1
|
Wang M, Wu Y, Li X, Dai M, Li S. IGJ suppresses breast cancer growth and metastasis by inhibiting EMT via the NF‑κB signaling pathway. Int J Oncol 2023; 63:105. [PMID: 37539706 PMCID: PMC10552693 DOI: 10.3892/ijo.2023.5553] [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: 04/04/2023] [Accepted: 07/06/2023] [Indexed: 08/05/2023] Open
Abstract
Breast cancer metastasis is the primary cause of mortality of patients with breast cancer. The present study aimed to explore the role and underlying mechanisms of IGJ in the invasion and metastasis of breast cancer. The Cancer Genome Atlas database was utilized to analyze the differential gene expression profiles in patients with breast cancer with or without metastasis; the target gene, joining chain of multimeric IgA and IgM (JCHAIN, also known as IGJ, as referred to herein), with significant expression and with prognostic value was screened. The expression levels of IGJ in human breast cancer paired tissues and cell lines were detected using reverse transcription‑quantitative PCR and western blot analysis. IGJ differential expression was detected in paired human breast cancer tissues using immunohistochemistry. The role of IGJ in breast cancer was verified using CCK‑8, invasion and migration assays, and scratch tests in vivo and in vitro. Further exploration of the role and mechanism of IGJ in breast cancer was conducted through Gene Set Enrichment Analysis, Kyoto Encyclopedia of Genes and Genomes analysis, western blot analysis and immunofluorescence experiments. Through the analysis of gene expression profiles, it was found that IGJ was poorly expressed in patients with breast cancer with metastasis compared to patients with non‑metastatic breast cancer. The overexpression of IGJ was associated with an improved distant metastasis‑free survival and overall survival (OS). COX multivariate regression analysis demonstrated that IGJ was an independent prognostic factor for the OS and relapse‑free survival of patients with breast cancer. In comparison to healthy breast cancer adjacent tissues and cell lines, IGJ was poorly expressed in breast cancer tissues and cell lines (P<0.05). Further analyses indicated that the overexpression of IGJ suppressed the proliferation, invasion and metastasis of breast cancer cells in vivo and in vitro by inhibiting the occurrence of epithelial‑to‑mesenchymal transition (EMT) and suppressing the nuclear translocation of p65. Finally, rescue experiments indicated that IGJ restricted the proliferation and metastasis of breast cancer cells by regulating the NF‑κB signaling pathway. On the whole, the present study demonstrates that IGJ suppresses the invasion and metastasis of breast cancer by inhibiting both the occurrence of EMT and the NF‑κB signaling pathway. These findings may provide novel biomarkers and potential therapeutic targets for the treatment of metastatic breast cancer.
Collapse
Affiliation(s)
- Mengxue Wang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016
| | - Yushen Wu
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016
| | - Xunjia Li
- Department of Nephrology, Chongqing Traditional Chinese Medicine Hospital, Chongqing 400013
| | - Meng Dai
- Department of Geriatric Oncology, Department of Palliative care, Chongqing University Cancer Hospital, Chongqing 400030, P.R. China
| | - Shengwei Li
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010
| |
Collapse
|
2
|
Wang ZX, Zhang GJ, Yang XF, Feng SJ, Ji SS, Qi YB. miRNA-633 and KAI1 as Potential Biomarkers of Malignant Melanoma with Gastric Cancer. Comb Chem High Throughput Screen 2023; 26:1001-1014. [PMID: 35713138 DOI: 10.2174/1386207325666220616125608] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 03/07/2022] [Accepted: 03/08/2022] [Indexed: 11/22/2022]
Abstract
OBJECTIVE Malignant melanoma with gastric cancer is one of the most malignant tumors. However, there have been no reports on the effects of KAI1 and miRNA-633 on the survival and prognosis of patients with malignant melanoma with gastric cancer. METHODS Fifty patients with malignant melanoma and gastric cancer were collected from October 2017 to December 2019. The clinical parameters included clinical information, such as sex, age, tumor size, and tumor staging. RT-qPCR was used to detect the expression of KAI1 and miRNA- 633. The role of KAI1 and miRNA-633 on the overall survival of melanoma was explored by the Pearson chi-square test, Spearman-rho correlation test, Univariate and multivariate cox regression analyses, and Kaplan-Meier method. Furthermore, the bioinformatic analysis was used to verify the role of KAI1 and miRNA-633 on malignant melanoma with gastric cancer. RESULTS The expression of KAI1 and miRNA-633 was significantly related with the tumor size and staging of tumor (p<0.05) based on the Pearson chi-square test. Spearman's correlation coefficient displayed that KAI1 was significantly correlated with the miRNA-633 (ρ=-0.439, p=0.001). The result of multivariate cox proportional regression analysis showed that KAI1 (HR =0.109, 95% CI: 0.031-0.375, p< 0.001), and miRNA-633 (HR = 13.315, 95% CI: 3.844-46.119, p<0.001) were significantly associated with overall survival. CONCLUSION The low expression level of KAI1 and high expression of miRNA-633 are significantly correlated with the poor overall survival prognosis of malignant melanoma with gastric cancer, to provide a basis for KAI1 and miRNA-633 to become novel molecular targets for malignant melanoma with gastric cancer.
Collapse
Affiliation(s)
- Zheng-Xiang Wang
- Department of Dermatology, Cangzhou Central Hospital, No. 16 Xinhua Western Road, Cangzhou, 061000 Hebei Province, P.R. China
| | - Guang-Jing Zhang
- Department of Dermatology, Cangzhou Central Hospital, No. 16 Xinhua Western Road, Cangzhou, 061000 Hebei Province, P.R. China
| | - Xiu-Fang Yang
- Department of Dermatology, Cangzhou Central Hospital, No. 16 Xinhua Western Road, Cangzhou, 061000 Hebei Province, P.R. China
| | - Shi-Jun Feng
- Department of Dermatology, Cangzhou Central Hospital, No. 16 Xinhua Western Road, Cangzhou, 061000 Hebei Province, P.R. China
| | - Shan-Shan Ji
- Department of Dermatology, Cangzhou Central Hospital, No. 16 Xinhua Western Road, Cangzhou, 061000 Hebei Province, P.R. China
| | - Ya-Bin Qi
- Department of Dermatology, Cangzhou Central Hospital, No. 16 Xinhua Western Road, Cangzhou, 061000 Hebei Province, P.R. China
| |
Collapse
|
3
|
Lack of Oestrogen Receptor Expression in Breast Cancer Cells Does Not Correlate with Kisspeptin Signalling and Migration. Int J Mol Sci 2022; 23:ijms23158744. [PMID: 35955878 PMCID: PMC9368979 DOI: 10.3390/ijms23158744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 08/01/2022] [Accepted: 08/02/2022] [Indexed: 12/03/2022] Open
Abstract
Kisspeptin is an anti-metastatic mediator in many cancer types, acting through its receptor, KISS1R. However, controversy remains regarding its role in breast cancer since both pro- and anti-metastatic roles have been ascribed to it. In KISS1R overexpressing triple-negative breast cancer (TNBC) cells, stimulation has been associated with increased invasion and MMP-9 expression, leading to the suggestion that hormone receptor status determines the metastatic effects of kisspeptin. To assess the veracity of this claim, we compared endogenous KISS1R signalling and physiological output in the hormone receptor-negative MDA-MB-231 and BT-20 cell lines after KP-10 (shortest active kisspeptin peptide) stimulation. MDA-MB-231 cells are metastatic when implanted in mice while BT-20 are not and remain epithelial-like. We show that both cell lines express KISS1R mRNA and respond to KP-10 by elevating calcium mobilisation. However, KP-10 stimulation induced migration of MDA-MB-231, but not BT-20 cells, in a calcium-dependent manner. Moreover, only BT-20 cells responded to KP-10 by increasing ERK phosphorylation in a β-arrestin-dependent manner. Interestingly, both cell lines displayed different complements of β-arrestin 1 and 2 expression. Overall, our data shows that, in TNBC, it is not universally true that kisspeptin/KISS1R stimulate migration or pro-metastatic behaviour, as divergent responses were observed in the two TNBC lines tested. Whether this divergence is related to the observed differences in β-arrestin complements warrants further investigation and may enable further stratification of the ability of kisspeptin to influence breast tumour behaviour.
Collapse
|
4
|
Sadoughi F, Dana PM, Homayoonfal M, Sharifi M, Asemi Z. Molecular basis of melatonin protective effects in metastasis: A novel target of melatonin. Biochimie 2022; 202:15-25. [DOI: 10.1016/j.biochi.2022.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Revised: 05/19/2022] [Accepted: 05/19/2022] [Indexed: 11/16/2022]
|
5
|
Viera M, Yip GWC, Shen HM, Baeg GH, Bay BH. Targeting CD82/KAI1 for Precision Therapeutics in Surmounting Metastatic Potential in Breast Cancer. Cancers (Basel) 2021; 13:4486. [PMID: 34503296 PMCID: PMC8431267 DOI: 10.3390/cancers13174486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/27/2021] [Accepted: 09/03/2021] [Indexed: 11/16/2022] Open
Abstract
Metastasis is the main cause of mortality in breast cancer patients. There is an unmet need to develop therapies that can impede metastatic spread. Precision oncology has shown great promise for the treatment of cancers, as the therapeutic approach is tailored to a specific group of patients who are likely to benefit from the treatment, rather than the traditional approach of "one size fits all". CD82, also known as KAI1, a glycoprotein belonging to the tetraspanin family and an established metastasis suppressor, could potentially be exploited to hinder metastases in breast cancer. This review explores the prospect of targeting CD82 as an innovative therapeutic approach in precision medicine for breast cancer patients, with the goal of preventing cancer progression and metastasis. Such an approach would entail the selection of a subset of breast cancer patients with low levels of CD82, and instituting an appropriate treatment scheme tailored towards restoring the levels of CD82 in this group of patients. Proposed precision treatment regimens include current modalities of treating breast cancer, in combination with either clinically approved drugs that could restore the levels of CD82, CD82 peptide mimics or non-coding RNA-based therapeutics.
Collapse
Affiliation(s)
- Maximillian Viera
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117594, Singapore; (M.V.); (G.W.C.Y.)
| | - George Wai Cheong Yip
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117594, Singapore; (M.V.); (G.W.C.Y.)
| | - Han-Ming Shen
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore;
- Faculty of Health Sciences, University of Macau, Taipa, China
| | - Gyeong Hun Baeg
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117594, Singapore; (M.V.); (G.W.C.Y.)
- Ciechanover Institute of Precision and Regenerative Medicine, School of Life and Health Sciences, Chinese University of Hong Kong, Shenzhen 518172, China
| | - Boon Huat Bay
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117594, Singapore; (M.V.); (G.W.C.Y.)
| |
Collapse
|
6
|
Shandiz SAS, Sharifian F, Behboodi S, Ghodratpour F, Baghbani-Arani F. Evaluation of Metastasis Suppressor Genes Expression and In Vitro Anti-Cancer Effects of Zinc Oxide Nanoparticles in Human Breast Cancer Cell Lines MCF-7 and T47D. Avicenna J Med Biotechnol 2021; 13:9-14. [PMID: 33680368 PMCID: PMC7903438 DOI: 10.18502/ajmb.v13i1.4576] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Background: Metallic nanoparticles are useful materials to be applied in biomedical research. In this study, the possible apoptotic and anti-metastatic activity of Zinc Oxide Nanoparticles (ZnONPs) was assessed in breast cancer cells. Methods: First, in vitro cell viability was investigated by MTT assay in two human breast cancer cells (MCF-7 and T47D) and normal Human Embryonic Kidney (HEK293) cells at 37°C overnight. Apoptosis induced by ZnONPs was evaluated by annexin V/PI staining, cell cycle analysis and caspase assay in cancerous cells. Moreover, quantitative real-time PCR was employed for the detection of two metastasis suppressor genes (KAI-1 and NM23) expression in cancerous cells. Results: Data demonstrated that ZnONPs exert a dose-dependent inhibitory effect on the viability of T47D and MCF-7 cells, while no cytotoxic effect was observed on normal HEK293 cells. The mRNA expression levels of KAI-1 and non-metastatic protein (NM23) genes were up-regulated in ZnONP-exposed cancerous cells. ZnONPs were also found to enhance the apoptosis properties of cells by annexin V/PI staining, and caspase assay in cancerous cells. Furthermore, ZnONPs can increase sub-G1 population as compared to negative control. Conclusion: Our findings showed that ZnONPs induce apoptotic activity and can modulate metastasis by up-regulating of KAI-1 and NM23 gene expression in two breast cancer (MCF-7 and T47D) cells.
Collapse
Affiliation(s)
| | - Faryad Sharifian
- Department of Genetics and Biotechnology, Faculty of Biological Sciences, Varamin-Pishva Branch, Islamic Azad University, Varamin, Iran
| | - Sorayya Behboodi
- Department of Biology, Tehran Shargh (East), Payam Noor University, Tehran, Iran
| | - Fatemeh Ghodratpour
- Department of Genetics and Biotechnology, Faculty of Biological Sciences, Varamin-Pishva Branch, Islamic Azad University, Varamin, Iran
| | - Fahimeh Baghbani-Arani
- Department of Genetics and Biotechnology, Faculty of Biological Sciences, Varamin-Pishva Branch, Islamic Azad University, Varamin, Iran
| |
Collapse
|
7
|
Maroufi NF, Ashouri N, Mortezania Z, Ashoori Z, Vahedian V, Amirzadeh-Iranaq MT, Fattahi A, Kazemzadeh H, Bizzarri M, Akbarzadeh M, Nejabati HR, Faridvand Y, Rashidi MR, Nouri M. The potential therapeutic effects of melatonin on breast cancer: An invasion and metastasis inhibitor. Pathol Res Pract 2020; 216:153226. [PMID: 32987338 DOI: 10.1016/j.prp.2020.153226] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 08/30/2020] [Accepted: 09/11/2020] [Indexed: 01/14/2023]
Abstract
Breast cancer is the most common cancer among women and its metastasis which generally observed at the last stage is the major cause of breast cancer-related death. Therefore, the agents that have the potential to prevent metastatic and invasive nature of breast cancer can open up new therapeutic strategies. Melatonin, a major hormone of pineal gland, is a powerful anti-cancer agent. There are growing evidence regarding the protective effect of melatonin against cancer invasion and metastasis. The anti-metastatic feature of melatonin accompanies with suppression of tumor proliferation, induction of tumor apoptosis, regulation of the cell cycle, modulating angiogenesis, impediment of invasion, and induction of cancer cells sensitivity to the chemotherapy agents. More recently, anti-metastatic effect of melatonin through affecting cancer stem cells and vascular mimicry has been identified. Thus, the aim of this review is to discuss the potential therapeutic effect of melatonin on breast cancer via modulating the cells invasion and metastasis.
Collapse
Affiliation(s)
- Nazila Fathi Maroufi
- Student Research Committee, Tabriz University of Medical Sciences, Iran; Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nima Ashouri
- Department of Medicine, Faculty of Cellular and Molecular Biology, University of Shiraz, Iran
| | | | - Zahra Ashoori
- Department of Medical, Faculty of Medical, University of Shahid Beheshti, Iran
| | - Vahid Vahedian
- Department of Experimental Biomedicine, Dr. Vahid Vahedian Medical Diagnostic Laboratory Gorgan, Iran; Department of Medical Laboratory Sciences, Faculty of Medicine, Islamic Azad University (IAU), Sari, Iran
| | | | - Amir Fattahi
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamid Kazemzadeh
- Department of Biology, Faculty of Basic Sciences, Hakim Sabzevari University, Iran
| | - Mariano Bizzarri
- University La Sapienza, Department of Experimental Medicine, System Biology Group, Rome, Italy
| | - Maryam Akbarzadeh
- Department of Biochemistry, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Hamid Reza Nejabati
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yousef Faridvand
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad-Reza Rashidi
- Stem Cell And Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mohammad Nouri
- Stem Cell And Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
8
|
Abstract
The significance of KISS1 goes beyond its original discovery as a metastasis suppressor. Its function as a neuropeptide involved in diverse physiologic processes is more well studied. Enthusiasm regarding KISS1 has cumulated in clinical trials in multiple fields related to reproduction and metabolism. But its cancer therapeutic space is unsettled. This review focuses on collating data from cancer and non-cancer fields in order to understand shared and disparate signaling that might inform clinical development in the cancer therapeutic and biomarker space. Research has focused on amino acid residues 68-121 (kisspeptin 54), binding to the KISS1 receptor and cellular responses. Evidence and counterevidence regarding this canonical pathway require closer look at the covariates so that the incredible potential of KISS1 can be realized.
Collapse
Affiliation(s)
- Thuc Ly
- Department of Cancer Biology, Kansas University Medical Center, 3901 Rainbow Blvd. - MS1071, Kansas City, KS, 66160, USA
| | - Sitaram Harihar
- Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Chennai, Tamil Nadu, 603203, India
| | - Danny R Welch
- Department of Cancer Biology, Kansas University Medical Center, 3901 Rainbow Blvd. - MS1071, Kansas City, KS, 66160, USA.
- University of Kansas Cancer Center, 3901 Rainbow Blvd., Kansas City, KS, 66160, USA.
| |
Collapse
|
9
|
Krishna Latha T, Verma A, Thakur GK, Banerjee B, Kaur N, Singh UR, Sharma S. Down Regulation of KAI1/CD82 in Lymph Node Positive and Advanced T-Stage Group in Breast Cancer Patients. Asian Pac J Cancer Prev 2019; 20:3321-3329. [PMID: 31759355 PMCID: PMC7063004 DOI: 10.31557/apjcp.2019.20.11.3321] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Metastasis represents a deadly aspect of any cancer including breast cancer, given its high prevalence; treatment of metastatic breast cancer remains a clinically unmet need, which necessitates the exploration of metastasis suppressor genes (MSGs). KAI-1/CD82 is an important member of MSGs; the role of KAI1 has been well explored in prostate cancer, however its role in breast cancer is not fully explored and in fact the results of breast cancer studies are contentious. Thus, the present study aimed to investigate expression of KAI1 at both transcriptional and translational levels in the tissue of breast cancer patients and benign breast disease. Further, we analysed the relationship between expression levels of KAI1 and clinicopathological parameters in breast cancer patients. MATERIALS AND METHODS mRNA expression was studied by Real time PCR and protein expression was analyzed by both Western blot and Immunohistochemistry. RESULTS The results of the study indicate that KAI1 expression was remarkably decreased in breast cancer both at the gene and the protein levels (P < 0.05) compared to benign breast disease. In addition, KAI1 expression levels were strongly associated with axillary lymph node status and advanced T stage (p < 0.05), however no association was found with tumor grade, age, menopausal status and receptor status like ER, PR and Her2. CONCLUSION Low expression of KAI1 might be helpful for predicting the lymph node metastasis and T staging, thus predicts malignant prognosis of breast cancer.<br />.
Collapse
Affiliation(s)
- Thammineni Krishna Latha
- Department of Biochemistry University College of Medical Sciences and GTB Hospital , University of Delhi, Dilshad Garden, Delhi, India
| | - Ankur Verma
- Department of Pathology, University College of Medical Sciences and GTB Hospital , University of Delhi, Dilshad Garden, Delhi, India
| | - Gaurav Kumar Thakur
- Department of Biochemistry University College of Medical Sciences and GTB Hospital , University of Delhi, Dilshad Garden, Delhi, India
| | - Basudev Banerjee
- Department of Biochemistry University College of Medical Sciences and GTB Hospital , University of Delhi, Dilshad Garden, Delhi, India
| | - Navneet Kaur
- Department of Surgery, University College of Medical Sciences and GTB Hospital , University of Delhi, Dilshad Garden, Delhi, India
| | - Usha Rani Singh
- Department of Pathology, University College of Medical Sciences and GTB Hospital , University of Delhi, Dilshad Garden, Delhi, India
| | - Sonal Sharma
- Department of Pathology, University College of Medical Sciences and GTB Hospital , University of Delhi, Dilshad Garden, Delhi, India
| |
Collapse
|
10
|
Wu YS, Lee ZY, Chuah LH, Mai CW, Ngai SC. Epigenetics in Metastatic Breast Cancer: Its Regulation and Implications in Diagnosis, Prognosis and Therapeutics. Curr Cancer Drug Targets 2019; 19:82-100. [PMID: 29714144 DOI: 10.2174/1568009618666180430130248] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 02/21/2018] [Accepted: 04/03/2018] [Indexed: 02/06/2023]
Abstract
Despite advances in the treatment regimen, the high incidence rate of breast cancer (BC) deaths is mostly caused by metastasis. Recently, the aberrant epigenetic modifications, which involve DNA methylation, histone modifications and microRNA (miRNA) regulations become attractive targets to treat metastatic breast cancer (MBC). In this review, the epigenetic alterations of DNA methylation, histone modifications and miRNA regulations in regulating MBC are discussed. The preclinical and clinical trials of epigenetic drugs such as the inhibitor of DNA methyltransferase (DNMTi) and the inhibitor of histone deacetylase (HDACi), as a single or combined regimen with other epigenetic drug or standard chemotherapy drug to treat MBCs are discussed. The combined regimen of epigenetic drugs or with standard chemotherapy drugs enhance the therapeutic effect against MBC. Evidences that epigenetic changes could have implications in diagnosis, prognosis and therapeutics for MBC are also presented. Several genes have been identified as potential epigenetic biomarkers for diagnosis and prognosis, as well as therapeutic targets for MBC. Endeavors in clinical trials of epigenetic drugs against MBC should be continued although limited success has been achieved. Future discovery of epigenetic drugs from natural resources would be an attractive natural treatment regimen for MBC. Further research is warranted in translating research into clinical practice with the ultimate goal of treating MBC by epigenetic therapy in the near future.
Collapse
Affiliation(s)
- Yuan Seng Wu
- School of Biosciences, Faculty of Science, University of Nottingham Malaysia Campus, Selangor, Malaysia
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia
| | - Zhong Yang Lee
- School of Biosciences, Faculty of Science, University of Nottingham Malaysia Campus, Selangor, Malaysia
| | - Lay-Hong Chuah
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia
- Advanced Engineering Platform, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia
| | - Chun Wai Mai
- Department of Pharmaceutical Chemistry, International Medical University, Bukit Jalil, Kuala Lumpur, Malaysia
| | - Siew Ching Ngai
- School of Biosciences, Faculty of Science, University of Nottingham Malaysia Campus, Selangor, Malaysia
| |
Collapse
|
11
|
Bell R, Barraclough R, Vasieva O. Gene Expression Meta-Analysis of Potential Metastatic Breast Cancer Markers. Curr Mol Med 2018; 17:200-210. [PMID: 28782484 PMCID: PMC5748874 DOI: 10.2174/1566524017666170807144946] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 07/20/2017] [Accepted: 07/30/2017] [Indexed: 12/15/2022]
Abstract
Background: Breast cancer metastasis is a highly prevalent cause of death for European females. DNA microarray analysis has established that primary tumors, which remain localized, differ in gene expression from those that metastasize. Cross-analysis of these studies allow to revile the differences that may be used as predictive in the disease prognosis and therapy. Objective: The aim of the project was to validate suggested prognostic and therapeutic markers using meta-analysis of data on gene expression in metastatic and primary breast cancer tumors. Method: Data on relative gene expression values from 12 studies on primary breast cancer and breast cancer metastasis were retrieved from Genevestigator (Nebion) database. The results of the data meta-analysis were compared with results of literature mining for suggested metastatic breast cancer markers and vectors and consistency of their reported differential expression. Results: Our analysis suggested that transcriptional expression of the COX2 gene is significantly downregulated in metastatic tissue compared to normal breast tissue, but is not downregulated in primary tumors compared with normal breast tissue and may be used as a differential marker in metastatic breast cancer diagnostics. RRM2 gene expression decreases in metastases when compared to primary breast cancer and could be suggested as a marker to trace breast cancer evolution. Our study also supports MMP1, VCAM1, FZD3, VEGFC, FOXM1 and MUC1 as breast cancer onset markers, as these genes demonstrate significant differential expression in breast neoplasms compared with normal breast tissue. Conclusion: COX2 and RRM2 are suggested to be prominent markers for breast cancer metastasis. The crosstalk between upstream regulators of genes differentially expressed in primary breast tumors and metastasis also suggests pathways involving p53, ER1, ERB-B2, TNF and WNT, as the most promising regulators that may be considered for new complex drug therapeutic interventions in breast cancer metastatic progression.
Collapse
Affiliation(s)
- R Bell
- Institute of Integrative Biology, University of Liverpool, Crown Street, Liverpool, L69 7ZB. United Kingdom
| | - R Barraclough
- Institute of Integrative Biology, University of Liverpool, Crown Street, Liverpool, L69 7ZB. United Kingdom
| | - O Vasieva
- Institute of Integrative Biology, University of Liverpool, Crown Street, Liverpool, L69 7ZB. United Kingdom
| |
Collapse
|
12
|
T lymphocytes facilitate brain metastasis of breast cancer by inducing Guanylate-Binding Protein 1 expression. Acta Neuropathol 2018; 135:581-599. [PMID: 29350274 PMCID: PMC5978929 DOI: 10.1007/s00401-018-1806-2] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 01/09/2018] [Accepted: 01/09/2018] [Indexed: 02/01/2023]
Abstract
The discovery of genes and molecular pathways involved in the formation of brain metastasis would direct the development of therapeutic strategies to prevent this deadly complication of cancer. By comparing gene expression profiles of Estrogen Receptor negative (ER-) primary breast tumors between patients who developed metastasis to brain and to organs other than brain, we found that T lymphocytes promote the formation of brain metastases. To functionally test the ability of T cells to promote brain metastasis, we used an in vitro blood–brain barrier (BBB) model. By co-culturing T lymphocytes with breast cancer cells, we confirmed that T cells increase the ability of breast cancer cells to cross the BBB. Proteomics analysis of the tumor cells revealed Guanylate-Binding Protein 1 (GBP1) as a key T lymphocyte-induced protein that enables breast cancer cells to cross the BBB. The GBP1 gene appeared to be up-regulated in breast cancer of patients who developed brain metastasis. Silencing of GBP1 reduced the ability of breast cancer cells to cross the in vitro BBB model. In addition, the findings were confirmed in vivo in an immunocompetent syngeneic mouse model. Co-culturing of ErbB2 tumor cells with activated T cells induced a significant increase in Gbp1 expression by the cancer cells. Intracardial inoculation of the co-cultured tumor cells resulted in preferential seeding to brain. Moreover, intracerebral outgrowth of the tumor cells was demonstrated. The findings point to a role of T cells in the formation of brain metastases in ER- breast cancers, and provide potential targets for intervention to prevent the development of cerebral metastases.
Collapse
|
13
|
Fratangelo F, Carriero MV, Motti ML. Controversial Role of Kisspeptins/KiSS-1R Signaling System in Tumor Development. Front Endocrinol (Lausanne) 2018; 9:192. [PMID: 29760678 PMCID: PMC5936968 DOI: 10.3389/fendo.2018.00192] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 04/09/2018] [Indexed: 02/01/2023] Open
Abstract
KiSS-1 was first described as a metastasis suppressor gene in malignant melanoma. KiSS-1 encodes a 145 amino-acid residue peptide that is further processed, producing the 54 amino acid metastin and shorter peptides collectively named kisspeptins (KPs). KPs bind and activate KiSS-1R (GPR54). Although the KPs system has been extensively studied for its role in endocrinology of reproductive axis in mammals, its role in cancer is still controversial. Experimental evidences show that KP system exerts an anti-metastatic effect by the regulation of cellular migration and invasion in several cancer types. However, the role of KPs/KiSS-1R is very complex. Genomic studies suggest that KiSS-1/KiSS-1R expression might be different in the various stages of tumor development. Furthermore, overexpression of KiSS-1R has been reported to elicit drug resistance in triple negative breast cancer. In this review, we focused on multiple functions exerted by the KPs/KiSS-1R system in regulating tumor progression.
Collapse
Affiliation(s)
| | | | - Maria Letizia Motti
- IRCCS Istituto Nazionale Tumori “Fondazione G. Pascale”, Naples, Italy
- Parthenope University of Naples, Naples, Italy
- *Correspondence: Maria Letizia Motti,
| |
Collapse
|
14
|
Kaverina N, Borovjagin AV, Kadagidze Z, Baryshnikov A, Baryshnikova M, Malin D, Ghosh D, Shah N, Welch DR, Gabikian P, Karseladze A, Cobbs C, Ulasov IV. Astrocytes promote progression of breast cancer metastases to the brain via a KISS1-mediated autophagy. Autophagy 2017; 13:1905-1923. [PMID: 28981380 PMCID: PMC5788498 DOI: 10.1080/15548627.2017.1360466] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 07/07/2017] [Accepted: 07/24/2017] [Indexed: 12/14/2022] Open
Abstract
Formation of metastases, also known as cancer dissemination, is an important stage of breast cancer (BrCa) development. KISS1 expression is associated with inhibition of metastases development. Recently we have demonstrated that BrCa metastases to the brain exhibit low levels of KISS1 expression at both mRNA and protein levels. By using multicolor immunofluorescence and coculture techniques here we show that normal adult astrocytes in the brain are capable of promoting metastatic transformation of circulating breast cancer cells localized to the brain through secretion of chemokine CXCL12. The latter was found in this study to downregulate KISS1 expression at the post-transcriptional level via induction of microRNA-345 (MIR345). Furthermore, we demonstrated that ectopic expression of KISS1 downregulates ATG5 and ATG7, 2 key modulators of autophagy, and works concurrently with autophagy inhibitors, thereby implicating autophagy in the mechanism of KISS1-mediated BrCa metastatic transformation. We also found that expression of KISS1 in human breast tumor specimens inversely correlates with that of MMP9 and IL8, implicated in the mechanism of metastatic invasion, thereby supporting the role of KISS1 as a potential regulator of BrCa metastatic invasion in the brain. This conclusion is further supported by the ability of KISS1, ectopically overexpressed from an adenoviral vector in MDA-MB-231Br cells with silenced expression of the endogenous gene, to revert invasive phenotype of those cells. Taken together, our results strongly suggest that human adult astrocytes can promote brain invasion of the brain-localized circulating breast cancer cells by upregulating autophagy signaling pathways via the CXCL12-MIR345- KISS1 axis.
Collapse
Affiliation(s)
- Natalya Kaverina
- Department of Tumor Immunology, Institute of Experimental Diagnostics and Therapy of Tumors, N.N. Blokhin Cancer Research Center, Moscow, Russia
| | - Anton V. Borovjagin
- University of Alabama at Birmingham School of Dentistry, Institute of Oral Health Research, Birmingham, AL, USA
| | - Zaira Kadagidze
- Department of Tumor Immunology, Institute of Experimental Diagnostics and Therapy of Tumors, N.N. Blokhin Cancer Research Center, Moscow, Russia
| | - Anatoly Baryshnikov
- Institute of Experimental Diagnostics and Therapy of Tumors, N.N. Blokhin Cancer Research Center, Moscow, Russia
| | - Maria Baryshnikova
- Institute of Experimental Diagnostics and Therapy of Tumors, N.N. Blokhin Cancer Research Center, Moscow, Russia
| | - Dmitry Malin
- Department of Endocrinology, University of Wisconsin-Madison, Madison, WI, USA
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Dhimankrishhna Ghosh
- Center for Advanced Brain Tumor Treatment, Swedish Neuroscience Institute, Seattle, WA, USA
| | - Nameeta Shah
- Center for Advanced Brain Tumor Treatment, Swedish Neuroscience Institute, Seattle, WA, USA
| | - Danny R. Welch
- Department of Cancer Biology, Kansas University Medical Center (KUMC), Kansas City, KS, USA
| | - Patrik Gabikian
- Department of Neurosurgery, Kaiser Permanente Los Angeles Medical Center, Los Angeles, CA, USA
| | - Apollon Karseladze
- Pathology, Institute of Experimental Diagnostics and Therapy of Tumors, N.N. Blokhin Cancer Research Center, Moscow, Russia
| | - Charles Cobbs
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Ilya V. Ulasov
- Center for Advanced Brain Tumor Treatment, Swedish Neuroscience Institute, Seattle, WA, USA
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russia
| |
Collapse
|
15
|
Kim TH, Cho SG. Melatonin-induced KiSS1 expression inhibits triple-negative breast cancer cell invasiveness. Oncol Lett 2017; 14:2511-2516. [PMID: 28781689 DOI: 10.3892/ol.2017.6434] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 04/06/2017] [Indexed: 12/21/2022] Open
Abstract
Breast cancer is one of the most common types of cancer in women, and its metastasis increases the risk of mortality. Melatonin, a hormone that regulates the circadian rhythm, has been revealed to inhibit breast cancer growth and metastasis. However, its involvement in highly metastatic triple-negative breast cancer cells is yet to be elucidated. The present study demonstrated that melatonin inhibited the metastatic abilities of triple-negative breast cancer cells and prolonged its inhibitory effect via the expression of kisspeptin (KiSS1), which is a suppressor of metastasis. Melatonin at concentrations ranging from 1 nM to 10 µM did not affect the proliferation of metastatic MDA-MB-231 and HCC-70 triple-negative breast cancer cells. However, melatonin repressed invasiveness in triple-negative breast cancer cells. Additionally, conditional medium from melatonin-treated MDA-MB-231 cells repressed the invasiveness of triple-negative breast cancer cells. Melatonin promoted the production of KiSS1, a metastasis suppressor encoded by the KiSS1 gene. In addition, melatonin increased KiSS1 expression via the expression and transcriptional activation of GATA binding protein 3. Silencing of KiSS1 weakened melatonin inhibition of breast cancer cell invasiveness. Therefore, the present study concluded that melatonin activates KiSS1 production in metastatic breast cancer cells, suggesting that melatonin activation of KiSS1 production may regulate the process of breast cancer metastasis.
Collapse
Affiliation(s)
- Tae-Hun Kim
- Department of Biotechnology, Korea National University of Transportation, Jeungpyeong, Chungcheongbuk 368-701, Republic of Korea
| | - Sung-Gook Cho
- Department of Biotechnology, Korea National University of Transportation, Jeungpyeong, Chungcheongbuk 368-701, Republic of Korea
| |
Collapse
|
16
|
Yan H, Ji X, Li J, Zhang L, Zhao P. Overexpression of KAI1 inhibits retinoblastoma metastasis in vitro. Oncol Lett 2017; 13:827-833. [PMID: 28356965 DOI: 10.3892/ol.2016.5507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 10/05/2016] [Indexed: 11/06/2022] Open
Abstract
The present study aimed to investigate the expression of cluster of differentiation 82 (KAI1), a gene involved in the suppression of tumor metastasis, in human retinoblastoma (RB) tissue and to study the effect of KAI1 expression on RB cell migration and invasion. KAI1 expression was examined in 26 patients with non-invasive and invasive retinoblastoma using reverse transcription-quantitative polymerase chain reaction and western blot analysis. A lentiviral vector containing KAI1 cDNA was used to transfect the two RB cell lines, HXO-Rb44-Gl and Y79. Following successful transfection, the migratory and invasive capacity of the two RB cell lines was evaluated using a Transwell® migration assay. KAI1 expression was observed to be downregulated in invasive RB compared to non-invasive RB. The migratory and invasive capacities of KAI1 transfected cell lines were significantly decreased compared to those of the control cells. KAI1 may be involved in retinoblastoma metastasis, and increased expression of KAI1 significantly inhibits the metastatic ability of RB cells in vitro.
Collapse
Affiliation(s)
- Hui Yan
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, P.R. China
| | - Xunda Ji
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, P.R. China
| | - Jing Li
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, P.R. China
| | - Lei Zhang
- Department of Ophthalmology, Tongji University Affiliated Yangpu Hospital, Shanghai 200090, P.R. China
| | - Peiquan Zhao
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, P.R. China
| |
Collapse
|
17
|
Singh R, Bhatt MLB, Singh SP, Kumar V, Goel MM, Mishra DP, Kumar R. Evaluation of KiSS1 as a Prognostic Biomarker in North Indian Breast Cancer Cases. Asian Pac J Cancer Prev 2017; 17:1789-95. [PMID: 27221854 DOI: 10.7314/apjcp.2016.17.4.1789] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Breast cancer is the commonest female cancer worldwide and its propensity to metastasize negatively impacts on therapeutic outcome. Several clinicopathological parameters with prognostic/predictive significance have been associated with metastatic suppressor expression levels. The role of metastatic suppressor gene (MSG) KiSS1 in breast cancer remains unclear. Our goal was to investigate the possible clinical significance of KiSS1 breast cancer. MATERIALS AND METHODS The study was conducted on 87 histologically proven cases of breast cancer and background normal tiisue. Quantitative reverse transcriptase polymerase chain reaction (qRT PCR) and immunohistochemistry (IHC) were used to investigate KiSS1 at gene and protein levels, respectively, for correlation with several patient characteristics including age, family history, hormonal receptor status, stage, tumor size, nodal involvement and metastatic manifestation and finally with median overall survival (OS). RESULTS Our study revealed (i) KiSS1 levels were generally elevated in breast cancer vs normal tissue (< 0.05). (ii) however, a statistically significant lower expression of KiSS1 was observed in metastatic vs non metastatic cases (P = 0.04). (iii) KiSS1 levels strongly correlated with T,N,M category, histological grade and advanced stage (<0.001) but not other studied parameters. (iv) Lastly, a significant correlation between expression of KiSS1 and median OS was found (P = 0.04). CONCLUSIONS Conclusively, less elevated KiSS1 expression is a negative prognostic factor for OS, advancing tumor stage, axillary lymph node status, metastatic propensity and advancing grade of the breast cancer patient. Patients with negative KiSS1 expression may require a more intensive therapeutic strategy.
Collapse
Affiliation(s)
- Richa Singh
- Department of Radiotherapy, King George's Medical University, India E-mail :
| | | | | | | | | | | | | |
Collapse
|
18
|
Feng J, Huang C, Wren JD, Wang DW, Yan J, Zhang J, Sun Y, Han X, Zhang XA. Tetraspanin CD82: a suppressor of solid tumors and a modulator of membrane heterogeneity. Cancer Metastasis Rev 2016; 34:619-33. [PMID: 26335499 DOI: 10.1007/s10555-015-9585-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Tetraspanin CD82 suppresses the progression and metastasis of a wide range of solid malignant tumors. However, its roles in tumorigenesis and hematopoietic malignancy remain unclear. Ubiquitously expressed CD82 restrains cell migration and cell invasion by modulating both cell-matrix and cell-cell adhesiveness and confining outside-in pro-motility signaling. This restraint at least contributes to, if not determines, the metastasis-suppressive activity and, also likely, the physiological functions of CD82. As a modulator of cell membrane heterogeneity, CD82 alters microdomains, trafficking, and topography of the membrane by changing the membrane molecular landscape. The functional activities of membrane molecules and the cytoskeletal interaction of the cell membrane are subsequently altered, followed by changes in cellular functions. Given its pathological and physiological importance, CD82 is a promising candidate for clinically predicting and blocking tumor progression and metastasis and also an emerging model protein for mechanistically understanding cell membrane organization and heterogeneity.
Collapse
Affiliation(s)
- Jin Feng
- Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Chao Huang
- Stephenson Cancer Center and Department of Physiology, University of Oklahoma Health Sciences Center, BRC 1474, 975 NE 10th Street, Oklahoma City, OK, 73104, USA
| | - Jonathan D Wren
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Dao-Wen Wang
- Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Jizhou Yan
- Institute for Marine Biosystem and Neurosciences, Shanghai Ocean University, Shanghai, China
| | - Jiexin Zhang
- Department of Biochemistry, Nanjing Medical University, Nanjing, China
| | - Yujie Sun
- Department of Biochemistry, Nanjing Medical University, Nanjing, China
| | - Xiao Han
- Department of Biochemistry, Nanjing Medical University, Nanjing, China
| | - Xin A Zhang
- Stephenson Cancer Center and Department of Physiology, University of Oklahoma Health Sciences Center, BRC 1474, 975 NE 10th Street, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
19
|
Uthaisar K, Vaeteewoottacharn K, Seubwai W, Talabnin C, Sawanyawisuth K, Obchoei S, Kraiklang R, Okada S, Wongkham S. Establishment and characterization of a novel human cholangiocarcinoma cell line with high metastatic activity. Oncol Rep 2016; 36:1435-46. [PMID: 27461717 DOI: 10.3892/or.2016.4974] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 05/11/2016] [Indexed: 11/06/2022] Open
Abstract
Cholangiocarcinoma (CCA) is a highly metastatic tumor, and the lung is a common site of metastasis. A greater understanding of the biology of metastases is needed to improve treatment outcomes. Herein, a highly metastatic human CCA subline, KKU-213L5 from an original cell line, KKU-213 that has marginally metastatic ability, was established and characterized. KKU-213L5 was selected in vivo through the fifth serial passage of pulmonary metastasized tissues via tail-vein injection in NOD/scid/Jak3 mice. The metastatic abilities of the KKU-213L5 cells were compared with the parental line in vitro and in vivo. The expression profile of this metastatic cell line was determined using real-time PCR. KKU-213L5 cells were found to possess higher metastatic phenotypes, i.e., growth rates, stem cell surface markers (CD133), migration and invasion characteristics when compared with the parental cells. Compared to the KKU-213 cells, KKU-213L5 cells formed larger tumors in subcutaneous xenografted mice and had a >10-fold increase in lung metastases in the tail-vein injected metastatic mouse model. Mice injected intravenously with KKU-213L5 cells had a significantly shorter survival. Analysis of the expressed genes related to progression of cancer revealed significant upregulation of anterior gradient protein-2 (AGR2) and suppression of KiSS-1 in the KKU-213L5 cells. The association of these two genes with metastasis was affirmed in CCA patient tissues since increased AGR2 expression and decreased KiSS-1 expression were found in higher stage patient tumors. In conclusion, a highly metastatic human CCA cell line was established and characterized. It is plausible that the differential expression between the parental KKU-213 and highly metastatic KKU-213L5 cells may be beneficial to classify novel genes associated with metastasis. The KKU-213L5 cell line should serve as a valued device for discovering the molecular mechanisms of CCA metastasis and enabling the search for an effective therapy for the unmet clinical need in CCA.
Collapse
Affiliation(s)
- Kwuntida Uthaisar
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | | | - Wunchana Seubwai
- Department of Forensic Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Chutima Talabnin
- School of Biochemistry, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand
| | - Kanlayanee Sawanyawisuth
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Sumalee Obchoei
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Ratthaphol Kraiklang
- Department of Nutrition, Faculty of Public Health, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Seiji Okada
- Division of Hematopoiesis, Center for AIDS Research, Kumamoto University, Honjo, Kumamoto 860-0811, Japan
| | - Sopit Wongkham
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| |
Collapse
|
20
|
Timologou A, Zafrakas M, Grimbizis G, Miliaras D, Kotronis K, Stamatopoulos P, Tarlatzis BC. Immunohistochemical expression pattern of metastasis suppressors KAI1 and KISS1 in endometriosis and normal endometrium. Eur J Obstet Gynecol Reprod Biol 2016; 199:110-5. [PMID: 26918694 DOI: 10.1016/j.ejogrb.2016.02.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2015] [Revised: 02/03/2016] [Accepted: 02/05/2016] [Indexed: 01/01/2023]
Abstract
OBJECTIVE To analyze the expression pattern of metastasis suppressors KAI1 and KISS1 in the endometrium of patients with and without endometriosis. STUDY DESIGN In this pilot study, tissue samples were prospectively collected from 38 patients with endometriosis and 29 without endometriosis, undergoing operative laparoscopy in the proliferative phase of the menstrual cycle; diagnosis or absence of endometriosis was confirmed histologically. Protein expression of KAI1 and KISS1 were analyzed immunohistochemically in endometriotic lesions and the eutopic endometrium of patients with endometriosis and without endometriosis. RESULTS KAI1 expression was significantly decreased in the glandular eutopic endometrium of endometriosis patients as compared with that of patients without endometriosis (p=0.008). On the other hand, in endometriosis patients, KAI1 expression was significantly increased in the ectopic as compared with the eutopic endometrial stroma (p=0.021). There were no other significant differences in KAI1 expression between different groups. KISS1 expression in the ectopic glandular endometrium was significantly increased as compared with the eutopic glandular endometrium from patients with (p=0.004) and without endometriosis (p=0.008). There was no significant difference in KISS1 protein expression in the stromal endometrium between the three groups. CONCLUSIONS KAI1 and KISS1 are implicated in the pathogenesis and maintenance of endometriosis. Future studies should investigate whether KAI1 and KISS1 could be used as markers for early and minimally invasive detection of endometriosis based on their differential protein expression pattern in the eutopic endometrium of patients with and without endometriosis.
Collapse
Affiliation(s)
- Anna Timologou
- 1st Department of Obstetrics and Gynecology, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Menelaos Zafrakas
- 1st Department of Obstetrics and Gynecology, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece; School of Health and Medical Care, Alexander Technological Educational Institute of Thessaloniki, Thessaloniki, Greece.
| | - Grigorios Grimbizis
- 1st Department of Obstetrics and Gynecology, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Dimosthenis Miliaras
- Laboratory of Histology & Embryology, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Konstantinos Kotronis
- 1st Department of Obstetrics and Gynecology, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Panayiotis Stamatopoulos
- 1st Department of Obstetrics and Gynecology, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Basil C Tarlatzis
- 1st Department of Obstetrics and Gynecology, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
21
|
Shandiz SAS, Khosravani M, Mohammadi S, Noorbazargan H, Mirzaie A, Inanlou DN, Jalali MD, Jouzaghkar H, Baghbani-Arani F, Keshavarz-Pakseresht B. Evaluation of imatinib mesylate (Gleevec) on KAI1/CD82 gene expression in breast cancer MCF-7 cells using quantitative real-time PCR. Asian Pac J Trop Biomed 2016. [DOI: 10.1016/j.apjtb.2015.10.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
22
|
Li Q, Xiao L, Harihar S, Welch DR, Vargis E, Zhou A. In vitro biophysical, microspectroscopic and cytotoxic evaluation of metastatic and non-metastatic cancer cells in responses to anti-cancer drug. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2015; 7:10162-10169. [PMID: 26744605 PMCID: PMC4699680 DOI: 10.1039/c5ay01810b] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
The Breast Cancer Metastasis Suppressor 1 (BRMS1) is a nucleo-cytoplasmic protein that suppresses cancer metastasis without affecting the growth of the primary tumor. Previous work has shown that it decreases the expression of protein mediators involved in chemoresistance. This study measured the biomechanical and biochemical changes in BRMS1 expression and the responses of BRMS1 to drug treatments on cancer cells in vitro. The results show that BRMS1 expression affects biomechanical properties by decreasing the Young's modulus and adhesion force of breast cancer cells after doxorubicin (DOX) exposure. Raman spectral bands corresponding to DNA/RNA, lipids and proteins were similar for all cells after DOX treatment. The expression of cytokines were similar for cancer cells after DOX exposure, although BRMS1 expression had different effects on the secretion of cytokines for breast cancer cells. The absence of significant changes on apoptosis, reactive oxygen species (ROS) expression and cell viability after BRMS1 expression shows that BRMS1 has little effect on cellular chemoresistance. Analyzing cancer protein expression is critical in evaluating therapeutics. Our study may provide evidence of the benefit of metastatic suppressor expression before chemotherapy.
Collapse
Affiliation(s)
- Qifei Li
- Department of Biological Engineering, Utah State University, Logan, UT 84322, USA
| | - Lifu Xiao
- Department of Biological Engineering, Utah State University, Logan, UT 84322, USA
| | - Sitaram Harihar
- Department of Cancer Biology, The University of Kansas Medical Center and The University of Kansas Cancer Center, Kansas City, KS 66160, USA
| | - Danny R. Welch
- Department of Cancer Biology, The University of Kansas Medical Center and The University of Kansas Cancer Center, Kansas City, KS 66160, USA
| | - Elizabeth Vargis
- Department of Biological Engineering, Utah State University, Logan, UT 84322, USA
| | - Anhong Zhou
- Department of Biological Engineering, Utah State University, Logan, UT 84322, USA
| |
Collapse
|
23
|
Pixberg CF, Schulz WA, Stoecklein NH, Neves RPL. Characterization of DNA Methylation in Circulating Tumor Cells. Genes (Basel) 2015; 6:1053-75. [PMID: 26506390 PMCID: PMC4690028 DOI: 10.3390/genes6041053] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Revised: 10/09/2015] [Accepted: 10/14/2015] [Indexed: 02/07/2023] Open
Abstract
Epigenetics contributes to molecular mechanisms leading to tumor cell transformation and systemic progression of cancer. However, the dynamics of epigenetic remodeling during metastasis remains unexplored. In this context, circulating tumor cells (CTCs) might enable a direct insight into epigenetic mechanisms relevant for metastasis by providing direct access to systemic cancer. CTCs can be used as prognostic markers in cancer patients and are regarded as potential metastatic precursor cells. However, despite substantial technical progress, the detection and molecular characterization of CTCs remain challenging, in particular the analysis of DNA methylation. As recent studies have started to address the epigenetic state of CTCs, we discuss here the potential of such investigations to elucidate mechanisms of metastasis and to develop tumor biomarkers.
Collapse
Affiliation(s)
- Constantin F Pixberg
- Department of General, Visceral and Pediatric Surgery, University Hospital and Medical Faculty of the Heinrich-Heine University Düsseldorf, Moorenstr. 5, 40225 Düsseldorf, Germany.
| | - Wolfgang A Schulz
- Department of Urology, University Hospital and Medical Faculty of the Heinrich-Heine University Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany.
| | - Nikolas H Stoecklein
- Department of General, Visceral and Pediatric Surgery, University Hospital and Medical Faculty of the Heinrich-Heine University Düsseldorf, Moorenstr. 5, 40225 Düsseldorf, Germany.
| | - Rui P L Neves
- Department of General, Visceral and Pediatric Surgery, University Hospital and Medical Faculty of the Heinrich-Heine University Düsseldorf, Moorenstr. 5, 40225 Düsseldorf, Germany.
| |
Collapse
|
24
|
Kodura MA, Souchelnytskyi S. Breast carcinoma metastasis suppressor gene 1 (BRMS1): update on its role as the suppressor of cancer metastases. Cancer Metastasis Rev 2015; 34:611-8. [DOI: 10.1007/s10555-015-9583-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
25
|
Progesterone and Src family inhibitor PP1 synergistically inhibit cell migration and invasion of human basal phenotype breast cancer cells. BIOMED RESEARCH INTERNATIONAL 2015; 2015:426429. [PMID: 26075237 PMCID: PMC4449873 DOI: 10.1155/2015/426429] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 11/19/2014] [Indexed: 01/01/2023]
Abstract
Basal phenotype breast cancer is one of the most aggressive breast cancers that frequently metastasize to brain. The role of sex hormones and their receptors in development of this disease is largely unclear. We demonstrated that mPRα was expressed at a moderate level in a brain metastatic BPBC cell line MB231Br, which was derived from the parent mPRα undetectable MB231 cells. It functioned as an essential mediator for progesterone induced inhibitory effects on cell migration of MB231Br and, when coincubated with PP1, synergistically enhanced the progesterone's inhibitory effect on cell migration and invasion in vitro. Progesterone and PP1 cotreatment induced a cascade of molecular signaling events, such as dephosphorylation of FAK, downregulation of MMP9, VEGF, and KCNMA1 expressions. Our in vitro study demonstrated that mPRα was expressed and functioned as an essential mediator for progesterone induced inhibitory effects on cell migration and invasion in BPBC cells. This inhibitory effect was enhanced by PP1 via FAK dephosphorylation, MMP9, VEGF, and KCNMA1 downregulation mechanisms. Our study provides a new clue toward the development of novel promising agents and pathways for inhibiting nuclear hormonal receptor-negative and endocrine-resistant breast cancers.
Collapse
|
26
|
Prabhu VV, Sakthivel KM, Guruvayoorappan C. Kisspeptins (KiSS-1): essential players in suppressing tumor metastasis. Asian Pac J Cancer Prev 2015; 14:6215-20. [PMID: 24377507 DOI: 10.7314/apjcp.2013.14.11.6215] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Kisspeptins (KPs) encoded by the KiSS-1 gene are C-terminally amidated peptide products, including KP- 10, KP-13, KP-14 and KP-54, which are endogenous agonists for the G-protein coupled receptor-54 (GPR54). Functional analyses have demonstrated fundamental roles of KiSS-1 in whole body homeostasis including sexual differentiation of brain, action on sex steroids and metabolic regulation of fertility essential for human puberty and maintenance of adult reproduction. In addition, intensive recent investigations have provided substantial evidence suggesting roles of Kisspeptin signalling via its receptor GPR54 in the suppression of metastasis with a variety of cancers. The present review highlights the latest studies regarding the role of Kisspeptins and the KiSS-1 gene in tumor progression and also suggests targeting the KiSS-1/GPR54 system may represent a novel therapeutic approach for cancers. Further investigations are essential to elucidate the complex pathways regulated by the Kisspeptins and how these pathways might be involved in the suppression of metastasis across a range of cancers.
Collapse
|
27
|
Rai A, Menon AV, Jalan S. Randomness and preserved patterns in cancer network. Sci Rep 2014; 4:6368. [PMID: 25220184 PMCID: PMC5376158 DOI: 10.1038/srep06368] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 08/26/2014] [Indexed: 01/16/2023] Open
Abstract
Breast cancer has been reported to account for the maximum cases among all female cancers till date. In order to gain a deeper insight into the complexities of the disease, we analyze the breast cancer network and its normal counterpart at the proteomic level. While the short range correlations in the eigenvalues exhibiting universality provide an evidence towards the importance of random connections in the underlying networks, the long range correlations along with the localization properties reveal insightful structural patterns involving functionally important proteins. The analysis provides a benchmark for designing drugs which can target a subgraph instead of individual proteins.
Collapse
Affiliation(s)
- Aparna Rai
- Centre for Bio-Science and Bio-Medical Engineering, Indian Institute of Technology Indore, M-Block, IET-DAVV Campus, Khandwa Road, Indore 452017, India
| | - A Vipin Menon
- Complex Systems Lab, Discipline of Physics, Indian Institute of Technology Indore, M-Block, IET-DAVV Campus, Khandwa Road, Indore 452017, India
| | - Sarika Jalan
- 1] Centre for Bio-Science and Bio-Medical Engineering, Indian Institute of Technology Indore, M-Block, IET-DAVV Campus, Khandwa Road, Indore 452017, India [2] Complex Systems Lab, Discipline of Physics, Indian Institute of Technology Indore, M-Block, IET-DAVV Campus, Khandwa Road, Indore 452017, India
| |
Collapse
|
28
|
Liu M, Wang S, Pan L, Yang D, Xie F, Liu P, Guo J, Zhang J, Zhou B. A new model for predicting non-sentinel lymph node status in Chinese sentinel lymph node positive breast cancer patients. PLoS One 2014; 9:e104117. [PMID: 25111296 PMCID: PMC4128817 DOI: 10.1371/journal.pone.0104117] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Accepted: 07/04/2014] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Our goal is to validate the Memorial Sloan-Kettering Cancer Center (MSKCC) nomogram and Stanford Online Calculator (SOC) for predicting non-sentinel lymph node (NSLN) metastasis in Chinese patients, and develop a new model for better prediction of NSLN metastasis. METHODS The MSKCC nomogram and SOC were used to calculate the probability of NSLN metastasis in 120 breast cancer patients. Univariate and multivariate analyses were performed to evaluate the relationship between NSLN metastasis and clinicopathologic factors, using the medical records of the first 80 breast cancer patients. A new model predicting NSLN metastasis was developed from the 80 patients. RESULTS The MSKCC and SOC predicted NSLN metastasis in a series of 120 patients with an area under the receiver operating characteristic curve (AUC) of 0.688 and 0.734, respectively. For predicted probability cut-off points of 10%, the false-negative (FN) rates of MSKCC and SOC were both 4.4%, and the negative predictive value (NPV) 75.0% and 90.0%, respectively. Tumor size, Kiss-1 expression in positive SLN and size of SLN metastasis were independently associated with NSLN metastasis (p<0.05). A new model (Peking University People's Hospital, PKUPH) was developed using these three variables. The MSKCC, SOC and PKUPH predicted NSLN metastasis in the second 40 patients from the 120 patients with an AUC of 0.624, 0.679 and 0.795, respectively. CONCLUSION MSKCC nomogram and SOC did not perform as well as their original researches in Chinese patients. As a new predictor, Kiss-1 expression in positive SLN correlated independently with NSLN metastasis strongly. PKUPH model achieved higher accuracy than MSKCC and SOC in predicting NSLN metastasis in Chinese patients.
Collapse
Affiliation(s)
- Miao Liu
- Breast Disease Center, Peking University People's Hospital, Beijing, China
| | - Shu Wang
- Breast Disease Center, Peking University People's Hospital, Beijing, China
| | - Lu Pan
- Breast Disease Center, Peking University People's Hospital, Beijing, China
| | - Deqi Yang
- Breast Disease Center, Peking University People's Hospital, Beijing, China
| | - Fei Xie
- Breast Disease Center, Peking University People's Hospital, Beijing, China
| | - Peng Liu
- Breast Disease Center, Peking University People's Hospital, Beijing, China
| | - Jiajia Guo
- Breast Disease Center, Peking University People's Hospital, Beijing, China
| | - Jiaqing Zhang
- Breast Disease Center, Peking University People's Hospital, Beijing, China
| | - Bo Zhou
- Breast Disease Center, Peking University People's Hospital, Beijing, China
- * E-mail:
| |
Collapse
|
29
|
Mei P, Bai J, Shi M, Liu Q, Li Z, Fan Y, Zheng J. BRMS1 suppresses glioma progression by regulating invasion, migration and adhesion of glioma cells. PLoS One 2014; 9:e98544. [PMID: 24879377 PMCID: PMC4039505 DOI: 10.1371/journal.pone.0098544] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 05/05/2014] [Indexed: 11/21/2022] Open
Abstract
Breast cancer metastasis suppressor 1 (BRMS1) is a metastasis suppressor gene in several solid tumors. However, the expression and function of BRMS1 in glioma have not been reported. In this study, we investigated whether BRMS1 play a role in glioma pathogenesis. Using the tissue microarray technology, we found that BRMS1 expression is significantly decreased in glioma compared with tumor adjacent normal brain tissue (P<0.01, χ2 test) and reduced BRMS1 staining is associated with WHO stages (P<0.05, χ2 test). We also found that BRMS1 was significantly downregulated in glioma cell lines compared to normal human astrocytes (P<0.01, χ2 test). Furthermore, we demonstrated that BRMS1 overexpression inhibited glioma cell invasion by suppressing uPA, NF-κB, MMP-2 expression and MMP-2 enzyme activity. Moreover, our data showed that overexpression of BRMS1 inhibited glioma cell migration and adhesion capacity compared with the control group through the Src-FAK pathway. Taken together, this study suggested that BRMS1 has a role in glioma development and progression by regulating invasion, migration and adhesion activities of cancer cells.
Collapse
Affiliation(s)
- Pengjin Mei
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, China
- Jiangsu Key Laboratory of Biological Cancer Therapy, Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Jin Bai
- Jiangsu Key Laboratory of Biological Cancer Therapy, Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Meilin Shi
- Jiangsu Key Laboratory of Biological Cancer Therapy, Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Qinghua Liu
- Jiangsu Key Laboratory of Biological Cancer Therapy, Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Zhonglin Li
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Yuechao Fan
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, China
- * E-mail: (JZ); (YF)
| | - Junnian Zheng
- Jiangsu Key Laboratory of Biological Cancer Therapy, Xuzhou Medical College, Xuzhou, Jiangsu, China
- Department of Medical Oncology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, China
- * E-mail: (JZ); (YF)
| |
Collapse
|
30
|
Cvetković D, Babwah AV, Bhattacharya M. Kisspeptin/KISS1R System in Breast Cancer. J Cancer 2013; 4:653-61. [PMID: 24155777 PMCID: PMC3805993 DOI: 10.7150/jca.7626] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Accepted: 09/22/2013] [Indexed: 01/18/2023] Open
Abstract
Kisspeptins (KP), peptide products of the kisspeptin-1 (KISS1) gene are the endogenous ligands for a G protein-coupled receptor (GPCR) - KP receptor (KISS1R). KISS1R couples to the Gαq/11 signaling pathway. KISS1 is a metastasis suppressor gene and the KP/KISS1R signaling has anti-metastatic and tumor-suppressant effects in numerous human cancers. On the other hand, recent studies indicate that KP/KISS1R pathway plays detrimental roles in breast cancer. In this review, we summarize recent developments in the understanding of the mechanisms regulating KP/KISS1R signaling in breast cancer metastasis.
Collapse
Affiliation(s)
- Donna Cvetković
- 1. Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada, N6A 5C1
| | | | | |
Collapse
|
31
|
Ji K, Ye L, Mason MD, Jiang WG. The Kiss-1/Kiss-1R complex as a negative regulator of cell motility and cancer metastasis (Review). Int J Mol Med 2013; 32:747-54. [PMID: 23969598 DOI: 10.3892/ijmm.2013.1472] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2013] [Accepted: 07/19/2013] [Indexed: 12/22/2022] Open
Abstract
Metastasis is a complex multistep process that involves the impairment of cell-cell adhesion in the neoplastic epithelium, invasion into adjacent tissues and the dissemination of cancer cells through the lymphatic and haematogenous routes. The inhibition of the metastatic process at an early stage has become a hot topic in cancer research. The Kiss-1 gene, initially described as a suppressor of metastasis in malignant melanoma, encodes the Kiss-1 protein which can be processed to other peptides, e.g., Kisspeptin-10, Kisspeptin-13, Kisspeptin-14 and Kisspeptin-54. These peptides are endogenous ligands of the Kiss‑1 receptor (Kiss-1R), a G protein-coupled receptor (GPR) also known as hOT7T175, AXOR12 or GPR54. The Kiss-1 gene has been suggested as a suppressor of metastasis in a various types of cancer, including gastric cancer, oesophageal carcinoma, pancreatic, ovarian, bladder and prostate cancer, through the regulation of cellular migration and invasion. In the current review, we summarise the current understanding of the role of Kiss‑1 and Kiss‑1R in cancer and cancer metastasis.
Collapse
Affiliation(s)
- Ke Ji
- Metastasis and Angiogenesis Research Group, Cardiff University School of Medicine, Cardiff, Wales CF14 4XN, UK
| | | | | | | |
Collapse
|
32
|
Stark AM, Schem C, Maass N, Hugo HH, Jonat W, Mehdorn HM, Held-Feindt J. Expression of metastasis suppressor gene maspin is reduced in breast cancer brain metastases and correlates with the estrogen receptor status. Neurol Res 2013; 32:303-8. [DOI: 10.1179/016164109x12518779082192] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
33
|
Okugawa Y, Inoue Y, Tanaka K, Toiyama Y, Shimura T, Okigami M, Kawamoto A, Hiro J, Saigusa S, Mohri Y, Uchida K, Kusunoki M. Loss of the metastasis suppressor gene KiSS1 is associated with lymph node metastasis and poor prognosis in human colorectal cancer. Oncol Rep 2013; 30:1449-54. [PMID: 23784200 DOI: 10.3892/or.2013.2558] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Accepted: 05/02/2013] [Indexed: 12/13/2022] Open
Abstract
Cancer research is currently focused on blocking the metastatic process at its early steps. Some particularly attractive targets are metastasis suppressor genes, which control cancer cell dissemination. The aim of this study was to clarify the relationship between the expression of KiSS1, a metastasis suppressor gene, and disease progression in colorectal cancer patients. One-hundred and seventy-five patients who underwent surgery for colorectal cancer were enrolled in this study. We analyzed KiSS1 mRNA expression by real-time reverse transcription PCR in colorectal cancer tissue and paired adjacent normal mucosa. KiSS1 protein expression in early- and advanced-stage colorectal cancer samples was determined by immunohistochemical analysis. Decreased KiSS1 expression was significantly associated with lymph node metastasis and was an independent prognostic factor. Logistic regression analysis revealed that decreased KiSS1 expression was an independent risk factor for lymph node metastasis. Immunohistochemical analysis indicated that KiSS1 was highly expressed in the cell cytoplasm of early-stage colorectal cancer cells. The loss of KiSS1 appears to correlate with the progression of lymph node metastasis. An assessment of KiSS1 expression may assist in the accurate colorectal cancer diagnosis and may contribute to predict clinical outcomes.
Collapse
Affiliation(s)
- Yoshinaga Okugawa
- Department of Gastrointestinal and Pediatric Surgery, Division of Reparative Medicine, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
KiSS1 and its cognate G-protein-coupled receptor, GPR54, have diverse functions. While KiSS1 and GPR54 have been intensively studied in physiology, their role in cancer is still unclear. In cancer, KiSS1 and GPR54 have been known to suppress metastasis by inhibiting cancer cell motility. However, recent studies suggest that KiSS1 and GPR54 have varied roles even in cancer development and metastasis. Here, we examine recent advances in understanding the roles of KiSS1 and GPR54 in cancer development and metastasis.
Collapse
|
35
|
Abstract
Metastasis is responsible for most cancer mortality. The process of metastasis is complex, requiring the coordinated expression and fine regulation of many genes in multiple pathways in both the tumor and host tissues. Identification and characterization of the genetic programs that regulate metastasis is critical to understanding the metastatic process and discovering molecular targets for the prevention and treatment of metastasis. Genomic approaches and functional genomic analyses can systemically discover metastasis genes. In this review, we summarize the genetic tools and methods that have been used to identify and characterize the genes that play critical roles in metastasis.
Collapse
Affiliation(s)
- Jinchun Yan
- University of Washington Medical Center, 1959 N. E. Pacific Street, Seattle, WA 98195, USA.
| | | |
Collapse
|
36
|
Roberts MR, Hong CC, Edge SB, Yao S, Bshara W, Higgins MJ, Freudenheim JL, Ambrosone CB. Case-only analyses of the associations between polymorphisms in the metastasis-modifying genes BRMS1 and SIPA1 and breast tumor characteristics, lymph node metastasis, and survival. Breast Cancer Res Treat 2013; 139:873-85. [PMID: 23771732 DOI: 10.1007/s10549-013-2601-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Accepted: 06/06/2013] [Indexed: 12/22/2022]
Abstract
Lymph node metastases and tumor characteristics predict breast cancer prognosis but correlate imperfectly with likelihood of metastatic relapse. Discovery of genetic polymorphisms affecting metastasis may improve identification of patients requiring aggressive adjuvant therapy to prevent recurrence. We investigated associations between several variants in the BRMS1 and SIPA1 metastasis-modifying genes and lymph node metastases, tumor subtype and grade, recurrence, disease-free survival, and overall survival. This cross-sectional and prospective prognostic analysis included 859 patients who received surgery for incident breast cancer at Roswell Park Cancer Institute, participated in the DataBank and BioRepository shared resource, and had DNA, clinical, and pathology data available for analysis. Genotyping for BRMS1 (rs11537993, rs3116068, and rs1052566) and SIPA1 (rs75894763, rs746429, rs3741378, and rs2306364) polymorphisms was performed using Sequenom(®) iPLEX Gold and Taqman(®) real-time PCR assays. Logistic and Cox proportional hazards regressions were used to estimate odds ratios (OR) and hazard ratios (HR), respectively. BRMS1 rs1052566 heterozygous individuals were more likely to have node-positive tumors (OR = 1.58, 95 % CI 1.13-2.23), although there was no dose-response relationship, and those with at least one variant allele were less likely to have the luminal B subtype (AG + AA: OR = 0.59, 95 % CI 0.36-0.98). BRMS1 rs3116068 was associated with increased likelihood of having the luminal B and the HER2-enriched tumor subtype (P trend = 0.03). Two SIPA1 SNPs, rs746429 and rs2306364, were associated with decreased risk of triple-negative tumors (P trend = 0.04 and 0.07, respectively). Presence of 8 or more risk alleles was associated with an increased likelihood of having a node-positive tumor (OR = 2.14, 95 % CI 1.18-3.36, P trend = 0.002). There were no significant associations with survival. Polymorphisms in metastasis-associated genes may be related to tumor characteristics and lymph node metastasis, but not survival. Future evaluation of metastasis-modifying gene variants is necessary to better understand the biology of metastasis.
Collapse
Affiliation(s)
- Michelle R Roberts
- Department of Cancer Prevention and Control, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA.
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Guo Y, Ma J, Wu L, Wang Q, Li X, Li X, Zhang Y, Zhang J, Yao L, Zhang J, Liu W. Hyperthermia-induced NDRG2 upregulation inhibits the invasion of human hepatocellular carcinoma via suppressing ERK1/2 signaling pathway. PLoS One 2013; 8:e61079. [PMID: 23630579 PMCID: PMC3632536 DOI: 10.1371/journal.pone.0061079] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Accepted: 03/05/2013] [Indexed: 12/23/2022] Open
Abstract
Hyperthermia (HT) has been proven to be able to alter the invasion capacity of cancer cells. However, the detailed mechanisms responsible for the anti-metastasis effects of HT have not been elucidated. N-myc downstream-regulated gene 2 (NDRG2), as a member of the NDRG family, has been suggested to be highly responsive to various stresses and is associated with tumor suppression. The present study aimed to investigate the biological role of NDRG2 in the invasion of human hepatocellular carcinoma (HCC) cells exposed to HT. We found that NDRG2 could be induced by HT at 45°C. In addition, NDRG2 overexpression inhibited the expression of matrix metallo proteinases-2 (MMP-2) and MMP-9 as well as the invasion of HCC cells, whereas knockingdown NDRG2 reversed the anti-invasion effect of HT in vivo. Further investigation revealed that the phosphorylation level of ERK1/2, but not that of JNK and p38MAPK, was reduced in NDRG2 overexpressing cells. Moreover, the knockdown of NDRG2 expression resulted in increased cell invasion, which was rescued by treating the HepG2 cells with the ERK1/2 inhibitor PD98059, but not with the p38MAPK inhibitor SB203580 or the JNK inhibitor SP600125. Finally, the synergistic cooperation of HT at 43°C and NDRG2 expression effectively reduced cytotoxicity and promoted the anti-invasion effect of HT at 45°C. Taken together, these data suggest that NDRG2 can be induced by HT and that it mediates the HT-caused inhibition of invasion in HCC cells by suppressing the ERK1/2 signaling pathway. The combined application of constitutive NDRG2 expression with HT may yield an optimized therapeutic benefit.
Collapse
Affiliation(s)
- Yan Guo
- Department of Oncology, State Key Discipline of Cell Biology, Xijing Hospital, the Fourth Military Medical University, Shaanxi, China
| | - Ji Ma
- Department of Oncology, State Key Discipline of Cell Biology, Xijing Hospital, the Fourth Military Medical University, Shaanxi, China
| | - Lin Wu
- The State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, the Fourth Military Medical University, Shaanxi, China
| | - Qianrong Wang
- Department of Oncology, State Key Discipline of Cell Biology, Xijing Hospital, the Fourth Military Medical University, Shaanxi, China
| | - Xia Li
- The State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, the Fourth Military Medical University, Shaanxi, China
| | - Xiaoming Li
- Department of Neurosurgery, Xijing Hospital, the Fourth Military Medical University, Shaanxi, China
| | - Yuan Zhang
- Department of Oncology, State Key Discipline of Cell Biology, Xijing Hospital, the Fourth Military Medical University, Shaanxi, China
| | - Jian Zhang
- The State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, the Fourth Military Medical University, Shaanxi, China
| | - Libo Yao
- The State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, the Fourth Military Medical University, Shaanxi, China
| | - Jing Zhang
- The State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, the Fourth Military Medical University, Shaanxi, China
- Experiment Teaching Center, School of Basic Medicine, the Fourth Military Medical University, Shaanxi, China
- * E-mail: (JZ); (WL)
| | - Wenchao Liu
- Department of Oncology, State Key Discipline of Cell Biology, Xijing Hospital, the Fourth Military Medical University, Shaanxi, China
- * E-mail: (JZ); (WL)
| |
Collapse
|
38
|
Daphu I, Sundstrøm T, Horn S, Huszthy PC, Niclou SP, Sakariassen PØ, Immervoll H, Miletic H, Bjerkvig R, Thorsen F. In vivo animal models for studying brain metastasis: value and limitations. Clin Exp Metastasis 2013; 30:695-710. [DOI: 10.1007/s10585-013-9566-9] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Accepted: 01/07/2013] [Indexed: 01/16/2023]
|
39
|
Zang YW, Gu XD, Xiang JB, Chen ZY. Brain metastases from colorectal cancer: microenvironment and molecular mechanisms. Int J Mol Sci 2012; 13:15784-800. [PMID: 23443093 PMCID: PMC3546661 DOI: 10.3390/ijms131215784] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Revised: 11/05/2012] [Accepted: 11/12/2012] [Indexed: 01/02/2023] Open
Abstract
Colorectal cancer is one of the most common digestive tract malignancies in the world. Owing to the newer and more effective systemic therapies, the life of colorectal cancer patients can be remarkably prolonged, and the incidence of brain metastases is increasing. However, little is known about the underlying mechanisms of brain metastasis from colorectal cancer. Here we review the tumor microenvironment and metastasis associated molecules in brain metastases from colorectal cancer. A further understanding of these mechanisms will help us to propose better strategies for colorectal cancer patients with brain metastasis and improve their life quality.
Collapse
Affiliation(s)
| | | | - Jian-Bin Xiang
- Department of General Surgery, Huashan Hospital, Fudan University, 12 Middle Wulumiqi Road, Shanghai 200040, China; E-Mails: (Y.-W.Z.); (X.-D.G.); (J.-B.X.)
| | - Zong-You Chen
- Department of General Surgery, Huashan Hospital, Fudan University, 12 Middle Wulumiqi Road, Shanghai 200040, China; E-Mails: (Y.-W.Z.); (X.-D.G.); (J.-B.X.)
| |
Collapse
|
40
|
Fokas E, Steinbach JP, Rödel C. Biology of brain metastases and novel targeted therapies: time to translate the research. Biochim Biophys Acta Rev Cancer 2012; 1835:61-75. [PMID: 23142311 DOI: 10.1016/j.bbcan.2012.10.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Revised: 10/26/2012] [Accepted: 10/30/2012] [Indexed: 01/23/2023]
Abstract
Brain metastases (BM) occur in 20% to 40% of patients with cancer and result in significant morbidity and poor survival. The main therapeutic options include surgery, whole brain radiotherapy, stereotactic radiosurgery and chemotherapy. Although significant progress has been made in diagnostic and therapeutic methods, the prognosis in these patients remains poor. Furthermore, the poor penetrability of chemotherapy agents through the blood brain barrier (BBB) continues to pose a challenge in the management of this disease. Preclinical evidence suggests that new targeted treatments can improve local tumor control but our clinical experience with these agents remains limited. In addition, several clinical studies with these novel agents have produced disappointing results. This review will examine the knowledge of targeted therapies in BM. The preclinical and clinical evidence of their use in BM induced by breast cancer, non-small cell lung cancer and melanoma will be presented. In addition, we will discuss the role of antiangiogenic and radiosensitising agents in the treatment of BM and the current strategies available to increase BBB permeability. A better understanding of the mechanism of action of these agents will help us to identify the best targets for testing in future clinical studies.
Collapse
Affiliation(s)
- Emmanouil Fokas
- Department of Radiation Therapy and Oncology, Johann Wolfgang Goethe University, Frankfurt, Germany.
| | | | | |
Collapse
|
41
|
Effect of breast-cancer metastasis suppressor 1 (BRMS1) on growth and metastasis of human gastric cancer cells in vivo. ACTA ACUST UNITED AC 2012. [DOI: 10.1007/s10330-011-0920-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
42
|
Nieder C, Grosu AL, Mehta MP. Brain metastases research 1990-2010: pattern of citation and systematic review of highly cited articles. ScientificWorldJournal 2012; 2012:721598. [PMID: 23028253 PMCID: PMC3458272 DOI: 10.1100/2012/721598] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Accepted: 08/26/2012] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND High and continuously increasing research activity related to different aspects of prevention, prediction, diagnosis and treatment of brain metastases has been performed between 1990 and 2010. One of the major databases contains 2695 scientific articles that were published during this time period. Different measures of impact, visibility, and quality of published research are available, each with its own pros and cons. For this overview, article citation rate was chosen. RESULTS Among the 10 most cited articles, 7 reported on randomized clinical trials. Nine covered surgical or radiosurgical approaches and the remaining one a widely adopted prognostic score. Overall, 30 randomized clinical trials were published between 1990 and 2010, including those with phase II design and excluding duplicate publications, for example, after longer followup or with focus on secondary endpoints. Twenty of these randomized clinical trials were published before 2008. Their median number of citations was 110, range 13-1013, compared to 5-6 citations for all types of publications. Annual citation rate appeared to gradually increase during the first 2-3 years after publication before reaching high levels. CONCLUSIONS A large variety of preclinical and clinical topics achieved high numbers of citations. However, areas such as quality of life, side effects, and end-of-life care were underrepresented. Efforts to increase their visibility might be warranted.
Collapse
Affiliation(s)
- Carsten Nieder
- Department of Oncology and Palliative Medicine, Nordland Hospital, 8092 Bodø, Norway.
| | | | | |
Collapse
|
43
|
Cui RX, Liu N, He QM, Li WF, Huang BJ, Sun Y, Tang LL, Chen M, Jiang N, Chen L, Yun JP, Zeng J, Guo Y, Wang HY, Ma J. Low BRMS1 expression promotes nasopharyngeal carcinoma metastasis in vitro and in vivo and is associated with poor patient survival. BMC Cancer 2012; 12:376. [PMID: 22931099 PMCID: PMC3517767 DOI: 10.1186/1471-2407-12-376] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2012] [Accepted: 08/14/2012] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Breast cancer metastasis suppressor 1 (BRMS1) is a metastasis suppressor gene. This study aimed to investigate the impact of BRMS1 on metastasis in nasopharyngeal carcinoma (NPC) and to evaluate the prognostic significance of BRMS1 in NPC patients. METHODS BRMS1 expression was examined in NPC cell lines using quantitative reverse transcription-polymerase chain reaction and Western blotting. NPC cells stably expressing BRMS1 were used to perform wound healing and invasion assays in vitro and a murine xenograft assay in vivo. Immunohistochemical staining was performed in 274 paraffin-embedded NPC specimens divided into a training set (n = 120) and a testing set (n = 154). RESULTS BRMS1 expression was down-regulated in NPC cell lines. Overexpression of BRMS1 significantly reversed the metastatic phenotype of NPC cells in vitro and in vivo. Importantly, low BRMS1 expression was associated with poor distant metastasis-free survival (DMFS, P < 0.001) and poor overall survival (OS, P < 0.001) in the training set; these results were validated in the testing set and overall patient population. Cox regression analysis demonstrated that low BRMS1 expression was an independent prognostic factor for DMFS and OS in NPC. CONCLUSIONS Low expression of the metastasis suppressor BRMS1 may be an independent prognostic factor for poor prognosis in NPC patients.
Collapse
Affiliation(s)
- Rui-Xue Cui
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
de Carvalho CCCR, Caramujo MJ. Tumour metastasis as an adaptation of tumour cells to fulfil their phosphorus requirements. Med Hypotheses 2012; 78:664-7. [PMID: 22391031 DOI: 10.1016/j.mehy.2012.02.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Accepted: 02/09/2012] [Indexed: 11/30/2022]
Abstract
Inorganic phosphate (Pi) is a vital component of nucleotides, membrane phospholipids, and phosphorylated intermediates in cellular signalling. The Growth Rate Hypothesis (GRH) states that fast growing organisms should be richer in phosphorus (relatively low C:P and N:P cell content) than slow developing organisms as a result of high ribosome biogenesis. Cells that proliferate rapidly, such as cancer cells, require a high amount of ribosomes and other P-rich RNA components that are necessary to manufacture proteins. The GRH hypothesis may be applied to cancer predicting that tumour cells are richer in phosphorus than the surrounding tissue, and that they resort to metastasis in order to meet their nutrient demands. Considering that the cells most P-deprived should be located in the inner parts of the tumour we propose that changes in the membrane of these cells favour the detachment of the more peripheral cells.
Collapse
Affiliation(s)
- Carla C C R de Carvalho
- IBB-Institute for Biotechnology and Bioengineering, Centre for Biological and Chemical Engineering, Instituto Superior Técnico, Av. Rovisco Pais, 1049-001 Lisboa, Portugal.
| | | |
Collapse
|
45
|
Attributes of brain metastases from breast and lung cancer. Int J Clin Oncol 2012; 18:396-401. [PMID: 22383025 DOI: 10.1007/s10147-012-0392-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2011] [Accepted: 02/12/2012] [Indexed: 10/28/2022]
Abstract
BACKGROUND Most brain metastases arise from breast and lung cancers. Few studies compare the brain regions they involve, their numbers and intrinsic attributes. METHODS Records of all patients referred to Radiation Oncology for treatment of symptomatic brain metastases were obtained. Computed tomography (n = 56) or magnetic resonance imaging (n = 72) brain scans were reviewed. RESULTS Data from 68 breast and 62 lung cancer patients were compared. Brain metastases presented earlier in the course of the lung than of the breast cancer patients (p = 0.001). There were more metastases in the cerebral hemispheres of the breast than of the lung cancer patients (p = 0.014). More breast than lung cancer patients had cerebellar metastases (p = 0.001). The number of cerebral hemisphere metastases and presence of cerebellar metastases were positively correlated (p = 0.001). The prevalence of at least one metastasis surrounded with >2 cm of edema was greater for the lung than for the breast patients (p = 0.019). The primary tumor type, rather than the scanning method, correlated with differences between these variables. CONCLUSIONS Brain metastases from lung occur earlier, are more edematous, but fewer in number than those from breast cancers. Cerebellar brain metastases are more frequent in breast cancer.
Collapse
|
46
|
Sheng XJ, Zhou YQ, Song QY, Zhou DM, Liu QC. Loss of breast cancer metastasis suppressor 1 promotes ovarian cancer cell metastasis by increasing chemokine receptor 4 expression. Oncol Rep 2011; 27:1011-8. [PMID: 22200669 PMCID: PMC3583538 DOI: 10.3892/or.2011.1596] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2011] [Accepted: 12/02/2011] [Indexed: 12/22/2022] Open
Abstract
Breast cancer metastasis suppressor 1 (BRMS1) is a predominantly nuclear protein that differentially regulates the expression of multiple genes, leading to suppression of metastasis without affecting orthotopic tumor growth. It has been demonstrated that BRMS1 may be correlated with advanced ovarian cancer. The aim of this study was to investigate the mechanisms of BRMS1 involvement in ovarian cancer metastasis. We constructed a plasmid containing a short hairpin RNA (shRNA) against BRMS1 and transfected it into the ovarian cancer cell line OVCAR3. Real-time reverse transcription polymerase chain reaction (real-time PCR) and Western blot analyses demonstrated that BRMS1 expression was efficiently downregulated. Stable suppression of BRMS1 significantly enhanced cell adhesion, migration, invasion and angiogenesis. We also found that chemokine receptor 4 (CXCR4) was upregulated at both the mRNA and protein levels. When approaching for the mechanism, we discovered that activation of the nuclear factor-κB (NF-κB) signaling pathway mediated CXCR4 upregulation, as demonstrated by the electrophoretic mobility shift assay (EMSA). Collectively, these results suggest that attenuation of BRMS1 may play a critical role in promoting migration, invasion and angiogenesis of ovarian cancer cells and BRMS1 may regulate the metastatic potential at least in part through upregulation of CXCR4 via NF-κB activation. Restoration of BRMS1 function is thus a potential new strategy for treating human ovarian cancer.
Collapse
Affiliation(s)
- Xiu-Jie Sheng
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical College, Guangzhou 510150, PR China.
| | | | | | | | | |
Collapse
|
47
|
Ulasov IV, Kaverina NV, Pytel P, Thaci B, Liu F, Hurst DR, Welch DR, Sattar HA, Olopade OI, Baryshnikov AY, Kadagidze ZG, Lesniak MS. Clinical significance of KISS1 protein expression for brain invasion and metastasis. Cancer 2011; 118:2096-105. [PMID: 21928364 DOI: 10.1002/cncr.26525] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2011] [Revised: 07/07/2011] [Accepted: 08/04/2011] [Indexed: 12/27/2022]
Abstract
BACKGROUND Metastases to the brain represent a feared complication and contribute to the morbidity and mortality of breast cancer. Despite improvements in therapy, prognostic factors for development of metastases are lacking. KISS1 is a metastasis suppressor that demonstrates inhibition of metastases formation in several types of cancer. The purpose of this study was to determine the importance of KISS1 expression in breast cancer progression and the development of intracerebral lesions. METHODS In this study, we performed a comparative analysis of 47 brain metastases and 165 primary breast cancer specimens by using the antihuman KISS1 antibody. To compare KISS1 expression between different groups, we used a 3-tier score and the automated score computer software (ACIS) evaluation. To reveal association between mRNA and protein expression, we used quantitative reverse transcription-polymerase chain reaction (qRT-PCR) analysis. Significance of immunohistochemistry stainings was correlated with clinicopathological data. RESULTS We identified that KISS1 expression is significantly higher in primary breast cancer compared with brain metastases (P < .05). The mRNA analysis performed on 33 selected ductal carcinoma brain metastatic lesions and 36 primary ductal carcinomas revealed a statistically significant down-regulation of KISS1 protein in metastatic cases (P = .04). Finally, we observed a significant correlation between expression of KISS1 and metastasis-free survival (P = .04) along with progression of breast cancer and expression of KISS1 in primary breast cancer specimens (P = .044). CONCLUSIONS In conclusion, our study shows that breast cancer expresses KISS1. Cytoplasmic expression of KISS1 may be used as a prognostic marker for increased risk of breast cancer progression.
Collapse
Affiliation(s)
- Ilya V Ulasov
- The Brain Tumor Cancer Center, The University of Chicago, Chicago, Illinois, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Knopeke MT, Ritschdorff ET, Clark R, Vander Griend DJ, Khan S, Thobe M, Shear JB, Rinker-Schaeffer CW. Building on the foundation of daring hypotheses: using the MKK4 metastasis suppressor to develop models of dormancy and metastatic colonization. FEBS Lett 2011; 585:3159-65. [PMID: 21925502 DOI: 10.1016/j.febslet.2011.09.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Accepted: 09/07/2011] [Indexed: 01/04/2023]
Abstract
The identification of a novel metastasis suppressor function for the MAP Kinase Kinase 4 protein established a role for the stress-activated kinases in regulating the growth of disseminated cancer cells. In this review, we describe MKK4's biological mechanism of action and how this information is being used to guide the development of new models to study cancer cell dormancy and metastatic colonization. Specifically, we describe the novel application of microvolume structures, which can be modified to represent characteristics similar to those that cancer cells experience at metastatic sites. Although MKK4 is currently one of many known metastasis suppressors, this field of research started with a single daring hypothesis, which revolutionized our understanding of metastasis, and opened up new areas of exploration for basic research. The combination of our increasing knowledge of metastasis suppressors and such novel technologies provide hope for possible clinical interventions to prevent suffering from the burden of metastatic disease.
Collapse
Affiliation(s)
- Matthew T Knopeke
- The Section of Urology, Department of Surgery, The University of Chicago, Chicago, IL 60637, United States
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Cho SG, Wang Y, Rodriguez M, Tan K, Zhang W, Luo J, Li D, Liu M. Haploinsufficiency in the prometastasis Kiss1 receptor Gpr54 delays breast tumor initiation, progression, and lung metastasis. Cancer Res 2011; 71:6535-46. [PMID: 21852382 DOI: 10.1158/0008-5472.can-11-0329] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Activation of KISS1 receptor (KISS1R or GPR54) by its ligands (Kisspeptins) regulates a diverse function both in normal physiology and pathophysiology. In cancer, KISS1R has been implicated in tumor angiogenesis and metastasis, but a broader evaluation of KISS1R in tumorigenesis and tumor progression is yet to be conducted. In this study, we used mouse models of Kiss1r gene knockout and mouse mammary tumor virus-polyoma virus middle T antigen (MMTV-PyMT)-induced breast cancer to conduct such an evaluation. Kiss1r heterozygosity in MMTV-PyMT mice was sufficient to attenuate breast cancer initiation, growth, latency, multiplicity, and lung metastasis. To confirm these effects and assess possible contributions of endogenous ligands, we isolated primary tumor cells from PyMT/Kiss1r(+/+) and PyMT/Kiss1r(+/-) mice and compared their phenotypes by in vitro and in vivo assays. Kiss1r loss attenuated in vitro tumorigenic properties as well as tumor growth in vivo in immunocompromised NOD.SCID/NCr mice. Kiss1r activation in these cells, resulting from the addition of its ligand Kisspeptin-10, resulted in RhoA activation and RhoA-dependent gene expression through the Gαq-p63RhoGEF signaling pathway. Anchorage-independent growth was tightly linked to dose-dependent regulation of RhoA by Kiss1r. In support of these results, siRNA-mediated knockdown of KISS1R or inactivation of RhoA in human MCF10A breast epithelial cells overexpressing H-RasV12 was sufficient to reduce Ras-induced anchorage-independent growth. In summary, we concluded that Kiss1r attenuation was sufficient to delay breast tumor initiation, progression, and metastasis through inhibitory effects on the downstream Gαq-p63RhoGEF-RhoA signaling pathway.
Collapse
Affiliation(s)
- Sung-Gook Cho
- Center for Cancer and Stem Cell Biology, Institute of Bioscience and Technology, Texas A&M System Health Science Center, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Promoter methylation of BRMS1 correlates with smoking history and poor survival in non-small cell lung cancer patients. Lung Cancer 2011; 74:305-9. [PMID: 21726917 DOI: 10.1016/j.lungcan.2011.03.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2010] [Revised: 02/22/2011] [Accepted: 03/06/2011] [Indexed: 01/29/2023]
Abstract
PURPOSE To investigate whether methylation of BRMS1 is associated with clinical outcomes in patients with NSCLC. METHODS Methylation status of BRMS1 was examined in 325 NSCLC patients who were treated with surgery. We analyzed associations between the methylation of BRMS1 genes separately and available epidemiologic and clinical information including smoking status, gender, age, and histological type, or the stage of the tumor. RESULTS In the cohort of 325 NSCLC cases, 152 samples were identified as methylated (46.77%). Promoter methylation of BRMS1 was present only in 6 specimens (8.42%) in adjacent non-cancerous tissues (P=2.257 × 10(-14)). Patient smoking history had a positive correlation with methylation rate of BRMS1 (OR=2.508, 95%CI(1.516, 4.151)). Compared with unmethylated group, methylated group showed the lower level of BRMS1 mRNA (P=0.013). And patients with a high level of BRMS1 mRNA expression had significantly better overall survival than those with low expression (P=0.002). Multivariate Cox proportional hazard regression analysis also showed that promoter methylation of BRMS1 was significantly unfavorable prognostic factors (hazard ratio, 1.912; 95% CI, and 1.341-2.726). CONCLUSIONS These results provide clinical evidence to support the notion that BRMS1 is a NSCLC metastasis suppressor gene. Measuring methylation status of BRMS1 promotor is a useful marker for identifying NSCLC patients with worse disease-free survival.
Collapse
|