1
|
Rivera IS, French JD, Bitar M, Sivakumaran H, Nair S, Kaufmann S, Hillman KM, Moradi Marjaneh M, Beesley J, Edwards SL. GWAS and 3D chromatin mapping identifies multicancer risk genes associated with hormone-dependent cancers. PLoS Genet 2024; 20:e1011490. [PMID: 39585897 PMCID: PMC11627375 DOI: 10.1371/journal.pgen.1011490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 12/09/2024] [Accepted: 11/06/2024] [Indexed: 11/27/2024] Open
Abstract
Hormone-dependent cancers (HDCs) share several risk factors, suggesting a common aetiology. Using data from genome-wide association studies, we showed spatial clustering of risk variants across four HDCs (breast, endometrial, ovarian and prostate cancers), contrasting with genetically uncorrelated traits. We identified 44 multi-HDC risk regions across the genome, defined as overlapping risk regions for at least two HDCs: two regions contained risk variants for all four HDCs, 13 for three HDCs and 28 for two HDCs. Integrating GWAS data, epigenomic profiling and promoter capture HiC maps from diverse cell line models, we annotated 53 candidate risk genes at 22 multi-HDC risk regions. These targets were enriched for established genes from the COSMIC Cancer Gene Census, but many had no previously reported pleiotropic roles. Additionally, we pinpointed lncRNAs as potential HDC targets and identified risk alleles in several regions that altered transcription factors motifs, suggesting regulatory mechanisms. Known drug targets were over-represented among the candidate multi-HDC risk genes, implying that some may serve as targets for therapeutic development or facilitate the repurposing of existing treatments for HDC. Our approach provides a framework for identifying common target genes driving complex traits and enhances understanding of HDC susceptibility.
Collapse
Affiliation(s)
- Isela Sarahi Rivera
- Cancer Research Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
- Faculty of Health, Queensland University of Technology, Brisbane, Australia
| | - Juliet D French
- Cancer Research Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
- Faculty of Health, Queensland University of Technology, Brisbane, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Mainá Bitar
- Cancer Research Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Haran Sivakumaran
- Cancer Research Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Sneha Nair
- Cancer Research Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Susanne Kaufmann
- Cancer Research Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Kristine M Hillman
- Cancer Research Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Mahdi Moradi Marjaneh
- Cancer Research Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Jonathan Beesley
- Cancer Research Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Stacey L Edwards
- Cancer Research Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
- Faculty of Health, Queensland University of Technology, Brisbane, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, Australia
| |
Collapse
|
2
|
Lin W, Yan Y, Huang Q, Zheng D. MDMX in Cancer: A Partner of p53 and a p53-Independent Effector. Biologics 2024; 18:61-78. [PMID: 38318098 PMCID: PMC10839028 DOI: 10.2147/btt.s436629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 12/08/2023] [Indexed: 02/07/2024]
Abstract
The p53 tumor suppressor protein plays an important role in physiological and pathological processes. MDM2 and its homolog MDMX are the most important negative regulators of p53. Many studies have shown that MDMX promotes the growth of cancer cells by influencing the regulation of the downstream target gene of tumor suppressor p53. Studies have found that inhibiting the MDMX-p53 interaction can effectively restore the tumor suppressor activity of p53. MDMX has growth-promoting activities without p53 or in the presence of mutant p53. Therefore, it is extremely important to study the function of MDMX in tumorigenesis, progression and prognosis. This article mainly reviews the current research progress and mechanism on MDMX function, summarizes known MDMX inhibitors and provides new ideas for the development of more specific and effective MDMX inhibitors for cancer treatment.
Collapse
Affiliation(s)
- Wu Lin
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, People’s Republic of China
| | - Yuxiang Yan
- Fujian Key Laboratory of Oral Diseases, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, People’s Republic of China
| | - Qingling Huang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, People’s Republic of China
| | - Dali Zheng
- Fujian Key Laboratory of Oral Diseases, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, People’s Republic of China
| |
Collapse
|
3
|
Ward SV, Autuori I, Luo L, LaPilla E, Yoo S, Sharma A, Busam KJ, Olilla DW, Dwyer T, Anton-Culver H, Zanetti R, Sacchetto L, Cust AE, Gallagher RP, Kanetsky PA, Rosso S, Begg CB, Berwick M, Thomas NE, Orlow I. Sex-Specific Associations of MDM2 and MDM4 Variants with Risk of Multiple Primary Melanomas and Melanoma Survival in Non-Hispanic Whites. Cancers (Basel) 2023; 15:2707. [PMID: 37345045 PMCID: PMC10216616 DOI: 10.3390/cancers15102707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/04/2023] [Accepted: 05/08/2023] [Indexed: 06/23/2023] Open
Abstract
MDM2-SNP309 (rs2279744), a common genetic modifier of cancer incidence in Li-Fraumeni syndrome, modifies risk, age of onset, or prognosis in a variety of cancers. Melanoma incidence and outcomes vary by sex, and although SNP309 exerts an effect on the estrogen receptor, no consensus exists on its effect on melanoma. MDM2 and MDM4 restrain p53-mediated tumor suppression, independently or together. We investigated SNP309, an a priori MDM4-rs4245739, and two coinherited variants, in a population-based cohort of 3663 primary incident melanomas. Per-allele and per-haplotype (MDM2_SNP309-SNP285; MDM4_rs4245739-rs1563828) odds ratios (OR) for multiple-melanoma were estimated with logistic regression models. Hazard ratios (HR) for melanoma death were estimated with Cox proportional hazards models. In analyses adjusted for covariates, females carrying MDM4-rs4245739*C were more likely to develop multiple melanomas (ORper-allele = 1.25, 95% CI 1.03-1.51, and Ptrend = 0.03), while MDM2-rs2279744*G was inversely associated with melanoma-death (HRper-allele = 0.63, 95% CI 0.42-0.95, and Ptrend = 0.03). We identified 16 coinherited expression quantitative loci that control the expression of MDM2, MDM4, and other genes in the skin, brain, and lungs. Our results suggest that MDM4/MDM2 variants are associated with the development of subsequent primaries and with the death of melanoma in a sex-dependent manner. Further investigations of the complex MDM2/MDM4 motif, and its contribution to the tumor microenvironment and observed associations, are warranted.
Collapse
Affiliation(s)
- Sarah V. Ward
- Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- School of Population and Global Health, The University of Western Australia, Perth, WA 6009, Australia
| | - Isidora Autuori
- Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Li Luo
- Department of Internal Medicine, The University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87106, USA
| | - Emily LaPilla
- Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Sarah Yoo
- Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ajay Sharma
- Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Klaus J. Busam
- Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - David W. Olilla
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Terence Dwyer
- Clinical Sciences Theme, Heart Group, Murdoch Children’s Research Institute, Melbourne, VIC 3052, Australia
- Nuffield Department of Women’s & Reproductive Health, University of Oxford, Oxford OX3 9DU, UK
- Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Carlton, VIC 3010, Australia
- Oxford Martin School, University of Oxford, Oxford OX1 3BD, UK
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
| | - Hoda Anton-Culver
- Department of Medicine, University of California, Irvine, CA 92617, USA
| | - Roberto Zanetti
- Piedmont Cancer Registry, Centre for Epidemiology and Prevention in Oncology in Piedmont, 10126 Turin, Italy
| | - Lidia Sacchetto
- Piedmont Cancer Registry, Centre for Epidemiology and Prevention in Oncology in Piedmont, 10126 Turin, Italy
| | - Anne E. Cust
- The Daffodil Centre, The University of Sydney, A Joint Venture with Cancer Council NSW, Sydney, NSW 2006, Australia
- Melanoma Institute Australia, The University of Sydney, Wollstonecraft, NSW 2065, Australia
| | - Richard P. Gallagher
- BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada
- Department of Dermatology and Skin Science, University of British Columbia, Vancouver, BC V5Z 4E8, Canada
| | - Peter A. Kanetsky
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Stefano Rosso
- Piedmont Cancer Registry, Centre for Epidemiology and Prevention in Oncology in Piedmont, 10126 Turin, Italy
| | - Colin B. Begg
- Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Marianne Berwick
- Department of Internal Medicine, The University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87106, USA
| | - Nancy E. Thomas
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27514, USA
- Department of Dermatology, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Irene Orlow
- Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
4
|
MDM4: What do we know about the association between its polymorphisms and cancer? MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 40:61. [PMID: 36566308 DOI: 10.1007/s12032-022-01929-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 12/13/2022] [Indexed: 12/25/2022]
Abstract
MDM4 is an important p53-negative regulator, consequently, it is involved in cell proliferation, DNA repair, and apoptosis regulation. MDM4 overexpression and amplification are described to lead to cancer formation, metastasis, and poor disease prognosis. Several MDM4 SNPs are in non-coding regions, and some affect the MDM4 regulation by disrupting the micro RNA binding site in 3'UTR (untranslated region). Here, we gathered several association studies with different MDM4 SNPs and populations to understand the relationship between its SNPs and solid tumor risk. Many studies failed to replicate their results regarding different populations, cancer types, and risk genotypes, leading to conflicting conclusions. We suggested that distinct haplotype patterns in different populations might affect the association between MDM4 SNPs and cancer risk. Thus, we propose to investigate some linkage SNPs in specific haplotypes to provide informative MDM4 markers for association studies with cancer.
Collapse
|
5
|
Mejía-Hernández JO, Raghu D, Caramia F, Clemons N, Fujihara K, Riseborough T, Teunisse A, Jochemsen AG, Abrahmsén L, Blandino G, Russo A, Gamell C, Fox SB, Mitchell C, Takano EA, Byrne D, Miranda PJ, Saleh R, Thorne H, Sandhu S, Williams SG, Keam SP, Haupt Y, Haupt S. Targeting MDM4 as a Novel Therapeutic Approach in Prostate Cancer Independent of p53 Status. Cancers (Basel) 2022; 14:3947. [PMID: 36010941 PMCID: PMC9405814 DOI: 10.3390/cancers14163947] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/11/2022] [Accepted: 08/12/2022] [Indexed: 11/17/2022] Open
Abstract
Metastatic prostate cancer is a lethal disease in patients incapable of responding to therapeutic interventions. Invasive prostate cancer spread is caused by failure of the normal anti-cancer defense systems that are controlled by the tumour suppressor protein, p53. Upon mutation, p53 malfunctions. Therapeutic strategies to directly re-empower the growth-restrictive capacities of p53 in cancers have largely been unsuccessful, frequently because of a failure to discriminate responses in diseased and healthy tissues. Our studies sought alternative prostate cancer drivers, intending to uncover new treatment targets. We discovered the oncogenic potency of MDM4 in prostate cancer cells, both in the presence and absence of p53 and also its mutation. We uncovered that sustained depletion of MDM4 is growth inhibitory in prostate cancer cells, involving either apoptosis or senescence, depending on the cell and genetic context. We identified that the potency of MDM4 targeting could be potentiated in prostate cancers with mutant p53 through the addition of a first-in-class small molecule drug that was selected as a p53 reactivator and has the capacity to elevate oxidative stress in cancer cells to drive their death.
Collapse
Affiliation(s)
- Javier Octavio Mejía-Hernández
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
- Tumour Suppression and Cancer Sex Disparity Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Dinesh Raghu
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
- Tumour Suppression and Cancer Sex Disparity Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
- Olivia Newton-John Cancer Research Institute, School of Cancer Medicine, La Trobe University, Heidelberg, VIC 3084, Australia
| | - Franco Caramia
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
- Tumour Suppression and Cancer Sex Disparity Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Nicholas Clemons
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Kenji Fujihara
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Thomas Riseborough
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
- Tumour Suppression and Cancer Sex Disparity Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Amina Teunisse
- Department of Cell and Chemical Biology, Leiden University Medical Centre, 2333 Leiden, The Netherlands
| | - Aart G. Jochemsen
- Department of Cell and Chemical Biology, Leiden University Medical Centre, 2333 Leiden, The Netherlands
| | | | - Giovanni Blandino
- Translational Oncology Research Unit, IRCSS Regina Elena National Cancer Institute, 0144 Rome, Italy
| | - Andrea Russo
- Surgical Pathology Unit, Department of Research, Advanced Diagnostics and Technological Innovation, IRCSS Regina Elena National Cancer Institute, 0144 Rome, Italy
| | - Cristina Gamell
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
- Tumour Suppression and Cancer Sex Disparity Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Stephen B. Fox
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
- Pathology Department, Peter MacCallum Cancer Centre, Parkville, VIC 3000, Australia
| | - Catherine Mitchell
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
- Pathology Department, Peter MacCallum Cancer Centre, Parkville, VIC 3000, Australia
| | - Elena A. Takano
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
- Pathology Department, Peter MacCallum Cancer Centre, Parkville, VIC 3000, Australia
| | - David Byrne
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
- Pathology Department, Peter MacCallum Cancer Centre, Parkville, VIC 3000, Australia
| | - Panimaya Jeffreena Miranda
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
- Tumour Suppression and Cancer Sex Disparity Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Reem Saleh
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
- Tumour Suppression and Cancer Sex Disparity Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Heather Thorne
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Shahneen Sandhu
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Parkville, VIC 3000, Australia
| | - Scott G. Williams
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
- Division of Radiation Oncology, Peter MacCallum Cancer Centre, Parkville, VIC 3000, Australia
| | - Simon P. Keam
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
- Tumour Suppression and Cancer Sex Disparity Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Ygal Haupt
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
- Tumour Suppression and Cancer Sex Disparity Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Sue Haupt
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
- Tumour Suppression and Cancer Sex Disparity Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| |
Collapse
|
6
|
Huang W, Zhang C, Xiong S, Zhou X, Wang G, Guo J. miR-1307-5p suppresses proliferation and tumorigenesis of bladder cancer via targeting MDM4 and the Hippo signaling pathway. Discov Oncol 2022; 13:57. [PMID: 35778656 PMCID: PMC9249964 DOI: 10.1007/s12672-022-00512-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 06/02/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Emerging evidence has shown that miR-1307-5p is involved in tumorigenesis of various types of cancer. This study aims to assess the role and mechanism of miR-1307-5p in bladder cancer. METHODS Bioinformatics analyses were carried out with clinical datasets in the public domains. To investigate the cellular functions of miR-1307-5p, assays of cell proliferation, cell cycle and cell apoptosis were conducted in bladder cancer cell lines and xenografts. The molecular mechanisms of miR-1307-5p were studied using luciferase reporter, RT-qPCR, and western blotting analyses. RESULTS We found that miR-1307-5p expression was significantly decreased in bladder cancer tissues, and its lower level was associated with poor prognosis. Cellular assays indicated the tumor-suppressor roles of miR-1307-5p were linked to cell proliferation, cell cycle inhibition, and cell apoptosis promotion. Conversely, anti-miR-1307-5p facilitated cell proliferation and cell cycle and antagonized cell apoptosis. In the in vivo setting, tumor growth was suppressed by miR-1307-5p overexpression. We found by bioinformatic and luciferase reporter assays that miR-1307-5p targets the 3'-UTR of MDM4, a well-known Inhibitor of TP53-mediated transactivation, cell cycle arrest and apoptosis. Specifically, miR-1307-5p markedly reduced MDM4 proteins expression, decreased the expression of Ki-67 and PCNA, and increased the expression of cleaved-caspase 3 and caspase 9. While in parallel assays, anti-miR-1307-5p had opposite effects. In addition, we found that miR-1307-5p overexpression would suppress bladder cancer cell growth by inhibiting MDM4 and its downstream Hippo pathway. CONCLUSION In bladder cancer, miR-1307-5p functions as a tumor suppressor and has the potentials as biomarker and therapeutical agent.
Collapse
Affiliation(s)
- Wei Huang
- Department of Urology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng street, Donghu District, Nanchang, 330006, Jiangxi, China
| | - Cheng Zhang
- Department of Urology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng street, Donghu District, Nanchang, 330006, Jiangxi, China
| | - Shida Xiong
- Department of Urology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng street, Donghu District, Nanchang, 330006, Jiangxi, China
| | - Xiaocheng Zhou
- Department of Urology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng street, Donghu District, Nanchang, 330006, Jiangxi, China
| | - Gongxian Wang
- Department of Urology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng street, Donghu District, Nanchang, 330006, Jiangxi, China.
| | - Ju Guo
- Department of Urology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng street, Donghu District, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
7
|
Alsheikh AJ, Wollenhaupt S, King EA, Reeb J, Ghosh S, Stolzenburg LR, Tamim S, Lazar J, Davis JW, Jacob HJ. The landscape of GWAS validation; systematic review identifying 309 validated non-coding variants across 130 human diseases. BMC Med Genomics 2022; 15:74. [PMID: 35365203 PMCID: PMC8973751 DOI: 10.1186/s12920-022-01216-w] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 03/17/2022] [Indexed: 02/08/2023] Open
Abstract
Background The remarkable growth of genome-wide association studies (GWAS) has created a critical need to experimentally validate the disease-associated variants, 90% of which involve non-coding variants. Methods To determine how the field is addressing this urgent need, we performed a comprehensive literature review identifying 36,676 articles. These were reduced to 1454 articles through a set of filters using natural language processing and ontology-based text-mining. This was followed by manual curation and cross-referencing against the GWAS catalog, yielding a final set of 286 articles. Results We identified 309 experimentally validated non-coding GWAS variants, regulating 252 genes across 130 human disease traits. These variants covered a variety of regulatory mechanisms. Interestingly, 70% (215/309) acted through cis-regulatory elements, with the remaining through promoters (22%, 70/309) or non-coding RNAs (8%, 24/309). Several validation approaches were utilized in these studies, including gene expression (n = 272), transcription factor binding (n = 175), reporter assays (n = 171), in vivo models (n = 104), genome editing (n = 96) and chromatin interaction (n = 33). Conclusions This review of the literature is the first to systematically evaluate the status and the landscape of experimentation being used to validate non-coding GWAS-identified variants. Our results clearly underscore the multifaceted approach needed for experimental validation, have practical implications on variant prioritization and considerations of target gene nomination. While the field has a long way to go to validate the thousands of GWAS associations, we show that progress is being made and provide exemplars of validation studies covering a wide variety of mechanisms, target genes, and disease areas. Supplementary Information The online version contains supplementary material available at 10.1186/s12920-022-01216-w.
Collapse
Affiliation(s)
- Ammar J Alsheikh
- Genomics Research Center, AbbVie Inc, North Chicago, Illinois, 60064, USA.
| | - Sabrina Wollenhaupt
- Information Research, AbbVie Deutschland GmbH & Co. KG, 67061, Knollstrasse, Ludwigshafen, Germany
| | - Emily A King
- Genomics Research Center, AbbVie Inc, North Chicago, Illinois, 60064, USA
| | - Jonas Reeb
- Information Research, AbbVie Deutschland GmbH & Co. KG, 67061, Knollstrasse, Ludwigshafen, Germany
| | - Sujana Ghosh
- Genomics Research Center, AbbVie Inc, North Chicago, Illinois, 60064, USA
| | | | - Saleh Tamim
- Genomics Research Center, AbbVie Inc, North Chicago, Illinois, 60064, USA
| | - Jozef Lazar
- Genomics Research Center, AbbVie Inc, North Chicago, Illinois, 60064, USA
| | - J Wade Davis
- Genomics Research Center, AbbVie Inc, North Chicago, Illinois, 60064, USA
| | - Howard J Jacob
- Genomics Research Center, AbbVie Inc, North Chicago, Illinois, 60064, USA
| |
Collapse
|
8
|
Zhou R, Li Y, Wang N, Niu C, Huang X, Cao S, Huo X. MDM4 polymorphisms associated with the risk but not the prognosis of esophageal cancer in Cixian high-incidence region from northern China. Asia Pac J Clin Oncol 2022; 18:e435-e441. [PMID: 35098683 DOI: 10.1111/ajco.13746] [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: 02/15/2021] [Accepted: 12/04/2021] [Indexed: 11/27/2022]
Abstract
AIM The mouse double minute 4 (MDM4) may contribute to tumorgenesis by inhibiting p53 tumor suppressor activity. This study was designed to investigate whether MDM4 polymorphisms could affect susceptibility to esophageal squamous cell carcinoma (ESCC) and the survival of ESCC patients in a population from Cixian high-incidence region of northern China, which has not been explored. METHODS MDM4 rs1380576 and rs4245739 were genotyped by polymerase chain reaction-ligase detection reaction (PCR-LDR) in 568 ESCC patients and 578 controls. RESULTS Compared to rs1380576 C/C genotype, C/G genotype was associated with decreased risk of ESCC (odds ratio [OR] = 0.761, 95% confidence interval [CI] = 0.595-0.973). Compared to rs4245739 A/A genotype, A/C or C/C genotype was related to increased susceptibility to ESCC (OR = 1.551, 95% CI = 1.001-2.402). Individuals with GC haplotype had significantly higher risk of ESCC than those with CA or GA haplotype (OR = 1.598, 95% CI = 1.048-2.438). Neither rs1380576 nor rs4245739 influenced the survival of ESCC patients. CONCLUSION rs1380576 and rs4245739 may be used to predict susceptibility to ESCC for population in Cixian high-incidence region.
Collapse
Affiliation(s)
- Rongmiao Zhou
- Hebei Provincial Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yan Li
- Hebei Provincial Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Na Wang
- Hebei Provincial Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Chaoxu Niu
- Department of Surgery, Shijiazhuang Ping'an Hospital, Shijiazhuang, China
| | - Xi Huang
- Hebei Provincial Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Shiru Cao
- Hebei Provincial Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiangran Huo
- Hebei Provincial Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
9
|
Lai X, Schmitz U, Vera J. The Role of MicroRNAs in Cancer Biology and Therapy from a Systems Biology Perspective. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1385:1-22. [DOI: 10.1007/978-3-031-08356-3_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
10
|
Chhichholiya Y, Suryan AK, Suman P, Munshi A, Singh S. SNPs in miRNAs and Target Sequences: Role in Cancer and Diabetes. Front Genet 2021; 12:793523. [PMID: 34925466 PMCID: PMC8673831 DOI: 10.3389/fgene.2021.793523] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 10/28/2021] [Indexed: 12/27/2022] Open
Abstract
miRNAs are fascinating molecular players for gene regulation as individual miRNA can control multiple targets and a single target can be regulated by multiple miRNAs. Loss of miRNA regulated gene expression is often reported to be implicated in various human diseases like diabetes and cancer. Recently, geneticists across the world started reporting single nucleotide polymorphism (SNPs) in seed sequences of miRNAs. Similarly, SNPs are also reported in various target sequences of these miRNAs. Both the scenarios lead to dysregulated gene expression which may result in the progression of diseases. In the present paper, we explore SNPs in various miRNAs and their target sequences reported in various human cancers as well as diabetes. Similarly, we also present evidence of these mutations in various other human diseases.
Collapse
Affiliation(s)
- Yogita Chhichholiya
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, India
| | - Aman Kumar Suryan
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, India
| | - Prabhat Suman
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, India
| | - Anjana Munshi
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, India
| | - Sandeep Singh
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, India
| |
Collapse
|
11
|
Mancini F, Giorgini L, Teveroni E, Pontecorvi A, Moretti F. Role of Sex in the Therapeutic Targeting of p53 Circuitry. Front Oncol 2021; 11:698946. [PMID: 34307167 PMCID: PMC8298065 DOI: 10.3389/fonc.2021.698946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 06/16/2021] [Indexed: 12/03/2022] Open
Abstract
Sex profoundly affects cancer incidence and susceptibility to therapy, with sex hormones highly contributing to this disparity. Various studies and omics data suggest a relationship between sex and the oncosuppressor p53 circuitry, including its regulators MDM2 and MDM4. Association of this network with genetic variation underlies sex-related altered cancer risk, age of onset, and cancer sensitivity to therapy. Moreover, sex-related factors, mainly estrogenic hormones, can affect the levels and/or function of the p53 network both in hormone-dependent and independent cancer. Despite this evidence, preclinical and clinical studies aimed to evaluate p53 targeted therapy rarely consider sex and related factors. This review summarizes the studies reporting the relationship between sex and the p53 circuitry, including its associated regulators, MDM2 and MDM4, with particular emphasis on estrogenic hormones. Moreover, we reviewed the evaluation of sex/hormone in preclinical studies and clinical trials employing p53-target therapies, and discuss how patients’ sex and hormonal status could impact these therapeutic approaches.
Collapse
Affiliation(s)
- Francesca Mancini
- Research Unit on Human Reproduction, International Scientific Institute Paul VI, Fondazione Policlinico A. Gemelli, IRCCS, Rome, Italy
| | - Ludovica Giorgini
- Institute of Biochemistry and Cell Biology, National Research Council of Italy, Monterotondo, Italy.,Catholic University of the Sacred Heart of Rome, Fondazione Policlinico A. Gemelli, IRCCS, Rome, Italy
| | - Emanuela Teveroni
- Research Unit on Human Reproduction, International Scientific Institute Paul VI, Fondazione Policlinico A. Gemelli, IRCCS, Rome, Italy
| | - Alfredo Pontecorvi
- Catholic University of the Sacred Heart of Rome, Fondazione Policlinico A. Gemelli, IRCCS, Rome, Italy
| | - Fabiola Moretti
- Institute of Biochemistry and Cell Biology, National Research Council of Italy, Monterotondo, Italy
| |
Collapse
|
12
|
Gansmo LB, Lie BA, Mæhlen MT, Vatten L, Romundstad P, Hveem K, Lønning PE, Knappskog S. Polymorphisms in the TP53-MDM2-MDM4-axis in patients with rheumatoid arthritis. Gene 2021; 793:145747. [PMID: 34077778 DOI: 10.1016/j.gene.2021.145747] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 04/30/2021] [Accepted: 05/27/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND In addition to being a tumour suppressor, TP53 is a suppressor of inflammation, and dysfunction of this gene has been related to autoimmune diseases. Patients with autoimmunity, such as rheumatoid arthritis (RA) have an increased risk of certain cancers, like lymphomas, indicating that some underlying mechanisms may modulate risk of both cancers and autoimmunity. METHODS We genotyped 5 common genetic variants in TP53 and its main regulators MDM2 and MDM4 in a sample of 942 RA patients and 3,747 healthy controls, and mined previously published GWAS-data, to assess the potential impact of these variants on risk of RA. RESULTS For the TP53 Arg72Pro polymorphism (rs1042522), MDM4 SNP34091 (rs4245739) and MDM2 SNP285C (rs117039649), we found no association to risk of RA. For MDM2 SNP309 (rs2279744), the minor G-allele was associated with a reduced risk of RA (OR: 0.87; CI: 0.79-0.97). This association was also seen in genotype models (OR: 0.86; CI: 0.74-0.99 and OR: 0.79; CI 0.63-0.99; dominant and recessive model, respectively), but was not validated in a large GWAS data set. For MDM2 del1518 (rs3730485), the minor del-allele was associated with an increased risk of RA in the dominant model (OR: 1.18; CI: 1.02-1.38). Stratifying RA cases and controls into phylogenetic subgroups according to the combined genotypes of all three MDM2 polymorphism, we found individuals with the del158-285-309 genotype del/ins-G/G-T/T to have an increased risk of RA as compared to those with the ins/ins-G/G-G/G genotype (OR: 1.56; CI: 1.18-2.06) indicating opposite effects of the del1518 del-allele and the SNP309 G-allele. CONCLUSION We find a potential association between the MDM2 del1518 variant and RA, and indications that combinatorial genotypes and haplotypes in the MDM2 locus may be related to RA.
Collapse
Affiliation(s)
- Liv B Gansmo
- K.G. Jebsen Center for Genome-Directed Cancer Therapy, Department of Clinical Science, University of Bergen, Bergen, Norway; Department of Oncology, Haukeland University Hospital, Bergen, Norway
| | - Benedicte A Lie
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Marthe T Mæhlen
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; Department of Rheumatology, Diakonhjemmet Hospital, Oslo, Norway
| | - Lars Vatten
- Department of Public Health, Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Pål Romundstad
- Department of Public Health, Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Kristian Hveem
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health, Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Per E Lønning
- K.G. Jebsen Center for Genome-Directed Cancer Therapy, Department of Clinical Science, University of Bergen, Bergen, Norway; Department of Oncology, Haukeland University Hospital, Bergen, Norway
| | - Stian Knappskog
- K.G. Jebsen Center for Genome-Directed Cancer Therapy, Department of Clinical Science, University of Bergen, Bergen, Norway; Department of Oncology, Haukeland University Hospital, Bergen, Norway.
| |
Collapse
|
13
|
Timmerman DM, Remmers TL, Hillenius S, Looijenga LHJ. Mechanisms of TP53 Pathway Inactivation in Embryonic and Somatic Cells-Relevance for Understanding (Germ Cell) Tumorigenesis. Int J Mol Sci 2021; 22:ijms22105377. [PMID: 34065345 PMCID: PMC8161298 DOI: 10.3390/ijms22105377] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/14/2021] [Accepted: 05/15/2021] [Indexed: 01/10/2023] Open
Abstract
The P53 pathway is the most important cellular pathway to maintain genomic and cellular integrity, both in embryonic and non-embryonic cells. Stress signals induce its activation, initiating autophagy or cell cycle arrest to enable DNA repair. The persistence of these signals causes either senescence or apoptosis. Over 50% of all solid tumors harbor mutations in TP53 that inactivate the pathway. The remaining cancers are suggested to harbor mutations in genes that regulate the P53 pathway such as its inhibitors Mouse Double Minute 2 and 4 (MDM2 and MDM4, respectively). Many reviews have already been dedicated to P53, MDM2, and MDM4, while this review additionally focuses on the other factors that can deregulate P53 signaling. We discuss that P14ARF (ARF) functions as a negative regulator of MDM2, explaining the frequent loss of ARF detected in cancers. The long non-coding RNA Antisense Non-coding RNA in the INK4 Locus (ANRIL) is encoded on the same locus as ARF, inhibiting ARF expression, thus contributing to the process of tumorigenesis. Mutations in tripartite motif (TRIM) proteins deregulate P53 signaling through their ubiquitin ligase activity. Several microRNAs (miRNAs) inactivate the P53 pathway through inhibition of translation. CCCTC-binding factor (CTCF) maintains an open chromatin structure at the TP53 locus, explaining its inactivation of CTCF during tumorigenesis. P21, a downstream effector of P53, has been found to be deregulated in different tumor types. This review provides a comprehensive overview of these factors that are known to deregulate the P53 pathway in both somatic and embryonic cells, as well as their malignant counterparts (i.e., somatic and germ cell tumors). It provides insights into which aspects still need to be unraveled to grasp their contribution to tumorigenesis, putatively leading to novel targets for effective cancer therapies.
Collapse
|
14
|
Yu D, Xu Z, Cheng X, Qin J. The role of miRNAs in MDMX-p53 interplay. J Evid Based Med 2021; 14:152-160. [PMID: 33988919 DOI: 10.1111/jebm.12428] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 04/16/2021] [Accepted: 04/16/2021] [Indexed: 02/06/2023]
Abstract
MicroRNAs (miRNAs) are endogenous noncoding RNAs of 19-24 nucleotides in length and are tightly related to tumorigenesis and progression. Recent studies have demonstrated that the tumor suppressor p53 and its negative controller MDMX are regulated by miRNAs in different ways. Some miRNAs directly target p53 and regulate its expression and function, whereas some miRNAs target MDMX and regulate p53's activity indirectly. The overexpression of several miRNAs can restore the activity of p53 by negatively regulating MDMX in cancer cells. Therefore, a better understanding of the miRNAs-MDMX-p53 network will put forward potential research directions for developing anticancer therapeutics. In the present review, we mainly focus on the regulatory effects of miRNAs on the MDMX-p53 interplay as well as the role of the miRNAs-MDMX-p53 network in human cancer.
Collapse
Affiliation(s)
- Dehua Yu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhiyuan Xu
- Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Xiangdong Cheng
- Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Jiangjiang Qin
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| |
Collapse
|
15
|
Klein AM, de Queiroz RM, Venkatesh D, Prives C. The roles and regulation of MDM2 and MDMX: it is not just about p53. Genes Dev 2021; 35:575-601. [PMID: 33888565 PMCID: PMC8091979 DOI: 10.1101/gad.347872.120] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In this review, Klein et al. discuss the p53-independent roles of MDM2 and MDMX. First, they review the structural and functional features of MDM2 and MDMX proteins separately and together that could be relevant to their p53-independent activities. Following this, they summarize how these two proteins are regulated and how they can function in cells that lack p53. Most well studied as proteins that restrain the p53 tumor suppressor protein, MDM2 and MDMX have rich lives outside of their relationship to p53. There is much to learn about how these two proteins are regulated and how they can function in cells that lack p53. Regulation of MDM2 and MDMX, which takes place at the level of transcription, post-transcription, and protein modification, can be very intricate and is context-dependent. Equally complex are the myriad roles that these two proteins play in cells that lack wild-type p53; while many of these independent outcomes are consistent with oncogenic transformation, in some settings their functions could also be tumor suppressive. Since numerous small molecules that affect MDM2 and MDMX have been developed for therapeutic outcomes, most if not all designed to prevent their restraint of p53, it will be essential to understand how these diverse molecules might affect the p53-independent activities of MDM2 and MDMX.
Collapse
Affiliation(s)
- Alyssa M Klein
- Integrated Program in Cellular, Molecular, and Biomedical Studies, Columbia University, New York, New York 10032, USA
| | | | - Divya Venkatesh
- Department of Biological Sciences, Columbia University, New York, New York 10027, USA
| | - Carol Prives
- Department of Biological Sciences, Columbia University, New York, New York 10027, USA
| |
Collapse
|
16
|
Chen RX, Lu WM, Lu MP, Wang ML, Zhu XJ, Wu ZF, Tian HQ, Zhu LP, Zhang ZD, Cheng L. Polymorphisms in MicroRNA Target Sites of TGF-β Signaling Pathway Genes and Susceptibility to Allergic Rhinitis. Int Arch Allergy Immunol 2021; 182:399-407. [PMID: 33596578 PMCID: PMC8117390 DOI: 10.1159/000511975] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Accepted: 09/29/2020] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND The polymorphisms inside microRNA target sites locating in the 3'-UTR region may introduce the micro-RNA-binding changes, which may regulate the gene expression and correlate with the potential diseases. OBJECTIVES We aimed to investigate whether the polymorphisms in microRNA target sites of transforming growth factor beta (TGF-β) signaling pathway genes are associated with the susceptibility of mite-sensitized allergic rhinitis (AR) in a Han Chinese population. METHODS In this case-control study, 454 AR patients and 448 healthy controls were recruited. Three HapMap single-nucleotide polymorphisms (SNPs) were mapped to putative microRNA recognition sites and genotyped by TaqMan allelic discrimination assay. RESULTS The genotype and allele frequencies of 3 SNPs (rs1590 in TGFBR1; rs1434536 and rs17023107 in BMPR1B) showed lack of significant association with AR. However, in the subgroup analysis, the TG, GG, and TG/GG genotypes of rs1590 exhibited significantly increased risk of AR in the male subgroup (TG: adjusted OR = 1.57, 95% CI = 1.08-2.31; GG: adjusted OR = 1.76, 95% CI = 1.09-2.86; TG/GG: adjusted OR = 1.62, 95% CI = 1.13-2.33). The CT genotypes of rs17023107 might have potential to protect against AR in the patients age of <15 years (adjusted OR = 0.37, 95% CI = 0.14-0.95) and the males (adjusted OR = 0.48, 95% CI = 0.25-0.95). No significant association was found between SNPs and the total serum IgE level. CONCLUSIONS In a Han Chinese population, stratified by age and gender, susceptibility to mite-sensitized AR may be associated with 2 SNPs (rs1590 and rs17023107) in microRNA target sites of TGF-β signaling pathway genes.
Collapse
Affiliation(s)
- Ruo-Xi Chen
- Department of Otorhinolaryngology and Clinical Allergy Center, The First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Wen-Min Lu
- Department of Otorhinolaryngology, The Affiliated Changzhou No. 2 People's Hospital, Nanjing Medical University, Changzhou, China
| | - Mei-Ping Lu
- Department of Otorhinolaryngology and Clinical Allergy Center, The First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Mei-Lin Wang
- Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Xin-Jie Zhu
- Department of Otorhinolaryngology and Clinical Allergy Center, The First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Zhong-Fei Wu
- Department of Otorhinolaryngology and Clinical Allergy Center, The First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Hui-Qin Tian
- Department of Otorhinolaryngology and Clinical Allergy Center, The First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Lu-Ping Zhu
- Department of Otorhinolaryngology, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Zheng-Dong Zhang
- Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Lei Cheng
- Department of Otorhinolaryngology and Clinical Allergy Center, The First Affiliated Hospital, Nanjing Medical University, Nanjing, China,
- International Centre for Allergy Research, Nanjing Medical University, Nanjing, China,
| |
Collapse
|
17
|
Association of KLK3, VAMP8 and MDM4 Genetic Variants within microRNA Binding Sites with Prostate Cancer: Evidence from Serbian Population. Pathol Oncol Res 2020; 26:2409-2423. [PMID: 32556890 DOI: 10.1007/s12253-020-00839-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 06/10/2020] [Indexed: 12/25/2022]
Abstract
A growing number of studies have suggested that genetic variants affecting the micro-RNA- binding mechanisms (miRSNPs) constitute a promising novel class of biomarkers for prostate cancer (PCa) biology. Among the most extensively studied miRSNPs in the context of cancer is the variation rs4245739 in the MDM4 gene, while a recent large-scale analysis revealed significant differences in genotype distributions between aggressive and non-aggressive disease for rs1058205 in KLK3 and rs1010 in VAMP8. In this study, we examined a total of 1083 subjects for these three variants using Taqman® SNP Genotyping Assays. Three hundred and fifty-five samples of peripheral blood were obtained from patients with PCa and 358 samples from patients with benign prostatic hyperplasia (BPH). The control group consisted of 370 healthy volunteers. Comparisons of genotype distributions among PCa and BPH patients, as well as between PCa patients and healthy controls, yielded no evidence of association between the analyzed genetic variants and the risk of developing PCa. However, all three tested genetic variants have shown the association with the parameters of PCa progression. For KLK3 variant rs1058205, minor allele C was found to associate with the lower serum PSA score in PCa patients (PSA > 20 ng/ml vs. PSA < 10 ng/ml comparison, Prec = 0.038; ORrec = 0.20, 95%CI 0.04-1.05). The obtained results point out the potential relevance of the tested genetic variants for the disease aggressiveness assessment.
Collapse
|
18
|
Association Analysis of TP53 rs1042522, MDM2 rs2279744, rs3730485, MDM4 rs4245739 Variants and Acute Myeloid Leukemia Susceptibility, Risk Stratification Scores, and Clinical Features: An Exploratory Study. J Clin Med 2020; 9:jcm9061672. [PMID: 32492903 PMCID: PMC7355701 DOI: 10.3390/jcm9061672] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 05/18/2020] [Accepted: 05/25/2020] [Indexed: 12/20/2022] Open
Abstract
This study aimed to explore the associations between the TP53 rs1042522 (TP53 Arg72Pro), MDM2 rs2279744 (MDM2 309T>G), rs3730485 (MDM2 del1518), MDM4 rs4245739 (MDM4 34091 C>A) variants and odds of developing acute myeloid leukemia (AML) in a cohort of 809 adult subjects, consisting of 406 healthy controls and 403 AML patients. Model-based multifactor dimensionality reduction (MB-MDR) framework was used to identify the interactions of the mentioned variants and their association with AML risk. Associations of the mentioned variants with clinical features of AML, somatic mutations, and response to treatment were also evaluated. Significant associations between TP53 rs1042522 and MDM4 rs4245739 variants and AML susceptibility were noticed. MB-MDR and logistic regression analysis revealed an interaction between MDM2 rs2279744 and TP53 rs1042522, between MDM4 rs4245739 and MDM2 rs3730485, as well as significant associations with AML susceptibility. Several associations between the mentioned variants and clinical features of AML and somatic mutations were also noticed. Individually, the variant genotypes of TP53 rs1042522 and MDM4 rs4245739 were associated with AML susceptibility, but their interaction with MDM2 rs2279744 and rs3730485 modulated the risk for AML. The variant genotypes of TP53 rs1042522 were associated with adverse molecular and cytogenetic risk and also with NPM1 mutations.
Collapse
|
19
|
Pathway Analysis of Genes Identified through Post-GWAS to Underpin Prostate Cancer Aetiology. Genes (Basel) 2020; 11:genes11050526. [PMID: 32397189 PMCID: PMC7291227 DOI: 10.3390/genes11050526] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 05/02/2020] [Accepted: 05/06/2020] [Indexed: 01/22/2023] Open
Abstract
Understanding the functional role of risk regions identified by genome-wide association studies (GWAS) has made considerable recent progress and is referred to as the post-GWAS era. Annotation of functional variants to the genes, including cis or trans and understanding their biological pathway/gene network enrichments, is expected to give rich dividends by elucidating the mechanisms underlying prostate cancer. To this aim, we compiled and analysed currently available post-GWAS data that is validated through further studies in prostate cancer, to investigate molecular biological pathways enriched for assigned functional genes. In total, about 100 canonical pathways were significantly, at false discovery rate (FDR) < 0.05), enriched in assigned genes using different algorithms. The results have highlighted some well-known cancer signalling pathways, antigen presentation processes and enrichment in cell growth and development gene networks, suggesting risk loci may exert their functional effect on prostate cancer by acting through multiple gene sets and pathways. Additional upstream analysis of the involved genes identified critical transcription factors such as HDAC1 and STAT5A. We also investigated the common genes between post-GWAS and three well-annotated gene expression datasets to endeavour to uncover the main genes involved in prostate cancer development/progression. Post-GWAS generated knowledge of gene networks and pathways, although continuously evolving, if analysed further and targeted appropriately, will have an important impact on clinical management of the disease.
Collapse
|
20
|
Goodwin AJ, Li P, Halushka PV, Cook JA, Sumal AS, Fan H. Circulating miRNA 887 is differentially expressed in ARDS and modulates endothelial function. Am J Physiol Lung Cell Mol Physiol 2020; 318:L1261-L1269. [PMID: 32321279 DOI: 10.1152/ajplung.00494.2019] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Circulating microRNAs (miRNAs) can be taken up by recipient cells and have been recently associated with the acute respiratory distress syndrome (ARDS). Their role in host predisposition to the syndrome is unknown. The objective of the study was to identify circulating miRNAs associated with the development of sepsis-related ARDS and examine their impact on endothelial cell gene expression and function. We determined miRNA levels in plasma collected from subjects during the first 24 h of admission to a tertiary intensive care unit for sepsis. A miRNA that was differentially expressed between subjects who did and did not develop ARDS was identified and was transfected into human pulmonary microvascular endothelial cells (HPMECs). RNA sequencing, in silico analysis, cytokine expression, and leukocyte migration assays were used to determine the impact of this miRNA on gene expression and cell function. In two cohorts, circulating miR-887-3p levels were elevated in septic patients who developed ARDS compared with those who did not. Transfection of miR-887-3p into HPMECs altered gene expression, including the upregulation of several genes previously associated with ARDS (e.g., CXCL10, CCL5, CX3CL1, VCAM1, CASP1, IL1B, IFNB, and TLR2), and activation of cellular pathways relevant to the response to infection. Functionally, miR-887-3p increased the endothelial release of chemokines and facilitated trans-endothelial leukocyte migration. Circulating miR-887-3p is associated with ARDS in critically ill patients with sepsis. In vitro, miR-887-3p regulates the expression of genes relevant to ARDS and neutrophil tracking. This miRNA may contribute to ARDS pathogenesis and could represent a novel therapeutic target.
Collapse
Affiliation(s)
- Andrew J Goodwin
- Division of Pulmonary, Critical Care, Allergy, and Sleep Medicine, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Pengfei Li
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Perry V Halushka
- Department of Medicine, Medical University of South Carolina, Charleston, South Carolina.,Department of Pharmacology, Medical University of South Carolina, Charleston, South Carolina
| | - James A Cook
- Department of Neurosciences, Medical University of South Carolina, Charleston, South Carolina
| | - Aman S Sumal
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Hongkuan Fan
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
21
|
Fehlmann T, Sahay S, Keller A, Backes C. A review of databases predicting the effects of SNPs in miRNA genes or miRNA-binding sites. Brief Bioinform 2020; 20:1011-1020. [PMID: 29186316 DOI: 10.1093/bib/bbx155] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 10/23/2017] [Indexed: 12/16/2022] Open
Abstract
Modern precision medicine comprises the knowledge and understanding of individual differences in the genomic sequence of patients to provide tailor-made treatments. Regularly, such variants are considered in coding regions only, and their effects are predicted based on their impact on the amino acid sequence of expressed proteins. However, assessing the effects of variants in noncoding elements, in particular microRNAs (miRNAs) and their binding sites, is important as well, as a single miRNA can influence the expression patterns of many genes at the same time. To analyze the effects of variants in miRNAs and their target sites, several databases storing variant impact predictions have been published. In this review, we will compare the core functionalities and features of these databases and discuss the importance of up-to-date data resources in the context of web applications. Finally, we will outline some recommendations for future developments in the field.
Collapse
Affiliation(s)
- Tobias Fehlmann
- Chair for Clinical Bioinformatics, Saarland University, Saarbrücken, Germany
| | - Shashwat Sahay
- Chair for Clinical Bioinformatics, Saarland University, Saarbrücken, Germany
| | - Andreas Keller
- Chair for Clinical Bioinformatics, Saarland University, Saarbrücken, Germany
| | - Christina Backes
- Chair for Clinical Bioinformatics, Saarland University, Saarbrücken, Germany
| |
Collapse
|
22
|
Cohen SJ, Papoulas M, Graubardt N, Ovdat E, Loewenstein S, Kania-Almog J, Pasmanik-Chor M, Brazowski E, Cagnano E, Nachmany I, Lahat G, Klausner JM, Lubezky N. Micro-RNA Expression Patterns Predict Metastatic Spread in Solid Pseudopapillary Neoplasms of the Pancreas. Front Oncol 2020; 10:328. [PMID: 32232006 PMCID: PMC7082878 DOI: 10.3389/fonc.2020.00328] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 02/25/2020] [Indexed: 12/17/2022] Open
Abstract
Solid pseudopapillary neoplasm (SPN) of pancreas is a rare pancreatic neoplasm with a low metastatic potential. Up to 10% of patients with localized disease at presentation will develop systemic metastases, usually in the peritoneum or the liver. Due to the rarity of SPNs and the overall excellent prognosis, reliable prognostic factors to predict malignant biological behavior remain undetermined. Therefore, we aimed to define clinical, histological, and microRNA patterns that are associated with metastatic disease. We conducted a retrospective single center study on all patients operated for SPN of pancreas between 1995 and 2018. Clinical and pathological data were collected, and expression patterns of 2,578 human microRNAs were analyzed using microRNA array (Affimetrix 4.1) in normal pancreases (NPs), localized tumors (LTs), and metastatic tumors (MTs). The diagnosis of SPN was confirmed in 35 patients who included 28 females and 3 males, with a mean age of 33.8 ± 13.9 years. The only clinical factor associated with metastases was tumor size (mean tumor size 5.20 ± 3.78 in LT vs. 8.13± 1.03 in MT, p < 0.012). Microscopic features of malignancy were not associated with metastases, nor were immunohistochemical stains, including the proliferative index KI67. Higher expressions of miR-184, miR-10a, and miR-887, and lower expressions of miR-375, miR-217, and miR-200c were observed in metastatic tissues on microarray, and validated by real-time polymerase chain reaction. Hierarchal clustering demonstrated that the microRNA expression pattern of MTs was significantly different from that of LTs. The only clinical factor associated with metastases of SPN of pancreas was tumor size. Histological features and immunohistological staining were not predictive of metastases. A panel of six microRNAs was differentially expressed in MTs, and these findings could potentially be used to predict tumor behavior. Validation of these results is needed in larger series.
Collapse
Affiliation(s)
- Shmuel Jaffe Cohen
- Surgical Division Research Laboratory, Tel-Aviv Sourasky Medical Center Affiliated to the Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Michail Papoulas
- Department of Surgery, Tel-Aviv Sourasky Medical Center Affiliated to the Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Nadine Graubardt
- Surgical Division Research Laboratory, Tel-Aviv Sourasky Medical Center Affiliated to the Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Esther Ovdat
- Department of Surgery, Tel-Aviv Sourasky Medical Center Affiliated to the Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Shelly Loewenstein
- Surgical Division Research Laboratory, Tel-Aviv Sourasky Medical Center Affiliated to the Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Juliane Kania-Almog
- Surgical Division Research Laboratory, Tel-Aviv Sourasky Medical Center Affiliated to the Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | | | - Eli Brazowski
- Tel-Aviv Sourasky Medical Center Affiliated to the Sackler Faculty of Medicine, Institute of Pathology, Tel-Aviv University, Tel-Aviv, Israel
| | - Emanuela Cagnano
- Tel-Aviv Sourasky Medical Center Affiliated to the Sackler Faculty of Medicine, Institute of Pathology, Tel-Aviv University, Tel-Aviv, Israel
| | - Ido Nachmany
- Department of Surgery, Tel-Aviv Sourasky Medical Center Affiliated to the Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Guy Lahat
- Department of Surgery, Tel-Aviv Sourasky Medical Center Affiliated to the Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Joseph M Klausner
- Department of Surgery, Tel-Aviv Sourasky Medical Center Affiliated to the Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Nir Lubezky
- Surgical Division Research Laboratory, Tel-Aviv Sourasky Medical Center Affiliated to the Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel.,Department of Surgery, Tel-Aviv Sourasky Medical Center Affiliated to the Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| |
Collapse
|
23
|
Ma R, Zhao Y, He M, Zhao H, Zhang Y, Zhou S, Gao M, Di D, Wang J, Ding J, Wei M. Identifying a ten-microRNA signature as a superior prognosis biomarker in colon adenocarcinoma. Cancer Cell Int 2019; 19:360. [PMID: 31892859 PMCID: PMC6937800 DOI: 10.1186/s12935-019-1074-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 12/13/2019] [Indexed: 12/16/2022] Open
Abstract
Background Increasing studies have suggested that aberrant expression of microRNAs might play essential roles in the progression of cancers. In this study, we sought to construct a high-specific and superior microRNAs signature to improve the survival prediction of colon adenocarcinoma (COAD) patients. Methods The genome-wide miRNAs, mRNA and lncRNA expression profiles and corresponding clinical information of COAD were collected from the TCGA database. Differential expression analysis, Kaplan–Meier curve and time-dependent ROC curve were calculated and performed using R software and GraphPad Prism7. Univariate and multivariate Cox analysis was performed to evaluate the prognostic ability of signature. Functional enrichment analysis was analyzed using STRING database. Results We identified ten prognosis-related microRNAs, including seven risky factors (hsa-miR-197, hsa-miR-32, hsa-miR-887, hsa-miR-3199-2, hsa-miR-4999, hsa-miR-561, hsa-miR-210) and three protective factors (hsa-miR-3917, hsa-miR-3189, hsa-miR-6854). The Kaplan–Meier survival analysis showed that the patients with high risk score had shorter overall survival (OS) in test series. And the similar results were observed in both validation and entire series. The time-dependent ROC curve suggested this signature have high accuracy of OS for COAD. The Multivariate Cox regression analysis and stratification analysis suggested that the ten-microRNA signature was an independent factor after being adjusted with other clinical characteristics. In addition, we also found microRNA signature have higher AUC than other signature. Furthermore, we identified some miRNA-target genes that affect lymphatic metastasis and invasion of COAD patients. Conclusion In this study, we established a ten-microRNA signature as a potentially reliable and independent biomarker for survival prediction of COAD patients.
Collapse
Affiliation(s)
- Rong Ma
- 1Department of Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning China.,2Liaoning Engineering Technology Research Center, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning People's Republic of China
| | - Yanyun Zhao
- 1Department of Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning China.,2Liaoning Engineering Technology Research Center, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning People's Republic of China
| | - Miao He
- 1Department of Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning China.,2Liaoning Engineering Technology Research Center, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning People's Republic of China
| | - Hongliang Zhao
- 1Department of Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning China.,2Liaoning Engineering Technology Research Center, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning People's Republic of China
| | - Yifan Zhang
- 1Department of Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning China.,2Liaoning Engineering Technology Research Center, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning People's Republic of China
| | - Shuqi Zhou
- 1Department of Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning China.,2Liaoning Engineering Technology Research Center, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning People's Republic of China
| | - Mengcong Gao
- 1Department of Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning China.,2Liaoning Engineering Technology Research Center, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning People's Republic of China
| | - Di Di
- 1Department of Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning China.,2Liaoning Engineering Technology Research Center, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning People's Republic of China
| | - Jue Wang
- 1Department of Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning China.,2Liaoning Engineering Technology Research Center, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning People's Republic of China
| | - Jian Ding
- 1Department of Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning China.,3Division of Anti-tumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Minjie Wei
- 1Department of Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning China.,2Liaoning Engineering Technology Research Center, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning People's Republic of China
| |
Collapse
|
24
|
Estrogens Counteract Platinum-Chemosensitivity by Modifying the Subcellular Localization of MDM4. Cancers (Basel) 2019; 11:cancers11091349. [PMID: 31547268 PMCID: PMC6770881 DOI: 10.3390/cancers11091349] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 09/05/2019] [Accepted: 09/07/2019] [Indexed: 12/28/2022] Open
Abstract
Estrogen activity towards cancer-related pathways can impact therapeutic intervention. Recent omics data suggest possible crosstalk between estrogens/gender and MDM4, a key regulator of p53. Since MDM4 can either promote cell transformation or enhance DNA damage-sensitivity, we analysed in vivo impact of estrogens on both MDM4 activities. In Mdm4 transgenic mouse, Mdm4 accelerates the formation of fibrosarcoma and increases tumor sensitivity to cisplatin as well, thus confirming in vivo Mdm4 dual mode of action. Noteworthy, Mdm4 enhances chemo- and radio-sensitivity in male but not in female animals, whereas its tumor-promoting activity is not affected by mouse gender. Combination therapy of transgenic females with cisplatin and fulvestrant, a selective estrogen receptor degrader, was able to recover tumor cisplatin-sensitivity, demonstrating the relevance of estrogens in the observed sexual dimorphism. Molecularly, estrogen receptor-α alters intracellular localization of MDM4 by increasing its nuclear fraction correlated to decreased cell death, in a p53-independent manner. Importantly, MDM4 nuclear localization and intra-tumor estrogen availability correlate with decreased platinum-sensitivity and apoptosis and predicts poor disease-free survival in high-grade serous ovarian carcinoma. These data demonstrate estrogen ability to modulate chemo-sensitivity of MDM4-expressing tumors and to impinge on intracellular trafficking. They support potential usefulness of combination therapy involving anti-estrogenic drugs.
Collapse
|
25
|
Somatic Mutations in miRNA Genes in Lung Cancer-Potential Functional Consequences of Non-Coding Sequence Variants. Cancers (Basel) 2019; 11:cancers11060793. [PMID: 31181801 PMCID: PMC6627760 DOI: 10.3390/cancers11060793] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 06/06/2019] [Accepted: 06/06/2019] [Indexed: 02/07/2023] Open
Abstract
A growing body of evidence indicates that miRNAs may either drive or suppress oncogenesis. However, little is known about somatic mutations in miRNA genes. To determine the frequency and potential consequences of miRNA gene mutations, we analyzed whole exome sequencing datasets of 569 lung adenocarcinoma (LUAD) and 597 lung squamous cell carcinoma (LUSC) samples generated in The Cancer Genome Atlas (TCGA) project. Altogether, we identified 1091 somatic sequence variants affecting 522 different miRNA genes and showed that half of all cancers had at least one such somatic variant/mutation. These sequence variants occurred in most crucial parts of miRNA precursors, including mature miRNA and seed sequences. Due to our findings, we hypothesize that seed mutations may affect miRNA:target interactions, drastically changing the pool of predicted targets. Mutations may also affect miRNA biogenesis by changing the structure of miRNA precursors, DROSHA and DICER cleavage sites, and regulatory sequence/structure motifs. We identified 10 significantly overmutated hotspot miRNA genes, including the miR-379 gene in LUAD enriched in mutations in the mature miRNA and regulatory sequences. The occurrence of mutations in the hotspot miRNA genes was also shown experimentally. We present a comprehensive analysis of somatic variants in miRNA genes and show that some of these genes are mutational hotspots, suggesting their potential role in cancer.
Collapse
|
26
|
Fernandes RC, Hickey TE, Tilley WD, Selth LA. Interplay between the androgen receptor signaling axis and microRNAs in prostate cancer. Endocr Relat Cancer 2019; 26:R237-R257. [PMID: 30817318 DOI: 10.1530/erc-18-0571] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 02/26/2019] [Indexed: 12/29/2022]
Abstract
The androgen receptor (AR) is a ligand-activated transcription factor that drives prostate cancer. Since therapies that target the AR are the mainstay treatment for men with metastatic disease, it is essential to understand the molecular mechanisms underlying oncogenic AR signaling in the prostate. miRNAs are small, non-coding regulators of gene expression that play a key role in prostate cancer and are increasingly recognized as targets or modulators of the AR signaling axis. In this review, we examine the regulation of AR signaling by miRNAs and vice versa and discuss how this interplay influences prostate cancer growth, metastasis and resistance to therapy. Finally, we explore the potential clinical applications of miRNAs implicated in the regulation of AR signaling in this prevalent hormone-driven disease.
Collapse
Affiliation(s)
- Rayzel C Fernandes
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia
- Freemasons Foundation Centre for Men's Health, Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Theresa E Hickey
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Wayne D Tilley
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia
- Freemasons Foundation Centre for Men's Health, Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Luke A Selth
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia
- Freemasons Foundation Centre for Men's Health, Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
27
|
Haupt S, Mejía-Hernández JO, Vijayakumaran R, Keam SP, Haupt Y. The long and the short of it: the MDM4 tail so far. J Mol Cell Biol 2019; 11:231-244. [PMID: 30689920 PMCID: PMC6478121 DOI: 10.1093/jmcb/mjz007] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 12/16/2018] [Accepted: 01/21/2019] [Indexed: 12/27/2022] Open
Abstract
The mouse double minute 4 (MDM4) is emerging from the shadow of its more famous relative MDM2 and is starting to steal the limelight, largely due to its therapeutic possibilities. MDM4 is a vital regulator of the tumor suppressor p53. It restricts p53 transcriptional activity and also, at least in development, facilitates MDM2's E3 ligase activity toward p53. These functions of MDM4 are critical for normal cell function and a proper response to stress. Their importance for proper cell maintenance and proliferation identifies them as a risk for deregulation associated with the uncontrolled growth of cancer. MDM4 tails are vital for its function, where its N-terminus transactivation domain engages p53 and its C-terminus RING domain binds to MDM2. In this review, we highlight recently identified cellular functions of MDM4 and survey emerging therapies directed to correcting its dysregulation in disease.
Collapse
Affiliation(s)
- Sue Haupt
- Tumor Suppression Laboratory, Peter MacCallum Cancer Centre, 305 Grattan Street, Melbourne, Victoria, Australia
- Department of Clinical Pathology, The University of Melbourne, Parkville, Victoria, Australia
| | | | - Reshma Vijayakumaran
- Tumor Suppression Laboratory, Peter MacCallum Cancer Centre, 305 Grattan Street, Melbourne, Victoria, Australia
| | - Simon P Keam
- Tumor Suppression Laboratory, Peter MacCallum Cancer Centre, 305 Grattan Street, Melbourne, Victoria, Australia
| | - Ygal Haupt
- Tumor Suppression Laboratory, Peter MacCallum Cancer Centre, 305 Grattan Street, Melbourne, Victoria, Australia
- Department of Clinical Pathology, The University of Melbourne, Parkville, Victoria, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton Campus, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
28
|
Farashi S, Kryza T, Clements J, Batra J. Post-GWAS in prostate cancer: from genetic association to biological contribution. Nat Rev Cancer 2019; 19:46-59. [PMID: 30538273 DOI: 10.1038/s41568-018-0087-3] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Genome-wide association studies (GWAS) have been successful in deciphering the genetic component of predisposition to many human complex diseases including prostate cancer. Germline variants identified by GWAS progressively unravelled the substantial knowledge gap concerning prostate cancer heritability. With the beginning of the post-GWAS era, more and more studies reveal that, in addition to their value as risk markers, germline variants can exert active roles in prostate oncogenesis. Consequently, current research efforts focus on exploring the biological mechanisms underlying specific susceptibility loci known as causal variants by applying novel and precise analytical methods to available GWAS data. Results obtained from these post-GWAS analyses have highlighted the potential of exploiting prostate cancer risk-associated germline variants to identify new gene networks and signalling pathways involved in prostate tumorigenesis. In this Review, we describe the molecular basis of several important prostate cancer-causal variants with an emphasis on using post-GWAS analysis to gain insight into cancer aetiology. In addition to discussing the current status of post-GWAS studies, we also summarize the main molecular mechanisms of potential causal variants at prostate cancer risk loci and explore the major challenges in moving from association to functional studies and their implication in clinical translation.
Collapse
Affiliation(s)
- Samaneh Farashi
- Cancer Program, School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
- Australian Prostate Cancer Research Centre - Queensland, Queensland University of Technology, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Thomas Kryza
- Cancer Program, School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
- Australian Prostate Cancer Research Centre - Queensland, Queensland University of Technology, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Judith Clements
- Cancer Program, School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
- Australian Prostate Cancer Research Centre - Queensland, Queensland University of Technology, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Jyotsna Batra
- Cancer Program, School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia.
- Australian Prostate Cancer Research Centre - Queensland, Queensland University of Technology, Translational Research Institute, Woolloongabba, Queensland, Australia.
| |
Collapse
|
29
|
Liu Z, Ren L, Tian J, Liu N, Hu Y, Zhang P. Comprehensive Analysis of Long Noncoding RNAs and Messenger RNAs Expression Profiles in Patients with Marjolin Ulcer. Med Sci Monit 2018; 24:7828-7840. [PMID: 30385735 PMCID: PMC6228116 DOI: 10.12659/msm.911177] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Background Marjolin ulcer (MU) is an aggressive cutaneous malignancy. Typically, MU occurs over a period of time in post-burn and/or post-traumatic lesions and scars. However, the pathogenesis of scar carcinogenesis and MU development remains to be elucidated. The present study aimed to investigate the long noncoding RNA (lncRNA) and messenger RNA (mRNA) expression profiling in MU, which could provide new information on the potential molecular mechanisms of MU development. Material/Methods The lncRNA microarray analysis was conducted in normal skin, scar, and MU tissue, and quantitative real-time PCR experiment was carried out to validate the reliability of the microarray data. Furthermore, a series of integrative bioinformatic approaches were applied to decipher the function of differentially expressed lncRNAs. Results A total of 7130 lncRNAs and 9867 mRNAs were differentially expressed among normal skin, scar, and MU tissues. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway analysis demonstrated that these aberrantly expressed transcripts were mainly involved in cell cycle, immune response, and the p53 signaling pathway. Series Test of Cluster analysis indicated certain dysregulated lncRNAs were expressed with a gradually increasing or decreasing trend and might participated in malignant transformation of scar tissue postburn. Co-expression analysis showed 5 selected lncRNAs might regulate cell proliferation through the p53 signaling pathway. Finally, the competing endogenous RNA (ceRNA) network indicated that lncRNA uc001oou.3 might be implicated in ceRNA mechanism during MU development. Conclusions Taken together, our study implied the aberrant expression of lncRNAs may play an important role in the pathogenesis and development of MU, and the exact mechanism warrants further investigation.
Collapse
Affiliation(s)
- Zan Liu
- Department of Burns and Reconstructive Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China (mainland)
| | - Licheng Ren
- Department of Burns and Reconstructive Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China (mainland)
| | - Jing Tian
- Department of Burns and Reconstructive Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China (mainland)
| | - Ning Liu
- Department of Burns and Reconstructive Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China (mainland)
| | - Yanke Hu
- Department of Burns and Reconstructive Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China (mainland)
| | - Pihong Zhang
- Department of Burns and Reconstructive Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China (mainland)
| |
Collapse
|
30
|
Association of Single Nucleotide Polymorphisms of the MDM4 Gene With the Susceptibility to Breast Cancer in a Southeast Iranian Population Sample. Clin Breast Cancer 2018; 18:e883-e891. [DOI: 10.1016/j.clbc.2018.01.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 01/12/2018] [Accepted: 01/13/2018] [Indexed: 01/14/2023]
|
31
|
Farina NH, Zingiryan A, Vrolijk MA, Perrapato SD, Ades S, Stein GS, Lian JB, Landry CC. Nanoparticle-based targeted cancer strategies for non-invasive prostate cancer intervention. J Cell Physiol 2018; 233:6408-6417. [PMID: 29663383 DOI: 10.1002/jcp.26593] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 03/09/2018] [Indexed: 01/07/2023]
Abstract
Prostate cancer is screened by testing circulating levels of the prostate-specific antigen (PSA) biomarker, monitoring changes over time, or a digital rectal exam. Abnormal results often lead to prostate biopsy. Prostate cancer positive patients are stratified into very low-risk, low-risk, intermediate-risk, and high-risk, based on clinical classification parameters, to assess therapy options. However, there remains a gap in our knowledge and a compelling need for improved risk stratification to inform clinical decisions and reduce both over-diagnosis and over-treatment. Further, current strategies for clinical intervention do not distinguish clinically aggressive prostate cancer from indolent disease. This mini-review takes advantage of a large number of functionally characterized microRNAs (miRNA), epigenetic regulators of prostate cancer, that define prostate cancer cell activity, tumor stage, and circulate as biomarkers to monitor disease progression. Nanoparticles provide an effective platform for targeted delivery of miRNA inhibitors or mimics specifically to prostate tumor cells to inhibit cancer progression. Several prostate-specific transmembrane proteins expressed at elevated levels in prostate tumors are under investigation for targeting therapeutic agents to prostate cancer cells. Given that prostate cancer progresses slowly, circulating miRNAs can be monitored to identify tumor progression in indolent disease, allowing identification of miRNAs for nanoparticle intervention before the crucial point of transition to aggressive disease. Here, we describe clinically significant and non-invasive intervention nanoparticle strategies being used in clinical trials for drug and nucleic acid delivery. The advantages of mesoporous silica-based nanoparticles and a number of candidate miRNAs for inhibition of prostate cancer are discussed.
Collapse
Affiliation(s)
- Nicholas H Farina
- Department of Biochemistry, Larner College of Medicine, University of Vermont, Burlington, Vermont.,UVM Cancer Center, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - Areg Zingiryan
- Department of Biochemistry, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - Michael A Vrolijk
- Department of Chemistry, College of Arts and Sciences, University of Vermont, Burlington, Vermont
| | - Scott D Perrapato
- UVM Cancer Center, Larner College of Medicine, University of Vermont, Burlington, Vermont.,Department of Surgery, Division of Urology, Larner College of Medicine, University of Vermont Medical Center, Burlington, Vermont
| | - Steven Ades
- UVM Cancer Center, Larner College of Medicine, University of Vermont, Burlington, Vermont.,Department of Medicine, Division of Hematology and Oncology, Larner College of Medicine, University of Vermont Medical Center, Burlington, Vermont
| | - Gary S Stein
- Department of Biochemistry, Larner College of Medicine, University of Vermont, Burlington, Vermont.,UVM Cancer Center, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - Jane B Lian
- Department of Biochemistry, Larner College of Medicine, University of Vermont, Burlington, Vermont.,UVM Cancer Center, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - Christopher C Landry
- Department of Chemistry, College of Arts and Sciences, University of Vermont, Burlington, Vermont
| |
Collapse
|
32
|
Wang MJ, Luo YJ, Shi ZY, Xu XL, Yao GL, Liu RP, Zhao H. The associations between MDM4 gene polymorphisms and cancer risk. Oncotarget 2018; 7:55611-55623. [PMID: 27742919 PMCID: PMC5342440 DOI: 10.18632/oncotarget.10877] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 07/10/2016] [Indexed: 01/01/2023] Open
Abstract
Considerable studies have investigated the associations between MDM4 gene polymorphisms and cancer risk recently, but with contradictory results. The aim of this meta-analysis was to evaluate the associations between MDM4 gene polymorphisms and cancer risk. Relevant studies were identified by a systematic search of PubMed, Embase, and CNKI databases. Crude odds ratios (ORs) and 95% confidence intervals (CIs) were used to describe the strength of the associations. Fifty-six studies published in 11 publications involving 18,910 cases and 51,609 controls were included in this meta-analysis. Five MDM4 gene polymorphisms were evaluated: rs4245739, rs1563828, rs11801299, rs10900598, and rs1380576. Our analyses suggested that the rs4245739 polymorphism was significantly associated with overall cancer risk. Furthermore, stratification analyses of ethnicity indicated that rs4245739 decreased the risk of cancer among the Asian population, and stratification analyses of smoking status indicated that rs4245739 decreased the risk of cancer among nonsmokers. However, stratification analyses of cancer type and sex suggested that rs4245739 was not related to cancer risk. There were no associations of rs1563828, rs11801299, rs10900598, or rs1380576 with overall cancer risk. In conclusion, our analyses indicated that rs4245739 polymorphism in the MDM4 gene may play an important role in the etiology of cancer.
Collapse
Affiliation(s)
- Ming-Jie Wang
- Department of Orthopedics, Affiliated Hospital of Nanjing Medical University, Changzhou Second People's Hospital, Changzhou 213003, China
| | - Yong-Jun Luo
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Zhi-Yong Shi
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, China National Clinical Research Center for Neurological Diseases, Beijing 100050, China
| | - Xiao-Liang Xu
- Liver Surgery of Jiangsu Province People's Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Guo-Liang Yao
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Rui-Ping Liu
- Department of Orthopedics, Affiliated Hospital of Nanjing Medical University, Changzhou Second People's Hospital, Changzhou 213003, China
| | - Hui Zhao
- Department of General Surgery, Third Affiliated Hospital of Nantong University, Wuxi, 214000, China
| |
Collapse
|
33
|
Qin JJ, Li X, Wang W, Zi X, Zhang R. Targeting the NFAT1-MDM2-MDMX Network Inhibits the Proliferation and Invasion of Prostate Cancer Cells, Independent of p53 and Androgen. Front Pharmacol 2017; 8:917. [PMID: 29311926 PMCID: PMC5735069 DOI: 10.3389/fphar.2017.00917] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 11/30/2017] [Indexed: 12/31/2022] Open
Abstract
The MDM2 and MDMX oncogenes are overexpressed in various types of human cancer and are highly associated with the initiation, progression, metastasis and chemotherapeutic resistance of these diseases, including prostate cancer. The present study was designed to test a natural MDM2 inhibitor, Inulanolide A (InuA), for anti-prostate cancer activity and to determine the underlying mechanism(s) of action. InuA directly bound to the RING domains of both MDM2 and MDMX with high affinity and specificity and disrupted MDM2-MDMX binding, markedly enhancing MDM2 protein degradation. We further discovered that InuA bound to the DNA binding domain of NFAT1, resulting in marked inhibition of MDM2 transcription. InuA inhibited the proliferation, migration, and invasion of prostate cancer cells, regardless of their p53 status and AR responsiveness. Double knockdown of MDM2 and NFAT1 also revealed that the expression of both of these molecules is important for InuA’s inhibitory effects on the proliferation and invasion of prostate cancer cells. In summary, InuA represents a novel class of bifunctional MDM2 inhibitors, and should be further investigated as a candidate lead compound for prostate cancer prevention and therapy.
Collapse
Affiliation(s)
- Jiang-Jiang Qin
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, United States.,Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, United States
| | - Xin Li
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, United States.,Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, United States
| | - Wei Wang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, United States.,Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, United States.,Center for Drug Discovery, University of Houston, Houston, TX, United States
| | - Xiaolin Zi
- Department of Urology, University of California, Irvine, Irvine, CA, United States.,Department of Pharmacology, University of California, Irvine, Irvine, CA, United States
| | - Ruiwen Zhang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, United States.,Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, United States.,Center for Drug Discovery, University of Houston, Houston, TX, United States
| |
Collapse
|
34
|
Anwar SL, Wulaningsih W, Watkins J. Profile of the breast cancer susceptibility marker rs4245739 identifies a role for miRNAs. Cancer Biol Med 2017; 14:387-395. [PMID: 29372105 PMCID: PMC5785168 DOI: 10.20892/j.issn.2095-3941.2017.0050] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 08/16/2017] [Indexed: 12/31/2022] Open
Abstract
OBJECTIVE To determine the influence of the single nucleotide polymorphism (SNP) rs4245739 on the binding and expression of microRNAs and subsequent MDM4 expression and the correlation of these factors with clinical determinants of ER-negative breast cancers. METHODS FindTar and miRanda were used to detect the manner in which potential microRNAs are affected by the SNP rs4245739-flanking sequence. RNA sequencing data for ER-negative breast cancer from The Cancer Genome Atlas (TCGA) were used to compare the expression of miR-184, miR-191, miR-193a, miR-378, and MDM4 in different rs4245739 genotypes. RESULTS Comparison of ER-negative cancer patients with and without the expression of miR-191 as well as profile microRNAs (miR-184, miR-191, miR-193a and miR-378 altogether) can differentiate the expression of MDM4 among different rs4245739 genotypes. Although simple genotyping alone did not reveal significant clinical relationships, the combination of genotyping and microRNA profiles was able to significantly differentiate individuals with larger tumor size and lower number of involved lymph nodes (P < 0.05) in the risk group (A allele). CONCLUSIONS We present two novel methods to analyze SNPs within 3'UTRs that use: (i) a single miRNA marker expression and (ii) an expression profile of miRNAs predicted to bind to the SNP region. We demonstrate that the application of these two methods, in particular the miRNA profile approach, permits detection of new molecular and clinical features related to the rs4245739 variant in ER-negative breast cancer.
Collapse
Affiliation(s)
- Sumadi Lukman Anwar
- PILAR Research Network, Cambridgeshire CB1 2JD, UK
- Division of Surgical Oncology, Department of Surgery, Faculty of Medicine Universitas Gadjah Mada, Yogyakarta 55281, Indonesia
| | - Wahyu Wulaningsih
- PILAR Research Network, Cambridgeshire CB1 2JD, UK
- Division of Surgical Oncology, Department of Surgery, Faculty of Medicine Universitas Gadjah Mada, Yogyakarta 55281, Indonesia
- Division of Hemato-Oncology, Department of Internal Medicine, Faculty of Medicine Universitas Gadjah Mada, Yogyakarta 55281, Indonesia
| | - Johnathan Watkins
- PILAR Research Network, Cambridgeshire CB1 2JD, UK
- Division of Surgical Oncology, Department of Surgery, Faculty of Medicine Universitas Gadjah Mada, Yogyakarta 55281, Indonesia
| |
Collapse
|
35
|
Variations within 3'-UTR of MDM4 gene contribute to clinical outcomes of advanced non-small cell lung cancer patients following platinum-based chemotherapy. Oncotarget 2017; 8:16313-16324. [PMID: 27462918 PMCID: PMC5369965 DOI: 10.18632/oncotarget.10771] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Accepted: 06/17/2016] [Indexed: 01/22/2023] Open
Abstract
Single-nucleotide polymorphism (SNPs) in microRNA (miRNA)-binding sites may modulate the posttranscriptional regulation of gene expression and explain individual sensitivity to platinum agents. This study aimed to investigate the impact of SNPs located at 3′-untranslated region (UTR) of MDM4 gene, on clinical outcomes of advanced non-small cell lung cancer (NSCLC) patients. Four SNPs were genotyped by using DNA from blood samples of advanced NSCLC patients (642 in the Discovery set and 330 in the Replication set) and were analyzed the relationships with clinical outcomes. Carriers with rs10900598 CC genotype and rs4245739 AC genotype showed increased overall survival (OS) than those with AA genotype (P = 0.017 and P = 0.037, respectively) in the Discovery set and after pooling results from the Replication set. A combined effect on survival of variant alleles was also concluded and validated. Stratification analysis revealed that the effect of MDM4 SNPs was more pronounced in lung adenocarcinoma (LAC) subgroups. A reduced expression of the reporter gene for the C allele of rs4245739 was observed in NSCLC cells using luciferase reporter gene assays. Taken together, our results demonstrate that genetic variations in 3′-UTR of MDM4 gene may influence outcomes of advanced NSCLC by miRNAs-mediated regulation.
Collapse
|
36
|
Moszyńska A, Gebert M, Collawn JF, Bartoszewski R. SNPs in microRNA target sites and their potential role in human disease. Open Biol 2017; 7:170019. [PMID: 28381629 PMCID: PMC5413909 DOI: 10.1098/rsob.170019] [Citation(s) in RCA: 150] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Accepted: 03/08/2017] [Indexed: 12/14/2022] Open
Abstract
In the post-genomic era, the goal of personalized medicine is to determine the correlation between genotype and phenotype. Developing high-throughput genotyping technologies such as genome-wide association studies (GWAS) and the 1000 Genomes Project (http://www.internationalgenome.org/about/#1000G_PROJECT) has dramatically enhanced our ability to map where changes in the genome occur on a population level by identifying millions of single nucleotide polymorphisms (SNPs). Polymorphisms, particularly those within the coding regions of proteins and at splice junctions, have received the most attention, but it is also now clear that polymorphisms in the non-coding regions are important. In these non-coding regions, the enhancer and promoter regions have received the most attention, whereas the 3'-UTR regions have until recently been overlooked. In this review, we examine how SNPs affect microRNA-binding sites in these regions, and how mRNA stability changes can lead to disease pathogenesis.
Collapse
Affiliation(s)
- Adrianna Moszyńska
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Gdansk, Poland
| | - Magdalena Gebert
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Gdansk, Poland
| | - James F Collawn
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Rafał Bartoszewski
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Gdansk, Poland
| |
Collapse
|
37
|
The role of MDM4 SNP34091 A>C polymorphism in cancer: a meta-analysis on 19,328 patients and 51,058 controls. Int J Biol Markers 2017; 32:e62-e67. [PMID: 27646776 DOI: 10.5301/jbm.5000228] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/21/2016] [Indexed: 12/30/2022]
Abstract
BACKGROUND Cancer is one of the leading causes of death in the world. Several observational studies have suggested a significant association of the MDM4 SNP34091 A>C polymorphism with cancers. However, the results of the published studies are inconsistent. MATERIALS AND METHODS PubMed, Embase/Ovid and the Chinese National Knowledge Infrastructure were searched for relevant studies with a time limit of April 20, 2016. Pooled odds ratios (ORs) with 95% confidence intervals (CIs) were used to evaluate the strength of the association between MDM4 polymorphism and cancer risk. Publication bias was estimated using Begg's funnel plots and Egger's regression test. RESULTS A total of 19,328 patients and 51,058 controls were included in the analysis. Overall, a significantly decreased risk of cancer was associated with MDM4 SNP34091 polymorphism for the allele model (C vs. A, OR = 0.715, 95% CI: 0.622-0.821, p = 0.000), dominant model (CC + AC vs. AA, OR = 0.684, 95% CI: 0.563-0.831, p = 0.000), recessive model (CC vs. AC + AA, OR = 1.139, 95% CI = 1.055-1.230, p = 0.001) and heterozygote model (AC vs. AA, OR = 0.687, 95% CI = 0.568-0.832). In the subgroup analysis by cancer type, no significant association was found in the breast cancer subgroup. In the subgroup analysis by geographical region, 2 genetic models, the allele and heterozygote models, showed a significant association in Chinese populations. CONCLUSIONS The results of our meta-analysis showed that the MDM4 SNP34091 A>C polymorphism may function as a protective factor against cancer risk.
Collapse
|
38
|
Essential Roles of E3 Ubiquitin Ligases in p53 Regulation. Int J Mol Sci 2017; 18:ijms18020442. [PMID: 28218667 PMCID: PMC5343976 DOI: 10.3390/ijms18020442] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2016] [Revised: 02/10/2017] [Accepted: 02/11/2017] [Indexed: 01/30/2023] Open
Abstract
The ubiquitination pathway and proteasomal degradation machinery dominantly regulate p53 tumor suppressor protein stability, localization, and functions in both normal and cancerous cells. Selective E3 ubiquitin ligases dominantly regulate protein levels and activities of p53 in a large range of physiological conditions and in response to cellular changes induced by exogenous and endogenous stresses. The regulation of p53’s functions by E3 ubiquitin ligases is a complex process that can lead to positive or negative regulation of p53 protein in a context- and cell type-dependent manner. Accessory proteins bind and modulate E3 ubiquitin ligases, adding yet another layer of regulatory control for p53 and its downstream functions. This review provides a comprehensive understanding of p53 regulation by selective E3 ubiquitin ligases and their potential to be considered as a new class of biomarkers and therapeutic targets in diverse types of cancers.
Collapse
|
39
|
Zhai Y, Dai Z, He H, Gao F, Yang L, Dong Y, Lu J. A PRISMA-compliant meta-analysis of MDM4 genetic variants and cancer susceptibility. Oncotarget 2016; 7:73935-73944. [PMID: 27738340 PMCID: PMC5342025 DOI: 10.18632/oncotarget.12558] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 10/04/2016] [Indexed: 12/31/2022] Open
Abstract
Molecular epidemiological research suggests that mouse double minute 4 (MDM4) polymorphisms may be associated with cancer susceptibility, but results remain controversial. To derive a more precise evaluation, we performed a PRISMA compliant meta-analysis focused on five single nucleotide polymorphisms (rs11801299, rs1380576, rs10900598, rs1563828, and rs4245739) of MDM4. Overall, 23 studies involving 22,218 cases and 55,033 controls were analyzed. The results showed that rs4245739 was significantly associated with a decreased cancer risk in the allelic (C vs. A: odds ratio [OR] = 0.848, 95% confidence interval [CI] = 0.765-0.941, P = 0.002), heterozygous (AC vs. AA: OR = 0.831, 95% CI = 0.735-0.939, P = 0.003), and dominant (AC+CC vs. A: OR = 0.823, 95% CI = 0.727-0.932, P = 0.002) models. The association was more prominent in Asians. No significant association was found using any genetic model for the rs11801299, rs1380576, rs10900598, and rs1563828 SNPs. These results indicate that the rs4245739 polymorphism may contribute to a decreased cancer susceptibility and support the hypothesis that genetic variants in the MDM4 genes act as important modifiers of cancer risk.
Collapse
Affiliation(s)
- Yajing Zhai
- Department of Pharmacy, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Zhijun Dai
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Hairong He
- Clinical Research Center, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Fan Gao
- Clinical Research Center, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Lihong Yang
- Clinical Research Center, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Yalin Dong
- Department of Pharmacy, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Jun Lu
- Clinical Research Center, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| |
Collapse
|
40
|
Matin F, Jeet V, Clements JA, Yousef GM, Batra J. MicroRNA Theranostics in Prostate Cancer Precision Medicine. Clin Chem 2016; 62:1318-33. [DOI: 10.1373/clinchem.2015.242800] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 06/10/2016] [Indexed: 01/07/2023]
Abstract
Abstract
BACKGROUND
Prostate cancer is the second most frequently diagnosed cancer in men worldwide. Theranostics, a combination of diagnostics and therapeutics, is an emerging concept in the field of precision medicine, and microRNAs (miRNAs) are predictive pioneers in this area.
CONTENT
miRNAs are small endogenous noncoding RNA molecules that regulate gene expression posttranscriptionally by targeting messenger RNAs. More than 60% of all protein coding genes are controlled by miRNAs, which makes them powerful regulators of the different cellular processes involved in the pathogenesis of various types of cancer, including prostate cancer. Growing evidence indicates the differential expression of miRNAs in tumor tissues. In addition, miRNAs in body fluids, known as circulating miRNAs, are present in remarkably stable forms and their alteration in prostate cancer has been well documented. Circulating miRNAs are known to originate from tumor tissues, thereby enabling intercellular communication via carriers to promote tumorigenesis and malignancy. In addition, fueled by recent advances, the use of miRNA-based anticancer therapies has been proposed with the onset of early phase clinical trials to assess the therapeutic efficacy of miRNAs.
SUMMARY
In this review, we summarize the theranostic utility of miRNAs and outline their diagnostic and prognostic potential in prostate cancer. In addition, we discuss the current detection methodologies and emerging innovative strategies for the detection of miRNAs in body fluids and tumor tissues in the clinical setting. We also provide insight into the current and future therapeutic potential of miRNAs in prostate cancer.
Collapse
Affiliation(s)
- Farhana Matin
- School of Biomedical Sciences, Faculty of Health, Institute of Health and Biomedical Innovation, Queensland University of Technology, Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), Australia
- Translational Research Institute, Queensland University of Technology, Brisbane, Australia
| | - Varinder Jeet
- School of Biomedical Sciences, Faculty of Health, Institute of Health and Biomedical Innovation, Queensland University of Technology, Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), Australia
- Translational Research Institute, Queensland University of Technology, Brisbane, Australia
| | - Judith A Clements
- School of Biomedical Sciences, Faculty of Health, Institute of Health and Biomedical Innovation, Queensland University of Technology, Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), Australia
- Translational Research Institute, Queensland University of Technology, Brisbane, Australia
| | - George M Yousef
- Molecular Diagnostics, Department of Laboratory Medicine, St. Michael's Hospital, Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Jyotsna Batra
- School of Biomedical Sciences, Faculty of Health, Institute of Health and Biomedical Innovation, Queensland University of Technology, Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), Australia
- Translational Research Institute, Queensland University of Technology, Brisbane, Australia
| |
Collapse
|
41
|
Abstract
The tumor suppressor gene TP53 is the most frequently mutated gene in human cancer; this gene is subject to inactivation by mutation or deletion in >50% of sporadic cancers. Genes that encode proteins that regulate p53 function, such as MDM2, MDM4, and CDKN2A (p14(ARF)) are also frequently altered in tumors, and it is generally believed that the p53 pathway is likely to be inactivated by mutation in close to 100% of human tumors. Unlike most other cancer-relevant signaling pathways, some of the genes in the p53 pathway contain functionally significant single nucleotide polymorphisms (SNPs) that alter the amplitude of signaling by this protein. These variants, thus, have the potential to impact cancer risk, progression, and the efficacy of radiation and chemotherapy. In addition, the p53 pathway plays a role in other biological processes, including metabolism and reproductive fitness, so these variants have the potential to modify other diseases as well. Here we have chosen five polymorphisms in three genes in the p53 pathway for review, two in TP53, two in MDM2, and one in MDM4. These five variants were selected based on the quality and reproducibility of functional data associated with them, as well as the convincingness of epidemiological data in support of their association with disease. We also highlight two other polymorphisms that may affect p53 signaling, but for which functional or association data are still forthcoming (KITLG and ANRIL). Finally, we touch on three questions regarding genetic modifiers of the p53 pathway: Why did these variants arise? Were they under selection pressure? And, is there compelling evidence to support genotyping these variants to better predict disease risk and prognosis?
Collapse
Affiliation(s)
- Subhasree Basu
- Program in Molecular and Cellular Oncogenesis, The Wistar Institute, Philadelphia, Pennsylvania 19104
| | - Maureen E Murphy
- Program in Molecular and Cellular Oncogenesis, The Wistar Institute, Philadelphia, Pennsylvania 19104
| |
Collapse
|
42
|
Hashemi M, Moradi N, Ziaee SAM, Narouie B, Soltani MH, Rezaei M, Shahkar G, Taheri M. Association between single nucleotide polymorphism in miR-499, miR-196a2, miR-146a and miR-149 and prostate cancer risk in a sample of Iranian population. J Adv Res 2016; 7:491-8. [PMID: 27222754 DOI: 10.1016/j.jare.2016.03.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 03/23/2016] [Accepted: 03/24/2016] [Indexed: 01/29/2023] Open
Abstract
MicroRNAs (miRNAs) play an important role in regulating gene expression at the post-transcriptional level and are involved in numerous physiological processes. Accumulating evidence suggests that single-nucleotide polymorphisms (SNPs) in human miRNA genes may affect miRNA biogenesis pathway and influence the susceptibility to several diseases such as cancer. The present study aimed to evaluate the impact of miR-499 rs3746444, miR-196a2 rs11614913, miR-149 rs2292832, and miR-146a rs2910164 polymorphisms on prostate cancer (PCa) risk in a sample of Iranian population. This case-control study was done on 169 patients with pathologically confirmed PCa and 182 benign prostatic hyperplasia (BPH). The genotyping assays were done using T-ARMS-PCR or PCR-RFLP methods. The findings indicated that CC genotype of miR-499 rs3746444 polymorphism increased the risk of PCa (OR = 1.76, 95% CI = 1.12-2.79, P = 0.019) compared to TT genotype. No statistically significant association was found between miR-196a2 rs11614913, miR-149 rs2292832, and miR-146a rs2910164 polymorphisms and PCa risk. In summary, the findings indicated that miR-499 rs3746444 polymorphism increased the risk of PCa in an Iranian population. Further studies with larger sample sizes and different ethnicities are necessary to verify the findings of the present study.
Collapse
Affiliation(s)
- Mohammad Hashemi
- Cellular and Molecular Research Center, Zahedan University of Medical Sciences, Zahedan, Iran; Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Nazanin Moradi
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Seyed Amir Mohsen Ziaee
- Urology and Nephrology Research Center, Department of Urology, Shahid Labbafinejad Medical Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Behzad Narouie
- Urology and Nephrology Research Center, Department of Urology, Shahid Labbafinejad Medical Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Hosein Soltani
- Urology and Nephrology Research Center, Department of Urology, Shahid Labbafinejad Medical Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Rezaei
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Ghazaleh Shahkar
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Mohsen Taheri
- Genetics of Non Communicable Disease Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| |
Collapse
|
43
|
Gansmo LB, Bjørnslett M, Halle MK, Salvesen HB, Dørum A, Birkeland E, Hveem K, Romundstad P, Vatten L, Lønning PE, Knappskog S. The MDM4 SNP34091 (rs4245739) C-allele is associated with increased risk of ovarian-but not endometrial cancer. Tumour Biol 2016; 37:10697-702. [PMID: 26867771 PMCID: PMC4999457 DOI: 10.1007/s13277-016-4940-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 01/29/2016] [Indexed: 12/17/2022] Open
Abstract
The MDM4 protein (also known as MDMX or HDMX) is a negative regulator of p53, not only by direct interaction but also through its interaction with MDM2. Further, MDM4 overexpression and amplification have been observed in several cancer forms. Recently, a single nucleotide polymorphism (SNP) in the 3’ untranslated region of the MDM4 gene, SNP34091A > C (rs4245739) was reported to alter MDM4 messenger RNA (mRNA) stability by modulating a microRNA binding site, thereby leading to decreased MDM4 levels. In this case-control study, we aimed to evaluate the possible association between MDM4 SNP34091 status and cancer risk by comparing the genotype frequencies in large hospital-based cohorts of endometrial- (n = 1404) and ovarian (n = 1385) cancer patients with healthy female controls (n = 1870). Genotype frequencies were compared by odds ratio (OR) estimates and Fisher exact tests. We found that individuals harboring the MDM4 SNP34091AC/CC genotypes had a significantly elevated risk for serous ovarian cancer (SOC) in general and high-grade serous ovarian cancer (HGSOC) in particular (SOC: OR = 1.18., 95 % CI = 1.01–1.39; HGSOC: OR = 1.25, CI = 1.02–1.53). No association between SNP34091 genotypes and endometrial cancer risk was observed. Our data indicate the MDM4 SNP34091AC/CC genotypes to be associated with an elevated risk for SOC and in particular the HGSOC type.
Collapse
MESH Headings
- 3' Untranslated Regions/genetics
- Adenocarcinoma, Clear Cell/epidemiology
- Adenocarcinoma, Clear Cell/genetics
- Adenocarcinoma, Mucinous/epidemiology
- Adenocarcinoma, Mucinous/genetics
- Alleles
- Carcinoma, Endometrioid/epidemiology
- Carcinoma, Endometrioid/genetics
- Case-Control Studies
- Cell Cycle Proteins
- Cystadenocarcinoma, Serous/epidemiology
- Cystadenocarcinoma, Serous/genetics
- Endometrial Neoplasms/epidemiology
- Endometrial Neoplasms/genetics
- Female
- Gene Frequency
- Genes, Neoplasm
- Genetic Predisposition to Disease
- Genotype
- Humans
- Neoplasm Proteins/genetics
- Norway/epidemiology
- Nuclear Proteins/genetics
- Odds Ratio
- Ovarian Neoplasms/epidemiology
- Ovarian Neoplasms/genetics
- Polymorphism, Single Nucleotide
- Proto-Oncogene Proteins/genetics
Collapse
Affiliation(s)
- Liv B Gansmo
- Section of Oncology, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Oncology, Haukeland University Hospital, Bergen, Norway
| | - Merete Bjørnslett
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Mari Kyllesø Halle
- Department of Gynecology and Obstetrics, Haukeland University Hospital, Bergen, Norway
- Center for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Helga B Salvesen
- Department of Gynecology and Obstetrics, Haukeland University Hospital, Bergen, Norway
- Center for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Anne Dørum
- Department of Gynecologic Oncology, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway
| | - Einar Birkeland
- Section of Oncology, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Oncology, Haukeland University Hospital, Bergen, Norway
| | - Kristian Hveem
- Department of Public Health, Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Pål Romundstad
- Department of Public Health, Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Lars Vatten
- Department of Public Health, Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Per Eystein Lønning
- Section of Oncology, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Oncology, Haukeland University Hospital, Bergen, Norway
| | - Stian Knappskog
- Section of Oncology, Department of Clinical Science, University of Bergen, Bergen, Norway.
- Department of Oncology, Haukeland University Hospital, Bergen, Norway.
| |
Collapse
|
44
|
Xu W, Luo F, Sun B, Ye H, Li J, Shi L, Liu Y, Lu X, Wang B, Wang Q, Liu Q, Zhang A. HIF-2α, acting via miR-191, is involved in angiogenesis and metastasis of arsenite-transformed HBE cells. Toxicol Res (Camb) 2016; 5:66-78. [PMID: 30090327 PMCID: PMC6060623 DOI: 10.1039/c5tx00225g] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 11/02/2015] [Indexed: 12/11/2022] Open
Abstract
Arsenic is a well established human carcinogen that causes diseases of the lung. Some studies have suggested that hypoxia-inducible factors (HIFs) and microRNAs (miRNAs) are involved in human lung cancer; however, their molecular mechanisms that causally contribute to arsenite-caused malignant transformation of cells remain unclear. To elucidate the mechanisms of angiogenesis and metastasis of lung cancer caused by arsenite, we investigated the role of HIF-2α regulation of miRNA-191 (miR-191) in the angiogenic and metastatic properties of human bronchial epithelial (HBE) cells transformed by arsenite. In HBE cells, HIF-2α binds to the hypoxia response element (HRE) in the promoter region of miR-191 and initiates transcription of miR-191. Blocking of HIF-2α with siRNA inhibited the up-regulation of miR-191, Wilms' tumor 1 (WT1) protein, matrix metalloproteinase 9 (MMP-9), vascular endothelial growth factor (VEGF), and the down-regulation of brain acid-soluble protein 1 (BASP1). In arsenite-transformed HBE (T-HBE) cells, down-regulation of HIF-2α by siRNA blocked the process of angiogenesis and decreased their neoplastic properties and metastatic capacity, which were reversed by over-expression of miR-191 or by up-regulating WT1. Thus, HIF-2α up-regulates WT1 via miR-191, both of which are involved in the angiogenesis and metastasis of T-HBE cells. The results present a better understanding of the processes involved in lung cancer caused by arsenite exposure.
Collapse
Affiliation(s)
- Wenchao Xu
- Institute of Toxicology , School of Public Health , Nanjing Medical University , Nanjing 211166 , Jiangsu , People's Republic of China
- The Key Laboratory of Modern Toxicology , Ministry of Education , School of Public Health , Nanjing Medical University , Nanjing 211166 , Jiangsu , People's Republic of China . ; ; Tel: +86-25-8686-8424
| | - Fei Luo
- Institute of Toxicology , School of Public Health , Nanjing Medical University , Nanjing 211166 , Jiangsu , People's Republic of China
- The Key Laboratory of Modern Toxicology , Ministry of Education , School of Public Health , Nanjing Medical University , Nanjing 211166 , Jiangsu , People's Republic of China . ; ; Tel: +86-25-8686-8424
| | - Baofei Sun
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control , Ministry of Education , School of Public Health , Guiyang Medical University , Guiyang 550025 , Guizhou , People's Republic of China . ; ; Tel: +86-851-8841-6171
| | - Hua Ye
- School of Medicine , Yangzhou University , Yangzhou 225009 , Jiangsu , People's Republic of China
| | - Jun Li
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control , Ministry of Education , School of Public Health , Guiyang Medical University , Guiyang 550025 , Guizhou , People's Republic of China . ; ; Tel: +86-851-8841-6171
| | - Le Shi
- Institute of Toxicology , School of Public Health , Nanjing Medical University , Nanjing 211166 , Jiangsu , People's Republic of China
- The Key Laboratory of Modern Toxicology , Ministry of Education , School of Public Health , Nanjing Medical University , Nanjing 211166 , Jiangsu , People's Republic of China . ; ; Tel: +86-25-8686-8424
| | - Yi Liu
- Institute of Toxicology , School of Public Health , Nanjing Medical University , Nanjing 211166 , Jiangsu , People's Republic of China
- The Key Laboratory of Modern Toxicology , Ministry of Education , School of Public Health , Nanjing Medical University , Nanjing 211166 , Jiangsu , People's Republic of China . ; ; Tel: +86-25-8686-8424
| | - Xiaolin Lu
- Institute of Toxicology , School of Public Health , Nanjing Medical University , Nanjing 211166 , Jiangsu , People's Republic of China
- The Key Laboratory of Modern Toxicology , Ministry of Education , School of Public Health , Nanjing Medical University , Nanjing 211166 , Jiangsu , People's Republic of China . ; ; Tel: +86-25-8686-8424
| | - Bairu Wang
- Institute of Toxicology , School of Public Health , Nanjing Medical University , Nanjing 211166 , Jiangsu , People's Republic of China
- The Key Laboratory of Modern Toxicology , Ministry of Education , School of Public Health , Nanjing Medical University , Nanjing 211166 , Jiangsu , People's Republic of China . ; ; Tel: +86-25-8686-8424
| | - Qingling Wang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control , Ministry of Education , School of Public Health , Guiyang Medical University , Guiyang 550025 , Guizhou , People's Republic of China . ; ; Tel: +86-851-8841-6171
| | - Qizhan Liu
- Institute of Toxicology , School of Public Health , Nanjing Medical University , Nanjing 211166 , Jiangsu , People's Republic of China
- The Key Laboratory of Modern Toxicology , Ministry of Education , School of Public Health , Nanjing Medical University , Nanjing 211166 , Jiangsu , People's Republic of China . ; ; Tel: +86-25-8686-8424
| | - Aihua Zhang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control , Ministry of Education , School of Public Health , Guiyang Medical University , Guiyang 550025 , Guizhou , People's Republic of China . ; ; Tel: +86-851-8841-6171
| |
Collapse
|
45
|
Vijayakumaran R, Tan KH, Miranda PJ, Haupt S, Haupt Y. Regulation of Mutant p53 Protein Expression. Front Oncol 2015; 5:284. [PMID: 26734569 PMCID: PMC4681805 DOI: 10.3389/fonc.2015.00284] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 12/02/2015] [Indexed: 12/19/2022] Open
Abstract
For several decades, p53 has been detected in cancer biopsies by virtue of its high protein expression level which is considered indicative of mutation. Surprisingly, however, mouse genetic studies revealed that mutant p53 is inherently labile, similar to its wild type (wt) counterpart. Consistently, in response to stress conditions, both wt and mutant p53 accumulate in cells. While wt p53 returns to basal level following recovery from stress, mutant p53 remains stable. In part, this can be explained in mutant p53-expressing cells by the lack of an auto-regulatory loop with Mdm2 and other negative regulators, which are pivotal for wt p53 regulation. Further, additional protective mechanisms are acquired by mutant p53, largely mediated by the co-chaperones and their paralogs, the stress-induced heat shock proteins. Consequently, mutant p53 is accumulated in cancer cells in response to chronic stress and this accumulation is critical for its oncogenic gain of functions (GOF). Building on the extensive knowledge regarding wt p53, the regulation of mutant p53 is unraveling. In this review, we describe the current understanding on the major levels at which mutant p53 is regulated. These include the regulation of p53 protein levels by microRNA and by enzymes controlling p53 proteasomal degradation.
Collapse
Affiliation(s)
- Reshma Vijayakumaran
- Tumour Suppression Laboratory, Peter MacCallum Cancer Centre , Melbourne, VIC , Australia
| | - Kah Hin Tan
- Tumour Suppression Laboratory, Peter MacCallum Cancer Centre , Melbourne, VIC , Australia
| | | | - Sue Haupt
- Tumour Suppression Laboratory, Peter MacCallum Cancer Centre , Melbourne, VIC , Australia
| | - Ygal Haupt
- Tumour Suppression Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; Sir Peter MacCallum Department of Oncology and Department of Pathology, The University of Melbourne, Parkville, VIC, Australia; Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| |
Collapse
|
46
|
Gansmo LB, Romundstad P, Birkeland E, Hveem K, Vatten L, Knappskog S, Lønning PE. MDM4 SNP34091 (rs4245739) and its effect on breast-, colon-, lung-, and prostate cancer risk. Cancer Med 2015; 4:1901-7. [PMID: 26471763 PMCID: PMC5123711 DOI: 10.1002/cam4.555] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 07/31/2015] [Accepted: 09/07/2015] [Indexed: 12/21/2022] Open
Abstract
The MDM4 protein plays an important part in the negative regulation of the tumor suppressor p53 through its interaction with MDM2. In line with this, MDM4 amplification has been observed in several tumor forms. A polymorphism (rs4245739 A>C; SNP34091) in the MDM4 3′ untranslated region has been reported to create a target site for hsa‐miR‐191, resulting in decreased MDM4 mRNA levels. In this population‐based case–control study, we examined the potential association between MDM4 SNP34091, alone and in combination with the MDM2 SNP309T>G (rs2279744), and the risk of breast‐, colon‐, lung‐, and prostate cancer in Norway. SNP34091 was genotyped in 7,079 cancer patients as well as in 3,747 gender‐ and age‐matched healthy controls. MDM4 SNP34091C was not associated with risk for any of the tumor forms examined, except for a marginally significant association with reduced risk for breast cancer in a recessive model (OR = 0.77: 95% CI = 0.59–0.99). Stratifying according to MDM2 SNP309 status, we observed a reduced risk for breast cancer related to MDM4 SNP34091CC among individuals harboring the MDM2 SNP309GG genotype (OR = 0.41; 95% CI = 0.21–0.82). We conclude, MDM4 SNP34091 status to be associated with reduced risk of breast cancer, in particular in individuals carrying the MDM2 SNP309GG genotype, but not to be associated with either lung‐, colon‐ or prostate cancer.
Collapse
Affiliation(s)
- Liv B Gansmo
- Section of Oncology, Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Oncology, Haukeland University Hospital, Bergen, Norway
| | - Pål Romundstad
- Faculty of Medicine, Department of Public Health, Norwegian University of Science and Technology, Trondheim, Norway
| | - Einar Birkeland
- Section of Oncology, Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Oncology, Haukeland University Hospital, Bergen, Norway
| | - Kristian Hveem
- Faculty of Medicine, Department of Public Health, Norwegian University of Science and Technology, Trondheim, Norway
| | - Lars Vatten
- Faculty of Medicine, Department of Public Health, Norwegian University of Science and Technology, Trondheim, Norway
| | - Stian Knappskog
- Section of Oncology, Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Oncology, Haukeland University Hospital, Bergen, Norway
| | - Per Eystein Lønning
- Section of Oncology, Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Oncology, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
47
|
Chen QG, Zhou W, Han T, Du SQ, Li ZH, Zhang Z, Shan GY, Kong CZ. MiR-378 suppresses prostate cancer cell growth through downregulation of MAPK1 in vitro and in vivo. Tumour Biol 2015; 37:2095-103. [PMID: 26346167 DOI: 10.1007/s13277-015-3996-8] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 08/25/2015] [Indexed: 01/09/2023] Open
Abstract
Prostate cancer is one of the biggest health problems for the aging male. To the present, the roles of dysregulated microRNAs in prostate cancer are still unclear. Here, we evaluated the anti-proliferative role of miR-378 in prostate cancer. And, we found that the expression of miR-378 was significantly downregulated in clinical prostate cancer tissues. In vitro assay suggested that overexpression of miR-378-suppressed prostate cancer cell migration and invasion promoted cell apoptosis. Furthermore, we identified and validated MAPK1 as a direct target of miR-378. Ectopic expression of MAPK1 rescues miR-378-suppressed cell migration and invasion. In vivo assay demonstrated that the stably miR-378-overexpressed prostate cancer cells displayed a significantly reduction in tumor growth. Taken together, our data suggested that miR-378 may act as a potential therapeutic target against human prostate cancer.
Collapse
Affiliation(s)
- Qi-Guang Chen
- Department of Urology, The First Affiliated Hospital of China Medical University, Nanjing Street 155#, Shenyang, 110001, Liaoning, China
| | - Wei Zhou
- Department of Diagnostic Radiology, General Hospital of Shenyang Military Region, Shenyang, Liaoning, China
| | - Tao Han
- Department of Oncology, General Hospital of Shenyang Military Region, Shenyang, Liaoning, China
| | - Shu-Qi Du
- Department of Urology, The First Affiliated Hospital of China Medical University, Nanjing Street 155#, Shenyang, 110001, Liaoning, China
| | - Zhen-Hua Li
- Department of Urology, The First Affiliated Hospital of China Medical University, Nanjing Street 155#, Shenyang, 110001, Liaoning, China
| | - Zhe Zhang
- Department of Urology, The First Affiliated Hospital of China Medical University, Nanjing Street 155#, Shenyang, 110001, Liaoning, China
| | - Guang-Yi Shan
- Department of Urology, LiaoNing Cancer Hospital and Institute, Shenyang, Liaoning, China
| | - Chui-Ze Kong
- Department of Urology, The First Affiliated Hospital of China Medical University, Nanjing Street 155#, Shenyang, 110001, Liaoning, China.
| |
Collapse
|
48
|
Gao F, Xiong X, Pan W, Yang X, Zhou C, Yuan Q, Zhou L, Yang M. A Regulatory MDM4 Genetic Variant Locating in the Binding Sequence of Multiple MicroRNAs Contributes to Susceptibility of Small Cell Lung Cancer. PLoS One 2015; 10:e0135647. [PMID: 26274820 PMCID: PMC4537101 DOI: 10.1371/journal.pone.0135647] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 07/24/2015] [Indexed: 11/24/2022] Open
Abstract
A functional rs4245739 A>C single nucleotide polymorphism (SNP) locating in the MDM43’-untranslated (3’-UTR) region creates a miR-191-5p or miR-887-3p targeting sites. This change results in decreased expression of oncogene MDM4. Therefore, we examined the association between this SNP and small cell lung cancer (SCLC) risk as well as its regulatory function in SCLC cells. Genotypes were determined in two independent case-control sets consisted of 520SCLC cases and 1040 controls from two regions of China. Odds ratios (ORs) and 95% confidence intervals (CIs) were estimated by logistic regression. The impact of the rs4245739 SNP on miR-191-5p/miR-887-3p mediated MDM4 expression regulation was investigated using luciferase reporter gene assays. We found that the MDM4 rs4245739AC and CC genotypes were significantly associated with decreased SCLC susceptibility compared with the AA genotype in both case-control sets (Shandong set: OR = 0.53, 95% CI = 0.32–0.89, P = 0.014; Jiangsu set: OR = 0.47, 95% CI = 0.26–0.879, P = 0.017). Stratified analyses indicated that there was a significantly multiplicative interaction between rs4245739 and smoking (Pinteractioin = 0.048). After co-tranfection of miRNAs and different allelic-MDM4 reporter constructs into SCLC cells, we found that the both miR-191-5p and miR-887-3p can lead to significantly decreased MDM4 expression activities in the construct with C-allelic 3’-UTR but not A-allelic 3’-UTR, suggesting a consistent genotype-phenotype correlation. Our data illuminate that the MDM4rs4245739SNP contributes to SCLC risk and support the notion that gene 3’-UTR genetic variants, impacting miRNA-binding, might modify SCLC susceptibility.
Collapse
Affiliation(s)
- Feng Gao
- Health Division of Guard Bureau, General Staff Department of Chinese PLA, Beijing, China
| | - Xiangyu Xiong
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Wenting Pan
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Xinyu Yang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Changchun Zhou
- Clinical Laboratory, Shandong Cancer Hospital, Shandong Academy of Medical Sciences, Jinan, Shandong Province, China
| | - Qipeng Yuan
- Health Division of Guard Bureau, General Staff Department of Chinese PLA, Beijing, China
| | - Liqing Zhou
- Department of Radiation Oncology, Huaian No. 2 Hospital, Huaian, Jiangsu Province, China
- * E-mail: (MY); (LZ)
| | - Ming Yang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
- * E-mail: (MY); (LZ)
| |
Collapse
|