1
|
Suvac A, Ashton J, Bristow RG. Tumour hypoxia in driving genomic instability and tumour evolution. Nat Rev Cancer 2025; 25:167-188. [PMID: 39875616 DOI: 10.1038/s41568-024-00781-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/21/2024] [Indexed: 01/30/2025]
Abstract
Intratumour hypoxia is a feature of all heterogenous solid tumours. Increased levels or subregions of tumour hypoxia are associated with an adverse clinical prognosis, particularly when this co-occurs with genomic instability. Experimental evidence points to the acquisition of DNA and chromosomal alterations in proliferating hypoxic cells secondary to inhibition of DNA repair pathways such as homologous recombination, base excision repair and mismatch repair. Cell adaptation and selection in repair-deficient cells give rise to a model whereby novel single-nucleotide mutations, structural variants and copy number alterations coexist with altered mitotic control to drive chromosomal instability and aneuploidy. Whole-genome sequencing studies support the concept that hypoxia is a critical microenvironmental cofactor alongside the driver mutations in MYC, BCL2, TP53 and PTEN in determining clonal and subclonal evolution in multiple tumour types. We propose that the hypoxic tumour microenvironment selects for unstable tumour clones which survive, propagate and metastasize under reduced immune surveillance. These aggressive features of hypoxic tumour cells underpin resistance to local and systemic therapies and unfavourable outcomes for patients with cancer. Possible ways to counter the effects of hypoxia to block tumour evolution and improve treatment outcomes are described.
Collapse
Affiliation(s)
- Alexandru Suvac
- Translational Oncogenomics Laboratory, Cancer Research UK Manchester Institute, University of Manchester, Manchester, UK
- Manchester Cancer Research Centre, University of Manchester, Manchester, UK
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Jack Ashton
- Translational Oncogenomics Laboratory, Cancer Research UK Manchester Institute, University of Manchester, Manchester, UK
- Manchester Cancer Research Centre, University of Manchester, Manchester, UK
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Robert G Bristow
- Translational Oncogenomics Laboratory, Cancer Research UK Manchester Institute, University of Manchester, Manchester, UK.
- Manchester Cancer Research Centre, University of Manchester, Manchester, UK.
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
| |
Collapse
|
2
|
Harryman WL, Hinton JP, Sainz R, Gard JMC, Ryniawec JM, Rogers GC, Warfel NA, Knudsen BS, Nagle RB, Chipollini JJ, Lee BR, Sun BL, Cress AE. Intermediate risk prostate tumors contain lethal subtypes. FRONTIERS IN UROLOGY 2025; 4:1487873. [PMID: 40129601 PMCID: PMC11932713 DOI: 10.3389/fruro.2024.1487873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/26/2025]
Abstract
In 2024, prostate cancer (PCa) remains the most common non-skin cancer in males within the United States, with an estimated 299,010 new cases, the highest increase incident trend rate (3.8%) of all cancers, and one of the eight deadliest. PCa cases are projected to double from 1.8 million to 2.9 million per year between 2020 and 2040. According to the National Comprehensive Cancer Network (NCCN) treatment guidelines, most cases (65%) are intermediate risk (Gleason sum score <7 [3 + 4, 4 + 3], prostate organ-confined, and PSA < 20) with treatment options limited to active surveillance, external beam radiation, and/or surgery to prevent metastasis in the long term (>10 years). It is increasingly recognized that the two most common subtypes of intermediate risk PCa are cribriform architecture (CA) and intraductal carcinoma of the prostate (IDC-P), which can occur together, and both are associated with increased metastatic risk, biochemical recurrence, and disease-specific mortality. Both subtypes display hypoxia, genomic instability, and are identified as Gleason 4 in pathology reports. However, since false negatives are common (up to 50%) in these subtypes on biopsy, more research is needed to reliably detect these subtypes that have an increased risk for invasive disease. We note that even with mpMRI-guided biopsies, the sensitivity is 54% for cribriform architecture and only 37% for IDC-P. The presence of these PCa subtypes in biopsy or radical prostatectomy (RP) tissue can exclude patients from active surveillance and from designation as intermediate risk disease, further underscoring the need for increased molecular understanding of these subtypes for diagnostic purposes. Understanding the heterogeneity of intermediate risk primary PCa phenotypes, using computational pathology approaches to evaluate the fixed biopsy specimen, or video microscopy of the surgical specimen with AI-driven analysis is now achievable. New research associating the resulting phenotypes with the different therapeutic choices and vulnerabilities will likely prevent extracapsular extension, the definition of high-risk disease, and upstaging of the final pathologic stage.
Collapse
Affiliation(s)
| | - James P. Hinton
- University of Arizona Cancer Center, Tucson, AZ, United States
| | - Rafael Sainz
- University of Arizona Cancer Center, Tucson, AZ, United States
| | | | - John M. Ryniawec
- University of Arizona Cancer Center, Tucson, AZ, United States
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, United States
| | - Gregory C. Rogers
- University of Arizona Cancer Center, Tucson, AZ, United States
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, United States
| | - Noel A. Warfel
- University of Arizona Cancer Center, Tucson, AZ, United States
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, United States
| | - Beatrice S. Knudsen
- Professor of Pathology and Biomedical Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, United States
| | | | - Juan J. Chipollini
- Department of Urology, University of Arizona College of Medicine, Tucson, AZ, United States
| | - Benjamin R. Lee
- Department of Urology, University of Arizona College of Medicine, Tucson, AZ, United States
| | - Belinda L. Sun
- Department of Pathology, University of Arizona College of Medicine, Tucson, AZ, United States
| | - Anne E. Cress
- University of Arizona Cancer Center, Tucson, AZ, United States
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
3
|
Yan W, Quan C, Mourad WF, Yuan J, Shi Z, Yang J, Lu Q, Zhang J. Application of radiomics in lung immuno-oncology. PRECISION RADIATION ONCOLOGY 2023; 7:128-136. [PMID: 40337267 PMCID: PMC11935008 DOI: 10.1002/pro6.1191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 02/22/2023] [Accepted: 02/26/2023] [Indexed: 04/08/2023] Open
Abstract
Radiomics is a rapidly evolving field of research that extracts and analyzes quantitative features within medical images. Those features are termed as radiomic features that can characterize a tumor in a comprehensive and quantitative manner with regard to its internal structure and heterogeneity. Radiomic features can be used, alone or in combination with demographic, histological, genomic, or proteomic data, for predicting prognosis or treatment response. Immunotherapy, or immune-oncology, is the study of cancer treatment by taking advantage of the body's immune system to prevent, control, and eliminate cancer. In this review, we first provide a brief introduction to both radiomics and immune-oncology in lung cancer. Then, we discuss the need for developing immune-oncology biomarkers, and the advantages of radiomics in identifying biomarkers related to immunotherapy. We also discuss potential areas in and out of tumors, such as the intra-tumoral hypoxic region and tumor microenvironment, where radiomic markers might be extracted, as well as a potential application of radiomic biomarkers in clinical lung cancer management. Finally, we present radiation and immune modulation in non-small cell lung cancer, clinical trials and their design to incorporate radiomic biomarkers, and radiomics-guided precision radiation therapy.
Collapse
Affiliation(s)
- Weisi Yan
- Baptist Health SystemLexingtonKentuckyUSA
| | - Chen Quan
- City of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Waleed F. Mourad
- Department of Radiation MedicineUniversity of KentuckyLexingtonKentuckyUSA
| | - Jianda Yuan
- Translational Oncology at Merck & CoKenilworthNew JerseyUSA
| | | | - Jun Yang
- Foshan Chancheng HospitalFoshanGuangdongChina
| | - Qiuxia Lu
- Foshan Chancheng HospitalFoshanGuangdongChina
| | - Jie Zhang
- Department of RadiologyUniversity of KentuckyLexingtonKentuckyUSA
| |
Collapse
|
4
|
Li X, Cao X, Zhao H, Guo M, Fang X, Li K, Qin L, He Y, Liu X. Hypoxia Activates Notch4 via ERK/JNK/P38 MAPK Signaling Pathways to Promote Lung Adenocarcinoma Progression and Metastasis. Front Cell Dev Biol 2022; 9:780121. [PMID: 34988077 PMCID: PMC8721100 DOI: 10.3389/fcell.2021.780121] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 11/22/2021] [Indexed: 12/12/2022] Open
Abstract
Hypoxia contributes to the progression and metastasis of lung adenocarcinoma (LUAD). However, the specific underlying molecular mechanisms have not been fully elucidated. Here we report that Notch4 is upregulated in lung tissue from lung cancer patients. Functionally, Hypoxia activates the expressions of Delta-like 4 and Notch4, resulting in the excessive proliferation and migration of LUAD cells as well as apoptotic resistance. Notch4 silencing reduced ERK, JNK, and P38 activation. Meanwhile, Notch4 overexpression enhanced ERK, JNK, and P38 activation in LUAD cells. Furthermore, Notch4 exerted pro-proliferation, anti-apoptosis and pro-migration effects on LUAD cells that were partly reversed by the inhibitors of ERK, JNK, and p38. The binding interaction between Notch4 and ERK/JNK/P38 were confirmed by the co-immunoprecipitation assay. In vivo study revealed that Notch4 played a key role in the growth and metastasis of LUAD using two xenograft models. This study demonstrates that hypoxia activates Notch4-ERK/JNK/P38 MAPK signaling pathways to promote LUAD cell progression and metastasis.
Collapse
Affiliation(s)
- Xiaochen Li
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Respiratory Diseases, National Ministry of Health of the People's Republic of China and National Clinical Research Center for Respiratory Disease, Wuhan, China
| | - Xiaopei Cao
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hanqiu Zhao
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Respiratory Diseases, National Ministry of Health of the People's Republic of China and National Clinical Research Center for Respiratory Disease, Wuhan, China
| | - Mingzhou Guo
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Respiratory Diseases, National Ministry of Health of the People's Republic of China and National Clinical Research Center for Respiratory Disease, Wuhan, China
| | - Xiaoyu Fang
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Respiratory Diseases, National Ministry of Health of the People's Republic of China and National Clinical Research Center for Respiratory Disease, Wuhan, China
| | - Ke Li
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Respiratory Diseases, National Ministry of Health of the People's Republic of China and National Clinical Research Center for Respiratory Disease, Wuhan, China
| | - Lu Qin
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Respiratory Diseases, National Ministry of Health of the People's Republic of China and National Clinical Research Center for Respiratory Disease, Wuhan, China
| | - Yuanzhou He
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Respiratory Diseases, National Ministry of Health of the People's Republic of China and National Clinical Research Center for Respiratory Disease, Wuhan, China
| | - Xiansheng Liu
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Respiratory Diseases, National Ministry of Health of the People's Republic of China and National Clinical Research Center for Respiratory Disease, Wuhan, China
| |
Collapse
|
5
|
Armstrong CWD, Coulter JA, Ong CW, Maxwell PJ, Walker S, Butterworth KT, Lyubomska O, Berlingeri S, Gallagher R, O'Sullivan JM, Jain S, Mills IG, Prise KM, Bristow RG, LaBonte MJ, Waugh DJJ. Clinical and functional characterization of CXCR1/CXCR2 biology in the relapse and radiotherapy resistance of primary PTEN-deficient prostate carcinoma. NAR Cancer 2020; 2:zcaa012. [PMID: 32743555 PMCID: PMC7380483 DOI: 10.1093/narcan/zcaa012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 06/16/2020] [Accepted: 06/22/2020] [Indexed: 01/20/2023] Open
Abstract
Functional impairment of the tumour suppressor PTEN is common in primary prostate cancer and has been linked to relapse post-radiotherapy (post-RT). Pre-clinical modelling supports elevated CXC chemokine signalling as a critical mediator of PTEN-depleted disease progression and therapeutic resistance. We assessed the correlation of PTEN deficiency with CXC chemokine signalling and its association with clinical outcomes. Gene expression analysis characterized a PTEN LOW/CXCR1HIGH/CXCR2HIGH cluster of tumours that associates with earlier time to biochemical recurrence [hazard ratio (HR) 5.87 and 2.65, respectively] and development of systemic metastasis (HR 3.51). In vitro, CXCL signalling was further amplified following exposure of PTEN-deficient prostate cancer cell lines to ionizing radiation (IR). Inhibition of CXCR1/2 signalling in PTEN-depleted cell-based models increased IR sensitivity. In vivo, administration of a CXCR1/2-targeted pepducin (x1/2pal-i3), or CXCR2-specific antagonist (AZD5069), in combination with IR to PTEN-deficient xenografts attenuated tumour growth and progression compared to control or IR alone. Post-mortem analysis confirmed that x1/2pal-i3 administration attenuated IR-induced CXCL signalling and anti-apoptotic protein expression. Interventions targeting CXC chemokine signalling may provide an effective strategy to combine with RT in locally advanced prostate cancer patients with known presence of PTEN-deficient foci.
Collapse
Affiliation(s)
- Chris W D Armstrong
- Movember FASTMAN Centre of Excellence, Patrick G Johnston Centre for Cancer Research, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast BT9 7AE, UK
| | | | - Chee Wee Ong
- Laboratory of Cancer Epigenome, Division of Medical Science, National Cancer Centre, Singapore, 169610
| | - Pamela J Maxwell
- Movember FASTMAN Centre of Excellence, Patrick G Johnston Centre for Cancer Research, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast BT9 7AE, UK
| | - Steven Walker
- Movember FASTMAN Centre of Excellence, Patrick G Johnston Centre for Cancer Research, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast BT9 7AE, UK
| | - Karl T Butterworth
- Movember FASTMAN Centre of Excellence, Patrick G Johnston Centre for Cancer Research, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast BT9 7AE, UK
| | - Oksana Lyubomska
- Movember FASTMAN Centre of Excellence, Patrick G Johnston Centre for Cancer Research, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast BT9 7AE, UK
| | - Silvia Berlingeri
- Movember FASTMAN Centre of Excellence, Patrick G Johnston Centre for Cancer Research, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast BT9 7AE, UK
| | - Rebecca Gallagher
- Movember FASTMAN Centre of Excellence, Patrick G Johnston Centre for Cancer Research, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast BT9 7AE, UK
| | - Joe M O'Sullivan
- Movember FASTMAN Centre of Excellence, Patrick G Johnston Centre for Cancer Research, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast BT9 7AE, UK
| | - Suneil Jain
- Movember FASTMAN Centre of Excellence, Patrick G Johnston Centre for Cancer Research, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast BT9 7AE, UK
| | - Ian G Mills
- Movember FASTMAN Centre of Excellence, Patrick G Johnston Centre for Cancer Research, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast BT9 7AE, UK
| | - Kevin M Prise
- Movember FASTMAN Centre of Excellence, Patrick G Johnston Centre for Cancer Research, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast BT9 7AE, UK
| | - Robert G Bristow
- Movember FASTMAN Centre of Excellence, Manchester CRUK Institute, Manchester, SK10 4TG, UK
| | - Melissa J LaBonte
- Movember FASTMAN Centre of Excellence, Patrick G Johnston Centre for Cancer Research, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast BT9 7AE, UK
| | - David J J Waugh
- Movember FASTMAN Centre of Excellence, Patrick G Johnston Centre for Cancer Research, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast BT9 7AE, UK
| |
Collapse
|
6
|
Angel CZ, Lynch SM, Nesbitt H, McKenna MM, Walsh CP, McKenna DJ. miR-210 is induced by hypoxia and regulates neural cell adhesion molecule in prostate cells. J Cell Physiol 2020; 235:6194-6203. [PMID: 31975433 DOI: 10.1002/jcp.29548] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 01/13/2020] [Indexed: 12/19/2022]
Abstract
Hypoxia in prostate tumours has been associated with disease progression and metastasis. MicroRNAs are short noncoding RNA molecules that are important in several cell processes, but their role in hypoxic signalling is still poorly understood. miR-210 has been linked with hypoxic mechanisms, but this relationship has been poorly characterised in prostate cancer. In this report, the link between hypoxia and miR-210 in prostate cancer cells is investigated. Polymerase chain reaction analysis demonstrates that miR-210 is induced by hypoxia in prostate cancer cells using in vitro cell models and an in vivo prostate tumour xenograft model. Analysis of The Cancer Genome Atlas prostate biopsy datasets shows that miR-210 is significantly correlated with Gleason grade and other clinical markers of prostate cancer progression. Neural cell adhesion molecule (NCAM) is identified as a target of miR-210, providing a biological mechanism whereby hypoxia-induced miR-210 expression can contribute to prostate cancer. This study provides evidence that miR-210 is an important regulator of cell response to hypoxic stress and proposes that its regulation of NCAM may play an important role in the pathogenesis of prostate cancer.
Collapse
Affiliation(s)
- Charlotte Zoe Angel
- Genomic Medicine Research Group, Biomedical Sciences Research Institute, Ulster University, Coleraine, UK
| | - Seodhna M Lynch
- Cancer Biology & Therapeutics Laboratory, School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Dublin, Ireland
| | - Heather Nesbitt
- Genomic Medicine Research Group, Biomedical Sciences Research Institute, Ulster University, Coleraine, UK
| | - Michael M McKenna
- Department of Cellular Pathology, Altnagelvin Area Hospital, Western Health & Social Care Trust, Co. Derry, UK
| | - Colum P Walsh
- Genomic Medicine Research Group, Biomedical Sciences Research Institute, Ulster University, Coleraine, UK
| | - Declan J McKenna
- Genomic Medicine Research Group, Biomedical Sciences Research Institute, Ulster University, Coleraine, UK
| |
Collapse
|
7
|
Han J, Chen M, Fang Q, Zhang Y, Wang Y, Esma J, Qiao H. Prediction of the Prognosis Based on Chromosomal Instability-Related DNA Methylation Patterns of ELOVL2 and UBAC2 in PTCs. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 18:650-660. [PMID: 31698312 PMCID: PMC6906861 DOI: 10.1016/j.omtn.2019.09.027] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 08/12/2019] [Accepted: 09/17/2019] [Indexed: 12/11/2022]
Abstract
Papillary thyroid carcinoma (PTC) is the most common malignant tumor of endocrine systems. Chromosomal instability (CIN) is crucial to the clinical prognoses of tumor patients. DNA methylation plays an important role in the regulation of gene expression and CIN. Based on PTC samples from The Cancer Genome Atlas database, we used multiple regression analyses to identify methylation patterns of CpG sites with the strongest correlation with gene expression. A total of 4,997 genes were obtained through combining the CpG sites, which were represented as featured DNA methylation patterns. In order to identify CIN-related epigenetic markers of PTC survival, we developed a method to characterize CIN based on DNA methylation patterns of genes using the Student’s t statistics. We found that 1,239 genes were highly associated with CIN. With the use of the log-rank test, univariate Cox regression analyses, and the Kaplan-Meier method, DNA methylation patterns of UBAC2 and ELOVL2, highly correlated with CIN, provided potential prognostic values for PTC. The higher these two genes, risk scores were correlated with worse PTC patient prognoses. Moreover, the ELOVL2 risk score was significantly different in the four stages of PTC, suggesting that it was related to the progress of PTC. The DNA methylation pattern associated with CIN may therefore be a good predictor of PTC survival.
Collapse
Affiliation(s)
- Jun Han
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital, Harbin Medical University, The Fourth Affiliated Hospital, Harbin Medical University, Harbin 150001, China
| | - Meijun Chen
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital, Harbin Medical University, Harbin 150001, China
| | - Qingxiao Fang
- Surgical Oncology, The Second Affiliated Hospital, Harbin Medical University, Harbin 150001, China
| | - Yanqing Zhang
- Hematological Department, The Second Affiliated Hospital, Harbin Medical University, Harbin 150001, China
| | - Yihan Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150001, China
| | - Jamaspishvili Esma
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital, Harbin Medical University, Harbin 150001, China
| | - Hong Qiao
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital, Harbin Medical University, Harbin 150001, China.
| |
Collapse
|
8
|
Tang LR, Wu JX, Cai SL, Huang YX, Zhang XQ, Fu WK, Zhuang QY, Li JL. Prolyl hydroxylase domain 3 influences the radiotherapy efficacy of pancreatic cancer cells by targeting hypoxia-inducible factor-1α. Onco Targets Ther 2018; 11:8507-8515. [PMID: 30555241 PMCID: PMC6278705 DOI: 10.2147/ott.s187615] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
PURPOSE Pancreatic cancer is characterized by a hypoxic microenvironment and resistance to most currently available treatment modalities. Prolyl hydroxylase domain 3 (PHD3) is a rate-limiting enzyme that regulates the degradation of hypoxia-inducible factors (HIFs) and is deregulated in pancreatic cancer cells. Whether such alteration of PHD3 expression contributes to the sustained growth and radioresistance of pancreatic cancer cells remains largely unknown. MATERIALS AND METHODS PHD3 was overexpressed in pancreatic cancer Mia-paca2 cells via lentiviral expression. Cell cycle progression and apoptosis were assayed by flow cytometry. HIF-1α, EGFR, and PHD3 protein expression was assessed by Western blotting. Cell survival was determined in a colony formation assay. RESULTS PHD3 overexpression suppressed HIF-1α protein expression and EGFR phosphorylation and enhanced the 2 Gy irradiation-mediated reductions in HIF-1α and phosphorylated (p)-EGFR under either normoxic or hypoxic conditions. PHD3 overexpression inhibited the growth and colony formation of Mia-paca2 cells in response to irradiation under either normoxic or hypoxic conditions. PHD3 overexpression exacerbated irradiation-induced apoptosis, with a greater effect under hypoxia than normoxia. Cell cycle distribution analysis demonstrated that PHD3 overexpression resulted in further shortened S phase and lengthened G2/M phase in response to irradiation. CONCLUSION PHD3 expression may contribute to the radiotherapy efficacy of pancreatic cancer cells and serve as a novel biomarker for improving radiotherapy efficacy in pancreatic cancer.
Collapse
Affiliation(s)
- Li-Rui Tang
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China,
| | - Jun-Xin Wu
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China,
| | - Shao-Li Cai
- Key Laboratories of Innate Immune Biology of Fujian Province, Biomedical Research Center of South China, Fujian Normal University, Fuzhou, China
| | - Yun-Xia Huang
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China,
| | - Xue-Qing Zhang
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China,
| | - Wan-Kai Fu
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China,
| | - Qing-Yang Zhuang
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China,
| | - Jin-Luan Li
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China,
| |
Collapse
|
9
|
Lybaert L, Vermaelen K, De Geest BG, Nuhn L. Immunoengineering through cancer vaccines – A personalized and multi-step vaccine approach towards precise cancer immunity. J Control Release 2018; 289:125-145. [DOI: 10.1016/j.jconrel.2018.09.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 09/11/2018] [Accepted: 09/12/2018] [Indexed: 02/07/2023]
|
10
|
Evaluation of tumor hypoxia prior to radiotherapy in intermediate-risk prostate cancer using 18F-fluoromisonidazole PET/CT: a pilot study. Oncotarget 2018. [PMID: 29515786 PMCID: PMC5839367 DOI: 10.18632/oncotarget.24234] [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] [Indexed: 12/20/2022] Open
Abstract
Purpose Hypoxia is a major factor in prostate cancer aggressiveness and radioresistance. Predicting which patients might be bad candidates for radiotherapy may help better personalize treatment decisions in intermediate-risk prostate cancer patients. We assessed spatial distribution of 18F-Misonidazole (FMISO) PET/CT uptake in the prostate prior to radiotherapy treatment. Materials and Methods Intermediate-risk prostate cancer patients about to receive high-dose (>74 Gy) radiotherapy to the prostate without hormonal treatment were prospectively recruited between 9/2012 and 10/2014. Prior to radiotherapy, all patients underwent a FMISO PET/CT as well as a MRI and 18F-choline-PET. 18F-choline and FMISO-positive volumes were semi-automatically determined using the fuzzy locally adaptive Bayesian (FLAB) method. In FMISO-positive patients, a dynamic analysis of early tumor uptake was performed. Group differences were assessed using the Wilcoxon signed rank test. Parameters were correlated using Spearman rank correlation. Results Of 27 patients (median age 76) recruited to the study, 7 and 9 patients were considered positive at 2.5h and 3.5h FMISO PET/CT respectively. Median SUVmax and SUVmax tumor to muscle (T/M) ratio were respectively 3.4 and 3.6 at 2.5h, and 3.2 and 4.4 at 3.5h. The median FMISO-positive volume was 1.1 ml. Conclusions This is the first study regarding hypoxia imaging using FMISO in prostate cancer showing that a small FMISO-positive volume was detected in one third of intermediate-risk prostate cancer patients.
Collapse
|
11
|
Nesbitt H, Worthington J, Errington RJ, Patterson LH, Smith PJ, McKeown SR, McKenna DJ. The unidirectional hypoxia-activated prodrug OCT1002 inhibits growth and vascular development in castrate-resistant prostate tumors. Prostate 2017; 77:1539-1547. [PMID: 28944496 DOI: 10.1002/pros.23434] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 09/09/2017] [Indexed: 11/07/2022]
Abstract
BACKGROUND OCT1002 is a unidirectional hypoxia-activated prodrug (uHAP) OCT1002 that can target hypoxic tumor cells. Hypoxia is a common feature in prostate tumors and is known to drive disease progression and metastasis. It is, therefore, a rational therapeutic strategy to directly target hypoxic tumor cells in an attempt to improve treatment for this disease. Here we tested OCT1002 alone and in combination with standard-of-care agents in hypoxic models of castrate-resistant prostate cancer (CRPC). METHODS The effect of OCT1002 on tumor growth and vasculature was measured using murine PC3 xenograft and dorsal skin fold (DSF) window chamber models. The effects of abiraterone, docetaxel, and cabazitaxel, both singly and in combination with OCT1002, were also compared. RESULTS The hypoxia-targeting ability of OCT1002 effectively controls PC3 tumor growth. The effect was evident for at least 42 days after exposure to a single dose (30 mg/kg) and was comparable to, or better than, drugs currently used in the clinic. In DSF experiments OCT1002 caused vascular collapse in the PC3 tumors and inhibited the revascularization seen in controls. In this model OCT1002 also enhanced the anti-tumor effects of abiraterone, cabazitaxel, and docetaxel; an effect which was accompanied by a more prolonged reduction in tumor vasculature density. CONCLUSIONS These studies provide the first evidence that OCT1002 can be an effective agent in treating hypoxic, castrate-resistant prostate tumors, either singly or in combination with established chemotherapeutics for prostate cancer.
Collapse
Affiliation(s)
- Heather Nesbitt
- Biomedical Sciences, Research Institute, University of Ulster, Cromore Road, Coleraine, Northern Ireland, United Kingdom
| | | | - Rachel J Errington
- School of Medicine, Cardiff University, Heath Park, Cardiff, United Kingdom
- BioStatus Ltd, Shepshed, Leicestershire, United Kingdom
| | | | - Paul J Smith
- OncoTherics Ltd, Shepshed, Leicestershire, United Kingdom
| | - Stephanie R McKeown
- Biomedical Sciences, Research Institute, University of Ulster, Cromore Road, Coleraine, Northern Ireland, United Kingdom
- OncoTherics Ltd, Shepshed, Leicestershire, United Kingdom
| | - Declan J McKenna
- Biomedical Sciences, Research Institute, University of Ulster, Cromore Road, Coleraine, Northern Ireland, United Kingdom
| |
Collapse
|
12
|
Hou R, Xu Y, Lu Q, Zhang Y, Hu B. Effect of low-frequency low-intensity ultrasound with microbubbles on prostate cancer hypoxia. Tumour Biol 2017; 39:1010428317719275. [PMID: 28974155 DOI: 10.1177/1010428317719275] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Affiliation(s)
- Rui Hou
- Department of Ultrasound in Medicine, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai Institute of Ultrasound in Medicine, Shanghai, P.R. China
| | - Yanjun Xu
- Department of Ultrasound in Medicine, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai Institute of Ultrasound in Medicine, Shanghai, P.R. China
| | - Qijie Lu
- Department of Ultrasound in Medicine, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai Institute of Ultrasound in Medicine, Shanghai, P.R. China
| | - Yang Zhang
- Department of Ultrasound in Medicine, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai Institute of Ultrasound in Medicine, Shanghai, P.R. China
| | - Bing Hu
- Department of Ultrasound in Medicine, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai Institute of Ultrasound in Medicine, Shanghai, P.R. China
| |
Collapse
|
13
|
Malla B, Zaugg K, Vassella E, Aebersold DM, Dal Pra A. Exosomes and Exosomal MicroRNAs in Prostate Cancer Radiation Therapy. Int J Radiat Oncol Biol Phys 2017; 98:982-995. [PMID: 28721912 DOI: 10.1016/j.ijrobp.2017.03.031] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 03/14/2017] [Accepted: 03/20/2017] [Indexed: 12/11/2022]
Abstract
Despite current risk stratification systems using traditional clinicopathologic factors, many localized and locally advanced prostate cancers fail radical treatment (ie, radical prostatectomy, radiation therapy with or without androgen deprivation therapy). Therefore, a pressing need exists for enhanced methods of disease stratification through novel prognostic and predictive tools that can reliably be applied in clinical practice. Exosomes are 50- to 150-nm small vesicles released by cancer cells that reflect the genetic and nongenetic materials of parent cancer cells. Cancer cells can contain distinct sets of microRNA profiles, the expression of which can change owing to stress such as radiation therapy. These alterations or distinctions in contents allow exosomes to be used as prognostic and/or predictive biomarkers and to monitor the treatment response. Additionally, microRNAs have been shown to influence multiple processes in prostate tumorigenesis, including cell proliferation, induction of apoptosis, migration, oncogene inhibition, and radioresistance. Thus, comparative exosomal microRNA profiling at different levels could help portray tumor aggressiveness and response to radiation therapy. Although technical challenges persist in exosome isolation and characterization, recent improvements in microRNA profiling have evolved toward in-depth analyses of the exosomal cargo and its functions. We have reviewed the role of exosomes and exosomal microRNAs in biologic processes of prostate cancer progression and radiation therapy response, with a particular focus on the development of clinical assays for treatment personalization.
Collapse
Affiliation(s)
- Bijaya Malla
- Department of Radiation Oncology, Bern University Hospital, Inselspital, Bern, Switzerland
| | - Kathrin Zaugg
- Department of Radiation Oncology, Bern University Hospital, Inselspital, Bern, Switzerland
| | - Erik Vassella
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - Daniel M Aebersold
- Department of Radiation Oncology, Bern University Hospital, Inselspital, Bern, Switzerland
| | - Alan Dal Pra
- Department of Radiation Oncology, Bern University Hospital, Inselspital, Bern, Switzerland.
| |
Collapse
|
14
|
Krause M, Dubrovska A, Linge A, Baumann M. Cancer stem cells: Radioresistance, prediction of radiotherapy outcome and specific targets for combined treatments. Adv Drug Deliv Rev 2017; 109:63-73. [PMID: 26877102 DOI: 10.1016/j.addr.2016.02.002] [Citation(s) in RCA: 225] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 01/05/2016] [Accepted: 02/03/2016] [Indexed: 12/26/2022]
Abstract
Inactivation of cancer stem cells (CSCs) is of utmost importance for tumor cure after radiotherapy. An increasing body of evidence complies with a higher radioresistance of CSCs compared to the mass of tumor cells, supporting the use of CSC related biomarkers for prediction of radiotherapy outcome. Treatment individualization strategies for patient groups with vastly different risk of recurrence will most likely require application of more than one biomarker. Specifically, inclusion of established biomarkers like tumor size for primary radio(chemo)therapy or human papilloma virus (HPV) infection status in head and neck squamous cell carcinoma seems to be of very high relevance. The high heterogeneity of CSC subclones along with changes of the functional behavior of individual tumors under treatment underlines the importance of the selection of the optimal timepoint(s) of biomarker evaluation, but also provides a potential therapeutic target for combined treatment approaches with irradiation.
Collapse
Affiliation(s)
- Mechthild Krause
- German Cancer Consortium (DKTK) Dresden, Germany; Dept. of Radiation Oncology, Technische Universität Dresden, Germany; OncoRay, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; Helmholtz-Zentrum Dresden-Rossendorf, Germany; German Cancer Research Center (DKFZ) Heidelberg, Germany.
| | - Anna Dubrovska
- German Cancer Consortium (DKTK) Dresden, Germany; OncoRay, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; German Cancer Research Center (DKFZ) Heidelberg, Germany
| | - Annett Linge
- German Cancer Consortium (DKTK) Dresden, Germany; Dept. of Radiation Oncology, Technische Universität Dresden, Germany; OncoRay, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; German Cancer Research Center (DKFZ) Heidelberg, Germany
| | - Michael Baumann
- German Cancer Consortium (DKTK) Dresden, Germany; Dept. of Radiation Oncology, Technische Universität Dresden, Germany; OncoRay, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; Helmholtz-Zentrum Dresden-Rossendorf, Germany; German Cancer Research Center (DKFZ) Heidelberg, Germany
| |
Collapse
|
15
|
Nesbitt H, Byrne NM, Williams SN, Ming L, Worthington J, Errington RJ, Patterson LH, Smith PJ, McKeown SR, McKenna DJ. Targeting Hypoxic Prostate Tumors Using the Novel Hypoxia-Activated Prodrug OCT1002 Inhibits Expression of Genes Associated with Malignant Progression. Clin Cancer Res 2016; 23:1797-1808. [PMID: 27697998 DOI: 10.1158/1078-0432.ccr-16-1361] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 08/11/2016] [Accepted: 08/25/2016] [Indexed: 11/16/2022]
Abstract
Purpose: To understand the role of hypoxia in prostate tumor progression and to evaluate the ability of the novel unidirectional hypoxia-activated prodrug OCT1002 to enhance the antitumor effect of bicalutamide.Experimental Design: The effect of OCT1002 on prostate cancer cells (LNCaP, 22Rv1, and PC3) was measured in normoxia and hypoxia in vitroIn vivo, tumor growth and lung metastases were measured in mice treated with bicalutamide, OCT1002, or a combination. Dorsal skin fold chambers were used to image tumor vasculature in vivo Longitudinal gene expression changes in tumors were analyzed using PCR.Results: Reduction of OCT1002 to its active form (OCT1001) decreased prostate cancer cell viability. In LNCaP-luc spheroids, OCT1002 caused increased apoptosis and decreased clonogenicity. In vivo, treatment with OCT1002 alone, or with bicalutamide, showed significantly greater tumor growth control and reduced lung metastases compared with controls. Reestablishment of the tumor microvasculature following bicalutamide-induced vascular collapse is inhibited by OCT1002. Significantly, the upregulation of RUNX2 and its targets caused by bicalutamide alone was blocked by OCT1002.Conclusions: OCT1002 selectively targets hypoxic tumor cells and enhances the antitumor efficacy of bicalutamide. Furthermore, bicalutamide caused changes in gene expression, which indicated progression to a more malignant genotype; OCT1002 blocked these effects, emphasizing that more attention should be attached to understanding genetic changes that may occur during treatment. Early targeting of hypoxic cells with OCT1002 can provide a means of inhibiting prostate tumor growth and malignant progression. This is of importance for the design and refinement of existing androgen-deprivation regimens in the clinic. Clin Cancer Res; 23(7); 1797-808. ©2016 AACR.
Collapse
Affiliation(s)
- Heather Nesbitt
- Biomedical Sciences Research Institute, University of Ulster, Cromore Road, Coleraine, Northern Ireland, United Kingdom
| | - Niall M Byrne
- Biomedical Sciences Research Institute, University of Ulster, Cromore Road, Coleraine, Northern Ireland, United Kingdom.,Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | | | - Louise Ming
- Biomedical Sciences Research Institute, University of Ulster, Cromore Road, Coleraine, Northern Ireland, United Kingdom
| | - Jenny Worthington
- Biomedical Sciences Research Institute, University of Ulster, Cromore Road, Coleraine, Northern Ireland, United Kingdom.,Axis Bioservices Ltd, Coleraine, Northern Ireland, United Kingdom
| | - Rachel J Errington
- School of Medicine, Cardiff University, Cardiff, United Kingdom.,BioStatus Ltd, Shepshed, Leicestershire, United Kingdom
| | | | - Paul J Smith
- OncoTherics Ltd, Shepshed, Leicestershire, United Kingdom
| | - Stephanie R McKeown
- Biomedical Sciences Research Institute, University of Ulster, Cromore Road, Coleraine, Northern Ireland, United Kingdom.,OncoTherics Ltd, Shepshed, Leicestershire, United Kingdom
| | - Declan J McKenna
- Biomedical Sciences Research Institute, University of Ulster, Cromore Road, Coleraine, Northern Ireland, United Kingdom.
| |
Collapse
|
16
|
Abstract
Tumours contain multiple different cell populations, including cells derived from the bone marrow as well as cancer-associated fibroblasts and various stromal populations including the vasculature. The microenvironment of the tumour cells plays a significant role in the response of the tumour to radiation treatment. Low levels of oxygen (hypoxia) caused by the poorly organized vasculature in tumours have long been known to affect radiation response; however, other aspects of the microenvironment may also play important roles. This article reviews some of the old literature concerning tumour response to irradiation and relates this to current concepts about the role of the tumour microenvironment in tumour response to radiation treatment. Included in the discussion are the role of cancer stem cells, radiation damage to the vasculature and the potential for radiation to enhance immune activity against tumour cells. Radiation treatment can cause a significant influx of bone marrow-derived cell populations into both normal tissues and tumours. Potential roles of such cells may include enhancing vascular recovery as well as modulating immune reactivity.
Collapse
Affiliation(s)
- Richard P Hill
- 1 Ontario Cancer Institute, Princess Margaret Cancer Centre, Toronto, ON, Canada.,2 Departments of Medical Biophysics and Radiation Oncology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
17
|
Espinoza I, Sakiyama MJ, Ma T, Fair L, Zhou X, Hassan M, Zabaleta J, Gomez CR. Hypoxia on the Expression of Hepatoma Upregulated Protein in Prostate Cancer Cells. Front Oncol 2016; 6:144. [PMID: 27379206 PMCID: PMC4908134 DOI: 10.3389/fonc.2016.00144] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 05/26/2016] [Indexed: 01/26/2023] Open
Abstract
Hepatoma upregulated protein (HURP) is a multifunctional protein with clinical promise. This protein has been demonstrated to be a predictive marker for the outcome in high-risk prostate cancer (PCa) patients, besides being a resistance factor in PCa. Although changes in oxygen tension (pO2) are associated with PCa aggressiveness, the role of hypoxia in the regulation of tumor progression genes such as HURP has not yet been described. We hypothesized that pO2 alteration is involved in the regulation of HURP expression in PCa cells. In the present study, PCa cells were incubated at 2% O2 (hypoxia) and 20% O2 (normoxia) conditions. Hypoxia reduced cell growth rate of PCa cells, when compared to the growth rate of cells cultured under normoxia (p < 0.05). The decrease in cell viability was accompanied by fivefold (p < 0.05) elevated rate of vascular endothelial growth factor (VEGF) release. The expression of VEGF and the hypoxia-inducible metabolic enzyme carbonic anhydrase 9 were elevated maximally nearly 61-fold and 200-fold, respectively (p < 0.05). Noted in two cell lines (LNCaP and C4-2B) and independent of the oxygen levels, HURP expression assessed at both mRNA and protein levels was reduced. However, the decrease was more pronounced in cells cultured under hypoxia (p < 0.05). Interestingly, the analysis of patients’ specimens by Western blot revealed a marked increase of HURP protein (fivefold), when compared to control (cystoprostatectomy) tissue (p < 0.05). Immunohistochemistry analysis showed an increase in the immunostaining intensity of HURP and the hypoxia-sensitive molecules, hypoxia-inducible factor 1-alpha (HIF-1α), VEGF, and heat-shock protein 60 (HSP60) in association with tumor grade. The data also suggested a redistribution of subcellular localization for HURP and HIF-1α from the nucleus to the cytoplasmic compartment in relation to increasing tumor grade. Analysis of HURP Promoter for HIF-1-binding sites revealed presence of four putative HIF binding sites on the promoter of DLGAP5/HURP gene in the non-translated region upstream from the start codon, suggesting association between HIF-1α and the regulation of HURP protein. Taken together, our findings suggest a modulatory role of hypoxia on the expression of HURP. Additionally our results provide basis for utilization of tumor-associated molecules as predictors of aggressive PCa.
Collapse
Affiliation(s)
- Ingrid Espinoza
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS, USA; Department of Preventive Medicine, University of Mississippi Medical Center, Jackson, MS, USA; Department of Biochemistry, University of Mississippi Medical Center, Jackson, MS, USA
| | - Marcelo J Sakiyama
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS, USA; Department of Pathology, University of Mississippi Medical Center, Jackson, MS, USA; CAPES Foundation, Ministry of Education of Brazil, Brasília, Brazil
| | - Tangeng Ma
- Cancer Institute, University of Mississippi Medical Center , Jackson, MS , USA
| | - Logan Fair
- School of Medicine, University of Mississippi Medical Center , Jackson, MS , USA
| | - Xinchun Zhou
- Department of Pathology, University of Mississippi Medical Center , Jackson, MS , USA
| | - Mohamed Hassan
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS, USA; Department of Pathology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Jovanny Zabaleta
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA, USA; Department of Pediatrics, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Christian R Gomez
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS, USA; Department of Pathology, University of Mississippi Medical Center, Jackson, MS, USA; Department of Radiation Oncology, University of Mississippi Medical Center, Jackson, MS, USA
| |
Collapse
|
18
|
Byrne NM, Nesbitt H, Ming L, McKeown SR, Worthington J, McKenna DJ. Androgen deprivation in LNCaP prostate tumour xenografts induces vascular changes and hypoxic stress, resulting in promotion of epithelial-to-mesenchymal transition. Br J Cancer 2016; 114:659-68. [PMID: 26954717 PMCID: PMC4800298 DOI: 10.1038/bjc.2016.29] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 01/04/2016] [Accepted: 01/14/2016] [Indexed: 12/21/2022] Open
Abstract
Background: When single-agent androgen deprivation therapy (ADT) is administered for locally advanced prostate cancer, men usually relapse within 1–2 years with more malignant castrate-resistant disease. The reason for this is currently unknown. We now hypothesise that an initial treatment response that increases tumour hypoxia drives selection of more malignant tumours. Methods: The LNCaP prostate tumour xenografts were analysed for physiological (oxygen and vasculature) and genetic (PCR array) changes during longitudinal treatment with ADT (bicalutamide, 6 or 2 mg kg−1 daily for 28 days). Results: Bicalutamide caused an immediate (within 24 h) dose-dependent fall in oxygenation in LNCaP-luc prostate tumours with a nadir of ≤0.1% oxygen within 3–7 days; this was attributed to a significant loss of tumour microvessels (window chamber study). The hypoxic nadir persisted for 10–14 days. During the next 7 days, tumours regrew, oxygenation improved and the vasculature recovered; this was inhibited by the VEGF inhibitor B20.4.1.1. Gene expression over 28 days showed marked fluctuations consistent with the physiological changes. Accompanying the angiogenic burst (day 21) was a particularly striking increase in expression of genes associated with epithelial-to-mesenchymal transition (EMT). In particular, insulin-like growth factor 1 (IGF-1) showed increases in mRNA and protein expression. Conclusions: Hypoxic stress caused by ADT promotes EMT, providing a mechanism for the cause of malignant progression in prostate cancer.
Collapse
Affiliation(s)
- N M Byrne
- Biomedical Science Research Institute, University of Ulster, Cromore Road, Coleraine, BT52 1SA Northern Ireland, UK.,Bone Biology Division, Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW 2010, Australia
| | - H Nesbitt
- Biomedical Science Research Institute, University of Ulster, Cromore Road, Coleraine, BT52 1SA Northern Ireland, UK
| | - L Ming
- Biomedical Science Research Institute, University of Ulster, Cromore Road, Coleraine, BT52 1SA Northern Ireland, UK
| | - S R McKeown
- Biomedical Science Research Institute, University of Ulster, Cromore Road, Coleraine, BT52 1SA Northern Ireland, UK
| | - J Worthington
- Axis Bioservices Ltd, Research Laboratory, Castleroe Road, Coleraine BT51 3RP, Northern Ireland, UK
| | - D J McKenna
- Biomedical Science Research Institute, University of Ulster, Cromore Road, Coleraine, BT52 1SA Northern Ireland, UK
| |
Collapse
|
19
|
Dhani N, Fyles A, Hedley D, Milosevic M. The clinical significance of hypoxia in human cancers. Semin Nucl Med 2015; 45:110-21. [PMID: 25704384 DOI: 10.1053/j.semnuclmed.2014.11.002] [Citation(s) in RCA: 151] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Hypoxia is present to some extent in most solid malignant human cancers because of an imbalance between the limited oxygen delivery capacity of the abnormal vasculature and the high oxygen consumption of tumor cells. This drives a complex and dynamic compensatory response to enable continued cell survival, including genomic changes leading to selection of hypoxia-adapted cells with a propensity to invade locally, metastasize, and recur following surgery or radiotherapy. There is indisputable clinical evidence from numerous observational and therapeutic studies across a range of tumor types to implicate hypoxia as a key determinant of cancer behavior and treatment outcome. Despite this, hypoxia-targeted treatment has failed to influence clinical practice. This is explained, in part, by emerging findings to indicate that hypoxia is not equally important in all patients even when present to the same extent. The impact of hypoxia on patient outcome and the benefit of hypoxia-targeted treatments are greatest in situations where hypoxia is a primary biological driver of disease behavior-patients with tumors having a "hypoxic driver" phenotype. The challenge for the clinical and scientific communities moving forward is to develop robust precision cancer medicine strategies for identifying these patients in the setting of other etiologic, genomic, and host-tumor factors, considering not only the state of the tumor at diagnosis but also changing patient and tumor characteristics over time.
Collapse
Affiliation(s)
- Neesha Dhani
- Department of Medical Oncology, Princess Margaret Cancer Centre, Toronto, Canada; Department of Medicine, University of Toronto, Toronto, Canada
| | - Anthony Fyles
- Department of Medical Oncology, Princess Margaret Cancer Centre, Toronto, Canada; Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, Canada; Department of Radiation Oncology, University of Toronto, Toronto, Canada
| | - David Hedley
- Department of Medical Oncology, Princess Margaret Cancer Centre, Toronto, Canada; Department of Medicine, University of Toronto, Toronto, Canada
| | - Michael Milosevic
- Department of Medical Oncology, Princess Margaret Cancer Centre, Toronto, Canada; Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, Canada; Department of Radiation Oncology, University of Toronto, Toronto, Canada.
| |
Collapse
|
20
|
Ma T, Schreiber CA, Knutson GJ, Khattouti AE, Sakiyama MJ, Hassan M, Charlesworth MC, Madden BJ, Zhou X, Vuk-Pavlović S, Gomez CR. Effects of oxygen on the antigenic landscape of prostate cancer cells. BMC Res Notes 2015; 8:687. [PMID: 26581192 PMCID: PMC4652345 DOI: 10.1186/s13104-015-1633-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 10/26/2015] [Indexed: 02/07/2023] Open
Abstract
Background Use of allogeneic cancer cells-based immunotherapy for treatment of established prostate cancer (PCa) has only been marginally effective. One reason for failure could stem from the mismatch of antigenic signatures of vaccine cells and cancer in situ. Hence, it is possible that vaccine cells expressed antigens differently than tumor cells in situ. We hypothesized that cells grown in vitro at low oxygen tension (pO2) provide a better antigen match to tumors in situ and could reveal a more relevant antigenic landscape than cells grown in atmospheric pO2. Methods We tested this hypothesis by comparing PCa cells propagated at pO2 = 2 kPa and 20 kPa. To identify potential tumor-associated antigens (TAAs), we prepared PCa cell lysates, resolved them by two-dimensional electrophoresis and immunoblotting using spontaneous antibodies from plasma derived from PCa patients and control subjects. Antibody-labeled spots were analyzed by MALDI-TOF mass spectrometry and validated by ELISA. We selected hypoxia-regulated HSP70 and hnRNP L and hypoxia-independent HSP60 and determined the frequency of plasma samples reacting with these molecules. Results Frequency of HSP60-reactive plasma was low in healthy controls [1.3 % (1/76)], while it was elevated in PCa patients [13.0 % (7/54); p < 0.05]. These data suggest a humoral immune response to HSP60 in PCa. Levels of autoantibodies to HSP70 did not differ from healthy controls [3.7 % (2/54)] in PCa patients [5.3 % (2/38)]. Similarly, hnRNP L autoantibodies did no differ between healthy controls [6.1 % (3/49)] and PCa patients [5.3 % (2/38)]. Conclusions Overall our results suggest the value of hypoxia as a modifier of the cellular and antigenic landscape of PCa cells. By modifying the immune reactivity of PCa cells in culture, manipulation of pO2 can be proposed as a new avenue for improving diagnosis, prognosis and immunotherapy for PCa.
Collapse
Affiliation(s)
- Tangeng Ma
- Cancer Institute, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA.
| | - Claire A Schreiber
- Stem Cell Laboratory, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA.
| | - Gaylord J Knutson
- Stem Cell Laboratory, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA.
| | - Abdelouahid El Khattouti
- Cancer Institute, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA.
| | - Marcelo J Sakiyama
- Cancer Institute, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA. .,Department of Pathology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA.
| | - Mohamed Hassan
- Cancer Institute, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA. .,Department of Pathology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA.
| | | | - Benjamin J Madden
- Proteomics Research Center, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA.
| | - Xinchun Zhou
- Department of Pathology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA.
| | - Stanimir Vuk-Pavlović
- Stem Cell Laboratory, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA. .,Division of Hematology, Department of Internal Medicine, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA. .,Division of Preventive and Occupational Medicine, Department of Internal Medicine, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA.
| | - Christian R Gomez
- Cancer Institute, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA. .,Department of Pathology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA. .,Department of Radiation Oncology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA. .,Stem Cell Laboratory, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA. .,Division of Hematology, Department of Internal Medicine, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA. .,Division of Preventive and Occupational Medicine, Department of Internal Medicine, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA.
| |
Collapse
|
21
|
Cell Death Conversion under Hypoxic Condition in Tumor Development and Therapy. Int J Mol Sci 2015; 16:25536-51. [PMID: 26512660 PMCID: PMC4632814 DOI: 10.3390/ijms161025536] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 10/07/2015] [Accepted: 10/19/2015] [Indexed: 12/29/2022] Open
Abstract
Hypoxia, which is common during tumor progression, plays important roles in tumor biology. Failure in cell death in response to hypoxia contributes to progression and metastasis of tumors. On the one hand, the metabolic and oxidative stress following hypoxia could lead to cell death by triggering signal cascades, like LKB1/AMPK, PI3K/AKT/mTOR, and altering the levels of effective components, such as the Bcl-2 family, Atg and p62. On the other hand, hypoxia-induced autophagy can serve as a mechanism to turn over nutrients, so as to mitigate the adverse condition and then avoid cell death potentially. Due to the effective role of hypoxia, this review focuses on the crosstalk in cell death under hypoxia in tumor progression. Additionally, the illumination of cell death in hypoxia could shed light on the clinical applications of cell death targeted therapy.
Collapse
|
22
|
Tang J, Chen Y, Cui R, Li D, Xiao L, Lin P, Du Y, Sun H, Yu X, Zheng X. Upregulation of fractalkine contributes to the proliferative response of prostate cancer cells to hypoxia via promoting the G1/S phase transition. Mol Med Rep 2015; 12:7907-14. [PMID: 26496926 PMCID: PMC4758273 DOI: 10.3892/mmr.2015.4438] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 08/25/2015] [Indexed: 12/31/2022] Open
Abstract
Hypoxia is a common phenomenon in prostate cancer, which leads to cell proliferation and tumor growth. Fractalkine (FKN) is a membrane-bound chemokine, which is implicated in the progression of human prostate cancer and skeletal metastasis. However, the association between FKN and hypoxia-induced prostate cancer cell proliferation remains to be elucidated. The present study demonstrated that hypoxia induced the expression and secretion of FKN in the DU145 prostate cancer cell line. Furthermore, inhibiting the activity of FKN with the anti-FKN FKN-specific antibody markedly inhibited hypoxia-induced DU145 cell proliferation. Under normoxic conditions, DU145 cell proliferation markedly increased following exogenous administration of human recombinant FKN protein, and the increase was significantly alleviated by anti-FKN, indicating the importance of FKN in DU145 cell proliferation. In addition, subsequent determination of cell cycle distribution and expression levels of two core cell cycle regulators, cyclin E and cyclin-dependent kinase (CDK)2, suggested that FKN promoted the G1/S phase transition by upregulating the expression levels of cyclin E and CDK2. The results of the present study demonstrated that hypoxia led to the upregulation of the secretion and expression of FKN, which enhanced cell proliferation by promoting cell cycle progression in the prostate cancer cells. These findings provide evidence of a novel function for FKN, and suggest that FKN may serve as a potential target for treating androgen-independent prostate cancer.
Collapse
Affiliation(s)
- Jiebing Tang
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Yuanyuan Chen
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Rongjun Cui
- Department of Biochemistry and Molecular Biology, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Dong Li
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Lijie Xiao
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Ping Lin
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Yandan Du
- Department of Clinical Laboratory, The Second Clinical Medical School of Inner Mongolia University for the Nationalities, Inner Mongolia Forestry General Hospital, Hulunbuir, Inner Mongolia 022150, P.R. China
| | - Hui Sun
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Xiaoguang Yu
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Xiulan Zheng
- Department of Ultrasonography, The Affiliated Tumor Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| |
Collapse
|
23
|
|
24
|
Hodzic J, Dingjan I, Maas MJ, van der Meulen-Muileman IH, de Menezes RX, Heukelom S, Verheij M, Gerritsen WR, Geldof AA, van Triest B, van Beusechem VW. A cell-based high-throughput screening assay for radiation susceptibility using automated cell counting. Radiat Oncol 2015; 10:55. [PMID: 25888875 PMCID: PMC4355372 DOI: 10.1186/s13014-015-0355-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Accepted: 02/13/2015] [Indexed: 01/04/2023] Open
Abstract
Background Radiotherapy is one of the mainstays in the treatment for cancer, but its success can be limited due to inherent or acquired resistance. Mechanisms underlying radioresistance in various cancers are poorly understood and available radiosensitizers have shown only modest clinical benefit. There is thus a need to identify new targets and drugs for more effective sensitization of cancer cells to irradiation. Compound and RNA interference high-throughput screening technologies allow comprehensive enterprises to identify new agents and targets for radiosensitization. However, the gold standard assay to investigate radiosensitivity of cancer cells in vitro, the colony formation assay (CFA), is unsuitable for high-throughput screening. Methods We developed a new high-throughput screening method for determining radiation susceptibility. Fast and uniform irradiation of batches up to 30 microplates was achieved using a Perspex container and a clinically employed linear accelerator. The readout was done by automated counting of fluorescently stained nuclei using the Acumen eX3 laser scanning cytometer. Assay performance was compared to that of the CFA and the CellTiter-Blue homogeneous uniform-well cell viability assay. The assay was validated in a whole-genome siRNA library screening setting using PC-3 prostate cancer cells. Results On 4 different cancer cell lines, the automated cell counting assay produced radiation dose response curves that followed a linear-quadratic equation and that exhibited a better correlation to the results of the CFA than did the cell viability assay. Moreover, the cell counting assay could be used to detect radiosensitization by silencing DNA-PKcs or by adding caffeine. In a high-throughput screening setting, using 4 Gy irradiated and control PC-3 cells, the effects of DNA-PKcs siRNA and non-targeting control siRNA could be clearly discriminated. Conclusions We developed a simple assay for radiation susceptibility that can be used for high-throughput screening. This will aid the identification of molecular targets for radiosensitization, thereby contributing to improving the efficacy of radiotherapy.
Collapse
Affiliation(s)
- Jasmina Hodzic
- Department of Medical Oncology, VU University Medical Center, De Boelelaan 1118, 1081HV, Amsterdam, The Netherlands.
| | - Ilse Dingjan
- Department of Medical Oncology, VU University Medical Center, De Boelelaan 1118, 1081HV, Amsterdam, The Netherlands.
| | - Mariëlle Jp Maas
- Department of Medical Oncology, VU University Medical Center, De Boelelaan 1118, 1081HV, Amsterdam, The Netherlands.
| | - Ida H van der Meulen-Muileman
- Department of Medical Oncology, VU University Medical Center, De Boelelaan 1118, 1081HV, Amsterdam, The Netherlands.
| | - Renee X de Menezes
- Department of Epidemiology and Biostatistics, VU University Medical Center, De Boelelaan 1118, 1081HV, Amsterdam, The Netherlands.
| | - Stan Heukelom
- Department of Radiology & Nuclear Medicine, VU University Medical Center, De Boelelaan 1118, 1081HV, Amsterdam, The Netherlands.
| | - Marcel Verheij
- Department of Radiotherapy, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands.
| | - Winald R Gerritsen
- Department of Medical Oncology, VU University Medical Center, De Boelelaan 1118, 1081HV, Amsterdam, The Netherlands. .,Present address: Department of Medical Oncology, The Radboud University Medical Center, Comeniuslaan 4, 6525 HP, Nijmegen, The Netherlands.
| | - Albert A Geldof
- Department of Radiology & Nuclear Medicine, VU University Medical Center, De Boelelaan 1118, 1081HV, Amsterdam, The Netherlands. .,Department of Urology, VU University Medical Center, De Boelelaan 1118, 1081HV, Amsterdam, The Netherlands.
| | - Baukelien van Triest
- Department of Radiotherapy, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands.
| | - Victor W van Beusechem
- Department of Medical Oncology, VU University Medical Center, De Boelelaan 1118, 1081HV, Amsterdam, The Netherlands.
| |
Collapse
|
25
|
Li J, Zhang G, Wang X, Li XF. Is carbonic anhydrase IX a validated target for molecular imaging of cancer and hypoxia? Future Oncol 2015; 11:1531-41. [PMID: 25963430 PMCID: PMC4976829 DOI: 10.2217/fon.15.11] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The presence of hypoxia is a general feature of most solid malignancies, and hypoxia is considered as one of major factors for anticancer therapy failure. Carbonic anhydrase IX (CAIX) has been reported to be an endogenous hypoxia marker, CAIX monoclonal antibodies, their segments and inhibitors are developed for CAIX imaging. However, growing evidence indicates that CAIX expression under hypoxia condition may be cancer cell lines or cancer-type dependent. Here we review the current literature on CAIX and discuss the advantage and limitation of CAIX as a target for tumor hypoxia imaging. Accordingly, CAIX would be unreliable as a universal target for cancer and tumor hypoxia visualization.
Collapse
Affiliation(s)
- Jianbo Li
- Department of Nuclear Medicine, Inner Mongolia Medical University Affiliated Hospital, Hohhot, Inner Mongolia, China
- Department of Diagnostic Radiology, University of Louisville, 530 S Jackson Street, CCB-C07, Louisville, KY 40202, USA
| | - Guojian Zhang
- Department of Nuclear Medicine, Inner Mongolia Medical University Affiliated Hospital, Hohhot, Inner Mongolia, China
- Department of Diagnostic Radiology, University of Louisville, 530 S Jackson Street, CCB-C07, Louisville, KY 40202, USA
| | - Xuemei Wang
- Department of Nuclear Medicine, Inner Mongolia Medical University Affiliated Hospital, Hohhot, Inner Mongolia, China
| | - Xiao-Feng Li
- Department of Diagnostic Radiology, University of Louisville, 530 S Jackson Street, CCB-C07, Louisville, KY 40202, USA
| |
Collapse
|
26
|
Ma Y, Yang HZ, Dong BJ, Zou HB, Zhou Y, Kong XM, Huang YR. Biphasic regulation of autophagy by miR-96 in prostate cancer cells under hypoxia. Oncotarget 2014; 5:9169-82. [PMID: 25333253 PMCID: PMC4253426 DOI: 10.18632/oncotarget.2396] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Accepted: 08/26/2014] [Indexed: 12/13/2022] Open
Abstract
Autophagy favors cell survival under hypoxia, and increasing evidence revealed that microRNAs regulate autophagy. We report here hypoxia increased the expression of miR-96 in prostate cancer cells, and miR-96 stimulated autophagy by suppressing MTOR. We found that inhibition of miR-96 abolished hypoxia-induced autophagy. Paradoxically, ectopic over-expression of miR-96 to a certain threshold, also abolished the hypoxia-induced autophagy. Further studies have shown that high levels of miR-96 inhibited autophagy through suppressing ATG7, a key autophagy-associated gene. Importantly, the miR-96 expression level threshold was determined, and the effects of miR-96 on autophagy on either side of the threshold were opposite. These data demonstrate hypoxia-induced autophagy is at least partially regulated by miR-96; miR-96 can promote or inhibit autophagy by principally inhibiting MTOR or ATG7 depending on the expression levels of miR-96. Our observation might reveal a novel regulatory mode of autophagy by microRNAs under hypoxia.
Collapse
Affiliation(s)
- Yi Ma
- Department of urology, Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, China
- Department of biobank, Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, China
| | - Hao-Zheng Yang
- Department of central laboratory, Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, China
| | - Bai-Jun Dong
- Department of urology, Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, China
| | - Han-Bing Zou
- Department of central laboratory, Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, China
| | - Yan Zhou
- Department of central laboratory, Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, China
| | - Xian-Ming Kong
- Department of biobank, Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, China
| | - Yi-Ran Huang
- Department of urology, Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, China
| |
Collapse
|