1
|
Qu R, Zhang Y, Qin S, Xiong J, Fu X, Li L, Tu D, Cai Y. Differences in tumor angiogenesis and related factors between lung adenocarcinomas manifesting as pure ground glass opacity and solid nodules. Discov Oncol 2025; 16:180. [PMID: 39948247 PMCID: PMC11825439 DOI: 10.1007/s12672-025-01898-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 02/03/2025] [Indexed: 02/16/2025] Open
Abstract
INTRODUCTION The prognosis of ground glass opacity featured lung adenocarcinomas (GGO-LUAD) is significantly better than that of solid nodule featured lung adenocarcinomas (SN-LUAD), but the specific reasons behind their indolent tumor behavior are still unclear. The purpose of this study is to investigate their differences in intratumoral microvessels, related angiogenic factors and important stromal cells. METHODS Thirty patients (15 paired patients only with GGO or SN) diagnosed with pathological stage 0-I lung adenocarcinoma who underwent surgical treatment were included into this study. Immunohistochemistry was performed to stain the blood vessel markers (CD31, CD34 and CD105), LYVE-1, the cancer-associated fibroblasts (CAFs) markers (α-SMA and S100A4), TGF-β and HIF-1α from 30 patients tissue sections. At the same time, Ki67 Labeling Index (LI) extracted from pathological report of all patients was also analyzed. RESULTS GGO-LUAD is more abundant than SN-LUAD in lymphatic vessel density (LVD), but similar in total microvessel density (CD31 + MVD). However, GGO-LUAD is significantly lower than SN-LUAD in CD34 + MVD and CD105 + MVD. In terms of TGF-β, HIF-1α expression and Ki67 LI level, GGO-LUAD was also significantly weaker than SN-LUAD. Moreover, the distribution of CAFs in GGO-LUAD is less than that in SN-LUAD. Regardless of the pathological type (adenocarcinoma in situ (AIS) or minimally invasive adenocarcinoma (MIA) or invasive lung adenocarcinoma (IAC)), there is no difference in any of the above indicators in GGO-LUAD. CONCLUSIONS Our finding displays that GGO-LUAD was significantly lower than SN-LUAD in CD34 + MVD and CD105 + MVD reflecting tumor angiogenesis, and the distribution of CAFs and factors related to tumor angiogenesis were also significantly lower in GGO-LUAD, which may indicate that the weak ability of angiogenesis might be the reason for the good prognosis of GGO-LUAD.
Collapse
Affiliation(s)
- Rirong Qu
- Thoracic Surgery Laboratory, Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Avenue, Wuhan, 430030, Hubei, China
| | - Yang Zhang
- Department of Pelvic Floor Rehabilitation, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shenghui Qin
- Department of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jing Xiong
- Department of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiangning Fu
- Thoracic Surgery Laboratory, Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Avenue, Wuhan, 430030, Hubei, China
| | - Lequn Li
- Thoracic Surgery Laboratory, Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Avenue, Wuhan, 430030, Hubei, China
| | - Dehao Tu
- Department of Thoracic Surgery, Yueyang Central Hospital, Yueyang, 41400, Hunan, China.
| | - Yixin Cai
- Thoracic Surgery Laboratory, Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Avenue, Wuhan, 430030, Hubei, China.
| |
Collapse
|
2
|
Shamis SAK, Quinn J, Al‐Badran S, McKenzie M, Hatthakarnkul P, Lynch G, Lian G, Numprasit W, Romics L, Andersen D, Mallon E, McMillan DC, Edwards J. Elucidation of Dysregulated Pathways Associated With Hypoxia in Oestrogen Receptor-Negative Breast Cancer. Cancer Med 2024; 13:e70274. [PMID: 39660488 PMCID: PMC11632397 DOI: 10.1002/cam4.70274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 07/06/2024] [Accepted: 09/20/2024] [Indexed: 12/12/2024] Open
Abstract
PURPOSE Carbonic anhydrase IX (CAIX) is a well-established prognostic marker in breast cancer (BC). Nevertheless, this prognostic value is yet to be confirmed in BC subtypes. This study aims to investigate the prognostic effects of CAIX in oestrogen receptor (ER)-negative (ER-) BCs and to establish pathways related to cytoplasmic CAIX expression in ER- and lymph node-negative BCs. METHODS Immunohistochemistry was performed to identify the prognostic role of CAIX protein expression in ER- tissue microarrays (TMAs) (n = 191). CAIX-positive samples (n = 37) were transcriptionally profiled by TempO-Seq and analysed by STRING. Real-time quantitative PCR (RT-qPCR) analysis was used to validate differentially expressed genes. RESULTS Overexpression of cytoplasmic CAIX was an independent predictor of recurrence free survival, disease-free survival and overall survival in ER- cohort. RNA transcriptomic analysis identified 10 significant genes in ER- cohort and 3 genes in the node-negative group. The STRING database demonstrated a significant interaction between MUCL1 and GALNT6, which were linked with extracellular matrix organisation, degradation of the extracellular matrix and disease of glycosylation pathways. In the node-negative group, SPNS2 is mainly involved in the sphingolipid de novo biosynthesis pathway. A significant correlation between cytoplasmic SphK1 and cytoplasmic hypoxia-inducible factor-1α was observed. Among the 10 genes, 7 genes (SERHL2, GALNT6, MUCL1, MMP7, PITX2, CEACAM6 and SPNS2) were selected, and their expression was quantitatively assessed by RT-qPCR. The PCR data of these genes showed that SERHL2, GALNT6, MUCL1, PITX2, and SPNS2 mRNA levels were expressed in MDA-MB-231 BC cell lines at variable levels of hypoxic exposure. CONCLUSION Cytoplasmic CAIX was independently associated with poor prognosis in ER- BC. Gene expression profiles shed light on the pathways and genes associated with hypoxia in ER- BC. In node-negative patients, SPNS2 was of particular interest.
Collapse
Affiliation(s)
- Suad A. K. Shamis
- Academic Unit of Surgery, School of MedicineUniversity of Glasgow, Royal InfirmaryGlasgowUK
- Wolfson Wohl Cancer Research Centre, School of Cancer SciencesUniversity of GlasgowGlasgowUK
| | - Jean Quinn
- Wolfson Wohl Cancer Research Centre, School of Cancer SciencesUniversity of GlasgowGlasgowUK
| | - Sara Al‐Badran
- Wolfson Wohl Cancer Research Centre, School of Cancer SciencesUniversity of GlasgowGlasgowUK
| | - Molly McKenzie
- Wolfson Wohl Cancer Research Centre, School of Cancer SciencesUniversity of GlasgowGlasgowUK
| | - Phimmada Hatthakarnkul
- Wolfson Wohl Cancer Research Centre, School of Cancer SciencesUniversity of GlasgowGlasgowUK
| | - Gerard Lynch
- Wolfson Wohl Cancer Research Centre, School of Cancer SciencesUniversity of GlasgowGlasgowUK
| | - Guang‐Yu Lian
- Wolfson Wohl Cancer Research Centre, School of Cancer SciencesUniversity of GlasgowGlasgowUK
| | - Warapan Numprasit
- Wolfson Wohl Cancer Research Centre, School of Cancer SciencesUniversity of GlasgowGlasgowUK
| | - Laszlo Romics
- NHS Greater Glasgow and Clyde (New Victoria Hospital)GlasgowUK
| | - Ditte Andersen
- BioClavis Ltd., Teaching & Learning CentreQueen Elizabeth University HospitalGlasgowUK
| | - Elizabeth Mallon
- Department of PathologyQueen Elizabeth University HospitalGlasgowUK
| | - Donald C. McMillan
- Academic Unit of Surgery, School of MedicineUniversity of Glasgow, Royal InfirmaryGlasgowUK
| | - Joanne Edwards
- Wolfson Wohl Cancer Research Centre, School of Cancer SciencesUniversity of GlasgowGlasgowUK
| |
Collapse
|
3
|
Lin X, Sun D, Yang S, Cheng K, Wang X, Meng W, Wu H, Liu W, Wu X, Yang H, Wang X, Zhou L. UBE2M forms a positive feedback loop with estrogen receptor to drive breast cancer progression and drug resistance. Cell Death Dis 2024; 15:590. [PMID: 39138151 PMCID: PMC11322533 DOI: 10.1038/s41419-024-06979-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 08/04/2024] [Accepted: 08/06/2024] [Indexed: 08/15/2024]
Abstract
UBE2M, a NEDD8-conjugating enzyme, is dysregulated in various human cancers and promotes tumor cell proliferation. However, its role in estrogen receptor-positive (ER+) breast cancer remains unknown. We found that UBE2M expression was significantly higher in ER+ breast cancer tissues than in ER-negative (ER-) breast cancer tissues. Higher expression of UBE2M indicated a poorer prognosis in patients with ER+ breast cancer but not in those with ER- breast cancer. Of interest, a positive feedback loop was observed between UBE2M and ERα. Specifically, ERα enhanced the HIF-1α-mediated transcription of UBE2M. In turn, UBE2M maintained ERα expression by inhibiting its ubiquitination and degradation through UBE2M-CUL3/4A-E6AP-ERα axis. Functionally, silencing of UBE2M suppressed the growth of breast cancer cells by inducing cell cycle arrest and apoptosis and improved their sensitivity to fulvestrant both in vitro and in vivo. Altogether, our findings reveal that the UBE2M-ERα feedback loop drives breast cancer progression and fulvestrant resistance, suggesting UBE2M as a viable target for endocrine therapy of ER+ breast cancer.
Collapse
Affiliation(s)
- Xiongzhi Lin
- Taizhou Central Hospital (Taizhou University Hospital), School of Medicine, Taizhou University, Taizhou, Zhejiang, China
- Graduate School of Medicine, Hebei North University, Zhangjiakou, Hebei, China
| | - Dongsheng Sun
- Taizhou Central Hospital (Taizhou University Hospital), School of Medicine, Taizhou University, Taizhou, Zhejiang, China
| | - Shuhan Yang
- Taizhou Central Hospital (Taizhou University Hospital), School of Medicine, Taizhou University, Taizhou, Zhejiang, China
| | - Kai Cheng
- Taizhou Central Hospital (Taizhou University Hospital), School of Medicine, Taizhou University, Taizhou, Zhejiang, China
| | - XingYi Wang
- Taizhou Central Hospital (Taizhou University Hospital), School of Medicine, Taizhou University, Taizhou, Zhejiang, China
| | - Weijia Meng
- Taizhou Central Hospital (Taizhou University Hospital), School of Medicine, Taizhou University, Taizhou, Zhejiang, China
| | - Haowei Wu
- Taizhou Central Hospital (Taizhou University Hospital), School of Medicine, Taizhou University, Taizhou, Zhejiang, China
| | - Wenlin Liu
- Taizhou Central Hospital (Taizhou University Hospital), School of Medicine, Taizhou University, Taizhou, Zhejiang, China
| | - Xiaoyu Wu
- Taizhou Central Hospital (Taizhou University Hospital), School of Medicine, Taizhou University, Taizhou, Zhejiang, China
| | - Hui Yang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaojun Wang
- Taizhou Central Hospital (Taizhou University Hospital), School of Medicine, Taizhou University, Taizhou, Zhejiang, China.
| | - Lisha Zhou
- Taizhou Central Hospital (Taizhou University Hospital), School of Medicine, Taizhou University, Taizhou, Zhejiang, China.
| |
Collapse
|
4
|
Carmona-Bozo JC, Manavaki R, Miller JL, Brodie C, Caracò C, Woitek R, Baxter GC, Graves MJ, Fryer TD, Provenzano E, Gilbert FJ. PET/MRI of hypoxia and vascular function in ER-positive breast cancer: correlations with immunohistochemistry. Eur Radiol 2023; 33:6168-6178. [PMID: 37166494 PMCID: PMC10415421 DOI: 10.1007/s00330-023-09572-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 12/16/2022] [Accepted: 02/08/2023] [Indexed: 05/12/2023]
Abstract
OBJECTIVES To explore the relationship between indices of hypoxia and vascular function from 18F-fluoromisonidazole ([18F]-FMISO)-PET/MRI with immunohistochemical markers of hypoxia and vascularity in oestrogen receptor-positive (ER +) breast cancer. METHODS Women aged > 18 years with biopsy-confirmed, treatment-naïve primary ER + breast cancer underwent [18F]-FMISO-PET/MRI prior to surgery. Parameters of vascular function were derived from DCE-MRI using the extended Tofts model, whilst hypoxia was assessed using the [18F]-FMISO influx rate constant, Ki. Histological tumour sections were stained with CD31, hypoxia-inducible factor (HIF)-1α, and carbonic anhydrase IX (CAIX). The number of tumour microvessels, median vessel diameter, and microvessel density (MVD) were obtained from CD31 immunohistochemistry. HIF-1α and CAIX expression were assessed using histoscores obtained by multiplying the percentage of positive cells stained by the staining intensity. Regression analysis was used to study associations between imaging and immunohistochemistry variables. RESULTS Of the lesions examined, 14/22 (64%) were ductal cancers, grade 2 or 3 (19/22; 86%), with 17/22 (77%) HER2-negative. [18F]-FMISO Ki associated negatively with vessel diameter (p = 0.03), MVD (p = 0.02), and CAIX expression (p = 0.002), whilst no significant relationships were found between DCE-MRI pharmacokinetic parameters and immunohistochemical variables. HIF-1α did not significantly associate with any PET/MR imaging indices. CONCLUSION Hypoxia measured by [18F]-FMISO-PET was associated with increased CAIX expression, low MVD, and smaller vessel diameters in ER + breast cancer, further corroborating the link between inadequate vascularity and hypoxia in ER + breast cancer. KEY POINTS • Hypoxia, measured by [18F]-FMISO-PET, was associated with low microvessel density and small vessel diameters, corroborating the link between inadequate vascularity and hypoxia in ER + breast cancer. • Increased CAIX expression was associated with higher levels of hypoxia measured by [18F]-FMISO-PET. • Morphologic and functional abnormalities of the tumour microvasculature are the major determinants of hypoxia in cancers and support the previously reported perfusion-driven character of hypoxia in breast carcinomas.
Collapse
Affiliation(s)
- Julia C Carmona-Bozo
- Department of Radiology, School of Clinical Medicine, University of Cambridge, Box 218 - Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK
| | - Roido Manavaki
- Department of Radiology, School of Clinical Medicine, University of Cambridge, Box 218 - Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK
| | - Jodi L Miller
- Cancer Research UK - Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
| | - Cara Brodie
- Cancer Research UK - Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
| | - Corradina Caracò
- Department of Radiology, School of Clinical Medicine, University of Cambridge, Box 218 - Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK
| | - Ramona Woitek
- Department of Radiology, School of Clinical Medicine, University of Cambridge, Box 218 - Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK
| | - Gabrielle C Baxter
- Department of Radiology, School of Clinical Medicine, University of Cambridge, Box 218 - Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK
| | - Martin J Graves
- Department of Radiology, School of Clinical Medicine, University of Cambridge, Box 218 - Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK
| | - Tim D Fryer
- Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, School of Clinical Medicine, University of Cambridge, Box 65 - Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK
| | - Elena Provenzano
- Cancer Research UK - Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
- Cambridge Breast Unit, Cambridge University Hospitals NHS Foundation Trust, Box 97 - Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK
| | - Fiona J Gilbert
- Department of Radiology, School of Clinical Medicine, University of Cambridge, Box 218 - Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK.
| |
Collapse
|
5
|
Wang T, Guo W, Zhang X, Ma J, Li F, Zheng S, Zhu M, Dong Y, Bai M. Correlation between conventional ultrasound features combined with contrast-enhanced ultrasound patterns and pathological prognostic factors in malignant non-mass breast lesions. Clin Hemorheol Microcirc 2023; 85:433-445. [PMID: 37781796 DOI: 10.3233/ch-231936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
OBJECTIVE To investigate the correlation between ultrasound performance and prognostic factors in malignant non-mass breast lesions (NMLs). MATERIALS AND METHODS This study included 106 malignant NMLs in 104 patients. Different US features and contrast enhancement patterns were evaluated. Prognostic factors, including histological types and grades, axillary lymph node and peritumoral lymphovascular status, estrogen and progesterone receptor status and the expression of HER-2 and Ki-67 were determined. A chi-square test and logistic regression analysis were used to analyse possible associations. RESULTS Lesion size (OR: 3.08, p = 0.033) and posterior echo attenuation (OR: 8.38, p < 0.001) were useful in reflecting malignant NMLs containing an invasive carcinoma component. Posterior echo attenuation (OR: 7.51, p = 0.003) and unclear enhancement margin (OR: 6.50, p = 0.018) were often found in tumors with axillary lymph node metastases. Peritumoural lymphovascular invasion mostly exhibited posterior echo attenuation (OR: 3.84, p = 0.049) and unclear enhancement margin (OR: 8.68, p = 0.042) on ultrasound images. Perfusion defect was a comparatively accurate enhancement indicator for negative ER (OR: 2.57, p = 0.041) and PR (OR: 3.04, p = 0.008) expression. Calcifications (OR: 3.03, p = 0.025) and enlarged enhancement area (OR: 5.36, p = 0.033) imply an increased risk of positive HER-2 expression. Similarly, Calcifications (OR: 4.13, p = 0.003) and enlarged enhancement area (OR: 11.05, p < 0.001) were valid predictors of high Ki-67 proliferation index. CONCLUSION Ultrasound performance is valuable for non-invasive prediction of prognostic factors in malignant NMLs.
Collapse
Affiliation(s)
- Tong Wang
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenjuan Guo
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Ultrasound, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xuemei Zhang
- Department of Pathology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ji Ma
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fang Li
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Siqi Zheng
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Miao Zhu
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi Dong
- Department of Ultrasound, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Min Bai
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
6
|
McAleese CE, Choudhury C, Butcher NJ, Minchin RF. Hypoxia-mediated drug resistance in breast cancers. Cancer Lett 2020; 502:189-199. [PMID: 33278499 DOI: 10.1016/j.canlet.2020.11.045] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 11/23/2020] [Accepted: 11/26/2020] [Indexed: 02/07/2023]
Abstract
Tissue hypoxia in solid tumors is caused by several pathological changes associated with tumor growth, including altered microvasculature structure, increased diffusional distances, and tumor-associated anemia. As the oxygen tension decreases, tumor cells adapt to the limited oxygen supply. Previous studies have shown that such adaptation leads to an aggressive phenotype that is resistant to many anti-cancer therapies. Induction of hypoxia inducible factors (HIFs) mediates many proteomic and genomic changes associated with tumor hypoxia. In breast cancers, HIFs not only predict poor prognosis, but also promote metastasis and drug resistance. Several studies have proposed HIF-1α as a druggable target in drug-resistant breast cancers, leading to the synthesis and development of small molecule inhibitors. Disappointingly, however, none of these small molecule inhibitors have progressed to clinical use. In this review, we briefly discuss the role of HIF-1α in breast cancer drug resistance and summarize the current and future approaches to targeting this transcription factor in breast cancer treatment.
Collapse
Affiliation(s)
- Courtney E McAleese
- School of Biomedical Sciences, University of Queensland, Brisbane, 4072, Australia
| | - Chandra Choudhury
- School of Biomedical Sciences, University of Queensland, Brisbane, 4072, Australia
| | - Neville J Butcher
- School of Biomedical Sciences, University of Queensland, Brisbane, 4072, Australia
| | - Rodney F Minchin
- School of Biomedical Sciences, University of Queensland, Brisbane, 4072, Australia.
| |
Collapse
|
7
|
Codelivery of HIF-1α siRNA and Dinaciclib by Carboxylated Graphene Oxide-Trimethyl Chitosan-Hyaluronate Nanoparticles Significantly Suppresses Cancer Cell Progression. Pharm Res 2020; 37:196. [PMID: 32944844 DOI: 10.1007/s11095-020-02892-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 07/24/2020] [Indexed: 01/07/2023]
Abstract
PURPOSE Hypoxia-inducible factor (HIF) is one of the critical components of the tumor microenvironment that is involved in tumor development. HIF-1α functionally and physically interacts with CDK1, 2, and 5 and stimulates the cell cycle progression and Cyclin-Dependent Kinase (CDK) expression. Therefore, hypoxic tumor microenvironment and CDK overexpression lead to increased cell cycle progression and tumor expansion. Therefore, we decided to suppress cancer cell expansion by blocking HIF-1α and CDK molecules. METHODS In the present study, we used the carboxylated graphene oxide (CGO) conjugated with trimethyl chitosan (TMC) and hyaluronate (HA) nanoparticles (NPs) loaded with HIF-1α-siRNA and Dinaciclib, the CDK inhibitor, for silencing HIF-1α and blockade of CDKs in CD44-expressing cancer cells and evaluated the impact of combination therapy on proliferation, metastasis, apoptosis, and tumor growth. RESULTS The results indicated that the manufactured NPs had conceivable physicochemical properties, high cellular uptake, and low toxicity. Moreover, combination therapy of cancer cells using CGO-TMC-HA NPs loaded with HIF-1α siRNA and Dinaciclib (SCH 727965) significantly suppressed the CDKs/HIF-1α and consequently, decreased the proliferation, migration, angiogenesis, and colony formation in tumor cells. CONCLUSIONS These results indicate the ability of CGO-TMC-HA NPs for dual drug/gene delivery in cancer treatment. Furthermore, the simultaneous inhibition of CDKs/HIF-1α can be considered as a novel anti-cancer treatment strategy; however, further research is needed to confirm this treatment in vivo. Graphical Abstract The suppression of HIF-1α and CDKs inhibits cancer growth. HIF-1α is overexpressed by the cells present in the tumor microenvironment. The hypoxic environment elevates mitochondrial ROS production and increases p38 MAP kinase, JAK/STAT, ERK, JNK, and Akt/PI3K signaling, resulting in cyclin accumulation and aberrant cell cycle progression. Furthermore, the overexpression of HIF-1α/CDK results in increased expression of genes such as BCL2, Bcl-xl, Ki-67, TGFβ, VEGF, FGF, MMP2, MMP9, and, HIF-1α and consequently raise the survival, proliferation, angiogenesis, metastasis, and invasion of tumor cells. In conclusion, HIF-1α-siRNA/Dinaciclib-loaded CGO-TMC-HA NPs can inhibit the tumor expansion by blockage of CDKs and HIF-1α (JAK: Janus kinase, STAT: Signal transducer and activator of transcription, MAPK: mitogen-activated protein kinase, ERK: extracellular signal-regulated kinase, JNK: c-Jun N-terminal kinase, PI3K: phosphatidylinositol 3-kinase).
Collapse
|
8
|
Macan AM, Harej A, Cazin I, Klobučar M, Stepanić V, Pavelić K, Pavelić SK, Schols D, Snoeck R, Andrei G, Raić-Malić S. Antitumor and antiviral activities of 4-substituted 1,2,3-triazolyl-2,3-dibenzyl-L-ascorbic acid derivatives. Eur J Med Chem 2019; 184:111739. [PMID: 31586832 PMCID: PMC7115614 DOI: 10.1016/j.ejmech.2019.111739] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 09/25/2019] [Accepted: 09/25/2019] [Indexed: 11/30/2022]
Abstract
Two series of 6-(1,2,3-triazolyl)-2,3-dibenzyl-l-ascorbic acid derivatives with the hydroxyethylene (8a−8u) and ethylidene linkers (10c−10p) were synthesized and evaluated for their antiproliferative activity against seven malignant tumor cell lines and antiviral activity against a broad range of viruses. Conformationally unrestricted spacer between the lactone and 1,2,3-triazole units in 8a−8u series had a profound effect on antitumor activity. Besides, the introduction of a long side chain at C-4 of 1,2,3-triazole that led to the synthesis of decyl-substituted 2,3-dibenzyl-l-ascorbic acid 8m accounted for a selective and potent antiproliferative activity on breast cancer MCF-7 cells cells in the nM range. Further analysis showed that compound 8m strongly enhanced expression of hypoxia inducible transcription factor 1 α (HIF-1α) and to some extent decreased expression of nitric oxide synthase 2 (NOS2) suggesting its role in regulating HIF-1α signalling pathway. The p-methoxyphenyl-substituted derivative 10g displayed specific anti-cytomegalovirus (CMV) potential, whereas aliphatic-substituted derivatives 8l and 8m had the most potent, yet relatively non-specific, anti-varicella-zoster (VZV) activity. Two series of 1,2,3-triazolyl 2,3-dibenzyl-l-ascorbic acid conjugates were synthesized. Conformationally unrestricted spacer had a major effect on antitumor activities. Decyl-substituted l-ascorbic acid 8m caused inhibition of breast cancer MCF-7 cells in the nM range. 8m increased the expression of hypoxia inducible transcription factor HIF-1α. p-Methoxyphenyl-substituted derivative 10g had specific anti-CMV activity.
Collapse
Affiliation(s)
- Andrijana Meščić Macan
- University of Zagreb, Department of Organic Chemistry, Faculty of Chemical Engineering and Technology, Marulićev Trg 20, HR-10000, Zagreb, Croatia
| | - Anja Harej
- University of Rijeka, Department of Biotechnology, Centre for High-throughput Technologies Radmile Matejčić 2, HR-51000, Rijeka, Croatia
| | - Ines Cazin
- University of Zagreb, Department of Organic Chemistry, Faculty of Chemical Engineering and Technology, Marulićev Trg 20, HR-10000, Zagreb, Croatia
| | - Marko Klobučar
- University of Rijeka, Department of Biotechnology, Centre for High-throughput Technologies Radmile Matejčić 2, HR-51000, Rijeka, Croatia
| | - Višnja Stepanić
- Ruđer Bošković Institute, Division of Molecular Medicine, Bijenička Cesta 54, 10 000, Zagreb, Croatia
| | - Krešimir Pavelić
- Juraj Dobrila University of Pula, Zagrebačka 30, 52100, Pula, Croatia
| | - Sandra Kraljević Pavelić
- University of Rijeka, Department of Biotechnology, Centre for High-throughput Technologies Radmile Matejčić 2, HR-51000, Rijeka, Croatia
| | - Dominique Schols
- Rega Institute for Medical Research, KU Leuven, Laboratory of Virology and Chemotherapy, Minderbroedersstraat 10, B-3000, Leuven, Belgium
| | - Robert Snoeck
- Rega Institute for Medical Research, KU Leuven, Laboratory of Virology and Chemotherapy, Minderbroedersstraat 10, B-3000, Leuven, Belgium
| | - Graciela Andrei
- Rega Institute for Medical Research, KU Leuven, Laboratory of Virology and Chemotherapy, Minderbroedersstraat 10, B-3000, Leuven, Belgium
| | - Silvana Raić-Malić
- University of Zagreb, Department of Organic Chemistry, Faculty of Chemical Engineering and Technology, Marulićev Trg 20, HR-10000, Zagreb, Croatia.
| |
Collapse
|
9
|
Vraka I, Panourgias E, Sifakis E, Koureas A, Galanis P, Dellaportas D, Gouliamos A, Antoniou A. Correlation Between Contrast-enhanced Ultrasound Characteristics (Qualitative and Quantitative) and Pathological Prognostic Factors in Breast Cancer. In Vivo 2018; 32:945-954. [PMID: 29936484 DOI: 10.21873/invivo.11333] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 04/21/2018] [Accepted: 04/23/2018] [Indexed: 12/19/2022]
Abstract
BACKGROUND/AIM Little is known about the correlation between contrast-enhanced ultrasound (CEUS) characteristics and pathological prognostic factors in breast cancer. The aim of this study was to explore the correlation between CEUS characteristics and pathological prognostic factors. PATIENTS AND METHODS A retrospective study with 34 malignant breast lesions was conducted. CEUS characteristics included qualitative characteristics (e.g. lesion's enhancement degree and order, internal lesion homogeneity etc.) and quantitative characteristics (e.g. peak intensity, time to peak etc.). Also, pathological prognostic factors were included (e.g. tumor grade, estrogen receptor status etc.). RESULTS Blurred lesion margins were observed more often in tumors of high histological grade (p=0.01) and in estrogen receptor-negative tumors (p=0.049). Furthermore, perilesional enhancement was associated with positive Ki-67 expression (p=0.049), while heterogeneous internal sentinel lymph node enhancement was associated with malignant infiltration of the node (p=0.002). CONCLUSION CEUS has the potential to provide a prevision of pathological prognostic factors in malignant breast lesions, helping in the better early patient management.
Collapse
Affiliation(s)
- Irene Vraka
- Department of Radiology, Aretaieion Hospital, Department of Medicine, National & Kapodistrian University of Athens, Athens, Greece
| | - Evangelia Panourgias
- Department of Radiology, Aretaieion Hospital, Department of Medicine, National & Kapodistrian University of Athens, Athens, Greece
| | - Emmanouil Sifakis
- Department of Oncology-Pathology, Jonas Bergh Research Group, Karolinska Institute, Stockholm, Sweden
| | - Andreas Koureas
- Department of Radiology, Aretaieion Hospital, Department of Medicine, National & Kapodistrian University of Athens, Athens, Greece
| | - Petros Galanis
- Center for Health Services Management and Evaluation, Department of Nursing, National & Kapodistrian University of Athens, Athens, Greece
| | - Dionysios Dellaportas
- 2nd Department of Surgery, Aretaieion Hospital, Department of Medicine, National & Kapodistrian University of Athens, Athens, Greece
| | - Athanasios Gouliamos
- Department of Radiology, Aretaieion Hospital, Department of Medicine, National & Kapodistrian University of Athens, Athens, Greece
| | - Aristides Antoniou
- Department of Radiology, Aretaieion Hospital, Department of Medicine, National & Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
10
|
Guerrini G, Criscuoli M, Filippi I, Naldini A, Carraro F. Inhibition of smoothened in breast cancer cells reduces CAXII expression and cell migration. J Cell Physiol 2018; 233:9799-9811. [PMID: 30132883 DOI: 10.1002/jcp.26947] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 06/12/2018] [Indexed: 12/13/2022]
Abstract
Breast cancer (BC) relapse and metastasis are the leading cause of death and, together with drug resistance, keep mortality still high. The Hedgehog (Hh) pathway is expressed during embryogenesis, organogenesis and in adult tissue homeostasis and its aberrant activation is often associated with cancer. Carbonic anhydrase (CA) enzymes are important during development; they play a key role in controlling several cellular mechanisms, such as pH regulation, survival, and migration, and they are aberrantly expressed in cancer. The goal of this study was to investigate the interplay between the Hh pathway and CAXII in terms of BC cell migration. We here demonstrated that smoothened (SMO) silencing resulted in a reduction of CAXII expression at mRNA and protein level. This led to a decrease in cell migration, which was restored when cells were treated with an SMO agonist, Sag dihydrochloride (SAG), but not when cells were cotreated with SAG and the CAs inhibitor Acetazolamide. This suggested that the ability of SAG to promote cell migration was impaired when CAXII was inhibited. The reduction was also confirmed within hypoxic and inflammatory microenvironment, typical of BC, indicating a key role of the Hh pathway in controlling CAXII expression. Our results may contribute to further understand the physiology of BC cells and indicate that the Hh pathway controls BC cell migration and cell invasion also through CAXII, with important implications in identifying novel therapeutic targets.
Collapse
Affiliation(s)
- Giuditta Guerrini
- Department of Molecular and Developmental Medicine, Cellular and Molecular Physiology Unit, University of Siena, Siena, Italy
| | - Mattia Criscuoli
- Department of Molecular and Developmental Medicine, Cellular and Molecular Physiology Unit, University of Siena, Siena, Italy
| | - Irene Filippi
- Department of Molecular and Developmental Medicine, Cellular and Molecular Physiology Unit, University of Siena, Siena, Italy.,Istituto Toscano Tumori, Firenze, Italy
| | - Antonella Naldini
- Department of Molecular and Developmental Medicine, Cellular and Molecular Physiology Unit, University of Siena, Siena, Italy
| | - Fabio Carraro
- Department of Molecular and Developmental Medicine, Cellular and Molecular Physiology Unit, University of Siena, Siena, Italy.,Istituto Toscano Tumori, Firenze, Italy
| |
Collapse
|
11
|
VRAKA IRENE, PANOURGIAS EVANGELIA, SIFAKIS EMMANOUIL, KOUREAS ANDREAS, GALANIS PETROS, DELLAPORTAS DIONYSIOS, GOULIAMOS ATHANASIOS, ANTONIOU ARISTIDES. Correlation Between Contrast-enhanced Ultrasound Characteristics (Qualitative and Quantitative) and Pathological Prognostic Factors in Breast Cancer. In Vivo 2018; 32. [PMID: 29936484 PMCID: PMC6117754 DOI: 10.21873/invivo.112333] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
BACKGROUND/AIM Little is known about the correlation between contrast-enhanced ultrasound (CEUS) characteristics and pathological prognostic factors in breast cancer. The aim of this study was to explore the correlation between CEUS characteristics and pathological prognostic factors. PATIENTS AND METHODS A retrospective study with 34 malignant breast lesions was conducted. CEUS characteristics included qualitative characteristics (e.g. lesion's enhancement degree and order, internal lesion homogeneity etc.) and quantitative characteristics (e.g. peak intensity, time to peak etc.). Also, pathological prognostic factors were included (e.g. tumor grade, estrogen receptor status etc.). RESULTS Blurred lesion margins were observed more often in tumors of high histological grade (p=0.01) and in estrogen receptor-negative tumors (p=0.049). Furthermore, perilesional enhancement was associated with positive Ki-67 expression (p=0.049), while heterogeneous internal sentinel lymph node enhancement was associated with malignant infiltration of the node (p=0.002). CONCLUSION CEUS has the potential to provide a prevision of pathological prognostic factors in malignant breast lesions, helping in the better early patient management.
Collapse
Affiliation(s)
- IRENE VRAKA
- Department of Radiology, Aretaieion Hospital, Department of Medicine, National & Kapodistrian University of Athens, Athens, Greece
| | - EVANGELIA PANOURGIAS
- Department of Radiology, Aretaieion Hospital, Department of Medicine, National & Kapodistrian University of Athens, Athens, Greece
| | - EMMANOUIL SIFAKIS
- Department of Oncology-Pathology, Jonas Bergh Research Group, Karolinska Institute, Stockholm, Sweden
| | - ANDREAS KOUREAS
- Department of Radiology, Aretaieion Hospital, Department of Medicine, National & Kapodistrian University of Athens, Athens, Greece
| | - PETROS GALANIS
- Center for Health Services Management and Evaluation, Department of Nursing, National & Kapodistrian University of Athens, Athens, Greece
| | - DIONYSIOS DELLAPORTAS
- 2nd Department of Surgery, Aretaieion Hospital, Department of Medicine, National & Kapodistrian University of Athens, Athens, Greece
| | - ATHANASIOS GOULIAMOS
- Department of Radiology, Aretaieion Hospital, Department of Medicine, National & Kapodistrian University of Athens, Athens, Greece
| | - ARISTIDES ANTONIOU
- Department of Radiology, Aretaieion Hospital, Department of Medicine, National & Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
12
|
Harrison H, Pegg HJ, Thompson J, Bates C, Shore P. HIF1-alpha expressing cells induce a hypoxic-like response in neighbouring cancer cells. BMC Cancer 2018; 18:674. [PMID: 29925335 PMCID: PMC6011406 DOI: 10.1186/s12885-018-4577-1] [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: 03/05/2018] [Accepted: 06/05/2018] [Indexed: 12/26/2022] Open
Abstract
Background Hypoxia stimulates metastasis in cancer and is linked to poor patient prognosis. In tumours, oxygen levels vary and hypoxic regions exist within a generally well-oxygenated tumour. However, whilst the heterogeneous environment is known to contribute to metastatic progression, little is known about the mechanism by which heterogeneic hypoxia contributes to cancer progression. This is largely because existing experimental models do not recapitulate the heterogeneous nature of hypoxia. The primary effector of the hypoxic response is the transcription factor Hypoxia inducible factor 1-alpha (HIF1-alpha). HIF1-alpha is stabilised in response to low oxygen levels in the cellular environment and its expression is seen in hypoxic regions throughout the tumour. Methods We have developed a model system in which HIF1-alpha can be induced within a sub-population of cancer cells, thus enabling us to mimic the effects of heterogeneic HIF1-alpha expression. Results We show that induction of HIF1-alpha not only recapitulates elements of the hypoxic response in the induced cells but also results in significant changes in proliferation, gene expression and mammosphere formation within the HIF1-alpha negative population. Conclusions These findings suggest that the HIF1-alpha expressing cells found within hypoxic regions are likely to contribute to the subsequent progression of a tumour by modifying the behaviour of cells in the non-hypoxic regions of the local micro-environment. Electronic supplementary material The online version of this article (10.1186/s12885-018-4577-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hannah Harrison
- Faculty of Biology, Medicine and Health, University of Manchester, Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK.
| | - Henry J Pegg
- Faculty of Biology, Medicine and Health, University of Manchester, Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK
| | - Jamie Thompson
- Faculty of Biology, Medicine and Health, University of Manchester, Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK
| | - Christian Bates
- Faculty of Biology, Medicine and Health, University of Manchester, Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK
| | - Paul Shore
- Faculty of Biology, Medicine and Health, University of Manchester, Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK.
| |
Collapse
|
13
|
Zhou F, Li M, Wei Y, Lin K, Lu Y, Shen J, Johanning GL, Wang-Johanning F. Activation of HERV-K Env protein is essential for tumorigenesis and metastasis of breast cancer cells. Oncotarget 2018; 7:84093-84117. [PMID: 27557521 PMCID: PMC5356647 DOI: 10.18632/oncotarget.11455] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 08/11/2016] [Indexed: 12/12/2022] Open
Abstract
Human endogenous retrovirus type K (HERV-K) Env protein was previously demonstrated to be overexpressed in human breast cancer (BC) cells and tissues. However, the molecular pathways driving the specific alterations are unknown. We now show that knockdown of its expression with an shRNA (shRNAenv) blocked BC cell proliferation, migration, and invasion. shRNAenv transduction also attenuated the ability of BC cells to form tumors, and notably prevented metastasis. Mechanistically, downregulation of HERV-K blocked expression of tumor-associated genes that included Ras, p-RSK, and p-ERK. The major upstream regulators influenced by HERV-K knockdown were p53, TGF- β1, and MYC. Of interest, when the HERV-K env gene was overexpressed in shRNAenv-transduced BC cells using an HERV-K env expression vector, Ras/Raf/MEK/ERK pathway signaling was restored. CDK5, which alters p53 phosphorylation in some cancers, was upregulated and p53 was downregulated when HERV-K was overexpressed. CDK5 is also a mediator of TGF-β1-induced epithelial-mesenchymal transition and migration in cancer cells, and is involved in tumor formation. Importantly, reductions in migration, invasion, and transformation of BC cells stably transduced with shRNAenv was reversed after adding back a vector with a synonymous mutation of HERV-K env. Taken together, these results indicate that HERV-K Env protein plays an important role in tumorigenesis and metastasis of BC.
Collapse
Affiliation(s)
- Fuling Zhou
- Viral Oncology Program, Center for Cancer and Metabolism, SRI International, Menlo Park, California, USA.,Department of Clinical Hematology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| | - Ming Li
- Viral Oncology Program, Center for Cancer and Metabolism, SRI International, Menlo Park, California, USA
| | - Yongchang Wei
- Viral Oncology Program, Center for Cancer and Metabolism, SRI International, Menlo Park, California, USA.,Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| | - Kevin Lin
- Department of Epigenetics and Molecular Carcinogenesis, Science Park, The University of Texas MD Anderson Cancer Center, Smithville, Texas, USA
| | - Yue Lu
- Department of Epigenetics and Molecular Carcinogenesis, Science Park, The University of Texas MD Anderson Cancer Center, Smithville, Texas, USA
| | - Jianjun Shen
- Department of Epigenetics and Molecular Carcinogenesis, Science Park, The University of Texas MD Anderson Cancer Center, Smithville, Texas, USA
| | - Gary L Johanning
- Viral Oncology Program, Center for Cancer and Metabolism, SRI International, Menlo Park, California, USA
| | - Feng Wang-Johanning
- Viral Oncology Program, Center for Cancer and Metabolism, SRI International, Menlo Park, California, USA
| |
Collapse
|
14
|
Crowley PD, Stuttgen V, O'Carroll E, Ash SA, Buggy DJ, Gallagher HC. Exposure to 60% oxygen promotes migration and upregulates angiogenesis factor secretion in breast cancer cells. Med Gas Res 2018; 7:226-235. [PMID: 29497482 PMCID: PMC5806442 DOI: 10.4103/2045-9912.222446] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Peri-operative factors, including anaesthetic drugs and techniques, may affect cancer cell biology and clinical recurrence. In breast cancer cells, we demonstrated that sevoflurane promotes migration and angiogenesis in high fractional oxygen but not in air. Follow-up analysis of the peri-operative oxygen fraction trial found an association between high inspired oxygen during cancer surgery and reduced tumor-free survival. Here we evaluated effects of acute, high oxygen exposure on breast cancer cell viability, migration and secretion of angiogenesis factors in vitro. MDA-MB-231 and MCF-7 breast cancer cells were exposed to 21%, 30%, 60%, or 80% v/v O2 for 3 hours. Cell viability at 24 hours was determined by MTT and migration at 24 hours with the Oris™ Cell Migration Assay. Secretion of angiogenesis factors at 24 hours was measured via membrane-based immunoarray. Exposure to 30%, 60% or 80% oxygen did not affect cell viability. Migration of MDA-MB-231 and MCF-7 cells was increased by 60% oxygen (P = 0.012 and P = 0.007, respectively) while 30% oxygen increased migration in MCF-7 cells (P = 0.011). These effects were reversed by dimethyloxaloylglycine. In MDA-MB-231 cells high fractional oxygen increased secretion of angiogenesis factors monocyte chemotactic protein 1, regulated on activation normal T-cell expressed and vascular endothelial growth factor. In MCF-7 cells, interleukin-8, angiogenin and vascular endothelial growth factor secretion was significantly increased by high fractional oxygen. High oxygen exposure stimulates migration and secretion of angiogenesis factors in breast cancer cells in vitro.
Collapse
Affiliation(s)
- Peter D Crowley
- School of Medicine, Conway Institute, University College Dublin, Dublin, Ireland
| | - Vivian Stuttgen
- School of Medicine, Conway Institute, University College Dublin, Dublin, Ireland.,School of Veterinary Medicine, Conway Institute, University College Dublin, Dublin, Ireland
| | - Emma O'Carroll
- School of Medicine, Conway Institute, University College Dublin, Dublin, Ireland
| | - Simon A Ash
- School of Medicine, Conway Institute, University College Dublin, Dublin, Ireland.,Discipline of Anesthesia, Memorial University, Canada
| | - Donal J Buggy
- School of Medicine, Conway Institute, University College Dublin, Dublin, Ireland.,Mater Misericordiae University Hospital, Dublin, Ireland.,Outcomes Research Consortium, Cleveland Clinic, Cleveland, OH, USA.,UCD-Mater Clinical Research Centre, Catherine McCauley Centre, Dublin, Ireland
| | - Helen C Gallagher
- School of Medicine, Conway Institute, University College Dublin, Dublin, Ireland.,UCD-Mater Clinical Research Centre, Catherine McCauley Centre, Dublin, Ireland
| |
Collapse
|
15
|
The ERβ4 variant induces transformation of the normal breast mammary epithelial cell line MCF-10A; the ERβ variants ERβ2 and ERβ5 increase aggressiveness of TNBC by regulation of hypoxic signaling. Oncotarget 2018; 9:12201-12211. [PMID: 29552303 PMCID: PMC5844739 DOI: 10.18632/oncotarget.24134] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Accepted: 11/05/2017] [Indexed: 01/03/2023] Open
Abstract
Triple negative breast cancer (TNBC) still remains a challenge to treat in the clinic due to a lack of good targets for treatment. Although TNBC lacks expression of ERα, the expression of ERβ and its variants are detected quite frequently in this cancer type and can represent an avenue for treatment. We show that two of the variants of ERβ, namely ERβ2 and ERβ5, control aggressiveness of TNBC by regulating hypoxic signaling through stabilization of HIF-1α. RNA-seq of patient derived xenografts (PDX) from TNBC shows expression of ERβ2, ERβ4 and ERβ5 variants in more than half of the samples. Furthermore, expression of ERβ4 in the immortalized, normal mammary epithelial cell line MCF-10A that is resistant to tumorsphere formation caused transformation and development of tumorspheres. By contrast, ERβ1, ERβ2 or ERβ5 were unable to support tumorsphere formation. We have previously shown that all variants except ERβ1 stabilize HIF-1α but only ERβ4 appears to have the ability to transform normal mammary epithelial cells, pointing towards a unique property of ERβ4. We propose that ERβ variants may be good diagnostic tools and also serve as novel targets for treatment of breast cancer.
Collapse
|
16
|
Park J, Lee Y. Hypoxia induced phosphorylation of estrogen receptor at serine 118 in the absence of ligand. J Steroid Biochem Mol Biol 2017; 174:146-152. [PMID: 28847747 DOI: 10.1016/j.jsbmb.2017.08.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 08/18/2017] [Accepted: 08/21/2017] [Indexed: 10/19/2022]
Abstract
The estrogen receptor (ER) plays an important role in breast cancer development and progression. Hypoxia modulates the level of ERα expression and induces ligand-independent transcriptional activation of ERα, which is closely related with the biology of breast carcinomas. Since phosphorylation itself affects the transcriptional activity and stabilization of ERα, we examined changes in ERα phosphorylation under hypoxic conditions. Hypoxia induced phosphorylation of ERα at serine residue 118 (S118) in the absence of estrogen through the mitogen-activated protein kinase (MAPK)/ERK1/2 pathway. Cell proliferation was significantly decreased under normoxia or hypoxia when ERα harboring the S118A mutation was overexpressed. Our previous studies showed that ER degradation is the most prominent phenomenon under hypoxia. E2-induced ER protein downregulation is dependent on phosphorylation of S118. However, hypoxia-induced ERα degradation did not involve S118 phosphorylation. Our study implies the existence of a differential mechanism between E2 and hypoxia-mediated ERα protein degradation. Understanding the mechanistic behavior of ER under hypoxia will likely facilitate understanding of endocrine therapy resistance and development of treatment strategies for breast cancer.
Collapse
Affiliation(s)
- Joonwoo Park
- College of Life Science, Department of Integrated Bioscience and Biotechnology, Sejong University, Seoul 143-747, South Korea
| | - YoungJoo Lee
- College of Life Science, Department of Integrated Bioscience and Biotechnology, Sejong University, Seoul 143-747, South Korea.
| |
Collapse
|
17
|
Ji CL, Li XL, He YP, Li DD, Gu XG, Xu HX. Quantitative parameters of contrast-enhanced ultrasound in breast invasive ductal carcinoma: The correlation with pathological prognostic factors. Clin Hemorheol Microcirc 2017; 66:333-345. [PMID: 28387663 DOI: 10.3233/ch-170251] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Chen-Li Ji
- Department of Medical Ultrasound, Shanghai Tenth People’s Hospital, Ultrasound Research and Education Institute, Tongji University School of Medicine, Shanghai, China
- Department of Medical Ultrasound, Shanghai Putuo District Central Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiao-Long Li
- Department of Medical Ultrasound, Shanghai Tenth People’s Hospital, Ultrasound Research and Education Institute, Tongji University School of Medicine, Shanghai, China
| | - Ya-Ping He
- Department of Medical Ultrasound, Shanghai Tenth People’s Hospital, Ultrasound Research and Education Institute, Tongji University School of Medicine, Shanghai, China
| | - Dan-Dan Li
- Department of Medical Ultrasound, Shanghai Tenth People’s Hospital, Ultrasound Research and Education Institute, Tongji University School of Medicine, Shanghai, China
| | - Xin-Gang Gu
- Department of Medical Ultrasound, Shanghai Putuo District Central Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hui-Xiong Xu
- Department of Medical Ultrasound, Shanghai Tenth People’s Hospital, Ultrasound Research and Education Institute, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
18
|
Nalwoga H, Ahmed L, Arnes JB, Wabinga H, Akslen LA. Strong Expression of Hypoxia-Inducible Factor-1α (HIF-1α) Is Associated with Axl Expression and Features of Aggressive Tumors in African Breast Cancer. PLoS One 2016; 11:e0146823. [PMID: 26760782 PMCID: PMC4711940 DOI: 10.1371/journal.pone.0146823] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 12/22/2015] [Indexed: 01/11/2023] Open
Abstract
PURPOSE Inhibition of hypoxia-inducible factor (HIF) and Axl receptor tyrosine kinase is being evaluated for targeted therapy in solid tumors. Both HIF-1α and Axl influence tumor growth and metastatic potential, and they have been linked to treatment failure in many cancers. However, there is a lack of reports on HIF-1α expression in African breast cancer, which has a poor prognosis, and novel treatment targets must therefore be established. Here, we aimed to evaluate HIF-1α in relation to Axl expression, angiogenesis markers, and other tumor characteristics in a series of African breast cancer. METHODS Using immunohistochemistry, we examined 261 invasive breast cancers on tissue microarrays for HIF-1α and Axl as well as several other markers, and a subset of 185 cases had information on VEGF (vascular endothelial growth factor) expression, microvessel density (MVD), proliferating microvessel density (pMVD) and vascular proliferation index (VPI) for important comparisons. RESULTS Strong HIF-1α expression was associated with increased Axl (p = 0.007), VEGF (p<0.0005), and p53 (p = 0.032) expression, as well as high tumor cell proliferation by Ki-67 (p = 0.006), and high tumor grade (p = 0.003). Tumors with strong HIF-1α expression had significantly higher MVD (p = 0.019) and higher pMVD (p = 0.027) than tumors with weak expression. CONCLUSIONS High HIF-1α expression is significantly associated with Axl and VEGF expression, and with markers of poor prognosis in this series of breast cancer, suggesting HIF-1α and Axl as potential therapeutic targets in African breast cancer.
Collapse
Affiliation(s)
- Hawa Nalwoga
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, University of Bergen, Haukeland University Hospital, Bergen, Norway
- Department of Pathology, School of Biomedical Sciences, Makerere University College of Health Sciences, Kampala, Uganda
| | - Lavina Ahmed
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, University of Bergen, Haukeland University Hospital, Bergen, Norway
- BerGenBio AS, Bergen, Norway
| | - Jarle B. Arnes
- Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Henry Wabinga
- Department of Pathology, School of Biomedical Sciences, Makerere University College of Health Sciences, Kampala, Uganda
| | - Lars A. Akslen
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, University of Bergen, Haukeland University Hospital, Bergen, Norway
- Department of Pathology, Haukeland University Hospital, Bergen, Norway
- * E-mail:
| |
Collapse
|
19
|
Yang J, AlTahan A, Jones DT, Buffa FM, Bridges E, Interiano RB, Qu C, Vogt N, Li JL, Baban D, Ragoussis J, Nicholson R, Davidoff AM, Harris AL. Estrogen receptor-α directly regulates the hypoxia-inducible factor 1 pathway associated with antiestrogen response in breast cancer. Proc Natl Acad Sci U S A 2015; 112:15172-7. [PMID: 26598706 PMCID: PMC4679044 DOI: 10.1073/pnas.1422015112] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
A majority of breast cancers are driven by estrogen via estrogen receptor-α (ERα). Our previous studies indicate that hypoxia-inducible factor 1α (HIF-1α) cooperates with ERα in breast cancer cells. However, whether ERα is implicated in the direct regulation of HIF-1α and the role of HIF-1α in endocrine therapy response are unknown. In this study we found that a subpopulation of HIF-1α targets, many of them bearing both hypoxia response elements and estrogen response elements, are regulated by ERα in normoxia and hypoxia. Interestingly, the HIF-1α gene itself also bears an estrogen response element, and its expression is directly regulated by ERα. Clinical data revealed that expression of the HIF-1α gene or a hypoxia metagene signature is associated with a poor outcome to endocrine treatment in ERα(+) breast cancer. HIF-1α was able to confer endocrine therapy resistance to ERα(+) breast cancer cells. Our findings define, for the first time to our knowledge, a direct regulatory pathway between ERα and HIF-1α, which might modulate hormone response in treatment.
Collapse
Affiliation(s)
- Jun Yang
- Growth Factor Group, Cancer Research UK, Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford OX3 9DS, United Kingdom; Department of Surgery, St. Jude Children's Research Hospital, Memphis, TN 38105;
| | - Alaa AlTahan
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, TN 38105
| | - Dylan T Jones
- Growth Factor Group, Cancer Research UK, Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford OX3 9DS, United Kingdom
| | - Francesca M Buffa
- Growth Factor Group, Cancer Research UK, Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford OX3 9DS, United Kingdom
| | - Esther Bridges
- Growth Factor Group, Cancer Research UK, Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford OX3 9DS, United Kingdom
| | - Rodrigo B Interiano
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, TN 38105
| | - Chunxu Qu
- Department of Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN 38105
| | - Nathan Vogt
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, TN 38105
| | - Ji-Liang Li
- Growth Factor Group, Cancer Research UK, Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford OX3 9DS, United Kingdom
| | - Dilair Baban
- Genomics Group, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, United Kingdom
| | - Jiannis Ragoussis
- Genomics Group, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, United Kingdom
| | - Robert Nicholson
- Tenovus Centre for Cancer Research, Welsh School of Pharmacy, Cardiff University, Cardiff CF10 3NB, United Kingdom
| | - Andrew M Davidoff
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, TN 38105
| | - Adrian L Harris
- Growth Factor Group, Cancer Research UK, Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford OX3 9DS, United Kingdom
| |
Collapse
|
20
|
Feitelson MA, Arzumanyan A, Kulathinal RJ, Blain SW, Holcombe RF, Mahajna J, Marino M, Martinez-Chantar ML, Nawroth R, Sanchez-Garcia I, Sharma D, Saxena NK, Singh N, Vlachostergios PJ, Guo S, Honoki K, Fujii H, Georgakilas AG, Bilsland A, Amedei A, Niccolai E, Amin A, Ashraf SS, Boosani CS, Guha G, Ciriolo MR, Aquilano K, Chen S, Mohammed SI, Azmi AS, Bhakta D, Halicka D, Keith WN, Nowsheen S. Sustained proliferation in cancer: Mechanisms and novel therapeutic targets. Semin Cancer Biol 2015; 35 Suppl:S25-S54. [PMID: 25892662 PMCID: PMC4898971 DOI: 10.1016/j.semcancer.2015.02.006] [Citation(s) in RCA: 464] [Impact Index Per Article: 46.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 02/20/2015] [Accepted: 02/23/2015] [Indexed: 02/08/2023]
Abstract
Proliferation is an important part of cancer development and progression. This is manifest by altered expression and/or activity of cell cycle related proteins. Constitutive activation of many signal transduction pathways also stimulates cell growth. Early steps in tumor development are associated with a fibrogenic response and the development of a hypoxic environment which favors the survival and proliferation of cancer stem cells. Part of the survival strategy of cancer stem cells may manifested by alterations in cell metabolism. Once tumors appear, growth and metastasis may be supported by overproduction of appropriate hormones (in hormonally dependent cancers), by promoting angiogenesis, by undergoing epithelial to mesenchymal transition, by triggering autophagy, and by taking cues from surrounding stromal cells. A number of natural compounds (e.g., curcumin, resveratrol, indole-3-carbinol, brassinin, sulforaphane, epigallocatechin-3-gallate, genistein, ellagitannins, lycopene and quercetin) have been found to inhibit one or more pathways that contribute to proliferation (e.g., hypoxia inducible factor 1, nuclear factor kappa B, phosphoinositide 3 kinase/Akt, insulin-like growth factor receptor 1, Wnt, cell cycle associated proteins, as well as androgen and estrogen receptor signaling). These data, in combination with bioinformatics analyses, will be very important for identifying signaling pathways and molecular targets that may provide early diagnostic markers and/or critical targets for the development of new drugs or drug combinations that block tumor formation and progression.
Collapse
Affiliation(s)
- Mark A Feitelson
- Department of Biology, Temple University, Philadelphia, PA, United States.
| | - Alla Arzumanyan
- Department of Biology, Temple University, Philadelphia, PA, United States
| | - Rob J Kulathinal
- Department of Biology, Temple University, Philadelphia, PA, United States
| | - Stacy W Blain
- Department of Pediatrics, State University of New York, Downstate Medical Center, Brooklyn, NY, United States
| | - Randall F Holcombe
- Tisch Cancer Institute, Mount Sinai School of Medicine, New York, NY, United States
| | - Jamal Mahajna
- MIGAL-Galilee Technology Center, Cancer Drug Discovery Program, Kiryat Shmona, Israel
| | - Maria Marino
- Department of Science, University Roma Tre, V.le G. Marconi, 446, 00146 Rome, Italy
| | - Maria L Martinez-Chantar
- Metabolomic Unit, CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Technology Park of Bizkaia, Bizkaia, Spain
| | - Roman Nawroth
- Department of Urology, Klinikum rechts der Isar der Technischen Universität München, Munich, Germany
| | - Isidro Sanchez-Garcia
- Experimental Therapeutics and Translational Oncology Program, Instituto de Biología Molecular y Celular del Cáncer, CSIC/Universidad de Salamanca, Salamanca, Spain
| | - Dipali Sharma
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Neeraj K Saxena
- Department of Oncology, Johns Hopkins University School of Medicine and the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, United States
| | - Neetu Singh
- Tissue and Cell Culture Unit, CSIR-Central Drug Research Institute, Council of Scientific & Industrial Research, Lucknow, India
| | | | - Shanchun Guo
- Department of Microbiology, Biochemistry & Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Kanya Honoki
- Department of Orthopedic Surgery, Nara Medical University, Kashihara 634-8521, Japan
| | - Hiromasa Fujii
- Department of Orthopedic Surgery, Nara Medical University, Kashihara 634-8521, Japan
| | - Alexandros G Georgakilas
- Physics Department, School of Applied Mathematical and Physical Sciences, National Technical University of Athens, Zografou 15780, Athens, Greece
| | - Alan Bilsland
- Institute of Cancer Sciences, University of Glasgow, UK
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Elena Niccolai
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Amr Amin
- Department of Biology, College of Science, UAE University, Al-Ain, United Arab Emirates
| | - S Salman Ashraf
- Department of Chemistry, College of Science, UAE University, Al-Ain, United Arab Emirates
| | - Chandra S Boosani
- Department of BioMedical Sciences, Creighton University, Omaha, NE, United States
| | - Gunjan Guha
- School of Chemical and Bio Technology, SASTRA University, Thanjavur, India
| | - Maria Rosa Ciriolo
- Department of Biology, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Katia Aquilano
- Department of Biology, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Sophie Chen
- Department of Research and Development, Ovarian and Prostate Cancer Research Trust Laboratory, Guildford, Surrey GU2 7YG, United Kingdom
| | - Sulma I Mohammed
- Department of Comparative Pathobiology, Purdue University Center for Cancer Research, West Lafayette, IN, United States
| | - Asfar S Azmi
- Department of Pathology, Karmonas Cancer Institute, Wayne State University School of Medicine, Detroit, MI, United States
| | - Dipita Bhakta
- School of Chemical and Bio Technology, SASTRA University, Thanjavur, India
| | - Dorota Halicka
- Brander Cancer Research Institute, Department of Pathology, New York Medical College, Valhalla, NY, United States
| | - W Nicol Keith
- Institute of Cancer Sciences, University of Glasgow, UK
| | - Somaira Nowsheen
- Mayo Graduate School, Mayo Medical School, Mayo Clinic Medical Scientist Training Program, Rochester, MN, United States
| |
Collapse
|
21
|
Jiwa LS, van Diest PJ, Hoefnagel LD, Wesseling J, Wesseling P, Moelans CB. Upregulation of Claudin-4, CAIX and GLUT-1 in distant breast cancer metastases. BMC Cancer 2014; 14:864. [PMID: 25417118 PMCID: PMC4247109 DOI: 10.1186/1471-2407-14-864] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 11/11/2014] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Several studies have shown that the immunophenotype of distant breast cancer metastases may differ significantly from that of the primary tumor, especially with regard to differences in the level of hormone receptor protein expression, a process known as receptor conversion. This study aimed to compare expression levels of several membrane proteins between primary breast tumors and their corresponding distant metastases in view of their potential applicability for molecular imaging and drug targeting. METHODS Expression of Claudin-4, EGFR, CAIX, GLUT-1 and IGF1R was assessed by immunohistochemistry on tissue microarrays composed of 97 paired primary breast tumors and their distant (non-bone) metastases. RESULTS In both the primary cancers and the metastases, Claudin-4 was most frequently expressed, followed by GLUT-1, CAIX and EGFR.From primary breast cancers to their distant metastases there was positive to negative conversion, e.g. protein expression in the primary tumor with no expression in its paired metastasis, in 6%, 19%, 12%, 38%, and 0% for Claudin-4 (n.s), GLUT-1 (n.s), CAIX (n.s), EGFR (n.s) and IGF1R (n.s) respectively. Negative to positive conversion was seen in 65%, 47%, 43%, 9% and 0% of cases for Claudin-4 (p = 0.049), GLUT-1 (p = 0.024), CAIX (p = 0.002), EGFR (n.s.) and IGF1R (n.s.) respectively. Negative to positive conversion of Claudin-4 in the metastasis was significantly associated with tumor size (p = 0.015), negative to positive conversion of EGFR with negative PR status (p = 0.046) and high MAI (p = 0.047) and GLUT-1 negative to positive conversion with (neo)adjuvant chemotherapy (p = 0.039) and time to metastasis formation (p = 0.034). CAIX and GLUT-1 expression in the primary tumor were significantly associated with high MAI (p = 0.008 and p = 0.038 respectively). CONCLUSION Claudin-4 is frequently expressed in primary breast cancers but especially in their metastases and is thereby an attractive membrane bound molecular imaging and drug target. Conversion in expression of the studied proteins from the primary tumor to metastases was fairly frequent, except for IGF1R, implying that the expression status of metastases cannot always be reliably predicted from the primary tumor, thereby necessitating biopsy for reliable assessment.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Cathy B Moelans
- Department of Pathology, University Medical Center Utrecht, Heidelberglaan 100, PO Box 85500, Utrecht 3508GA, The Netherlands.
| |
Collapse
|
22
|
Liu Y, Hu Z. Identification of collaborative driver pathways in breast cancer. BMC Genomics 2014; 15:605. [PMID: 25034939 PMCID: PMC4111852 DOI: 10.1186/1471-2164-15-605] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 07/07/2014] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND An important challenge in cancer biology is to computationally screen mutations in cancer cells, separating those that might drive cancer initiation and progression, from the much larger number of bystanders. Since mutations are large in number and diverse in type, the frequency of any particular mutation pattern across a set of samples is low. This makes statistical distinctions and reproducibility across different populations difficult to establish. RESULTS In this paper we develop a novel method that promises to partially ameliorate these problems. The basic idea is although mutations are highly heterogeneous and vary from one sample to another, the processes that are disrupted when cells undergo transformation tend to be invariant across a population for a particular cancer or cancer subtype. Specifically, we focus on finding mutated pathway-groups that are invariant across samples of breast cancer subtypes. The identification of informative pathway-groups consists of two steps. The first is identification of pathways significantly enriched in genes containing non-synonymous mutations; the second uses pathways so identified to find groups that are functionally related in the largest number of samples. An application to 4 subtypes of breast cancer identified pathway-groups that can highly explicate a particular subtype and rich in processes associated with transformation. CONCLUSIONS In contrast to previous methods that identify pathways across a set of samples without any further validation, we show that mutated pathway-groups can be found in each breast cancer subtype and that such groups are invariant across the majority of samples. The algorithm is available at http://www.visantnet.org/misi/MUDPAC.zip.
Collapse
Affiliation(s)
- Yang Liu
- Bioinformatics Graduate Program and Department of Biomedical Engineering, Boston University, 24 Cummington Mall, Boston, MA 02215 USA
| | - Zhenjun Hu
- Bioinformatics Graduate Program and Department of Biomedical Engineering, Boston University, 24 Cummington Mall, Boston, MA 02215 USA
| |
Collapse
|
23
|
Feng L, Tao L, Dawei H, Xuliang L, Xiaodong L. HIF-1α expression correlates with cellular apoptosis, angiogenesis and clinical prognosis in rectal carcinoma. Pathol Oncol Res 2013; 20:603-10. [PMID: 24374863 DOI: 10.1007/s12253-013-9738-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Accepted: 12/12/2013] [Indexed: 12/24/2022]
Abstract
Regional hypoxia caused by accelerated cell proliferation and overgrowth is an important characteristic of neoplasm. Hypoxia can cause a series of changes in gene transcription and protein expression, thereby not only inducing tumor cell resistance to radiotherapy and chemotherapy but also promoting tumor invasion and metastasis. This study aimed to investigate the relationship between HIF-1α expression and cellular apoptosis, angiogenesis and clinical prognosis in rectal carcinoma. In 113 rectal carcinoma cases, cellular apoptosis was analyzed by the in situ terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling (TUNEL) assay, whereas the levels of HIF-1α expression, VEGF expression, microvessel density (MVD) and lymphatic vessel density(LVD) were examined by immunohistochemical staining. HIF-1 expression was detected in 67 of 113 rectal carcinoma cases (59.3 %). A positive correlation was found among HIF-1α expression, cellular apoptosis and angiogenesis. The 5-year survival rate in the HIF-1α-negative group was significantly higher than that in the HIF-1α-positive group (81.34 % versus 50 %, P < 0.05). According to the Cox regression analysis, HIF-1α expression, VEGF expression and cellular apoptosis index were independent risk factors for clinical prognosis in rectal carcinoma. Aberrant HIF-1α expression correlates with apoptosis inhibition, angiogenesis and poor prognosis in rectal carcinoma.
Collapse
Affiliation(s)
- Liu Feng
- Department of Urinary Surgery, the Children's Hospital, Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorder, Key Laboratory of Pediatrics in Chongqing (CSTC2009CA5002), Chongqing, 400014, China
| | | | | | | | | |
Collapse
|
24
|
Abstract
There are areas of limited oxygen availability in most solid tumours, including breast cancer. Hypoxia in solid tumours is mainly a consequence of poor perfusion. Structural and functional abnormalities of newly formed tumour vessels cause spatial and temporal heterogeneity of tissue perfusion. The two principal mediators of hypoxia response, HIF-1 and HIF-2, are known to be stabilized at different oxygen levels and to have different temporal responses to hypoxia. Recently, stromal HIF-1 and HIF-2 have been suggested to have opposing roles in breast cancer progression. There is an established link between intralesional, severe hypoxia near areas of necrosis with high levels of HIF-1 and poor prognosis in breast cancer. However, the biological effects of moderate hypoxia and the hypoxic response of stromal cells are currently topics of intense investigation.
Collapse
Affiliation(s)
- H Rundqvist
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden.
| | | |
Collapse
|
25
|
How does hypoxia inducible factor-1α participate in enhancing the glycolysis activity in cervical cancer? Ann Diagn Pathol 2013; 17:305-11. [DOI: 10.1016/j.anndiagpath.2012.12.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2012] [Revised: 11/15/2012] [Accepted: 12/18/2012] [Indexed: 01/27/2023]
|
26
|
Bol GM, Raman V, van der Groep P, Vermeulen JF, Patel AH, van der Wall E, van Diest PJ. Expression of the RNA helicase DDX3 and the hypoxia response in breast cancer. PLoS One 2013; 8:e63548. [PMID: 23696831 PMCID: PMC3656050 DOI: 10.1371/journal.pone.0063548] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Accepted: 04/03/2013] [Indexed: 12/11/2022] Open
Abstract
Aims DDX3 is an RNA helicase that has antiapoptotic properties, and promotes proliferation and transformation. In addition, DDX3 was shown to be a direct downstream target of HIF-1α (the master regulatory of the hypoxia response) in breast cancer cell lines. However, the relation between DDX3 and hypoxia has not been addressed in human tumors. In this paper, we studied the relation between DDX3 and the hypoxic responsive proteins in human breast cancer. Methods and Results DDX3 expression was investigated by immunohistochemistry in breast cancer in comparison with hypoxia related proteins HIF-1α, GLUT1, CAIX, EGFR, HER2, Akt1, FOXO4, p53, ERα, COMMD1, FER kinase, PIN1, E-cadherin, p21, p27, Transferrin receptor, FOXO3A, c-Met and Notch1. DDX3 was overexpressed in 127 of 366 breast cancer patients, and was correlated with overexpression of HIF-1α and its downstream genes CAIX and GLUT1. Moreover, DDX3 expression correlated with hypoxia-related proteins EGFR, HER2, FOXO4, ERα and c-Met in a HIF-1α dependent fashion, and with COMMD1, FER kinase, Akt1, E-cadherin, TfR and FOXO3A independent of HIF-1α. Conclusions In invasive breast cancer, expression of DDX3 was correlated with overexpression of HIF-1α and many other hypoxia related proteins, pointing to a distinct role for DDX3 under hypoxic conditions and supporting the oncogenic role of DDX3 which could have clinical implication for current development of DDX3 inhibitors.
Collapse
Affiliation(s)
- Guus M. Bol
- Departments of Pathology, University Medical Center Utrecht Cancer Center, Utrecht, The Netherlands
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Venu Raman
- Departments of Pathology, University Medical Center Utrecht Cancer Center, Utrecht, The Netherlands
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Petra van der Groep
- Departments of Pathology, University Medical Center Utrecht Cancer Center, Utrecht, The Netherlands
- Division of Internal Medicine and Dermatology, University Medical Center Utrecht Cancer Center, Utrecht, The Netherlands
| | - Jeroen F. Vermeulen
- Departments of Pathology, University Medical Center Utrecht Cancer Center, Utrecht, The Netherlands
| | - Arvind H. Patel
- MRC, University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Elsken van der Wall
- Division of Internal Medicine and Dermatology, University Medical Center Utrecht Cancer Center, Utrecht, The Netherlands
| | - Paul J. van Diest
- Departments of Pathology, University Medical Center Utrecht Cancer Center, Utrecht, The Netherlands
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
27
|
van der Groep P, van Diest PJ, Smolders YHCM, Ausems MGEM, van der Luijt RB, Menko FH, Bart J, de Vries EGE, van der Wall E. HIF-1α overexpression in ductal carcinoma in situ of the breast in BRCA1 and BRCA2 mutation carriers. PLoS One 2013; 8:e56055. [PMID: 23409121 PMCID: PMC3568038 DOI: 10.1371/journal.pone.0056055] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Accepted: 01/05/2013] [Indexed: 11/18/2022] Open
Abstract
Recent studies have revealed that BRCA1 and BRCA2 germline mutation-related breast cancers show frequent overexpression of hypoxia inducible factor-1α (HIF-1α), the key regulator of the hypoxia response. However, the question remained whether hypoxia is a late stage bystander or a true carcinogenetic event in patients with hereditary predisposition. We therefore studied HIF-1α overexpression in ductal carcinoma in situ (DCIS), an established precursor of invasive breast cancer. We used immunohistochemistry to examine the expression of the hypoxia markers HIF-1α, CAIX and Glut-1 in DCIS and available invasive carcinoma lesions of 32 BRCA1, 16 BRCA2 and 77 non-BRCA mutation-related cases. HIF-1α expression was detected in 63% of BRCA1 and 62% of BRCA2 as compared to 34% of non-BRCA mutation-related DCIS cases (p = 0.005). CAIX overexpression was present in 56% of BRCA1 and 44% of BRCA2 as compared to 6% of non-BRCA mutation-related DCIS cases (p = 0.000). Glut-1 overexpression was observed in 59% of BRCA1, 75% of BRCA2 and 67% of non-BRCA mutation-related DCIS cases (p = 0.527). Overall, HIF-1α, CAIX and Glut-1 expression in BRCA mutation-related DCIS matched the expression in the accompanying invasive cancers in 60% or more of cases. In non-BRCA mutation-related cases the expression of the hypoxia markers in DCIS matched the expression in the invasive part in 46% or more of the cases. Although BRCA1 and BRCA2 germline mutation-related invasive breast cancers are different in many ways, the hypoxia-related proteins HIF-1α, CAIX and Glut-1 are expressed in both DCIS and invasive lesions of BRCA1 and BRCA2 mutation carriers. This suggests that hypoxia may already play a role in the DCIS stage of BRCA1 and BRCA2 germline mutation related breast carcinogenesis, and may also drive cancer progression. Hypoxia-related proteins are therefore putative targets for therapy and molecular imaging for early detection and monitoring therapy response in BRCA mutation patients.
Collapse
Affiliation(s)
- Petra van der Groep
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands.
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Harrison H, Rogerson L, Gregson HJ, Brennan KR, Clarke RB, Landberg G. Contrasting Hypoxic Effects on Breast Cancer Stem Cell Hierarchy Is Dependent on ER-α Status. Cancer Res 2012; 73:1420-33. [DOI: 10.1158/0008-5472.can-12-2505] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
29
|
Dobrzyńska I, Skrzydlewska E, Figaszewski ZA. Changes in electric properties of human breast cancer cells. J Membr Biol 2012; 246:161-6. [PMID: 23135059 PMCID: PMC3555248 DOI: 10.1007/s00232-012-9516-5] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2012] [Accepted: 10/15/2012] [Indexed: 12/01/2022]
Abstract
Studies of the electrical surface properties of biological cells have provided fundamental knowledge about the cell surface. The change in biological functions of cells may affect the surface properties and can be detected by electrokinetic measurements. The surface density of fibroblasts and breast cancer cells (MDA-MB-231 and MCF-7) as a function of pH was measured by electrophoresis. The interaction between solution ions and the breast cancer cell or fibroblast surface was described by a four-component equilibrium model. The agreement between the experimental and theoretical charge variation curves of the breast cancer cells and fibroblasts was good at pH 2.5–9. The extent of fibroblast and breast cancer cell lipid peroxidation was estimated by HPLC measurement of the malondialdehyde level. The acid (CTA) and basic (CTB) functional group concentrations and the average association constant with hydroxyl (KBOH) ions values of the breast cancer cell membranes were higher than in normal cells, while the average association constant with hydrogen (KAH) value was smaller. The level of lipid peroxidation products was higher in breast cancer cells than in normal cells.
Collapse
Affiliation(s)
- Izabela Dobrzyńska
- Institute of Chemistry, University in Białystok, Al. Piłsudskiego 11/4, 15-443 Białystok, Poland.
| | | | | |
Collapse
|
30
|
Generali D, Symmans WF, Berruti A, Fox SB. Predictive immunohistochemical biomarkers in the context of neoadjuvant therapy for breast cancer. J Natl Cancer Inst Monogr 2012; 2011:99-102. [PMID: 22043052 DOI: 10.1093/jncimonographs/lgr030] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The adoption of personalized medicine has led to the search for prognostic and predictive markers that can be applied to individual patients to give optimal information for their clinical management. We have used samples from randomized clinical trials of hormonal and chemotherapy to identify relevant markers of sensitivity and resistance using a neoadjuvant approach by linking expression of a panel of proteins involved in growth factor receptor signaling, angiogenesis, estrogen receptor signaling, and hypoxia to individual patient response. We evaluated samples from randomized clinical trials of epirubicin with or without tamoxifen, and letrozole with or without metronomic cyclophosphamide, to study chemotherapy, hormonal therapy, and antiangiogenic effects. We present a proof of principle of this approach in identifying several key pathways that are associated with clinical and pathological response. Thus, we have shown that the hypoxia-inducible factor (HIF) pathway, mitogen activated protein kinase, and phosphorylated estrogen receptor-α can identify patients who are likely to respond to hormonal therapy and that HIF signaling is also a marker of resistance for anthracycline-based chemotherapy. To redress the role of HIF, we then evaluated samples from a randomized control trial of an anthracycline chemotherapy with and without erythropoietin. These studies demonstrate that the approach of using primary systemic therapy in breast can identify markers of response and potentially targets for rationale design of new therapies.
Collapse
Affiliation(s)
- Daniele Generali
- Unità di Patologia Mammaria-Breast Cancer Unit, Azienda Instituti Ospitalieri di Cremona, Cremona, Italy
| | | | | | | |
Collapse
|
31
|
Wan CF, Du J, Fang H, Li FH, Zhu JS, Liu Q. Enhancement patterns and parameters of breast cancers at contrast-enhanced US: correlation with prognostic factors. Radiology 2012; 262:450-9. [PMID: 22282183 DOI: 10.1148/radiol.11110789] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE To investigate the correlation between enhancement patterns and parameters of contrast material-enhanced ultrasonography (US) with prognostic factors in breast cancers. MATERIALS AND METHODS The study was approved by the institutional ethics committee, and written informed consent was obtained. Surgical resection specimens of 74 malignant breast lesions in 74 women (mean age, 55 years; age range, 32-78 years) who had undergone contrast-enhanced US were included. Different contrast enhancement patterns (enhancement degree, order, and margin; internal homogeneity; perfusion defect; and radial or penetrating vessels) and parameters (wash-in time, peak intensity, time to peak, area under the time-intensity curve, ascending slope, and descending slope) were evaluated. Pathologic prognostic factors, including histologic grade, lymph node status, tumor diameter, microvessel density (MVD), estrogen and progesterone receptor status, and c-erb-B2, p53, and Ki-67 expression were determined. Correlation of enhancement patterns and parameters with prognostic factors was analyzed with the Pearson χ2 test, Spearman rank correlation test, and logistic regression analysis. RESULTS Some enhancement features were associated, albeit not significantly, with prognostic factors. Perfusion defect was the most accurate enhancement criterion for higher histologic grade (grade III) (P=.016), negative estrogen receptor expression (P=.006), positive c-erb-B2 expression (P=.013), larger tumor diameter (≥2 cm) (P=.016), and increased MVD (P=.019). Radial or penetrating vessels were associated with lymph node status (P=.010). Hyperenhancement may be useful in reflecting increased MVD (P=.008) and positive p53 expression (P=.037). For contrast enhancement parameters, ascending slope was the best discrimination criterion for proliferative activity (P=.003). CONCLUSION Enhancement patterns and parameters of contrast-enhanced US may be useful in the noninvasive prediction of prognostic factors of breast cancers.
Collapse
Affiliation(s)
- Cai Feng Wan
- Department of Ultrasound, Renji Hospital, School of Medicine, Shanghai Jiaotong University, 1630 Dongfang Rd, Shanghai 200127, China
| | | | | | | | | | | |
Collapse
|
32
|
Doublier S, Belisario DC, Polimeni M, Annaratone L, Riganti C, Allia E, Ghigo D, Bosia A, Sapino A. HIF-1 activation induces doxorubicin resistance in MCF7 3-D spheroids via P-glycoprotein expression: a potential model of the chemo-resistance of invasive micropapillary carcinoma of the breast. BMC Cancer 2012; 12:4. [PMID: 22217342 PMCID: PMC3262753 DOI: 10.1186/1471-2407-12-4] [Citation(s) in RCA: 135] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2011] [Accepted: 01/04/2012] [Indexed: 11/30/2022] Open
Abstract
Background Invasive micropapillary carcinoma (IMPC) of the breast is a distinct and aggressive variant of luminal type B breast cancer that does not respond to neoadjuvant chemotherapy. It is characterized by small pseudopapillary clusters of cancer cells with inverted cell polarity. To investigate whether hypoxia-inducible factor-1 (HIF-1) activation may be related to the drug resistance described in this tumor, we used MCF7 cancer cells cultured as 3-D spheroids, which morphologically simulate IMPC cell clusters. Methods HIF-1 activation was measured by EMSA and ELISA in MCF7 3-D spheroids and MCF7 monolayers. Binding of HIF-1α to MDR-1 gene promoter and modulation of P-glycoprotein (Pgp) expression was evaluated by ChIP assay and FACS analysis, respectively. Intracellular doxorubicin retention was measured by spectrofluorimetric assay and drug cytotoxicity by annexin V-FITC measurement and caspase activity assay. Results In MCF7 3-D spheroids HIF-1 was activated and recruited to participate to the transcriptional activity of MDR-1 gene, coding for Pgp. In addition, Pgp expression on the surface of cells obtained from 3-D spheroids was increased. MCF7 3-D spheroids accumulate less doxorubicin and are less sensitive to its cytotoxic effects than MCF7 cells cultured as monolayer. Finally, HIF-1α inhibition either by incubating cells with 3-(5'-hydroxymethyl-2'-furyl)-1-benzylindazole (a widely used HIF-1α inhibitor) or by transfecting cells with specific siRNA for HIF-1α significantly decreased the expression of Pgp on the surface of cells and increased the intracellular doxorubicin accumulation in MCF7 3-D spheroids. Conclusions MCF7 breast cancer cells cultured as 3-D spheroids are resistant to doxorubicin and this resistance is associated with an increased Pgp expression in the plasma membrane via activation of HIF-1. The same mechanism may be suggested for IMPC drug resistance.
Collapse
Affiliation(s)
- Sophie Doublier
- Department of Genetics, Biology and Biochemistry, University of Turin, Via Santena, 5/bis, 10126 Turin, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Choi JY, Jang YS, Min SY, Song JY. Overexpression of MMP-9 and HIF-1α in Breast Cancer Cells under Hypoxic Conditions. J Breast Cancer 2011; 14:88-95. [PMID: 21847402 PMCID: PMC3148536 DOI: 10.4048/jbc.2011.14.2.88] [Citation(s) in RCA: 104] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2010] [Accepted: 02/24/2011] [Indexed: 12/27/2022] Open
Abstract
Purpose Hypoxia, which is a loss of oxygen in tissues, is a common condition in solid tumors due to the tumor outgrowing existing vasculature. Under hypoxic conditions, hypoxia-inducible factor (HIF)-1α rapidly accumulates and transactivates hundreds of genes, such as matrix metalloproteinases (MMPs). MMPs contribute to invasion and metastasis of tumor cells by degrading the surrounding basement membrane and extracellular matrix barriers, which enables the easy migration and spread of cancer cells. We examined whether hypoxia increases tumor cell invasion, and whether increased invasiveness was due to HIF-1α and MMP-9 expression. Methods Transwell invasion assays were performed to demonstrate whether hypoxia enhance tumor invasion by use of MDA-MB-231 breast cancer cells. An immunofluorescence assay was used to demonstrate expression of HIF-1α and MMP-9 under hypoxic conditions. Luciferase and ChiP assays were performed to demonstrate that MMP-9 promoter activity was regulated by HIF-1α. Results HIF-1α was stabilized under hypoxic conditions and stimulated MMP-9 expression, which affected the tumor invasiveness of breast cancer cells. HIF-1α transactivated the MMP-9 promoter by forming a transcriptional unit with p300, thus increasing expression of MMP-9 transcripts. Zymography indicated that MMP-9 had more gelatinase activity under hypoxic conditions than normoxic conditions. Furthermore, the small GTPase Ras was also activated in response to hypoxia, which then aids stabilization of HIF-1α, and in turn upregulates MMP-9 expression. We also demonstrate that MMP-9 is upregulated concurrently with HIF-1α in tumor tissues from patients with breast cancer. Conclusion These results suggest that HIF-1α promotes cell invasion through a MMP-9-dependent mechanism and that future antitumor agents could be used to target HIF-1α and MMP-9.
Collapse
|
34
|
Ryu K, Park C, Lee Y. Hypoxia-inducible factor 1 alpha represses the transcription of the estrogen receptor alpha gene in human breast cancer cells. Biochem Biophys Res Commun 2011; 407:831-6. [PMID: 21458421 DOI: 10.1016/j.bbrc.2011.03.119] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2011] [Accepted: 03/26/2011] [Indexed: 01/13/2023]
Abstract
The estrogen receptor is one of the most important transcription factors in breast tumor growth and development. We, and others, have previously shown that hypoxia induces rapid ERα protein degradation by a proteasome-mediated pathway in breast cancer cells, which is linked with ERα activation. However, no report has shown the effect of hypoxia on ESR1 gene regulation at the transcriptional level. In this report, we show that hypoxia repressed the expression of ERα mRNA in MCF-7 and T47D human breast cancer cells, but not in human endometrial Ishikawa cells, although ERα degradation under hypoxia was also observed in Ishikawa cells. This indicates that ESR1 transcriptional repression and ERα protein downregulation by hypoxia are regulated by distinct mechanisms. Repression of ESR1 gene transcription occurred at the transcriptional level as a result of decreased recruitment of RNA polymerase II at the proximal promoter of the ESR1 locus in response to stabilization of the HIF-1α protein under hypoxia. Our data show that hypoxia induces repression of the ESR1 gene, which may facilitate hormone insensitivity in the tumor microenvironment.
Collapse
Affiliation(s)
- Kwanghee Ryu
- College of Life Science, Institute of Biotechnology, Department of Bioscience and Biotechnology, Sejong University, Seoul 143-747, Republic of Korea
| | | | | |
Collapse
|
35
|
Prabhu V, Guruvayoorappan C. Prolyl hydroxylase and hypoxia inducible factor: potential targets for cancer therapy. Immunopharmacol Immunotoxicol 2011; 33:568-75. [DOI: 10.3109/08923973.2010.545418] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
36
|
Kanczuga-Koda L, Koda M, Tomaszewski J, Jarzabek K, Lotowska J, Baltaziak M, Sulkowska U, Sobaniec-Lotowska M, Sulkowski S. ERα and ERβ expression in correlation with Ki-67, Bcl-2 and Bak in primary tumors and lymph node metastases of breast cancer: The effect of pre-operative chemotherapy. Oncol Lett 2010; 1:1067-1071. [PMID: 22870114 DOI: 10.3892/ol.2010.178] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2010] [Accepted: 09/02/2010] [Indexed: 11/06/2022] Open
Abstract
This study aimed to assess the pre-operative chemotherapy impact on the relationship between estrogen receptor (ER) expression and markers of proliferation and apoptosis in primary and metastatic breast cancer. Immunohistochemical examinations were conducted on surgically removed ductal invasive breast cancers and their lymph node metastases in 135 patients. A total of 64 patients from this group underwent pre-operative chemotherapy and in 71 cases the surgery was performed without primary chemotherapy. A negative correlation between ERα and Ki-67 was found in primary tumors and lymph node metastases. A positive correlation was observed between ERα and Bcl-2. A positive correlation was also noted between ERβ and Bak, suggesting that the two ERs were involved in the regulation of proteins responsible for the control of the apoptotic process. Assessment of the expression of the proteins conducted separately in primary tumors and lymph node metastases did not reveal a significant effect of pre-operative chemotherapy on the correlations of ERs with Ki-67, Bcl-2 and Bak. However, the analysis of the correlations between the receptor expression in primary tumors and Ki-67, Bcl-2 and Bak in lymph node metastases showed a statistically significant impact of pre-operative chemotherapy on the correlations of ERα and Bcl-2 with ERβ and Bak, confirming involvement of the two ERs in the regulation of apoptosis during breast carcinogenesis.
Collapse
Affiliation(s)
- Luiza Kanczuga-Koda
- Department of Pathology, Maria Sklodowska-Curie Memorial Bialystok Oncology Center, 15-027 Bialystok
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Ogba N, Doughman YQ, Chaplin LJ, Hu Y, Gargesha M, Watanabe M, Montano MM. HEXIM1 modulates vascular endothelial growth factor expression and function in breast epithelial cells and mammary gland. Oncogene 2010; 29:3639-49. [PMID: 20453883 PMCID: PMC2892028 DOI: 10.1038/onc.2010.110] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Recently, we found that mutation of the C-terminus of transcription factor Hexamethylene bisacetamide inducible protein 1 (HEXIM1) in mice leads to abnormalities in cardiovascular development due to aberrant vascular endothelial growth factor (VEGF) expression. HEXIM1 regulation of some genes has also been shown to be positive transcription elongation factor b (P-TEFb)-dependent. However, it is not known whether HEXIM1 regulates VEGF in the mammary gland. We demonstrate that HEXIM1 regulates estrogen-induced VEGF transcription via inhibition of Estrogen Receptor alpha recruitment to the VEGF promoter in a P-TEFb-independent manner in MCF-7 cells. Under hypoxic conditions, HEXIM1 inhibits estrogen-induced Hypoxia-inducible factor-1 alpha (HIF-1α) protein expression and recruitment of HIF-1α to the hypoxia response element in the VEGF promoter. In the mouse mammary gland, increased HEXIM1 expression decreased estrogen-driven VEGF and HIF-1α expression. Conversely, a mutation in the C-terminus of HEXIM1 (HEXIM11-312) led to increased VEGF and HIF-1α expression and vascularization in mammary glands of heterozygous HEXIM11-312 mice when compared to their wild-type littermates. Additionally, HEXIM11-312 mice have a higher incidence of carcinogen-induced mammary tumors with increased vascularization, suggesting an inhibitory role for HEXIM1 during angiogenesis. Taken together, our data provide evidence to suggest a novel role for HEXIM1 in cancer progression.
Collapse
Affiliation(s)
- N Ogba
- Department of Pharmacology, Rainbow Babies and Children's Hospital, Cleveland, OH, USA
| | | | | | | | | | | | | |
Collapse
|
38
|
Ogane N, Yasuda M, Shimizu M, Miyazawa M, Kamoshida S, Ueda A, Takata K, Sakuma Y, Miyagi Y, Kameda Y. Clinicopathological implications of expressions of hypoxia-related molecules in esophageal superficial squamous cell carcinoma. Ann Diagn Pathol 2010; 14:23-9. [PMID: 20123453 DOI: 10.1016/j.anndiagpath.2009.10.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2009] [Revised: 09/25/2009] [Accepted: 10/13/2009] [Indexed: 11/25/2022]
Abstract
This study was conducted to clarify whether or not expressions of hypoxia-related molecules would have clinicopathological significance in squamous cell carcinoma (SCC) of the esophagus. Expressions of hypoxia inducible factor-1 alpha (HIF-1alpha), glucose transporter 1 (GLUT-1) and RAC-1 were immunohistochemically analyzed in 96 surgically resected SCCs at pT1b (sm1, 12 cases; sm2, 35 cases; sm3, 49 cases). They were divided into a lymph node metastasis (LNM)-positive group composed of 44 cases and an LNM-negative group composed of 52 cases. Immunohistochemical profiles were estimated based on the staining extent (score: 1+, 2+, 3+) and intensity (score: 1+, 2+, 3+). A significant expression pattern was found in the nucleus for HIF-1alpha, cell membrane for GLUT-1 and cytoplasm for RAC-1. The cases were categorized into a high score group (total score of 4 or more) and a low score group (total score of 3 or less) in each maker, respectively. A comparison made between the LNM-positive group and the LNM-negative group showed that the proportion of cases with a high score was larger in the LNM-positive group than in the LNM-negative group (HIF-1alpha, P = .02; GLUT-1, P = .008; RAC-1, P = .001). Among them, HIF-1alpha was found to be significantly related to the disease-free survival (P = .019) and overall survival (P = .034) as well as LNM (disease-free survival, P = .030; overall survival, P = .030). The multivariate analysis demonstrated that the HIF-1alpha expression would be an independent indicator for prognosis. In the superficial SCCs of the esophagus, GLUT-1 and RAC-1 may be involved in LNM, and HIF-1alpha overexpression is expected to predict an unfavorable clinical outcome.
Collapse
|
39
|
Lant B, Storey KB. An overview of stress response and hypometabolic strategies in Caenorhabditis elegans: conserved and contrasting signals with the mammalian system. Int J Biol Sci 2010; 6:9-50. [PMID: 20087441 PMCID: PMC2808051 DOI: 10.7150/ijbs.6.9] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2009] [Accepted: 11/25/2009] [Indexed: 12/21/2022] Open
Abstract
Studies of the molecular mechanisms that are involved in stress responses (environmental or physiological) have long been used to make links to disease states in humans. The nematode model organism, Caenorhabditis elegans, undergoes a state of hypometabolism called the 'dauer' stage. This period of developmental arrest is characterized by a significant reduction in metabolic rate, triggered by ambient temperature increase and restricted oxygen/ nutrients. C. elegans employs a number of signal transduction cascades in order to adapt to these unfavourable conditions and survive for long times with severely reduced energy production. The suppression of cellular metabolism, providing energetic homeostasis, is critical to the survival of nematodes through the dauer period. This transition displays molecular mechanisms that are fundamental to control of hypometabolism across the animal kingdom. In general, mammalian systems are highly inelastic to environmental stresses (such as extreme temperatures and low oxygen), however, there is a great deal of conservation between the signal transduction pathways of nematodes and mammals. Along with conserving many of the protein targets in the stress response, many of the critical regulatory mechanisms are maintained, and often differ only in their level of expression. Hence, the C. elegans model outlines a framework of critical molecular mechanisms that may be employed in the future as therapeutic targets for addressing disease states.
Collapse
Affiliation(s)
| | - Kenneth B. Storey
- Institute of Biochemistry, Carleton University, Ottawa, Ont., Canada
| |
Collapse
|
40
|
Abstract
In this review we summarize the evidence for a role for hypoxic response in the biology of metastasis, with a particular emphasis on the metastasis of breast cancer and the function of the hypoxia inducible factor (HIF).
Collapse
Affiliation(s)
- Helene Rundqvist
- Molecular Biology Section, Division of Biological Sciences, University of California, San Diego, CA 92093, USA
| | | |
Collapse
|
41
|
Miyazawa M, Yasuda M, Fujita M, Kajiwara H, Hirabayashi K, Takekoshi S, Hirasawa T, Murakami M, Ogane N, Kiguchi K, Ishiwata I, Mikami M, Osamura RY. Therapeutic strategy targeting the mTOR-HIF-1α-VEGF pathway in ovarian clear cell adenocarcinoma. Pathol Int 2009; 59:19-27. [DOI: 10.1111/j.1440-1827.2008.02320.x] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
42
|
Thornburg JM, Nelson KK, Clem BF, Lane AN, Arumugam S, Simmons A, Eaton JW, Telang S, Chesney J. Targeting aspartate aminotransferase in breast cancer. Breast Cancer Res 2008; 10:R84. [PMID: 18922152 PMCID: PMC2614520 DOI: 10.1186/bcr2154] [Citation(s) in RCA: 219] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2008] [Revised: 09/19/2008] [Accepted: 10/15/2008] [Indexed: 01/15/2023] Open
Abstract
Introduction Glycolysis is increased in breast adenocarcinoma cells relative to adjacent normal cells in order to produce the ATP and anabolic precursors required for survival, growth and invasion. Glycolysis also serves as a key source of the reduced form of cytoplasmic nicotinamide adenine dinucleotide (NADH) necessary for the shuttling of electrons into mitochondria for electron transport. Lactate dehydrogenase (LDH) regulates glycolytic flux by converting pyruvate to lactate and has been found to be highly expressed in breast tumours. Aspartate aminotransferase (AAT) functions in tandem with malate dehydrogenase to transfer electrons from NADH across the inner mitochondrial membrane. Oxamate is an inhibitor of both LDH and AAT, and we hypothesised that oxamate may disrupt the metabolism and growth of breast adenocarcinoma cells. Methods We examined the effects of oxamate and the AAT inhibitor amino oxyacetate (AOA) on 13C-glucose utilisation, oxygen consumption, NADH and ATP in MDA-MB-231 cells. We then determined the effects of oxamate and AOA on normal human mammary epithelial cells and MDA-MB-231 breast adenocarcinoma cell proliferation, and on the growth of MDA-MB-231 cells as tumours in athymic BALB/c female mice. We ectopically expressed AAT in MDA-MB-231 cells and examined the consequences on the cytostatic effects of oxamate. Finally, we examined the effect of AAT-specific siRNA transfection on MDA-MB-231 cell proliferation. Results We found that oxamate did not attenuate cellular lactate production as predicted by its LDH inhibitory activity, but did have an anti-metabolic effect that was similar to AAT inhibition with AOA. Specifically, we found that oxamate and AOA decreased the flux of 13C-glucose-derived carbons into glutamate and uridine, both products of the mitochondrial tricarboxylic acid cycle, as well as oxygen consumption, a measure of electron transport chain activity. Oxamate and AOA also selectively suppressed the proliferation of MDA-MB-231 cells relative to normal human mammary epithelial cells and decreased the growth of MDA-MB-231 breast tumours in athymic mice. Importantly, we found that ectopic expression of AAT in MDA-MB-231 cells conferred resistance to the anti-proliferative effects of oxamate and that siRNA silencing of AAT decreased MDA-MB-231 cell proliferation. Conclusions We conclude that AAT may be a valid molecular target for the development of anti-neoplastic agents.
Collapse
|
43
|
Mizobuchi H, García-Castellano JM, Philip S, Healey JH, Gorlick R. Hypoxia markers in human osteosarcoma: an exploratory study. Clin Orthop Relat Res 2008; 466:2052-9. [PMID: 18528739 PMCID: PMC2493019 DOI: 10.1007/s11999-008-0328-y] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2007] [Accepted: 05/16/2008] [Indexed: 01/31/2023]
Abstract
Neoplastic cells growing under hypoxic conditions exhibit a more aggressive phenotype by activating a cascade of molecular events partly mediated by hypoxia-inducible transcription factor (HIF-1alpha) and vascular endothelial growth factor (VEGF). The roles of these markers have been studied previously in several cancer lines. We ascertained the frequency of HIF-1alpha expression, VEGF expression, the degree of neovascularization, and cell proliferation in osteosarcoma samples. Samples from osteosarcoma patients were assessed for HIF-1alpha and VEGF protein expression using immunohistochemistry, neovascularization using antibodies for Factor VIII, and cell proliferation using the Ki-67 labeling index. Associations between these parameters and clinical features were examined. HIF-1alpha staining was positive in 35% of patients and metastases were present in 61% of these HIF-1alpha-positive patients. VEGF protein expression was detected in 69% of patients, 92% of whom were female. We observed an insignificant trend for a higher frequency of VEGF expression in the high-grade as compared to low-grade osteosarcoma. We observed no association between vascular density and proliferation index and any clinical parameters. We found an association between HIF-1alpha expression and metastatic disease and between VEGF expression and female gender.
Collapse
Affiliation(s)
- Hiroo Mizobuchi
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY USA
| | | | - Shaji Philip
- Division of Hematology/Oncology, Department of Pediatrics, The Children’s Hospital at Montefiore, 3415 Bainbridge Avenue, Rosenthal 3rd Floor, Bronx, NY 10467 USA
| | - John H. Healey
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY USA
| | - Richard Gorlick
- Division of Hematology/Oncology, Department of Pediatrics, The Children’s Hospital at Montefiore, 3415 Bainbridge Avenue, Rosenthal 3rd Floor, Bronx, NY 10467 USA
| |
Collapse
|
44
|
Jiang YG, Luo Y, He DL, Li X, Zhang LL, Peng T, Li MC, Lin YH. Role of Wnt/beta-catenin signaling pathway in epithelial-mesenchymal transition of human prostate cancer induced by hypoxia-inducible factor-1alpha. Int J Urol 2008; 14:1034-9. [PMID: 17956532 DOI: 10.1111/j.1442-2042.2007.01866.x] [Citation(s) in RCA: 170] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
OBJECTIVES Epithelial-mesenchymal transition (EMT) is an important process in tumor development, and several studies suggest that the Wnt/beta-catenin signal pathway may play an important role in EMT. However, there is no direct evidence showing that the Wnt/beta-catenin pathway actually determines the EMT induced by an exogenous signal. Our previous research has successfully proved that overexpression of hypoxia-inducible factor-1alpha (HIF-1alpha) could induce EMT in LNCaP cells, but not in PC-3. The present study aims to determine whether the signal of HIF-1alpha for inducing prostate cancer cells to undergo EMT might possibly pass through the Wnt/beta-catenin pathway. METHODS Epithelial-mesenchymal transition associated proteins were detected in several human prostate carcinoma cell lines by Western blot, and then we distinguished the EMT positive cell lines from the EMT negative cell lines. Furthermore, we evaluated the possible correlation between potency of invasiveness and proliferation among these cell lines with different characteristics of EMT using Matrigel transwell and thiazolyl blue tetrazolium bromide (MTT) assays. Finally, the different expression of some critical proteins and genes in Wnt/beta-catenin signaling pathway were analyzed by Western blot and reverse transcription-polymerase chain reaction (RT-PCR) in these cells with different characteristics of EMT. RESULTS Among several prostate cancer cell lines, PC-3, LNCaP and PC-3/HIF-1alpha are EMT negative cell lines, whereas LNCaP/HIF-1alpha and IA8 have undergone the EMT process. EMT positive cells (LNCaP/HIF-1alpha and IA8) exhibit much stronger potency of invasiveness and proliferation than those of PC-3 and LNCaP, which belong to EMT negative cells. Interestingly, although PC-3/HIF-1alpha had not completed the EMT process, it still displayed stronger potency of invasion and proliferation, resembling EMT positive cells. The protein expression level of total glycogensynthase kinase 3beta (GSK-3beta) and phospho-GSK-3beta in LNCaP/HIF-1alpha, IA8 and PC-3/HIF-1alpha cells significantly decreased; however, the relative ratios of p-GSK3beta/t-GSK3beta in LNCaP/HIF-1alpha, IA8 and PC-3/HIF-1alpha cells were significantly higher than PC-3 and LNCaP. Consistently, beta-catenin protein expression increased in LNCaP/HIF-1alpha and IA8 cells, but not in PC-3/HIF-1alpha; RT-PCR confirmed these results, except for the enhanced transcription activity of beta-catenin mRNA in PC-3/HIF-1alpha. CONCLUSION Our data suggests that activation of the Wnt/beta-catenin signaling pathway correlates with the characteristic of EMT and potency of invasiveness and proliferation. This may be the critical factor that directly controls the process of EMT induced by HIF-1alpha in prostate cancer cells.
Collapse
Affiliation(s)
- Yong-Guang Jiang
- Department of Urology, Affiliated Beijing Anzhen Hospital of Capital Medical University, Anzhenli Street, Chaoyang District, Beijing, China
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Tao J, Liu YQ, Li Y, Peng JL, Li L, Liu J, Shen X, Shen GX, Tu YT. Hypoxia: dual effect on the expression of transferrin receptor in human melanoma A375 cell line. Exp Dermatol 2008; 16:899-904. [PMID: 17927572 DOI: 10.1111/j.1600-0625.2007.00601.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND Over-expression of transferrin receptor (TfR) is a common feature of human malignancies. Therapeutic strategies designed to interfere with tumor iron metabolism have targeted TfR. In previous studies, our laboratory successfully constructed the human-mouse chimeric antibody against TfR and it displayed a tumor-specificity distribution, and has a strong anti-tumor effect. We also found that there were still some limitations to anti-tumor effect in vivo. Oxygen and iron have a very tight relationship, and hypoxia is considered a fundamentally important characteristic of the tumor microenvironment. To exploit the target molecule TfR more rationally and effectively, we were prompted to explore TfR expression under hypoxia. OBJECTIVE To examine the expressing alteration of TfR of human melanoma A375 cell line under hypoxia at various time points (0, 12, 24, 36, 48 and 60 h). DESIGN The expressing alteration of TfR of A375 cell line under hypoxia at various time points (0, 12, 24, 36, 48 and 60 h) was assayed by flow cytometry, real-time RT-PCR and Western blot. RESULTS Hypoxia has dual effect on the expression of TfR in human melanoma A375 cell line. CONCLUSIONS These findings may have important implications for more rational, individualized gene-based therapy using TfR as target receptor in melanoma.
Collapse
Affiliation(s)
- Juan Tao
- Department of Dermatology, Affiliated Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Rare variant of hypoxia-inducible factor-1alpha (HIF-1A) and breast cancer risk in Korean women. Clin Chim Acta 2007; 389:167-70. [PMID: 18160046 DOI: 10.1016/j.cca.2007.12.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2007] [Revised: 12/04/2007] [Accepted: 12/04/2007] [Indexed: 11/20/2022]
Abstract
BACKGROUND Hypoxia inducible factor 1 alpha (HIF-1A) is activated by low oxygen condition tension, a key regulator of the gene involved in the cellular response to hypoxia. Tumors exhibiting extensive hypoxia are more aggressive than tumors oxygenized better. METHODS To evaluate the potential role of the polymorphisms of HIF-1A in the etiology of breast cancer, histologically confirmed incident breast cancer cases (n=1599) and control subjects (n=1536) were recruited. RESULTS Two selected SNPs (Ex15+197C>T and P582S) were not associated with overall breast cancer risk (TT vs. CC: OR=0.9, 95% CI=0.6-1.5, Ser/Ser vs. Pro/Pro: OR=5.5, 95% CI=0.7-45.4, respectively). However, when stratified analyses were performed, significant associations were observed between Ser/Ser genotype at codon 582 and breast cancer risk among women with larger tumor size (>2 cm) (OR=10.1, 95% CI=1.1-91.1) or without lymph node involvement (OR=9.3, 95% CI=1.1-79.4), although confidence intervals were wide. CONCLUSIONS Our findings support the hypothesis that the HIF-1Alpha P582S variant may confer susceptibility to subgroups of breast cancer in Korean women. However, further study is warranted due to low statistical power caused by very low minor allele frequencies.
Collapse
|
47
|
High frequency of HIF-1alpha overexpression in BRCA1 related breast cancer. Breast Cancer Res Treat 2007; 111:475-80. [PMID: 18030615 DOI: 10.1007/s10549-007-9817-z] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2007] [Accepted: 11/08/2007] [Indexed: 10/22/2022]
Abstract
Hypoxia is a hallmark of cancer. Hypoxia inducible factor-1alpha (HIF-1alpha) is the key regulator of the hypoxia response. HIF-1alpha is overexpressed during sporadic breast carcinogenesis and correlated with poor prognosis. Little is known on the role of HIF-1alpha in hereditary breast carcinogenesis. A recent study suggests a role for BRCA1 in HIF-1alpha regulation. We therefore examined the expression of HIF-1alpha in BRCA1 related breast cancers. By immunohistochemistry we studied expression of HIF-1alpha and some of its downstream targets in 30 hereditary invasive breast cancers in comparison with 200 sporadic controls. HIF-1alpha overexpression was significantly more frequent in BRCA1 related breast cancers (27/30, 90%) than in sporadic controls (88/200, 44%) (P < 0.0001). 19/30 (63%) of BRCA1 tumors showed perinecrotic (hypoxia induced) and 8/30 (27%) a diffuse HIF-1alpha overexpression pattern, the latter more likely related to genetic alterations in oncogenes and tumor suppressor genes. In contrast, sporadic breast cancer HIF-1 expressing tumors showed an inverse ratio of perinecrotic/diffuse expression (31 vs. 69%, P = 0.0002). Glut-1 and CAIX, downstream HIF1 targets, were expressed in 27/30 (90%) and 15/21 (71%) of hereditary cases versus 54/183 (29%) and 24/183 (13%) in sporadic cases. p300 levels, necessary for HIF-1 downstream activation, were significantly higher in hereditary cases (20/21, 95%) compared to sporadic cases (91/183, 50%, P = 0.0001). In conclusion, in BRCA1 germline mutation related breast cancer, functional HIF-1alpha overexpression is seen at a much higher frequency than in sporadic breast cancer, mostly hypoxia induced. This points to an important role of hypoxia and its key regulator HIF-1alpha in hereditary breast carcinogenesis.
Collapse
|
48
|
Yamamoto Y, Ibusuki M, Okumura Y, Kawasoe T, Kai K, Iyama K, Iwase H. Hypoxia-inducible factor 1α is closely linked to an aggressive phenotype in breast cancer. Breast Cancer Res Treat 2007; 110:465-75. [PMID: 17805961 DOI: 10.1007/s10549-007-9742-1] [Citation(s) in RCA: 118] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2007] [Accepted: 08/21/2007] [Indexed: 01/02/2023]
Abstract
PURPOSE The aim of this study is to clarify characteristics of invasive breast cancer with expression of Hypoxia-induced factor 1alpha (HIF-1alpha) which is induced by hypoxia and signal transduction of growth factors. EXPERIMENTAL DESIGN We examined, by immunohistochemical analysis, the expression of HIF-1alpha in normal breast tissue, benign disorders and breast cancer. In invasive breast cancer, we investigated the correlation between expression of HIF-1alpha and clinicopathological and biological factors. We also studied the prognostic value of HIF-1alpha in breast cancer. RESULTS HIF-1alpha was mainly detected in tumor cell nuclei. In the 171 cases of invasive breast cancer examined, nuclear HIF-1alpha expression was detected in 63 (36.8%) cases. Immunoreactive nuclear HIF-1alpha was correlated with tumor size (p = 0.0013), lymph node metastasis (p = 0.0005), tumor stage (p = 0.0031) and histological grade (p = 0.0074). Elevated HIF-1alpha levels was also associated with hormone receptor negativity (p = 0.0025), HER2 overexpression (p = 0.0053), high Ki67 labeling index (p = 0.0002), increased levels of VEGF (p < 0.0001), COX-2 overexpression (p = 0.0029) and increased nuclear p53 (p = 0.0048). In terms of the possible use of HIF-1alpha as a prognostic indicator, patients who had increased HIF-1alpha levels in their tumor showed shorter disease-free survival (DFS) (p < 0.0001) and overall survival (OS) (p = 0.0002) than those lacking HIF-1alpha in univariate analysis. In multivariate analysis of DFS and OS, HIF-1alpha was identified an independent prognostic factor. CONCLUSIONS These findings suggest that HIF-1alpha was closely linked to an aggressive phenotype in breast cancer. It may be useful to study the expression of HIF-1alpha using immunohistochemical analysis for better understanding of the tumor characteristics of breast cancer.
Collapse
Affiliation(s)
- Yutaka Yamamoto
- Department of Breast & Endocrine Surgery, Faculty of Medical and Pharmaceutical Sciences, Kumamoto University Graduate School of Medical Sciences, 1-1-1 Honjo, Kumamoto 860-8556 Kumamoto, Japan.
| | | | | | | | | | | | | |
Collapse
|
49
|
Luo Y, He DL, Ning L, Shen SL, Li L, Li X, Zhau HE, Chung LWK. Over-expression of hypoxia-inducible factor-1? increases the invasive potency of LNCaP cells in vitro. BJU Int 2006; 98:1315-9. [PMID: 16956360 DOI: 10.1111/j.1464-410x.2006.06480.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To evaluate the effect of hypoxia-inducible factor-1alpha (HIF-1alpha) over-expression on the invasion-associated proteins in human prostate cancer cells, as HIF-1alpha is a transcriptional factor that could activate genes involved in the response to hypoxia, but might also enhance the invasive potency of prostate cancer cells. MATERIALS AND METHODS Prostate cancer (LNCaP) cells were transfected with recombinant plasmid pcDNA3.1(-)/HIF-1alpha and pcDNA3.1(-) control vector using a commercial system, designated as LNCaP/HIF-1alpha and LNCaP/pcDNA3.1, respectively. The positive clones were selected with G418 and confirmed by Western blot and indirect immunofluorescence labelling. A polycarbonate filter, coated with a matrix gel, was used to analyse the invasive potency. The expression of E-cadherin, vimentin, cathepsin D, matrix metalloproteinase (MMP2) and urokinase plasminogen activator receptor (uPAR) was detected by Western blot. RESULTS The expression level of HIF-1alpha in LNCaP/HIF-1alpha was distinctly higher than that in LNCaP/pcDNA3.1 and LNCaP. Many more cells of LNCaP/HIF-1alpha penetrated through the polycarbonate filter than those of LNCaP/pcDNA3.1 and LNCaP. Compared with the LNCaP/pcDNA3.1 and LNCaP cells, the expression of vimentin, cathepsin D, MMP-2 and uPAR were up-regulated in LNCaP/HIF-1alpha, whereas the expression of E-cadherin was down-regulated. CONCLUSION These results show that over-expression of HIF-1alpha directly stimulates the invasive potency of human prostate carcinoma LNCaP cells through the matrix gel. The expression of E-cadherin, vimentin, cathepsin D, MMP-2 and uPAR, which are proteins with established roles in the pathophysiology of invasion, could be regulated by HIF-1alpha in human prostate cancer cells.
Collapse
Affiliation(s)
- Yong Luo
- Department of Urology, First Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Ministry of Education, Xi'an, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Atkin GK, Daley FM, Bourne S, Glynne-Jones R, Northover JMA, Wilson GD. The impact of surgically induced ischaemia on protein levels in patients undergoing rectal cancer surgery. Br J Cancer 2006; 95:928-33. [PMID: 17016487 PMCID: PMC2360543 DOI: 10.1038/sj.bjc.6603362] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The goal of targeted therapy has driven a search for markers of prognosis and response to adjuvant therapy. The surgical resection of a solid tumour induces tissue ischaemia and acidosis, both potent mediators of gene expression. This study investigated the impact of colorectal cancer (CRC) surgery on prognostic and predictive marker levels. Tumour expression of thymidylate synthase, thymidine phosphorylase, cyclin A, vascular endothelial growth factor (VEGF), carbonic anhydrase-9, hypoxia inducible factor-1α, and glucose transporter-1 (GLUT-1) proteins was determined before and after rectal cancer surgery. Spectral imaging of tissue sections stained by immunohistochemistry provided quantitative data. Surgery altered thymidylate synthase protein expression (P=0.02), and this correlated with the change in the proliferation marker cyclin A. The expression of hypoxia inducible factor-1α, VEGF, and GLUT-1 proteins was also different following surgery. Colorectal cancer surgery significantly impacts on intratumoral gene expression, suggesting archival specimens may not accurately reflect in situ marker levels. Although rectal cancer was the studied model, the results may be applicable to any solid tumour undergoing extirpation in which molecular markers have been proposed to guide patient therapy.
Collapse
Affiliation(s)
- G K Atkin
- Gray Cancer Institute, Mount Vernon Hospital, Northwood, Middlesex HA6 2RN, UK, and Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201-2013, USA.
| | | | | | | | | | | |
Collapse
|