russia
| | - Mikhail V Kibanov
- Laboratory of Biochemical Genetics of Animals, Institute of Molecular Genetics, Russian Academy of Sciences, Kurchatov Sq. 2, Moscow 123182, Russia
| | - Ludmila V Olenina
- Laboratory of Biochemical Genetics of Animals, Institute of Molecular Genetics, Russian Academy of Sciences, Kurchatov Sq. 2, Moscow 123182, Russia.
| |
Collapse
|